Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 288
Filtrar
1.
Cells ; 12(14)2023 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-37508570

RESUMO

Tumor therapy escape due to undesired side effects induced by treatment, such as prosurvival autophagy or cellular senescence, is one of the key mechanisms of resistance that eventually leads to tumor dormancy and recurrence. Glioblastoma is the most frequent and practically incurable neoplasm of the central nervous system; thus, new treatment modalities have been investigated to find a solution more effective than the currently applied standards based on temozolomide. The present study examined the newly synthesized compounds of aziridine-hydrazide hydrazone derivatives to determine their antineoplastic potential against glioblastoma cells in vitro. Although the output of our investigation clearly demonstrates their proapoptotic activity, the cytotoxic effect appeared to be blocked by treatment-induced autophagy, the phenomenon also detected in the case of temozolomide action. The addition of an autophagy inhibitor, chloroquine, resulted in a significant increase in apoptosis triggered by the tested compounds, as well as temozolomide. The new aziridine-hydrazide hydrazone derivatives, which present cytotoxic potential against glioblastoma cells comparable to or even higher than that of temozolomide, show promising results and, thus, should be further investigated as antineoplastic agents. Moreover, our findings suggest that the combination of an apoptosis inducer with an autophagy inhibitor could optimize chemotherapeutic efficiency, and the addition of an autophagy inhibitor should be considered as an optional adjunctive therapy minimizing the risk of tumor escape from treatment.


Assuntos
Antineoplásicos , Aziridinas , Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Cloroquina/farmacologia , Hidrazonas/farmacologia , Hidrazinas/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Autofagia , Aziridinas/farmacologia , Aziridinas/uso terapêutico
2.
Cancer Gene Ther ; 29(7): 1021-1032, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34837065

RESUMO

Advances in the field of cancer immunotherapy have stimulated renewed interest in adenoviruses as oncolytic agents. Clinical experience has shown that oncolytic adenoviruses are safe and well tolerated but possess modest single-agent activity. One approach to improve the potency of oncolytic viruses is to utilise their tumour selectivity to deliver genes encoding prodrug-activating enzymes. These enzymes can convert prodrugs into cytotoxic species within the tumour; however, these cytotoxins can interfere with viral replication and limit utility. In this work, we evaluated the activity of a nitroreductase (NTR)-armed oncolytic adenovirus ONYX-411NTR in combination with the clinically tested bioreductive prodrug PR-104. Both NTR-expressing cells in vitro and xenografts containing a minor population of NTR-expressing cells were highly sensitive to PR-104. Pharmacologically relevant prodrug exposures did not interfere with ONYX-411NTR replication in vitro. In vivo, prodrug administration increased virus titre and improved virus distribution within tumour xenografts. Colonisation of tumours with high ONYX-411NTR titre resulted in NTR expression and prodrug activation. The combination of ONYX-411NTR with PR-104 was efficacious against HCT116 xenografts, whilst neither prodrug nor virus were active as single agents. This work highlights the potential for future clinical development of NTR-armed oncolytic viruses in combination with bioreductive prodrugs.


Assuntos
Aziridinas , Neoplasias , Terapia Viral Oncolítica , Pró-Fármacos , Adenoviridae , Aziridinas/farmacologia , Aziridinas/uso terapêutico , Humanos , Neoplasias/terapia , Compostos de Mostarda Nitrogenada , Nitrorredutases/genética , Nitrorredutases/metabolismo , Vírus Oncolíticos , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico
3.
Curr Urol Rep ; 19(12): 101, 2018 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-30357541

RESUMO

PURPOSE OF REVIEW: As our molecular understanding of bladder cancer continues to advance, more and more novel agents are entering clinical trials across the spectrum of bladder cancer stages. The clinical trial activity for non-muscle invasive bladder cancer (NMIBC) has been boosted further by the evolution of specific disease states that set more uniform inclusion criteria for clinical trial design. Here, we aimed to review the current clinical trials landscape in non-muscle invasive bladder cancer with respect to these disease states. RECENT FINDINGS: Most active clinical trials focus on high-risk NMIBC in either the BCG-naïve or BCG-unresponsive setting. Strict criteria to define the disease state and a clear pathway to drug registration have encouraged trials for patients with BCG-unresponsive NMIBC. The most promising potential breakthroughs for BCG-naïve patients include alternative BCG strains, immune-priming with intradermal BCG vaccination, and systemic immune checkpoint blockade. The latter therapy is also being actively investigated in multiple trials in BCG-unresponsive NMIBC, along with novel viral agents such as INSTILADRIN (nadofaragene firadenovec) and targeted agents such as oportuzumab monatox. After many years of relative stagnation, multiple new therapies currently under investigation in well-designed clinical trials appear poised for routine clinical implementation in the near future. These therapies should dramatically improve the outcome of patients with NMIBC. We can look forward to the challenges of biomarker-driven drug selection, optimal drug sequencing, and rational combination therapies.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Vacina BCG/uso terapêutico , Carcinoma de Células de Transição/tratamento farmacológico , Neoplasias da Bexiga Urinária/tratamento farmacológico , Administração Intravesical , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/uso terapêutico , Antineoplásicos Hormonais/uso terapêutico , Aziridinas/uso terapêutico , Carcinoma de Células de Transição/patologia , Quimioterapia Adjuvante , Ensaios Clínicos como Assunto , Terapia Combinada , Humanos , Indolquinonas/uso terapêutico , Injeções Intradérmicas , Mitomicina/uso terapêutico , Músculo Liso/patologia , Invasividade Neoplásica , Polissacarídeos Bacterianos/uso terapêutico , Proteínas/uso terapêutico , Proteínas Recombinantes de Fusão , Tamoxifeno/uso terapêutico , Vacinas Tíficas-Paratíficas/uso terapêutico , Neoplasias da Bexiga Urinária/patologia , Procedimentos Cirúrgicos Urológicos
4.
Mini Rev Med Chem ; 16(17): 1374-1391, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27156518

RESUMO

Human African Trypanosomiasis (HAT) is an endemic parasitic disease of sub-Saharan Africa, caused by two subspecies of protozoa belonging to Trypanosoma genus: T. brucei gambiense and T. brucei rhodesiense. In this context the inhibition of the papain-family cysteine proteases rhodesain and TbCatB has to be considered a promising strategy for HAT treatment. Rhodesain, the major cathepsin L-like cysteine protease of T. brucei rhodesiense, is a lysosomal protease essential for parasite survival. It is involved in parasite invasivity, allowing it to cross the blood-brain barrier (BBB) of the human host, causing the second lethal stage of the disease. Moreover, it plays an important role in immunoevasion, being involved in the turnover of variant surface glycoproteins of the T. brucei coat and in the degradation of immunoglobulins, avoiding a specific immune response by the host cells. On the other hand TbCatB, a cathepsin B-like cysteine protease, present in minor abundance in T. brucei, showed a key role in the degradation of host transferrin, which is necessary for iron acquisition by the parasite. In this review article we now discuss the most active peptide, peptidomimetic and non-peptide rhodesain and TbCatB inhibitors as valuable strategy to treat HAT, due also to the complementary role of the two T. brucei proteases.


Assuntos
Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Proteínas de Protozoários/metabolismo , Tripanossomicidas/metabolismo , Aziridinas/química , Aziridinas/metabolismo , Aziridinas/farmacologia , Aziridinas/uso terapêutico , Barreira Hematoencefálica/metabolismo , Cisteína Endopeptidases/química , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/farmacologia , Inibidores de Cisteína Proteinase/uso terapêutico , Humanos , Proteínas de Protozoários/antagonistas & inibidores , Sulfonas/química , Sulfonas/metabolismo , Sulfonas/farmacologia , Sulfonas/uso terapêutico , Tripanossomicidas/química , Tripanossomicidas/farmacologia , Tripanossomicidas/uso terapêutico , Trypanosoma brucei brucei/efeitos dos fármacos , Trypanosoma brucei brucei/enzimologia , Tripanossomíase Africana/tratamento farmacológico
5.
J Control Release ; 222: 9-17, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26655063

RESUMO

Some chemotherapeutic drugs (prodrugs) require activation by an enzyme for efficacy. We and others have demonstrated the ability of probiotic bacteria to grow specifically within solid tumours following systemic administration, and we hypothesised that the natural enzymatic activity of these tumour-localised bacteria may be suitable for activation of certain such chemotherapeutic drugs. Several wild-type probiotic bacteria; Escherichia coli Nissle, Bifidobacterium breve, Lactococcus lactis and Lactobacillus species, were screened against a panel of popular prodrugs. All strains were capable of activating at least one prodrug. E. coli Nissle 1917 was selected for further studies because of its ability to activate numerous prodrugs and its resistance to prodrug toxicity. HPLC data confirmed biochemical transformation of prodrugs to their toxic counterparts. Further analysis demonstrated that different enzymes can complement prodrug activation, while simultaneous activation of multiple prodrugs (CB1954, 5-FC, AQ4N and Fludarabine phosphate) by E. coli was confirmed, resulting in significant efficacy improvement. Experiments in mice harbouring murine tumours validated in vitro findings, with significant reduction in tumour growth and increase in survival of mice treated with probiotic bacteria and a combination of prodrugs. These findings demonstrate the ability of probiotic bacteria, without the requirement for genetic modification, to enable high-level activation of multiple prodrugs specifically at the site of action.


Assuntos
Antineoplásicos/administração & dosagem , Proteínas de Bactérias/metabolismo , Neoplasias/tratamento farmacológico , Probióticos , Pró-Fármacos/administração & dosagem , Animais , Antraquinonas/administração & dosagem , Antraquinonas/uso terapêutico , Antineoplásicos/uso terapêutico , Aziridinas/administração & dosagem , Aziridinas/uso terapêutico , Bifidobacterium/enzimologia , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Enzimas , Escherichia coli/enzimologia , Feminino , Lactobacillus/enzimologia , Lactococcus/enzimologia , Camundongos Endogâmicos BALB C , Neoplasias/patologia , Pró-Fármacos/uso terapêutico , Carga Tumoral/efeitos dos fármacos , Fosfato de Vidarabina/administração & dosagem , Fosfato de Vidarabina/análogos & derivados , Fosfato de Vidarabina/uso terapêutico
6.
Biochem J ; 471(2): 131-53, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26431849

RESUMO

This review examines the vast catalytic and therapeutic potential offered by type I (i.e. oxygen-insensitive) nitroreductase enzymes in partnership with nitroaromatic prodrugs, with particular focus on gene-directed enzyme prodrug therapy (GDEPT; a form of cancer gene therapy). Important first indications of this potential were demonstrated over 20 years ago, for the enzyme-prodrug pairing of Escherichia coli NfsB and CB1954 [5-(aziridin-1-yl)-2,4-dinitrobenzamide]. However, it has become apparent that both the enzyme and the prodrug in this prototypical pairing have limitations that have impeded their clinical progression. Recently, substantial advances have been made in the biodiscovery and engineering of superior nitroreductase variants, in particular development of elegant high-throughput screening capabilities to enable optimization of desirable activities via directed evolution. These advances in enzymology have been paralleled by advances in medicinal chemistry, leading to the development of second- and third-generation nitroaromatic prodrugs that offer substantial advantages over CB1954 for nitroreductase GDEPT, including greater dose-potency and enhanced ability of the activated metabolite(s) to exhibit a local bystander effect. In addition to forging substantial progress towards future clinical trials, this research is supporting other fields, most notably the development and improvement of targeted cellular ablation capabilities in small animal models, such as zebrafish, to enable cell-specific physiology or regeneration studies.


Assuntos
Aziridinas/uso terapêutico , Proteínas de Escherichia coli , Terapia Genética/métodos , Neoplasias Experimentais/terapia , Nitrorredutases , Pró-Fármacos/uso terapêutico , Animais , Evolução Molecular Direcionada , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/uso terapêutico , Humanos , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Nitrorredutases/biossíntese , Nitrorredutases/genética , Nitrorredutases/uso terapêutico
7.
Theranostics ; 4(5): 460-74, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24669276

RESUMO

Metastatic breast cancer is an obdurate cancer type that is not amenable to chemotherapy regimens currently used in clinic. There is a desperate need for alternative therapies to treat this resistant cancer type. Gene-Directed Enzyme Prodrug Therapy (GDEPT) is a superior gene therapy method when compared to chemotherapy and radiotherapy procedures, proven to be effective against many types of cancer in pre-clinical evaluations and clinical trials. Gene therapy that utilizes a single enzyme/prodrug combination targeting a single cellular mechanism needs significant overexpression of delivered therapeutic gene in order to achieve therapy response. Hence, to overcome this obstacle we recently developed a dual therapeutic reporter gene fusion that uses two different prodrugs, targeting two distinct cellular mechanisms in order to achieve effective therapy with a limited expression of delivered transgenes. In addition, imaging therapeutic reporter genes offers additional information that indirectly correlates gene delivery, expression, and functional effectiveness as a theranostic approach. In the present study, we evaluate the therapeutic potential of HSV1-sr39TK-NTR fusion dual suicide gene therapy system that we recently developed, in MDA-MB-231 triple negative breast cancer lung-metastatic lesions in a mouse model. We compared the therapeutic potential of HSV1-sr39TK-NTR fusion with respective dual prodrugs GCV-CB1954 with HSV1-sr39TK/GCV and NTR/CB1954 single enzyme prodrug system in this highly resistant metastatic lesion of the lungs. In vitro optimization of dose and duration of exposure to GCV and CB1954 was performed in MDA-MB-231 cells. Drug combinations of 1 µg/ml GCV and 10 µM CB1954 for 3 days was found to be optimal regimen for induction of significant cell death, as assessed by FACS analysis. In vivo therapeutic evaluation in animal models showed a complete ablation of lung metastatic nodules of MDA-MB-231 triple negative breast cancer cells following two consecutive doses of a combination of GCV (40 mg/kg) and CB1954 (40 mg/kg) administered at 5 day intervals. In contrast, the respective treatment condition in animals expressing HSV1-sr39TK or NTR separately, showed minimal or no effect on tumor reduction as measured by bioluminescence (tumor mass) and [(18)F]-FHBG microPET (TK expression) imaging. These highlight the strong therapeutic effect of the dual fusion prodrug therapy and its use in theranostic imaging of tumor monitoring in living animals by multimodality molecular imaging.


Assuntos
Antineoplásicos/uso terapêutico , Terapia Genética/métodos , Neoplasias Pulmonares/tratamento farmacológico , Metástase Neoplásica/tratamento farmacológico , Pró-Fármacos/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Aziridinas/uso terapêutico , Modelos Animais de Doenças , Glutamina/análogos & derivados , Glutamina/uso terapêutico , Neoplasias Pulmonares/diagnóstico por imagem , Camundongos , Metástase Neoplásica/diagnóstico por imagem , Nitrorredutases/metabolismo , Imagem Óptica/métodos , Compostos Organometálicos/uso terapêutico , Radiografia , Timidina Quinase/metabolismo , Resultado do Tratamento , Neoplasias de Mama Triplo Negativas/diagnóstico por imagem , Neoplasias de Mama Triplo Negativas/secundário
8.
Curr Med Chem ; 19(33): 5745-53, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22998528

RESUMO

Following the demonstration that addition of a 2-cyano group to aziridines prevented DNA alkylation and thus reduced toxicity, many novel 2-cyanoaziridines were synthesized and evaluated as immunomodulating and antitumor agents. They typically reacted with thiols such as cysteine, depleting them and allowing the accumulation of reactive oxygen species. Two of these compounds, azimexon and ciamexon, showed activity against tumors in clinical trials. Imexon was produced by cyclization of 2-cyanoaziridine-1- carboxamide in the presence of hydroxide ions. The two enantiomers were prepared by a process involving chiral chromatography. They were equipotent against cultured tumor cells. Imexon also reacts with thiols and it is especially potent against multiple myeloma in cell cultures. An efficient chemical synthesis and a lyophilization formulation of imexon as a water soluble, injectible drug, were developed. In Phase I and I/II clinical trials imexon showed hints of activity against a variety of tumors, but a randomized double-blind Phase II trial of imexon plus gemcitabine versus gemcitabine alone in pancreatic cancer showed no enhancement of activity above that of gemcitabine alone. This result was disappointing because in cell culture and mice the two compounds were synergistic. Based on a complete response in a Phase I trial, a new Phase II clinical trial of imexon is underway in non-Hodgkins lymphoma.


Assuntos
Antineoplásicos/química , Antineoplásicos/uso terapêutico , Aziridinas/química , Aziridinas/uso terapêutico , Hexanonas/química , Hexanonas/uso terapêutico , Animais , Antineoplásicos/farmacologia , Aziridinas/farmacologia , Ensaios Clínicos como Assunto , Hexanonas/farmacologia , Humanos , Imunomodulação/efeitos dos fármacos , Camundongos , Neoplasias/tratamento farmacológico
9.
Curr Med Chem ; 19(3): 364-85, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22335513

RESUMO

DNA interstrand cross-linking (ICL) agents are an important group of cytotoxic drugs with the capability of binding covalently between two strands of DNA, thereby preventing vital processes such as replication or transcription in dividing cells. In anticancer therapy however, their potential is limited due to the resistance by various mechanisms. In order to develop highly effective antitumor drugs it is necessary to study both effective ICL formations and their subsequent repair mechanisms. This review presents an overview of development over the past decade and the use of both well-known and new DNA interstrand cross-linking agents. Their potential in applications especially as anticancer chemotherapeutics in the framework of current knowledge of repair mechanisms and development of combined chemotherapy is discussed.


Assuntos
Antineoplásicos/química , Reagentes de Ligações Cruzadas/química , DNA/metabolismo , Alcaloides/química , Alcaloides/uso terapêutico , Antraquinonas/química , Antraquinonas/uso terapêutico , Antineoplásicos/uso terapêutico , Azepinas/química , Azepinas/uso terapêutico , Aziridinas/química , Aziridinas/uso terapêutico , Complexos de Coordenação/química , Complexos de Coordenação/uso terapêutico , Reagentes de Ligações Cruzadas/uso terapêutico , DNA/química , Reparo do DNA , Humanos , Mecloretamina/química , Mecloretamina/uso terapêutico , Neoplasias/tratamento farmacológico
10.
Urol Oncol ; 30(1): 64-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-19945311

RESUMO

OBJECTIVES: Apaziquone used intravesically showed significant activity in phase I and II marker lesion studies in non-muscle-invasive bladder cancer. The objective of this study was to assess antitumor activity and safety of 3 different formulations of intravesical apaziquone in an orthotopic rat bladder cancer model. MATERIALS AND METHODS: Female Fischer F344 rats were instilled with 1.5 × 10(6) AY-27 urothelial cell carcinoma cells and divided in 3 treatment groups (n = 10) and 1 placebo group (n = 6). Intravesical treatment was administered for 1 hour on days 2 and 5. Rats were treated with apaziquone in the formulation used in phase I/II clinical trials (group 1); apaziquone with an altered buffering capacity being used in phase III clinical trials (group 2), and apaziquone as in group 2, but without propylene glycol in the diluent (group 3). On days 5 and 14, the bladder wall was inspected by cystoscopy and evaluated for macroscopic tumor growth. After sacrificing the rats (day 14), cystectomy was performed and the bladders were investigated. RESULTS: There were no signs of any toxicity due to the study drug. On histopathologic examination of the bladders 0, 1, and 2 tumors per group were found in group 1, 2, and 3, respectively. In the placebo-treated group, 60% of animals developed tumor, which is comparable to untreated animals. CONCLUSIONS: Apaziquone showed an excellent antitumor activity. The effectiveness of apaziquone in this orthotopic rat bladder tumor model corroborates the clinical observations and implies the validity of this model.


Assuntos
Antineoplásicos/uso terapêutico , Aziridinas/uso terapêutico , Carcinoma de Células de Transição/tratamento farmacológico , Indolquinonas/uso terapêutico , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Carcinoma de Células de Transição/patologia , Modelos Animais de Doenças , Feminino , Ratos , Ratos Endogâmicos F344 , Neoplasias da Bexiga Urinária/patologia
11.
Gene Ther ; 19(3): 295-302, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21753794

RESUMO

Gene-directed enzyme prodrug therapy (GDEPT) is a promising and emerging strategy that attempts to limit the systemic toxicity inherent to cancer chemotherapy by means of tumor-targeted delivery and expression of an exogenous gene whose product converts nontoxic prodrug(s) into activated cytotoxic agent(s). The bacterial nitroreductase (NTR) enzyme, coupled with its substrate prodrug 5-(azaridin-1-yl)-2,4-dinitrobenzamide (CB1954), is a promising GDEPT strategy that has reached clinical trials. However, no strategy exists to visually monitor and quantitatively evaluate the therapeutic efficacy of NTR/CB1954 prodrug therapy in cells and imaging in living animals. As the success of any GDEPT is dependent upon the efficiency of transgene expression in vivo, we developed a safe, sensitive and reproducible noninvasive imaging method to monitor NTR transgene expression that would allow quantitative assessment of both therapeutic efficacy and diagnostic outcome of NTR/CB1954 prodrug therapy in the future. Here, we investigate the use of a novel fluorescent imaging dye CytoCy5S (a Cy5-labeled quenched substrate of NTR enzyme) on various cancer cell lines in vitro and in NTR-transfected tumor-bearing animals in vivo. CytoCy5S-labeled cells become fluorescent at 'red-shifted' wavelengths (638 nm) when reduced by cellular NTR enzyme and remains trapped within the cells for extended periods of time. The conversion and entrapment was dynamically recorded using a time-lapsed microscopy. Systemic and intratumoral delivery of CytoCy5S to NTR-expressing tumors in animals indicated steady and reproducible signals even 16 h after delivery (P<0.001). This is the first study to address visual monitoring and quantitative evaluation of NTR activity in small animals using CytoCy5S, and establishes the capability of NTR to function as an imageable reporter gene.


Assuntos
Aziridinas/metabolismo , Imagem Molecular , Nitrorredutases/genética , Nitrorredutases/metabolismo , Pró-Fármacos/metabolismo , Animais , Aziridinas/uso terapêutico , Linhagem Celular , Ativação Enzimática/genética , Expressão Gênica , Ordem dos Genes , Vetores Genéticos , Humanos , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Cinética , Metagenoma/genética , Camundongos , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/genética , Pró-Fármacos/uso terapêutico , Transfecção , Transplante Heterólogo
12.
Langmuir ; 27(23): 14300-7, 2011 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-22014024

RESUMO

Directed enzyme prodrug therapy is an extensive area of research in cancer chemotherapy. Although very promising, the current directed approaches are still hampered by inefficient enzyme expression and tumor targeting. This work investigates the viability of using metal nanoparticles as a novel delivery vehicle for prodrug-activating enzymes. Using genetically incorporated amino acid sequences, a nitroreductase from E. coli was directly immobilized onto a 50 nm gold colloid, as confirmed by gel electrophoresis, DLS, and UV-vis spectroscopy. The resulting conjugates showed excellent stability in changing proton and sodium chloride environments, including PBS at 37 °C. Remarkably, the immobilized nitroreductase retained more than 99% activity to the CB1954 prodrug without the need for stabilizers. This work provides the foundation for attaching prodrug-activating enzymes to metal nanoparticles for future use in directed enzyme prodrug therapy.


Assuntos
Aziridinas/uso terapêutico , Sistemas de Liberação de Medicamentos , Ouro/química , Neoplasias/tratamento farmacológico , Nitrorredutases/química , Pró-Fármacos/uso terapêutico , Engenharia de Proteínas , Coloides/química , Humanos , Modelos Moleculares , Nitrorredutases/isolamento & purificação , Nitrorredutases/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Cloreto de Sódio/química
13.
Ann Oncol ; 22(7): 1653-1660, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21378203

RESUMO

BACKGROUND: This trial describes a first-in-man evaluation of RH1, a novel bioreductive drug activated by DT-diaphorase (DTD), an enzyme overexpressed in many tumours. PATIENTS AND METHODS: A dose-escalation phase I trial of RH1 was carried out. The primary objective was to establish the maximum tolerated dose (MTD) of RH1. Secondary objectives were assessment of toxicity, pharmacokinetic determination of RH1 and pharmacodynamic assessment of drug effect through measurement of DNA cross linking in peripheral blood mononuclear cells (PBMCs) and tumour, DTD activity in tumour and NAD(P)H:quinone oxidoreductase 1 (NQO1) polymorphism status. RESULTS: Eighteen patients of World Health Organization performance status of zero to one with advanced refractory solid malignancies were enrolled. MTD was 1430 µg/m(2)/day with reversible bone marrow suppression being dose limiting. Plasma pharmacokinetic analysis showed RH1 is rapidly cleared from blood (t(1/2) = 12.3 min), with AUC increasing proportionately with dose. The comet-X assay demonstrated dose-related increases in DNA cross linking in PBMCs. DNA cross linking was demonstrated in tumours, even with low levels of DTD. Only one patient was homozygous for NQO1 polymorphism precluding any conclusion of its effect. CONCLUSIONS: RH1 was well tolerated with predictable and manageable toxicity. The MTD of 1430 µg/m(2)/day is the dose recommended for phase II trials. The biomarkers of DNA cross linking, DTD activity and NQO1 status have been validated and clinically developed.


Assuntos
Aziridinas/uso terapêutico , Benzoquinonas/uso terapêutico , NAD(P)H Desidrogenase (Quinona)/metabolismo , Neoplasias/tratamento farmacológico , Adulto , Idoso , Aziridinas/farmacocinética , Benzoquinonas/farmacocinética , Feminino , Seguimentos , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , NAD(P)H Desidrogenase (Quinona)/genética , Neoplasias/enzimologia , Neoplasias/patologia , Polimorfismo Genético/genética , Estudos Retrospectivos , Distribuição Tecidual , Resultado do Tratamento
14.
Sci Transl Med ; 2(40): 40ra50, 2010 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-20630857

RESUMO

DNA-damaging agents are widely used in cancer treatment despite their lack of tumor specificity. Human NQO2 (quinone oxidoreductase-2) is an atypical oxidoreductase because no endogenous electron donor has been identified to date. The enzyme converts CB1954 [5-(aziridin-1-yl)-2,4-dinitrobenzamide], in the presence of the synthetic nicotinamide cofactor analog EP0152R, to a cytotoxic bifunctional alkylating agent. NQO2 activity in hepatocellular tumor tissue is higher than that in other cancer types by a factor of 6 and higher than that in bone marrow by a factor of 20. Structural data from x-ray crystallography and nuclear magnetic resonance spectroscopy allowed us to construct a model of CB1954 and EP0152R binding to NQO2, which suggested an optimal infusion schedule for a phase I trial combining the two agents. Thirty-two patients were treated, and diarrhea and serum transaminase concentrations defined a maximum tolerated dose for the drug combination. There was a clear pharmacokinetic interaction, with EP0152R inducing a marked increase in clearance of CB1954, in keeping with model predictions. We detected DNA interstrand cross-links caused by nitroreduced CB1954 in tumor biopsies from treated patients, demonstrating that the activated prodrug exerts its cytotoxic properties through DNA base alkylation.


Assuntos
Antineoplásicos/uso terapêutico , Aziridinas/uso terapêutico , Neoplasias/tratamento farmacológico , Pró-Fármacos/uso terapêutico , Quinona Redutases/metabolismo , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Antineoplásicos/farmacocinética , Aziridinas/efeitos adversos , Aziridinas/química , Aziridinas/farmacocinética , Morte Celular/efeitos dos fármacos , Reagentes de Ligações Cruzadas/farmacologia , Cristalografia por Raios X , DNA de Neoplasias/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Quinona Redutases/química
15.
Biochem Pharmacol ; 79(5): 678-87, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19852945

RESUMO

Gene-directed enzyme prodrug therapy (GDEPT) aims to achieve highly selective tumor-cell killing through the use of tumor-tropic gene delivery vectors coupled with systemic administration of otherwise inert prodrugs. Nitroaromatic prodrugs such as CB1954 hold promise for GDEPT as they are readily reduced to potent DNA alkylating agents by bacterial nitroreductase enzymes (NTRs). Transfection with the nfsB gene from Escherichia coli can increase the sensitivity of tumor cells to CB1954 by greater than 1000-fold. However, poor catalytic efficiency limits the activation of CB1954 by NfsB at clinically relevant doses. A lack of flexible, high-throughput screening technology has hindered efforts to discover superior NTR candidates. Here we demonstrate how the SOS chromotest and complementary screening technologies can be used to evaluate novel enzymes that activate CB1954 and other bioreductive and/or genotoxic prodrugs. We identify the major E. coli NTR, NfsA, as 10-fold more efficient than NfsB in activating CB1954 as purified protein (k(cat)/K(m)) and when over-expressed in an E. coli nfsA(-)/nfsB(-) gene deleted strain. NfsA also confers sensitivity to CB1954 when expressed in HCT-116 human colon carcinoma cells, with similar efficiency to NfsB. In addition, we identify two novel E. coli NTRs, AzoR and NemA, that have not previously been characterized in the context of nitroaromatic prodrug activation.


Assuntos
Antineoplásicos/metabolismo , Aziridinas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Nitrorredutases/metabolismo , Pró-Fármacos/metabolismo , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Antineoplásicos/uso terapêutico , Aziridinas/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Escherichia coli/genética , Inativação Gênica , Terapia Genética , Humanos , Cinética , Pró-Fármacos/uso terapêutico , Resposta SOS em Genética/efeitos dos fármacos , Resposta SOS em Genética/genética , Transfecção , Células Tumorais Cultivadas
16.
Molecules ; 14(11): 4517-45, 2009 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-19924084

RESUMO

Gene directed enzyme prodrug therapy (GDEPT) of cancer aims to improve the selectivity of chemotherapy by gene transfer, thus enabling target cells to convert nontoxic prodrugs to cytotoxic drugs. A zone of cell kill around gene-modified cells due to transfer of toxic metabolites, known as the bystander effect, leads to tumour regression. Here we discuss the implications of either striving for a strong bystander effect to overcome poor gene transfer, or avoiding the bystander effect to reduce potential systemic effects, with the aid of three successful GDEPT systems. This review concentrates on bystander effects and drug development with regard to these enzyme prodrug combinations, namely herpes simplex virus thymidine kinase (HSV-TK) with ganciclovir (GCV), cytosine deaminase (CD) from bacteria or yeast with 5-fluorocytodine (5-FC), and bacterial nitroreductase (NfsB) with 5-(azaridin-1-yl)-2,4-dinitrobenzamide (CB1954), and their respective derivatives.


Assuntos
Terapia Genética/métodos , Pró-Fármacos/uso terapêutico , Animais , Aziridinas/uso terapêutico , Citosina Desaminase/genética , Citosina Desaminase/fisiologia , Flucitosina/uso terapêutico , Ganciclovir/uso terapêutico , Humanos , Nitrorredutases/genética , Nitrorredutases/fisiologia , Timidina Quinase/genética
17.
Hum Gene Ther ; 20(11): 1249-58, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19619056

RESUMO

We have completed a phase I/II suicide gene therapy clinical trial in patients with prostate cancer, using an E1/E3-deleted replication-deficient adenovirus (CTL102) encoding the bacterial nitroreductase enzyme in combination with prodrug CB1954. This study has provided an opportunity to monitor and characterize vector- and tumor-specific adaptive immunity before and after single or repeat injections of adenovirus. Here we report robust vector-specific humoral and cellular immune responses in all patients monitored. However, we found no correlation between preexisting immunity or the magnitude of the immune response to vector and the clinical outcome as measured by changes in serum prostate-specific antigen (PSA) level. Increased frequency of T cells recognizing prostate-specific antigens PSA or prostate-specific membrane antigen (PSMA) was detected in 3 of 11 patients after therapy, suggesting that this direct cytotoxic strategy can also stimulate tumor-specific immunity.


Assuntos
Aziridinas/uso terapêutico , Genes Transgênicos Suicidas/genética , Terapia Genética/métodos , Vetores Genéticos/imunologia , Pró-Fármacos/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/terapia , Adenoviridae , Ensaio de Imunoadsorção Enzimática , Genes Transgênicos Suicidas/imunologia , Humanos , Interferon gama/imunologia , Masculino , Nitrorredutases , Antígeno Prostático Específico/sangue , Antígeno Prostático Específico/imunologia , Neoplasias da Próstata/imunologia , Linfócitos T/imunologia
18.
Biochemistry ; 48(32): 7665-72, 2009 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-19580253

RESUMO

The enzyme nitroreductase, NfsB, from Escherichia coli has entered clinical trials for cancer gene therapy with the prodrug CB1954 [5-(aziridin-1-yl)-2,4-dinitrobenzamide]. However, CB1954 is a poor substrate for the enzyme. Previously we made several NfsB mutants that show better activity with CB1954 in a cell-killing assay in E. coli. Here we compare the kinetic parameters of wild-type NfsB with CB1954 to those of the most active single, double, and triple mutants isolated to date. For wild-type NfsB the global kinetic parameters for both k(cat) and K(m) for CB1954 are about 20-fold higher than previously estimated; however, the measured specificity constant, k(cat)/K(m) is the same. All of the mutants are more active with CB1954 than the wild-type enzyme, the most active mutant showing about 100-fold improved specificity constant with CB1954 over the wild-type protein with little effect on k(cat). This enhancement in specificity constants for the mutants is not seen with the antibiotic nitrofurazone as substrate, leading to reversed nitroaromatic substrate selectivity for the double and triple mutants. However, similar enhancements in specificity constants are found with the quinone menadione. Stopped-flow kinetic studies suggest that the rate-determining step of the reaction is likely to be the release of products. The most active mutant is also selective for the 4-nitro group of CB1954, rather than the 2-nitro group, giving the more cytotoxic reduction product. The double and triple mutants should be much more effective enzymes for use with CB1954 in prodrug-activation gene therapy.


Assuntos
Antineoplásicos/metabolismo , Aziridinas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Mutação , Nitrorredutases/metabolismo , Pró-Fármacos/metabolismo , Anti-Infecciosos/química , Anti-Infecciosos/metabolismo , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Aziridinas/química , Aziridinas/uso terapêutico , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Modelos Moleculares , Estrutura Molecular , Nitrofurazona/química , Nitrofurazona/metabolismo , Nitrorredutases/genética , Pró-Fármacos/química , Pró-Fármacos/uso terapêutico , Estrutura Terciária de Proteína , Vitamina K 3/química , Vitamina K 3/metabolismo , Vitaminas/química , Vitaminas/metabolismo
19.
Eur J Med Chem ; 44(9): 3373-87, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19540628

RESUMO

The present review describes research on natural aziridine alkaloids isolated from both terrestrial and marine species, as well as their lipophilic semi-synthetic, and/or synthetic analogs. Over 130 biologically active aziridine-containing compounds demonstrate confirmed pharmacological activity including antitumor, antimicrobial, antibacterial effects. The structures, origin, and biological activities of aziridine alkaloids are reviewed. Consequently this review emphasizes the role of aziridine alkaloids as an important source of drug prototypes and leads for drug discovery.


Assuntos
Alcaloides/uso terapêutico , Antibacterianos/uso terapêutico , Antineoplásicos/uso terapêutico , Aziridinas/uso terapêutico , Alcaloides/química , Alcaloides/isolamento & purificação , Alcaloides/farmacologia , Animais , Antibacterianos/química , Antibacterianos/isolamento & purificação , Antibacterianos/farmacologia , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Aziridinas/química , Aziridinas/isolamento & purificação , Aziridinas/farmacologia , Humanos
20.
Anticancer Res ; 29(6): 2159-66, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19528476

RESUMO

BACKGROUND: HSV1790 is an oncolytic virus generated by inserting the enzyme nitroreductase (NTR) into the virus HSV1716. NTR converts the prodrug CB1954 into an active alkylating agent. MATERIALS AND METHODS: In vitro, 3T6 cells (non permissive to HSV) were used in order to distinguish between virus-induced cytopathic effect and cell death due to activated prodrug. In vivo, xenograft models were injected with HSV1790 (10(5)-10(9) PFU) with or without CB1954 (max 80mg/kg) and tumor volume recorded regularly. Biodistribution of HSV1790 was determined immunohistochemically and by PCR. RESULTS: HSV1790 + CB1954 in vitro was more effective at killing tumor cells than the virus or the prodrug alone. In vivo, the combination reduced tumor volume and increased survival compared to treatment with HSV1790 or CB1954 alone. Following systemic administration of HSV1790, viral replication was detected in tumors, but not organs. CONCLUSION: HSV1790 + prodrug enhances tumor cell killing in vitro and reduces tumor volume and increases survival in vivo.


Assuntos
Antineoplásicos/uso terapêutico , Aziridinas/uso terapêutico , Herpesvirus Humano 1/patogenicidade , Neoplasias Experimentais/terapia , Vírus Oncolíticos/metabolismo , Pró-Fármacos/uso terapêutico , Animais , Western Blotting , Terapia Combinada , Feminino , Herpes Simples/genética , Herpes Simples/patologia , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Nus , Neoplasias Experimentais/genética , Neoplasias Experimentais/virologia , Vírus Oncolíticos/genética , Reação em Cadeia da Polimerase , Pró-Fármacos/farmacocinética , Distribuição Tecidual , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...