Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cancer Chemother Pharmacol ; 48(6): 435-45, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11800023

ABSTRACT

PURPOSE: Radicicol is a novel hsp90 antagonist, distinct from the chemically unrelated benzoquinone ansamycin compounds, geldanamycin and herbimycin. Both geldanamycin and radicicol bind in the aminoterminal nucleotide-binding pocket of hsp90, destabilizing the hsp90 client proteins, many of which are essential for tumor cell growth. We describe here antitumor activity of a novel oxime derivative of radicicol, KF58333. We also investigated the mechanism of antitumor activity of KF58333 in comparison with its oxime isomer KF58332. METHODS: Antiproliferative activities were determined in a panel of breast cancer cell lines in vitro. We also examined inhibition of hsp90 function and apoptosis induction in erbB2-overexpressing human breast carcinoma KPL-4 cells in vitro. Direct binding activity to hsp90 was assessed by hsp90-binding assays using geldanamycin or radicicol beads. In animal studies, we investigated plasma concentrations of these compounds after i.v. injection in BALB/c mice and antitumor activity against KPL-4 cells transplanted into nude mice. Inhibition of hsp90 function and induction of apoptosis in vivo were investigated using tumor specimens from drug-treated animals. RESULTS: KF58333 showed potent antiproliferative activity against all breast cancer cell lines tested in vitro, and was more potent than its stereoisomer KF58332. These results are consistent with the ability of KF58333 to deplete hsp90 client proteins and the induction of apoptosis in KPL-4 cells in vitro. Interestingly, KF58333, but not KF58332, showed significant in vivo antitumor activity accompanied by induction of apoptosis in KPL-4 human breast cancer xenografts. Although the plasma concentrations of these compounds were equivalent, KF58333, but not KF58332, depleted hsp90 client proteins such as erbB2, raf-1 and Akt in the tumor specimen recovered from nude mice. CONCLUSIONS: These results suggest that inhibition of hsp90 function, which causes depletion of hsp90 client proteins in tumor, contributes to the antitumor activity of KF58333, and that the stereochemistry of the oxime moiety is important for the biological activity of radicicol oxime derivatives.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Lactones/pharmacology , Animals , Breast Neoplasms/pathology , Female , Gene Expression Regulation , Genes, erbB-2 , Humans , Injections, Intravenous , Macrolides , Mice , Mice, Inbred BALB C , Mice, Nude , Oximes/pharmacology , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
2.
Blood ; 96(6): 2284-91, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10979978

ABSTRACT

Chronic myelogenous leukemia (CML) is a clonal disorder of a pluripotent hematopoietic stem cells characterized by a chimeric bcr-abl gene giving rise to a p210(Bcr-Abl) protein with dysregulated tyrosine kinase activity. Radicicol, a macrocyclic antifungal antibiotic, binds to the N-terminal of heat shock protein 90 (Hsp90) and destabilizes Hsp90-associated proteins such as Raf-1. This study investigated the effect of radicicol, novel oxime derivatives of radicicol (KF25706 and KF58333), and herbimycin A (HA), a benzoquinoid ansamycin antibiotic, on the growth and differentiation of human K562 CML cells. Although KF25706 and KF58333 induced the expression of glycophorin A in K562 cells, radicicol and HA caused erythroid differentiation transiently. Cell cycle analysis showed that G(1) phase accumulation was observed in K562 cells treated with KF58333. KF58333 treatment depleted p210(Bcr-Abl), Raf-1, and cellular tyrosine phosphorylated proteins in K562 cells, whereas radicicol and HA showed transient depletion of these proteins. KF58333 also down-regulated the level of cell cycle-dependent kinases 4 and 6 and up-regulated cell cycle-dependent kinase inhibitor p27(Kip1) protein without an effect on the level of Erk and Hsp90 proteins. Immunoprecipitation analysis showed that p210(Bcr-Abl) formed multiple complexes with Hsp90, some containing p23 and others Hsp70; KF58333 treatment dissociated p210(Bcr-Abl) from Hsp90/p23 chaperone complexes. Furthermore, KF58333 induced apoptosis in K562 cells and administration of KF58333 prolonged the survival time of SCID mice inoculated with K562 cells. These results suggest that KF58333 may have therapeutic potential for the treatment of CML that involves abnormal cellular proliferation induced by p210(Bcr-Abl).


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antifungal Agents/pharmacology , Fusion Proteins, bcr-abl/metabolism , G1 Phase/drug effects , HSP90 Heat-Shock Proteins/metabolism , Lactones/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Quinones/pharmacology , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Antifungal Agents/chemistry , Antifungal Agents/therapeutic use , Benzoquinones , Cell Differentiation/drug effects , Erythroblasts/drug effects , Erythroblasts/pathology , Humans , K562 Cells , Lactams, Macrocyclic , Lactones/chemistry , Lactones/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Macrolides , Mice , Quinones/chemistry , Quinones/therapeutic use , Rifabutin/analogs & derivatives
3.
Blood ; 95(12): 3771-80, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10845909

ABSTRACT

We examined the chemoprotective effects of KF41399, a novel derivative of carbazole compounds, on severe thrombocytopenia induced by nimustine (ACNU, 45 mg/kg administered for 2 consecutive days intravenously) in mice. Administration schedule studies revealed that pretreatment of mice with KF41399 was necessary to improve thrombocytopenia. Oral administration of KF41399 ameliorated thrombocytopenia induced by ACNU and accelerated the rate of platelet recovery in a dose-dependent fashion. In addition, KF41399 pretreatment improved the decrease in body weight and spleen weight and in the colony-forming activity of bone marrow mononuclear cells (MNC). Oral administration of KF41399 to normal mice induced G(0)/G(1)-phase accumulation of MNC as well as hematopoietic progenitor cells (lineage negative cells [Lin(-)]) and reduced the colony-forming activity of MNC. In Lin(-) cells derived from KF41399-treated mice, up-regulation of Bcl-2 and down-regulation of cyclin E and cyclin A proteins were observed. In the same cells, a decrease in the phosphorylated form of Rb protein and an increase in the p130 protein were observed without changes in the protein level of cell cycle-dependent kinase 2 (Cdk2), Cdk4, and Cdk6. More important, KF41399 did not affect the antitumor activity of ACNU against mouse Sarcoma180 and human lung cancer LC-6. However, 25-mg/kg KF41399 treatment reduced the antitumor activity of ACNU against human lung cancer Lu-65, and 5 mg/kg KF41399 caused a slight reduction of the antitumor activity of ACNU without inducing thrombocytopenia. These results suggest that KF41399 might be useful as a chemoprotective agent to improve chemotherapy-induced thrombocytopenia and types of other toxicity. (Blood. 2000;95:3771-3780)


Subject(s)
Bone Marrow Cells/cytology , Carbazoles/pharmacology , Hematopoietic Stem Cells/cytology , Nimustine/toxicity , Sarcoma 180/drug therapy , Thrombocytopenia/prevention & control , Adenocarcinoma/drug therapy , Administration, Oral , Animals , Bone Marrow Cells/drug effects , Carbazoles/administration & dosage , Carbazoles/therapeutic use , Cell Cycle/drug effects , Cell Survival/drug effects , Colony-Forming Units Assay , Hematopoietic Stem Cells/drug effects , Humans , K562 Cells , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Lung Neoplasms/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nimustine/therapeutic use , Platelet Count/drug effects , Spleen/drug effects , Thrombocytopenia/chemically induced , Transplantation, Heterologous
4.
Bioorg Med Chem ; 8(2): 381-91, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10722161

ABSTRACT

A series of the 8-O-substituted A-ring pyrrole derivatives of duocarmycin bearing the simplified DNA-binding moieties such as cinnamoyl or heteroarylacryloyl groups were synthesized, and evaluated for in vitro anticellular activity against HeLa S3 cells and in vivo antitumor activity against murine sarcoma 180 in mice. In addition, the stability of the 8-O-substituted analogues in aqueous solution and the conversion to their active form (cyclopropane compound) from the 8-O-substituted analogues in mice or human serum were examined. The 8-O-substituted A-ring pyrrole derivatives bearing the simplified DNA-binding moieties showed remarkably potent in vivo antitumor activity and low peripheral blood toxicity compared with the 8-O-substituted A-ring pyrrole derivatives having the trimethoxyindole skeleton in segment-B (Seg-B), which were equal to 8-O-[(N-methylpiperazinyl)carbonyl] derivatives of 4'-methoxycinnamates and 4'-methoxy-beta-heteroarylacrylates. Moreover, among 8-O-substituted analogues, several compounds can be chemically or enzymatically converted to their active form in human serum. This result indicated that new 8-O-substituted derivatives were different prodrugs from KW-2189 and 8-O-substituted analogues being the same type of prodrug as KW-2189.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Pyrroles/chemistry , Pyrrolidinones/chemistry , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/chemical synthesis , Antibiotics, Antineoplastic/pharmacology , Blood Cell Count/drug effects , DNA/drug effects , Drug Screening Assays, Antitumor , Humans , Magnetic Resonance Spectroscopy , Male , Mice , Pyrrolidinones/adverse effects , Pyrrolidinones/chemical synthesis , Pyrrolidinones/pharmacology , Spectrometry, Mass, Fast Atom Bombardment , Tumor Cells, Cultured
5.
J Neurosci ; 20(1): 43-50, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10627579

ABSTRACT

We have studied the mechanisms of auditory hair cell death after insults in vitro and in vivo. We show DNA fragmentation of hair cell nuclei after ototoxic drug and intense noise trauma. By using phospho-specific c-Jun-N-terminal kinase (JNK) and c-Jun antibodies in immunohistochemistry, we show that the JNK pathway, associated with stress, injury, and apoptosis, is activated in hair cells after trauma. CEP-1347, a derivative of the indolocarbazole K252a, is a small molecule that has been shown to attenuate neurodegeneration by blocking the activation of JNK (). Subcutaneously delivered CEP-1347 attenuated noise-induced hearing loss. The protective effect was demonstrated by functional tests, which showed less hearing threshold shift in CEP-1347-treated than in nontreated guinea pigs, and by morphometric methods showing less hair cell death in CEP-1347-treated cochleas. In organotypic cochlear cultures, CEP-1347 prevented neomycin-induced hair cell death. In addition to hair cells, CEP-1347 promoted survival of dissociated cochlear neurons. These results suggest that therapeutic intervention in the JNK signaling cascade, possibly by using CEP-1347, may offer opportunities to treat inner ear injuries.


Subject(s)
Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/cytology , Hearing Loss, Noise-Induced/drug therapy , Indoles/pharmacology , Neurons, Afferent/cytology , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Aminoglycosides/toxicity , Animals , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/physiology , Hearing Loss, Noise-Induced/chemically induced , Hearing Loss, Noise-Induced/pathology , Neomycin/toxicity , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Noise/adverse effects , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
6.
J Med Chem ; 42(15): 2946-60, 1999 Jul 29.
Article in English | MEDLINE | ID: mdl-10425104

ABSTRACT

A series of 3-substituted A-ring pyrrole compounds of duocarmycin were synthesized and evaluated for in vitro anticellular activity against HeLa S(3) cells and in vivo antitumor activity against murine sarcoma 180 in mice. These compounds were evaluated on the peripheral blood toxicity and delayed lethal toxicity. Further, to expand our investigation of their peripheral blood toxicity, the toxicity to bone marrow cells (CFU-GM, CFU-Meg) was investigated. Among 3-substituted A-ring pyrrole compounds of duocarmycin bearing a 5',6',7'-trimethoxy-2'-indolecarboxyl group as segment-B (Seg-B), several analogues showed remarkably potent antitumor activity with low peripheral blood toxicity. The 3-formyl compound 12h, one of such analogues, showed stronger antitumor activity with lower toxicity to bone marrow cells compared to DU-86 (2a), an active metabolite of KW-2189 (2b). However, compound 12h caused delayed death. On the other hand, the 3-bromo compound 15f, one of the 3-substituted A-ring pyrrole derivatives bearing a 4'-methoxycinnamoyl group as Seg-B, showed the most potent antitumor activity among the 4'-methoxycinnamate analogues with low toxicity to bone marrow cells. Furthermore, compound 15f did not cause delayed death similarly to 2d. These results would indicate the importance of the C-3 substituents of A-ring pyrrole duocarmycin derivatives for exhibiting antitumor activity and decreasing toxicity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Indoles/chemical synthesis , Pyrrolidinones/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Bone Marrow Cells/drug effects , Colony-Forming Units Assay , Drug Screening Assays, Antitumor , Humans , Indoles/chemistry , Indoles/pharmacology , Indoles/toxicity , Inhibitory Concentration 50 , Leukocyte Count/drug effects , Male , Mice , Neoplasm Transplantation , Platelet Count/drug effects , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacology , Pyrrolidinones/toxicity , Sarcoma 180/drug therapy , Structure-Activity Relationship , Tumor Cells, Cultured
7.
Cancer Res ; 59(12): 2931-8, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10383157

ABSTRACT

Radicicol, a macrocyclic antifungal antibiotic, has been shown to bind to the heat shock protein 90 (Hsp90) chaperone, interfering with its function. Hsp90 family chaperones have been shown to associate with several signaling molecules and play an essential role in signal transduction, which is important for tumor cell growth. Because radicicol lacks antitumor activity in vivo in experimental animal models, we examined the antitumor activity of a novel radicicol oxime derivative, radicicol 6-oxime (KF25706), on human tumor cell growth both in vitro and in vivo. KF25706 showed potent antiproliferative activities against various human tumor cell lines in vitro and inhibited v-src- and K-ras-activated signaling as well as radicicol. In addition, Hsp90 family chaperone-associated proteins, such as p185erbB2, Raf-1, cyclin-dependent kinase 4, and mutant p53, were depleted by KF25706 at a dose comparable to that required for antiproliferative activity. KF25706 was also shown to compete with geldanamycin for binding to Hsp90. KF29163, which is an inactive derivative of radicicol, was less potent both in p185erbB2 depletion and Hsp90 binding. More importantly, KF25706 showed significant growth-inhibitory activity against human breast carcinoma MX-1 cells transplanted into nude mice at a dose of 100 mg/kg twice daily for five consecutive i.v. injections. KF25706 was also shown to possess antitumor activity against human breast carcinoma MCF-7, colon carcinoma DLD-1, and vulval carcinoma A431 cell lines in vivo in an animal model. Finally, we confirmed the depletion of Hsp90-associated signaling molecules (Raf-1 and cyclin-dependent kinase 4) with ex vivo Western blotting analysis using MX-1 xenografts. In agreement with in vivo antitumor activity, KF25706 depleted Hsp90-associated molecules in vivo, whereas KF29163 and radicicol did not show this activity in vivo. Taken together, these results suggest that antitumor activity of KF25706 may be mediated, at least in part, by binding to Hsp90 family proteins and destabilization of Hsp90-associated signaling molecules.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Lactones/chemistry , Lactones/pharmacology , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Benzoquinones , Cell Line , Drug Screening Assays, Antitumor , Genes, ras , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic , Lactones/metabolism , Macrolides , Mice , Mice, Inbred BALB C , Mice, Nude , Oncogene Protein pp60(v-src)/metabolism , Quinones/pharmacology , Signal Transduction/drug effects , Tumor Cells, Cultured
8.
Cancer Res ; 59(10): 2395-401, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10344749

ABSTRACT

The indolocarbazole analogue CEP-751 is a potent and selective tyrosine kinase inhibitor of the neurotrophin-specific trk receptors that has demonstrated antitumor activity in nine different models of prostate cancer growth in vivo. In the slow-growing, androgen-sensitive Dunning H prostate cancers, which express trk receptors, CEP-751 induced transient regressions independent of effects on cell cycle. Because androgen ablation is the most commonly used treatment for prostate cancer, we examined whether the combination treatment of CEP-751 with castration would lead to better antitumor efficacy than either treatment alone. For a 60-day period, H tumor-bearing rats received treatment with either castration, CEP-751 (10 mg/kg once a day s.c. for 5 days every 2 weeks), a combination of both, or vehicle. Castration caused tumor regression, followed by tumor regrowth in 4-6 weeks, whereas intermittent CEP-751 treatments resulted in tumor regressions during each treatment, which were followed by a period of regrowth between intermittent drug treatment cycles. Overall, both monotherapies significantly inhibited tumor growth compared with the vehicle-treated control group. However, the combination of castration and concomitant CEP-751 produced the most dramatic results: sigificantly greater tumor regression than either therapy alone, with no signs of regrowth. A related experiment using an orally administered CEP-751 analogue (CEP-701), as the trk inhibitor, and a gonadotrophin-releasing hormone agonist, Leuprolide, to induce androgen ablation demonstrated similar results, indicating that these effects could be generalized to other forms of androgen ablation and other trk inhibitors within this class. In addition, when CEP-701 was given sequentially to rats bearing H tumors, which were progressing in the presence of continuous androgen ablation induced by Leuprolide, regression of the androgen-independent tumors occurred. In summary, these data demonstrate that CEP-751 or CEP-701, when combined with surgically or chemically induced androgen ablation, offer better antitumor efficacy than either monotherapy and suggest that each therapy produces prostate cancer cell death through complementary mechanisms.


Subject(s)
Adenocarcinoma/drug therapy , Androgens , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carbazoles/therapeutic use , Indoles , Leuprolide/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Hormone-Dependent/drug therapy , Orchiectomy , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Nerve Growth Factor/antagonists & inhibitors , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Carbazoles/administration & dosage , Combined Modality Therapy , Drug Screening Assays, Antitumor , Drug Synergism , Furans , Injections, Subcutaneous , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Rats , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptor, trkA , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/genetics
9.
Exp Hematol ; 26(13): 1195-201, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9845374

ABSTRACT

We investigated the in vitro and in vivo effects of KT6352, a derivative of indolocarbazole compound, on murine megakaryocytopoiesis. When serum-free megakaryocyte (Meg) colony assay was performed with 100 U/mL of recombinant mouse interleukin-3 (rmIL-3), the addition of 1x10(-11)M to 1x10(-9)M of KT6352 increased the number of Meg colonies. An additional increase of Meg colonies by KT6352 was observed in the serum-free culture containing rmIL-3 plus recombinant mouse interleukin-6 or rmIL-3 plus recombinant mouse stem cell factor. KT6352 did not stimulate Meg colony formation without rmIL-3. When KT6352 was administered intraperitoneally to normal BALB/c male mice at a dose of 10 mg/kg daily for 5 consecutive days, a 2.1-fold increase in the platelet count was observed on day 14, and the prolonged thrombocytopoiesis was detectable from 9 to 27 days after KT6352 administration. A marked increase in the white blood cell count was also observed from 5 to 14 days after KT6352 treatment. Before the gradual increase of platelet counts, 8 days after KT6352 administration, a marked increase in the number of colony-forming units of megakaryocytes (CFU-Megs) in bone marrow and spleen was observed, and a substantial increase in the number of splenic CFU-Megs was observed 14 and 23 days after KT6352 administration. Bone marrow Meg ploidy analysis by two-color flow cytometry showed a shift in the modal ploidy class from 16 to 32 and an increase in the frequency of 64 cells in KT6352-treated mice. These results suggest a possible therapeutic benefit of KT6352 in the management of thrombocytopenia.


Subject(s)
Carbazoles/pharmacology , Hematopoiesis/drug effects , Indoles/pharmacology , Megakaryocytes/drug effects , Animals , Bone Marrow Cells/drug effects , Cells, Cultured , Colony-Forming Units Assay , Enzyme Inhibitors/pharmacology , Flow Cytometry , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Leukocyte Count , Male , Mice , Mice, Inbred BALB C , Platelet Count , Ploidies , Protein Kinase C/antagonists & inhibitors , Recombinant Proteins/pharmacology , Spleen/cytology , Spleen/drug effects , Staurosporine/pharmacology
10.
Clin Cancer Res ; 4(8): 1887-98, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9717816

ABSTRACT

Advanced prostate cancer remains largely incurable, primarily because the very low growth fraction present in these tumors makes them generally resistant to treatment with standard chemotherapeutic agents that target cell division. Effective therapies should therefore induce death of prostate cancer cells, independent of their growth rate. trkA, the high-affinity tyrosine kinase-linked receptor for nerve growth factor, has been implicated in prostatic cancer growth and may represent a molecular target for therapeutic agents. At low mg/kg doses, the trk tyrosine kinase inhibitor CEP-751 (KT6587) inhibits prostatic cancer growth in nine different animal models independent of the tumor growth rate, androgen sensitivity, metastatic ability, or state of tumor differentiation. CEP-751 is selective for cancerous versus normal prostate cells and affects the growth of only a limited number of nonprostate tumors. Importantly, CEP-751 induces cell death of prostate cancer cells in a cell cycle-independent fashion and, therefore, represents a novel therapeutic approach to the management of both hormone-dependent and hormone-independent prostate cancer.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Nerve Growth Factor/antagonists & inhibitors , Adenocarcinoma/pathology , Animals , Cell Cycle/drug effects , Cell Death/drug effects , Disease Models, Animal , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/biosynthesis , Rats , Rats, Inbred Strains , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor, trkA , Receptors, Nerve Growth Factor/biosynthesis , Transplantation, Heterologous , Tumor Cells, Cultured
11.
J Neurobiol ; 35(4): 361-70, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9624618

ABSTRACT

CEP-1347, also known as KT7515, a derivative of a natural product indolocarbazole, inhibited motor neuronal death in vitro, inhibited activation of the stress-activated kinase JNK1 (c-jun NH terminal kinase) in cultured spinal motor neurons, but had no effect on the mitogen-activated protein kinase ERK1 in these cells. Results reported here profile the functional activity of CEP-1347/KT7515 in vivo in models of motor neuronal death or dedifferentiation. Application of CEP-1347/KT7515 to the chorioallantoic membrane of embryonic chicks rescued 40% of the lumbar motor neurons that normally die during the developmental period assessed. Peripheral administration of low doses (0.5 and 1 mg/kg daily) of CEP-1347/KT7515 reduced death of motor neurons of the spinal nucleus of the bulbocavernosus in postnatal female rats, with efficacy comparable to testosterone. Strikingly, daily administration of CEP-1347/KT7515 during the 4-day postnatal window of motor neuronal death resulted in persistent long-term motor neuronal survival in adult animals that received no additional CEP-1347/KT7515. In a model of adult motor neuronal dedifferentiation following axotomy, local application of CEP-1347/KT7515 to the transected hypoglossal nerve substantially reduced the loss of choline acetyl transferase immunoreactivity observed 7 days postaxotomy compared to untreated animals. Results from these experiments demonstrate that a small organic molecule that inhibits a signaling pathway associated with stress and injury also reduces neuronal death and degeneration in vivo.


Subject(s)
Apoptosis/drug effects , Axotomy , Carbazoles/pharmacology , Indoles/pharmacology , Mitogen-Activated Protein Kinases , Motor Neurons/drug effects , Motor Neurons/physiology , Animals , Animals, Newborn/physiology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Differentiation/drug effects , Chick Embryo , Choline O-Acetyltransferase/metabolism , Enzyme Inhibitors/pharmacology , Female , Hypoglossal Nerve/drug effects , Hypoglossal Nerve/enzymology , Hypoglossal Nerve/pathology , JNK Mitogen-Activated Protein Kinases , Motor Neurons/pathology , Rats , Rats, Sprague-Dawley
12.
Neuroscience ; 86(2): 461-72, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9881861

ABSTRACT

We have identified a class of small organic molecules, derived from the indolocarbazole K-252a, that promote the survival of cultured neurons. However, many of these indolocarbazoles inhibit protein kinase C and neurotrophin-activated tyrosine kinase receptors. These kinase inhibitory activities may limit the utility of these compounds for neurological disorders. A bis-ethyl-thiomethyl analogue of K-252a, CEP-1347/KT-7515, has been identified that lacks protein kinase C and tyrosine kinase receptor inhibitory activities, yet retains the ability to promote survival of cultured neurons, including cholinergic neurons derived from the basal forebrain. In the present studies, CEP-1347/KT-7515 was assessed for neurotrophic activity on basal forebrain neurons of in vivo rats following excitotoxic insult. Ibotenate infusion into the nucleus basalis magnocellularis reduced levels of choline acetyltransferase activity in the cortex, as well as reduced numbers of choline acetyltransferase-immunoreactive and retrogradely (FluoroGold)-labelled cortically-projecting neurons in the nucleus basalis. Systemically administered CEP-1347/KT-7515 attenuated the loss of cortical choline acetyltransferase activity and the loss of the number of choline acetyltransferase-immunoreactive and retrogradely-labelled FluoroGold neurons in the nucleus basalis. Moreover, CEP-1347/KT-7515 ameliorated the loss of cortical choline acetyltransferase if administration was initiated one day, but not seven days post-lesion. Together, these results demonstrate that CEP-1347/KT-7515 protects damaged cortically-projecting basal forebrain neurons from degeneration. Thus, CEP-1347/KT-7515 may have therapeutic potential in neurodegenerative diseases, such as Alzheimer's disease, in which basal forebrain cholinergic neurons degenerate.


Subject(s)
Carbazoles/pharmacology , Choline O-Acetyltransferase/metabolism , Indoles/pharmacology , Neurons/physiology , Stilbamidines , Substantia Innominata/physiology , Animals , Axonal Transport/drug effects , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Frontal Lobe/drug effects , Frontal Lobe/enzymology , Frontal Lobe/physiology , Functional Laterality , Ibotenic Acid/toxicity , Male , Neurons/cytology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Substantia Innominata/drug effects , Substantia Innominata/pathology
13.
Bioorg Med Chem Lett ; 8(14): 1873-6, 1998 Jul 21.
Article in English | MEDLINE | ID: mdl-9873450

ABSTRACT

Prodrug esters of the indolocarbazole CEP-751 (KT-6587) were prepared with the goal of identifying water soluble, stable but cleavable forms for intravenous dosing. A dipeptide proform Lys-beta-Ala (16, CEP-2563/KT-8391) was identified for advancement to clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Prodrugs/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Carbazoles/pharmacokinetics , Drug Screening Assays, Antitumor , Esters , Humans , Prodrugs/pharmacokinetics
14.
Int J Cancer ; 72(4): 673-9, 1997 Aug 07.
Article in English | MEDLINE | ID: mdl-9259409

ABSTRACT

The present report describes the in vitro and in vivo profile of CEP-751, a novel receptor tyrosine kinase inhibitor. CEP-751 at 100 nM inhibits the receptor tyrosine kinase activity of the neurotrophin receptors trkA, trkB and trkC. CEP-751 has no effect on activity of receptors for EGF, IGF-I, insulin or on erbB2; inhibition of receptors for PDGF and bFGF was observed but occurred with lesser potency than inhibition of trk. CEP-751 exhibited anti-tumor efficacy against tumors derived from NIH3T3 cells transfected with trkA. Inhibition of trk phosphorylation could also be measured in these tumors, suggesting that anti-tumor efficacy of CEP-751 is related to inhibition of trk receptor tyrosine kinase activity. CEP-751 was found to be without effect when administered to nude mice bearing SK-OV-3 tumors, which overexpress erbB2 receptors, providing further evidence that inhibition of tumor growth may be related to inhibition of trk receptor tyrosine kinase activity. Our data indicate that CEP-751 is a potent trk inhibitor which possesses anti-tumor activity.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Nerve Growth Factor/antagonists & inhibitors , 3T3 Cells/metabolism , 3T3 Cells/physiology , Animals , Mice , Nerve Growth Factors/pharmacology , PC12 Cells , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rats , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, trkA , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Stimulation, Chemical , Transfection , Tyrosine/metabolism
15.
J Med Chem ; 40(12): 1863-9, 1997 Jun 06.
Article in English | MEDLINE | ID: mdl-9191963

ABSTRACT

A series of 3,9 disubstituted [(alkylthio)methyl]- and (alkoxymethyl)-K-252a derivatives was synthesized with the aim of enhancing and separating the neurotrophic properties from the undesirable NGF (trk A kinase) and PKC inhibitory activities of K-252a. Data from this series reveal that substitution in the 3- and 9-positions of K-252a with these groups reduces trk A kinase inhibitory properties approximately 100- to > 500-fold while maintaining or in certain cases enhancing the neurotrophic activity. From this research, 3,9-bis[(ethylthio)methyl]-K-252a (8) was identified as a potent and selective neurotrophic agent in vitro as measured by enhancement of choline acetyltransferase activity in embryonic rat spinal cord and basal forebrain cultures. Compound 8 was found to have weak kinase inhibitory activity for trk A, protein kinase C1 protein kinase A, and myosin light chain kinase. On the basis of the in vitro profile, 8 was evaluated in in vivo models suggestive of neurological diseases. Compound 8 was active in preventing degeneration of cholinergic neurons of the nucleus basalis magnocellularis (NBM) and reduced developmentally programmed cell death (PCD) of female rat spinal nucleus of the bulbocavernosus motoneurons and embryonic chick lumbar motoneurons.


Subject(s)
Carbazoles/chemistry , Carbazoles/chemical synthesis , Carbazoles/pharmacology , Indoles/chemical synthesis , Indoles/pharmacology , Neurons/drug effects , Neurons/physiology , Animals , Apoptosis/drug effects , Chick Embryo , Choline O-Acetyltransferase/metabolism , Enzyme Inhibitors/pharmacology , Female , Humans , Indole Alkaloids , Motor Neurons/drug effects , Motor Neurons/physiology , Nerve Degeneration/drug effects , Nerve Growth Factors/antagonists & inhibitors , Prosencephalon/embryology , Prosencephalon/enzymology , Protein Kinase C/antagonists & inhibitors , Proto-Oncogene Proteins , Rats , Receptor Protein-Tyrosine Kinases , Receptor, trkA , Receptors, Nerve Growth Factor , Spinal Cord/embryology , Spinal Cord/enzymology , Substantia Innominata/cytology
16.
Eur J Cancer ; 31A(3): 380-8, 1995.
Article in English | MEDLINE | ID: mdl-7786606

ABSTRACT

T-25-Adh cells, cell variants derived from S49 mouse lymphoma, were transduced with a retrovirus containing the human MDR1 cDNA. The resultant cells (HU-1) are cross-resistant to colchicine, doxorubicin, vinblastine and actinomycin D, and their resistance to colchicine is reversed by verapamil. HU-1 cells were used to screen several protein kinase modulators for their ability to reverse multidrug resistance. Among the tested indole carbazole (K-252a) family of protein kinase inhibitors, only the antibiotic alkaloid KT-5720 (9-n-hexyl derivative of K-252a) could overcome the multidrug resistance of HU-1 cells and KB-V1 human carcinoma cells. Since other protein kinase A, C and G modulators did not reverse multidrug resistance in the tested multidrug-resistant cells, the chemosensitising activity of KT-5720 on these cells is apparently independent of its kinase inhibitory effects. Since KT-5720 fully reversed multidrug resistance at non-toxic concentrations, it might be a candidate for clinical chemosensitisation in combination chemotherapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Anti-Bacterial Agents/pharmacology , Carbazoles , Drug Resistance, Multiple/genetics , Indoles/pharmacology , Lymphoma/genetics , Protein Kinase C/antagonists & inhibitors , Pyrroles/pharmacology , Animals , Blotting, Southern , Carcinoma/genetics , Carcinoma/pathology , DNA, Complementary/genetics , Humans , Lymphoma/pathology , Mice , Transduction, Genetic , Tumor Cells, Cultured/drug effects
17.
Biochemistry ; 31(48): 12069-75, 1992 Dec 08.
Article in English | MEDLINE | ID: mdl-1333791

ABSTRACT

DNA topoisomerases have been shown to be important therapeutic targets in cancer chemotherapy. We found that KT6006 and KT6528, synthetic antitumor derivatives of indolocarbazole antibiotic K252a, were potent inducers of a cleavable complex with topoisomerase I. In DNA cleavage assay using purified calf thymus DNA topoisomerase I and supercoiled pBR322 DNA, KT6006 induced topoisomerase I mediated DNA cleavage in a dose-dependent manner at drug concentrations up to 50 microM, while DNA cleavage induced by KT6528 was saturated at 5 microM. The maximal amount of nicked DNA produced by KT6006 was more than 50% of substrate DNA, which was comparable to that of camptothecin. Heat treatment (65 degrees C) of the reaction mixture containing these compounds and topoisomerase I resulted in a substantial reduction in DNA cleavage, suggesting that topoisomerase I mediated DNA cleavage induced by KT6006 and KT6528 is through the mechanism of stabilizing the reversible enzyme-DNA "cleavable complex". Both KT6006 and KT6528 did not induce topoisomerase II mediated DNA cleavage in vitro. KT6006 and KT6528 were found to induce nearly identical topoisomerase I mediated DNA cleavage patterns, which was distinctly different from that with camptothecin. In contrast to the similarity between KT6006 and KT6528 in their structures and the nature of their cleavable complex with topoisomerase I, these drugs have different properties with respect to their interaction with DNA: KT6006 is a very weak intercalator whereas KT6528 is a strong intercalator with potentials comparable to that of adriamycin. These results indicate that KT6006 and KT6528 represent a new distinct class of mammalian DNA topoisomerase I active antitumor drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , DNA Topoisomerases, Type I/biosynthesis , Animals , Camptothecin/pharmacology , Catalysis , Cattle , DNA Helicases/metabolism , DNA, Superhelical/drug effects , DNA, Superhelical/metabolism , Enzyme Induction/drug effects , Hydrolysis , Indoles/pharmacology , Thymus Gland/enzymology
18.
Carbohydr Res ; 235: 95-114, 1992 Nov 04.
Article in English | MEDLINE | ID: mdl-1473115

ABSTRACT

The total synthesis of O-(O-[6-O-(2-aminoethylphosphono)-alpha-D-mannopyranosyl]-(1-->2)- O-alpha- D-mannopyranosyl-(1-->6)-O-[O-alpha-D-galactopyranosyl-(1-->6)-alpha-D- galactopyranosyl-(1-->3)]-O-alpha-D-mannopyranosyl-(1-->4)-2-amino-2-deo xy- alpha-D-glucopyranosyl)-(1-->6)-[1-O-(1,2-dimyristoyl-sn-glycero-3-phosp hono) - 1D-myo-inositol], the GPI anchor of Trypanosoma brucei was achieved for the first time. The core structure of the GPI molecule, the glycoheptaosyl part, was constructed in a highly stereocontrolled manner from O-[O-(2,4-di-O-benzyl-alpha-D-mannopyranosyl-(1-->4)-2-azido-3,6-di-O-be nzyl- 2-deoxy-D-glucopyranosyl]-(1-->6)-2,3,4,5-tetra-O-benzyl-1-O-(4- methoxybenzyl)-D-myo-inositol, O-(2,3,4,6-tetra-O-benzyl-alpha-D-galactopyranosyl)-(1-->6)-2,3,4-tri-O- benzyl-D-galactopyranosyl fluoride, 2-O-acetyl-3,4,6-tri-O-benzyl-alpha-D-mannopyranosyl chloride, and 6-O-acetyl-2,3,4-tri-O-benzyl-alpha-D-mannopyranosyl fluoride. The introduction of two phosphodiester functions was efficiently achieved using the H-phosphonate method.


Subject(s)
Glycosylphosphatidylinositols/chemical synthesis , Oligosaccharides/chemical synthesis , Trypanosoma brucei brucei , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Glycosylphosphatidylinositols/chemistry , Indicators and Reagents , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Oligosaccharides/chemistry , Stereoisomerism
19.
Carbohydr Res ; 234: 75-91, 1992 Oct 09.
Article in English | MEDLINE | ID: mdl-1468082

ABSTRACT

O-alpha-D-Mannopyranosyl-(1-->4)-O-2-amino-2-deoxy-alpha-D-glucopyranosy l- (1-->6)-1D-myo-inositol 1-(1,2-di-O-myristoyl-sn-glycer-3-yl hydrogen phosphate), a part structure of the glycosyl-phosphatidylinositol (GPI) anchor of Trypanosoma brucei, was synthesised efficiently by the phosphonate approach. The glycobiosylinositol core was prepared in a stereocontrolled manner from 1D-2,3,4,5-tetra-O-benzyl-1-O-(4-methoxybenzyl)-myo-inositol, tert-butyldimethylsilyl 2-azido-3,6-di-O-benzyl-2-deoxy-alpha-D-glucopyranoside, and methyl 3,6-di-O-acetyl-2,6-di-O-benzyl-2-thio-alpha-D-mannopyranoside.


Subject(s)
Glycosylphosphatidylinositols/chemistry , Glycosylphosphatidylinositols/chemical synthesis , Trypanosoma brucei brucei/chemistry , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Glycosylation , Molecular Sequence Data , Molecular Structure
20.
Cancer Chemother Pharmacol ; 29(4): 266-72, 1992.
Article in English | MEDLINE | ID: mdl-1537071

ABSTRACT

Novel derivatives of K-252a, (8R*,9S*,11S*)-(-)-9-hydroxy-9-methoxycarbonyl- 8-methyl-2,3,9,10-tetrahydro-8,11-epoxy-1H,8H,11H-2,7b,11a-triazadibe nzo[a,g]-cycloocta[cde]trinden-1-one, an inhibitor of protein kinases and calmodulin-dependent phosphodiesterase, were synthesized and evaluated for their antitumor activity in vitro and in vivo. Of ten derivatives tested, four were active against the P388 murine leukemia i.p.-i.p. system, although K-252a was inactive. Among these derivatives, KT6124 was selected for further biological evaluation studies because its efficacy was the highest. KT6124 was also active against sarcoma 180 and B16 melanoma. It exerted a relatively broad spectrum of antiproliferative activity against 20 human tumor cell lines in vitro. To determine the mechanism(s) of action underlying the antitumor activity of KT6124, we tested the drug for inhibition of protein kinases, including Ca(2+)- and phospholipid-dependent protein kinase (PKC), in intact A431 human epidermoid carcinoma cells in comparison with the PKC-inhibitory activity of K-252a. KT6124 did not antagonize the action of phorbol 12-myristate 13-acetate (PMA) in A431 cells, whereas K-252a did, suggesting that KT6124 may not act on protein kinases in the cells. The interaction of KT6124 with DNA in living cells was examined by the alkaline elution method. KT6124 apparently exhibited DNA scission both dose- and time-dependently in the target cells. The DNA breakage was dependent on proteinase K treatment, suggesting its possible interaction with DNA-related enzyme(s). These results indicate that KT6124 exerts antitumor activity by acting on DNA or on DNA-related enzyme(s) in tumor cells rather than via the inhibition of protein kinases.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Indoles/pharmacology , Protein Kinase C/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Carbazoles/therapeutic use , Cell Division/drug effects , DNA Damage , DNA, Neoplasm/drug effects , DNA, Single-Stranded/drug effects , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , ErbB Receptors/drug effects , Humans , Indole Alkaloids , Indoles/therapeutic use , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...