Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Control Release ; 275: 217-228, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29477351

ABSTRACT

The ability to modify existing microbiota at different sites presents enormous potential for local or indirect management of various diseases. Because bacteria can be maintained for lengthy periods in various regions of the body, they represent a platform with enormous potential for targeted production of biomolecules, which offer tremendous promise for therapeutic and diagnostic approaches for various diseases. While biological medicines are currently limited in the clinic to patient administration of exogenously produced biomolecules from engineered cells, in situ production of biomolecules presents enormous scope in medicine and beyond. The slow pace and high expense of traditional research approaches has particularly hampered the development of biological medicines. It may be argued that bacterial-based medicine has been "waiting" for the advent of enabling technology. We propose that this technology is Synthetic Biology, and that the wait is over. Synthetic Biology facilitates a systematic approach to programming living entities and/or their products, using an approach to Research and Development (R&D) that facilitates rapid, cheap, accessible, yet sophisticated product development. Full engagement with the Synthetic Biology approach to R&D can unlock the potential for bacteria as medicines for cancer and other indications. In this review, we describe how by employing Synthetic Biology, designer bugs can be used as drugs, drug-production factories or diagnostic devices, using oncology as an exemplar for the concept of in situ biomolecule production in medicine.


Subject(s)
Bacteria/metabolism , Neoplasms/therapy , Animals , Biological Products/metabolism , Biological Products/therapeutic use , Humans , Neoplasms/microbiology , Synthetic Biology
2.
Adv Drug Deliv Rev ; 118: 8-23, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28916496

ABSTRACT

Bacterial-directed enzyme prodrug therapy (BDEPT) is an emerging form of treatment for cancer. It is a biphasic variant of gene therapy in which a bacterium, armed with an enzyme that can convert an inert prodrug into a cytotoxic compound, induces tumour cell death following tumour-specific prodrug activation. BDEPT combines the innate ability of bacteria to selectively proliferate in tumours, with the capacity of prodrugs to undergo contained, compartmentalised conversion into active metabolites in vivo. Although BDEPT has undergone clinical testing, it has received limited clinical exposure, and has yet to achieve regulatory approval. In this article, we review BDEPT from the system designer's perspective, and provide detailed commentary on how the designer should strategize its development de novo. We report on contemporary advancements in this field which aim to enhance BDEPT in terms of safety and efficacy. Finally, we discuss clinical and regulatory barriers facing BDEPT, and propose promising approaches through which these hurdles may best be tackled.


Subject(s)
Bacteria/enzymology , Biological Factors/metabolism , Neoplasms/drug therapy , Prodrugs/metabolism , Prodrugs/therapeutic use , Animals , Enzyme Therapy/methods , Humans
3.
PLoS One ; 12(6): e0180034, 2017.
Article in English | MEDLINE | ID: mdl-28662099

ABSTRACT

Systemic administration of the highly potent anticancer therapeutic, tumour necrosis factor alpha (TNFα) induces high levels of toxicity and is responsible for serious side effects. Consequently, tumour targeting is required in order to confine this toxicity within the locality of the tumour. Bacteria have a natural capacity to grow within tumours and deliver therapeutic molecules in a controlled fashion. The non-pathogenic E. coli strain MG1655 was investigated as a tumour targeting system in order to produce TNFα specifically within murine tumours. In vivo bioluminescence imaging studies and ex vivo immunofluorescence analysis demonstrated rapid targeting dynamics and prolonged survival, replication and spread of this bacterial platform within tumours. An engineered TNFα producing construct deployed in mouse models via either intra-tumoural (i.t.) or intravenous (i.v.) administration facilitated robust TNFα production, as evidenced by ELISA of tumour extracts. Tumour growth was impeded in three subcutaneous murine tumour models (CT26 colon, RENCA renal, and TRAMP prostate) as evidenced by tumour volume and survival analyses. A pattern of pro-inflammatory cytokine induction was observed in tumours of treated mice vs. CONTROLS: Mice remained healthy throughout experiments. This study indicates the therapeutic efficacy and safety of TNFα expressing bacteria in vivo, highlighting the potential of non-pathogenic bacteria as a platform for restricting the activity of highly potent cancer agents to tumours.


Subject(s)
Colorectal Neoplasms/microbiology , Colorectal Neoplasms/therapy , Escherichia coli/metabolism , Kidney Neoplasms/microbiology , Kidney Neoplasms/therapy , Prostatic Neoplasms/microbiology , Prostatic Neoplasms/therapy , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Escherichia coli/growth & development , Male , Mice , Mice, Inbred BALB C
4.
J Control Release ; 222: 9-17, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26655063

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Bacterial Proteins/metabolism , Neoplasms/drug therapy , Probiotics , Prodrugs/administration & dosage , Animals , Anthraquinones/administration & dosage , Anthraquinones/therapeutic use , Antineoplastic Agents/therapeutic use , Aziridines/administration & dosage , Aziridines/therapeutic use , Bifidobacterium/enzymology , Cell Line, Tumor , Drug Delivery Systems , Enzymes , Escherichia coli/enzymology , Female , Lactobacillus/enzymology , Lactococcus/enzymology , Mice, Inbred BALB C , Neoplasms/pathology , Prodrugs/therapeutic use , Tumor Burden/drug effects , Vidarabine Phosphate/administration & dosage , Vidarabine Phosphate/analogs & derivatives , Vidarabine Phosphate/therapeutic use
5.
Sci Rep ; 5: 14554, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26416623

ABSTRACT

In this study, the potential effects of bacteria on the efficacy of frequently used chemotherapies was examined. Bacteria and cancer cell lines were examined in vitro and in vivo for changes in the efficacy of cancer cell killing mediated by chemotherapeutic agents. Of 30 drugs examined in vitro, the efficacy of 10 was found to be significantly inhibited by certain bacteria, while the same bacteria improved the efficacy of six others. HPLC and mass spectrometry analyses of sample drugs (gemcitabine, fludarabine, cladribine, CB1954) demonstrated modification of drug chemical structure. The chemoresistance or increased cytotoxicity observed in vitro with sample drugs (gemcitabine and CB1954) was replicated in in vivo murine subcutaneous tumour models. These findings suggest that bacterial presence in the body due to systemic or local infection may influence tumour responses or off-target toxicity during chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Aziridines/pharmacokinetics , Cladribine/pharmacokinetics , Colonic Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Vidarabine/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Aziridines/pharmacology , Biotransformation , Cladribine/pharmacology , Colonic Neoplasms/metabolism , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Escherichia coli/growth & development , Female , Injections, Intralesional , Injections, Subcutaneous , Listeria/growth & development , Mice , Neoplasm Transplantation , Skin/drug effects , Skin/microbiology , Skin/pathology , Treatment Outcome , Vidarabine/pharmacokinetics , Vidarabine/pharmacology , Gemcitabine
6.
Curr Gene Ther ; 15(3): 277-88, 2015.
Article in English | MEDLINE | ID: mdl-25619884

ABSTRACT

The feasibility of utilising bacteria as vectors for gene therapy is becoming increasingly recognised. This is primarily due to a number of intrinsic properties of bacteria such as their tumour targeting capabilities, their ability to carry large genetic or protein loads and the availability of well-established genetic engineering tools for a range of common lab strains. However, a number of issues relating to the use of bacteria as vectors for gene therapy need to be addressed in order for the field to progress. Amongst these is the need for the development of non-invasive detection/imaging systems for bacteria within a living host. In vivo optical imaging has advanced preclinical research greatly, and typically involves engineering of bacteria with genetic expression constructs for luminescence (e.g. the lux operon) or fluorescent proteins (GFP etc.). This requirement for genetic modification can be restrictive, where engineering is not experimentally appropriate or technologically feasible (e.g. due to lack of suitable engineering tools). We describe a novel strategy exploiting endogenous bacterial enzymatic activity to specifically activate an exogenously administered fluorescent imaging probe. The red shifted, quenched fluorophore CytoCy5S is reduced to a fluorescent form by bacterial-specific nitroreductase (NTR) enzymes. NTR enzymes are present in a wide range of bacterial genera and absent in mammalian systems, permitting highly specific detection of Gram-negative and Gram-positive bacteria in vivo. In this study, dose-responsive bacterial-specific signals were observed in vitro from all genera examined - E. coli, Salmonella, Listeria, Bifidobacterium and Clostridium difficile. Examination of an NTR-knockout strain validated the enzyme specificity of the probe. In vivo whole-body imaging permitted specific, dose-responsive monitoring of bacteria over time in various infection models, and no toxicity to bacteria or host was observed. This study demonstrates the concept of exploiting innate NTR activity as a reporting strategy for wild-type bacteria using optical imaging, while the concept may also be extended to NTR-specific probes for use with other imaging modalities.


Subject(s)
Bacteria/genetics , Fluorescent Dyes , Nitroreductases/metabolism , Animals , Bacteria/metabolism , Female , Genetic Therapy/methods , Genetic Vectors , Mice , Mice, Inbred BALB C , Neoplasms/therapy , Prodrugs/metabolism
7.
J Control Release ; 170(1): 120-31, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23688772

ABSTRACT

Current conventional treatments for cancer lack tumour selectivity resulting in the destruction of healthy tissue and severe adverse effects to the patient in addition to limiting the administration dose and efficacy. Hence, it is imperative that we seek alternative approaches to treat cancer that localise therapeutic agents to the site of the tumour and spare normal tissue. The use of bacteria in cancer therapy represents one such approach. Bacteria were first used as anti-cancer agents over a century ago. Today, this field has re-emerged from the past and is progressing at a rapid rate. Bacteria are used as anticancer agents either alone or in combination with conventional treatments and have been armed with an arsenal of therapeutic genes, which enhance their efficacy. Bacterial directed enzyme prodrug therapy (BDEPT) is one of the most promising approaches, which harnesses the tumour-specific location of bacteria to locally activate systemically administered 'prodrugs' within the tumour in order to induce selective tumour destruction. BDEPT is a relatively new concept. It was originally conceived more than 10years ago but it is only until recently that we witness a surge in activity in this field. In this review, we provide a full account of developments in the field of BDEPT since its inception. We share technical knowhow and discuss optimization strategies for vector and enzyme combinations, provide a clear view of the research landscape and suggest possible directions for the field.


Subject(s)
Antineoplastic Agents/administration & dosage , Bacteria , Enzyme Therapy/methods , Neoplasms/therapy , Prodrugs/administration & dosage , Animals , Humans
8.
Clin Cancer Res ; 14(13): 4259-66, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18594008

ABSTRACT

PURPOSE: We engineered the oncolytic Salmonella typhimurium-derived bacterium VNP20009 as a vector to target delivery to tumors of the prodrug-activating enzyme carboxypeptidase G2 (CPG2) and to show enhanced antitumor efficacy on administration of different prodrugs. EXPERIMENTAL DESIGN: We characterized CPG2 expression in vectors by immunoblotting, immunofluorescence, and enzyme activity. We assessed prodrug activation by high-performance liquid chromatography. Target human tumor cell and bacterial vector cell cytotoxicity was measured by flow cytometry and colony-forming assays. Therapy was shown in two human tumor xenografts and one mouse allograft with postmortem analysis of bacterial and CPG2 concentration in the tumors. RESULTS: CPG2 is expressed within the bacterial periplasm. It activates prodrugs and induces cytotoxicity in human tumor cells but not in host bacteria. Following systemic administration, bacteria multiply within xenografts reaching 2 x 10(7)/g to 2 x 10(8)/g at 40 days postinoculation. The concentration of CPG2 in these tumors increases steadily to therapeutic levels of 1 to 6 units/g. The bacteria alone reduce the growth of the tumors. Subsequent administration of prodrugs further reduces significantly the growth of the xenografts. CONCLUSIONS: The bacteria multiply within tumors, resulting in a selective expression of CPG2. The CPG2-expressing bacteria alone reduce the growth of tumors. However, in the presence of prodrugs activated by CPG2, this oncolytic effect is greatly increased. We conclude that bacterial oncolytic therapy, combined with CPG2-mediated prodrug activation, has great potential in the treatment of a range of cancers.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/therapy , Genetic Therapy/methods , Melanoma/drug therapy , Melanoma/genetics , Prodrugs/metabolism , Salmonella typhimurium/metabolism , gamma-Glutamyl Hydrolase/genetics , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Flow Cytometry , Genetic Vectors , Humans , Mice , Neoplasm Transplantation
9.
Curr Med Chem ; 10(20): 2175-84, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12871091

ABSTRACT

Regulation of expression in gene therapy is considered to be a very desirable goal, preventing toxic effects and improving biological efficacy. A variety of systems have been reported in an ever widening range of applications, this paper describes these systems with specific reference to cancer gene therapy.


Subject(s)
Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Genetic Therapy/methods , Animals , Genetic Vectors , Humans , Neoplasms/therapy
10.
J Med Chem ; 46(9): 1690-705, 2003 Apr 24.
Article in English | MEDLINE | ID: mdl-12699387

ABSTRACT

Nineteen novel potential self-immolative prodrugs and their corresponding drugs have been synthesized for gene-directed enzyme prodrug therapy (GDEPT) with carboxypeptidase G2 (CPG2) as the activating enzyme. The compounds are derived from o- and p-amino and p-methylamino aniline nitrogen mustards. Their aqueous stability, kinetics of drug release by CPG2, and cytotoxicity in the colon carcinoma cell line WiDr, expressing either surface-tethered CPG2 (stCPG2(Q)3) or control beta-galactosidase, are assessed. The effect of various structural features on stability, kinetics of activation, and biological activity is discussed. The p-methylamino prodrugs are the most stable compounds from this series, with the largest cytotoxicity differentials between CPG2-expressing and nonexpressing cells. The most potent compounds in all series are prodrugs of bis-iodo nitrogen mustards. 4-[N-[4'-Bis(2' '-iodoethyl)aminophenyl]-N'-methylcarbamoyloxymethyl]phenylcarbamoyl-l-glutamic acid, compound 39b, is 124-fold more cytotoxic to WiDr cells expressing CPG2 than to cells expressing beta-galactosidase. An additional six compounds show better cytotoxicity differential than the published N-[4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl]-l-glutamic acid (CMDA) prodrug.


Subject(s)
Aniline Mustard/chemical synthesis , Antineoplastic Agents, Alkylating/chemical synthesis , Carbamates/chemical synthesis , Glutamic Acid/chemical synthesis , Prodrugs/chemical synthesis , gamma-Glutamyl Hydrolase/metabolism , Aniline Mustard/chemistry , Aniline Mustard/pharmacokinetics , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacokinetics , Carbamates/chemistry , Carbamates/pharmacokinetics , Gene Transfer Techniques , Glutamic Acid/analogs & derivatives , Glutamic Acid/chemistry , Glutamic Acid/pharmacokinetics , Half-Life , Humans , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Quantitative Structure-Activity Relationship , Tumor Cells, Cultured , gamma-Glutamyl Hydrolase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...