Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 7(7): 2124-33, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11448932

ABSTRACT

The compound bis(4,7-dimethyl-1,10-phenanthroline) sulfatooxovanadium(IV) (METVAN [VO(SO4)(Me2-Phen)2]), exhibits potent cytotoxicity against human cancer cells at low micromolar concentrations. At concentrations > or = 1 microM, METVAN treatment was associated with a nearly complete loss of the adhesive, migratory, and invasive properties of the treated tumor cell populations. METVAN did not cause acute or subacute toxicity in mice at dose levels ranging from 12.5 mg/kg to 100 mg/kg. Therapeutic plasma concentrations > or = 5 microM were rapidly achieved and maintained in mice for at least 24 h after i.p. bolus injection of a single 10 mg/kg nontoxic dose of METVAN. At this dose level, the maximum plasma METVAN concentration was 37.0 microM, which was achieved with a t(max) of 21.4 min. Plasma samples (diluted 1:16) from METVAN-treated mice killed 85% of human breast cancer cells in vitro. METVAN was slowly eliminated with an apparent plasma t(1/2) of 17.5 h and systemic clearance of 42.1 ml/h/kg. In accordance with its potent in vitro activity and favorable in vivo pharmacokinetics, METVAN exhibited significant antitumor activity and delayed tumor progression in CB.17 severe combined immunodeficient (SCID) mouse xenograft models of human glioblastoma and breast cancer. In these experiments, METVAN was administered in daily injections of a single nontoxic 10 mg/kg i.p. dose on 5 consecutive days per week for 4 consecutive weeks beginning the day after the s.c. inoculation of U87 glioblastoma or MDA-MB-231 breast cancer cells. At 40 days after the inoculation of tumor cells, the U87 tumor xenografts in the vehicle-treated control SCID mice were much larger than those of the mice treated with METVAN (4560 +/- 654 mm(3) versus 1688 +/- 571 mm(3); P = 0.003). Similarly, the MDA-MB-231 tumors in SCID mice treated with METVAN were much smaller 40 days after tumor cell inoculation than those of the vehicle-treated control SCID mice (174 +/- 29 mm(3) versus 487 +/- 82 mm(3); P = 0.002). The favorable in vivo pharmacodynamic features and antitumor activity of METVAN warrants further development of this novel oxovanadium compound as a potential new anticancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Organometallic Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Survival/genetics , Dose-Response Relationship, Drug , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Injections, Intraperitoneal , Mice , Mice, SCID , Neoplasm Transplantation , Organometallic Compounds/blood , Organometallic Compounds/pharmacokinetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Curr Pharm Des ; 7(13): 1291-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11472268

ABSTRACT

Recently identified novel agents that disrupt tubulin polymerization include synthetic spiroketal pyrans (SPIKET) targeting the spongistatin binding site of b-tubulin. These agents exhibit anticancer activity by disrupting normal mitotic spindle assembly and cell division as well as inducing apoptosis. At nanomolar concentrations, the SPIKET compound SPIKET-P caused tubulin depolymerization in cell-free turbidity assays and exhibited potent cytotoxic activity against cancer cells as evidenced by destruction of microtubule organization, and prevention of mitotic spindle formation in human breast cancer cells. SPIKET compounds represent a new class of tubulin targeting agents that show promise as anti-cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Pyrans/pharmacology , Spiro Compounds/pharmacology , Tubulin/drug effects , Animals , Binding Sites , Humans , Pyrans/metabolism , Spiro Compounds/metabolism , Tubulin/metabolism
3.
Anticancer Drugs ; 12(4): 369-76, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11335794

ABSTRACT

We present experimental data which establish the organometallic compounds vanadocene dichloride (VDC) and vanadocene acetylacetonate (VDacac) as potent anti-proliferative agents. We first examined the effects of VDC and VDacac on the rapid embryonic cell division and development of Zebrafish. Both compounds were capable of causing cell division block at the 8-16 cell stage of embryonic development followed by total cell fusion and developmental arrest. We next examined the effect of VDC and VDacac on proliferation of human breast cancer and glioblastoma cell lines using MTT assays. VDC inhibited the proliferation of the breast cancer cell line BT-20 as well as the glioblastoma cell line U373 in a concentration-dependent fashion with IC50 values of 11.0, 14.9 and 18.6 microM, respectively. VDacac inhibited cellular proliferation with IC50 values of 9.1, 26.9 and 35.5 microM, respectively. Whereas in vehicle-treated control cancer cells mitotic spindles were organized as a bipolar microtubule array and the DNA was organized on a metaphase plate, vanadocene-treated cancer cells had aberrant monopolar mitotic structures where microtubules were detected only on one side of the chromosomes and the chromosomes were arranged in a circular pattern. In contrast to control cells which showed a single focus of gamma-tubulin at each pole of the bipolar mitotic spindle, VDC- or VDacac-treated cells had two foci of gamma-tubulin on the same side of the chromosomes resulting in a broad centrosome at one pole. All monopolar spindles examined had two foci of gamma-tubulin labeling consistent with a mechanism in which the centrosomes duplicate but do not separate properly to form a bipolar spindle. These results provide unprecedented evidence that organometallic compounds can block cell division in human cancer cells by disrupting bipolar spindle formation. In accordance with these results vanadocene treatment caused an arrest at the G2/M phase of the cell cycle. This unique mechanism of anti-mitotic function warrants further development of vanadocene complexes as anti-cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Organometallic Compounds/pharmacology , Spindle Apparatus/drug effects , Vanadium Compounds/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Division/drug effects , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Female , Flow Cytometry , G2 Phase/drug effects , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Inhibitory Concentration 50 , Microscopy, Confocal , Mitosis/drug effects , Nervous System Neoplasms/drug therapy , Nervous System Neoplasms/pathology , Spindle Apparatus/ultrastructure , Tumor Cells, Cultured , Zebrafish/embryology
4.
Leuk Lymphoma ; 41(5-6): 625-34, 2001 May.
Article in English | MEDLINE | ID: mdl-11378580

ABSTRACT

Bis(4,7-dimethyl-1,10 phenanthroline) sulfatooxovanadium(IV) [VO(SO(4) )(Me(2)-Phen)(2)] induces apoptosis in human NALM-6 leukemia cells. In the present report, we demonstrate that VO(SO(4) )(Me(2)-Phen)(2)-induced apoptosis is mediated through the generation of reactive oxygen species (ROS), depletion of glutathione and depolarization of mitochondrial membrane potential (DeltaPsim). Using multilaser flow cytometry methods, we further mapped out the death sequence that occurs in VO(SO(4))(Me(2)-Phen)(2)-treated leukemic cells. Triple labeling method to measure ROS, DeltaPsim and glutathione coupled with multilaser excitation flow cytometry showed that induction of ROS took place before the loss of mitochondrial permeability transition and depletion of glutathione. Correlated two parameter plots of glutathione content versus DeltaPsim showed that loss of DeltaPsim and depletion of glutathione closely follows each other. Translocation of phosphatidylserine to the outer leaflet of the cell membrane was the final step in the process before the cells became apoptotic. These results demonstrate that the mitochondrial permeability transition takes place during VO(SO(4))(Me(2)-Phen)(2)-induced apoptosis and is mediated through induction of ROS and depletion of glutathione.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Membrane Potentials/drug effects , Mitochondria/ultrastructure , Organometallic Compounds/pharmacology , Flow Cytometry , Glutathione/metabolism , Glutathione/pharmacology , Humans , Intracellular Membranes/chemistry , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Kinetics , Leukemia/drug therapy , Phosphatidylserines/metabolism , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
5.
J Inorg Biochem ; 84(3-4): 241-53, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11374587

ABSTRACT

The structure of [V(eta5-C5H5)2(CH3C(O)CHC(O)CH3)](O3SCF3) (1) (=[VCp2(acac)](O3SCF3)), a dual-function anti-cancer agent with anti-angiogenic and anti-mitotic properties, was determined by single-crystal X-ray diffraction. The geometry is well described as a pseudo-tetrahedral like structure with the centroids of the cyclopentadienyl rings and the two oxygen atoms of the acetylacetonate ring in the ancillary positions of the central vanadium (IV) atom. The bisector of the V(acac) fragment deviates from the C2 axis of the ligand framework by only 4 degrees, compared to a deviation of 7 degrees for the V(acac) fragment in the tetramethylethano-bridged vanadocene acetyl acetonate complex. Crystal data for 1: space group, P2(1)/c; a=7.5544(9) A, b=14.936(2) A, c=16.193(2) A, beta=102.901(2) degrees, V= 1781.0(4) A3; Z=4; R=0.0506 for 2310 reflections with I> 2sigma(I). This report also details the electron paramagnetic resonance, UV/Vis spectroscopy, electrochemical properties and the biological activity profile of this potent anti-cancer agent.


Subject(s)
Angiogenesis Inhibitors/chemistry , Antineoplastic Agents/chemistry , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Vanadium/chemistry , Vanadium/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Chick Embryo/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Stability , Female , Humans , Hydroxybutyrates/chemistry , Immunohistochemistry , Male , Molecular Structure , Neovascularization, Physiologic/drug effects , Pentanones/chemistry , Spindle Apparatus/drug effects , Tumor Cells, Cultured/drug effects , Zebrafish , Zygote/drug effects
6.
Clin Cancer Res ; 7(4): 1094-101, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309362

ABSTRACT

We have examined the in vitro anticancer activity of METVAN [bis(4,7-dimethyl-1,10 phenanthroline) sulfatooxovanadium(IV); VO(SO(4))(Me(2)-Phen)(2)] against acute lymphoblastic leukemia (ALL; NALM-6 and MOLT-3), acute myeloid leukemia (AML; HL-60), Hodgkin's disease (HS445), and multiple myeloma (ARH-77, U266BL, and HS-SULTAN) cell lines as well as primary leukemic cells from patients with ALL, AML, and chronic acute myeloid leukemia (CML). METVAN induced apoptosis in NALM-6, MOLT-3, and HL-60 cells in a concentration-dependent fashion with EC(50) values of 0.19 +/- 0.03 microM, 0.19 +/- 0.01 microM, and 1.1 +/- 0.2 microM, respectively. METVAN induced apoptosis at low micromolar concentrations in primary leukemic cells from patients with ALL, AML, and CML. METVAN inhibited the constitutive expression of matrix metalloproteinase (MMP)-9 protein and its gelatinolytic activity in HL-60 cells and MMP-2 as well as MMP-9 gelatinolytic activities in leukemic cells from ALL, AML, and CML patients. Furthermore, METVAN inhibited the leukemic cell adhesion to the extracellular matrix proteins laminin, type IV collagen, vitronectin, and fibronectin and the invasion through Matrigel matrix. Further preclinical development of METVAN may provide the basis for the development of more effective chemotherapy programs.


Subject(s)
Antineoplastic Agents/pharmacology , Matrix Metalloproteinase Inhibitors , Organometallic Compounds/pharmacology , Cell Adhesion/drug effects , Cell Survival/drug effects , HL-60 Cells , Hodgkin Disease , Humans , Leukemia , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness , Tumor Cells, Cultured
7.
Curr Cancer Drug Targets ; 1(1): 59-71, 2001 May.
Article in English | MEDLINE | ID: mdl-12188892

ABSTRACT

Recently identified agents that interact with cytoskeletal elements such as tubulin include synthetic spiroketal pyrans (SPIKET) and monotetrahydrofuran compounds (COBRA compounds). SPIKET compounds target the spongistatin binding site of beta-tubulin and COBRA compounds target a unique binding cavity on alpha-tubulin. At nanomolar concentrations, the SPIKET compound SPIKET-P causes tubulin depolymerization and exhibits potent cytotoxic activity against cancer cells. COBRA-1 inhibits GTP-induced tubulin polymerization. Treatment of human breast cancer and brain tumor cells with COBRA-1 caused destruction of microtubule organization and apoptosis. Other studies have identified some promising protein tyrosine kinase inhibitors as anti-cancer agents. These include EGFR inhibitors such as the quinazoline derivative WHI-P97 and the leflunomide metabolite analog LFM-A12. Both LFM-A12 and WHI-P97 inhibit the in vitro invasiveness of EGFR positive human breast cancer cells at micromolar concentrations and induce apoptotic cell death. Dimethoxyquinazoline compounds WHI-P131 and WHI-P154 inhibit tyrosine kinase JAK3 in leukemia cells. Of particular interest is WHI-P131, which inhibits JAK3 but not JAK1, JAK2, SYK, BTK, LYN, or IRK at concentrations as high as 350 microM. Studies of BTK inhibitors showed that the leflunomide metabolite analog LFM-A13 inhibited BTK in leukemia and lymphoma cells. Consistent with the anti-apoptotic function of BTK, treatment of leukemic cells with LFM-A13 enhanced their sensitivity to chemotherapy-induced apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , ErbB Receptors/antagonists & inhibitors , Furans/pharmacology , Neoplasms/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrans/pharmacology , Quinazolines/pharmacology , Spiro Compounds/pharmacology , Tubulin/drug effects , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Furans/therapeutic use , Humans , Protein-Tyrosine Kinases/metabolism , Pyrans/therapeutic use , Spiro Compounds/therapeutic use , Structure-Activity Relationship , Tubulin/metabolism
8.
J Inorg Biochem ; 78(4): 321-30, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10857913

ABSTRACT

In a systematic effort to identify and develop effective anticancer agents, four oxovanadium(IV) complexes with 1,10-phenanthroline (Phen) or 4,7-dimethyl-1,10-phenanthroline (Me2-Phen) as ligand(s) were synthesized and characterized. Among the four oxovanadium(IV) complexes synthesized, the crystal structure of the bis(phenanthroline)oxovanadium(IV) complex bis(1,10-phenanthroline)sulfatooxovanadium(IV) ([VO(SO4)(Phen)2], compound 1) has been determined. Compound 1 crystallized in the space group P2(1)/n with unit cell parameters a = 14.2125(17) A, b = 10.8628(13) A, c = 20.143(2) A, alpha = 90 degrees, beta = 102.569(2) degrees, gamma = 90 degrees, V = 3035.3(6) A3, and Z = 4. The refinement of compound 1 by full-matrix least-squares techniques gave an R factor of 0.0785 for 4356 independent reflections. The structure contains two enantiomorphous molecules, lambda and delta, which are related by an inversion center. Compound 1 exhibited 3.5-fold more potent cytotoxic activity against NALM-6 human leukemia cells than the mono(phenanthroline)oxovanadium(IV) complex (diaqua)(1,10-phenanthroline)sulfatooxovanadium(IV) ([VO(SO4)(Phen)(H2O)2], compound 2) (IC50 values: 0.97+/-0.10 microM versus 3.40+/-0.20 microM: P=0.0004). Methyl substitution in the phenanthroline ligand enhanced the anti-leukemic activity of the mono(phenanthroline)oxovanadium(IV) complex 4.4-fold (IC50 values: 0.78+/-0.10 microM, compound 4, versus 3.40+/-0.20 microM, compound 2; P=0.0003) and the anti-leukemic activity of the bis(phenanthroline)oxovanadium(IV) complex 5.7-fold (IC50 values: 0.17+/-0.02 microM, compound 3, versus 0.97+/-0.10 microM, compound 1; P=0.001). The leading oxovanadium compound, bis(4,7-dimethyl-1,10-phenanthroline)sulfatooxovanadium(IV) ([VO(SO4)(Me2-Phen)2], compound 3) triggered the production of reactive oxygen species (ROS) in human leukemia cells, caused G1-arrest and inhibited clonogenic growth at nanomolar concentrations.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Vanadates/chemistry , Vanadates/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Ligands , Methylcellulose/metabolism , Models, Chemical , Reactive Oxygen Species , Time Factors , Tumor Cells, Cultured
9.
Bioorg Med Chem Lett ; 10(10): 1015-8, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10843205

ABSTRACT

A novel mono-THF containing synthetic anticancer drug (WHI-261) was designed for targeting a previously unrecognized unique narrow binding cavity on the surface of tubulin. The anti-cancer activity of WHI-261 was confirmed using MTT assays. The structure-based design, synthesis, and biological activity of WHI-261 are reported.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Fatty Alcohols/chemistry , Fatty Alcohols/pharmacology , Furans/chemistry , Furans/pharmacology , Tubulin/metabolism , Antineoplastic Agents/metabolism , Drug Design , Drug Screening Assays, Antitumor , Fatty Alcohols/metabolism , Furans/metabolism , Humans , Models, Molecular , Protein Conformation , Tubulin/chemistry , Tumor Cells, Cultured
10.
J Cardiovasc Pharmacol ; 35(4): 595-605, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10774791

ABSTRACT

A murine model of vascular injury-induced neointimal hyperplasia was developed by using a photoactive dye, rose bengal. Photoactivation of rose bengal induced vascular injury to the femoral arteries of C57B1/6 mice and resulted in an occlusive neointimal hyperplasia after 4 weeks. The cellular elements of the hyperplastic neointima were found to be alpha-actin-positive vascular smooth muscle cells expressing epidermal growth factor (EGF) receptor at high levels. EGF-Gen, an EGF-R-specific inhibitor with potent anticancer activity, suppressed the formation of hyperplastic neointima. Morphometric analysis of serial tissue sections at 4 weeks after vascular injury showed that in 75% of the EGF-Gen-treated mice, the maximal stenosis index was only 0.44 +/- 0.13, whereas in 75% of phosphate-buffered saline (PBS)-treated mice, the maximal stenosis index was 1.20 +/- 0.25. The mean neointima/media ratios for areas of maximum neointimal hyperplasia were 0.59 +/- 0.16 (n = 24) for the EGF-Gen-treated group, 0.99 +/- 16 (n = 45) for the PBS group (EGF-Gen vs. PBS, p = 0.0017), and 1.03 +/- 18 (n = 8) for group treated with unconjugated genistein (EGF-Gen vs. Gen, p = 0.0088). EGF-Gen treatment of mice with vascular injury to the left femoral artery was not associated with any clinical signs of toxicity or histopathologic lesions in any of the organs, including the uninjured right femoral artery. EGF-Gen also inhibited VSMC migration in vitro, without affecting VSMC proliferation and viability, suggesting that EGF-Gen is blocking neointima formation by inhibiting cellular migration to vascular injury sites. In conclusion, EGF-Gen may be useful as a nontoxic prophylactic agent for prevention of restenosis in clinical settings.


Subject(s)
Antineoplastic Agents/pharmacology , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Genistein/pharmacology , Tunica Intima/pathology , Vascular Diseases/prevention & control , Animals , Antineoplastic Agents/chemistry , Cell Movement , Constriction, Pathologic , Disease Models, Animal , Epidermal Growth Factor/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Genistein/chemistry , Hyperplasia/prevention & control , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Tunica Intima/drug effects , Up-Regulation , Vasoconstriction/drug effects
11.
Clin Cancer Res ; 6(4): 1546-56, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10778988

ABSTRACT

In a systematic effort to identify a potent anticancer agent, we synthesized 15 oxovanadium(IV) complexes and examined their cytotoxic activity against 14 different human cancer cell lines. The oxovanadium compounds included mono and bis ancillary ligands of 1,10-phenanthroline (phen) [VO(phen), VO(phen)2, VO(Me2-phen), VO (Me2-phen)2, VO(Cl-phen), VO(Cl-phen)2, VO(NO2-phen), VO(NO2-phen)2], 2,2'-bipyridyl (bipy) [VO(bipy), VO(bipy)2, VO(Me2-bipy), VO(Me2-bipy)2], and 2-2'-bipyrimidine(bipym) [VO(bipym) and VO-(bipym)2], linked via nitrogen atoms, and 5'-bromo-2'-hydroxyacetophenone (acph) [VO(acph)2], linked via oxygen donor atom. The mono-chelated [VO(Me2-phen), compound 3] and bis-chelated-phen[VO(Me2-phen)2, compound 4] complexes were the most potent oxovanadium compounds and killed target cancer cells at low micromolar concentrations. Notably, the dimethyl substitution of the phenanthroline rings was essential for the anticancer activity of both compound 4 [VO(Me2-phen)2] and compound 3 [VO(Me2-phen)] because unsubstituted bis-chelated and mono-chelated phen oxovanadium(IV) complexes [VO(phen), compound 1, or VO(phen)2, compound 2] were less active. Addition of a chloro or nitro group to the phen complexes did not significantly improve the cytotoxic activity of the unsubstituted oxovanadium(IV) complexes. Irrespective of the ligands, bis-chelated phenanthroline containing compounds showed better activity than the mono-chelated phenanthroline containing complexes. The marked differences in the cytotoxic activity of oxovanadium(IV) complexes containing different heterocyclic ancillary ligands suggest that the cytotoxic activity of these compounds is determined by the identity of the five-member bidentate ligands, as well as the nature of the substitutents on the heterocyclic aromatic rings. Our results presented herein provide experimental evidence that oxovanadium compounds induce apoptosis in human cancer cells. Oxovanadium compounds, especially the lead compound VO(Me2-phen)2, may be useful in the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Organometallic Compounds/pharmacology , Vanadates/pharmacology , Cell Cycle/drug effects , Cell Survival/drug effects , DNA/drug effects , DNA/genetics , DNA/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , HL-60 Cells , Humans , In Situ Nick-End Labeling , Inhibitory Concentration 50 , Organometallic Compounds/chemistry , Tumor Cells, Cultured , Vanadates/chemistry
12.
Clin Cancer Res ; 6(4): 1536-45, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10778987

ABSTRACT

We systematically assessed the cytotoxic effects of five metallocene dichlorides containing vanadium (vanadocene dichloride), titanium (titanocene dichloride), zirconium (zircodocene dichloride), molybdenum (molybdocene dichloride), and hafnium (hafnocene dichloride) as the central metal atom and 19 other vanadocene complexes. These compounds were tested against the human testicular cancer cell lines Tera-2 and Ntera-2 using both 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays and apoptosis assays. Notably, only the vanadium(IV)-containing metallocenes exhibited significant cytotoxicity against Tera-2 and Ntera-2 cells and induced apoptosis within 24 h. Vanadocenes with dithiocyanate [VCp2(SCN)2 x 0.5 H2O] and diselenocyanate [VCp2(NCSe)2] as ancillary ligands were identified as the most potent cytotoxic compounds. Vanadocenes, especially the lead compound VCp2(NCSe)2, may be useful in the treatment of testicular cancer.


Subject(s)
Apoptosis/drug effects , Organometallic Compounds/pharmacology , Testicular Neoplasms/drug therapy , Vanadium Compounds/pharmacology , Cell Cycle/drug effects , Cell Survival/drug effects , DNA/drug effects , DNA/genetics , DNA/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , In Situ Nick-End Labeling , Inhibitory Concentration 50 , Male , Organometallic Compounds/chemistry , Testicular Neoplasms/pathology , Tumor Cells, Cultured , Vanadium Compounds/chemistry
13.
Cell Tissue Res ; 299(2): 185-92, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10741459

ABSTRACT

Transgenic mice in which overexpression of the transforming growth factor alpha (TGF-alpha) gene was directed by the keratin-14 promoter were used to study the regulation of cell cycle progression and proliferation in vivo in the olfactory epithelium. The level of TGF-alpha protein was 73% greater in the nasal-olfactory epithelium of the transgenic mice than in that of nontransgenic littermate controls. Increased levels of TGF-alpha protein were accompanied by a 5.8-fold selective increase in the proliferation of phenotypically characterized horizontal basal cells in the transgenics compared with nontransgenics; in contrast, globose basal cells exhibited a similar low level of proliferation in both transgenics and nontransgenics. The level of expression of epidermal growth factor receptor protein, the receptor for TGF-alpha, was also upregulated in the transgenics, indicating a role for the ErbB tyrosine kinase receptor family in the response to TGF-alpha in the olfactory epithelium. TGF-alpha overexpression was also associated with increased expression of several early cell-cycle-associated proteins, including the growth factor sensor cyclin D1, retinoblastoma, E2F-1 transcription factor, and cyclin E, indicating the progression of relatively quiescent progenitor cells in the G1 phase of the cell cycle toward the G1/S restriction point, after which the cells become refractive to mitogens. These results demonstrate a role for the growth factor TGF-alpha in the in vivo regulation of cell cycle progression and proliferation in the mitotically active olfactory epithelium in these transgenic mice.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Olfactory Mucosa/cytology , Olfactory Receptor Neurons/cytology , Transforming Growth Factor alpha/physiology , Animals , Cell Cycle , Cell Division , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cyclin E/biosynthesis , Cyclin E/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression Regulation , Genes, Retinoblastoma , Genes, Synthetic , Human Growth Hormone/genetics , Humans , Keratin-14 , Keratins/genetics , Mice , Mice, Transgenic , Promoter Regions, Genetic , Retinoblastoma Protein/biosynthesis , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transforming Growth Factor alpha/genetics , Transgenes
14.
Bioorg Med Chem Lett ; 10(6): 551-2, 2000 Mar 20.
Article in English | MEDLINE | ID: mdl-10741551

ABSTRACT

A series of cysteine chloromethyl ketone compounds with a systematic variation of the S-alkyl chain length have been synthesized in order to gauge the effect of the alkyl chain length on the cytotoxicity of these compounds against human acute lymphoblastic leukemia cells. Comparable activities were observed for compounds with S-alkyl chains ranging from pentyl to dodecyl, with the best being undecyl (IC50= 1.7 microM) and dodecyl (IC50=2.0 microM) against B-lineage leukemia cells and hexyl (IC50 = 0.7 microM) against T-lineage leukemia cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Alkylation , Antineoplastic Agents/pharmacology , Cell Line , Coloring Agents , Humans , Structure-Activity Relationship , Tetrazolium Salts , Thiazoles , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
15.
Bioorg Med Chem Lett ; 10(6): 547-9, 2000 Mar 20.
Article in English | MEDLINE | ID: mdl-10741550

ABSTRACT

A series of cysteine diazomethyl- and chloromethyl ketone derivatives has been synthesized and evaluated against human B-lineage (Nalm-6) and T-lineage (Molt-3) acute lymphoblastic leukemia cell lines. The chloromethyl ketone compounds showed potent cytotoxicity against these cell lines, with IC50 values in the low micromolar range. The best compounds were N-acetyl-S-dodecyl-Cys chloromethyl ketone (IC50 = 2.0 microM against Nalm-6, 2.3 microM against Molt-3) and N-acetyl-S-trans,trans-farnesyl-Cys chloromethyl ketone (IC50 = 3.0 microM against Nalm-6 and 1.4 microM against Molt-3).


Subject(s)
Amino Acid Chloromethyl Ketones/chemical synthesis , Antineoplastic Agents/chemical synthesis , Cysteine/analogs & derivatives , Cysteine/chemical synthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Amino Acid Chloromethyl Ketones/pharmacology , Antineoplastic Agents/pharmacology , Cell Line , Coloring Agents , Cysteine/pharmacology , Humans , Tetrazolium Salts , Thiazoles , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
16.
Bioorg Med Chem Lett ; 10(1): 87-90, 2000 Jan 03.
Article in English | MEDLINE | ID: mdl-10636251

ABSTRACT

In a systematic search for novel dual function antioxidants with potent anti-HIV activity, we evaluated 9 rationally designed non-nucleoside inhibitors (NNI) of HIV-1 RT for antioxidant and anti-HIV activities. Our lead phenethyl-5-bromopyridyl thiourea (PEPT) compounds, N-[2-(2-methoxyphenylethyl)]-N'-[2-(5-bromopyridyl)]-thioure a (2) and N-[2-(2-chlorophenylethyl)]-N'-[2-(5-bromopyridyl)]-thiourea (9), inhibited the oxidation of ABTS to ABTS*+ by metmyoglobin in the presence of hydrogen peroxide with EC50 values of 79 and 75 microM, respectively. Both compounds effectively inhibited the oxidation-induced green fluorescence emission from the free radical-sensitive indicator dye 2',7'-dichlorodihydrofluorescein diacetate in CEM human T-cells and Nalm-6 human B-cells exposed to hydrogen peroxide. To our knowledge, compounds 2 and 9 are the first NNI of HIV-1 RT with potent anti-oxidant activity. Furthermore, the activity center was defined as the sulfhydryl group since alkylated PEPT derivatives were inactive. The presence of a free thiourea group was also essential for the anti-HIV activity of the PEPT compounds.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Thiourea/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Crystallography, X-Ray , Flow Cytometry , Fluorescence , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , Humans , Hydrogen Peroxide/toxicity , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Thiourea/analogs & derivatives
17.
Curr Opin Investig Drugs ; 1(2): 252-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11249582

ABSTRACT

Agents that either promote or inhibit tubulin polymerization exhibit anticancer activity by disrupting normal mitotic spindle assembly and cell division as well as inducing apoptosis. Recently identified novel agents that target tubulin include synthetic spiroketal pyrans (SPIKET), targeting the spongistatin binding site of beta-tubulin, and COBRA compounds, targeting a unique binding cavity on alpha-tubulin. At nanomolar concentrations, the SPIKET compound SPIKET-P caused tubulin depolymerization in cell-free turbidity assays and exhibited potent cytotoxic activity against cancer cells as evidenced by destruction of microtubule organization, and prevention of mitotic spindle formation in human breast cancer cells. Molecular modeling studies predicted a high-affinity interaction of the first COBRA compounds, COBRA-0 and COBRA-1, with a unique hydrophobic binding site on alpha-tubulin located between the GTP/GDP binding site and the M-loop. Further studies showed that COBRA-1 inhibited GTP-induced tubulin polymerization in cell-free tubulin turbidity assays. Treatment of human breast cancer and brain tumor (glioblastoma) cells with COBRA-1 caused destruction of microtubule organization and apoptosis. COBRA-1 activated the pro-apoptotic c-Jun N-terminal kinase (JNK) signal transduction pathway. COBRA and SPIKET compounds represent two new classes of tubulin targeting agents that show promise as anticancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Furans/pharmacology , Pyrans/pharmacology , Spiro Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Furans/chemical synthesis , Furans/therapeutic use , Humans , Neoplasms/drug therapy , Pyrans/chemical synthesis , Pyrans/therapeutic use , Spiro Compounds/chemical synthesis , Spiro Compounds/therapeutic use
18.
Clin Cancer Res ; 5(6): 1569-82, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10389946

ABSTRACT

A novel homology model of the kinase domain of Janus kinase (JAK) 3 was used for the structure-based design of dimethoxyquinazoline compounds with potent and specific inhibitory activity against JAK3. The active site of JAK3 in this homology model measures roughly 8 A x 11 A x 20 A, with a volume of approximately 530 A3 available for inhibitor binding. Modeling studies indicated that 4-(phenyl)-amino-6,7-dimethoxyquinazoline (parent compound WHI-258) would likely fit into the catalytic site of JAK3 and that derivatives of this compound that contain an OH group at the 4' position of the phenyl ring would more strongly bind to JAK3 because of added interactions with Asp-967, a key residue in the catalytic site of JAK3. These predictions were consistent with docking studies indicating that compounds containing a 4'-OH group, WHI-P131 [4-(4'-hydroxyphenyl)-amino-6,7-dimethoxyquinazoline], WHI-P154 [4-(3'-bromo-4'-hydroxylphenyl)-amino-6,7-dimethoxyquinazoline], and WHI-P97 [4-(3',5'-dibromo-4'-hydroxylphenyl)-amino-6,7-dimethoxyquinazolin e], were likely to bind favorably to JAK3, with estimated K(i)s ranging from 0.6 to 2.3 microM. These compounds inhibited JAK3 in immune complex kinase assays in a dose-dependent fashion. In contrast, compounds lacking the 4'-OH group, WHI-P79 [4-(3'-bromophenyl)-amino-6,7-dimethoxyquinazoline], WHI-P111 [4-(3'-bromo-4'-methylphenyl)-amino-6,7-dimethoxyquinazoline], WHI-P112 [4-(2',5'-dibromophenyl)-amino-6,7-dimethoxyquinazoline], WHI-P132 [4-(2'-hydroxylphenyl)-amino-6,7-dimethoxyquinazoline], and WHI-P258 [4-(phenyl)-amino-6,7-dimethoxyquinazoline], were predicted to bind less strongly, with estimated K(i)s ranging from 28 to 72 microM. These compounds did not show any significant JAK3 inhibition in kinase assays. Furthermore, the lead dimethoxyquinazoline compound, WHI-P131, which showed potent JAK3-inhibitory activity (IC50 of 78 microM), did not inhibit JAK1 and JAK2, the ZAP/SYK family tyrosine kinase SYK, the TEC family tyrosine kinase BTK, the SRC family tyrosine kinase LYN, or the receptor family tyrosine kinase insulin receptor kinase, even at concentrations as high as 350 microM. WHI-P131 induced apoptosis in JAK3-expressing human leukemia cell lines NALM-6 and LC1;19 but not in melanoma (M24-MET) or squamous carcinoma (SQ20B) cells. Leukemia cells were not killed by dimethoxyquinazoline compounds that were inactive against JAK3. WHI-P131 inhibited the clonogenic growth of JAK3-positive leukemia cell lines DAUDI, RAMOS, LC1;19, NALM-6, MOLT-3, and HL-60 (but not JAK3-negative BT-20 breast cancer, M24-MET melanoma, or SQ20B squamous carcinoma cell lines) in a concentration-dependent fashion. Potent and specific inhibitors of JAK3 such as WHI-P131 may provide the basis for the design of new treatment strategies against acute lymphoblastic leukemia, the most common form of childhood cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Leukemia/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins , Quinazolines/pharmacology , Binding Sites/drug effects , Computer Simulation , Drug Design , Drug Evaluation, Preclinical , Humans , Janus Kinase 1 , Janus Kinase 2 , Janus Kinase 3 , Leukemia/metabolism , Models, Molecular , Protein-Tyrosine Kinases/metabolism , Substrate Specificity , Tumor Cells, Cultured , Tumor Stem Cell Assay
19.
Radiat Res ; 151(2): 133-41, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9952297

ABSTRACT

Mutations of the SOD1 gene (formerly known as Cu,Zn-SOD) are frequently associated with the familial form of amyotrophic lateral sclerosis (ALS). The G93A mutation of SOD1 with substitution of Gly to Ala at residue 93 results in gain of a peroxidative function. Here we report that transfection of PC12 neuron precursor cells with the G93A mutation of SOD1 results in increased production of hydroxyl radicals (*OH) and an enhanced rate of cell death by apoptosis. Notably, PC12 cells transfected with the H63C/G93A mutant of SOD1 with a mutation in the catalytic site that converts histidine at position 63 to cysteine showed a dramatically reduced production of *OH and rate of death by apoptosis. Thus the gain of function of the mutant G93A SOD1 can be reduced by an active site mutation. These results provide additional genetic evidence for the hypothesis that the increased *OH production and induced cytotoxicity in neuron cells expressing the mutant G93A SOD1 results from the gain of peroxidative function by the enzyme's catalytic site.


Subject(s)
Apoptosis/physiology , Hydroxyl Radical/metabolism , Neurons/enzymology , Superoxide Dismutase/physiology , Animals , Binding Sites , Blotting, Northern , Blotting, Western , Gene Expression , Mutation , Neurons/cytology , PC12 Cells/metabolism , Protein Conformation , Rats , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics
20.
Toxicol Appl Pharmacol ; 154(3): 228-35, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9931282

ABSTRACT

Metallocene complexes containing vanadium induce apoptosis in human cancer cells by an as yet unknown mechanism and may therefore be useful as a new class of cytotoxic anticancer drugs. Ultrastructural studies showing the formation of metallocene-DNA complexes prompted the hypothesis that their mechanism of action may resemble the DNA damage induced by cisplatin. Molecular genotoxicity testing provides insights into the mechanisms of action of new chemotherapeutic agents. Therefore, we determined the effects of three cytotoxic vanadocene complexes, vanadocene dichloride, vanadocene dithiocyanate, and vanadocene dioxycyanate, on genomic stability using the yeast DEL recombination assay and transcriptional activation of genotoxic stress-specific promoters in human HepG2 cells using the CAT-Tox(L) assay. Cisplatin caused an 11-fold increase of recombination frequency in yeast and induced transcriptional activation of the DNA damage-associated promoters such as the minimum promoter containing p53 response elements and the GADD45 promoter in addition to activating the promoters for c-fos, heat shock protein 70, metallothionine IIa, and the minimum promoter containing nuclear factor kappa(kappa)B response elements. In contrast to cisplatin, vanadocene complexes did not increase the DEL recombination frequency in yeast nor did they activate any of the DNA damage-associated promoters in HepG2 cells. Vanadocene complexes triggered activation of the c-fos promoter without affecting the minimum promoter containing p53 response elements or the GADD45 promoter. These results indicate that the apoptotic signal of vanadocene complexes is not triggered by primary DNA damage and it does not require p53 induction, thereby disproving the hypothesis that it mechanistically resembles the cytotoxic action of cisplatin.


Subject(s)
Apoptosis , Cisplatin/toxicity , Organometallic Compounds/toxicity , Vanadium Compounds/toxicity , Antineoplastic Agents/toxicity , Cell Line , DNA/metabolism , Dose-Response Relationship, Drug , Gene Expression/drug effects , Genes, Reporter , Heat-Shock Proteins/genetics , Humans , In Situ Nick-End Labeling , Linear Models , Mutagenicity Tests , NF-kappa B/genetics , Saccharomyces cerevisiae/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...