Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Biochim Biophys Acta ; 1776(1): 32-57, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17706876

ABSTRACT

The sodium pump, Na(+)/K(+)-ATPase, could be an important target for the development of anti-cancer drugs as it serves as a versatile signal transducer, it is a key player in cell adhesion and its aberrant expression and activity are implicated in the development and progression of different cancers. Cardiotonic steroids, known ligands of the sodium pump have been widely used for the treatment of heart failure. However, early epidemiological evaluations and subsequent demonstration of anti-cancer activity in vitro and in vivo have indicated the possibility of developing this class of compound as chemotherapeutic agents in oncology. Their development to date as anti-cancer agents has however been impaired by a narrow therapeutic margin resulting from their potential to induce cardiovascular side-effects. The review will thus discuss (i) sodium pump structure, function, expression in diverse cancers and its chemical targeting and that of its sub-units, (ii) reported in vitro and in vivo anti-cancer activity of cardiotonic steroids, (iii) managing the toxicity of these compounds and the limitations of existing preclinical models to adequately predict the cardiotoxic potential of new molecules in man and (iv) the potential of chemical modification to reduce the cardiovascular side-effects and improve the anti-cancer activity of new molecules.


Subject(s)
Antineoplastic Agents/therapeutic use , Cardiac Glycosides/therapeutic use , Neoplasms/drug therapy , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Amino Acid Sequence , Animals , Humans , Models, Biological , Molecular Sequence Data , Neoplasms/enzymology , Protein Conformation , Sequence Homology, Amino Acid , Sodium-Potassium-Exchanging ATPase/metabolism
2.
J Org Chem ; 73(17): 6816-23, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18681408

ABSTRACT

A divergent and regioselective synthesis of 1,2,4- and 1,2,5-trisubstituted imidazoles from a readily available (two steps) common intermediate has been developed. This methodology is based on the regiocontrolled N-alkylation of 1-(N,N-dimethylsulfamoyl)-5-iodo-2-phenylthio-1H-imidazole (10). When this intermediate is engaged in reaction with methyl triflate, selective formation of the corresponding 1,2,5-trisubsituted 1H-imidazole is observed. NMR studies have revealed that this regioselectivity can be accounted for by in situ rapid isomerization of 10 into its 1,2,4-isomer (13) followed by regiospecific N-alkylation of the latter. Conversely, when key intermediate 10 is slowly added to Meerwein's salt, isomerization can be constrained and regiospecific N-alkylation of 10 leads to 1,2,4-trisubstituted 1H-imidazole with a high selectivity. The general character of this methodology has been illustrated by showing that iodine in position 4 or 5 could be easily substituted by an aryl group by Suzuki coupling, whereas the phenylthio group at position 2 could, after oxidation into sulfone, be displaced by nucleophilic substitution.


Subject(s)
Hydrocarbons, Iodinated/chemistry , Imidazoles/chemical synthesis , Sulfhydryl Compounds/chemistry , Sulfonamides/chemistry , Alkylation , Isomerism , Magnetic Resonance Spectroscopy , Models, Chemical , Oxidation-Reduction , Sulfones/chemistry
3.
J Med Chem ; 50(17): 4122-34, 2007 Aug 23.
Article in English | MEDLINE | ID: mdl-17658777

ABSTRACT

Amonafide (1), a naphthalimide which binds to DNA by intercalation and poisons topoisomerase IIalpha, has demonstrated activity in phase II breast cancer trials, but has failed thus far to enter clinical phase III because of dose-limiting bone marrow toxicity. Compound 17 (one of 41 new compounds synthesized) is a novel anticancer naphthalimide with a distinct mechanism of action, notably inducing autophagy and senescence in cancer cells. Compound 17 (2,2,2-trichloro-N-({2-[2-(dimethylamino)ethyl]-1,3-dioxo-2,3-dihydro-1H-benzo[de]isoquinolin-5-yl}carbamoyl)acetamide (UNBS3157)) was found to have a 3-4-fold higher maximum tolerated dose compared to amonafide and not to provoke hematotoxicity in mice at doses that display significant antitumor effects. Furthermore, 17 has shown itself to be superior to amonafide in vivo in models of (i) L1210 murine leukemia, (ii) MXT-HI murine mammary adenocarcinoma, and (iii) orthotopic models of human A549 NSCLC and BxPC3 pancreatic cancer. Compound 17, therefore, merits further investigation as a potential anticancer agent.


Subject(s)
Acetamides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Isoquinolines/chemical synthesis , Naphthalimides/chemical synthesis , Urea/analogs & derivatives , Acetamides/pharmacology , Acetamides/toxicity , Adenine , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Apoptosis , Autophagy , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , Cellular Senescence , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Screening Assays, Antitumor , Erythrocyte Count , Female , Humans , Imides/pharmacology , Imides/toxicity , Irinotecan , Isoquinolines/pharmacology , Isoquinolines/toxicity , Leukocyte Count , Maximum Tolerated Dose , Mice , Naphthalimides/pharmacology , Naphthalimides/toxicity , Neoplasm Transplantation , Organophosphonates , Platelet Count , Structure-Activity Relationship , Topoisomerase I Inhibitors , Urea/chemical synthesis , Urea/pharmacology , Urea/toxicity , Gemcitabine
4.
Mol Cancer Ther ; 5(2): 391-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16505114

ABSTRACT

Non-small cell lung cancers (NSCLC) are associated with very dismal prognoses, and adjuvant chemotherapy, including irinotecan, taxanes, platin, and Vinca alkaloid derivatives, offers patients only slight clinical benefits. Part of the chemoresistance of NSCLCs results from the constitutive or anticancer drug-induced activation of the nuclear factor-kappaB (NF-kappaB) signaling pathways. The present study shows that human A549 NSCLC cells display highly activated cytoprotective NF-kappaB signaling pathways. UNBS1450, which is a cardenolide belonging to the same class of chemicals as ouabain and digitoxin, affected the expression and activation status of different constituents of the NF-kappaB pathways in these A549 tumor cells. The modifications brought about by UNBS1450 led to a decrease in both the DNA-binding capacity of the p65 subunit and the NF-kappaB transcriptional activity. Using the 3-(4,5-dimethylthiazol-2yl)-dephenyltetrazolium bromide colorimetric assay, we observed in vitro that UNBS1450 was as potent as taxol and SN38 (the active metabolite of irinotecan) in reducing the overall growth levels of the human A549 NSCLC cell line, and was more efficient than platin derivatives, including cisplatin, carboplatin, and oxaliplatin. The chronic in vivo i.p. and p.o. UNBS1450 treatments of human A549 orthotopic xenografts metastasizing to the brains and the livers of immunodeficient mice had a number of significant therapeutic effects on this very aggressive model.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cardenolides/pharmacology , Cytoprotection/drug effects , Lung Neoplasms/metabolism , NF-kappa B/metabolism , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cardenolides/therapeutic use , DNA/metabolism , Humans , Lung Neoplasms/drug therapy , Mice , Transcription Factor RelA/metabolism , Transcription, Genetic , Up-Regulation , Xenograft Model Antitumor Assays
5.
Curr Med Chem ; 13(29): 3513-27, 2006.
Article in English | MEDLINE | ID: mdl-17168720

ABSTRACT

Galectins form a family of carbohydrate-binding proteins defined by their affinity for beta-galactosides containing glycoconjugates. The carbohydrate recognition domain (CRD) is responsible for the specificity of galectins for saccharides. This binding may result in modulated cell proliferation, cell death and cell migration, three processes that are intimately involved in cancer initiation and progression. Galectins can also display protein-protein types of interactions with their binding partners. Certain galectins directly involved in cancer progression seem to be promising targets for the development of novel therapeutic strategies to combat cancer. Indeed, migrating cancer cells resistant to apoptosis still constitute the principal target for the cytotoxic drugs used to treat cancer patients. Reducing the levels of migration in apoptosis-resistant cancer cells can restore certain levels of sensitivity to apoptosis (and so to pro-apoptotic drugs) in restricted-migration cancer cells. Anti-galectin agents can restrict the levels of migration of several types of cancer cell and should therefore be used in association with cytotoxic drugs to combat metastatic cancer. We provide experimental proof in support of this concept. While the present review focuses on various experimental strategies to impair cancer progression by targeting certain types of galectins, it pays particular attention to glioblastomas, which constitute the ultimate level of malignancy in primary brain tumors. Glioblastomas form the most common type of malignant brain tumor in children and adults, and no glioblastoma patient has been cured to date.


Subject(s)
Galectins/antagonists & inhibitors , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Galectins/physiology , Glioblastoma/drug therapy , Humans , Neoplasm Metastasis/drug therapy , Neoplasms/pathology
6.
J Med Chem ; 49(5): 1800-7, 2006 Mar 09.
Article in English | MEDLINE | ID: mdl-16509595

ABSTRACT

Various mono- and disaccharides were grafted onto a steroid backbone. Whereas in vitro these glycosylated steroids had no cytotoxic effects on six different human cancer cell lines, several of the glycosylated steroids under study did significantly modify the levels of in vitro migration of the human U373 glioblastoma, the A549 non-small-cell-lung cancer (NSCLC), and the PC-3 prostate cancer cells, with more pronounced effects in the case of a monosubstituted beta-L-fucopyranosyl-steroid (19), a monosubstituted beta-D-isomaltosyl-steroid (22), and a monosubstituted beta-D-lactosyl-steroid (24). These three compounds significantly increased the survival of conventional mice grafted subcutaneously with the P388 lymphoma, a lymphoma that metastasizes toward the liver. In vivo, the monosubstituted beta-D-lactosyl-steroid (24) also increased the antitumor effectiveness of cisplatin, a cytotoxic pro-apoptotic drug, in the case of the P388 lymphoma model. This compound also increased the survival of immunodeficient mice into whose brains human U373 glioblastoma cells had been orthotopically grafted.


Subject(s)
Antineoplastic Agents/chemical synthesis , Brain Neoplasms/drug therapy , Fucose/chemistry , Glioblastoma/drug therapy , Lactose/chemistry , Lymphoma/drug therapy , Steroids/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cisplatin/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Glioblastoma/mortality , Glioblastoma/pathology , Glycosylation , Humans , Immunocompromised Host , Lymphoma/mortality , Lymphoma/pathology , Mice , Neoplasm Transplantation , Steroids/chemistry , Steroids/pharmacology , Structure-Activity Relationship , Survival Rate , Transplantation, Heterologous
7.
J Med Chem ; 48(3): 849-56, 2005 Feb 10.
Article in English | MEDLINE | ID: mdl-15689169

ABSTRACT

Analysis of the methanolic extract of Calotropis procera root barks enabled the identification of a novel cardenolide (2''-oxovoruscharin) to be made. Of the 27 compounds that we hemisynthesized, one (23) exhibited a very interesting profile with respect to its hemisynthetic chemical yield, its in vitro antitumor activity, its in vitro inhibitory influence on the Na+/K+-ATPase activity, and its in vivo tolerance. Compound 23 displayed in vitro antitumor activity on a panel of 57 human cancer cell lines similar to taxol, and higher than SN-38 (the active metabolite of irinotecan), two of the most potent drugs used in hospitals to combat cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/isolation & purification , Calotropis/chemistry , Camptothecin/analogs & derivatives , Cardenolides/chemical synthesis , Cardenolides/isolation & purification , Thiazoles/chemical synthesis , Thiazoles/isolation & purification , Animals , Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , Cardenolides/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cerebral Cortex/chemistry , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Etoposide/pharmacology , Humans , Irinotecan , Magnetic Resonance Spectroscopy , Maximum Tolerated Dose , Mice , Organoplatinum Compounds/pharmacology , Oxaliplatin , Paclitaxel/pharmacology , Sodium-Potassium-Exchanging ATPase/chemistry , Structure-Activity Relationship , Swine , Thiazoles/pharmacology
8.
Int J Oncol ; 27(3): 607-16, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16077908

ABSTRACT

To establish the pharmacological profile of a molecule with anti-cancer potential, it seems essential to add an in vivo approach to the first pharmacological experiments carried out in vitro. The present study aims to characterize the degree of sensitivity of seven syngeneic models (two leukemias and five solid tumors) to eleven molecules which have proven to be clinically reliable. We also used some of these models to investigate whether the molecular effects on the extent of growth in a subcutaneously grafted experimental model correlate with the effects of the same drug on the survival of the animals so grafted. Our data show that all the molecules demonstrated significant anti-tumor activities in two mouse leukemia models (with some discrepancies between the two). Two lymphoma models displayed weaker chemosensitivity profiles than the two leukemia models from which they were developed. Two other models, namely the MXT-HS mammary carcinoma and the B16 melanoma, appeared to be rather chemoresistant. However, a direct relationship was evident between the drug-induced decrease in the tumor growth rate and the increase observed in the survival periods of the MXT tumor-bearing mice. This relationship was also observed in the L1210_LYM lymphoma, though to a lesser extent, and was completely absent from the B16 melanoma model. Finally, our data indicated that we had developed a pair of metastasizing, as opposed to non-metastasizing, lymphoma and mammary carcinoma models. In conclusion, the present study shows that syngeneic mouse tumor models can be used as valuable in vivo experimental models for the screening of potential anti-cancer agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor/methods , Neoplasms, Experimental/drug therapy , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor/statistics & numerical data , Female , Leukemia L1210/drug therapy , Leukemia L1210/pathology , Leukemia P388/drug therapy , Leukemia P388/pathology , Male , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasms, Experimental/pathology , Reproducibility of Results
9.
J Neurosurg ; 102(4): 706-14, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15871514

ABSTRACT

OBJECT: Malignant gliomas consist of both heterogeneous proliferating and migrating cell subpopulations, with migrating glioma cells exhibiting less sensitivity to antiproliferative or proapoptotic drugs than proliferative cells. Therefore, the authors combined cimetidine, an antiinflammatory agent already proven to act against migrating epithelial cancer cells, with temozolomide to determine whether the combination induces antitumor activities in experimental orthotopic human gliomas compared with the effects of temozolomide alone. METHODS: Cimetidine added to temozolomide compared with temozolomide alone induced survival benefits in nude mice with U373 human glioblastoma multiforme (GBM) cells orthotopically xenografted in the brain. Computer-assisted phase-contrast microscopy analyses of 9L rat and U373 human GBM cells showed that cimetidine significantly decreased the migration levels of these tumor cells in vitro at concentrations at which tumor growth levels were not modified (as revealed on monotetrazolium colorimetric assay). Computer-assisted microscope analyses of neoglycoconjugate-based glycohistochemical staining profiles of 9L gliosarcomas grown in vivo revealed that cimetidine significantly decreased expression levels of endogenous receptors for fucose and, to a lesser extent, for N-acetyl-lactosamine moieties. Endogenous receptors of this specificity are known to play important roles in adhesion and migration processes of brain tumor cells. CONCLUSIONS: Cimetidine, acting as an antiadhesive and therefore an antimigratory agent for glioma cells, could be added in complement to the cytotoxic temozolomide compound to combat both migrating and proliferating cells in GBM.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Cimetidine/pharmacology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Glioblastoma/drug therapy , Adjuvants, Immunologic/administration & dosage , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/pathology , Brain Neoplasms/veterinary , Cimetidine/administration & dosage , Dacarbazine/administration & dosage , Female , Glioblastoma/pathology , Glioblastoma/veterinary , Mice , Mice, Nude , Temozolomide , Transplantation, Heterologous
10.
J Neuropathol Exp Neurol ; 61(7): 585-96, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12125737

ABSTRACT

We show that high-grade astrocytic tumors with high levels of galectin-1 expression are associated with dismal prognoses. The immunohistochemical analysis of galectin-1 expression of human U87 and U373 glioblastoma xenografts from the brains of nude mice revealed a higher level of galectin-1 expression in invasive areas rather than non-invasive areas of the xenografts. Nude mice intracranially grafted with U87 or U373 cells constitutively expressing low levels of galectin-1 (by stable transfection of an expression vector containing the antisense mRNA of galectin-1) had longer survival periods than those grafted with U87 or U373 cells expressing normal levels of galectin-1. Galectin-1 added to the culture media markedly and specifically increased cell motility levels in human neoplastic astrocytes. These effects are related to marked modifications in the organization of the actin cytoskeleton and the increase in small GTPase RhoA expression. All the data obtained indicate that galectin-1 enhances the migratory capabilities of tumor astrocytes and, therefore, their biological aggressiveness.


Subject(s)
Actin Cytoskeleton/metabolism , Brain Neoplasms/metabolism , Cell Movement/physiology , Glioblastoma/metabolism , Hemagglutinins/metabolism , Neoplasm Invasiveness/physiopathology , Tumor Cells, Cultured/metabolism , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/pathology , Animals , Brain/metabolism , Brain/pathology , Brain/surgery , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Brain Tissue Transplantation , Cell Movement/drug effects , Disease Progression , Female , Galectin 1 , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/pathology , Glioblastoma/physiopathology , Graft Survival/drug effects , Graft Survival/physiology , Hemagglutinins/genetics , Hemagglutinins/pharmacology , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , RNA, Antisense/genetics , Survival Rate , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/transplantation , rhoA GTP-Binding Protein/drug effects
11.
J Med Chem ; 45(12): 2543-55, 2002 Jun 06.
Article in English | MEDLINE | ID: mdl-12036363

ABSTRACT

Among 25 3-aryl-2-quinolone derivatives synthesized, the antitumor activity of some of them was characterized both in vitro and in vivo. In this series, no compound appeared to be cytotoxic in vitro, as was known by the colorimetric MTT assay carried out on 12 distinct human cancer cell lines obtained from the American Type Culture Collection. Indeed, the concentration values decreasing the growth of the 12 cell lines by at least 50% (IC(50) index) were always higher than 10(-5) M. We then made use of a computer-assisted phase-contrast videomicroscopy system to quantitatively determine in vitro the level of migration of living MCF-7 human breast cancer cells. For example, at 10(-7) M, compounds 7, 13, 16, and 28 markedly decreased the migration level of these MCF-7 human breast cancer cells. The in vivo determination of the maximum tolerated dose showed that all compounds tested were definitively nontoxic. When the nontoxic, antimigratory compound 16 was combined with either doxorubicin or etoposide, two cytotoxic compounds routinely used in the clinic, this led to additive in vivo benefits from this treatment (as compared to individual administrations of the drugs) when the MXT mouse mammary adenocarcinoma was used. Thus, nontoxic antimigratory compounds, including the 2-quinolone derivatives synthesized here, can actually improve the efficiency of antitumor treatment when combined with conventional cytotoxic agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Quinolones/chemical synthesis , Tetrazoles/chemical synthesis , Adenocarcinoma/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Etoposide/pharmacology , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mice , Microscopy, Phase-Contrast , Neoplasm Transplantation , Quinolones/chemistry , Quinolones/pharmacology , Structure-Activity Relationship , Tetrazoles/chemistry , Tetrazoles/pharmacology , Tumor Cells, Cultured
12.
J Med Chem ; 46(16): 3536-45, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12877592

ABSTRACT

A series of substituted pyrido[4,3,2-de][1,7] or [1,10]-phenanthrolin-7-ones, analogues of the marine pyridoacridines meridine and ascididemin, have been synthesized on the basis of Diels-Alder reactions involving different quinoline-5,8-diones and N,N-aldehyde-dimethylhydrazones. All the compounds were evaluated for in vitro cytotoxic activity against 12 distinct human cancer cell lines. They all exhibit cytotoxic activity with IC(50) values at least of micromolar order.


Subject(s)
Alkaloids/chemical synthesis , Antineoplastic Agents/chemical synthesis , Phenanthrolines , Alkaloids/chemistry , Alkaloids/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Drug Screening Assays, Antitumor , Humans , Hydrazones/chemistry , Quinolines/chemistry , Structure-Activity Relationship
13.
J Med Chem ; 45(17): 3765-71, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12166949

ABSTRACT

Marine compounds with pyridoacridine skeletons are known to exhibit interesting antitumor activities. Ascididemin has already been reported as displaying significant antitumor activities in vitro and has also been found to have a relatively high global toxicity in vivo. We synthesized a series of 16 analogues (among which 11 compounds were different from previously described ones) with the aim of developing new anticancer agents with significantly improved efficacy/tolerability ratios. These compounds were obtained either by total synthesis from 5,8-quinolinedione and substituted 2-aminoacetophenones or by the direct substitution of ascididemin. The different compounds and ascididemin used as the control compound were tested at six different concentrations on 12 different human cancer cell lines of various histopathological types (glioblastomas and breast, colon, lung, prostate, and bladder cancers). The IC(50) value (i.e., the drug concentration inhibiting the mean growth value of the 12 cell lines by 50%) of these compounds ranged over five log concentrations, i.e., between 10 000 and 0.1 nM. For several new chemical entities, the antitumor activity (determined in vitro) and tolerability (determined in vivo) were superior to those of the parent alkaloids, i.e., ascididemin and 2-bromoleptoclinidone.


Subject(s)
Acridines/chemical synthesis , Alkaloids/chemical synthesis , Antineoplastic Agents/chemical synthesis , Phenanthrolines , Quinolines , Acridines/pharmacology , Acridines/toxicity , Alkaloids/pharmacology , Alkaloids/toxicity , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Cell Division/drug effects , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Mice , Structure-Activity Relationship , Tumor Cells, Cultured
14.
Int J Oncol ; 21(2): 417-25, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12118340

ABSTRACT

The aim of the present work is to investigate whether microtubule-affecting drugs including vincristine, vinblastine, vindesine and vinorelbine are able to produce an anti-angiogenic effect at non-cytotoxic doses in the same way of taxol. The cytotoxic effects were determined by means of the colorimetric MTT assay, and the anti-angiogenic effects on HUVEC cells growing on Matrigel and forming capillary networks. Sixteen additional drugs (camptothecin, SN38, topothecan, adriamycin, daunomycin, etoposide, bleomycin, melphalan, mitomycin C, TNP-470, cisplatin, carboplatin, 5-fluorouracil, methotrexate, suramin and batimastat) were used as control in order to test the specificity of the microtubule-affecting drug effects. We also investigated by means of videomicroscopy whether microtubule-affecting drugs could produce anti-migratory effects at non-cytotoxic doses on tumor cells. Finally, we used computer-assisted fluorescence microscopy to characterize the influence of microtubule-affecting drugs on the polymerization/depolymerization dynamics of the actin cytoskeleton in tumor cells. Our results show that taxol, vincristine and vindesine behave similarly in their ability to reduce the capillary network formation by HUVEC cells cultured on Matrigel. These anti-angiogenic effects appear at non-cytotoxic concentrations. In contrast, vinblastine and vinorelbine produce apparent anti-angiogenic effects by direct cytotoxicity. Microtubule-affecting agents are also able to significantly reduce the level of migration of tumor cells at non-cytotoxic concentrations, some of these effects may occur via modifications to the actin cytoskeleton organization. Several types of microtubule-affecting agents could be used as anti-angiogenic agents by administering them at non-cytotoxic concentrations, and some microtubule-affecting agents abandoned in pharmacological assays could turn out to be potent anti-migratory drugs acting on tumor cells, though without being too cytotoxic.


Subject(s)
Actins/metabolism , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Cell Movement/drug effects , Colonic Neoplasms/blood supply , Colonic Neoplasms/drug therapy , Endothelium, Vascular/drug effects , Microtubules/metabolism , Tumor Cells, Cultured/drug effects , Breast Neoplasms/pathology , Cell Division/drug effects , Collagen , Colonic Neoplasms/pathology , Drug Combinations , Endothelium, Vascular/cytology , Endothelium, Vascular/growth & development , Humans , In Vitro Techniques , Laminin , Microtubules/pathology , Neovascularization, Pathologic/drug therapy , Proteoglycans
15.
Transl Oncol ; 1(1): 1-13, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18607503

ABSTRACT

This review covers the isolation, total synthesis, biologic activity, and more particularly the in vitro and in vivo antitumor activities of naturally occurring isocarbostyril alkaloids from the Amaryllidaceae family. Starting from these natural products, new derivatives have been synthesized to explore structure-activity relationships within the chemical class and to obtain potential candidates for preclinical development. This approach appears to be capable of providing novel promising anticancer agents.

16.
Neoplasia ; 10(6): 573-86, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18516294

ABSTRACT

Several naphthalimides have been evaluated clinically as potential anticancer agents. UNBS3157, a naphthalimide that belongs to the same class as amonafide, was designed to avoid the specific activating metabolism that induces amonafide's hematotoxicity. The current study shows that UNBS3157 rapidly and irreversibly hydrolyzes to UNBS5162 without generating amonafide. In vivo UNBS5162 after repeat administration significantly increased survival in orthotopic human prostate cancer models. Results obtained by the National Cancer Institute (NCI) using UNBS3157 and UNBS5162 against the NCI 60 cell line panel did not show a correlation with any other compound present in the NCI database, including amonafide, thereby suggesting a unique mechanism of action for these two novel naphthalimides. Affymetrix genome-wide microarray analysis and enzyme-linked immunosorbent assay revealed that in vitro exposure of PC-3 cells to UNBS5162 (1 microM for 5 successive days) dramatically decreased the expression of the proangiogenic CXCL chemokines. Histopathology additionally revealed antiangiogenic properties in vivo for UNBS5162 in the orthotopic PC-3 model. In conclusion, the present study reveals UNBS5162 to be a pan-antagonist of CXCL chemokine expression, with the compound displaying antitumor effects in experimental models of human refractory prostate cancer when administered alone and found to enhance the activity of taxol when coadministered with the taxoid.


Subject(s)
Antineoplastic Agents/pharmacology , Chemokines, CXC/metabolism , Naphthalimides/pharmacology , Prostatic Neoplasms/drug therapy , Urea/analogs & derivatives , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Disease Models, Animal , Humans , Kinetics , Male , Mice , Neoplasm Transplantation , Urea/pharmacology
17.
Neoplasia ; 9(9): 766-76, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17898872

ABSTRACT

Our study has shown that the Amaryllidaceae isocarbostyril narciclasine induces marked apoptosis-mediated cytotoxic effects in human cancer cells but not in normal fibroblasts by triggering the activation of the initiator caspases of the death receptor pathway (caspase-8 and caspase-10) at least in human MCF-7 breast and PC-3 prostate carcinoma cells. The formation of the Fas and death receptor 4 (DR4) death-inducing signaling complex was clearly evidenced in MCF-7 and PC-3 cancer cells. Caspase-8 was found to interact with Fas and DR4 receptors on narciclasine treatment. However, narciclasine-induced downstream apoptotic pathways in MCF-7 cells diverged from those in PC-3 cells, where caspase-8 directly activated effector caspases such as caspase-3 in the absence of any further release of mitochondrial proapoptotic effectors. In contrast, in MCF-7 cells, the apoptotic process was found to require an amplification step that is mitochondria-dependent, with Bid processing, release of cytochrome c, and caspase-9 activation. It is postulated that the high selectivity of narciclasine to cancer cells might be linked, at least in part, to this activation of the death receptor pathway. Normal human fibroblasts appear approximately 250-fold less sensitive to narciclasine, which does not induce apoptosis in these cells probably due to the absence of death receptor pathway activation.


Subject(s)
Amaryllidaceae Alkaloids/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Carcinoma/pathology , Mitochondria/physiology , Phenanthridines/pharmacology , Prostatic Neoplasms/pathology , Receptors, Tumor Necrosis Factor/physiology , Amaryllidaceae Alkaloids/isolation & purification , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/physiology , Breast Neoplasms/metabolism , Carcinoma/metabolism , Caspases/physiology , Cytochromes c/analysis , DNA Fragmentation , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Female , Fibroblasts/drug effects , Humans , Male , Mitochondria/enzymology , Narcissus/chemistry , Neoplasm Proteins/drug effects , Neoplasm Proteins/physiology , Phenanthridines/isolation & purification , Prostatic Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand , fas Receptor/drug effects , fas Receptor/physiology
18.
Neoplasia ; 8(5): 402-12, 2006 May.
Article in English | MEDLINE | ID: mdl-16790089

ABSTRACT

Non-small cell lung cancers (NSCLCs) are the leading cause of cancer deaths in most developed countries. Targeting heat shock protein 70 (Hsp70) expression and function, together with the induction of lysosomal membrane permeabilization (LMP), could overcome the multiple anti-cell death mechanisms evidenced in NSCLCs that are responsible for the failure of currently used chemotherapeutic drugs. Because cardenolides bind to the sodium pump, they affect multiple signaling pathways and thus have a number of marked effects on tumor cell behavior. The aim of the present study was to characterize in vitro and in vivo the antitumor effects of a new cardenolide (UNBS1450) on experimental human NSCLCs. UNBS1450 is a potent source of in vivo antitumor activity in the case of paclitaxel-and oxaliplatin-resistant subcutaneous human NCI-H727 and orthotopic A549 xenografts in nude mice. In vitro UNBS1450-mediated antitumor activity results from the induction of nonapoptotic cell death. UNBS1450 mediates the decrease of Hsp70 at both mRNA and protein levels, and this is at least partly due to UNBS1450-induced downregulation of NFAT5/TonEBP (a factor responsible for the transcriptional control of Hsp70). These effects were paralleled by the induction of LMP, as evidenced by acridine orange staining and immunofluorescence analysis for cathepsin B accumulation.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cardenolides/pharmacology , Lung Neoplasms/drug therapy , Lysosomes/chemistry , Acridine Orange/pharmacology , Animals , Cathepsin B/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , HSP70 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Nude
19.
Bioorg Med Chem ; 10(9): 2845-53, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12110304

ABSTRACT

4-Substituted-7H-pyrido-[4,3,2-de][1,8] or [1,9]-phenanthroline-7-ones and 9-methyl-1,4-diazanaphtacene-3,10-dione, analogues of the marine pyridoacridine amphimedine were synthesised from isoquinoline-5,8-dione. The first compounds were obtained starting from a Diels-Alder reaction whereas the synthesis of the last compound was initiated by a reaction of condensation with 2-aminoacetophenone. The different tetra- and pentacyclic compounds were evaluated for in vitro cytotoxic activities against six distinct human cancer cell lines. All the compounds exhibit cytotoxic activity with IC(50) values (i.e., the drug concentration inhibiting the mean growth value of the six cell lines by 50%)<10(-7)M for two of them.


Subject(s)
Acridines/chemical synthesis , Acridines/pharmacology , Animals , Aza Compounds , Cell Division/drug effects , Humans , Isoquinolines , Magnetic Resonance Spectroscopy , Phenanthrolines/chemical synthesis , Phenanthrolines/pharmacology , Porifera/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
20.
Bioorg Med Chem ; 11(20): 4351-6, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-13129571

ABSTRACT

The isomer (9H-quino[4,3,2-de][1,7]phenanthroline-9-one) (2) of the marine alkaloid ascididemin (9H-quino[4,3,2-de][1,10]phenanthroline-9-one) (1) has been synthesized in six steps from 1,4-dimethoxyacridine (10) with an overall yield of 12%. Different related compounds were prepared and tested in vitro at six different concentrations on 12 different human cancer cell lines of various histopathological types (glioblastomas and breast, colon, lung, prostate and bladder cancers). Almost all the compounds present cytotoxic activity of micromolar order.


Subject(s)
Alkaloids/chemical synthesis , Antineoplastic Agents/chemical synthesis , Acridines/chemical synthesis , Acridines/chemistry , Acridines/pharmacology , Alkaloids/chemistry , Alkaloids/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Isomerism , Marine Biology , Phenanthrolines/chemical synthesis , Phenanthrolines/chemistry , Phenanthrolines/pharmacology , Quinolines/chemical synthesis , Quinolines/chemistry , Quinolines/pharmacology , Structure-Activity Relationship , Urochordata/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL