Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters










Publication year range
1.
Pharmacol Rep ; 70(5): 972-980, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30107347

ABSTRACT

BACKGROUND: The compound 9-(2'-hydroxyethylamino)-4-methyl-1-nitroacridine (C-1748), the promising antitumor agent developed in our laboratory was determined to undergo phase I metabolic pathways. The present studies aimed to know its biotransformation with phase II enzymes - UDP-glucuronosyltransferases (UGTs) and its potential to be engaged in drug-drug interactions arising from the modulation of UGT activity. METHODS: UGT-mediated transformations with rat liver (RLM), human liver (HLM), and human intestine (HIM) microsomes and with 10 recombinant human isoenzymes were investigated. Studies on the ability of C-1748 to inhibit UGT were performed with HLM, HT29 colorectal cancer cell homogenate and the selected recombinant UGT isoenzymes. The reactions were monitored using HPLC-UV/Vis method and the C-1748 metabolite structure was determined with ESI-TOF-MS/MS analysis. RESULTS: Pseudo-molecular ion (m/z 474.1554) and the experiment with ß-glucuronidase indicated that O-glucuronide of C-1748 was formed in the presence of microsomal fractions. This reaction was selectively catalyzed by UGT2B7 and 2B17. High inhibitory effect of C-1748 was shown towards isoenzyme UGT1A9 (IC50=39.7µM) and significant but low inhibitory potential was expressed in HT29 cell homogenate (IC50=84.5µM). The mixed-type inhibition mechanism (Ki=17.0µM;Ki'=81.0µM), induced by C-1748 was observed for recombinant UGT1A9 glucuronidation, whereas HT29 cell homogenate resulted in noncompetitive inhibition (Ki=94.6µM). CONCLUSIONS: The observed UGT-mediated metabolism of C-1748 and its ability to inhibit UGT activity should be considered as the potency for drug resistance and drug-drug interactions in the prospective multidrug therapy.


Subject(s)
Glucuronosyltransferase/metabolism , Nitracrine/analogs & derivatives , Animals , Biotransformation , Cell Line, Tumor , Glucuronosyltransferase/antagonists & inhibitors , Humans , Microsomes, Liver/enzymology , Nitracrine/pharmacokinetics , Nitracrine/pharmacology , Rats , UDP-Glucuronosyltransferase 1A9
2.
Biochem Pharmacol ; 142: 21-38, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28645477

ABSTRACT

Drug resistance is one of the major causes of pancreatic cancer treatment failure. Thus, it is still imperative to develop new active compounds and novel approach to improve drug efficacy. Here we present 9-amino-1-nitroacridine antitumor agent, C-1748, developed in our laboratory, as a candidate for pancreatic cancer treatment. We examined (i) the cellular response of pancreatic cancer cell lines: Panc-1, MiaPaCa-2, BxPC-3 and AsPC-1, differing in expression levels of commonly mutated genes for this cancer type, to C-1748 treatment and (ii) the role of P450 3A4 isoenzyme and cytochrome P450 reductase (CPR) in the modulation of this response. C-1748 exhibited the highest cytotoxic activity against MiaPaCa-2, while AsPC-1 cells were the most resistant (IC50: 0.015, 0.075µM, respectively). A considerable amount of apoptosis was detected in Panc-1 and MiaPaCa-2 cells but only limited apoptosis was observed in AsPC-1 and BxPC-3 cells as indicated by morphological changes and biochemical markers. Furthermore, only AsPC-1 cells underwent senescence. Since AsPC-1 cells were the most resistant to C-1748 as evidenced by the lowest P450 3A4 and CPR protein levels, this cell line was subjected to transient transfection either with P450 3A4 or CPR gene. The overexpression of P450 3A4 or CPR changed the pro-apoptotic activity of C-1748 and sensitized AsPC-1 cells to this drug compared to wild-type cells. However, metabolism was changed significantly only for CPR overexpressing cells. In conclusion, the antitumor effectiveness of C-1748 would be improved by multi-drug therapy with chemotherapeutics, that are able to induce P450 3A4 and/or CPR gene expression.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cytochrome P-450 CYP3A/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Nitracrine/analogs & derivatives , Pancreatic Neoplasms , Cell Culture Techniques , Cell Line, Tumor , Cell Survival/drug effects , Cytochrome P-450 CYP3A/genetics , Drug Resistance, Neoplasm/genetics , Flow Cytometry , Gene Expression , Humans , Membrane Potential, Mitochondrial/drug effects , NADPH-Ferrihemoprotein Reductase/genetics , Nitracrine/pharmacology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Transfection , Up-Regulation
3.
Biochem Pharmacol ; 86(2): 231-41, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23688499

ABSTRACT

Induction of proteins involved in drug metabolism and in drug delivery has a significant impact on drug-drug interactions and on the final therapeutic effects. Two antitumor acridine derivatives selected for present studies, C-1748 (9-(2'-hydroxyethylamino)-4-methyl-1-nitroacridine) and C-1305 (5-dimethylaminopropylamino-8-hydroxy-triazoloacridinone), expressed high and low susceptibility to metabolic transformations with liver microsomes, respectively. In the current study, we examined the influence of these compounds on cytochrome P450 3A4 (CYP3A4) and 2C9 (CYP2C9) enzymatic activity and gene expression in HepG2 tumor cells. Luminescence and HPLC examination, real-time RT-PCR and western blot analyses along with transfection of pregnane X receptor (PXR) siRNA and CYP3A4 reporter gene assays were applied. We found that both compounds strongly induced CYP3A4 and CYP2C9 activity and expression as well as expression of UGT1A1 and MDR1 in a concentration- and time-dependent manner. C-1748-mediated CYP3A4 and CYP2C9 mRNA induction equal to rifampicin occurred at extremely low concentrations (0.001 and 0.01µM), whereas 10µM C-1305 induced three-times higher CYP3A4 and CYP2C9 mRNA levels than rifampicin did. CYP3A4 and CYP2C9 expressions were shown to be PXR-dependent; however, neither compound influenced PXR expression. Thus, the observed drug-mediated induction of isoenzymes occurs on a PXR-mediated regulatory level. Furthermore, C-1748 and C-1305 were demonstrated to be selective PXR agonists. These effects are hypoxia-inhibited only in the case of C-1748, which is sensitive to P450 metabolism. In summary, PXR was found to be a new target of the studied compounds. Thus, possible combinations of these compounds with other therapeutics might lead to the PXR-dependent enzyme-mediated drug-drug interactions.


Subject(s)
Acridines/pharmacology , Antineoplastic Agents/pharmacology , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP3A/metabolism , Hypoxia/metabolism , Nitracrine/analogs & derivatives , Receptors, Steroid/physiology , Triazoles/pharmacology , Up-Regulation , Base Sequence , Blotting, Western , Cytochrome P-450 CYP2C9 , DNA Primers , Hep G2 Cells , Humans , Hypoxia/enzymology , Nitracrine/pharmacology , Pregnane X Receptor , Real-Time Polymerase Chain Reaction
4.
Biochem Pharmacol ; 84(1): 30-42, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22484277

ABSTRACT

The narrow "therapeutic window" of anti-tumour therapy may be the result of drug metabolism leading to the activation or detoxification of antitumour agents. The aim of this work is to examine (i) whether the diminished toxicity of a potent antitumour drug, C-1748, 9-(2'-hydroxyethylamino)-4-methyl-1-nitroacridine, compared with its 4-demethyl analogue, C-857, results from the differences between the metabolic pathways for the two compounds and (ii) the impact of reducing and/or hypoxic conditions on studied metabolism. We investigated the metabolites of C-1748 and C-857 formed in rat and human liver microsomes, with human P450 reductase (POR) and in HepG2 cells under normoxia and hypoxia. The elimination rate of C-1748 from POR knockout mice (HRN) was also evaluated. Three products, 1-amino-9-hydroxyethylaminoacridine, 1-aminoacridinone and a compound with an additional 6-membered ring, were identified for C-1748 and C-857 in all studied metabolic systems. The new metabolite was found in HepG2 cells. We showed that metabolic rate and the reactivity of metabolites of C-1748 were considerably lower than those of C-857, in all investigated metabolic models. Compared with metabolism under normoxia, cellular metabolism under hypoxia led to higher levels of 1-aminoacridine and aza-acridine derivatives of both compounds and of the 6-membered ring metabolite of C-1748. In conclusion, the crucial role of hypoxic conditions and the direct involvement of POR in the metabolism of both compounds were demonstrated. Compared with C-857, the low reactivity of C-1748 and the stability of its metabolites are postulated to contribute significantly to the diminished toxicity of this compound observed in animals.


Subject(s)
Aminoacridines/metabolism , Aminoacridines/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Nitracrine/analogs & derivatives , Aminoacridines/chemistry , Animals , Antineoplastic Agents/chemistry , Biotransformation , Cell Culture Techniques , Cell Hypoxia/physiology , Chromatography, High Pressure Liquid , Hep G2 Cells , Humans , Male , Mice , Mice, Knockout , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Molecular Structure , NADPH-Ferrihemoprotein Reductase/genetics , NADPH-Ferrihemoprotein Reductase/physiology , Nitracrine/chemistry , Nitracrine/metabolism , Nitracrine/pharmacology , Rats , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
5.
Biochem Pharmacol ; 79(9): 1231-41, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20026080

ABSTRACT

C-1748 is a DNA-binding agent with potent antitumor activity, especially towards prostate and colon carcinoma xenografts in mice. Here, we elucidated the nature of cellular response of human colon carcinoma HCT8 and HT29 cells to C-1748 treatment, at biologically relevant concentrations (EC(90) and their multiplicity). Cell cycle analysis showed gradual increase in HCT8 cells with sub-G1 DNA content (25% after 72h) considered as apoptotic. Hypodiploid cell population increased up to 60% upon treatment with 4x EC(90) concentration of the drug. Compared with HCT8 cells, the fraction of sub-G1 HT29 cells did not exceed 14%, even following 4-fold dose escalation. Morphological changes and biochemical markers such as: phosphatydylserine externalization, apoptotic DNA breaks, mitochondrial dysfunction and caspase activation confirmed the presence of considerable amount of apoptotic HCT8 cells but only a low amount of apoptotic HT29 cells. Next, we demonstrated that HCT8 cells surviving after exposure to C-1748 were in the state of senescence, based on altered cell morphology and expression of a pH 6-dependent beta-galactosidase. On the contrary, no beta-galactosidase staining was observed in HT29 cells after C-1748 treatment. Moreover, prolonged drug incubation (up to 168h) resulted in massive detachment of cells from culture plates, which together with Annexin V/PI results, indicated that necrosis was the main response of HT29 cells to C-1748 treatment. We also determined the ability of C-1748 to induce reactive oxygen species (ROS) in colon cancer cells and demonstrated, that generation of ROS was not essential for C-1748-induced apoptosis and cytotoxic activity of this drug.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Necrosis/chemically induced , Nitracrine/analogs & derivatives , Antineoplastic Agents/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Molecular Structure , Nitracrine/chemistry , Nitracrine/pharmacology
6.
Cancer Biol Ther ; 6(10): 1632-7, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17921700

ABSTRACT

Chemotherapy in prostate cancer (CaP) even as an adjunct has not been a success. In this communication, we report the pre-clinical efficacy of a nitroacridine derivative, C-1748 (9[2'-hydroxyethylamino]-4-methyl-1-nitroacridine) in CaP cell culture and human xenograft animal models. C-1748, a DNA intercalating agent has been derived from its precursor C-857 that was a potent anti-cancer drug, but failed clinical development due to "high" systemic toxicities. Chemical modifications such as the introduction of a "methyl" group imparted novel properties, the most interesting of which is the difference in the IC(50) values between LnCaP (22.5 nM), a CaP cell line and HL-60, a leukemia cell line (>100 nM). Using gammaH2AX as an intervention marker of DNA double strand breaks, we concluded that C-1748 is more efficacious in CaP cells than in HL-60 cells. In hormone dependent cells, the androgen receptor (AR) was identified as an additional target of C-1748. In xenograft studies, administration of C-1748 intra-peritoneally inhibited tumor growth by 80-90% with minimal toxicity. These studies identify C-1748 as a novel acridine drug that has a high therapeutic index and low cytotoxicity on myelocytic cells with potential for clinical development.


Subject(s)
Antineoplastic Agents/therapeutic use , Intercalating Agents/therapeutic use , Nitracrine/analogs & derivatives , Prostatic Neoplasms/drug therapy , Androgen Receptor Antagonists , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Down-Regulation , Drug Evaluation, Preclinical , Histones/metabolism , Humans , Intercalating Agents/pharmacology , Male , Mice , Mice, Inbred BALB C , Nitracrine/pharmacology , Nitracrine/therapeutic use , Prostatic Neoplasms/metabolism , Xenograft Model Antitumor Assays
7.
Neuroreport ; 15(16): 2495-9, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15538182

ABSTRACT

Brain-derived neurotrophic factor (BDNF) modulates glutamate receptors of the NMDA type in many areas of the brain. We assessed whether BDNF exerts an effect on NMDA receptor properties in retinal ganglion cells during early postnatal development. Electrophysiological responses to the glutamate agonist NMDA (500 microM-2 mM) in retinal slices of wildtype and BDNF deficient mice (bdnf-/-) were recorded using the whole-cell patch-clamp technique. Retinal ganglion cells of bdnf-/- mice displayed significantly smaller NMDA currents than those of age-matched wildtype mice. Remarkably, NMDA receptor activity was restored by incubating retinal slices of bdnf-/- mice in BDNF (50 ng/ml) for 1-3 h. We suggest that BDNF plays a role in the activation of functional NMDA receptors in early ganglion cell development.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Nitracrine/analogs & derivatives , Receptors, N-Methyl-D-Aspartate/physiology , Retina/cytology , Retinal Ganglion Cells/metabolism , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Drug Interactions , Electric Capacitance , Heterozygote , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Knockout , N-Methylaspartate/pharmacology , Nitracrine/pharmacology , Patch-Clamp Techniques/methods , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Retina/growth & development
8.
J Photochem Photobiol B ; 75(1-2): 27-32, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15246347

ABSTRACT

The cytotoxicity of two nitroheterocyclic compounds (NHCD), Nitracrine, 1-nitro-9(3-3-dimethylaminopropylamino) acridine and Quinifuryl, 2-(5'-nitro-2'-furanyl) ethenyl-4-[N-[4-(N,N-diethylamino)-1'-methylbutyl] carbamoyl] quinoline, towards two lines of leukaemic cells and a line of non-transformed cells, was measured in comparison, on the dark and under illumination with visible light (350-450 nm). Both drugs showed highly elevated cytotoxicity when illuminated with LC(50) values 7-35 times lower after 1 h illumination compared to 1 h incubation of cells incubation with drug on the dark. Cytotoxicity of Nitracrine toward all cell lines studied exceeded that of Quinifuryl, both on the dark and under illumination, so that approximately 10 times lower concentration of former drug was needed to reach the same toxicity as the latter. General toxic effect was calculated as a direct cell kill and a cell proliferation arrest. The effect >80% for both drugs was achieved after 1 h cell illumination with as low drug concentrations as 0.2 microM for Quinifuryl and 0.02 microM for Nitracrine.


Subject(s)
Antineoplastic Agents/pharmacology , Light , Nitracrine/pharmacology , Quinolines/pharmacology , Animals , Antineoplastic Agents/toxicity , Cell Survival/drug effects , Darkness , Drug Evaluation, Preclinical , Humans , K562 Cells , Leukemia P388 , Mice , NIH 3T3 Cells , Nitracrine/toxicity , Quinolines/toxicity
10.
Apoptosis ; 7(4): 347-59, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12101394

ABSTRACT

Nitracrine (Ledakrin) is an antitumor drug which is activated by cellular enzymes and binds covalently to DNA. Previous studies have shown that covalent binding and crosslinking of DNA is associated with the cytotoxic and antitumor activities of this compound. In this study, cell cycle perturbations, effects on DNA synthesis and the cell death process initiated by Nitracrine were studied in murine leukemia L1210 cells. We show that exposure of L1210 cells to Nitracrine at the IC(99) concentration delayed progression through the S phase and transiently arrested cells in G(2)/M as found by flow cytometry. Higher drug concentration (2 x IC(99)) inhibited cell cycle progression in the S phase and induced rapid cell death. Both studied concentrations of the drug produced different effects on DNA synthesis as determined by bromodeoxyuridine incorporation, with a delay in the S phase progression at EC(99) concentration and irreversible arrest in early S phase at the higher dose (2 x IC(99)). At both concentrations of Nitracrine cell death occurred preferentially in the S phase as revealed by the TUNEL assay. When cells treated with the drug for 4 hours were post-incubated in the presence of 1 mM caffeine this led to rapid cell death and suppression of the G(2) arrest. This was associated with a about 10-fold increase in the cytotoxicity of Nitracrine. Similar effects were observed for another DNA crosslinking agent, cis-platinum, and to a lesser extent, for DNA topoisomerase I inhibitor, camptothecin. Together, our studies show that suppression of G(2) arrest induced by Nitracrine greatly enhances its cytotoxicity toward L1210 cells.


Subject(s)
Apoptosis/drug effects , G2 Phase/drug effects , Nitracrine/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Camptothecin/pharmacology , Cisplatin/pharmacology , DNA Fragmentation , Dose-Response Relationship, Drug , Flow Cytometry , HeLa Cells , Humans , In Situ Nick-End Labeling , S Phase/drug effects
11.
Biochem Pharmacol ; 60(7): 969-78, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-10974206

ABSTRACT

Some N-oxide derivatives of DNA intercalators are bioreductive prodrugs that are selectively toxic under hypoxic conditions. The hypoxic selectivity is considered to result from an increase in DNA binding affinity when the N-oxide moiety is reduced. This study investigated whether differences in DNA binding affinity between N-oxides and their corresponding amines, measured by equilibrium dialysis, can account for the hypoxic cytotoxicity ratios (HCR) of tertiary amine N-oxide (-tO) and aromatic N-oxide (-aO) derivatives of the 1-nitroacridine nitracrine (NC) and its non-nitro analogue 9-[3-(N,N-dimethylamino)propylamino]acridine (DAPA). Cytotoxicity was measured in aerobic and hypoxic suspensions of Chinese hamster ovary (CHO) AA8 cells by clonogenic assay. HCR were much greater for NC-tO (820-fold) than for NC (5-fold) or NC-aO (4-fold), whereas DAPA and its N-oxides lacked hypoxic selectivity (1-fold). DNA binding measurements demonstrated that binding affinity is lowered more by aromatic than tertiary amine (side-chain) N-oxides, an observation that does not correlate with HCR. Compounds were accumulated in cells to high concentrations (C(i)/C(e) approximately 10-200), with the exception of the tertiary amine N-oxides, for which the ratio of intracellular to extracellular drug was less than unity. For NC-tO this probably resulted from low pK(a) values for both the acridine chromophore and the side-chain, whereas DAPA-tO may be too hydrophilic for efficient membrane permeation. Bioreductive drug metabolism, assessed by HPLC, was faster for the NC than the DAPA N-oxides. The high HCR of NC-tO relative to NC-aO is ascribed to the rapid and selective reduction of its N-oxide moiety, followed by activation of the NC intermediate by O(2)-sensitive reduction of its 1-nitro group to the corresponding 1-amine. The metabolism studies suggest that unmasking of DNA binding affinity by reductive removal of the N-oxide moiety, although not the only determinant, is important and needs to occur before nitroreduction for optimal effect.


Subject(s)
DNA/drug effects , Intercalating Agents/pharmacology , Nitracrine/analogs & derivatives , Nitracrine/pharmacology , Animals , Biological Transport , CHO Cells , Cell Survival/drug effects , Cells, Cultured , Cricetinae , DNA/metabolism , Intercalating Agents/chemistry , Intercalating Agents/metabolism , Nitracrine/metabolism
12.
Neoplasma ; 46(1): 50-3, 1999.
Article in English | MEDLINE | ID: mdl-10355534

ABSTRACT

The anticancer drug, nitracrine, a 1-nitro-9-aminoalkyl derivative of acridine exhibits potent cytotoxic effects which are due to its metabolic activation, followed by covalent binding to macromolecules--DNA being the target for the drug. The renaturable fraction of DNA from L-1210 cells pretreated with nitracrine is assayed by means of ethidium bromide fluorescence assay and chromatography on hydroxyapatite column. The effect of the drug was compared with furocoumarins of different DNA crosslinking potencies. The existence of crosslinks in DNA upon incubation of cells with nitracrine (1-4 microM) have been confirmed with two different methods under the conditions where 8-methoxypsoralen, a classic crosslinking agent induced the renaturation. The DNA preparation isolated from the drug pretreated cells exhibited decreased transcriptional template activity with E. coli DNA-dependent RNA polymerase.


Subject(s)
Antineoplastic Agents/pharmacology , Cross-Linking Reagents/pharmacology , DNA/drug effects , Furocoumarins/pharmacology , Nitracrine/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Animals , Cattle , Cells, Cultured , DNA/analysis , DNA/isolation & purification , Mice
13.
Anticancer Drug Des ; 14(6): 487-97, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10834270

ABSTRACT

The tertiary amine N-oxide (nitracrine-N-oxide, 1b) of the 1-nitroacridine nitracrine is a bis-bioreductive agent showing very high hypoxic selectivity (approximately 1000-fold) against tumour cells in culture, but only modest activity against the hypoxic subfraction of tumours in vivo. Because the hypoxic selectivity of 1b was considered to depend significantly on the rate of enzyme-mediated reduction of the N-oxide group, this paper reports the preparation and evaluation of a series of analogues in which the environment of this group was modified. Three analogues contained more weakly basic N-oxides, while two others had varying degrees of steric bulk around the N-oxide. In all but one case (an aromatic N-oxide), the N-oxides were much less cytotoxic (10- to 300-fold) than the corresponding tertiary amines towards AA8 Chinese hamster cells under aerobic conditions. Both the N-oxides and the corresponding amines were more cytotoxic to an ERCC-1 mutant defective in nucleotide excision repair, indicating that DNA alkylation was the cytotoxic event. However, there was no apparent correlation of these parameters with structure. All of the aliphatic N-oxides, with the exception of the aromatic N-oxide example, showed substantial (70- to 800-fold) hypoxic selectivity against AA8 cells in a clonogenic assay. While the weakly basic derivatives were the least selective, there was no apparent relationship between hypoxic selectivity and the steric environment of the N-oxide. Selectivity for hypoxic cells in culture is shown to depend on the hypoxic selectivity of the corresponding tertiary amine (reflecting O2-inhibitable reduction of the 1-nitro group) and the differential in aerobic toxicity between amine and N-oxide (a measure of the potential toxicity increase achievable by reducing the N-oxide). Four analogues whose structures fairly represented the range of steric and electronic modifications of the N-oxide site were evaluated against the hypoxic subfraction of cells in KHT tumours in vivo, but were inactive. These results suggest that either such modifications do not exert significant effects on N-oxide reduction, or that the rate of such reduction is not a factor limiting the in vivo activity of the parent analogue 1b.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Hypoxia/drug effects , Nitracrine/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Clone Cells , Drug Design , Humans , Indicators and Reagents , Kinetics , Male , Mice , Mice, Inbred C3H , Nitracrine/chemistry , Oxides/chemistry , Oxides/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured
14.
Chem Biol Interact ; 103(2): 131-9, 1997 Feb 28.
Article in English | MEDLINE | ID: mdl-9055871

ABSTRACT

Adducts generated in vitro by the reaction of 1-nitroacridines with poly(dN)s in the presence of dithiothreitol were used to identify a kind of nucleic base involved in the formation of individual adducts. The patterns of chromatographic spots corresponding to modified nucleotides obtained by 32P-post-labelling assay for synthetic homopolymers of four deoxyribonucleotides were compared with the fingerprints detected in the case of calf thymus DNA reacted with 1-nitroacridines under conditions in which the formation of identical DNA adducts as in cellular models was demonstrated in earlier investigations. Both compounds studied (Ledakrin and C-857) turned out to bind covalently only with purine nucleotides. Ledakrin formed with dG four and C-857 five different adducts. All of them were also detected in ctDNA. The incubation with poly(dA) resulted in four Ledakrin-dA species, two of which were found in ctDNA, and in two C-857-dA adducts that were not, however, observed in DNA containing samples. Modification of purines accounted for all adducts observed in ctDNA. For both compounds studied, the level of total binding to poly(dA) was about one order of magnitude lower than to poly(dG) for which it was comparable with the extent of ctDNA modification. This indicates that dG represents a preferential site of covalent binding of 1-nitroacridines to DNA.


Subject(s)
Aminoacridines/metabolism , Aminoacridines/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , DNA Adducts/biosynthesis , Nitracrine/metabolism , Nitracrine/pharmacology , Nucleic Acid Synthesis Inhibitors/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Animals , Cattle , DNA/drug effects , DNA/metabolism , DNA Fingerprinting , Dithiothreitol/pharmacology , Isotope Labeling , Phosphorus Radioisotopes , Poly A/metabolism , Poly G/metabolism , Sulfhydryl Reagents/pharmacology
15.
Chem Biol Interact ; 103(2): 141-51, 1997 Feb 28.
Article in English | MEDLINE | ID: mdl-9055872

ABSTRACT

Using agarose gel electrophoresis we confirmed that Ledakrin is capable of incurring covalent crosslinking in pBR322 plasmid DNA and also in poly(dGdC) in the presence of a simple activating system containing DTT. The identification of adducts resulting from DNA crosslinking was carried out by 32P-post-labelling assay. We assumed that such adduct(s) should be brought about more readily with double-stranded than with single-stranded polynucleotides or nucleotides. Since our earlier experiments had shown that guanine is a major site of covalent binding of 1-nitroacridines, we compared DNA adduct formation by Ledakrin for ctDNA, dG-containing synthetic homopolymers and 3'-pdG. 32P-Post-labelling assay revealed two adduct spots that were enhanced in samples containing double-stranded substrates in which interstrand crosslinking between guanines was possible, namely ctDNA and poly(dGdC).


Subject(s)
Antineoplastic Agents/pharmacology , Cross-Linking Reagents/pharmacology , DNA Adducts/biosynthesis , DNA/drug effects , DNA/metabolism , Intercalating Agents/pharmacology , Nitracrine/pharmacology , Animals , Antineoplastic Agents/metabolism , Cattle , Cross-Linking Reagents/metabolism , DNA, Single-Stranded/drug effects , DNA, Single-Stranded/metabolism , Dithiothreitol/pharmacology , Intercalating Agents/metabolism , Nitracrine/metabolism , Poly C/metabolism , Poly G/metabolism , Sulfhydryl Reagents/metabolism
16.
Br J Cancer Suppl ; 27: S43-7, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8763844

ABSTRACT

Tertiary amine N-oxides of DNA intercalators with alkylamino sidechains are a new class of bioreductive drugs. N-oxidation masks the cationic charge of the amines, forming prodrugs with low DNA binding affinity and low toxicity which can be activated selectively by metabolic reduction under hypoxic conditions. This study compares three intercalator N-oxides (NC-NO, DACA-NO and AQ4N), which, respectively, give nitracrine (NC), DACA and AQ4 on reduction. In aerobic cell culture all three N-oxide were much less toxic than the corresponding amines, and showed large increases in cytotoxicity under hypoxia. The topoisomerase poisons DACA and AQ4 (and their N-oxides) were less active against non-cycling than cycling cells. However, only AQ4N was active against the mouse mammary tumour MDAH-MCa-4. This dialkylaminoanthraquinone-di-N-oxide has activity at least as great as the reference bioreductive drug RB 6145 against this tumour, both with and without radiation and when combined with the tumour blood flow inhibitor 5,6-dimethylxanthenone-4-acetic acid (DMXAA). It is suggested that the high in vivo activity of AQ4N relative to the other topoisomerase-targeted N-oxide, DACA-NO, may be in part due to release in hypoxic cells of an intracalator with sufficiently high DNA binding affinity that it is retained long enough to kill non-cycling cells when they eventually re-enter the cell cycle.


Subject(s)
Acridines/pharmacology , Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , Intercalating Agents/pharmacology , Nitracrine/analogs & derivatives , Prodrugs/pharmacology , Xanthones , Animals , Cell Survival/drug effects , Humans , Mammary Neoplasms, Experimental/drug therapy , Mice , Nitracrine/pharmacology , Oxidation-Reduction , Xanthenes/pharmacology
17.
J Med Chem ; 39(13): 2508-17, 1996 Jun 21.
Article in English | MEDLINE | ID: mdl-8691448

ABSTRACT

A series of nuclear-substituted derivatives of nitracrine N-oxide (2; a bis-bioreductive hypoxia-selective cytotoxin) were prepared and evaluated, seeking analogues of lower nitroacridine reduction potential. Disubstitution with Me or OMe groups at the 4- and 5-positions did not provide analogues with one-electron reduction potentials significantly lower than those of the corresponding monosubstituted derivatives (E(1) ca. -350 mV for both the 4-OMe and 4,5-diOMe compounds). This appears not to be due to a concomitant raising of the acridine pKa but to a lack of direct electronic effect of substituents in the ring not bearing the nitro group. Conversely, placing two OMe groups in the nitro-bearing ring does result in a substantial further lowering of reduction potential (the 2,4-diOMe analogue has an E(1) of -401 mV). The mono- and disubstituted N-oxides have substantially lower cytotoxicities than the parent nitracrine N-oxide 2 but generally retain very high hypoxic selectivity. The OMe-substituted N-oxides all showed greater metabolic stability than 2 in hypoxic AA8 cell cultures, and the 4-OMe compound 6 had improved activity in EMT6 multicellular spheroids suggesting that this metabolic stabilization may allow more efficient diffusion in tumor tissue. The parent compound 2 was selectively toxic to hypoxic cells in KHT tumors in vivo and clearly superior to nitracrine itself (although only at doses which would eventually be lethal to the host). The analogues of lower E(1), including 6, were not superior to 2 in vivo, indicating that metabolic stabilization of the nitro group is not alone sufficient to improve therapeutic utility.


Subject(s)
Acridines/pharmacology , Antineoplastic Agents/pharmacology , Nitracrine/analogs & derivatives , Acridines/chemical synthesis , Acridines/metabolism , Acridines/toxicity , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Division/drug effects , Cell Hypoxia , Cell Line , Cell Survival/drug effects , Cricetinae , Cricetulus , Culture Media , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C3H , Molecular Structure , Nitracrine/chemical synthesis , Nitracrine/chemistry , Nitracrine/metabolism , Nitracrine/pharmacology , Oxidation-Reduction , Tumor Cells, Cultured
18.
Gen Pharmacol ; 26(3): 473-81, 1995 May.
Article in English | MEDLINE | ID: mdl-7789719

ABSTRACT

1. An anticancer drug, nitracrine 1-nitro-9(3'3'-dimethylaminopropylamino)acridine (Ledakrin, C-283) exhibits potent cytostatic effects which can be ascribed to interactions of the drug with DNA. 2. The reduction of the nitro group of nitracrine is one of the activation steps leading to the drug covalent binding to DNA and proteins both in subcellular systems and in the cell. 3. DNA-drug non-covalent interactions and covalent complexes are examined in several model systems and compared with the properties of a number of derivatives with programmed structural changes. 4. DNA-protein crosslinks and interstrand crosslinks are detected in the cells following exposition to the drug. 5. The drug exhibits selective toxicity and radiosensitization effects to hypoxic mammalian cells.


Subject(s)
Antineoplastic Agents/pharmacology , Nitracrine/analogs & derivatives , Nitracrine/pharmacology , Animals , Humans
20.
Int J Radiat Oncol Biol Phys ; 22(4): 693-6, 1992.
Article in English | MEDLINE | ID: mdl-1544839

ABSTRACT

Drugs with two reducible centers, both of which must be metabolized by oxygen-inhibitable processes for full activation ("bis-bioreductive agents"), offer potential for the development of hypoxia-selective cytotoxins with improved oxygen sensitivity. The sidechain N-oxide (1-NCO) of the (mono)bioreductive agent nitracrine (1-NC) has been synthesized and evaluated as a potential example of such an approach. The association constant for reversible DNA binding of 1-NCO was 15-fold lower than that of 1-NC, as measured by equilibrium dialysis in a low ionic strength buffer, indicating that the N-oxide has the potential to act as a less toxic pro-drug of 1-NC. Cell uptake and aerobic cytotoxicity of 1-NCO were much lower than for 1-NC whereas its hypoxic selectivity as a cytotoxin was greatly increased. In stirred suspension cultures of AA8 cells, pure (less than 0.02% 1-NC) 1-NCO was 1000-1500 times more potent under hypoxia than in 20% O2. For 1-NC the corresponding ratio was 10 +/- 1. 1-NCO had greater hypoxic selectivity in this system than misonidazole (ratio 11), RSU 1069 (ratio 25), 8Me-5NQ (ratio 60), or SR 4233 (ratio 80). Studies of 1-NCO metabolism indicate rapid, O2-inhibited reduction to 1-NC. The data are consistent with a two-step bioactivation mechanism, with reduction of the N-oxide generating a DNA intercalator of increased binding affinity, followed by reduction of the nitro group of this DNA-targeted cytotoxin to form reactive cytotoxic metabolites.


Subject(s)
Antineoplastic Agents/pharmacology , Nitracrine/analogs & derivatives , Prodrugs/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Cell Hypoxia , Cell Survival/drug effects , Cricetinae , In Vitro Techniques , Nitracrine/chemical synthesis , Nitracrine/pharmacology , Prodrugs/chemical synthesis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...