Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Invest Ophthalmol Vis Sci ; 46(9): 3323-30, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16123436

ABSTRACT

PURPOSE: Polyamine analogues inhibit tumor growth in vitro and in vivo, and oligoamines with a chain length of 10, 12, or 14 are particularly potent. This study was conducted to investigate the effect of the decamines CGC-11144 and CGC-11150 in a mouse model of choroidal neovascularization (CNV). METHODS: Mice with laser-induced rupture of Bruch's membrane were given intraperitoneal, intravitreous, or periocular injection of CGC-11144, CGC-11150, or vehicle, and after 14 days, they were perfused with fluorescein-labeled dextran, and the area of CNV was measured on choroidal flatmounts by image analysis. In some groups of mice, treatments were started 7 days after rupture of Bruch's membrane to determine the effect of the agent on established CNV. Electroretinograms (ERGs) were performed to assess the effects on retinal function, and histopathology was used to evaluate retinal structure. RESULTS: Intraperitoneal injection of 10 or 20 mg/kg CGC-11144 or CGC-11150 resulted in small but significant reductions in the area of CNV. Intravitreous injection of 20 microg CGC-11144 or CGC-11150 on days 0 and 7 after rupture of Bruch's membrane resulted in a approximately 40% reduction in the area of CNV, with a similar reduction after periocular injections of 0.2 mg CGC-11144 three times a week for 2 weeks. Both intravitreous and periocular delivery of CGC-11144 also caused significant regression of established CNV. Within 2 days of periocular injection of CGC-11144, there was apoptosis in CNV lesions, but not in normal blood vessels or other retinal cells. Periocular injections of d,l-alpha-difluoromethyl-ornithine (DFMO), which decreases polyamine levels by a different mechanism, also inhibited CNV. There was no decline in ERG amplitudes or abnormal retinal morphology after daily injections of 0.2 mg CGC-11144 for 2 weeks, but a single intravitreous injection compromised retinal structure and function. CONCLUSIONS: Periocular delivery of the polyamine analogues may be a useful approach for the treatment of CNV.


Subject(s)
Biogenic Polyamines/therapeutic use , Choroidal Neovascularization/drug therapy , Animals , Apoptosis , Biogenic Polyamines/administration & dosage , Bruch Membrane/surgery , Choroidal Neovascularization/diagnosis , Choroidal Neovascularization/physiopathology , Dextrans , Disease Models, Animal , Drug Administration Routes , Eflornithine/therapeutic use , Electroretinography , Female , Fluorescein Angiography , Fluoresceins , In Situ Nick-End Labeling , Injections, Intraperitoneal , Laser Therapy , Mice , Mice, Inbred C57BL , Orbit , Retina/physiology , Vitreous Body
2.
Cancer Chemother Pharmacol ; 56(1): 83-90, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15791459

ABSTRACT

The recent cloning of the mammalian gene coding for N(1)-acetylpolyamine oxidase (PAO) provides the opportunity to directly examine the role of human PAO (hPAO) in polyamine homeostasis as well as its potential role in determining cellular response to antitumor polyamine analogues. To facilitate the study of this enzyme, the production, purification, and characterization of the recombinant hPAO is reported. hPAO oxidizes N(1)-acetylspermidine (K(m)=2.1 microM, K(cat)=15.0 s(-1)) and has very high affinity for N(1)-acetylspermine (K(m)=0.85 microM, K(cat)=31.7 s(-1)). The recombinant hPAO does not efficiently oxidize spermine, thereby demonstrating a significant difference in substrate specificity from the previously described human spermine oxidase PAOh1/SMO. Importantly, hPAO demonstrates the ability to oxidize a subset of antitumor polyamine analogues, suggesting that this oxidase activity could have a significant effect on determining tumor sensitivity to these or similar agents. Transfection of A549 human lung cancer cells with an hPAO-expressing plasmid leads to a profound decrease in sensitivity to those analogues which act as substrates, confirming its potential to alter drug response. One similarity that hPAO shares with human PAOh1/SMO, is that certain oligoamine analogues are potent inhibitors of its oxidase activity. The results of these studies demonstrate how changes in polyamine catabolism may affect drug response.


Subject(s)
Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/pharmacology , Polyamines/metabolism , Drug Resistance, Neoplasm , Homeostasis , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Oxidation-Reduction , Recombinant Proteins , Spermine/metabolism , Transfection , Tumor Cells, Cultured , Polyamine Oxidase
3.
Biochem J ; 384(Pt 2): 271-9, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15315476

ABSTRACT

Spermidine, spermine and putrescine are essential for mammalian cell growth, and there has been a pervasive effort to synthesize analogues of these polyamines that will disrupt their function and serve as tools to inhibit cell proliferation. Recently, we demonstrated that a number of such polyamine analogues are also capable of inducing the regulatory protein AZ (antizyme). In the present study the incorporation of a few sample analogues [mimics of bis(ethyl)spermine] was shown to be significantly limited by a decrease in the V(max) for the polyamine transport system in response to analogue-induced AZ. This creates an unusual circumstance in which compounds that are being designed for therapeutic use actually inhibit their own incorporation into targeted cells. To explore the impact of this feedback system, cultures of rat hepatoma HTC cells were pre-treated to exhibit either low or high polyamine uptake activity and then exposed to polyamine analogues. As predicted, regardless of initial uptake activity, all cultures eventually achieved the same steady-state levels of the cellular analogue and AZ. Importantly, analogue-induced AZ levels remained elevated with respect to controls even after the native polyamines were reduced by more than 70%. To model the insufficient AZ expression found in certain tumours, GS-CHO (GS Chinese-hamster ovary) cells were transfected to express high levels of exogenic AZI (AZ inhibitor). As anticipated, this clone incorporated significantly higher levels of the polyamine analogues examined. This study reveals a potential limitation in the use of polyamine-based compounds as therapeutics, and strategies are presented to either circumvent or exploit this elegant transport feedback system.


Subject(s)
Feedback, Physiological/physiology , Polyamines/metabolism , Proteins/physiology , Animals , CHO Cells/chemistry , CHO Cells/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carrier Proteins/genetics , Cells, Cultured , Cricetinae , Cricetulus , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Rats , Transfection , Tumor Cells, Cultured
4.
Mol Cancer Res ; 2(2): 81-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14985464

ABSTRACT

Several polyamine analogues have efficacy against a variety of epithelial tumor models including breast cancer. Recently, a novel class of polyamine analogues designated as oligoamines has been developed. Here, we demonstrate that several representative oligoamine compounds inhibit in vitro growth of human breast cancer MDA-MB-435 cells. The activator protein-1 (AP-1) transcriptional factor family members, c-Jun and c-Fos, are up-regulated by oligoamines in MDA-MB-435 cells, suggesting a possible AP-1-dependent induction of apoptosis. However, the use of a novel c-Jun NH(2)-terminal kinase (JNK) inhibitor, SP600125, suggests that inhibition of c-Jun activity sensitized tumor cells to oligoamine-induced cell death. To directly test this hypothesis, cells were stably transfected with the dominant-negative mutant c-Jun (TAM67), which lacks the NH(2)-terminal transactivation domain. Cells overexpressing TAM67 exhibit normal growth kinetics but demonstrate a significantly increased sensitivity to oligoamine cytotoxicity and attenuated colony formation after oligoamine treatment. Furthermore, oligoamine treatment leads to more profound caspase-3 activation and poly(ADP-ribose) polymerase cleavage in TAM67 transfectants, suggesting that c-Jun acts as an antiapoptosis factor in MDA-MB-435 cells in response to oligoamine treatment. These findings indicate that oligoamine-inducible AP-1 plays a prosurvival role in oligoamine-treated MDA-MB-435 cells and that JNK/AP-1 might be a potential target for enhancing the therapeutic efficacy of polyamine analogues in human breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Peptide Fragments/metabolism , Polyamines/chemistry , Polyamines/toxicity , Proto-Oncogene Proteins c-jun/metabolism , Anthracenes/pharmacology , Apoptosis/drug effects , Breast Neoplasms/genetics , Cell Division/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mutation/genetics , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins c-jun/genetics , Transfection , Tumor Cells, Cultured , Up-Regulation/drug effects
5.
J Med Chem ; 46(21): 4586-600, 2003 Oct 09.
Article in English | MEDLINE | ID: mdl-14521420

ABSTRACT

Polyamine analogues 7, 10, 18, 27, and 32 containing cyclopropane rings were obtained by chemical synthesis. Their antineoplastic activities were assessed against the cultured human prostate tumor cell lines DU-145, DuPro, and PC-3. Decamines 32 and 27 exhibited variable levels of cytotoxicity against all three cell lines, while 7, 10, and 18 were efficacious against DU-145 and DuPro. Maximum tolerated doses (MTD) for all five compounds in a NCr-nu mouse model were determined at dosing schedules of q1d x 5 (ip) in two cycles with a break of 10 days between cycles. Their antitumor efficacies were then tested against DU-145 tumor xenografts in mice treated with all five agents at their respective MTDs. In addition, the efficacies of 7 and 10 against the same tumor xenograft were assessed at doses below their respective MTDs. In all experiments, administration began two weeks after tumor implantation. All compounds efficiently inhibited tumor growth for up to 50 days postimplantation, with negligible animal body weight loss. Tetramine 10 and hexamine 18 were the most efficient among the five analogues in arresting tumor growth. Tetramine 10 containing two cyclopropane rings had the lowest systemic toxicity as reflected in animal body weight loss. It was further assessed at a weekly administration regimen of (q1w x 4) in two cycles with a four-week break between the cycles. At this dosing schedule, 10 again efficiently arrested tumor growth with negligible effect on animal body weight. Tetramine 10 also arrested the growth of large tumors (ca. 2000 mm(3)) treated 66 days postimplantation. Studies on the metabolism of 10 showed that it accumulates in tumor within 6 h after the end of administration and reached a maximum level 72 h after cessation of dosing. Intracellular concentrations of 10 in liver and kidney were much smaller when compared to those in the tumor when measured 72 h after cessation of dosing. In liver and kidney, the deethyl metabolites of 10 accumulated over a 96 h period after cessation of dosing.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cyclopropanes/chemistry , Polyamines/chemical synthesis , Polyamines/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Body Weight , Cell Division/drug effects , Humans , Indicators and Reagents , Kidney/metabolism , Liver/metabolism , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Nude , Neoplasm Transplantation , Polyamines/pharmacokinetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
6.
Bioorg Med Chem ; 11(18): 4121-31, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12927874

ABSTRACT

alpha N,(omega)N-bis(ethyl) octamine SL-11160, decamine SL-11159, dodecamine SL-11226, and tetradecamine SL-11175 were chemically synthesized. We called this class of compounds 'oligoamines'. In these compounds, each -NH(2)(+) residue is separated by four CH(2) residues. trans-Unsaturation was also introduced into the center of the oligoamine chain resulting in the trans-octamine SL-11158, trans-decamine SL-11144, trans-dodecamine SL-11172 and trans-tetradecamine SL-11227. cis-Unsaturation gave the cis-octamine SL-11157 and cis-decamine SL-11150. When assayed for their growth inhibitory effect against four human prostate cancer cell lines LnCap, DU-145, DuPro, and PC-3 by a MTT assay, the ID(50) values were less than 1 microM in all four cell lines. On day 6 of treatment, 2 microM SL-11159, SL-11144 and SL-11175 killed over five logs of DuPro cells while SL-11172 killed over four logs as determined by a colony forming efficiency (CFE) assay. In addition, SL-11159, SL-11226 and SL-11227 killed four logs of PC-3 cells. PC-3 cells are generally resistant to shorter chain polyamine analogues. Such a level of cytotoxicity in any of the prostate tumor cell lines has not been observed for any other polyamine analogues tested thus far. The DU-145 cell line was too sensitive to oligoamines to perform a CFE analysis and the DuPro cell line was too sensitive to SL-11227 treatment to obtain reproducible CFE data. Interestingly, all 10 oligoamines were efficient DNA aggregators in a cell-free system and their cytotoxicities generally parallel their capacities to aggregate DNA.


Subject(s)
Antineoplastic Agents/chemical synthesis , Propylamines/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Line, Tumor/drug effects , Cell Survival , DNA/chemistry , DNA/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Propylamines/pharmacology , Prostatic Neoplasms , Stereoisomerism , Structure-Activity Relationship , Time Factors
7.
Clin Cancer Res ; 9(7): 2769-77, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12855657

ABSTRACT

Polyamine analogs have demonstrated considerable activity against many important solid tumor models including breast cancer. However, the precise mechanisms of antitumor activities of polyamine analogs are not entirely understood. The cytotoxicity of a newly developed polyamine analog compound, SL11144, against human breast cancer was assessed. Treatment of human breast cancer cell lines in culture with SL11144 decreased cell proliferation and induced programmed cell death in a time- and dose-dependent manner. SL11144 also profoundly inhibited the growth of MDA-MB-231 xenografts in host nude mice without overt toxic effects. Treatment of MDA-MB-435 cells with SL11144 led to the release of cytochrome c from mitochondria into cytosol, activation of caspase-3, and poly(ADP-ribose) polymerase cleavage. SL11144 decreased Bcl-2 and increased Bax protein levels in MDA-MB-231 cells. Furthermore, activator protein 1 transcriptional factor family member c-Jun was up-regulated by SL11144 in MDA-MB-435 and MDA-MB-231 cells, but not in MCF7 cells. In addition, significant inhibition of ornithine decarboxylase activity and a decrease in polyamine pools were demonstrated. These results demonstrate that the novel polyamine analog SL11144 has effective antineoplastic action against human breast cancer cells in vitro and in vivo and that multiple apoptotic mechanisms are associated with its cytotoxic effect in specific human breast cancer cell lines.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Polyamines/chemistry , Polyamines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Caspase 3 , Caspases/metabolism , Cell Death , Cell Division , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival , Chromatin/metabolism , Cytochromes c/metabolism , Cytoplasm/metabolism , Cytosol/metabolism , DNA Damage , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Models, Chemical , Neoplasm Transplantation , Nucleosomes/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Up-Regulation , bcl-2-Associated X Protein
8.
Cancer Chemother Pharmacol ; 52(5): 383-90, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12827295

ABSTRACT

PURPOSE: The induction of polyamine catabolism has been directly associated with the cytotoxic response of various tumor types to the antitumor polyamine analogues. Initially, human polyamine catabolism was assumed to be under the control of a rate-limiting spermidine/spermine N1-acetyltransferase (SSAT) that provides substrate for an acetylpolyamine oxidase (PAO). We have recently cloned a new polyamine analogue-inducible human polyamine oxidase (PAOh1/SMO) that efficiently uses spermine as a substrate. The induction of PAOh1/SMO in response to multiple polyamine analogues was examined in representative lung tumor cell lines. METHODS: Representatives of three different classes of antitumor polyamine analogues were examined for their ability to induce PAOh1/SMO. RESULTS: The human adenocarcinoma line, NCI A549 was found to be the most responsive line with respect to induction of PAOh1/SMO in response to analogue exposure. Similar to previous observations with SSAT expression, PAOh1/SMO induction was found to occur primarily in non-small-cell lung cancers cell lines. Using a series of polyamine analogues, it was found that the most potent inducers of PAOh1/SMO possessed multiple three-carbon linkers between nitrogens, as typified by N1,N11-bis(ethyl)norspermine. CONCLUSIONS: Since PAOh1/SMO is an analogue-inducible enzyme that produces H2O2 as a metabolic product, it may play a significant role in determining the sensitivity of various human tumors to specific polyamine analogues.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Small Cell/enzymology , Lung Neoplasms/enzymology , Oxidoreductases Acting on CH-NH Group Donors/biosynthesis , Polyamines/pharmacology , Acetyltransferases/metabolism , Blotting, Northern , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Humans , Polyamines/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Substrate Specificity , Time Factors , Polyamine Oxidase
9.
Cancer Chemother Pharmacol ; 51(6): 488-92, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12695854

ABSTRACT

PURPOSE: We tested the polyamine analog SL-11093 (3,8,13,18-tetraaza-10,11-[(E)-1,2-cyclopropyl]eicosane tetrahydrochloride) as an effective chemotherapeutic agent against human prostate cancer grown in nude mice. METHODS: NCr-nu mice grafted with DU-145 human prostate tumor cells were treated i.p. with SL-11093 at 50 mg/kg q1dx5 for either three or five cycles separated by intervals of about 10-15 days. RESULTS: In treated animals, tumor growth remained arrested for up to 100 days with minimal animal weight loss. None of the animals died during the treatment and in one experiment two out of six animals showed no palpable tumor. SL-11093 was readily taken up by the tumors, where its levels remained elevated for about 48 h after the end of drug administration. In liver and in kidney, SL-11093 (a (alpha)N,(omega)N-bisethyl derivative) was oxidatively N-deethylated predominantly to its monoethyl and di-deethyl derivatives. In time, the monoethyl derivative was further dealkylated, with a loss of an aminobutyl chain to form an aminomethyl cyclopropyl derivative. In tumor (and in lung), N-dealkylation reactions were less evident. CONCLUSION: SL-11093 is an effective chemotherapeutic agent against a human prostate tumor xenograft grown in nude mice. The drug accumulation and slow metabolism in tumor compared to other tissues would most likely reduce systemic toxicity of the drug and contribute to a larger therapeutic window for SL-11093 as compared to other cytotoxic polyamine analogs.


Subject(s)
Antineoplastic Agents/pharmacology , Polyamines/pharmacology , Prostatic Neoplasms/drug therapy , Acetyltransferases/metabolism , Animals , Biogenic Polyamines/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Ornithine Decarboxylase/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , S-Adenosylmethionine/metabolism , Tissue Distribution , Transplantation, Heterologous , Tumor Cells, Cultured
10.
Biochem J ; 366(Pt 2): 663-71, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-11972449

ABSTRACT

The polyamines spermidine and spermine and their diamine precursor putrescine are essential for mammalian cell growth and viability, and strategies are sought for reducing polyamine levels in order to inhibit cancer growth. Several structural analogues of the polyamines have been found to decrease natural polyamine levels and inhibit cell growth, probably by stimulating normal feedback mechanisms. In the present study, a large selection of spermine analogues has been tested for their effectiveness in inducing the production of antizyme, a key protein in feedback inhibition of putrescine synthesis and cellular polyamine uptake. Bisethylnorspermine, bisethylhomospermine, 1,19-bis-(ethylamino)-5,10,15-triazanonadecane, longer oligoamine constructs and many conformationally constrained analogues of these compounds were found to stimulate antizyme synthesis to different levels in rat liver HTC cells, with some producing far more antizyme than the natural polyamine spermine. Uptake of the tested compounds was found to be dependent on, and limited by, the polyamine transport system, for which all these have approximately equal affinity. These analogues differed in their ability to inhibit HTC cell growth during 3 days of exposure, and this ability correlated with their antizyme-inducing potential. This is the first direct evidence that antizyme is induced by several polyamine analogues. Selection of analogues with this potential may be an effective strategy for maximizing polyamine deprivation and growth inhibition.


Subject(s)
Polyamines/pharmacology , Protein Biosynthesis , Proteins , Animals , Cell Division/drug effects , Cell Survival/drug effects , Enzyme Induction/drug effects , Kinetics , Liver/enzymology , Liver Neoplasms, Experimental , Ornithine Decarboxylase Inhibitors , Polyamines/chemical synthesis , Polyamines/chemistry , Rats , Spermine/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Antimicrob Agents Chemother ; 46(1): 55-61, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11751111

ABSTRACT

Microsporidia are eukaryotic obligate intracellular protists that are emerging pathogens in immunocompromised hosts, such as patients with AIDS or patients who have undergone organ transplantation. We have demonstrated in vitro and in vivo that synthetic polyamine analogs are effective antimicrosporidial agents with a broad therapeutic window. CD8-knockout mice or nude mice infected with the microsporidian Encephalitozoon cuniculi were cured when they were treated with four different novel polyamine analogs at doses ranging from 1.25 to 5 mg/kg of body weight/day for a total of 10 days. Cured animals demonstrated no evidence of parasitemia by either PCR or histologic staining of tissues 30 days after untreated control animals died.


Subject(s)
AIDS-Related Opportunistic Infections/drug therapy , Microsporidiosis/drug therapy , Polyamines/therapeutic use , Animals , Disease Models, Animal , Encephalitozoon cuniculi/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Parasitic Sensitivity Tests , Polyamines/adverse effects , Polyamines/chemistry , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL