Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
J Thromb Haemost ; 12(11): 1788-800, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25196897

ABSTRACT

BACKGROUND: Routine prophylaxis with replacement factor VIII (FVIII) - the standard of care for severe hemophilia A - often requires frequent intravenous infusions (three or four times weekly). An FVIII molecule with an extended half-life could reduce infusion frequency. The A-LONG study established the safety, efficacy and prolonged pharmacokinetics of recombinant FVIII Fc fusion protein (rFVIIIFc) in previously treated adolescents and adults with severe hemophilia A. OBJECTIVE: In this post hoc analysis, we investigated the relationship between subjects' prestudy (FVIII) and on-study (rFVIIIFc) regimens. METHODS: We analyzed two subgroups of subjects: prior prophylaxis and on-study individualized prophylaxis (n = 80), and prior episodic treatment and on-study weekly prophylaxis (n = 16). Subjects' prestudy dosing regimens and bleeding rates were compared with their final rFVIIIFc regimens and annualized bleeding rates (ABRs) in the last 3 months on-study. Dosing regimen simulations based on population pharmacokinetics models for rFVIII and rFVIIIFc were performed. RESULTS: As compared with their prestudy regimen, 79 of 80 (98.8%) subjects on individualized rFVIIIFc prophylaxis decreased their infusion frequency. Overall ABRs were low, with comparable factor consumption. Longer dosing intervals, including 5-day dosing, were associated with higher baseline von Willebrand factor antigen levels. Simulated dosing regimens predicted a greater proportion of subjects with steady-state FVIII activity trough levels of ≥ 1 IU dL(-1) (1%) with rFVIIIFc than with equivalent rFVIII regimens. CONCLUSION: These results suggest that patients on rFVIIIFc prophylaxis can reduce their infusion frequency as compared with their prior FVIII regimen while maintaining low bleeding rates, affording more patients trough levels of ≥ 1 IU dL(-1) than with rFVIII products requiring more frequent dosing regimens.


Subject(s)
Coagulants/administration & dosage , Factor VIII/administration & dosage , Hemophilia A/drug therapy , Hemorrhage/prevention & control , Hemostasis/drug effects , Immunoglobulin Fc Fragments/administration & dosage , Recombinant Fusion Proteins/administration & dosage , von Willebrand Factor/metabolism , Adolescent , Adult , Biomarkers/blood , Coagulants/adverse effects , Coagulants/blood , Coagulants/pharmacokinetics , Computer Simulation , Drug Administration Schedule , Drug Monitoring , Factor VIII/adverse effects , Factor VIII/pharmacokinetics , Half-Life , Hemophilia A/blood , Hemophilia A/diagnosis , Hemorrhage/chemically induced , Humans , Immunoglobulin Fc Fragments/adverse effects , Immunoglobulin Fc Fragments/blood , Infusions, Intravenous , Male , Models, Biological , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/pharmacokinetics , Severity of Illness Index , Time Factors , Treatment Outcome , Young Adult
2.
Hum Reprod ; 20(7): 1805-13, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15817590

ABSTRACT

BACKGROUND: The alpha and beta subunits of FSH were fused to the Fc domain of IgG1 either in a single chain or a heterodimer format. These molecules were absorbed through the epithelium in lung and intestine by neonatal Fc receptor (FcRn)-mediated transcytosis. METHODS AND RESULTS: Single chain and heterodimer FSH-Fc were made recombinantly in Chinese hamster ovary cells. Treatment of rats with a single s.c. dose of single chain or heterodimer FSH-Fc resulted in greater stimulation of ovarian weight (20.8+/-3.9 and 26.9+/-6.1 mg respectively) compared to those receiving vehicle (12.1+/-1.0 mg) or an equimolar dose of recombinant human FSH (14.3+/-1.7 mg). Both FSH-Fc fusion proteins were absorbed after oral dosing of newborn rats with long terminal half-lives of approximately 60 h, and pulmonary delivery in four cynomolgus monkeys produced maximum serum concentrations between 69 and 131 ng/ml with long terminal half-lives between 55 and 210 h. Serum inhibin levels increased after pulmonary dosing with single chain FSH-Fc (1.3- and 1.4-fold) and heterodimer FSH-Fc (5.9- and 7.1-fold) and remained elevated for >12 days after treatment with heterodimer FSH-Fc. CONCLUSIONS: We have shown that FSH-Fc fusion proteins have increased stability in blood and improved bioactivity in vivo, and that heterodimer FSH-Fc is more active in rats and monkeys than single chain FSH-Fc. These data suggest that Fc fusion proteins offer the potential for oral and pulmonary delivery of FSH.


Subject(s)
Follicle Stimulating Hormone/administration & dosage , Immunoglobulin Fc Fragments/administration & dosage , Administration, Oral , Aerosols , Animals , Animals, Newborn , Base Sequence , Biological Transport, Active , CHO Cells , Cricetinae , DNA, Complementary/genetics , Female , Follicle Stimulating Hormone/pharmacokinetics , Humans , Immunoglobulin Fc Fragments/metabolism , Macaca fascicularis , Male , Organ Size/drug effects , Ovary/anatomy & histology , Ovary/drug effects , Rats , Receptors, Fc/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Testis/anatomy & histology , Testis/drug effects
3.
J Med Chem ; 44(4): 524-30, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11170642

ABSTRACT

Cyclin-dependent kinases (CDKs) are regulatory proteins of the eukaryotic cell cycle. They act after association with different cyclins, the concentrations of which vary throughout the progression of the cell cycle. As central mediators of cell growth, CDKs are potential targets for inhibitory molecules that would allow disruption of the cell cycle in order to evoke an antiproliferative effect and may therefore be useful as cancer therapeutics. We synthesized several inhibitory 2,6,9-trisubstituted purine derivatives and solved the crystal structure of one of these compounds, H717, in complex with human CDK2 at 2.6 A resolution. The orientation of the C2-p-diaminocyclohexyl portion of the inhibitor is strikingly different from those of similar moieties in other related inhibitor complexes. The N9-cyclopentyl ring fully occupies a space in the enzyme which is otherwise empty, while the C6-N-aminobenzyl substituent points out of the ATP-binding site. The structure provides a basis for the further development of more potent inhibitory drugs.


Subject(s)
Adenine/chemistry , CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/chemistry , Enzyme Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Adenine/analogs & derivatives , Crystallography, X-Ray , Cyclin-Dependent Kinase 2 , Humans , Models, Molecular , Molecular Structure
4.
Circulation ; 100(6): 659-65, 1999 Aug 10.
Article in English | MEDLINE | ID: mdl-10441105

ABSTRACT

BACKGROUND: Smooth muscle cell (SMC) proliferation is a critical component of neointimal formation in many models of vascular injury and in human lesions as well. Cell-cycle inhibition by gene transfer techniques can block SMC proliferation and lesion formation in animal models, although these methods are not yet applicable to the treatment of human disease. Flavopiridol is a recently identified, potent, orally available cyclin-dependent kinase inhibitor. METHODS AND RESULTS: Using human aortic SMCs, we found that flavopiridol in concentrations as low as 75 nmol/L resulted in nearly complete inhibition of basic fibroblast growth factor-induced and thrombin-induced proliferation. At this dose, flavopiridol inhibited cyclin-dependent kinase activity, as measured by histone H1 phosphorylation, but had no effect on mitogen-activated protein kinase activation. Induction of the cell cycle-related proteins cyclin D1, proliferating cell nuclear antigen, and phosphorylated retinoblastoma protein was also blocked by flavopiridol. Flavopiridol had no effect on cellular viability. To test whether flavopiridol had a similar activity in vivo when administered orally, we examined neointimal formation in rat carotid arteries after balloon injury. Flavopiridol 5 mg/kg reduced neointimal area by 35% and 39% at 7 and 14 days, respectively, after injury. CONCLUSIONS: Flavopiridol inhibits SMC growth in vitro and in vivo. Its oral availability and selectivity for cyclin-dependent kinases make it a potential therapeutic tool in the treatment of SMC-rich vascular lesions.


Subject(s)
Carotid Artery Injuries , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Growth Inhibitors/pharmacology , Muscle Proteins/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Neovascularization, Physiologic/drug effects , Piperidines/pharmacology , Wound Healing/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Carotid Arteries/drug effects , Carotid Arteries/enzymology , Carotid Arteries/pathology , Catheterization/adverse effects , Cell Cycle/drug effects , Cell Division/drug effects , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Humans , Hyperplasia , Male , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rats , Rats, Sprague-Dawley , Thrombin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Tunica Intima/drug effects , Tunica Intima/enzymology , Tunica Intima/injuries , Tunica Intima/pathology
5.
J Steroid Biochem Mol Biol ; 58(1): 21-30, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8809183

ABSTRACT

Steroidal antiestrogens appear to have at least two major modes of action in breast cancer cells, direct antagonism of estrogen binding to its receptor and depletion of estrogen receptors (ER) due to inhibition of dimerization of the receptor and a resultant destabilization of the receptor protein. In a search for other classes of compounds which would act as dimerization inhibitors, a novel substituted indole (8-{2-[1-(4-chlorobenzoyl)-5-hydroxy-2-methyl-1H-indol-3-yl]-acetylamino} octanoic acid butyl-methyl amide, MDL 101,906) was synthesized. Binding of the ER to its consensus response element (ERE) was apparently decreased in nuclear extracts from MCF-7 human breast cancer cell treated with MDL 101,906. This decreased binding was found to be due to depletion of ER based on direct measurement of ER using an enzyme-linked immunoassay. Other transcription factors were apparently unaffected by MDL 101,906 treatment. Whereas depletion of ER with a steroidal antiestrogen was almost complete after 3 h of treatment of MCF-7 cells, the effect of MDL 101,906 took significantly longer to occur, suggesting a fundamental difference in the mechanisms of action of the two drugs. This was also evident in the lack of binding of MDL 101,906 to the hormone binding domain of ER. MDL 101,906 treatment also caused depletion of ER mRNA in MCF-7 cells. Depletion of ER mRNA was noted by 3 h of drug treatment and was apparently almost complete after 24 h of treatment. Depletion of ER from MCF-7 cells led to a dose-dependent decrease in the expression of luciferase by an ERE-driven luciferase reporter gene assay system. The mechanism of MDL 101,906 appears to be unique and additional studies with this chemical class seem to be warranted to assess the potential for therapeutic utility.


Subject(s)
Breast Neoplasms/chemistry , Estrogen Antagonists/pharmacology , Indoles/pharmacology , Receptors, Estrogen/analysis , Base Sequence , Binding, Competitive , Breast Neoplasms/metabolism , Cell Extracts , Cell Nucleus , DNA/metabolism , Dose-Response Relationship, Drug , Estradiol/analogs & derivatives , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Antagonists/chemical synthesis , Gene Expression , Genes, Reporter/genetics , Humans , Indoles/chemical synthesis , Molecular Sequence Data , Polyunsaturated Alkamides , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Receptors, Estradiol/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Time Factors , Transcription Factors/metabolism , Tumor Cells, Cultured
6.
Cell Growth Differ ; 7(3): 351-9, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8838865

ABSTRACT

We have isolated a variant of the MCF-7 human breast tumor that is characterized by a hormone-independent, yet hormone-responsive, phenotype. This tumor, designated MCF-WES, was derived from MCF-7 tumor cells implanted in the mammary fat pad of a nude mouse in the absence of estradiol supplementation. MCF-WES tumors remain responsive to estradiol; however, unlike the parental MCF-7 tumors, they are stimulated to grow by tamoxifen. Additionally, MCF-WES cells are resistant to the pure steroidal antiestrogen, ICI 182,780. To our knowledge, a tumor with this combination of properties has not yet been described. Nuclear estrogen receptor (ER) levels in MCF-WES cells were 10% of those for MCF-7 under steroid-depleted conditions. MCF-WES tumor ER levels were 32% of those in MCF-7 tumors. Similarly, in vivo expression of ER mRNA for MCF-WES was 20% of levels determined for MCF-7. Further characterization of MCF-WES cells showed that they have increased levels of AP-1 DNA-binding activity. The marked increase in AP-1 binding activity may act to bypass the hormone dependence that is a characteristic of MCF-7 cells. It is also probable that the increase in AP-1 binding activity is responsible for the finding that MCF-WES cells secrete greater quantities of metalloproteinase activity in comparison to parental MCF-7 cells, suggesting progression to a more invasive, malignant phenotype. More complete characterization of this new cell line will help elucidate hormone-independent breast cancer and possibly identify targets for therapy.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , DNA, Neoplasm/metabolism , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Transcription Factor AP-1/metabolism , Animals , Cell Division , DNA, Neoplasm/analysis , Drug Resistance, Neoplasm , Estradiol/pharmacology , Fulvestrant , Gelatinases/metabolism , Humans , Mice , Mice, Nude , Phenotype , Protein Binding , RNA, Messenger/analysis , Receptors, Estrogen/analysis , Receptors, Estrogen/genetics , Receptors, Progesterone/analysis , Tamoxifen/pharmacology , Tumor Cells, Cultured
7.
Biochem Biophys Res Commun ; 204(1): 264-72, 1994 Oct 14.
Article in English | MEDLINE | ID: mdl-7945369

ABSTRACT

A correlation between changes in protein kinase C (PKC) activity and tumor metastasis has been reported previously with several murine tumor cell lines. Treatment of a human metastatic melanoma cell line, M24met, with phorbol ester, phorbol-12-myristate-13-acetate (PMA), followed by injection into the tail vein of scid mice doubled pulmonary metastasis. Adhesion of M24met cells exposed to PMA, was enhanced to collagens I and IV, but not to laminin or fibronectin, suggesting a change in specific adhesion receptors on the tumor cells. Treatment of M24met cells with PMA did not affect de novo synthesis of integrin subunits (alpha 2, alpha 3, beta 1) known to form collagen receptors. However, PMA stimulated the phosphorylation of integrin subunits alpha 3 and beta 1 on serine. Therefore, PMA effects on metastasis and cell adhesion may occur through PKC-mediated phosphorylation of integrins.


Subject(s)
Cell Adhesion , Integrins/metabolism , Lung Neoplasms/secondary , Melanoma/pathology , Melanoma/physiopathology , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Amino Acids/analysis , Animals , Cell Line , Collagen , Fibronectins , Humans , Laminin , Lung Neoplasms/pathology , Melanoma/enzymology , Mice , Mice, SCID , Phosphorylation , Transplantation, Heterologous , Tumor Cells, Cultured
8.
Cancer Res ; 54(6): 1485-90, 1994 Mar 15.
Article in English | MEDLINE | ID: mdl-8137252

ABSTRACT

(E)-2'-Deoxy-2'-(fluoromethylene)cytidine (MDL 101,731) is a mechanism-based inhibitor of ribonucleoside diphosphate reductase (J. Stubbe, personal communication), an enzyme involved in DNA synthesis and therefore a potential target for cancer chemotherapy. In the present report, we show that MDL 101,731 inhibits the proliferation of several human breast cancer cell lines, including the estrogen-dependent cell line, MCF-7, and the estrogen-independent cell lines MDA-MB-231, MDA-MB-468, and MDA-MB-435 in vitro at nanomolar concentrations (50% inhibitory concentration, 15-26 nM). Administration of MDL 101,731 caused marked regression of tumors which formed after s.c. inoculation of all four of the cell lines in athymic (nude) mice. MDA-MB-231 tumors were found to be most sensitive to MDL 101,731 with a 90-100% cure rate at doses of MDL 101,731 between 2 and 20 mg/kg, given as once daily i.p. injections, 5 days/week for as little as 3 weeks. Almost complete cessation of MDA-MB-231 tumor growth was obtained with a dose of 0.5 mg/kg MDL 101,731 following the same dosing regimen. MDA-MB-468, MDA-MB-435, and MCF-7 tumors were not as sensitive as MDA-MB-231, but tumor regression of 50, 65, and 80%, respectively, was obtained after 5-6 weeks of treatment. The effects of MDL 101,731 on spontaneous metastasis of MDA-MB-435 cells from the mammary fat pad to the lung was also examined, and it was found that the number of lung metastases was significantly decreased if mice received MDL 101,731 while the primary tumors were growing and after primary tumors were surgically excised. Additionally, preliminary evidence raises the possibility that MDL 101,731 may induce apoptosis in MDA-MB-231 tumors. Our data suggest that the use of MDL 101,731 for the treatment of breast cancer and possibly other solid tumors should be pursued.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Deoxycytidine/analogs & derivatives , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/enzymology , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Animals , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Death/physiology , Cell Division/drug effects , Deoxycytidine/pharmacology , Estrogens , Female , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/pathology , Transplantation, Heterologous , Tumor Cells, Cultured/drug effects
9.
Cancer Res ; 52(5): 1195-200, 1992 Mar 01.
Article in English | MEDLINE | ID: mdl-1737379

ABSTRACT

Phorbol esters which activate protein kinase C (PKC) have been shown to enhance experimental lung metastasis. Therefore, it was reasoned that inhibitors of PKC might also modulate metastasis. We have investigated this possibility using a PKC inhibitor, MDL 27,032 [4-propyl-5(4-pyridinyl)-2(3H)-oxazolone], as well as staurosporine and H-7. Treatment of B16F1 murine melanoma cells with MDL 27,032 for 24 h in culture and subsequent i.v. injection of the cells into mice resulted in greater than 90% inhibition of lung metastasis. Inhibition of metastasis was time dependent, with 90% of maximum inhibition occurring by 8 h of incubation. The 50% inhibitory concentration (IC50) for inhibition of metastasis with MDL 27,032 was 7 microM, a value similar to that for the inhibition of B16F1 membrane-associated PKC (IC50 = 13 microM) but not cytosolic PKC (IC50 = 54 microM). B16F1 cells treated with MDL 27,032 for 24 h were less adherent than untreated cells to extracellular matrix/basement membrane proteins. Adhesion to fibrinogen and collagen IV was inhibited (IC50 = 6 microM and 48 microM, respectively) by MDL 27,032, whereas adherence to laminin and fibronectin was not affected, indicating that the drug affects specific adhesion molecules. MDL 27,032-treated cells were also found to be less adherent than untreated cells to human umbilical vein endothelial cells. The phosphorylation of an 80-kDa B16F1 cell plasma membrane protein was stimulated under conditions known to stimulate PKC activity, and MDL 27,032 inhibited this phosphorylation in a dose-dependent manner. MDL 27,032 was more potent than H-7 for the inhibition of metastasis but was significantly less potent than staurosporine. These results support the hypothesis that there is a critical role for PKC-mediated phosphorylation of cell surface adhesion receptors in metastasis.


Subject(s)
Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/prevention & control , Melanoma, Experimental/secondary , Oxazoles/pharmacology , Oxazolone/analogs & derivatives , Protein Kinase C/antagonists & inhibitors , Pyridines/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , Alkaloids/pharmacology , Animals , Cell Adhesion/drug effects , Isoquinolines/pharmacology , Lung Neoplasms/enzymology , Melanoma, Experimental/enzymology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Phosphorylation , Piperazines/pharmacology , Staurosporine , Time Factors
10.
J Med Chem ; 34(2): 569-74, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1995879

ABSTRACT

A series of novel tetraamines of the general formula RNH(CH2)xNH(CH2)yNH(CH2)xNHR was synthesized and examined for activity against growth of Plasmodium falciparum in vitro. Within the series, dibenzyl analogues (R = benzyl) were found to be the most effective growth inhibitors, with IC50 values of about 10(-6) M. Further modifications of the tetraamine provided the optimum chain length for antimalarial activity of y = 7, x = 3. Compound 8 (MDL 27,695) with the structure y = 7, x = 3, R = benzyl, in combination with the ornithine decarboxylase inhibitor alpha-(difluoromethyl)ornithine, resulted in radical cures when tested against experimental Plasmodium berghei infections in mice. The structure-activity relationships of the series are discussed.


Subject(s)
Antimalarials/chemical synthesis , Polyamines/chemical synthesis , Animals , Antimalarials/therapeutic use , Chemical Phenomena , Chemistry , Malaria/drug therapy , Mice , Plasmodium falciparum/drug effects , Polyamines/therapeutic use , Structure-Activity Relationship
11.
J Med Chem ; 33(5): 1369-75, 1990 May.
Article in English | MEDLINE | ID: mdl-2329556

ABSTRACT

A series of tetraamines derived from 1,8-diaminooctane was prepared and tested as antitumor agents. The reaction of 1,8-diaminooctane with acrylonitrile gave N,N'-bis(cyanoethyl)-1,8-diaminooctane, which was reduced to tetraamine 20. Alkylation of the terminal nitrogen atoms of the tetra-Boc derivative of this compound by methyl or ethyl halide followed by removal of the Boc groups gave the bis(alkyl)polyamines 26a and 26b, respectively. These three compounds exhibit promising antitumor activity in the mouse L1210 leukemia model. Coadministration of a polyamine oxidase inhibitor potentiated the antitumor activity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Polyamines/chemical synthesis , Animals , Chemical Phenomena , Chemistry , Leukemia L1210/drug therapy , Mice , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Polyamines/therapeutic use , Structure-Activity Relationship , Polyamine Oxidase
12.
Article in English | MEDLINE | ID: mdl-2399280
13.
J Biol Chem ; 265(1): 382-8, 1990 Jan 05.
Article in English | MEDLINE | ID: mdl-2294109

ABSTRACT

N,N'-Bis(benzyl)polyamine analogs were found to be substrates for highly purified polyamine oxidase. Metabolism of these analogs was apparently dependent on molecular O2 and resulted in the formation of benzaldehyde, H2O2, and a polyamine analog with free terminal amines. The debenzylation reaction was optimal between pH 9 and 10, identical to the pH optimum for polyamine oxidase activity when N1-acetylspermine was used as the substrate. On a molecular sieve column the debenzylating activity co-eluted with N1-acetylspermine oxidizing activity, at an apparent molecular mass of approximately 65 kDa. The purified enzyme also appeared to have a molecular mass of approximately 65 kDa on sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Debenzylation of the bis(benzyl)polyamines was competitively inhibited by N1-acetylspermine and N1-acetylspermidine. The specific irreversible inhibitor of polyamine oxidase, N1,N4-bis(buta-2,3-dienyl)butanediamine also inhibited the debenzylation, whereas inhibitors of diamine and monoamine oxidases did not. The evolution of benzaldehyde from bis(benzyl)polyamine analogs by polyamine oxidase allowed the development of a simple rapid spectrophotometric assay for use in the measurement of polyamine oxidase activity in partially purified tissue or cell extracts. Further, metabolism of a bis(benzyl)polyamine analog by polyamine oxidase was found to be an important element in the growth inhibitory properties of the compound in a mouse model of malaria.


Subject(s)
Liver/enzymology , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Polyamines/metabolism , Animals , Antimalarials , Benzaldehydes/metabolism , Chromatography, High Pressure Liquid , Erythrocytes/metabolism , Hydrogen Peroxide/metabolism , Hydrogen-Ion Concentration , Male , Mice , Molecular Weight , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Polyamines/pharmacokinetics , Rats , Rats, Inbred Strains , Substrate Specificity , Polyamine Oxidase
15.
Proc Natl Acad Sci U S A ; 86(2): 651-5, 1989 Jan.
Article in English | MEDLINE | ID: mdl-2463635

ABSTRACT

A number of bis(benzyl)polyamine analogs were found to be potent inhibitors of both chloroquine-resistant and chloroquine-sensitive strains of the human malaria parasite Plasmodium falciparum in vitro (IC50 values = 0.2-14 microM). Administration of one of the compounds, MDL 27695, which is N,N'-bis(3-[(phenylmethyl)amino]propyl)-1,7-diaminoheptane (C6H5CH2NH(CH2)3NH(CH2)7NH(CH2)3NHCH2C6H5), at 10-15 mg/kg i.p. three times per day for 3 days in combination with 2% alpha-difluoromethylornithine (DFMO; eflornithine) in drinking water effected cures of 47/54 mice infected with Plasmodium berghei. Cured mice were found to be immune upon rechallenge with the same P. berghei strain 4 months after the initial infection and drug-induced cure. MDL 27695 rapidly inhibited the incorporation of [3H]hypoxanthine into P. falciparum RNA and DNA, whereas the incorporation of [3H]isoleucine was not affected until much later. We conclude, therefore, that the major cytotoxic event may be direct binding of MDL 27695 to DNA with subsequent disruption of macromolecular biosynthesis and cell death. These compounds offer a lead in the search for new agents for chemotherapy of malaria.


Subject(s)
Antimalarials/pharmacology , Eflornithine/therapeutic use , Malaria/drug therapy , Plasmodium falciparum/drug effects , Polyamines/pharmacology , Animals , Antimalarials/therapeutic use , Chloroquine/pharmacology , DNA/biosynthesis , Drug Resistance , Drug Therapy, Combination , Humans , Mice , Plasmodium berghei/drug effects , Plasmodium falciparum/growth & development , Polyamines/therapeutic use , Protein Biosynthesis , RNA/biosynthesis
17.
Biochem J ; 237(3): 685-9, 1986 Aug 01.
Article in English | MEDLINE | ID: mdl-3800910

ABSTRACT

Trypanosoma brucei brucei S-adenosyl-L-methionine (AdoMet) decarboxylase was found to be relatively insensitive to activation by putrescine as compared with the mammalian enzyme, being stimulated by only 50% over a 10,000-fold range of putrescine concentrations. The enzyme was not stimulated by up to 10 mM-Mg2+. The Km for AdoMet was 30 microM, similar to that of other eukaryotic AdoMet decarboxylases. T.b. brucei AdoMet decarboxylase activity was apparently irreversibly inhibited in vitro by Berenil and reversibly by pentamidine and methylglyoxal bis(guanylhydrazone). Berenil also inhibited trypanosomal AdoMet decarboxylase by 70% within 4 h after administration to infected rats and markedly increased the concentration of putrescine in trypanosomes that were exposed to the drug in vivo. Spermidine and spermine blocked the curative effect of Berenil on model mouse T.b. brucei infections. This effect of the polyamines was probably not due to reversal of Berenil's inhibitory effects on the AdoMet decarboxylase.


Subject(s)
Adenosylmethionine Decarboxylase/antagonists & inhibitors , Amidines/pharmacology , Carboxy-Lyases/antagonists & inhibitors , Diminazene/pharmacology , Mitoguazone/pharmacology , Pentamidine/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/enzymology , Animals , Diminazene/analogs & derivatives , Diminazene/therapeutic use , Male , Putrescine/pharmacology , Rats , Rats, Inbred Strains , Spermidine/pharmacology , Spermine/pharmacology , Trypanocidal Agents/therapeutic use
18.
Arch Biochem Biophys ; 236(1): 167-75, 1985 Jan.
Article in English | MEDLINE | ID: mdl-3881082

ABSTRACT

The marine heteronemertine Cerebratulus lacteus produces a family of protein cytolysins designated as A-toxins. Limited subtilisin digests of the most abundant homolog, toxin A-III, yield two major products which may be purified by high-performance liquid chromatography. One product is shown to represent residues 1-86 and the other contains the entire toxin sequence (1-95). Both polypeptides are shown to lack internal protease nicks. The 1-95 polypeptide retains full cytolytic activity in comparison to native toxin, whereas 1-86 has an activity that is approximately four times lower. Extensive treatment of A-III with carboxypeptidase Y yields a polypeptide containing residues 1-75 which is totally devoid of hemolytic activity. Residues 63-95 of native A-III have been predicted to form a relatively hydrophobic alpha-helix which is potentially important for activity. The circular dichroism spectrum of 1-95 is in excellent agreement with both experimental and Chou-Fasman-predicted secondary structures of native A-III, while the spectra of 1-86 and 1-75 indicate a loss of helicity quantitatively consistent with the removal of residues 87-95 and 76-95, respectively. Combined with our earlier data on bilayer penetration by N-terminal sequences (K. M. Blumenthal (1982) Biochemistry 21, 4229-4233], the current results indicate a direct involvement of both ends of A-III in lytic activity. The C-terminal region may function by contributing a membrane binding site in the form of an amphipathic helix.


Subject(s)
Marine Toxins , Amino Acids/analysis , Animals , Chemical Phenomena , Chemistry , Hemolysis/drug effects , Humans , In Vitro Techniques , Marine Toxins/toxicity , Peptide Fragments/analysis , Peptide Hydrolases , Platyhelminths , Protein Conformation , Subtilisins
19.
Urban Health ; 12(3): 45-8, 1983 Mar.
Article in English | MEDLINE | ID: mdl-10260559

ABSTRACT

Project Health Care is a program which uses college students considering careers in health care, as summer volunteers in the Emergency Department of a large, municipal hospital. It began with a small group in 1981, and was expanded the following year to include 16 students. The program was designed to provide opportunities for service and observation in various out-patient settings, seminars, lectures and individual and group projects. The project benefitted all participants. For the Emergency Department staff it stimulated better communication with non-professionals; for the hospital it provided intelligent, enthusiastic volunteer help while aiding community relations, and for the volunteers it provided an opportunity to make educated career decisions.


Subject(s)
Emergency Service, Hospital , Hospital Volunteers/statistics & numerical data , Hospital Bed Capacity, 500 and over , Hospitals, Municipal , New York City , Students , Workforce
SELECTION OF CITATIONS
SEARCH DETAIL