Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Dose Response ; 12(1): 1-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24659931

ABSTRACT

UNLABELLED: Targeted radiotherapy is a potentially useful treatment for some cancers and may be potentiated by bystander effects. However, without estimation of absorbed dose, it is difficult to compare the effects with conventional external radiation treatment. METHODS: Using the Vynckier - Wambersie dose point kernel, a model for dose rate evaluation was created allowing for calculation of absorbed dose values to two cell lines transfected with the noradrenaline transporter (NAT) gene and treated with [(131)I]MIBG. RESULTS: The mean doses required to decrease surviving fractions of UVW/NAT and EJ138/NAT cells, which received medium from [(131)I]MIBG-treated cells, to 25 - 30% were 1.6 and 1.7 Gy respectively. The maximum mean dose rates achieved during [(131)I]MIBG treatment were 0.09 - 0.75 Gy/h for UVW/NAT and 0.07 - 0.78 Gy/h for EJ138/NAT. These were significantly lower than the external beam gamma radiation dose rate of 15 Gy/h. In the case of control lines which were incapable of [(131)I]MIBG uptake the mean absorbed doses following radiopharmaceutical were 0.03 - 0.23 Gy for UVW and 0.03 - 0.32 Gy for EJ138. CONCLUSION: [(131)I]MIBG treatment for ICCM production elicited a bystander dose-response profile similar to that generated by external beam gamma irradiation but with significantly greater cell death.

2.
Br J Radiol ; 81 Spec No 1: S28-35, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18819996

ABSTRACT

The radiopharmaceutical [(131)I]meta-iodobenzylguanidine ([(131)I]MIBG) and the topoisomerase I inhibitor topotecan are both effective as single-agent treatments of neuroblastoma. Our purpose was to assess the therapeutic potential of [(131)I]MIBG and topotecan in combination using SK-N-BE(2c) neuroblastoma cells and UVW/NAT glioma cells expressing the noradrenaline transporter transgene. Topotecan treatment was given (i) before, (ii) after or (iii) simultaneously with [(131)I]MIBG. DNA fragmentation was evaluated by comet assay and cell cycle redistribution was determined by fluorescence-activated cell sorting. Combination index analysis indicated that delivery schedules (ii) and (iii) were more effective than schedule (i) with respect to clonogenic cell kill. Similarly, significant DNA damage was observed following treatment schedules (ii) and (iii) (p <0.005), but not (i). Prior exposure to topotecan did not significantly enhance [(131)I]MIBG uptake in athymic mice bearing tumour xenografts. We conclude that the enhancement of the efficacy of [(131)I]MIBG by combining it with topotecan was the result of inhibition of DNA damage repair rather than an increase in expression of the noradrenaline transporter by tumour.


Subject(s)
3-Iodobenzylguanidine/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Glioma/therapy , Neuroblastoma/therapy , Radiopharmaceuticals/administration & dosage , Topotecan/administration & dosage , Animals , DNA Fragmentation/drug effects , DNA Fragmentation/radiation effects , Mice , Mice, Nude , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Treatment Outcome
3.
Gene Ther ; 14(3): 246-55, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17006546

ABSTRACT

Cancer gene therapy that utilizes toxic transgene products requires strict transcriptional targeting to prevent adverse normal tissue effects. We report on the use of a promoter derived from the cyclin dependent kinase inhibitor, p21((WAF1)), to control transgene expression. We demonstrate that this promoter is relatively silent in normal cells (L132, FSK, HMEC-1) compared to the almost constitutive expression obtained in tumour cells (DU145, LNCaP, HT29 and MCF-7) of varying p53 status, a characteristic that will be important in gene therapy protocols. In addition, we found that the p21((WAF1)) promoter could be further induced by both external beam radiation (up to eight-fold in DU145 cells), intracellular-concentrated radionuclides ([(211)At]MABG) (up to 3.5-fold in SK-N-BE(2c) cells) and hypoxia (up to four-fold in DU145 cells). We have previously achieved significant radiosensitization of tumour cells both in vitro and in vivo by using inducible nitric oxide synthase (iNOS) gene therapy to generate the potent radiosensitizer, nitric oxide (NO(.-)). Here, we report that a clinically relevant schedule of p21((WAF1))-driven iNOS gene therapy significantly sensitized both p53 wild-type RIF-1 tumours and p53 mutant HT29 tumours to fractionated radiotherapy. Our data highlight the utility of this p21((WAF1))/iNOS-targeted approach.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Genetic Therapy/methods , Neoplasms/therapy , Nitric Oxide Synthase Type II/genetics , Promoter Regions, Genetic , Radiation-Sensitizing Agents/therapeutic use , Animals , Cell Hypoxia , Cell Line, Tumor , Combined Modality Therapy , Female , Gene Expression , Gene Targeting , Genes, p53/genetics , Mice , Neoplasms/enzymology , Neoplasms/radiotherapy , Neoplasms, Experimental , Nitric Oxide Synthase Type II/metabolism , Transfection/methods
5.
Med Chem ; 1(5): 423-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16787326

ABSTRACT

Genetically engineered herpes simplex virus ICP34.5 null mutants replicate only in dividing cells and have shown potential for the treatment of malignant disease, including glioma. Phase I trials have demonstrated the safety of these viruses in various clinical settings but it is envisaged that for full efficacy they will be used in combination with other therapeutic modalities. To enhance virus-induced tumour cytotoxicity, we have engineered an ICP34.5 null mutant (HSV1716) of HSV1 which expresses the noradrenaline transporter gene (NAT). This virus is designated HSV1716/NAT. We have shown previously that introduction of the NAT gene into a range of tumour cells, via plasmid-mediated transfection, conferred the capacity for active uptake of the radiopharmaceutical [131I]MIBG and resulted in dose-dependent toxicity. In this study, combination therapy utilising HSV1716/NAT and [131I]MIBG was assessed in vitro by the MTT assay. We demonstrate that the NAT gene, introduced by HSV1716/NAT into cultured glioma cells, was expressed 1 h after viral infection, enabling active uptake of [131I]MIBG. The combination of viral oncolysis and induced radiopharmaceutical uptake resulted in significantly enhanced cytotoxicity compared to either agent alone and the response was dose- and time-dependent. These studies show that the combination of oncolytic HSV therapy with targeted radiotherapy has the potential for effective tumour cell kill and warrants further investigation as a treatment for malignant glioma.


Subject(s)
3-Iodobenzylguanidine/pharmacokinetics , Gene Transfer Techniques , Glioma/therapy , Herpesvirus 1, Human/physiology , Oncolytic Virotherapy/methods , Animals , Cell Line , Cell Survival/drug effects , Combined Modality Therapy , Cricetinae , Dose-Response Relationship, Drug , Genetic Engineering , Glioma/genetics , Glioma/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/growth & development , Humans , In Vitro Techniques , Kinetics , Mice , Norepinephrine Plasma Membrane Transport Proteins/genetics , Norepinephrine Plasma Membrane Transport Proteins/physiology , Radiotherapy/methods , Structure-Activity Relationship , Time Factors , Transduction, Genetic , Tumor Cells, Cultured
6.
Prostate Cancer Prostatic Dis ; 7(4): 355-63, 2004.
Article in English | MEDLINE | ID: mdl-15477875

ABSTRACT

A targeted radiotherapy/gene therapy approach for prostate cancer, using the radiopharmaceutical [(131)I]meta-iodobenzylguanidine ([(131)I]MIBG), would restrict the effects of radiotherapy to malignant cells, thereby increasing efficacy and decreasing morbidity of radiotherapy. Prostate cancer cells were transfected with a transgene encoding the noradrenaline transporter (NAT) under the control of tumour-specific telomerase promoters, enabling them to actively take up [(131)I]MIBG. This led to tumour-specific cell kill. This strategy has the advantage of generating a radiological bystander effect, leading to the destruction of neighbouring tumour cells that have escaped transfection. This targeted approach could be a promising tumour-specific treatment option for prostate cancer.


Subject(s)
3-Iodobenzylguanidine/therapeutic use , Adenocarcinoma/therapy , Genetic Therapy , Prostatic Neoplasms/therapy , Radiopharmaceuticals/therapeutic use , Symporters/therapeutic use , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Bystander Effect , Combined Modality Therapy , Humans , Iodine Radioisotopes , Male , Norepinephrine Plasma Membrane Transport Proteins , Plasmids , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Telomerase/genetics , Transfection , Tumor Cells, Cultured , Tumor Stem Cell Assay
7.
Curr Gene Ther ; 3(4): 319-39, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12871020

ABSTRACT

Radiotherapy is, after surgery, the most widely used form of cancer treatment but the main limitation of radiation treatment is damage to normal tissues. This may be overcome by targeted radiotherapy - the selective delivery to malignant deposits of cytotoxic radionuclides bound to tumour-seeking agents. Several gene transfer techniques are being evaluated which combine gene therapy and targeted radiotherapy, rendering malignant cells more sensitive to radiation or causing them to take up radioactive drugs - providing tumour cell kill with reduced damage to normal tissues. This review examines several aspects of targeted radiotherapy/gene therapy strategies. As well as identification of suitable gene/radiopharmaceutical combinations, expression of the transgenes must be confined to tumour cells via tumour specific transcriptional regulation or via delivery vehicles which specifically target tumour cells. The inability of current delivery vehicles to target every cell within a tumour mass must be addressed by maximising collateral cell damage in targeted radiotherapy strategies via radiation mediated bystander effects and suitable in vitro models are described, which allow assessment of promising gene transfer strategies in scenarios where tumour heterogeneity of transgene expression and cell proliferative state are considered.


Subject(s)
Combined Modality Therapy/methods , Genetic Therapy/methods , Neoplasms/radiotherapy , Neoplasms/therapy , Animals , Apoptosis , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Transfer Techniques , Humans , Microscopy, Confocal , Models, Biological , Radiopharmaceuticals/therapeutic use , Telomerase/metabolism , Transfection , Transgenes
8.
J Biomed Biotechnol ; 2003(2): 102-109, 2003.
Article in English | MEDLINE | ID: mdl-12721515

ABSTRACT

Targeted radionuclide therapy is an alternative method of radiation treatment which uses a tumor-seeking agent carrying a radioactive atom to deposits of tumor, wherever in the body they may be located. Recent experimental data signifies promise for the amalgamation of gene transfer with radionuclide targeting. This review encompasses aspects of the integration of gene manipulation and targeted radiotherapy, highlighting the possibilities of gene transfer to assist the targeting of cancer with low molecular weight radiopharmaceuticals.

9.
J Gene Med ; 4(5): 567-76, 2002.
Article in English | MEDLINE | ID: mdl-12221650

ABSTRACT

BACKGROUND: We describe an in vitro tumour model for targeted radiotherapy and gene therapy that incorporates cell population heterogeneity. MATERIALS AND METHODS: Transfectant mosaic spheroids (TMS) and transfected mosaic monolayers (TMM) are composed of two cell populations derived from a single cell line. The cells of one population were transfected with the noradrenaline transporter gene (NAT), allowing active uptake of a radiolabelled targeting agent meta-[131I]iodobenzylguanidine ([131I]MIBG); the other population of cells was derived from the same parent line and transfected with a marker gene - green fluorescent protein (GFP). After treatment with [131I]MIBG, cell kill was determined in TMM by clonogenic assay and in TMS by clonogenic assay and spheroid growth delay. RESULTS: We have used the TMS model to assess the 'radiological bystander effect' (radiation cross-fire) conferred by the beta-emitting radiopharmaceutical [131I] MIBG whose cellular uptake is facilitated by the transfected gene encoding NAT. We show that cell killing by [131I]MIBG in both TMS and TMM cultures increased in direct proportion to the fraction of NAT-transfected cells and that the degree of cell killing against fraction transfected was greater in TMS, suggestive of a greater bystander effect in the three-dimensional culture system. CONCLUSIONS: TMS provide a useful model for assessment of the effectiveness of targeted radiotherapy in combination with gene therapy when less than 100% of the target cell population is expressing the NAT transgene. Further, this novel model offers the unique opportunity to investigate radiation-induced bystander effects and their contribution to cell cytotoxicity in radiotherapy and other gene therapy applications.


Subject(s)
Genetic Therapy , Glioma/genetics , Glioma/radiotherapy , Mosaicism/genetics , 3-Iodobenzylguanidine/metabolism , Cell Death/genetics , Cell Death/radiation effects , Flow Cytometry , Glioma/pathology , Humans , Iodine Radioisotopes/metabolism , Iodine Radioisotopes/toxicity , Spheroids, Cellular
10.
Oncogene ; 20(53): 7804-8, 2001 Nov 22.
Article in English | MEDLINE | ID: mdl-11753659

ABSTRACT

One of the most effective ways to kill cancer cells is by treatment of tumours with radiation. However, the administered dose of radiation to the tumour is limited by normal tissue toxicity. Strategies which decrease normal tissue exposure relative to tumour dose are urgently sought. One such promising scheme involves gene transfer, leading to the introduction of transporters specific for pharmaceuticals which can be labelled with radionuclides. We have previously demonstrated in vitro, that transfer of the noradrenaline transporter (NAT) gene, under viral promoter control, induces in host cells the active accumulation of the radiopharmaceutical [131I]meta-iodobenzylguanidine ([131I]MIBG) which results in kill of clonogens. We now report 17-fold enhancement of [131I]MIBG uptake by UVW glioma cells transfected with the NAT gene whose expression is driven by the human telomerase RNA (hTR) promoter (70% the uptake achieved by the strong viral promoter). Multicellular spheroids composed of hTR-NAT-transfected UVW cells exhibited dose-dependent susceptibility to treatment with [131I]MIBG. This was demonstrated by decreased survival of clonogens and complete sterilization of clonogens derived from spheroids and also failure of spheroids to regrow after administration of 7 MBq/ml [131I]MIBG. These data suggest hTR regulated expression of NAT may be an effective gene therapy strategy.


Subject(s)
3-Iodobenzylguanidine/metabolism , Genetic Therapy/methods , Glioma/genetics , Glioma/radiotherapy , Promoter Regions, Genetic/genetics , Radiopharmaceuticals/metabolism , Symporters/genetics , 3-Iodobenzylguanidine/administration & dosage , 3-Iodobenzylguanidine/pharmacology , 3-Iodobenzylguanidine/therapeutic use , Animals , Cattle , Cell Death/drug effects , Cell Death/radiation effects , Cell Division/drug effects , Cell Division/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Glioma/pathology , Humans , Norepinephrine Plasma Membrane Transport Proteins , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacology , Radiopharmaceuticals/therapeutic use , Tumor Cells, Cultured
11.
J Theor Biol ; 210(1): 93-100, 2001 May 07.
Article in English | MEDLINE | ID: mdl-11343433

ABSTRACT

A multistage mathematical model of tumorigenesis has been developed to explore the effects of target cell growth pattern on the proportions of tumours deriving from different tissues (the tumour spectrum). Analytical modelling techniques have shown that the effect of the target cell growth pattern on the tumour spectrum also depends on the number of stages (gene mutations) necessary for malignant change in cells of each tissue type. This suggests the existence of temporal "windows of opportunity" for tumours of different types in relation to stage number and growth kinetics. Models of this kind are applicable to cancer-prone transgenic (e.g. p53 deficient) mice, where homozygotes and heterozygotes differ in one carcinogenic stage, and differ also in the spectrum of tumours observed. Generally, tumours deriving from target stem cells which are developmentally short-lived will arise more frequently in homozygotes than heterozygotes. Such models may also be applicable to human syndromes (e.g. Li-Fraumeni) in which susceptibility to cancer is inherited.


Subject(s)
Models, Genetic , Neoplasms/genetics , Neoplastic Stem Cells/physiology , Animals , Cell Division , Genes, p53 , Genetic Predisposition to Disease , Humans , Mice , Mice, Transgenic , Models, Animal , Mutagenesis , Neoplasms/pathology
12.
J Gene Med ; 3(2): 165-72, 2001.
Article in English | MEDLINE | ID: mdl-11318115

ABSTRACT

BACKGROUND: Although [131I]meta-iodobenzylguanidine (MIBG) is currently one of the best agents available for targeted radiotherapy, its use is confined to a few neural crest derived tumours which accumulate the radiopharmaceutical via the noradrenaline transporter (NAT). To determine whether this drug could be used for the treatment of non-NAT expressing tumours following genetic manipulation, we previously showed that plasmid mediated transfection of NAT into a non-NAT expressing glioblastoma cell line, UVW, endowed the host cells with the capacity to actively accumulate [131I]MIBG. We now present data defining the conditions required for complete sterilisation of NAT transfected cells cultured as multicellular spheroids and treated with [131I]MIBG. METHODS: NAT transfected UVW cells, grown as monolayers and spheroids, were treated with various doses of [131I]MIBG and assessed for cell kill by clonogenic survival and measurement of spheroid volume over time (growth delay). Spheroids were left intact for different time periods to assess the effect of radiation crossfire on cell death. RESULTS AND CONCLUSIONS: Total clonogen sterilisation was observed when the cells were grown as three-dimensional spheroids and treated with 7 MBq/ml [131I]MIBG. The added benefit of radiation crossfire was demonstrated by the improvement in cell kill achieved by prolongation of the maintenance of [131I]MIBG treated spheroids in their three-dimensional form, before disaggregation and clonogenic assay. When left intact for 48 h after treatment, spheroid cure was achieved by exposure to 6 MBq/ml [131I]MIBG. These results demonstrate that the efficiency of cell kill by [131I]MIBG targeted therapy is strongly dependent on beta-particle crossfire irradiation. This gene therapy/targeted radiotherapy strategy has potential for [131I]MIBG mediated cell kill in tumours other than those derived from the neural crest.


Subject(s)
3-Iodobenzylguanidine/pharmacology , Cell Death/drug effects , Cell Death/radiation effects , Genetic Therapy/methods , Iodine Radioisotopes/pharmacology , Radiopharmaceuticals/pharmacology , Radiotherapy/methods , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/genetics , Glioma/pathology , Humans , Tumor Cells, Cultured
13.
Med Pediatr Oncol ; 35(6): 708-11, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11107152

ABSTRACT

BACKGROUND: The aims of this study were to determine whether the introduction and expression of the noradrenaline transporter (NAT) gene into NAT-negative neuroblastoma cell lines would make them amenable to targeted radiotherapy using [(131)I]MIBG. PROCEDURE: Neuroblastoma cell lines were transfected with a eukaryotic expression vector containing the bovine noradrenaline transporter cDNA under the expression of the CMV promoter. Stable transfectants were created by selection in geneticin (G418) and were characterised for their MIBG uptake ability and susceptibility to [(131)I]MIBG therapy. RESULTS: The cell line SK-N-MC, which normally shows no ability to take up MIBG, was successfully transfected with bNAT. SK-N-MC.bNAT transfectants exhibited uptake and release kinetics similar to those of the natural NAT-expressing cell line SK-N-BE(2c). Levels of [(131)I]MIBG uptake were 33% of those of the highest naturally NAT-expressing cell line SK-N-BE(2c). Growth delay assays using multicellular spheroids indicated that this degree of [(131)I]MIBG uptake was sufficient to inhibit growth at radioactive concentrations of 4 Mbq/ml. CONCLUSIONS: These results demonstrate the feasibility of combining gene therapy with targeted radiotherapy to enhance uptake, and hence radiation dose, to neuroblastoma tumours using [(131)I]MIBG. With the appropriate delivery vehicle and tumour-specific control of expression, the introduction of noradrenaline transporter molecules may be a viable means of enhancing the response of neuroblastoma tumours to [(131)I]MIBG therapy.


Subject(s)
Carrier Proteins/genetics , Genetic Therapy/methods , Neuroblastoma/genetics , Neuroblastoma/radiotherapy , Norepinephrine/genetics , Symporters , Child , Humans , Norepinephrine Plasma Membrane Transport Proteins , Tumor Cells, Cultured
14.
Int J Radiat Biol ; 76(7): 963-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10923620

ABSTRACT

PURPOSE: To seek the delayed appearance of chromosomal abnormalities in human fibroblasts exposed to the Auger electron emitter 125I. MATERIALS AND METHODS: Normal untransformed human fibroblasts, HF19, were exposed to a concentration of [I125]IUdR, which allowed the survival of 37% of clonogens. Chromosomal analysis using both conventional Giemsa and fluorescence in situ hybridization (FISH) was undertaken on non-clonal bulk cultures from 2 to 39 days after treatment. RESULTS: The data show a declining level of unstable aberrations in the progeny of HF19 fibroblasts exposed to [I125]IUdR, eventually reaching control levels. CONCLUSIONS: The results provide evidence that [125I]IUdR does not induce ongoing chromosomal instability in long-term culture, and gives further support to the use of Auger-electron emitting radionuclides in the treatment and diagnosis of tumours.


Subject(s)
Chromosome Aberrations , Iodine Radioisotopes/toxicity , Cell Line , Fibroblasts/radiation effects , Humans , Idoxuridine/toxicity , In Situ Hybridization, Fluorescence
15.
Curr Pharm Des ; 6(14): 1419-32, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10903401

ABSTRACT

For targeted radionuclide therapy to succeed as a single modality treatment, schemes must be devised which will enable the deposition in malignant cells of sterilising doses of radiation. Until such methods have been perfected, it is necessary to combine targeted radiotherapy in a rational manner with conventional anti-cancer treatments. Several means of delivery of therapeutic radionuclides are being evaluated but none of these yet appears to be as powerful as the simplest and most effective example, viz: sodium [131I]iodide treatment of disseminated thyroid carcinoma. The radiopharmaceutical [131I]meta-iodobenzylguanidine ([131I]MIBG) is an effective single agent for the treatment of neuroblastoma. However, uptake of the drug in malignant sites is heterogeneous, suggesting that this therapy alone is unlikely to cure disease. A growing body of experimental evidence indicates exciting possibilities for the integration of gene transfer with radionuclide targeting. This review covers aspects of the combination of gene manipulation and targeted radiotherapy, emphasising the potential of gene transfer to facilitate tumour targeting with low molecular weight radiopharmaceuticals.


Subject(s)
Gene Transfer Techniques , Neoplasms/radiotherapy , Radiotherapy/methods , Symporters , 3-Iodobenzylguanidine/therapeutic use , Animals , Carrier Proteins/genetics , Humans , Norepinephrine Plasma Membrane Transport Proteins , Receptors, Somatostatin/genetics
16.
Nucl Med Commun ; 21(5): 437-40, 2000 May.
Article in English | MEDLINE | ID: mdl-10874700

ABSTRACT

Radioiodinated meta-iodobenzylguanidine (MIBG) is used routinely for imaging and targeted radiotherapy of tumours derived from the neural crest. Since active uptake of MIBG by the noradrenaline transporter (NAT) makes a greater contribution to total drug accumulation than passive uptake when MIBG is present at low concentrations, tumour-specific uptake should be enhanced by the administration of lower molar amounts of MIBG. This could be achieved through the use of MIBG with a high specific activity. Commercially available preparations of 123I-MIBG have specific activities of approximately 200 MBq.mg-1. We have synthesized and used no-carrier-added (n.c.a.) 123I-MIBG produced by an iododesilylation reaction (specific activity 0.7 TBq.mg-1). We report here the first clinical studies comparing the commercially available and n.c.a. MIBG diagnostic preparations. Five patients with known phaeochromocytoma were studied. Unlike studies in animal models, no consistent improvement in tumour uptake was observed with the n.c.a. material. A larger patient group is required to determine whether there are significant differences between the two preparations, before proceeding to studies at therapeutic activity levels of n.c.a. 131I-MIBG. Even with no improvement in tumour uptake, n.c.a. MIBG may be the favoured formulation for therapeutic applications to reduce the molar amount of drug injected.


Subject(s)
3-Iodobenzylguanidine/pharmacokinetics , Adrenal Gland Neoplasms/metabolism , Pheochromocytoma/metabolism , Radiopharmaceuticals/pharmacokinetics , Adrenal Gland Neoplasms/diagnostic imaging , Adult , Chemistry, Pharmaceutical , Female , Humans , Iodine Radioisotopes , Male , Middle Aged , Pheochromocytoma/diagnostic imaging , Tomography, Emission-Computed, Single-Photon/methods
17.
Int J Radiat Biol ; 76(2): 169-76, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10716638

ABSTRACT

PURPOSE: The study was designed to determine the dose-response relationship for radiation induction of mutations at mini- and microsatellite loci in human somatic cells. Mutations induced by graded doses of gamma-irradiation were quantified by screening clones derived from single irradiated cells for micro- and minisatellite alterations following irradiation with 1, 2 or 3 Gy. MATERIALS AND METHODS: After irradiation, the moderately radioresistant glioma cell line UVW was seeded at low density into Petri dishes to allow formation of discrete colonies, 100 of which were examined at each dose. All the cells within a colony were presumed to have arisen from a single irradiated cell. Radiation-induced microsatellite alterations were determined at 16 different loci, by PCR amplification and visualization on polyacrylamide gels. Minisatellite alterations were identified at four different minisatellite loci by restriction enzyme digestion and Southern blotting. RESULTS: A dose-response curve for mutation frequency was obtained by analysis of 100 clones, yielding a minisatellite mutation rate of 5.5x10(-3) mutations/locus/Gy/cell and a microsatellite mutation rate of 8.75x10(-4) mutations/locus/ Gy/cell. At microsatellite loci, alterations were predominantly simple loss or gain of repeat units and loss of heterozygosity (LOH). The mutations in minisatellite loci resulted predominantly in LOH and variation in repeat number. The background instability at each locus was determined by analysis of non-irradiated clones. Only 2% and 1% of the micro-and minisatellite loci respectively showed altered bands. CONCLUSIONS: This is the first report of a dose-response relationship for radiation-induced micro- and minisatellite mutations in human somatic cells. Described is a sensitive method for analysis of low-dose radiation mutagenesis in somatic cells that may prove to be a useful tool for radiation protection and dosimetry.


Subject(s)
Microsatellite Repeats , Mutation , Dose-Response Relationship, Radiation , Humans , Loss of Heterozygosity , Polymerase Chain Reaction , Tumor Cells, Cultured
18.
Br J Cancer ; 82(1): 74-80, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10638969

ABSTRACT

The Auger electron emitting agent 5-[125I]iodo-2'-deoxyuridine (i.e. [125I]IUdR) holds promise for the treatment of residual glioma after surgery because this thymidine analogue kills only proliferating cells. However, malignant cells which are not synthesizing DNA during exposure to the radiopharmaceutical will be spared. To determine whether tumour incorporation of [125I]IUdR could be enhanced by protracted administration, we used a C6 cell line, growing in the brains of Wistar rats, as a glioma model and compared three methods of intracerebral delivery of [125I]IUdR. Twenty-four hours after administration of drug, autoradiography of brain sections demonstrated nuclear uptake of the radiopharmaceutical in cells throughout tumour while normal brain cells remained free of radioactivity. The [125I]IUdR labelling indices (% +/- s.e.m.) achieved were 6.2 (0.4) by single injection, 22.5 (4.1) using a sustained release polymer implant (poly(lactide-co-glycolide)) and 34.3 (2.0) by mini-osmotic pump. These results emphasize the need for a sustained delivery system as a prerequisite for effective treatment. These findings are also encouraging for the development of a sustained release system for radiolabelled IUdR for use in the treatment of intracranial tumours, particularly in the immediate postoperative setting.


Subject(s)
Brain Neoplasms/radiotherapy , Coated Materials, Biocompatible/administration & dosage , Drug Delivery Systems/methods , Glioma/radiotherapy , Idoxuridine/administration & dosage , Iodine Radioisotopes/administration & dosage , Lactic Acid/administration & dosage , Polyglycolic Acid/administration & dosage , Polymers/administration & dosage , Radiopharmaceuticals/administration & dosage , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Female , Glioma/metabolism , Glioma/pathology , Idoxuridine/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Male , Polylactic Acid-Polyglycolic Acid Copolymer , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Wistar , Tissue Distribution
19.
Cancer Gene Ther ; 7(12): 1529-36, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11228531

ABSTRACT

To evaluate the potential of the expression of the sodium/iodide symporter (NIS) as a means of targeting radioiodine to tumor cells, we have employed plasmid-mediated transfection of the NIS gene into a range of mammalian cell hosts. We observed perchlorate-inhibitable iodide uptake up to 41-fold over control in all NIS-transfected cells. We assessed the effect of NIS expression followed by exposure to 131I- on the clonogenic survival of UVW glioma cells. After exposure of two-dimensional monolayer cultures of UVW-NIS cells to 131I- at a radioactive concentration of 4 MBq/mL, clonogenic survival was reduced to 21%. Similar treatment of UVW-NIS cells in three-dimensional spheroid cultures resulted in a reduction of clonogenic survival to 2.5%. This increase in sensitivity to 131I- exposure is likely to be due to a radiological bystander effect. These results are very encouraging for the development of a novel cytotoxic gene-therapy strategy in which a radiological bystander effect plays a significant role in tumor cell sterilization.


Subject(s)
Carrier Proteins/genetics , Iodine Radioisotopes/therapeutic use , Membrane Proteins/genetics , Neoplasms/radiotherapy , Sodium Iodide/metabolism , Symporters , Transfection , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/radiotherapy , Carrier Proteins/metabolism , Disease Models, Animal , Female , Gene Expression , Glioma/genetics , Glioma/metabolism , Glioma/radiotherapy , Humans , In Vitro Techniques , Membrane Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/radiotherapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/radiotherapy , Thyroid Gland/metabolism , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/radiation effects , Tumor Stem Cell Assay
20.
Gene Ther ; 6(6): 1147-52, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10455418

ABSTRACT

Meta-iodobenzylguanidine conjugated to 131I-iodine is an effective agent for the targeted radiotherapy of tumors of neural crest origin which express the noradrenaline transporter (NAT). The therapeutic application of 131I MIBG is presently limited to the treatment of phaeochromocytoma, neuroblastoma, carcinoid and medullary thyroid carcinoma. To determine the feasibility of MIBG targeting for a wider range of tumor types, we employed plasmid-mediated transfer of the NAT gene into a human glioblastoma cell line (UVW) which does not express the NAT gene. This resulted in a 15-fold increase in uptake of MIBG by the host cells. A dose-dependent toxicity of 131I MIBG to the transfectants was demonstrated using three methods: (1) survival of clonogens derived from monolayer culture; (2) survival of clonogens derived from disaggregated multicellular spheroids; and (3) spheroid growth delay. 131I MIBG was twice as toxic to cells in spheroids compared with those in monolayers, consistent with a greater effect of radiation cross-fire (radiological bystander effect) from 131I beta-radiation in the three-dimensional tumor spheroids. The highest concentration of 131I MIBG tested (1 MBq/ml) was nontoxic to UVW control cells or spheroids transfected with the NAT gene in reverse orientation. These findings are encouraging for the development of NAT gene transfer-mediated 131I MIBG therapy.


Subject(s)
3-Iodobenzylguanidine/metabolism , Antineoplastic Agents/therapeutic use , Carrier Proteins/genetics , Glioblastoma/radiotherapy , Radiopharmaceuticals/therapeutic use , Symporters , Cell Division , Cell Survival , Dose-Response Relationship, Drug , Gene Transfer Techniques , Glioblastoma/pathology , Humans , Norepinephrine Plasma Membrane Transport Proteins , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...