Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 21(4A): 2753-60, 2001.
Article in English | MEDLINE | ID: mdl-11724351

ABSTRACT

BACKGROUND: The anthracycline antitumor drug, doxorubicin (DOX), is proposed to catalyze the production of formaldehyde and bond to the formaldehyde at its amino sugar to produce an active metabolite that subsequently crosslinks DNA as part of the cytotoxic mechanism. Doxoform (DOXF), a synthetic formaldehyde conjugate of DOX, exhibits enhanced toxicity to numerous sensitive and resistant cancer cell lines. The aim of this study was to demonstrate that DOXF, at much lower drug levels, retains the apoptosis-inducing characteristics of DOX, consistent with DOXF being a prodrug to the DOX active metabolite. MATERIALS AND METHODS: HeLa S3 and MCF-7 cells were treated with IC50-equivalent concentrations of DOX and DOXF and analyzed for DNA fragmentation and phosphatidylserine externalization, common morphological features of apoptosis. DNA fragmentation was detected by gel electrophoresis and TUNEL assay; phosphatidylserine externalization was detected by annexin V binding. RESULTS: DNA fragmentation and phosphatidylserine externalization were detected in HeLa S3 cells following a 3 h treatment with either 86 nM equiv. DOXF or 1 microM DOX. No apoptotic features were observed for MCF-7 cells following a 3 h treatment with either DOXF (100 nM equiv.) or DOX (1 microM). CONCLUSIONS: DOXF induced cell death in both cell lines at drug levels an order of magnitude lower than DOX. The similar behavior of DOXF and DOX supports the role of formaldehyde in the cytotoxic mechanism of the clinical anthracycline antitumor agents and provides further support for the proposition that DOXF is a prodrug to the DOX active metabolite.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Doxorubicin/pharmacology , Annexin A5/metabolism , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Death/drug effects , DNA Fragmentation , Doxorubicin/pharmacokinetics , Electrophoresis , Fluorescein-5-isothiocyanate/metabolism , Fluorescent Dyes/metabolism , Formaldehyde/pharmacology , HeLa Cells , Humans , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Tumor Cells, Cultured
2.
Anal Chem ; 73(13): 2992-7, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11467545

ABSTRACT

A rapid and highly sensitive method for the detection of formaldehyde utilizing selected ion flow tube-chemical ionization mass spectrometry is reported. Formaldehyde in aqueous biological samples is preconcentrated by distillation and directly analyzed using gas-phase thermal energy proton transfer from H30+; this procedure can be performed in 30 min. The method detection limit for formaldehyde based on seven replicate measurements of reference water samples (2.5 mL) is 80 nM at the 99% confidence level. Detection is linear up to 130 microM. This technique allows the first measurement of natural formaldehyde levels in human cancer cells in vitro. Elevated levels of formaldehyde relative to the reference water are observed for doxorubicin-sensitive cells (MCF-7 breast cancer, K562 leukemia, HeLa S3 cervical cancer) with estimated intracellular formaldehyde concentrations ranging from 1.5 to 4.0 microM, whereas formaldehyde in doxorubicin-resistant MCF-7/Adr breast cancer cells is essentially at reference level. This trend is inverted for prostate cancer cells LNCaP (sensitive) and DU-145 (resistant). Correlation of natural formaldehyde level with doxorubicin cytotoxicity is a function of the expression of enzymes that neutralize oxidative stress and the drug efflux pump, P-170 glycoprotein.


Subject(s)
Formaldehyde/analysis , Mass Spectrometry/methods , Neoplasms/chemistry , Humans , Neoplasms/pathology , Sensitivity and Specificity , Tumor Cells, Cultured
3.
Curr Med Chem ; 8(1): 15-29, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11172689

ABSTRACT

Recent and new results which support a drug-DNA covalent bonding mechanism for cell toxicity of the clinical antitumor drugs, daunorubicin, doxorubicin, and epidoxorubicin, are summarized. The mechanism involves the iron complex of the drugs inducing oxidative stress to yield formaldehyde, which then mediates covalent attachment to G-bases of DNA. At NGC sites the combination of covalent and non-covalent drug interactions serve to virtually crosslink the DNA. Structural data for virtual crosslinks are compared as a function of drug structure. Elucidation of the mechanism led to the synthesis and evaluation of drug formaldehyde conjugates, Daunoform, Doxoform, and Epidoxoform, as improved chemotherapeutics. Drug uptake, nuclear targeting, drug release, and cytotoxicity of the clinical drugs by sensitive and resistant breast and prostate cancer cells are contrasted with those of the corresponding formaldehyde conjugates. Conjugates are taken up better, retained longer, and are more toxic to a wide variety of tumor cells. The kinetics of drug release from Doxoform and Epidoxoform treated MCF-7/Adr cells are biexponential and correlate with the biexponential kinetics of drug release from extracellular DNA. The results of the lead conjugate, Epidoxoform, in the National Cancer Institute 60 human tumor cell screen are presented and discussed in terms of some resistance mechanisms. Epidoxoform shows increased toxicity to all panels relative to doxorubicin and epidoxorubicin, and this enhanced toxicity is especially evident with the more resistant cell lines.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacology , Cell Nucleus/drug effects , Neoplasms/drug therapy , Neoplasms/ultrastructure , Animals , Cell Nucleus/metabolism , Drug Resistance, Neoplasm , Humans , Neoplasms/metabolism , Tumor Cells, Cultured
4.
Biochem Pharmacol ; 60(12): 1915-23, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11108808

ABSTRACT

One of the proposed mechanisms for multidrug resistance relies on the ability of resistant tumor cells to efficiently promote glutathione S-transferase (GST)-catalyzed GSH conjugation of the antitumor drug. This type of conjugation, observed in several families of drugs, has never been documented satisfactorily for anthracyclines. Adriamycin-resistant human breast cancer MCF-7/DOX cells, presenting a comparable GSH concentration, but a 14-fold increase of the GST P1-1 activity relative to the sensitive MCF-7 cells, have been treated with adriamycin in the presence of verapamil, an inhibitor of the 170 P-glycoprotein (P-gp) drug transport protein, and scrutinized for any production of GSH-adriamycin conjugates. HPLC analysis of cell content and culture broths have shown unequivocally that no GSH conjugates are present either inside the cell or in the culture broth. The only anthracycline present inside the cells after 24 hr of incubation was > 98% pure adriamycin. Confocal laser scanning microscopic observation showed that in MCF-7/DOX cells adriamycin was localized mostly in the Golgi apparatus rather than in the nucleus, the preferred site of accumulation for sensitive MCF-7 cells. These findings rule out GSH conjugation or any other significant biochemical transformation as the basis for resistance to adriamycin and as a ground for the anomalous localization of the drug in the cell. Adriamycin, daunomycin, and menogaril did not undergo meaningful conjugation to GSH in the presence of GST P1-1 at pH 7.2. Indeed, their synthetic C(7)-aglycon-GSH conjugates exerted a strong inhibitory effect on GST P1-1, with K(i) at 25 degrees in the 1-2 microM range, scarcely dependent on their stereochemistry at C(7).


Subject(s)
Breast Neoplasms/metabolism , Doxorubicin/metabolism , Glutathione Transferase/metabolism , Glutathione/metabolism , Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/enzymology , Doxorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Female , Glutathione Transferase/antagonists & inhibitors , Humans , Tumor Cells, Cultured
5.
J Biotechnol ; 81(2-3): 167-78, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10989176

ABSTRACT

High sensitivity and specificity of two modified ssDNA aptamers capable of photocross-linking recombinant human basic fibroblast growth factor (bFGF((155))) were demonstrated. The aptamers were identified through a novel, covalent, in vitro selection methodology called photochemical systematic evolution of ligands by exponential enrichment (PhotoSELEX). The aptamers exhibited high sensitivity for bFGF((155)) comparable with commercially available ELISA monoclonal antibodies with an absolute sensitivity of at least 0.058 ppt bFGF((155)) under prevailing test conditions. The aptamers exquisitely distinguished bFGF((155)) from consanguine proteins, vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF). A commercially viable diagnostic system incorporating PhotoSELEX-evolved aptamers capable of simultaneous quantification of a large number of analyte molecules is also described. Such a system benefits from covalent bonding of aptamer to target protein allowing vigorous washing with denaturants to improve signal to noise.


Subject(s)
Bromodeoxyuridine/chemistry , DNA, Single-Stranded/chemistry , Fibroblast Growth Factor 2/analysis , Base Sequence , DNA, Single-Stranded/metabolism , Enzyme-Linked Immunosorbent Assay , Escherichia coli , Fibroblast Growth Factor 2/chemistry , Molecular Sequence Data , Photochemistry , Sensitivity and Specificity
7.
Chem Res Toxicol ; 13(6): 509-16, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10858324

ABSTRACT

Selected ion flow tube-chemical ionization mass spectrometry was used to measure formaldehyde levels in human breast cancer cells in comparison with levels in cells treated with the antitumor drugs doxorubicin (DOX) and daunorubicin (DAU) and the daunorubicin-formaldehyde conjugate Daunoform (DAUF). The measurement was performed on cell lysates and showed only background levels of formaldehyde in untreated cells and drug-treated resistant cells (MCF-7/Adr cells) but levels above background in DOX- and DAU-treated sensitive cells (MCF-7 cells). The level of formaldehyde above background was a function of drug concentration (0.5-50 microM), treatment time (3-24 h), cell density (0.3 x 10(6) to 7 x 10(6) cells/mL), and cell viability (0-100%). Higher levels of formaldehyde were observed in lysates of MCF-7 cells treated at higher drug levels, unless the treatment resulted in low cell viability. Elevated levels were directly related to cell density and were observed even with 0.5 microM drug. A lower limit for excess formaldehyde in MCF-7 cells treated with 0.5 microM DAU for 24 h is 0.3 mM. Control experiments showed that formaldehyde was not produced after cell lysis. Lysates of sensitive and resistant cells treated with 0.5 micromolar equiv of the formaldehyde conjugate (DAUF) for 3 h showed only background levels of formaldehyde. The results support a mechanism for drug cytotoxicity which involves drug induction of metabolic processes leading to formaldehyde production followed by drug utilization of formaldehyde to virtually cross-link DNA.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Doxorubicin/pharmacology , Formaldehyde/metabolism , Tumor Cells, Cultured/drug effects , Animals , Cross-Linking Reagents , Daunorubicin/pharmacology , Female , Humans , Mass Spectrometry
8.
Inorg Chem ; 39(3): 562-7, 2000 Feb 07.
Article in English | MEDLINE | ID: mdl-11229577

ABSTRACT

Ferricyanide oxidation of 1,4,5,6-tetrahydro-2,4-dimethyl-6-(2'-pyridyl)-1,2,4,5-tetrazin-3(2H)-one (pvdH3) produces the stable chelating free radical 1,5-dimethyl-3-(2'-pyridyl)-6-oxoverdazyl (pvd) as an orange solid. Combination of group 12 metal halides with the ligand pvdH3 in acetonitrile results in precipitation of metal complexes. The mercuric chloride complex crystallizes in the monoclinic space group P2(1/c) with unit cell dimensions a = 8.5768(8) A, b = 19.1718(17) A, c = 8.5956(8) A, beta = 90.405 degrees, and V = 1413.4(2) A3. The mercuric ion is tricoordinate with a distorted trigonal planar geometry. Cadmium iodide and zinc chloride induce ring opening of the tetrazine resulting in pentacoordinate complexes of a hydrazone ligand. The cadmium iodide complex crystallizes in the triclinic space group P1 with cell dimensions a = 7.7184(8) A, b = 8.0240(9) A, c = 13.348(2) A, alpha = 97.876(4) degrees, beta = 95.594(6) degrees, gamma = 107.304(6) degrees, and V = 773.40(21) A3. Oxidation of all three metal complexes produces verdazyl radicals. Metal coordination is indicated by small changes in the EPR spectrum and by changes in the UV-visible spectrum, in particular the changes in the position of bands in the visible region. The metal halide-pvd complexes can also be synthesized by direct combination of metal halides with the free radical.

9.
Acta Crystallogr D Biol Crystallogr ; 55(Pt 9): 1516-23, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10489446

ABSTRACT

Epidoxorubicin and daunorubicin are proposed to be cytotoxic to tumor cells by catalyzing production of formaldehyde through redox cycling and using the formaldehyde for covalent attachment to DNA at G bases. The crystal structure of epidoxorubicin covalently bound to a d(CGCGCG) oligomer was determined to 1.6 A resolution. The structure reveals slightly distorted B-form DNA bearing two molecules of epidoxorubicin symmetrically intercalated at the termini, with each covalently attached from its N3' to N2 of a G base via a CH2 group from the formaldehyde. The structure is analogous to daunorubicin covalently bound to d(CGCGCG) determined previously, except for additional hydrogen bonding from the epimeric O4' to O2 of a C base. The role of drug-DNA covalent bonding in cells was investigated with synthetic epidoxorubicin-formaldehyde conjugate (Epidoxoform) and synthetic daunorubicin-formaldehyde conjugate (Daunoform). Uptake and location of drug fluorophore in doxorubicin-resistant human breast-cancer cells (MCF-7/ADR cells) was observed by fluorescence microscopy and flow cytometry. The fluorophore of Daunoform appeared more rapidly in cells and was released more rapidly from cells than the fluorophore of Epidoxoform over a 3 h exposure period. The fluorophore appeared predominantly in the nucleus of cells treated with both conjugates. The difference in uptake is explained in terms of the slower rate of hydrolysis of Epidoxoform to the species reactive with DNA and a proposed slower release from DNA based upon the crystal structures.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/chemistry , DNA Adducts/chemistry , DNA Adducts/metabolism , DNA, Neoplasm/metabolism , Epirubicin/analogs & derivatives , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Cross-Linking Reagents/chemistry , Crystallography, X-Ray , Daunorubicin/analogs & derivatives , Daunorubicin/chemistry , Daunorubicin/metabolism , Daunorubicin/pharmacokinetics , Daunorubicin/pharmacology , Epirubicin/chemistry , Epirubicin/metabolism , Epirubicin/pharmacokinetics , Epirubicin/pharmacology , Humans , Intercalating Agents/chemistry , Intercalating Agents/metabolism , Intercalating Agents/pharmacokinetics , Intercalating Agents/pharmacology , Nucleic Acid Conformation , Oligonucleotides/chemistry , Tissue Distribution , Tumor Cells, Cultured
10.
Chem Res Toxicol ; 12(7): 588-96, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10409398

ABSTRACT

The anthracycline, antitumor drugs doxorubicin (DOX), daunorubicin (DAU), and epidoxorubicin (EPI) catalyze production of formaldehyde through induction of oxidative stress. The formaldehyde then mediates covalent bonding of the drugs to DNA. Synthetic formaldehyde conjugates of DOX, DAU, and EPI, denoted Doxoform (DOXF), Daunoform (DAUF), and Epidoxoform (EPIF), exhibit enhanced toxicity to anthracycline-sensitive and -resistant tumor cells. Uptake and retention of parent anthracycline antitumor drugs (DOX, DAU, and EPI) relative to those of their formaldehyde conjugates (DOXF, DAUF, and EPIF) were assessed by flow cytometry in both drug-sensitive MCF-7 cells and drug-resistant MCF-7/ADR cells. The MCF-7 cells took up more than twice as much drug as the MCF-7/ADR cells, and both cell lines took up substantially more of the formaldehyde conjugates than the parent drugs. Both MCF-7 and MCF-7/ADR cells retained fluorophore from DOXF, DAUF, and EPIF hours after drug removal, while both cell lines almost completely expelled DOX, DAU, and EPI within 1 h. Longer treatment with DOX, DAU, and EPI resulted in modest drug retention in MCF-7 cells following drug removal but poor retention of DOX, DAU, and EPI in MCF-7/ADR cells. Fluorescence microscopy showed that the formaldehyde conjugates targeted the nuclei of both sensitive and resistant cells, and remained in the nucleus hours after drug removal. Experiments in which [(3)H]Doxoform was used, synthesized from doxorubicin and [(3)H]formaldehyde, also indicated that Doxoform targeted the nucleus. Elevated levels of (3)H were observed in DNA isolated from [(3)H]Doxoform-treated MCF-7 and MCF-7/ADR cells relative to controls. The results implicate drug-DNA covalent bonding in the tumor cell toxicity mechanism of these anthracyclines.


Subject(s)
Antineoplastic Agents/pharmacology , DNA, Neoplasm/metabolism , Daunorubicin/pharmacology , Doxorubicin/pharmacology , Epirubicin/pharmacology , Formaldehyde/metabolism , Alkylation , Antineoplastic Agents/metabolism , Cell Nucleus/metabolism , DNA, Neoplasm/analysis , Daunorubicin/analogs & derivatives , Daunorubicin/metabolism , Doxorubicin/analogs & derivatives , Doxorubicin/metabolism , Epirubicin/analogs & derivatives , Epirubicin/metabolism , Humans , RNA, Neoplasm/analysis , Spectrometry, Fluorescence , Tumor Cells, Cultured
11.
Anticancer Res ; 19(2A): 1201-8, 1999.
Article in English | MEDLINE | ID: mdl-10368676

ABSTRACT

Recent data indicate that the clinical anthracycline anti-tumor drugs, doxorubicin (DOX), daunorubicin (DAU), and epidoxorubicin (EPI), catalyze the production of formaldehyde through induction of oxidative stress and bind the formaldehyde to form a metabolite which covalently bonds to DNA. Based upon this discovery, anthracycline-formaldehyde conjugates were synthesized and evaluated in three metastatic prostate cancer cell lines, LNCaP, PC-3, and DU-145. The doxorubicin-formaldehyde conjugate, Doxoform (DOXF), inhibits the growth of PC-3 and DU-145 cells 50- and 80-fold better, respectively, than the corresponding clinical drug, DOX. Daunorubicin- and epidoxorubicin-formaldehyde conjugates, Daunoform and Epidoxoform (DAUF and EPIF), inhibit the growth about 6 to 10-fold better than the clinical drugs, DAU and EPI. In addition, DAUF, DOXF, and EPIF are 2- to 20-fold more toxic to the doxorubicin-sensitive metastatic prostate cancer cell line, LNCaP. Fluorescence microscopy indicates that the nucleus is the major target for all six drugs. Flow cytometry together with fluorescence microscopy shows that DOXF and EPIF are taken up more rapidly and more abundantly and are retained in the nucleus longer than DOX and EPI, respectively, especially in DU-145 cells. The enhanced toxicity of the anthracycline-formaldehyde conjugates is attributed to their increased nuclear uptake and retention and suggests that DOXF, DAUF, and EPIF are prodrugs to the active metabolites of the clinical drugs DOX, DAU, and EPI.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Cell Nucleus/metabolism , Formaldehyde/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Antibiotics, Antineoplastic/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Drug Carriers , Flow Cytometry , Humans , Male , Tumor Cells, Cultured
12.
Protein Sci ; 8(12): 2806-12, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10631998

ABSTRACT

A photocrosslink between basic fibroblast growth factor (bFGF155) and a high affinity ssDNA oligonucleotide was characterized by positive ion electrospray ionization mass spectrometry (ESIMS). The DNA was a 61-mer oligonucleotide photoaptamer bearing seven bromodeoxyuridines, identified by in vitro selection. Specific photocrosslinking of the protein to the oligonucleotide was achieved by 308 nm XeCl excimer laser excitation. The cross-linked protein nucleic acid complex was proteolyzed with trypsin. The resulting peptide crosslink was purified by PAGE, eluted, and digested by snake venom phosphodiesterase/alkaline phosphatase. Comparison of the oligonucleotide vs. the degraded peptide crosslink by high performance liquid chromatography coupled to an electrospray ionization triple quadrupole mass spectrometer showed a single ion unique to the crosslinked material. Sequencing by collision induced dissociation (MS/MS) on a triple quadrupole mass spectrometer revealed that this ion was the nonapeptide TGQYKLGSK (residues 130-138) crosslinked to a dinucleotide at Tyr133. The MS/MS spectrum indicated sequential fragmentation of the oligonucleotide to uracil covalently attached to the nonapeptide followed by fragmentation of the peptide bonds. Tyr133 is located within the heparin binding pocket, suggesting that the in vitro selection targeted this negative ion binding region of bFGF155.


Subject(s)
Fibroblast Growth Factor 2/chemistry , Oligonucleotides/chemistry , Alkaline Phosphatase/chemistry , Amino Acid Sequence , Bromodeoxyuridine , Chromatography, High Pressure Liquid , Cross-Linking Reagents , Fibroblast Growth Factor 2/radiation effects , Humans , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Oligonucleotides/radiation effects , Peptide Fragments/chemistry , Phosphodiesterase I , Phosphoric Diester Hydrolases/chemistry , Radiation-Sensitizing Agents , Recombinant Proteins/chemistry , Recombinant Proteins/radiation effects , Trypsin/chemistry , Ultraviolet Rays
13.
Anticancer Res ; 18(2A): 1159-66, 1998.
Article in English | MEDLINE | ID: mdl-9615782

ABSTRACT

The accumulation of adriamycin (ADR), daunomycin (DAUNO) and their glutathione (GSH)-conjugates, recently obtained by anaerobic reaction of the parent anthracyclines with reduced GSH, was examined in drug-sensitive and multidrug resistant (MDR) cell lines using confocal laser scanning microscopy. In all drug-sensitive lines used (TVM-A12 and TVM-A197 human melanoma cells, K562 lymphoblastoid cells and MCF-7 human breast cancer cells) ADR and DAUNO were mostly located in the nuclei, while their GSH-conjugates were found only in the cytoplasm, predominantly in the Golgi region. On the contrary, in MDR MCF-7/Dox cells, both conjugated and non conjugated anthracyclines gave fluorescence only in the cytoplasm, mostly in the Golgi region, the intensity of the fluorescence being stronger in cells pretreated with verapamil. Viability assay showed that the GSH-conjugate are significantly less cytotoxic than the parent anthracyclines in sensitive cells and showed the same scarce cytotoxicity in MDR MCF-7/Dox cells. These results demonstrate that conjugation of anthracycline antitumor drugs with GSH prevents their access to the nucleus and decreases their cytotoxicity. Furthermore, the observations on MCF-7/Dox suggest that GSH-conjugation of anthracycline might occur in resistant cells and can be in part responsible for the MDR in breast cancer cells.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Cytoplasm/metabolism , Drug Resistance, Neoplasm , Glutathione/metabolism , Antibiotics, Antineoplastic/pharmacology , Drug Resistance, Multiple , Golgi Apparatus/metabolism , Humans , Microscopy, Confocal , Tumor Cells, Cultured
14.
J Med Chem ; 41(8): 1306-14, 1998 Apr 09.
Article in English | MEDLINE | ID: mdl-9548820

ABSTRACT

The recent discovery that the formaldehyde conjugates of doxorubicin and daunorubicin, Doxoform and Daunoform, are cytotoxic to resistant human breast cancer cells prompted the search for hydrolytically more stable anthracycline-formaldehyde conjugates. Doxoform and Daunoform consist of two molecules of the parent drug bound together with three methylene groups, two forming oxazolidine rings and one binding the oxazolidines together at their 3'-amino nitrogens. The 4'-epimer of doxorubicin, epidoxorubicin, reacts with formaldehyde at its amino alcohol functionality to produce a conjugate, Epidoxoform, in 59% yield whose structure consists of two molecules of epidoxorubicin bound together with three methylene groups in a 1, 6-diaza-4,9-dioxabicyclo[4.4.1]undecane ring system. The structure was established from spectroscopic data and is consistent with products from reaction of simpler vicinal trans-amino alcohols with formaldehyde. Epidoxoform hydrolyzes at pH 7.3 to an equilibrium mixture with dimeric and monomeric epidoxorubicin-formaldehyde conjugates without release of formaldehyde or epidoxorubicin. The hydrolysis follows the rate law (A if B) if C + D where A (Epidoxoform) is in rapid equilibrium with B, and B is in slow equilibrium with C and D. The forward rate constant for A/B going to C+D gives a half-life of approximately 2 h at 37 degrees C. At equilibrium the mixture is stable for at least 2 days. At pH 6.0, hydrolysis proceeds with first-order kinetics to epidoxorubicin and formaldehyde with a half-life of 15 min at 37 degrees C. Epidoxoform and epidoxorubicin plus formaldehyde react with the self-complementary DNA octamer (GC)4 to yield five drug-DNA adducts which have structures analogous to the doxorubicin-DNA adducts from reaction of Doxoform with (GC)4. Epidoxoform is 3-fold more toxic to MCF-7 human breast cancer cells and greater than 120-fold more toxic to MCF-7/ADR resistant cells than epidoxorubicin. Epidoxoform in equilibrium with its hydrolysis products is greater than 25-fold more toxic to resistant cells with respect to epidoxorubicin.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Epirubicin/analogs & derivatives , Antibiotics, Antineoplastic/chemical synthesis , Antibiotics, Antineoplastic/chemistry , Cell Division/drug effects , DNA Adducts/chemistry , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Epirubicin/chemical synthesis , Epirubicin/chemistry , Epirubicin/pharmacology , Humans , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Magnetic Resonance Spectroscopy , Mass Spectrometry , Oligonucleotides/chemistry , Tumor Cells, Cultured
15.
Curr Pharm Des ; 4(3): 203-18, 1998 Jun.
Article in English | MEDLINE | ID: mdl-10197040

ABSTRACT

Doxorubicin has been a constituent of antitumor drug protocols for a broad spectrum of cancers for more than two decades. Side effects and resistance continue to be important limitations. Drug targets responsible for both side effects and anti-tumor activity are cell membrane receptors, cell membrane lipids, nucleic acids and topoisomerase. Induction of oxidative stress is responsible for most if not all biological activity. An important consequence of oxidative stress is the production of formaldehyde which can subsequently be utilized by the drug for covalent bonding to nucleic acids and other targets as shown by in vitro experiments. Multidrug resistance mechanisms inhibit drug-induced DNA damage, drug uptake, and drug-induced oxidative stress. Synthetic anthracyclines conjugated to formaldehyde circumvent some if not all of the resistance mechanisms. Consequently, anthracycline-formaldehyde conjugates have potential for the treatment of resistant cancer.


Subject(s)
Alkylating Agents/metabolism , Anthracyclines/metabolism , Antineoplastic Agents/therapeutic use , Doxorubicin/metabolism , Drug Design , Anthracyclines/therapeutic use , Antineoplastic Agents/metabolism , Chemistry, Pharmaceutical , DNA Adducts , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Molecular Structure , Nucleic Acids/metabolism , Oxidation-Reduction
16.
Chem Res Toxicol ; 10(9): 953-61, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9305576

ABSTRACT

The reaction of the antitumor drugs adriamycin and daunomycin with the self-complementary DNA oligonucleotide (GC)4 to generate DNA-drug adducts was investigated as a function of redox reaction conditions. The redox systems dithiothreitol (DTT)/Fe(III) and xanthine oxidase/ NADH both gave the same distribution of four DNA-anthracycline adducts. In each of these adducts the anthracycline is bonded via a methylene linkage between the 3'-amino group of the drug and the 2-amino group of a deoxyguanosine of the DNA. The methylene linkage results from reaction of the drug and DNA with in situ-generated formaldehyde via Schiff base chemistry [Taatjes, D.J., Gaudiano, G., Resing, K., and Koch, T.H. (1997) J. Med. Chem. 40, 1276-1286]. Formaldehyde production is promoted by iron, inhibited by metal-chelating agents, and does not require drug. Iron enhances formaldehyde production by a factor of 30, EDTA inhibits its formation by a factor of 2, and Desferal inhibits its formation by a factor of more than 20. Hydrogen peroxide accumulates in significant quantities only with xanthine oxidase/NADH in the presence of Desferal. The results are explained in terms of Fenton oxidation of Tris buffer to formaldehyde. Biological reagents also cause DNA-drug adduct formation; reduction of ferric ion with glutathione in phosphate buffer in the presence of spermine produced the same DNA-drug adducts. The observations are discussed in terms of cytotoxicity resulting from iron chelated to adriamycin catalyzing in vivo production of formaldehyde which links adriamycin to DNA and tumor cell resistance resulting from factors which decrease formaldehyde.


Subject(s)
Antibiotics, Antineoplastic/chemistry , DNA Adducts/chemistry , Daunorubicin/chemistry , Doxorubicin/chemistry , Formaldehyde/chemistry , Anaerobiosis , Buffers , Chromatography, High Pressure Liquid , Dithiothreitol , Glutathione/chemistry , Hydrogen Peroxide , Indicators and Reagents , Iron/chemistry , NAD/chemistry , Oxidation-Reduction , Xanthine Oxidase/chemistry
17.
J Med Chem ; 40(16): 2452-61, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9258351

ABSTRACT

The recent discovery that the clinically important antitumor drugs doxorubicin and daunorubicin alkylate DNA via catalytic production of formaldehyde prompted the synthesis of derivatives bearing formaldehyde. Reaction of the parent drugs with aqueous formaldehyde at pH 6 produced in 40-50% yield conjugates consisting of two molecules of the parent drug as oxazolidine derivatives bound together at their 3'-nitrogens by a methylene group. The structures were established as bis(3'-N-(3'-N,4'-O-methylenedoxorubicinyl)) methane (Doxoform) and bis(3'-N-(3'-N,4'-O-methylenedaunorubicinyl))methane (Daunoform) from spectroscopic data. Both derivatives are labile with respect to hydrolysis to the parent drugs. 3'-N,4'-O-Methylenedoxorubicin and 3'-N,4'-O-methylenedaunorubicin are intermediates in the hydrolysis. Daunoform reacts with the self-complementary deoxyoligonucleotide (GC)4 faster than the combination of daunorubicin and formaldehyde at an equivalent concentration to given drug-DNA adducts. In spite of hydrolytic instability, Doxoform is 150-fold more toxic to MCF-7 human breast cancer cells and 10000-fold more toxic to MCF-7/ADR resistant cells. Toxicity to resistant cancer cells is interpreted in terms of higher lipophilicity of the derivatives and circumvention of catalytic formaldehyde production.


Subject(s)
Daunorubicin/analogs & derivatives , Doxorubicin/analogs & derivatives , Formaldehyde/chemistry , Oxazoles/chemistry , Alkylation , Catalysis , Chromatography, High Pressure Liquid , DNA/metabolism , Daunorubicin/chemistry , Daunorubicin/pharmacology , Dimerization , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Formaldehyde/pharmacology , Humans , Hydrolysis , Mass Spectrometry , Models, Chemical , Oxazoles/pharmacology , Tumor Cells, Cultured
18.
J Med Chem ; 40(8): 1276-86, 1997 Apr 11.
Article in English | MEDLINE | ID: mdl-9111302

ABSTRACT

Reaction of the anthracycline, antitumor drugs adriamycin and daunomycin with the self-complementary DNA oligonucleotide GCGCGCGC, (GC)4, in the presence of the reducing agent dithiothreitol, the oxidizing agent hydrogen peroxide, or the alkylating agent formaldehyde gives a similar mixture of DNA-drug adducts. Negative ion electrospray mass spectra indicate that adduct formation involves coupling of the DNA to the anthracycline via a methylene group and that the major adduct is duplex DNA containing two molecules of anthracycline, each bound to a separate strand of the DNA via a methylene group. The source of the methylene group is formaldehyde. A molecular structure with each anthracycline intercalated at a 5'-CpG-3' site and covalently bound from its 3'-amino group to a 2-amino group of a 2'-deoxyguanosine nucleotide is proposed based upon spectral data and a relevant crystal structure. The reaction of (GC)4 with the anthracyclines and formaldehyde forms an equilibrium mixture with DNA-drug adducts which is shifted toward free DNA by dilution. The results suggest a pathway to the inhibition of transcription by reductively activated adriamycin and daunomycin. Reductive activation in the presence of oxygen yields hydrogen peroxide; hydrogen peroxide oxidizes constituents in the reaction mixture to formaldehyde; and formaldehyde couples the drug to DNA. In this regard, hydrogen peroxide reacts with adriamycin via Baeyer-Villiger reactions at the 13-position to yield 2, 3, and formaldehyde. Formaldehyde also results from hydrogen peroxide oxidation of Tris [tris(hydroxymethyl)aminomethane] present in transcription buffer and spermine, a polyamine commonly associated with DNA in vivo, presumably via the Fenton reaction.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Antineoplastic Agents/metabolism , DNA/metabolism , Daunorubicin/metabolism , Doxorubicin/metabolism , Alkylation , Chromatography, High Pressure Liquid , DNA Adducts/chemistry , DNA Adducts/metabolism , Mass Spectrometry , Models, Chemical , Oxidation-Reduction
19.
Crit Rev Biochem Mol Biol ; 32(2): 101-40, 1997.
Article in English | MEDLINE | ID: mdl-9145285

ABSTRACT

Photocross-linking is a useful technique for the partial definition of the nucleic acid-protein interface of nucleoprotein complexes. It can be accomplished by one or two photon excitations of wild-type nucleoprotein complexes or by one photon excitation of nucleoprotein complexes bearing one or more substitutions with photoreactive chromophores. Chromophores that have been incorporated into nucleic acids for this purpose include aryl azides, 5-azidouracil, 8-azidoadenine, 8-azidoguanine, 4-thiouracil, 5-bromouracil, 5-iodouracil, and 5-iodocytosine. The various techniques and chromophores are described and compared, with attention to the photochemical mechanism.


Subject(s)
Nuclear Proteins/chemistry , Nucleic Acids/chemistry , Photochemistry/methods , Animals , Cross-Linking Reagents , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Nuclear Proteins/metabolism , Nucleic Acids/metabolism , Protein Binding , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...