Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Br J Cancer ; 109(5): 1223-9, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23942068

ABSTRACT

BACKGROUND: Pt-(GpG) intrastrand crosslinks are the major DNA adducts induced by platinum-based anticancer drugs. In the cell lines and mouse models, the persistence of these lesions correlates significantly with cell damage. Here we studied Pt-(GpG) DNA adducts in circulating tumour cells (CTC) treated with cisplatin in medium upfront to systemic therapy from patients with advanced non-small-cell lung cancer (NSCLC). METHODS: Blood was drawn before systemic treatment and the CD45/CD15-depleted fraction of mononuclear cells was exposed to cisplatin, verified for the presence of CTC by pan-cytokeratin (pCK) staining and immunoanalysed for the level of Pt-(GpG) in DNA. RESULTS: Immunostaining for pCK, CD45 and subsequently for Pt-(GpG) adducts in the cisplatin-exposed cells (ex vivo) at different time points depicted distinct differences for adduct persistence in CTC between responders vs non-responders. CONCLUSION: Pt-(GpG) adducts can be detected in CTC from NSCLC patients and assessing their kinetics may constitute a clinically feasible biomarker for response prediction and dose individualisation of platinum-based chemotherapy. This functional pre-therapeutic test might represent a more biological approach than measuring protein factors or other molecular markers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , DNA Adducts/biosynthesis , Dinucleoside Phosphates/chemistry , Lung Neoplasms/drug therapy , Platinum/chemistry , Aged , Cisplatin/pharmacology , Cisplatin/therapeutic use , DNA Adducts/genetics , Humans , Leukocyte Common Antigens/metabolism , Leukocytes, Mononuclear/drug effects , Lewis X Antigen/metabolism , Middle Aged , Neoplastic Cells, Circulating , Tumor Cells, Cultured
2.
Br J Cancer ; 108(2): 334-41, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23257893

ABSTRACT

BACKGROUND: Sorafenib has recently been shown to reduce tumour growth in hepatoblastoma (HB) xenografts. The effect of a combined administration with cytostatic agents was now investigated. METHODS: Cell viability after treatment with sorafenib and different cytostatic agents was evaluated in two HB cell lines (HUH6 and HepT1) using MTT assay. ERK signalling was investigated by western blot, NOXA expression by rt-PCR, and formation of DNA adducts using immunocytology. NMRI mice bearing subcutaneous HUH6-derived tumours were treated with sorafenib alone or in combination with cisplatin. Tumour progression, viability, apoptosis, and vascularisation were monitored by tumour volume, AFP levels, TUNEL assay, and CD31 immunostaining, respectively. RESULTS: The combination of sorafenib and cisplatin led to a remarkable decrease in cell viability. The cisplatin-induced enhanced ERK1/2 activation, but not NOXA expression and the formation of DNA adducts was partly abrogated by sorafenib. In HB xenografts, both, sorafenib and alternated application of sorafenib and cisplatin significantly reduced tumour growth (P<0.05). Levels of AFP were lower in both treated groups (P=0.08). Relative apoptotic areas were increased (P=0.003). Mean vascular density was the lowest in the sorafenib/CDDP group (P=0.02). CONCLUSION: The combination of sorafenib with cisplatin might be a promising treatment option for high risk or recurrent HB.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/therapeutic use , Cytostatic Agents/therapeutic use , Hepatoblastoma/drug therapy , Hepatoblastoma/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , DNA Adducts/metabolism , Enzyme Activation/drug effects , Female , Humans , Liver Neoplasms/drug therapy , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/pharmacology , Neovascularization, Pathologic , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Sorafenib , Xenograft Model Antitumor Assays
3.
Neuroscience ; 169(1): 463-74, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20417692

ABSTRACT

Bone-cancer-related pain is one of the most disabling factors in patients suffering from primary bone cancer or bone metastases. Recent studies point toward an important role of proinflammatory cytokines, example tumor necrosis factor-alpha (TNF), for tumor growth and bone-cancer-associated pain. Mechanisms by which TNF, through its receptor subtypes, TNF receptor 1 (TNFR1) and -2 (TNFR2), elicits altered sensation and pain behavior, are still incompletely understood. To look for a potential role of TNF in bone cancer pain, cancer-related pain was analyzed in fibrosarcoma-bearing C57Bl/6J wild type mice after systemic antagonism of TNF. To further clarify the role of TNF receptor (TNFR) in bone-cancer pain, naive and fibrosarcoma-bearing C57Bl/ 6J wild type and transgenic mice with a deficiency of TNFR1 (TNFR1ko), TNFR2 (TNFR2ko), and TNFR1+2 (TNFR1+2ko) were compared regarding cancer-related pain and hyperalgesia, tumor growth, osteoclast activation, and spinal astrogliosis. Systemic antagonism of TNF significantly alleviated tactile hypersensitivity and spontaneous bone-cancer-related pain behavior. Most interestingly, combined deletion of the TNFR1 and TNFR2, but not of either gene alone, almost completely inhibited the development of tactile hypersensitivity, whereas spontaneous pain behavior was transiently increased. Accordingly, spinal astrogliosis was markedly reduced, whereas tumor growth was significantly increased in TNFR1+2ko mice. In contrast, deletion of the TNFR1 or TNFR2 gene alone did not change tumor growth or spinal astrogliosis. Our findings suggest that the combined absence of TNFR1 and TNFR2 is necessary for the attenuation of cancer-related tactile hypersensitivity and concomitant spinal astrogliosis, whereas tumor growth seems to be inhibited by combined TNFR activation. These findings support the hypothesis of cytokine-dependent pain development in cancer pain. Differential targeting of TNFR activation could be an interesting strategy in bone-cancer-related pain conditions.


Subject(s)
Bone Neoplasms/secondary , Fibrosarcoma/secondary , Gliosis/physiopathology , Neoplasm Proteins/physiology , Pain/physiopathology , Receptors, Tumor Necrosis Factor, Type I/physiology , Receptors, Tumor Necrosis Factor/physiology , Spinal Cord/pathology , Tumor Necrosis Factor-alpha/physiology , Animals , Astrocytes/pathology , Behavior, Animal , Bone Neoplasms/physiopathology , Etanercept , Fibrosarcoma/physiopathology , Gliosis/etiology , Gliosis/prevention & control , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , Lameness, Animal/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoclasts/drug effects , Osteoclasts/pathology , Pain/etiology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/therapeutic use , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Touch , Tumor Necrosis Factor-alpha/antagonists & inhibitors
4.
Leukemia ; 20(10): 1774-82, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16932341

ABSTRACT

B-cell chronic lymphocytic leukaemia (B-CLL) is a heterogenous disease with a highly variable clinical course and analysis of zeta-associated protein 70 (ZAP-70) and CD38 expression on B-CLL cells allowed for identification of patients with good (ZAP-70-CD38-) and poor (ZAP-70+CD38+) prognosis. DNA microarray technology was employed to compare eight ZAP-70+CD38+ with eight ZAP-70-CD38- B-CLL cases. The expression of 358 genes differed significantly between the two subgroups, including genes involved in B-cell receptor signaling, angiogenesis and lymphomagenesis. Three of these genes, that is, immune receptor translocation-associated protein 4 (IRTA4)/Fc receptor homologue 2 (FcRH2), angiopoietin 2 (ANGPT2) and Pim2 were selected for further validating studies in a cohort of 94 B-CLL patients. IRTA4/FcRH2 expression as detected by flow cytometry was significantly lower in the poor prognosis subgroup as compared to ZAP-70-CD38- B-CLL cells. In healthy individuals, IRTA4/FcRH2 protein expression was associated with a CD19+CD27+ memory cell phenotype. ANGPT2 plasma concentrations were twofold higher in the poor prognosis subgroup (P<0.05). Pim2 was significantly overexpressed in poor prognosis cases and Binet stage C. Disease progression may be related to proangiogenic processes and strong Pim2 expression.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Membrane Glycoproteins/genetics , ZAP-70 Protein-Tyrosine Kinase/genetics , ADP-ribosyl Cyclase 1/metabolism , Aged , Aged, 80 and over , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , B-Lymphocytes/pathology , B-Lymphocytes/physiology , Cell Differentiation , Cohort Studies , Female , Flow Cytometry , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/classification , Leukemia, Lymphocytic, Chronic, B-Cell/epidemiology , Male , Membrane Glycoproteins/metabolism , Middle Aged , Neovascularization, Pathologic/genetics , Oligonucleotide Array Sequence Analysis , Prognosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcr/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Risk Factors , Signal Transduction/genetics , ZAP-70 Protein-Tyrosine Kinase/metabolism
5.
Mutat Res ; 450(1-2): 139-53, 2000 May 30.
Article in English | MEDLINE | ID: mdl-10838139

ABSTRACT

In this contribution we discuss the gene- and cell type-specific repair of miscoding DNA alkylation products as a risk parameter in both mutation induction and malignant transformation by N-nitroso carcinogens. Upon exposure to N-nitroso compounds such as N-methyl-N-nitrosourea (MeNU) or N-ethyl-N-nitrosourea (EtNU), about a dozen different alkylation products are formed in cellular DNA. Among these are O(6)-methylguanine (O(6)-MeGua) and O(6)-ethylguanine (O(6)-EtGua), respectively, which differ only by one CH(2) group in their alkyl residue and, when unrepaired, cause G:C-->A:T transition mutations by anomalous base pairing during DNA replication. We have analyzed the global and gene-specific repair of O(6)-MeGua and O(6)-EtGua in target cell DNA, ras gene mutation frequencies, and tumor incidence, in the model of mammary carcinogenesis induced in 50-day-old female Sprague-Dawley rats by a single application of MeNU or EtNU. Both carcinogens induce histologically indistinguishable mammary adenocarcinomas at high yield. In the target mammary epithelia, O(6)-MeGua is repaired at similar slow rates in both transcriptionally active genes (Ha-ras, beta-actin), silent genes (lgE heavy chain), and in bulk DNA, by the one-step repair protein O(6)-alkylguanine-DNA alkyltransferase (MGMT; low level of expression in the target cells). The slow repair of O(6)-MeGua translates into a high frequency of mutations at the central position of Ha-ras codon 12 (GGA) in MeNU-induced tumors. O(6)-EtGua, however, is removed approximately 20 times faster than O(6)-MeGua selectively from transcribed genes via an MGMT independent, as yet uncharacterized excision mechanism. Accordingly, no Ha-ras codon 12 mutations are found in the EtNU-induced mammary tumors. Neither MeNU- nor EtNU-induced tumors exhibit mutations at codons 13 and 61 of Ha-ras or at codons 12, 13 and 61 of Ki-ras. While a moderate surplus MGMT activity of the target cells - contributed by a bacterial MGMT transgene (ada) - significantly counteracts mammary tumorigenesis in MeNU-exposed rats, this is not the case in the EtNU-treated animals. Differential repair of structurally distinct DNA lesions in transcribed or (temporarily) silent genes thus determines the probability of mutation and, together with cell type-specific and interindividual differences in DNA repair capacity, influences carcinogenic risk.


Subject(s)
Carcinogens/toxicity , DNA Repair/genetics , Genes, ras/drug effects , Mutation , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , DNA/drug effects , DNA/genetics , DNA/metabolism , Ethylnitrosourea/toxicity , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Methylnitrosourea/toxicity , Models, Genetic , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Rats , Rats, Sprague-Dawley
6.
Mutat Res ; 462(2-3): 101-5, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10767621

ABSTRACT

DNA-reactive carcinogens and anticancer drugs induce many structurally distinct mutagenic and cytotoxic DNA lesions. The varying capability of normal and malignant cells to recognize and repair specific DNA lesions influences both cancer risk and the relative sensitivity or resistance of cancer cells towards cytotoxic agents. Using monoclonal antibody-based immunoanalytical assays, very low amounts of defined carcinogen-DNA adducts can be quantified in bulk genomic DNA, in individual genes, and in the nuclear DNA of single cells. DNA repair kinetics can, thus, be measured in a lesion-, gene-, and cell type-specific manner, and the DNA repair profiles of malignant cells can be monitored in individual patients. Even structurally very similar DNA lesions may be repaired with strongly differing efficiency. The miscoding DNA alkylation products O(6)-methylguanine and O(6)-ethylguanine, for example, differ only by one CH(2) group. These lesions are formed in DNA upon exposure to N-methyl-N-nitrosourea or N-ethyl-N-nitrosourea, both of which induce mammary adenocarcinomas in female rats at high yield. Unrepaired O(6)-alkylguanines in DNA cause G:C-->A:T transition mutations via mispairing during DNA replication. O(6)-methylguanines are repaired at a similar slow rate in both transcriptionally active (H-ras, beta-actin) and inactive genes (IgE heavy chain; bulk DNA) of the target mammary epithelia (which express the repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT) at a very low level). In contrast, O(6)-ethylguanines are repaired approximately 20 times faster than O(6)-methylguanines in both DNA strands of the transcribed genes selectively via an AGT-independent, as yet unclarified excision mechanism. Accordingly, G:C-->A:T transitions resulting from the misreplication of an O(6)-methylated guanine at the second position of codon 12 (GGA) of H-ras represent a frequent "signature" mutation in rat mammary adenocarcinomas that develop after exposure to N-methyl-N-nitrosourea. However, this mutation is not observed when these tumors are induced by N-ethyl-N-nitrosourea, due to the fast repair of O(6)-ethylguanines in the H-ras gene. The key importance of "conventional" and "conditional" gene knockout technology for resolving the intricacies of the complex network of DNA repair pathways is briefly discussed.


Subject(s)
DNA Repair , DNA/metabolism , Adenocarcinoma/chemically induced , Adenocarcinoma/metabolism , Alkylating Agents/administration & dosage , Alkylating Agents/toxicity , Animals , Carcinogens/administration & dosage , Carcinogens/toxicity , DNA/chemistry , DNA/drug effects , DNA Adducts/chemistry , DNA Adducts/metabolism , Drug Resistance, Neoplasm , Ethylnitrosourea/adverse effects , Ethylnitrosourea/toxicity , Female , Genes, ras/drug effects , Genes, ras/genetics , Guanine/analogs & derivatives , Guanine/chemistry , Guanine/metabolism , Humans , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/metabolism , Methylnitrosourea/administration & dosage , Methylnitrosourea/toxicity , Mutagens/administration & dosage , Mutagens/toxicity , Mutation , Rats
7.
Mutat Res ; 425(1): 125-34, 1999 Mar 10.
Article in English | MEDLINE | ID: mdl-10082923

ABSTRACT

N-Nitrosodialkylamines are potent carcinogens in experimental animals. Previously, we reported that the mutagenicity of N-nitrosodimethylamine (NDMA) was 10 times higher than that of N-nitrosodiethylamine (NDEA) in the Drosophila wing spot test. To find out how to explain this difference, we have measured the levels of O-alkylated bases in the DNA of exposed Drosophila larvae. Third instar larvae were fed for 3 or 6 h with NDMA or NDEA. Part of the treated larvae were grown to adult flies to score their wings for the presence of mutant spots. From the remaining larvae, DNA was isolated and digested to deoxyribonucleosides, and the digest fractionated by high-performance liquid chromatography (HPLC). The amounts of specific alkyldeoxyribonucleosides present in the fractions were quantified by a radioimmunoassay (RIA) using monoclonal antibodies. Dose-dependent O6-methylguanine, O6-ethylguanine and O4-ethylthymine formations were found to be correlated with the induction frequencies of mutant wing spots. At the same exposure dose, the values of O6-alkylde- oxyguanosine/106 deoxyguanosine were similar for NDMA and NDEA: on feeding 20 micromol/1.5 ml feeding solution, the values for NDMA were 4.0 with 3 h and 18.5 with 6 h of exposure; with 20 micromol NDEA, the corresponding values were 5.4 with 3 h and 14.6 with 6 h of exposure. The wing spot frequencies were very different; however, with NDMA, the total numbers of spots/wing were 3.5 (3 h) and 15 (6 h), and with NDEA 0.8 (3 h) and 0.9 (6 h). Similar discrepancies exist as well between the mutagenicities and the alkylation rates observed for O4-alkylthymidines. These results suggest that the difference between the mutagenic potencies of NDMA and NDEA cannot be explained by the amounts of O-alkyl adducts formed. Different mechanisms are considered by which NDMA and NDEA may produce the genetic effects observed.


Subject(s)
DNA Adducts , Diethylnitrosamine/toxicity , Dimethylnitrosamine/toxicity , Drosophila melanogaster/drug effects , Mutagens/toxicity , Alkylation , Animals , Drosophila melanogaster/genetics , Female , Male , Mutation , Recombination, Genetic
8.
Proc Natl Acad Sci U S A ; 95(4): 1635-40, 1998 Feb 17.
Article in English | MEDLINE | ID: mdl-9465068

ABSTRACT

Differential repair of structurally distinct mutagenic lesions in critical genes may influence the cellular risk of malignant conversion. We have investigated rat mammary tumorigenesis induced by N-ethyl-N-nitrosourea (EtNU) versus N-methyl-N-nitrosourea (MeNU) with respect to tumor incidence, ras gene mutation, and gene-specific repair. Both carcinogens induced mammary adenocarcinomas at high yield. In mammary epithelia (very low expression of O6-alkylguanine-DNA alkyltransferase, MGMT), O6-methylguanine (O6-MeGua) was eliminated from transcribed (H-ras and beta-actin) and inactive genes (IgE heavy chain) at the same slow rate as determined for bulk genomic DNA. The persistence of O6-MeGua in DNA correlated with a high frequency of G:C --> A:T transition mutations at codon 12 of the H-ras gene in MeNU-induced tumors. Repair of O6-ethylguanine (O6-EtGua), too, was slow in the IgE heavy chain gene as in bulk DNA. Contrasting with O6-MeGua, however, O6-EtGua was removed approximately 20 times faster from the active H-ras and beta-actin genes via MGMT-independent mechanism(s). Accordingly, no H-ras codon 12 mutations were found in EtNU-induced tumors, and 5- to 8-fold surplus alkyltransferase activity of the mammary epithelia-via a bacterial ada transgene-did not significantly counteract tumorigenesis in EtNU-exposed contrary to MeNU-treated animals. Neither MeNU- nor EtNU-induced tumors exhibited mutations at codons 13 and 61 of H-ras or codons 12, 13, and 61 of K-ras. Fast repair of O6-EtGua, but not O6-MeGua, in transcribed genes thus prevents mutational activation of H-ras when rat mammary carcinogenesis is initiated by EtNU in place of MeNU.


Subject(s)
DNA Adducts/metabolism , DNA Repair , Ethylnitrosourea , Genes, ras , Guanine/analogs & derivatives , Mammary Neoplasms, Experimental/genetics , Methylnitrosourea , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Alkylation , Animals , Animals, Genetically Modified , DNA, Neoplasm/genetics , Female , Guanine/metabolism , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/chemically induced , Point Mutation , Rats , Transcription, Genetic
9.
Recent Results Cancer Res ; 154: 127-46, 1998.
Article in English | MEDLINE | ID: mdl-10026996

ABSTRACT

DNA-reactive carcinogens and anticancer drugs induce many structurally distinct cytotoxic and potentially mutagenic DNA lesions. The capability of normal and malignant cells to recognize and repair different DNA lesions is an important variable influencing the risk of mutation and cancer as well as therapy resistance. Using monoclonal antibody-based immunoanalytical assays, very low amounts of defined carcinogen-DNA adducts can be quantified in bulk genomic DNA, individual genes, and in the nuclear DNA of single cells. The kinetics of DNA repair can thus be measured in a lesion-, gene-, and cell type-specific manner, and the DNA repair profiles of malignant cells can be monitored in individual patients. Even structurally very similar DNa lesions may be repaired with extremely different efficiency. The miscoding DNA alkylation products O6-methylguanine (O6-MeGua) and O6-ethylguanine (O6-EtGua), for example, differ only by one CH2 group. These lesions are formed in DNA upon exposure to N-methyl-N-nitrosourea (MeNU) or N-ethyl-N-nitrosourea (EtNU), both of which induce mammary adenocarcinomas in female rats at high yield. Unrepaired O6-alkylguanines cause transition mutations via mispairing during DNA replication. O6-MeGua is repaired at a similar slow rate in transcribed (H-ras, beta-actin) and inactive genes (IgE heavy chain; bulk DNA) of the target mammary epithelia (which express the repair protein O6-alkylguanine-DNA alkyltransferase at a very low level). O6-EtGua, however, via an alkyltransferase-independent mechanism, is excised approximately 20 times faster than O6-MeGua from the transcribed genes selectively. Correspondingly, G:C-->A:T transitions arising from unrepaired O6-MeGua at the second nucleotide of codon 12 (GGA) of the H-ras gene are frequently found in MeNU-induced mammary tumors, but are absent in their EtNU-induced counterparts.


Subject(s)
DNA Repair , Neoplasms/etiology , Animals , Humans , Neoplasms/therapy , Transcription, Genetic
10.
Cytotechnology ; 27(1-3): 175-85, 1998 Sep.
Article in English | MEDLINE | ID: mdl-19002791

ABSTRACT

Most cytotoxic agents exert their action via damage of DNA. Therefore, the repair of such lesions is of major importance for the sensitivity of malignant cells to chemotherapeutic agents. The underlying mechanisms of various DNA repair pathways have extensively been studied in yeast, bacteria and mammalian cells. Sensitive and drug resistant cancer cell lines have provided models for analysis of the contribution of DNA repair to chemosensitivity. However, the validity of results obtained by laboratory experiments with regard to the clinical situation is limited. In both acute and chronic leukaemias, the emergence of drug resistant cells is a major cause for treatment failure. Recently, assays have become available to measure cellular DNA repair capacity in clinical specimens at the single-cell level. Application of these assays to isolated lymphocytes from patients with chronic lymphatic leukaemia (CLL) revealed large interindividual differences in DNA repair rates. Accelerated O(6)-ethylguanine elimination from DNA and faster processing of repair-induced single-strand breaks were found in CLL lymphocytes from patients nonresponsive to chemotherapy with alkylating agents compared to untreated or treated sensitive patients. Moreover, modulators of DNA repair with different target mechanisms were identified which also influence the sensitivity of cancer cells to alkylating agents. In this article, we review the current knowledge about the contribution of DNA repair to drug resistance in human leukaemia.

11.
Cancer Res ; 57(4): 651-8, 1997 Feb 15.
Article in English | MEDLINE | ID: mdl-9044842

ABSTRACT

Recent evidence has linked cellular DNA repair capacity to the chemosensitivity of cancer cells to alkylating agents. Using single-cell gel electrophoresis ("comet assay"), we have analyzed the induction and differential processing of DNA damage in human lymphocytes derived from healthy donors and from patients with chronic lymphatic leukemia (CLL) after exposure to N-ethyl-N-nitrosourea in vitro. The extent of comet formation in lymphocytes after N-ethyl-N-nitrosourea exposure appears to depend predominantly on the processing of DNA repair intermediates, because strand breaks in plasmid DNA were not induced by ethylation before the addition of nuclear proteins. Although the initial level of a specific alkylation product (O6-ethylguanine) in nuclear DNA was uniform, different dose-response curves were obtained for the comet size in individual cell samples immediately after exposure, with small intercellular variation. The individual kinetics of DNA repair varied significantly between specimens derived from both healthy individuals and CLL patients; for the DNA repair half-time (t1/2), large difference was found. Pretreatment of cells with methoxyamine as a DNA repair modifier blocking the base excision repair pathway revealed a quite similar extent of base excision repair-independent DNA incision in almost all normal lymphocyte samples. In contrast, this portion varied relatively and absolutely to a great extent among individual samples of CLL lymphocytes, suggesting a loss of stringent control of DNA repair processes in these cells. The comet assay can thus be used to gain information about interindividual variation in the efficiency of different DNA repair processes in small samples of normal cells and their malignant counterparts.


Subject(s)
DNA Damage , DNA Repair , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Carcinogens , DNA/drug effects , Electrophoresis/methods , Ethylnitrosourea , Humans , Hydroxylamines/pharmacology , Lymphocytes , Xeroderma Pigmentosum/genetics
12.
Fundam Appl Toxicol ; 35(1): 131-7, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9024680

ABSTRACT

Hydrazine belongs to a group of compounds for which there is evidence that the in vivo genotoxic effects become manifest only upon exposure to toxic dose levels. The present study was performed to investigate whether this phenomenon is also reflected in the pattern of DNA methylation. The induction of N7- and O6-methylguanine (MeGua) was studied in liver DNA of rats, 16 hr after treatment with various doses of hydrazine. After DNA isolation, the presence of N7-MeGua in DNA was assessed with an immunochemical method and with a physicochemical technique (HPLC with electrochemical detection). Application of these two methods resulted in almost identical patterns of dose-dependent induction of guanine N7-methylation in rats dosed orally with 0.1 to 10 mg hydrazine per kilogram of body weight, increasing from 1.1-1.3 to 39-45 N7-MeGua per 10(6) nucleotides. At lower dosages a constant adduct level was observed, equivalent to that in untreated rats (background level). The O6-MeGua level was analyzed by a combination of HPLC separation and competitive radioimmunoassay. A background level was observed for untreated rats and no increase was visible up to the 0.2 mg/kg dose group. After hydrazine doses from 0.2 to 10 mg/kg, O6-MeGua increased from 0.29 to 134 per 10(9) nucleotides. These data show that even at dosages below the maximum tolerated dose (0.6 mg/kg/day), for which carcinogenic effects have not been described, DNA adducts are formed. A comparison is made of the data obtained in this study with models that describe the mechanism of hydrazine-induced DNA methylation.


Subject(s)
DNA Adducts/biosynthesis , Guanine/analogs & derivatives , Guanine/metabolism , Hydrazines/toxicity , Liver/metabolism , Animals , Chromatography, High Pressure Liquid , Electrochemistry/methods , Guanine/analysis , Hydrazines/administration & dosage , Immunochemistry , Liver/chemistry , Liver/drug effects , Rats , Rats, Wistar
13.
Clin Cancer Res ; 3(11): 2055-61, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9815597

ABSTRACT

The time course of the formation and persistence of repair-induced DNA lesions such as single-strand breaks (SSBs) were determined in isolated lymphocytes derived from 32 patients with chronic lymphocytic leukemia (CLL) using the single-cell gel electrophoresis (SCGE, "comet") assay. After pulse-exposure to N-ethyl-N-nitrosourea (EtNU), the initial amount of SSBs (t0 SCGE values) and the time periods required to reduce DNA damage by 50% (t50% SCGE values) were determined in nuclear DNA of individual cells. The t0 SCGE and t50% SCGE values varied interindividually between CLL specimens by factors of 16.6 and 8.2, respectively. Regarding cell-to-cell variation, no major subpopulations with significantly different DNA repair capacities were observed in cell specimens from a given patient. In addition, a monoclonal antibody-based immunocytological assay was used to determine the elimination kinetics for the cytotoxic alkylation product O6-ethylguanine from nuclear DNA. A strong correlation was observed between the relative times for SSB repair and the elimination of O6-ethylguanine from nuclear DNA. Because SCGE and immunocytological assay measure different steps of DNA repair, this observation suggests coordinated regulation of the respective repair pathways. With regard to chemosensitivity profiles, a "fast" repair phenotype corresponded to enhanced in vitro resistance to EtNU, 1,3-bis(2-chloroethyl)-1-nitrosourea, or chlorambucil. Accelerated SSB repair and pronounced in vitro resistance to chlorambucil, 1,3-bis(2-chloroethyl)-1-nitrosourea, and EtNU were found in lymphocytes from CLL patients nonresponsive to chemotherapy with alkylating agents. Distinct DNA repair processes thus mediate resistance to alkylating agents in CLL lymphocytes.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Carmustine/toxicity , Chlorambucil/toxicity , DNA Damage , DNA Repair , Drug Resistance, Neoplasm , Ethylnitrosourea/toxicity , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Lymphocytes/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Nucleus/drug effects , Cyclophosphamide/administration & dosage , DNA, Neoplasm/blood , DNA, Neoplasm/chemistry , Guanine/analogs & derivatives , Guanine/analysis , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphocytes/pathology , Prednisone/administration & dosage , Vincristine/administration & dosage
14.
Mutat Res ; 385(3): 205-11, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9506889

ABSTRACT

The cellular parameters which modulate trans germ-line carcinogenesis by DNA-reactive agents have not yet been studied in detail. Therefore, we have measured in this study the formation and repair kinetics of the miscoding alkylation product O6-ethylguanine (O6-EtGua) in nuclear DNA of spermatogonial cells of the Syrian golden hamster (SGH) after exposure to either of two potent N-nitroso carcinogens, ethylnitrosourea (ENU) or diethylnitrosamine (DEN). Both compounds, the spontaneously decomposing ENU, and DEN, which has to be converted by cellular enzymes to the reactive ethyl diazonium ion, induce the same pattern of alkylation products in nuclear DNA. Adduct analyses were performed at the single-cell level by using a quantitative immunocytological assay and anti-(O6-EtGua) monoclonal antibodies. 1.5 h after intraperitoneal application of ENU (100 microg/g body weight) O6-EtGua levels in the nuclear DNA of spermatogonia were similar to those in other cell types of the same hamster. About 30% of the initially formed DNA adducts were still persistent in spermatogonial cells even 4 days after ENU exposure. The presence of O6-EtGua in DNA after exposure to DEN (100 microg/g body weight) implies the capability of hamster spermatogonial cells to convert nitrosamines into DNA-alkylating metabolites. This capability of male germ cells in combination with their limited repair capacity for a critical DNA adduct and their high rate of proliferation may be considered as a major risk factor for genetic effects including carcinogenesis in subsequent generation(s).


Subject(s)
Carcinogens/pharmacology , DNA/metabolism , Diethylnitrosamine/pharmacology , Ethylnitrosourea/pharmacology , Guanine/analogs & derivatives , Animals , Antibodies, Monoclonal/immunology , Cricetinae , DNA Adducts/analysis , DNA Adducts/immunology , DNA Adducts/metabolism , DNA Damage/drug effects , DNA Repair/drug effects , Fluorescent Antibody Technique, Indirect , Guanine/immunology , Guanine/metabolism , Male , Mesocricetus , Spermatogonia/drug effects , Spermatogonia/metabolism
15.
Cancer Res ; 56(24): 5615-9, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8971165

ABSTRACT

Normal and malignant cells of the oligodendrocyte lineage show increased sensitivity to alkylating agents compared to astrocytes. One of the most mutagenic DNA lesions formed following exposure to alkylating agents is O6-alkylguanine. To determine whether the increased sensitivity to nitrosoureas seen in oligodendrocytes is due to decreased repair capacity for O6-alkylguanine, removal of this lesion from DNA was assessed in primary cultures of rat oligodendrocytes, astrocytes, and microglia. Glial cells were exposed to 1 mM N-methyl-N-nitrosourea for 1 h and allowed 8 or 24 h for repair. Repair was evaluated using an immunoslot blot technique and a monoclonal antibody which recognizes O6-methylguanine (O6MeGua). Astrocytes removed O6MeGua more efficiently (approximately 80% in 24 h) than either oligodendrocytes (approximately 20%) or microglia (approximately 4%). Determination of O6-alkylguanine-DNA-alkyltransferase (AT) activity revealed that astrocytes contain 0.4 pmol/mg protein, which is average by comparison to other cell types. Both oligodendrocytes and microglia exhibited very low levels of AT (oligodendrocytes, 0.08; microglia, 0.01 pmol/mg protein). These data are the first to show that within different populations of glial cells, O6MeGua adduct removal is substantially reduced in both oligodendrocytes and microglia. Rapid removal of O6MeGua in astrocytes coupled with persistence of this mutagenic lesion in oligodendrocytes following exposure of the developing central nervous system to nitrosoureas could contribute to the observed formation of oligodendrogliomas. Inefficient removal of O6MeGua in oligodendrogliomas might also account for their response to chemotherapeutic regimens involving alkylating agents such as procarbazine, lomustine, and carmustine. The lack of repair of O6MeGua in microglia suggests that primary lymphomas of the central nervous system might be sensitive to treatment with alkylating drugs whose toxicity depends on repair of this adduct.


Subject(s)
Astrocytes/metabolism , DNA Repair , Guanine/analogs & derivatives , Microglia/metabolism , Oligodendroglia/metabolism , Alkylating Agents/toxicity , Animals , Astrocytes/drug effects , DNA Damage , Guanine/metabolism , Methylnitrosourea/toxicity , Methyltransferases/metabolism , Microglia/drug effects , O(6)-Methylguanine-DNA Methyltransferase , Oligodendroglia/drug effects , Rats , Time Factors
17.
Carcinogenesis ; 17(11): 2449-54, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8968062

ABSTRACT

LacZ transgenic mice are suitable for short-term mutagenicity studies in vivo. Mutagenicity in these mice is determined in the lacZ transgene. Since the lacZ gene is of bacterial origin the question has been raised whether DNA-adduct formation and repair in the transgene are comparable to those in total genomic DNA. Mice were treated with N-ethyl-N-nitrosourea (ENU) and killed at several time points following treatment. Some mice were pretreated with O6-benzylguanine to inactivate the repair protein O6-alkylguanine-DNA alkyltransferase (AGT). O6-ethylguanine (O6-EtG) was determined in lacZ in liver and brain by means of a monoclonal antibody-based immunoaffinity assay. In addition, O6-EtG and N7-ethylguanine (N7-EtG) were assayed in total genomic DNA of liver and brain with an immunoslotblot procedure. In liver, the initial O6-EtG level in total genomic DNA was 1.6 times that in lacZ. The extent of repair of O6-EtG during the first 1.5 h after treatment was 2.1 times that in lacZ. At later time points, O6-EtG repair was the same. N7-EtG repair in genomic DNA was evident. In contrast to the liver, little repair of O6-EtG in total genomic and lacZ DNA occurred in the brain while N7-EtG was repaired. No initial difference in O6-EtG levels were found in lacZ and genomic brain DNA. These findings indicate that in the liver, total genomic DNA is more accessible than lacZ to ENU and/or the AGT protein, during the first 1.5 h following treatment. Because the difference in O6-EtG levels in the transgene and genomic DNA in the liver is restricted to the first 1.5 h after treatment, while the fixation of mutations occurs at later time points, O6-EtG-induced mutagenesis most likely is also very similar in both types of DNA.


Subject(s)
Brain/drug effects , DNA/drug effects , DNA/genetics , Ethylnitrosourea/toxicity , Guanine/analogs & derivatives , Lac Operon , Liver/drug effects , Mutagens/toxicity , Animals , Bacteriophage lambda/genetics , Brain/metabolism , Brain/physiology , DNA/metabolism , DNA Repair , Female , Genome , Guanine/analysis , Guanine/biosynthesis , Guanine/metabolism , Immunoblotting , Liver/metabolism , Liver/physiology , Mice , Mice, Transgenic , Mutagenicity Tests , Polymerase Chain Reaction , Sensitivity and Specificity
18.
Mutat Res ; 353(1-2): 177-218, 1996 Jun 12.
Article in English | MEDLINE | ID: mdl-8692191

ABSTRACT

Previous studies on structure-activity relationships (SARs) between types of DNA modifications and tumour incidence revealed linear positive relationships between the log TD50 estimates and s-values for a series of mostly monofunctional alkylating agents. The overall objective of this STEP project was to further elucidate the mechanistic principles underlying these correlations, because detailed knowledge on mechanisms underlying the formation of genotoxic damage is an absolute necessity for establishing guidance values for exposures to genotoxic agents. The analysis included: (1) the re-calculation and further extension of TD50 values in mmol/kg body weight for chemicals carcinogenic in rodents. This part further included the checking up data for Swain-Scott s-values and the use of the covalent binding index (CBI); (2) the elaboration of genetic toxicity including an analysis of induced mutation spectra in specific genes at the DNA level, i.e., the vermilion gene of Drosophila, a plasmid system (pX2 assay) and the HPRT gene in cultured mammalian cells (CHO-9); and (3) the measurement of specific DNA alkylation adducts in animal models (mouse, rat, hamster) and mammalian cells in culture. The analysis of mechanisms controlling the expression of mammalian DNA repair genes (alkyltransferases, glycosylases) as a function of the cell type, differentiation stage, and cellular microenvironment in mammalian cells. The 3 classes of genotoxic carcinogens selected for the project were: (1) chemicals forming monoalkyl adducts upon interaction with DNA; (2) genotoxins capable of forming DNA etheno-adducts; and (3) N-substituted aryl compounds forming covalent adducts at the C8 position of guanine in DNA. In general, clear SARs and AARs (activity-activity relationships) between physiochemical parameters (s-values, O6/N7-alkylguanine ratios, CBI), carcinogenic potency in rodents and several descriptors of genotoxic activity in germ cells (mouse, Drosophila) became apparent when the following descriptors were used: TD50 estimates (lifetime doses expressed in mg/kg b.wt. or mmol/kg b.wt.) from cancer bioassays in rodents; the degree of germ-cell specificity, i.e., the ability of a genotoxic agent to induce mutations in practically all cell stages of the male germ-cell cycle of Drosophila (this project) and the mouse (literature search), as opposed to a more specific response in postmeiotic stages of both species; the Mexr-/Mexr+ hypermutability ratio, determined in a repair assay utilizing Drosophila germ cells; mutation spectra induced at single loci (the 7 loci used in the specific-locus test of the mouse (published data), and the vermilion gene of Drosophila); and doubling doses (DD) in mg/kg (mmol/kg) for specific locus test results on mice. By and large, the TD50 values, the inverse of which can be considered as measures of carcinogenic potency, were shown to be predictable from knowledge of the in vivo doses associated with the absorbed amounts of the investigated alkylators and with the second-order constant, kc, reaction at a critical nucleophilic strength, nc. For alkylating agents kc can be expressed as the second-order rate constant for hydrolysis, kH2O, and the substrate constant s:kH2OTD50 is a function of a certain accumulated degree of alkylation, here given as the (average) daily increment, ac, for 2 years exposure of the rodents. The TD*50 in mmol/kg x day) could then be written: [formula: see text] This expression would be valid for monofunctional alkylators provided the reactive species are uncharged. This is the case for most SN2 reagents. Although it appears possible to predict carcinogenic potency from measured in vivo doses and from detailed knowledge of reaction-kinetic parameter values, it is at present not possible to quantify the uncertainty of such predictions. One main reason for this is the complication due to uneven distribution in the body, with effects on the dose in target tissues. The estimation can be impro


Subject(s)
Carcinogens/toxicity , DNA Damage , DNA Repair , Mutagens/toxicity , Alkylating Agents/toxicity , Animals , Cricetinae , Humans , Male , Mice , Rats , Structure-Activity Relationship
19.
Exp Toxicol Pathol ; 47(6): 443-5, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8871082

ABSTRACT

17-Week-old Syrian golden hamsters received a single intraperitoneal injection of diethylnitrosamine (DEN) at a dose of 100 mg/kg body weight. The DNA alkylation product O6-ethylguanine was formed in the spermatogonia. The data demonstrate that DEN can pass the blood-testis barrier and be metabolized in the spermatogonia to yield an alkylating derivative or that externally activated metabolites themselves can pass the barrier.


Subject(s)
DNA Adducts/metabolism , DNA/drug effects , Diethylnitrosamine/toxicity , Guanine/analogs & derivatives , Spermatogonia/drug effects , Spermatogonia/metabolism , Animals , Biotransformation , Cricetinae , DNA/metabolism , Diethylnitrosamine/administration & dosage , Guanine/biosynthesis , Injections, Intraperitoneal , Male , Mesocricetus
20.
Cancer Res ; 54(16): 4524-31, 1994 Aug 15.
Article in English | MEDLINE | ID: mdl-8044803

ABSTRACT

The elimination kinetics of the alkylation product O6-ethylguanine (O6eGua) from nuclear DNA were determined in individual lymphocytes or blast cells isolated from 27 patients with chronic lymphatic leukemia (CLL) and 26 patients with de novo acute myeloid leukemia (AML). A monoclonal antibody-based immunocytological assay was used for quantification of O6eGua in DNA of individual cells after pulse exposure of cells to N-ethyl-N-nitrosourea (EtNU). In cell specimens from a given patient, no major subpopulations with significantly different capacities for repair of O6eGua were observed. The time required to remove 50% of induced O6eGua residues varied interindividually between 0.5 and 8.4 h in CLL lymphocytes and between 0.8 and 6.3 h in leukemic blast cells. An inverse relationship was found between the rate of removal of O6eGua from DNA and the chemosensitivity of cells to EtNU, 1,3-bis(2-chloroethyl)-1-nitrosourea or mafosfamide in vitro. High rates of O6eGua repair and pronounced resistance to mafosfamide, 1,3-bis(2-chloroethyl)-1-nitrosourea, and EtNU in vitro were found in samples from 8 CLL patients nonresponsive to chemotherapy with alkylating agents. In AML patients treated with anthracyclines and 1-beta-D-arabinofuranosylcytosine, no relation was found between DNA repair capacity and treatment outcome. However, increased P-glycoprotein expression was observed between specimens derived from AML patients who had failed to reach complete remission (n = 12) after chemotherapy versus responsive patients (n = 14). DNA repair rate was not related to chemosensitivity to Adriamycin and 1-beta-D-arabinofuranosylcytosine in vitro, nor were cellular glutathione content, glutathione S-transferases activity, or P-glycoprotein expression.


Subject(s)
DNA Repair , Guanine/analogs & derivatives , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Myeloid/genetics , Acute Disease , Carmustine/pharmacology , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/pharmacology , Cytarabine/pharmacology , Doxorubicin/pharmacology , Drug Resistance , Ethylnitrosourea/pharmacology , Glutathione/analysis , Guanine/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/pathology , Lymphocytes/chemistry , Lymphocytes/drug effects , Lymphocytes/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...