Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Pharm Sci ; 100(9): 3977-84, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21344411

ABSTRACT

BNP7787, an investigational drug undergoing global Phase III development, appears to have potential advantages over other cytoprotective compounds that have been evaluated for preventing and mitigating cisplatin-induced nephrotoxicity. Herein, we characterized the in vitro accumulation of BNP7787 in human renal proximal tubule cells (HK-2) in which cisplatin is known to be taken up and accumulate. HK-2 cells were incubated with pharmacological concentrations of BNP7787 for varying times. Temperature-dependent accumulation of BNP7787 in cells was observed and the BNP7787-derived metabolite, mesna, formed intracellularly was directly monitored. The peak level of BNP7787-derived mesna measured in HK-2 cells was approximately 0.6 nmol/10(6) cells; this is pharmacologically similar to reported platinum concentrations in kidney cells and may be sufficient to afford nephroprotection. Therefore, in addition to previously suggested glomerular filtration, the cellular accumulation of BNP7787 by HK-2 cells is a plausible newly identified mechanism by which BNP7787 may accumulate in renal tubular cells, where it can exert its pharmacological effects to protect against cisplatin-induced nephrotoxicity by direct covalent conjugation of mesna with cisplatin, or by the formation of BNP7787-derived mesna-disulfide heteroconjugates that exert nephroprotective effects by inhibition of the key toxification enzyme targets γ-glutamyltranspeptidase and aminopeptidase N.


Subject(s)
Kidney Tubules, Proximal/metabolism , Mesna/analogs & derivatives , Cell Line , Chromatography, High Pressure Liquid , Electrochemistry , Humans , Kidney Tubules, Proximal/cytology , Mesna/pharmacokinetics
2.
Cancer Chemother Pharmacol ; 67(2): 381-91, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20440617

ABSTRACT

PURPOSE: Previous studies from our laboratory have identified a role for gamma-glutamyl transpeptidase (GGT) in BNP7787 (disodium 2,2'-dithio-bis ethane sulfonate, dimesna, Tavocept™)-mediated cisplatin nephroprotection. Dekant has proposed that gamma-glutamyl transpeptidase (GGT), aminopeptidase N (APN) and cysteine-conjugate-ß-lyase (CCBL) comprise a multi-enzyme pathway that acts on xenobiotic-glutathione conjugates converting them to nephrotoxic metabolites. We report modulation of APN activity within this pathway by BNP7787-derived mesna-disulfide heteroconjugates. METHODS: A fluorimetric assay was used to determine the effect of BNP7787, BNP7787-derived mesna-disulfide heteroconjugates, and the BNP7787 metabolite, mesna (sodium 2-mercaptoethane sulfonate), on the initial velocity and overall progress curve of the human APN reaction in vitro. RESULTS: Neither BNP7787 nor mesna-cysteinyl-glutamate inhibited human APN. Select BNP7787-derived mesna-disulfide heteroconjugates (mesna-cysteine, mesna-glutathione, mesna-cysteinyl-glycine) and high concentrations of mesna inhibited APN activity. Allosteric effects on the enzyme progress curve outside of the linear initial velocity region were observed for mesna-cysteinyl-glycine, mesna-glutathione and mesna-cysteinyl-glutamate and appeared to be a function of having both mesna and di- or tri-peptide functionalities in one molecule. In situ-generated mesna-cisplatin conjugates were not a substrate for human APN. CONCLUSIONS: BNP7787-mediated prevention or mitigation of cisplatin-induced nephrotoxicity may involve APN inhibition by certain BNP7787-derived mesna-disulfide heteroconjugates and appears correlated to the presence of a glycinate moiety and/or an anionic group. Two general mechanisms for BNP7787-mediated nephroprotection of cisplatin-induced nephrotoxicity involving the GGT, APN and CCBL nephrotoxigenic pathway are proposed which acting in a concerted and/or synergistic manner, and thereby prevent or mitigate cisplatin-induced renal toxicity.


Subject(s)
CD13 Antigens/antagonists & inhibitors , Cisplatin/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Mesna/analogs & derivatives , Protective Agents/pharmacology , Allosteric Regulation , Biocatalysis/drug effects , CD13 Antigens/genetics , CD13 Antigens/metabolism , Cisplatin/analogs & derivatives , Cisplatin/metabolism , Cisplatin/pharmacology , Cysteine/analogs & derivatives , Cysteine/metabolism , Cysteine/pharmacology , Dipeptides/pharmacology , Glutathione/analogs & derivatives , Glutathione/metabolism , Glutathione/pharmacology , Glycine/analogs & derivatives , Glycine/metabolism , Glycine/pharmacology , Humans , Kinetics , Mesna/metabolism , Mesna/pharmacology , Models, Biological , Protective Agents/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
3.
Br J Cancer ; 90(8): 1654-9, 2004 Apr 19.
Article in English | MEDLINE | ID: mdl-15083199

ABSTRACT

In preclinical studies, BNP7787 (disodium 2,2'-dithio-bis-ethane sulphonate), the disulphide form of mesna, has demonstrated selective protection against cisplatin-induced nephrotoxicity due to conversion into mesna inactivating toxic platinum species. Mesna (sodium 2-mercapto ethane sulphonate), however, can affect the antitumour activity of cisplatin, while BNP7787 does not interfere with the antitumour activity. To understand the difference in interference with cisplatin-induced antitumour activity between BNP7787 and mesna as well to characterise the selective nephroprotection by BNP7787, the pharmacokinetics of BNP7787 and mesna, each given i.v. 1000 mg x kg(-1), were determined in plasma, kidney, liver, red blood cells (RBC), skeletal muscle and tumour of Fischer rats bearing subcutaneously implanted WARD colon tumours. The following results were obtained: (1). high concentrations of BNP7787 and mesna were observed in the plasma and kidney after administration of BNP7787 or mesna, except for mesna in plasma after BNP7787 administration; (2). in all other sampled compartments, the AUC values of both compounds were at least 5.5-fold lower than the corresponding values in kidney; (3). the AUC of mesna in plasma after mesna administration was comparable to the AUC of mesna in kidney after a dose of BNP7787 that can completely prevent cisplatin-induced nephrotoxicity in rats; (4). the AUC of mesna in plasma was five-fold higher relative to the AUC of mesna following BNP7787 administration (P<0.01). In conclusion, the five-fold higher AUC of mesna in plasma after mesna administration and the fact that mesna is more reactive with (hydrated) cisplatin than its disulphide form BNP7787 represent a plausible explanation as to why mesna administration can reduce the antitumour activity of cisplatin. After BNP7787 administration, the distribution of BNP7787 and mesna was restricted to the kidney, which confirmed the selective protection of the kidney by BNP7787.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Mesna/analogs & derivatives , Mesna/pharmacology , Mesna/pharmacokinetics , Protective Agents/pharmacology , Protective Agents/pharmacokinetics , Animals , Area Under Curve , Colonic Neoplasms/drug therapy , Colonic Neoplasms/veterinary , Female , Humans , Injections, Intravenous , Kidney/drug effects , Kidney/pathology , Mesna/administration & dosage , Neoplasms, Experimental , Protective Agents/administration & dosage , Rats , Rats, Inbred F344
4.
Eur J Cancer ; 38(8): 1148-56, 2002 May.
Article in English | MEDLINE | ID: mdl-12008205

ABSTRACT

BNP7787 (2',2'-dithio-bis-ethane sulphonate sodium), a water-soluble disulphide, is chemically and mechanistically different from other sulphur-containing chemoprotective agents. Presently, BNP7787 is under investigation for its protective properties with regard to the side-effects of platinum compounds. In this study, we evaluated BNP7787, mesna and amifostine for their effects on the antitumour activity of platinum compounds. Continuous exposure to BNP7787 did not affect the antiproliferative effects of cisplatin or carboplatin, but the efficacy of both compounds was reduced in the presence of mesna in vitro in two human ovarian cancer cell lines. BNP7787 or amifostine combined with cisplatin or carboplatin given in standard schedules for the treatment of nude mice bearing well-established OVCAR-3 xenografts did not interfere with platinum-induced inhibition of tumour growth. Of interest, BNP7787 or amifostine co-administered with carboplatin was significantly more effective than carboplatin alone (P<0.01). In the presence of amifostine, doses of cisplatin and carboplatin could be safely increased by factors of 1.6 and 1.5, respectively. Unlike in a previous study of BNP7787 in tumour-bearing rats, BNP7787 did not protect against additional weight loss following treatment with higher doses of cisplatin in OVCAR-3-bearing mice. Pharmacokinetics of (mixed) disulphides including BNP7787 and extractable mesna in deproteinised plasma revealed a rapid disappearance of BNP7787 and an AUC(5-60) value of mesna 9-fold lower than that calculated after an equivalent dose of mesna by weight. We can conclude that BNP7787 does not interfere with the antitumour activity of platinum compounds in vitro and in vivo. Clinical trials are underway to evaluate the protection of normal tissues by BNP7787 when combined with cisplatin.


Subject(s)
Antineoplastic Agents/therapeutic use , Carboplatin/therapeutic use , Cisplatin/therapeutic use , Mesna/analogs & derivatives , Mesna/pharmacology , Ovarian Neoplasms/drug therapy , Protective Agents/pharmacology , Amifostine/pharmacology , Animals , Cell Division/drug effects , Drug Interactions , Female , Humans , Lethal Dose 50 , Mesna/blood , Mesna/pharmacokinetics , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Radiation-Protective Agents/pharmacology , Transplantation, Heterologous , Weight Loss
5.
Clin Cancer Res ; 7(4): 935-41, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309344

ABSTRACT

This study was aimed at characterizing the role of BCRP/MXR/ABCP (BCRP) in resistance of the human ovarian tumor cell lines T8 and MX3 to camptothecins more extensively and investigating whether resistance can be reversed by inhibiting BCRP by GF120918. Camptothecins studied were topotecan, CPT-11, and its active metabolite SN-38, 9-aminocamptothecin, and the novel experimental camptothecins NX211, DX8951f, and BNP1350. Notably, DX8951f and BNP1350 appeared to be very poor substrates for BCRP, with much lower resistance factors observed both in T8 and MX3 cells than observed for the other camptothecins tested. In the presence of a nontoxic dose level of GF120918, the intracellular accumulation of topotecan in the T8 and MX3 cells was completely restored to the intracellular levels observed in the sensitive IGROV1 parental cell line. This resulted in almost complete reversal of drug resistance to topotecan and to most of the other topoisomerase I drugs tested in the T8 cell line and to complete reversal in the MX3 cells. However, coincubation of DX8951f or BNP1350 with GF120918 did not affect the cytotoxicity of either of these drugs significantly. From the combined data, we conclude that the affinities of topoisomerase I drugs for BCRP are, in decreasing order: SN-38 > topotecan > 9-aminocamptothecin approximately CPT-11 > NX211 > DX8951f > BNP1350. Furthermore, GF120918 appears to be a potent reversal agent of BCRP-mediated resistance to camptothecins, with almost complete reversal noted at 100 nM. Potential BCRP-mediated resistance to topoisomerase I inhibitors can also be avoided by using the BCRP-insensitive drugs DX8951f or BNP1350. This observation may have important clinical implications for future development of novel camptothecins.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Acridines/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Isoquinolines/pharmacology , Neoplasm Proteins , Tetrahydroisoquinolines , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Cell Survival/drug effects , Drug Interactions , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Tumor Cells, Cultured
6.
J Mol Biol ; 303(5): 679-92, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-11061968

ABSTRACT

Tubulin, the major structural component of the microtubules, participates actively in mitotic spindle formation and chromosomal organization during cell division. Tubulin is the major target for a variety of anti-mitotic drugs. Some of the drugs, such as Vinca alkaloids and taxol, are routinely used for cancer chemotherapy. It is unfortunate that our knowledge of the binding sites on tubulin of these drugs is limited because of lack of a useful and appropriate tool. The photoaffinity labeling approach is the major technique available at present to detect the binding sites of drugs on tubulin. This method, however, has several limitations. First, only part of the binding site can be identified, namely, the residues which react with the photoaffinity label. Second, there are regions of tubulin which are not at the binding site but are affected by the binding of the drug; these regions can not be detected by the photoaffinity labeling approach. The third, and perhaps most serious, limitation is that the traditional approach can detect areas which have nothing to do with the binding of the ligand but which are within a certain distance of the binding site, that distance being less than the length of the photoreactive moiety attached to the ligand. There has been a great deal of controversy on the localization of the binding site of colchicine on tubulin, with some reports suggesting that the binding site is on alpha and some supporting a binding site on beta. Colchicine also has significant effects on tubulin conformation, but the regions which are affected have not been identified. We have attempted here to address these questions by a novel "footprinting" method by which the drug-binding sites and as well as the domain of tubulin affected by drug-induced conformational changes could be determined. Here, we report for the first time that the interaction of the B-ring of colchicine with the alpha-subunit affects a domain of tubulin which appears to be far from its binding site. This domain includes the cysteine residues at positions 295, 305, 315 and 316 on alpha-tubulin; these residues are located well away from the alpha/beta interface where colchicine appears to bind. This is correlated with the stabilizing effect of colchicine on the tubulin molecule. Furthermore, we also found that the B-ring of colchicine plays a major role in the stability of tubulin while the A and the C-rings have little effect on it. Our results therefore, support a model whereby colchicine binds at the alpha/beta interface of tubulin with the B-ring on the alpha-subunit and the A and the C-rings on the beta-subunit.


Subject(s)
Colchicine/chemistry , Colchicine/metabolism , Protein Footprinting , Stilbenes , Tropolone/analogs & derivatives , Tubulin/chemistry , Tubulin/metabolism , Amino Acid Sequence , Animals , Bibenzyls/chemistry , Bibenzyls/metabolism , Bibenzyls/pharmacology , Binding Sites , Cattle , Chromatography, High Pressure Liquid , Colchicine/pharmacology , Fluorescence , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Podophyllotoxin/chemistry , Podophyllotoxin/metabolism , Podophyllotoxin/pharmacology , Protein Conformation/drug effects , Tropolone/chemistry , Tropolone/metabolism , Tropolone/pharmacology , Tryptophan/metabolism , Urea/pharmacology
7.
Int J Cancer ; 88(2): 260-6, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11004678

ABSTRACT

BNP1350, 7-[(2-trimethylsilyl)ethyl]-20(S)-camptothecin, is a novel semi-synthetic, highly lipophilic, silicon-containing camptothecin and an inhibitor of topoisomerase I. It has been supercomputer engineered for superior oral bioavailability, superior lactone stability, broad anti-tumor activity, increased potency and insensitivity to Pgp/MRP/LRP drug resistance. We determined the efficacy of BNP1350 in experimental human colon cancer and compared its anti-tumor effects with those of CPT-11/SN-38. We also determined a possible influence of Pgp, MRP and LRP on the efficacy of BNP1350. The in vitro anti-proliferative capacity of the compounds using various exposure times was assessed in five colon cancer cell lines and indicated that BNP1350 was similarly effective or slightly more potent than SN-38. Four cell lines of other origin with sublines expressing Pgp, MRP and/or LRP showed that BNP1350 was significantly more effective than SN-38 (p < 0.05) and that the activity of BNP1350 was not reduced in multidrug-resistant cells. For in vivo experiments, BNP1350 was given 1.0 mg/kg i.p. or 1.5 mg/kg p.o. daily x 5 and CPT-11 20 mg/kg i.p. daily x 5 being equitoxic schedules in nude mice bearing s.c. human tumor xenografts. The schedules were studied in colon cancer xenografts COLO320, COLO205 or WiDr as well as in two Pgp-positive xenografts 2780AD and BRO/mdr1.1 and the parental Pgp-negative A2780 ovarian cancer xenografts and BRO melanoma xenografts. Growth inhibition of >50% was obtained for BNP1350 given i.p. in six out of the seven xenografts studied. BNP1350 was similarly effective when given i.p. or p.o. CPT-11 was as effective as BNP1350, except in BRO and BRO/mdr1.1 xenografts. Pgp expression in xenografts in vivo confirmed that there was no negative influence on the efficacy of BNP1350. In conclusion, BNP1350 shows a broad spectrum of activity in experimental human tumors and is a suitable candidate for oral treatment of cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/analogs & derivatives , Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Animals , Camptothecin/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Small Cell/drug therapy , Colonic Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Resistance, Multiple , Enzyme Inhibitors/therapeutic use , Female , Humans , Irinotecan , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Eur J Cancer ; 35(6): 984-93, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10533483

ABSTRACT

We developed a combination protocol for inhibitors of thymidylate synthase (TS) and DNA topoisomerase I (Topo I) that can exert highly lethal effects in vitro against HCT-8 human colorectal cancer cells. The specific schedule was constructed so that a TS inhibitor could induce not only primary DNA damage but also cellular conditions optimal for the efficient action of a Topo I inhibitor. The initial drug treatment consisted of a brief exposure to a quinazoline-based antifolate, ZD1694. After an interval of approximately one cell-doubling time, cells were exposed for 8-24 h to BNP1100, a Karenitecin-class 7-thiomethyl-camptothecin, in the presence of 1-10 microM thymidine; the latter acted as a crucial factor to promote the collision of moving replication forks with the drug-stabilised DNA-Topo I cleavable complexes even under continuous TS inhibition. Clonogenic analyses confirmed that these mechanistically distinct drugs at clinically achievable concentrations worked in a highly synergistic manner, with a maximum effect abolishing the viability of virtually all cancer cells (> 99.9%). The pretreatment with ZD1694 increased the amount of DNA-bound Topo I by up to 4-fold and the DNA-damaging capability of BNP1100 by up to 15-fold. The possibility of at least four DNA-damaging pathways is proposed which might have resulted from the individual actions of TS and Topo I inhibitors as well as their concerted actions. Taken together, the present findings provided a logically permissible explanation as to why TS and Topo I inhibitors in concerted interactions induced a highly lethal effect which was more than a simple additive effect. Since these drugs are effective specifically on actively proliferating cancer cells, but not on non-cycling G0/G1 cells, this mechanism-based protocol may warrant consideration for clinical verification.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Camptothecin/analogs & derivatives , Colonic Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Quinazolines/therapeutic use , Thiophenes/therapeutic use , Thymidylate Synthase/antagonists & inhibitors , Topoisomerase I Inhibitors , Camptothecin/therapeutic use , Cell Division , Cell Survival , Colonic Neoplasms/pathology , DNA Damage , Drug Synergism , Humans , Tumor Cells, Cultured
9.
Semin Oncol ; 25(5): 584-99, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9783598

ABSTRACT

Platinum-type drugs have proven to be valuable in the treatment of a variety of solid tumors, beginning with the commercial approval of cisplatin 18 years ago. There are several clinically important toxicities commonly associated with the administration of these drugs. Despite the extensive use of cisplatin and carboplatin, the fundamental chemical transformations and mechanisms that underlie their antitumor and toxic effects have not been fully characterized. Several first-generation protective thiols have been clinically studied in an attempt to reduce the toxicity of platinum-type drugs; while some of these agents appear to protect against certain toxicities, nearly all platinum-protecting drugs have their own intrinsic toxicities, which can be additive to the toxicity of platinum-type drugs. Tumor protection by platinum-protecting drugs is an additional untoward effect that is associated with certain types of agents and must be addressed with care. Recent advances in theoretical and laboratory methods and the use of supercomputers have extended our understanding of the possible major mechanisms underlying platinum drug antitumor activity and toxicity; we present strong evidence that there are two classes of chemical species of platinum drug. One class appears to predominantly account for the antitumor activity, and the other class of chemical species produces many of the toxic effects of platinum drugs. We have discovered a new nontoxic, second-generation platinum-protecting agent, known as BNP7787, which appears to selectively inactivate and eliminate toxic platinum species. BNP7787 has recently entered phase I clinical testing in cancer patients.


Subject(s)
Amifostine/pharmacology , Antineoplastic Agents/adverse effects , Mesna/analogs & derivatives , Platinum Compounds/adverse effects , Sulfhydryl Compounds/pharmacology , Amifostine/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cisplatin/adverse effects , Cisplatin/chemistry , Cisplatin/pharmacology , Drug Interactions , Humans , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Mesna/pharmacology , Mesna/therapeutic use , Platinum Compounds/chemistry , Platinum Compounds/pharmacology , Protective Agents/pharmacology , Sulfhydryl Compounds/chemistry
10.
Cancer Res ; 58(17): 3782-6, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9731484

ABSTRACT

We report the first experimental observation that a clinically important camptothecin [CPT; topotecan (TPT), a water-soluble CPT] binds directly and noncovalently to double-stranded DNA and single-stranded DNA structures in the absence of topoisomerase I, but only in the lactone form. We observed clear DNA sequence specificity of the TPT lactone binding to duplex DNA, which was comprised of alternating purine-pyrimidine sequences that contained dT. These structural studies of direct TPT lactone-DNA binding support several important considerations involving possible mechanism(s) of anticancer activity of CPT-type drugs containing a 20(S) lactone moiety.


Subject(s)
Antineoplastic Agents/metabolism , DNA Topoisomerases, Type I/physiology , DNA, Single-Stranded/metabolism , DNA/metabolism , Topotecan/metabolism , Hydrogen-Ion Concentration , Lactones/metabolism , Magnetic Resonance Spectroscopy
11.
J Med Chem ; 41(4): 515-29, 1998 Feb 12.
Article in English | MEDLINE | ID: mdl-9484502

ABSTRACT

NMR (1H and 31P) and HPLC techniques were used to study the partitioning of phosphoramide mustard (PM) and its aziridinium ions among alkylation and P-N bond hydrolysis reactions as a function of the concentration and strength of added nucleophiles at 37 degrees C and pH 7.4. With water as the nucleophile, bisalkylation accounted for only 10-13% of the product distribution given by PM. The remainder of the products resulted from P-N bond hydrolysis reactions. With 50 mM thiosulfate or 55-110 mM glutathione (GSH), bisalkylation by a strong nucleophile increased to 55-76%. The rest of the PM was lost to either HOH alkylation or P-N bond hydrolysis reactions. Strong experimental and theoretical evidence was obtained to support the hypothesis that the P-N bond scission observed at neutral pH does not occur in the parent PM to produce nornitrogen mustard; rather it is an aziridinium ion derived from PM which undergoes P-N bond hydrolysis to give chloroethylaziridine. In every buffer studied (bis-Tris, lutidine, triethanolamine, and Tris), the decomposition of PM (with and without GSH) gave rise to 31P NMR signals which could not be attributed to products of HOH or GSH alkylation or P-N bond hydrolysis. The intensities of these unidentified signals were dependent on the concentration of buffer.


Subject(s)
Aziridines/chemistry , Phosphoramide Mustards/chemistry , Alkylation , Chromatography, High Pressure Liquid , Hydrogen , Hydrolysis , Kinetics , Models, Chemical , Nuclear Magnetic Resonance, Biomolecular , Phosphorus , Structure-Activity Relationship
12.
J Chromatogr B Biomed Sci Appl ; 695(2): 401-8, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9300877

ABSTRACT

Gamma-methylene-10-deazaaminopterin (MDAM), a unique dihydrofolate reductase inhibitor, has demonstrated antitumor activity against a broad spectrum of human solid tumors in preclinical studies. A novel reversed-phase, ion-pair high-performance liquid chromatography (HPLC) assay that uses fluorescence detection has been developed to quantitate levels of MDAM and its major metabolite, 7-hydroxy-gamma-methylene-10-deazaaminopterin (7-OH-MDAM), in human plasma. The recovery of MDAM and 7-OH-MDAM from plasma was >97% by a simple one-step deproteinization process using tetrabutylammonium bromide (TBABr) and methanol. MDAM and 7-OH-MDAM remained stable in plasma over a 28-day test period at ambient temperatures, and neither compound was light-sensitive. The limit of quantitation was 0.005 microM for both MDAM and 7-OH-MDAM. This assay has been found to be simple, sensitive and reproducible in determining plasma concentrations of MDAM and 7-OH-MDAM in patients with solid cancers in a phase I trial.


Subject(s)
Aminopterin/analogs & derivatives , Antineoplastic Agents/blood , Neoplasms/blood , Pterins/blood , Aminopterin/blood , Aminopterin/therapeutic use , Antineoplastic Agents/therapeutic use , Chromatography, High Pressure Liquid , Humans , Neoplasms/drug therapy , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Fluorescence
13.
Comput Chem ; 20(4): 459-67, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8800001

ABSTRACT

Gemcitabine 2',2'-difluoro 2'-deoxy cytosine (GEM) is a novel nucleoside which has demonstrated broad preclinical anti-cancer activity and appears promising in early stage human clinical trials. One purpose of this study was to characterize the energetically favored conformational modes of GEM by means of ab initio quantum mechanical studies with comparison to a novel X-ray crystallographic structure, and to determine the performance of ab initio quantum mechanical theory by comparison with X-ray structural data for GEM and 2'-deoxy cytosine (CYT). Another objective of this study was to attempt to determine key structural and electronic atomic interactions relating to the 2',2'-difluoro substitution in GEM by the application of ab initio quantum mechanical methods. To our knowledge, these are the first reported ab initio quantum mechanical geometry optimizations of nucleosides using large (e.g. 6-31G*) slit valence function basis sets. The development of accurate physicochemical models on a small scale enables us to extend our studies of GEM to more complex studies including DNA incorporation, deamination, ribonucleotide reductase inhibition, and triphosphorylation.


Subject(s)
Antimetabolites, Antineoplastic/chemistry , Cytosine/analogs & derivatives , Deoxycytidine/analogs & derivatives , Chemical Phenomena , Chemistry, Physical , Crystallography, X-Ray , Cytosine/chemistry , Deoxycytidine/chemistry , Models, Molecular , Gemcitabine
14.
Clin Cancer Res ; 2(4): 707-12, 1996 Apr.
Article in English | MEDLINE | ID: mdl-9816221

ABSTRACT

As part of a continuing program aimed at developing nonpolyglutamylatable inhibitors of dihydrofolate reductase that are less toxic and more specific in their action, we herein report the therapeutic efficacy and toxicity of gamma-methylene-10-deazaaminopterin (MDAM) in athymic nude mice bearing advanced human HCT-8 ileocecal xenografts and its antitumor activity in C57BL/6 x DBA/2 F1 (hereafter called B6D2F1) mice bearing P388 murine leukemia. For the xenograft study, MDAM was administered at the maximum tolerated dose by the following dose schedules: (a) 5-day continuous i.v. infusion at 1.0 mg/kg/day (schedule I); and (b) i.v. push, daily for 5 days at 50 mg/kg/day (schedule II). The maximum tolerated dose values for methotrexate (MTX) under these conditions were 0.2 and 1.0 mg/kg/day for schedule I and schedule II, respectively. MTX did not exhibit any significant antitumor activity in this model system by both schedules; however, MDAM induced complete responses of 13 and 25% and partial responses of 25 and 50% by schedules I and II, respectively. MDAM also exhibited antitumor activity significantly superior to that of MTX in the P388 tumor model. One of the enantiomers of MDAM, which possesses the natural configuration at the gamma-methyleneglutamate moiety (l-MDAM), has been shown to be a better inhibitor of human recombinant dihydrofolate reductase and H35 hepatoma cell growth than D,L-MDAM. L-MDAM inhibited the uptake of radiolabeled folinic acid to H35 hepatoma cells eight times more efficiently than MTX. The results indicate that the superior activity of MDAM relative to MTX may be partially due to a combination of enhanced transport to tumor cells and slower deactivation by aldehyde oxidase.


Subject(s)
Aminopterin/analogs & derivatives , Antineoplastic Agents/pharmacology , Folic Acid Antagonists/pharmacology , Polyglutamic Acid/metabolism , Aminopterin/metabolism , Aminopterin/pharmacology , Animals , Female , Humans , Leucovorin/pharmacokinetics , Methotrexate/pharmacology , Mice , Mice, Inbred DBA
15.
J Biomol Struct Dyn ; 10(4): 675-80, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8466672

ABSTRACT

Molecular modeling of the system composed of ethidium cation initially intercalated through the major and minor grooves of the B-DNA form of 5'GCATGC3' into the central AT pairs, with the phenyl group oriented either toward or away from the helical axis, led upon energy minimization to intercalation complexes only when the phenyl group was initially oriented away from the helical axis. For the other two cases, energy minimization led to extrusion of the ethidium from the helix to form unanticipated outside-bound complexes stabilized by specific hydrogen bonds. It is suggested that outside-bound major groove complexes involving the GXXXG sequence may play important roles in the reaction mechanism of ethidium intercalation and in its action as a drug.


Subject(s)
DNA/chemistry , Ethidium/metabolism , Base Composition , Base Sequence , Binding Sites , DNA/metabolism , Hydrogen Bonding , Models, Molecular , Nucleic Acid Conformation
16.
Anticancer Drug Des ; 5(2): 159-67, 1990 May.
Article in English | MEDLINE | ID: mdl-2357262

ABSTRACT

Molecular dynamics simulations of oligonucleoside methylphosphonates (MP) and naturally occurring oligodeoxynucleotides (ODN) as the third strand to double-stranded (ds) DNA targets predict that the third strand with MP backbone is more favorable to triple helix formation than the native phosphodiester ODN third strand. In contrast to experimental data, the calculated DNA conformations in both fully solvated triple helical system are found to be in hybrid A and B conformations. The calculations predict that a third strand with MP backbone is readily accommodated in the major groove of the ds DNA target, and adopts a different conformation from a helical triplet formed with native ODN as the third strand through the Hoogsteen base pairing scheme.


Subject(s)
Computer Simulation , DNA/metabolism , Models, Molecular , Oligonucleotides/metabolism , Organophosphorus Compounds/metabolism , Chemical Phenomena , Chemistry
17.
Anticancer Drug Des ; 4(4): 281-94, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2619866

ABSTRACT

The anti-cancer drug cyclophosphamide produces cytotoxic effects by DNA interstrand crosslink formation. Current knowledge of local physicochemical factors in DNA influencing crosslink formation in DNA has been based on physical models of Watson-Crick DNA. Specific interactions between DNA and cyclophosphamide metabolites determining formation and stabilization of interstrand crosslinks have been identified by advanced molecular computational methods. These results predict that the more favorable DNA sequence for interstrand crosslinking is 5'-GC-3' rather than the previously proposed 5'-CG-3' and that thymine methyl groups adjacent to guanine will inhibit interstrand crosslinking. In addition the conformational and energetic consequences of formamido-pyrimidine adducts and local water interactions with the drug-DNA complex were identified. These simulations of interstrand crosslinking provide a fundamental basis for understanding the mechanism of action of nitrogen mustard-type alkylating agents, and a rationale for the prospective design of more effective anti-tumor agents.


Subject(s)
Cyclophosphamide/metabolism , DNA/metabolism , Base Sequence , Models, Molecular , Nucleic Acid Conformation , Nucleic Acid Denaturation
18.
J Mol Graph ; 7(3): 160-4, 157-8, 1989 Sep.
Article in English | MEDLINE | ID: mdl-2488855

ABSTRACT

Ray tracing is an elegant and powerful technique for creating computer-generated images. The wide variety of geometric primitives and realistic effects, such as reflections, transparency, and shadows, make it one of the most popular rendering methods in use today. We present a brief introduction to ray tracing and discuss some of the computational issues involved. Examples illustrate ray tracing's effectiveness for producing high-quality visualizations of molecular structures.


Subject(s)
Computer Graphics , Molecular Structure , Models, Molecular
19.
Carcinogenesis ; 10(6): 1131-7, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2720906

ABSTRACT

The conformational and energetic consequences of cytosine methylation in eukaryotes related to transcription or formation of chromatin structure are not well understood. Structures of methylated and unmethylated DNA sequences from biologically relevant sources were studied by theoretical methods under different ionic conditions and demonstrate that cytosine methylation produces a localized pattern of steric, hydrophobic, energetic, conformational and electrostatic alterations in DNA. These findings suggest how this modification may influence protein-DNA interactions and support current hypotheses. The results reveal previously unrecognized potential effects of cytosine methylation which could critically affect normal and neoplastic cellular processes by altering transcriptional events, histone binding, chromosomal stability and cellular differentiation.


Subject(s)
DNA/genetics , Models, Molecular , Animals , Base Sequence , Cytosine , DNA/metabolism , Genes , Genes, Synthetic , Humans , Methylation , Models, Theoretical , Molecular Sequence Data , Nucleic Acid Conformation , Restriction Mapping , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...