Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
1.
Arch Toxicol ; 93(7): 2035-2043, 2019 07.
Article in English | MEDLINE | ID: mdl-31209508

ABSTRACT

Paraoxonase 1 (PON1) is a high-density lipoprotein (HDL)-associated enzyme that by hydrolysing exogenous and endogenous substrates can provide protection against substrate induced toxicity. To investigate the extent to which PON1 provides protection against lactone induced DNA damage, DNA damage was measured in HepG2 cells using the neutral Comet assay following lactone treatment in the presence and absence of exogenous recombinant PON1 (rPON1). Low dose lactones (10 mM) caused little or no damage while high doses (100 mM) induced DNA damage in the following order of potency: α-angelica lactone > γ-butyrolactone ~ γ-hexalactone > γ-heptalactone ~ γ-octaclactone ~ γ-furanone ~ γ-valerolactone > γ-decalactone. Co-incubation of 100 mM lactone with rPON1, resulted in almost all cells showing extensive DNA damage, particularly with those lactones that decreased rPON1 activity by > 25%. In contrast, with the lactones that are poor rPON1 subtrates (γ-decalactone and γ-furanone), rPON1 did not increase DNA damage. DNA damage induced by a 1 h co-treatment with 10 mM α-angelica lactone and rPON1 was reduced when cells when incubated for a further 4 h in fresh medium suggesting break formation was due to induced DNA damage rather than apoptosis. Preincubation (1-6 h) of α-angelica lactone with rPON1 in the absence of cells, decreased cellular DNA damage by around 40%  in comparison to cells treated without preincubation. These results suggest that in addition to its well-recognised detoxification effects, PON1 can increase genotoxicity potentially by hydrolysing certain lactones to reactive intermediates that increase DNA damage via the formation of DNA adducts.


Subject(s)
Aryldialkylphosphatase/metabolism , DNA Damage/drug effects , Lactones/toxicity , Aryldialkylphosphatase/administration & dosage , Comet Assay , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Lactones/administration & dosage , Time Factors
2.
Neurotoxicology ; 59: 105-109, 2017 03.
Article in English | MEDLINE | ID: mdl-28163087

ABSTRACT

BACKGROUND: L-ß-N-methylamino-l-alanine (BMAA) is a non-proteinic amino acid, that is neurotoxic in vitro and in animals, and is implicated in the causation of amyotrophic lateral sclerosis and parkinsonism-dementia complex (ALS-PDC) on Guam. Given that natural amino acids can be N-nitrosated to form toxic alkylating agents and the structural similarity of BMAA to other amino acids, our hypothesis was that N-nitrosation of BMAA might result in a toxic alkylating agent, providing a novel mechanistic hypothesis for BMAA action. FINDINGS: We have chemically nitrosated BMAA with sodium nitrite to produce nitrosated BMAA (N-BMAA) which was shown to react with the alkyl-trapping agent, 4-(p-nitrobenzyl)pyridine, cause DNA strand breaks in vitro and was toxic to the human neuroblastoma cell line SH-SY5Y under conditions in which BMAA itself was minimally toxic. CONCLUSIONS: Our results indicate that N-BMAA is an alkylating agent and toxin suggesting a plausible and previously unrecognised mechanism for the neurotoxic effects of BMAA.


Subject(s)
Alkylating Agents/toxicity , Amino Acids, Diamino/chemistry , DNA Damage/drug effects , Nitrates , Pyridines/toxicity , Cell Line, Tumor , Cyanobacteria Toxins , DNA Breaks, Single-Stranded/drug effects , Dose-Response Relationship, Drug , Humans , Neuroblastoma , Nitrosation/drug effects
3.
Curr Med Chem ; 20(19): 2389-401, 2013.
Article in English | MEDLINE | ID: mdl-23521681

ABSTRACT

Acute myeloid leukemia (AML) is a highly lethal disease, especially in old patients. Chemoresistance and the absence of host immune responses against autochthonous malignancy play a major role in the poor prognosis of AML. The triazene compounds Dacarbazine and Temozolomide are monofunctional alkylators that donate methyl groups to many sites in DNA, including the O(6)-position of guanine producing O(6)-methylguanine (O(6)-MeG). If not repaired, O(6)-MeG frequently mispairs with thymine during DNA duplication. O(6)-MeG:T mismatches can be recognized by the mismatch repair (MMR) system which activates a cascade of molecular events leading to cell cycle arrest and cell death. If MMR is defective, cells continue to divide and GC → AT transition mutations occur. In preclinical models, such mutations can lead to the appearance of abnormal proteins containing non-self peptides ("chemical xenogenization" CX) that can be recognized by host cell-mediated immunity. Repair of O(6)-MeG is achieved by the DNA repair protein, O(6)-methylguanine-DNA methyltransferase (MGMT), which removes the methyl adduct in an autoinactivating stoichiometric reaction. High MGMT levels attenuate the pharmacodynamic effects of triazenes. In the last few years, triazenes, alone or with MGMT inhibitors, have been tested in AML. In view of their potential activity as CX inducers, triazenes could offer the additional advantage of host anti-leukemia immune responses. The present paper describes several studies of leukemia treatment with triazenes and a case of acute refractory leukemia with massive skin infiltration by malignant cells. Treatment with Temozolomide and Lomeguatrib, a potent MGMT inhibitor, produced a huge, although transient, blastolysis and complete disappearance of all skin lesions.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Triazenes/therapeutic use , Animals , Antineoplastic Agents, Alkylating/chemistry , Dacarbazine/chemistry , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Temozolomide , Triazenes/chemistry
4.
Int J Cancer ; 131(1): 59-69, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-21805478

ABSTRACT

Myxoid Liposarcomas (MLS), characterized by the expression of FUS-CHOP fusion gene are clinically very sensitive to the DNA binding antitumor agent, trabectedin. However, resistance eventually occurs, preventing disease eradication. To investigate the mechanisms of resistance, a trabectedin resistant cell line, 402-91/ET, was developed. The resistance to trabectedin was not related to the expression of MDR related proteins, uptake/efflux of trabectedin or GSH levels that were similar in parental and resistant cells. The 402-91/ET cells were hypersensitive to UV light because of a nucleotide excision repair defect: XPG complementation decreased sensitivity to UV rays, but only partially to trabectedin. 402-91/ET cells showed collateral sensitivity to temozolomide due to the lack of O(6) -methylguanine-DNA-methyltransferase (MGMT) activity, related to the hypermethylation of MGMT promoter. In 402-91 cells chromatin immunoprecipitation (ChIP) assays showed that FUS-CHOP was bound to the PTX3 and FN1 gene promoters, as previously described, and trabectedin caused FUS-CHOP detachment from DNA. Here we report that, in contrast, in 402-91/ET cells, FUS-CHOP was not bound to these promoters. Differences in the modulation of transcription of genes involved in different pathways including signal transduction, apoptosis and stress response between the two cell lines were found. Trabectedin activates the transcription of genes involved in the adipogenic-program such as c/EBPα and ß, in 402-91 but not in 402-91/ET cell lines. The collateral sensitivity of 402-91/ET to temozolomide provides the rationale to investigate the potential use of methylating agents in MLS patients resistant to trabectedin.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Dioxoles/pharmacology , Liposarcoma, Myxoid/genetics , Liposarcoma, Myxoid/metabolism , Tetrahydroisoquinolines/pharmacology , Apoptosis , C-Reactive Protein/genetics , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , DNA Methylation , DNA Modification Methylases/deficiency , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Resistance, Neoplasm , Fibronectins/genetics , Humans , Liposarcoma, Myxoid/drug therapy , Liposarcoma, Myxoid/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Serum Amyloid P-Component/genetics , Signal Transduction , Temozolomide , Trabectedin , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ultraviolet Rays
5.
Br J Cancer ; 104(5): 750-5, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21326243

ABSTRACT

BACKGROUND: Poly adenosine diphosphate (ADP)-ribose polymerase (PARP) is essential in cellular processing of DNA damage via the base excision repair pathway (BER). The PARP inhibition can be directly cytotoxic to tumour cells and augments the anti-tumour effects of DNA-damaging agents. This study evaluated the optimally tolerated dose of olaparib (4-(3--4-fluorophenyl) methyl-1(2H)-one; AZD2281, KU0059436), a potent PARP inhibitor, with dacarbazine and assessed safety, toxicity, clinical pharmacokinetics and efficacy of combination treatment. PATIENTS AND METHODS: Patients with advanced cancer received olaparib (20-200 mg PO) on days 1-7 with dacarbazine (600-800 mg m(-2) IV) on day 1 (cycle 2, day 2) of a 21-day cycle. An expansion cohort of chemonaive melanoma patients was treated at an optimally tolerated dose. The BER enzyme, methylpurine-DNA glycosylase and its substrate 7-methylguanine were quantified in peripheral blood mononuclear cells. RESULTS: The optimal combination to proceed to phase II was defined as 100 mg bd olaparib with 600 mg m(-2) dacarbazine. Dose-limiting toxicities were neutropaenia and thrombocytopaenia. There were two partial responses, both in patients with melanoma. CONCLUSION: This study defined a tolerable dose of olaparib in combination with dacarbazine, but there were no responses in chemonaive melanoma patients, demonstrating no clinical advantage over single-agent dacarbazine at these doses.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dacarbazine/administration & dosage , Neoplasms/drug therapy , Phthalazines/administration & dosage , Phthalazines/adverse effects , Piperazines/administration & dosage , Piperazines/adverse effects , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Dacarbazine/adverse effects , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Melanoma/drug therapy , Middle Aged , Neutropenia/chemically induced , Poly(ADP-ribose) Polymerase Inhibitors , Thrombocytopenia/chemically induced
6.
Cancer Chemother Pharmacol ; 66(5): 829-35, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20039040

ABSTRACT

BACKGROUND: Expression of the DNA repair protein O (6)-methylguanine-DNA methyltransferase (MGMT) correlates with resistance to irinotecan in colorectal cancer cell lines. This phase I study evaluated the maximum tolerated dose (MTD) of lomeguatrib, an inactivating pseudosubstrate of MGMT, in combination with irinotecan in patients with metastatic colorectal cancer and assessed the safety, toxicity and clinical pharmacology of combination treatment. PATIENTS AND METHODS: Patients with metastatic colorectal cancer received lomeguatrib (10-80 mg PO) on days 1-5 with irinotecan (250-350 mg/m(2) IV) on day 4 of a 21-day cycle. RESULTS: Twenty-four patients, pre-treated with a median of 2 lines of chemotherapy, received 104 cycles of treatment. The MTD was defined as 80 mg/day lomeguatrib with 300 mg/m(2) irinotecan. The main toxicities observed were neutropaenia and diarrhoea. Lomeguatrib of 80 mg/day produced complete MGMT depletion in all available peripheral blood mononuclear cells (PBMCs) and paired tumour biopsies (one patient). There was no pharmacokinetic interaction between the drugs. In 22 patients assessable for tumour response, one achieved a partial response and 16 had stable disease. CONCLUSION: This study defined a tolerable dose of irinotecan in combination with lomeguatrib in patients with metastatic colorectal cancer. Combination treatment gave a similar response rate to irinotecan monotherapy in this heavily pre-treated patient group.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , O(6)-Methylguanine-DNA Methyltransferase/drug effects , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Interactions , Female , Humans , Irinotecan , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Purines/administration & dosage , Treatment Outcome
7.
Br J Cancer ; 100(8): 1245-9, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19367282

ABSTRACT

Lomeguatrib, an O(6)-methylguanine-DNA methyltransferase inactivator, was evaluated in an extended dosing regimen with temozolomide, designed according to pharmacodynamic data from previous studies. Patients with unresectable stage 3 or 4 cutaneous or unknown primary melanoma metastases were treated with lomeguatrib 40 mg, b.i.d. for 10 or 14 days and temozolomide 75-100 mg m(-2) on days 1-5. Drugs were administered orally with cycles repeated every 28 days, for up to six cycles. A total of 32 patients were recruited to the study. Lomeguatrib for 10 days with temozolomide 75 mg m(-2) was established as the optimal extended lomeguatrib dosing schedule, with haematological toxicity being dose limiting. There were two partial responses to treatment giving an overall response rate of 6.25%. Extending lomeguatrib administration beyond that of temozolomide requires a reduced dose of the latter agent. Only limited clinical activity was seen, suggesting no advantage for this regimen over conventional temozolomide administration in the treatment of melanoma.


Subject(s)
Antineoplastic Agents/toxicity , Dacarbazine/analogs & derivatives , Melanoma/drug therapy , Purines/toxicity , Skin Neoplasms/drug therapy , Aged , Aged, 80 and over , Anemia/chemically induced , Child , Dacarbazine/toxicity , Dose-Response Relationship, Drug , Female , Humans , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Neutropenia/chemically induced , Patient Selection , Skin Neoplasms/pathology , Temozolomide , Thrombocytopenia/chemically induced
8.
Br J Cancer ; 100(8): 1250-6, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19367283

ABSTRACT

We evaluated the pharmacodynamic effects of the O(6)-methylguanine-DNA methyltransferase (MGMT) inactivator lomeguatrib (LM) on patients with melanoma in two clinical trials. Patients received temozolomide (TMZ) for 5 days either alone or with LM for 5, 10 or 14 days. Peripheral blood mononuclear cells (PBMCs) were isolated before treatment and during cycle 1. Where available, tumour biopsies were obtained after the last drug dose in cycle 1. Samples were assayed for MGMT activity, total MGMT protein, and O(6)-methylguanine (O(6)-meG) and N7-methylguanine levels in DNA. MGMT was completely inactivated in PBMC from patients receiving LM, but detectable in those on TMZ alone. Tumours biopsied on the last day of treatment showed complete inactivation of MGMT but there was recovery of activity in tumours sampled later. Significantly more O(6)-meG was present in the PBMC DNA of LM/TMZ patients than those on TMZ alone. LM/TMZ leads to greater MGMT inactivation, and higher levels of O(6)-meG than TMZ alone. Early recovery of MGMT activity in tumours suggested that more protracted dosing with LM is required. Extended dosing of LM completely inactivated PBMC MGMT, and resulted in persistent levels of O(6)-meG in PBMC DNA during treatment.


Subject(s)
DNA Damage , Dacarbazine/analogs & derivatives , Melanoma/drug therapy , Melanoma/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Purines/toxicity , Antineoplastic Agents/toxicity , Biopsy , DNA Damage/drug effects , DNA Repair/drug effects , DNA Replication/drug effects , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Dacarbazine/toxicity , Disease Progression , Humans , Kinetics , Melanoma/pathology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/drug effects , Temozolomide
9.
Carcinogenesis ; 30(4): 615-20, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19168588

ABSTRACT

Components of human diets may influence the incidence of colorectal adenomas, by modifying exposure or susceptibility to DNA-damaging alkylating agents. To examine this hypothesis, a food frequency questionnaire was used to assess the diet of patients recruited for a case-referent study where biopsies of normal colorectal mucosa were collected during colonoscopy and subsequently analysed for DNA N7-methylguanine (N7-MeG) levels, as an indicator of exposure, and activity of the DNA repair protein O6-alkylguanine DNA-alkyltransferase (MGMT), as an indicator of potential susceptibility. Cases with histologically proven colorectal adenomas (n = 38) were compared with referents (n = 35) free of gastrointestinal neoplasia. The case group consumed significantly more red meat (4.5 versus 3.4 servings/week, P < 0.05), processed meats, (4.7 versus 3.2 servings/week, P < 0.05) and % food energy as fat (34.9 versus 30.7%, P < 0.001). N7-MeG [mean: 95% confidence interval (CI)] levels were significantly lower in the group that consumed the highest proportion of dietary fibre/1000 kcal in comparison with the group with the lowest intake (0.61; 0.35-0.86 versus 1.88; 0.88-2.64 micromol/mol dG, P < 0.05). N7-MeG levels were also inversely associated with folate consumption (P < 0.05). MGMT activity (mean; 95% CI) was significantly higher in the group with the lowest consumption of vegetables than in the group with the greatest vegetable consumption (7.02; 5.70-8.33 versus 4.93; 3.95-5.91 fmol/microg DNA, P < 0.05). Our results are consistent with the hypothesis that dietary factors may modify exposure or susceptibility, respectively, to DNA damage by alkylating agents.


Subject(s)
Adenoma/metabolism , Colorectal Neoplasms/metabolism , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , DNA/metabolism , Diet , Gastrointestinal Neoplasms/metabolism , Guanine/analogs & derivatives , Tumor Suppressor Proteins/metabolism , Adenoma/pathology , Aged , Case-Control Studies , Cohort Studies , Colonic Polyps/enzymology , Colonic Polyps/genetics , Colonic Polyps/pathology , Colonoscopy , Colorectal Neoplasms/pathology , Female , Gastrointestinal Neoplasms/pathology , Guanine/metabolism , Humans , Male , Middle Aged
10.
Br J Cancer ; 98(10): 1614-8, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18475294

ABSTRACT

To evaluate the tumour response to lomeguatrib and temozolomide (TMZ) administered for 5 consecutive days every 4 weeks in patients with metastatic colorectal carcinoma. Patients with stage IV metastatic colorectal carcinoma received lomeguatrib (40 mg) and TMZ (50-200 mg m(-2)) orally for 5 consecutive days every 4 weeks. Response was determined every two cycles. Pharmacokinetics of lomeguatrib and TMZ as well as their pharmacodynamic effects in peripheral blood mononuclear cells (PBMC) were determined. Nineteen patients received 49 cycles of treatments. Despite consistent depletion of O(6)-methylguanine-DNA methyltransferase in PBMC, none of the patients responded to treatment. Three patients had stable disease, one for the duration of the study, and no fall in carcinoembryonic antigen was observed in any patient. Median time to progression was 50 days. The commonest adverse effects were gastrointestinal and haematological and these were comparable to those of TMZ when given alone. This combination of lomeguatrib and TMZ is not efficacious in metastatic colorectal cancer. If further studies are to be performed, emerging data suggest that higher daily doses of lomeguatrib and a dosing period beyond that of TMZ should be evaluated.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Dacarbazine/analogs & derivatives , Purines/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Disease Progression , Female , Humans , Male , Middle Aged , Neoplasm Staging , Purines/adverse effects , Temozolomide , Treatment Failure
11.
Gut ; 56(3): 380-4, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16891355

ABSTRACT

BACKGROUND AND AIMS: O(6)-alkylguanine DNA-alkyltransferase (MGMT) provides protection against alkylating agent-induced GC-->AT transition mutations. Such mutations are frequently seen in the KRAS oncogene of large colorectal adenomas, but whether adenoma or mutational risk in humans is influenced by MGMT activity and alkylating agent exposure is unclear. Hence, MGMT activity and, as an indicator of alkylating agent exposure, DNA-N7-methylguanine (N7-MeG) levels were determined in the normal tissue of patients with and without adenomas. METHODS: Biopsy specimens of normal colorectal mucosa were collected during colonoscopy from 85 patients with histologically proved colorectal adenomas (cases) and from 85 patients free of gastrointestinal neoplasia (referents) matched by age, sex and biopsy location. MGMT activity and N7-MeG levels were measured in colorectal tissue extracts and DNA, respectively. RESULTS: MGMT activity was higher in the normal mucosa of cases than in referents (6.65+/-3.03 vs 5.61+/-2.74 fmol/micro g DNA, p = 0.01). On stratification of cases, MGMT activity was found to be considerably greater in the normal mucosa of cases with large adenomas (p = 0.003) and slightly higher in cases with a GC-->AT transition mutation in the K-ras gene (p = 0.03). Elevated MGMT levels were associated with an increased risk of adenoma (OR 1.17, 95% CI 1.03 to 1.33 per unit increase in activity). Detectable levels of N7-MeG were found in DNA from 89% of cases and 93% of referents, with levels ranging from <0.1 to 7.7 micro mol/mol dG. Cases and referents had similar DNA-N7-MeG levels. CONCLUSIONS: Human exposure to methylating agents is widespread. MGMT activity is increased in the normal mucosa of patients with adenomas.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , Guanine/analogs & derivatives , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Adenoma/metabolism , Aged , Aged, 80 and over , Carrier Proteins/genetics , Carrier Proteins/metabolism , Case-Control Studies , Colonic Polyps/genetics , Colonic Polyps/metabolism , Colonoscopy , Colorectal Neoplasms/metabolism , DNA Mutational Analysis/methods , DNA Repair , DNA, Neoplasm/genetics , Female , Guanine/metabolism , Humans , Intestinal Mucosa/enzymology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
12.
Br J Cancer ; 95(7): 822-8, 2006 Oct 09.
Article in English | MEDLINE | ID: mdl-16969346

ABSTRACT

To determine the efficacy and tolerability of SPI-77 (sterically stabilised liposomal cisplatin) at three dose levels in patients with advanced non-small-cell lung cancer (NSCLC). Patients had Stage IIIB or IV NSCLC and were chemo-naïve, and Eastern Oncology Cooperative Group 0-2. The first cohort received SPI-77 at 100 mg m-2, the second 200 mg m-2 and the final cohort 260 mg m-2. Patients had also pharmacokinetics and analysis of leucocyte platinum (Pt)-DNA adducts performed. Twenty-six patients were treated, with 22 patients being evaluable for response. Only one response occurred at the 200 mg m-2 dose level for an overall response rate of 4.5% (7.1% at >or=200 mg m-2). No significant toxicity was noted including nephrotoxicity or ototoxicity aside from two patients with Grade 3 nausea. No routine antiemetics or hydration was used. The pharmacokinetic profile of SPI-77 was typical for a liposomally formulated drug, and the AUC appeared to be proportional to the dose of SPI-77. Plasma Pt levels and leucocyte DNA adduct levels did not appear to rise with successive doses. SPI-77 demonstrates only modest activity in patients with NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/administration & dosage , Lung Neoplasms/drug therapy , Aged , Area Under Curve , Carcinoma, Non-Small-Cell Lung/mortality , Cisplatin/adverse effects , Cisplatin/metabolism , Cisplatin/pharmacokinetics , DNA Adducts/metabolism , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Leukocytes/drug effects , Lung Neoplasms/mortality , Male , Metabolic Clearance Rate , Middle Aged , Neoplasm Staging , Platinum/blood , Survival Analysis , Treatment Outcome
13.
Gene Ther ; 13(13): 1037-47, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16541120

ABSTRACT

The failure of pharmacological approaches to cure infection with the human immunodeficiency virus (HIV) has renewed the interest in gene-based therapies. Among the various strategies that are currently explored, the blockade of HIV entry into susceptible T cells and macrophages promises to be the most powerful intervention. For long-term protection of both of these lineages, genetic modification of hematopoietic stem cells (HSCs) would be required. Here, we tested whether HSCs and their progeny can be modified to express therapeutic levels of M87o, a gammaretroviral vector encoding an artificial transmembrane molecule that blocks fusion-mediated uptake of HIV. In serial murine bone marrow transplantations, efficient and multilineage expression of M87o was observed for more than 1 year (range 37-75% of mononuclear cells), without signs of toxicity related to the transmembrane molecule. To allow enrichment of M87o-modified HSCs after transplant, we constructed vectors coexpressing the P140K mutant of O(6)-methylguanine-DNA-methyltransferase (MGMT-P140K). This clinically relevant selection marker mediates a survival advantage in HSCs if exposed to combinations of methylguanine-methyltransferase (MGMT) inhibitors and alkylating agents. A bicistronic vector mediated sufficient expression of both M87o and MGMT to confer a selective survival advantage in the presence of HIV and alkylating agents, respectively. These data encourage further investigations in large animal models and clinical trials.


Subject(s)
AIDS Vaccines/administration & dosage , Genetic Therapy/methods , HIV Fusion Inhibitors/therapeutic use , HIV Infections/prevention & control , HIV-1 , Hematopoietic Stem Cells/metabolism , AIDS Vaccines/genetics , Animals , Flow Cytometry , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HIV Infections/metabolism , Hematopoietic Stem Cell Transplantation/methods , Male , Mice , Mice, Inbred C57BL , Models, Animal , O(6)-Methylguanine-DNA Methyltransferase/analysis , Retroviridae/genetics , Transduction, Genetic/methods
14.
Br J Cancer ; 93(10): 1152-6, 2005 Nov 14.
Article in English | MEDLINE | ID: mdl-16278661

ABSTRACT

Tumour resistance to chemotherapy involving methylating agents such as DTIC (dacarbazine) and temozolomide is linked to expression of the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (MGMT). There is considerable interest in improving the efficacy of such O(6)-alkylating chemotherapy by the prior inactivation of MGMT. We have examined the effect of the modified guanine base, O(6)-(4-bromothenyl)guanine (PaTrin-2, Patrin, Lomeguatrib) on MGMT activity and cell or xenograft tumour growth inhibition by temozolomide in the human breast carcinosarcoma cell line, MCF-7. PaTrin-2 effectively inactivated MGMT in MCF-7 cells (IC(50) approximately 6 nM) and in xenografts there was complete inactivation of MGMT within 2 h of dosing (20 mg kg(-1) i.p.) and only slight recovery by 24 h. MGMT inactivation in a range of murine host tissues varied between complete and approximately 60%, with extensive recovery by 24 h. PaTrin-2 (10 microM) substantially increased the growth inhibitory effects of temozolomide in MCF-7 cells (D(60)=10 microM with PaTrin-2 vs 400 microM without). In MCF-7 xenografts, neither temozolomide (100 mg kg(-1) day(-1) for 5 days) nor PaTrin-2 (20 mg kg(-1) day(-1) for 5 days) had any significant effect on tumour growth. In contrast, the PaTrin-2-temozolomide combination produced a substantial tumour growth delay: median tumour quintupling time was increase by 22 days (P<0.005) without any significant increase in toxicity as assessed from animal weight. A PaTrin-2-temozolomide combination may therefore be beneficial in the treatment of human breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Guanine/analogs & derivatives , Animals , Body Weight/drug effects , Breast Neoplasms/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Guanine/pharmacology , Humans , Male , Mice , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Temozolomide , Xenograft Model Antitumor Assays
15.
Br J Cancer ; 93(5): 529-37, 2005 Sep 05.
Article in English | MEDLINE | ID: mdl-16136028

ABSTRACT

Cisplatin may have additive activity with temozolomide due to ablation of the DNA repair protein O6-alkylguanine-DNA alkyltransferase (MGMT). This phase I/II study determined recommended combination doses using the Continual Reassessment Method, toxicities and antitumour activity in paediatric patients, and evaluated MGMT in peripheral blood mononuclear cells (PBMCs) in order to correlate with haematological toxicity. In total, 39 patients with refractory or recurrent solid tumours (median age approximately 13 years; 14 pretreated with high-dose chemotherapy, craniospinal irradiation, or having bone marrow involvement) were treated with cisplatin, followed the next day by oral temozolomide for 5 days every 4 weeks at dose levels 80 mg m(-2)/150 mg m(-2) day(-1), 80/200, and 100/200, respectively. A total of 38 patients receiving 113 cycles (median 2, range 1-7) were evaluable for toxicity. Dose-limiting toxicity was haematological in all but one case. Treatment-related toxicities were thrombocytopenia, neutropenia, nausea-vomiting, asthenia. Hearing loss was experienced in five patients with prior irradiation to the brain stem or posterior fossa. Partial responses were observed in two malignant glioma, one brain stem glioma, and two neuroblastoma. Median MGMT activity in PBMCs decreased after 5 days of temozolomide treatment: low MGMT activity correlated with increased severity of thrombocytopenia. Cisplatin-temozolomide combinations are well tolerated without additional toxicity to single-agent treatments; the recommended phase II dosage is 80 mg m(-2) cisplatin and 150 mg m(-2) x 5 temozolomide in heavily treated, and 200 mg m(-2) x 5 temozolomide in less-heavily pretreated children.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Adolescent , Adult , Child , Child, Preschool , Cisplatin/administration & dosage , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Female , Humans , Infant , Male , Maximum Tolerated Dose , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/epidemiology , Neoplasm Staging , Neoplasms/enzymology , Salvage Therapy , Temozolomide , Treatment Outcome
16.
Int J Biol Markers ; 20(1): 69-72, 2005.
Article in English | MEDLINE | ID: mdl-15832776

ABSTRACT

Previous studies suggest that bladder cancer risk may vary with GST genotype but these results are inconsistent. The aim of this study was to explore whether GSTM1, GSTT1 and GSTP polymorphisms were associated with increased bladder cancer risk in an Egyptian population. GSTM1, GSTT1 and GSTP1 genotype frequencies were determined in bladder cancer cases (n=72) and healthy controls with no history of malignancies (n=82) using PCR-based techniques. The GSTT1*2 genotype was particularly associated with increased risk (OR 2.71, 95%CI 1.27-5.73) and the GSTM1*2 genotype to a lesser extent (OR 1.63, 95%CI 0.85-3.10). 18.1% of cases but only 7.3% of controls were GSTP1*B*B homozygotes (OR 2.38, 95%CI 0.83-6.87). The presence of two or more a priori at-risk genotypes was associated with increased bladder cancer risk (OR 2.42; 95%CI 1.47-3.97). These results suggest that polymorphisms in the GST genes are associated with increased risk of bladder cancer among Egyptians.


Subject(s)
Genetic Predisposition to Disease/genetics , Glutathione Transferase/genetics , Isoenzymes/genetics , Polymorphism, Genetic/genetics , Urinary Bladder Neoplasms/genetics , Adult , Aged , Egypt , Glutathione S-Transferase pi , Humans , Middle Aged
17.
Br J Cancer ; 92(1): 125-30, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15611794

ABSTRACT

The epidermal growth factor receptor (EGFR) is expressed in a wide variety of epithelial tumours including carcinoma of the bladder. Stimulation of the EGFR pathway is blocked by ZD1839 (Iressa), a highly selective EGFR tyrosine kinase inhibitor. Radical radiotherapy is an established organ sparing treatment option for muscle invasive bladder cancer and this study has explored the possibility for the use of ZD1839 as a radiosensitiser in this scenario. The effect of combination treatment with ZD1839 (0.01 microM) and ionising radiation in the established bladder cancer cell lines MGH-U1 and its radiosensitive mutant clone S40b was measured by clonogenic assays. A highly significant radiosensitising effect was seen in both cell lines (P < 0.001 for MGH-U1 and S40b cell lines). This effect was independent of the concentration of the drug and the duration of exposure prior to treatment with ionising radiation. Cell cycle kinetics of both cell lines was not significantly altered with ZD1839 (0.01 microM) as a single agent. A modest induction of apoptosis was observed with ZD1839 (0.01 microM) as a single agent, but a marked induction was observed with the combination treatment of ZD1839 and ionising radiation. These results suggest a potentially important role for ZD1839 in combination with radiotherapy in the treatment of muscle invasive bladder cancer.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/radiotherapy , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Enzyme Inhibitors/pharmacology , Gefitinib , Humans , Tumor Cells, Cultured
18.
Br J Cancer ; 90(2): 542-8, 2004 Jan 26.
Article in English | MEDLINE | ID: mdl-14735206

ABSTRACT

The aim of this study was to establish the radiosensitising properties of gemcitabine in a pair of related bladder tumour cell lines with differential radiosensitivity. The radioresistant bladder tumour cell line MGH-U1 and its radiosensitive mutant clone, S40b (both p53 mutant), had SF(2) values (surviving fraction at 2 Gy) of 0.98 and 0.64, respectively (P<0.001). Colony-forming assays showed that at 0.01 microM gemcitabine radiosensitisation occurred only in the S40b cell line (dose-modifying factor (DMF)=1.4). At 0.3 microM (killing 50% of cells), both cell lines were radiosensitised; DMF=2.25 and 1.2 for MGH-U1 and S40b, respectively. These data suggest that gemcitabine is an effective radiosensitiser in bladder cancer cell lines, with greater sensitisation in the radioresistant parental line-a feature that should be useful in a clinical setting.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Radiation-Sensitizing Agents/pharmacology , Urinary Bladder Neoplasms/pathology , Colony-Forming Units Assay , Humans , Radiation Tolerance , Tumor Cells, Cultured , Gemcitabine
19.
Carcinogenesis ; 24(10): 1691-4, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12896905

ABSTRACT

O6-Alkylguanine-DNA alkyltransferase (MGMT) confers resistance to many of the mutagenic and toxic effects of certain classes of alkylating agents by repairing the DNA lesions responsible. The levels of expression of this protein are of interest in relation to the prevention and treatment of cancer in man. They vary widely between individuals, and the basis of this variation is not understood. RT-PCR-RFLP analysis of mRNA from normal human lung tissue reveals that the two MGMT alleles are frequently expressed at different levels, indicating that there is a genetic component to inter-individual variation of MGMT levels and that at least some of this variation maps close to or within the MGMT locus.


Subject(s)
Lung/physiology , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/genetics , RNA, Messenger/analysis , Alleles , DNA Repair/genetics , Humans , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
20.
Mini Rev Med Chem ; 3(5): 471-85, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12769698

ABSTRACT

The protein O(6)-alkylguanine-DNA alkyltransferase is the basis of an important process for repairing damage to cellular DNA, which renders cells resistant to drugs that alkylate at the O(6)-position of guanine residues. The development of various pseudosubstrates which inactivate this protein is reviewed, from a chemical standpoint. Study of the influence of pseudosubstrate molecular structure on their interaction with the active site cysteine has progressed together with direct investigation of protein structure. Combination therapy using a powerful inactivator with a suitable alkylating agent shows great clinical promise in the treatment of cancer, particularly when some degree of selectivity is possible.


Subject(s)
Neoplasms/drug therapy , Neoplasms/enzymology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Alkylation , Nucleosides/pharmacology , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/metabolism , O(6)-Methylguanine-DNA Methyltransferase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...