Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Clin Cancer Res ; 7(8): 2237-45, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489797

ABSTRACT

During the progression of prostate cancer, molecular changes occur resulting in the autocrine production of a series of neurotrophins by the malignant cells. This is coupled with expression of high-affinity cognate receptors for these ligands, termed trk receptors, by these cancer cells. The binding of the neurotrophins to their trk receptors activates the receptor's latent tyrosine kinase activity inducing a series of signal transduction pathways within these prostate cancer cells. These molecular changes result in the acquisition by prostate cancer cells of a restricted requirement for these trk signaling pathways for optimal survival. CEP-701 is an indolocarbazole compound specifically designed as a potent inhibitor (IC(50), 4 nM) of the tyrosine kinase activity of the trk receptors required for initiation of these survival pathways. In the present studies, the consequences of CEP-701 inhibition of these trk signaling survival pathways were tested in vivo using both rat (R3327 AT 6.3 and H) and human (TSU-pr1 and CWR-22Rv1) prostatic cancer models. These in vivo studies demonstrated that treatment with CEP-701 inhibits the growth of both rodent and human prostate cancers, without being toxic to the normal tissue including the host prostate. Because of this selective effect, CEP-701 inhibits metastasis and growth of both primary and metastatic sites of prostate cancer. Based upon this profile, long-term survival studies were performed using the slow-growing Dunning H rat prostate cancer model. For these latter studies, the dosing regimen was 10 mg CEP-701/kg/dose twice a day via gavage 5 days a week. This regimen maintains CEP-701 tumor tissue concentrations of 25-50 nM. Such chronic dosing increased (P < 0.001) the median survival of rats bearing the slow growing H prostate cancers from 408 days (395-432 days, 95% confidence interval) for the vehicle group (n = 18) to 566 days (497-598 days, 95% confidence interval) for the CEP-701-treated group (n = 24).


Subject(s)
Apoptosis/drug effects , Indoles , Neoplasm Metastasis/prevention & control , Prostatic Neoplasms/drug therapy , Receptor, trkA/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Cell Division/drug effects , Cell Line , Disease Models, Animal , Furans , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis/pathology , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/drug effects , Prostatic Neoplasms/pathology , Rats , Survival Analysis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Clin Cancer Res ; 5(11): 3594-602, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10589776

ABSTRACT

Neuroblastoma (NBL) and medulloblastoma (MBL) are tumors of the neuroectoderm that occur in children. NBL and MBL express Trk family tyrosine kinase receptors, which regulate growth, differentiation, and cell death. CEP-751 (KT-6587), an indolocarbazole derivative, is an inhibitor of Trk family tyrosine kinases at nanomolar concentrations. This study was designed to determine the effect of CEP-751 on the growth of NBL and MBL cell lines as xenografts. In vivo studies were conducted on four NBL cell lines (IMR-5, CHP-134, NBL-S, and SY5Y) and three MBL cell lines (D283, D341, and DAOY) using two treatment schedules: (a) treatment was started after the tumors were measurable (therapeutic study); or (b) 4-6 days after inoculation, before tumors were palpable (prevention study). CEP-751 was given at 21 mg/kg/dose administered twice a day, 7 days a week; the carrier vehicle was used as a control. In therapeutic studies, a significant difference in tumor size was seen between treated and control animals with IMR-5 on day 8 (P = 0.01), NBL-S on day 17 (P = 0.016), and CHP-134 on day 15 (P = 0.034). CEP-751 also had a significant growth-inhibitory effect on the MBL line D283 (on day 39, P = 0.031). Inhibition of tumor growth of D341 did not reach statistical significance, and no inhibition was apparent with DAOY. In prevention studies, CEP-751 showed a modest growth-inhibitory effect on IMR5 (P = 0.062) and CHP-134 (P = 0.049). Furthermore, inhibition of growth was greater in the SY5Y cell line transfected with TrkB compared with the untransfected parent cell line expressing no detectable TrkB. Terminal deoxynucleotidyl transferase-mediated nick end labeling studies showed CEP-751 induced apoptosis in the treated CHP-134 tumors, whereas no evidence of apoptosis was seen in the control tumors. Finally, there was no apparent toxicity identified in any of the treated mice. These results suggest that CEP-751 may be a useful therapeutic agent for NBL or MBL.


Subject(s)
Antineoplastic Agents/therapeutic use , Carbazoles/therapeutic use , Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Neuroblastoma/drug therapy , Animals , Antineoplastic Agents/toxicity , Carbazoles/toxicity , Cell Division/drug effects , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/prevention & control , Child , Female , Humans , Medulloblastoma/pathology , Medulloblastoma/prevention & control , Mice , Mice, Nude , Neuroblastoma/pathology , Transplantation, Heterologous , Tumor Cells, Cultured
3.
Proc Natl Acad Sci U S A ; 96(22): 12536-41, 1999 Oct 26.
Article in English | MEDLINE | ID: mdl-10535957

ABSTRACT

Human cytomegalovirus (CMV), a herpesvirus that causes congenital disease and opportunistic infections in immunocompromised individuals, encodes functions that facilitate efficient viral propagation by altering host cell behavior. Here we show that CMV blocks apoptosis mediated by death receptors and encodes a mitochondria-localized inhibitor of apoptosis, denoted vMIA, capable of suppressing apoptosis induced by diverse stimuli. vMIA, a product of the viral UL37 gene, inhibits Fas-mediated apoptosis at a point downstream of caspase-8 activation and Bid cleavage but upstream of cytochrome c release, while residing in mitochondria and associating with adenine nucleotide translocator. These functional properties resemble those ascribed to Bcl-2; however, the absence of sequence similarity to Bcl-2 or any other known cell death suppressors suggests that vMIA defines a previously undescribed class of anti-apoptotic proteins.


Subject(s)
Apoptosis/genetics , Cytomegalovirus/genetics , Viral Structural Proteins/genetics , Cell Line , Cytomegalovirus/physiology , HeLa Cells , Humans , Virus Replication/genetics
4.
Clin Cancer Res ; 5(8): 2205-12, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10473107

ABSTRACT

The aggressive behavior and poor prognosis of pancreatic ductal adenocarcinoma (PDAC) is associated with an increased expression of many growth factors and their cognate receptors. We have previously demonstrated the aberrant expression of the Trk receptors (Trks A, B, and C), enhanced tumor stromal expression of neurotrophins in primary PDAC specimens and human PDAC-derived cell lines, and a dose-dependent biological response of PDAC cells (in vitro invasiveness) to selective neurotrophins (Miknyoczki, S. J., et al., Int. J. Cancer, 81: 417-427, 1999). On the basis of these data, we have evaluated the therapeutic potential of inhibiting neurotrophin-Trk interactions using a selective and potent Trk tyrosine kinase inhibitor (CEP-701) in several preclinical models of human PDAC. CEP-701 is currently approved for clinical trials within the United States We demonstrate that CEP-701 administration at 10 mg/kg s.c. b.i.d. 5 days a week for 21-28 days inhibited tumor growth in a statistically significant manner in Panc-1, AsPc-1, BxPc-3, Colo 357, and MiaPaCa2 s.c. xenografts in athymic nude mice compared with vehicle-treated controls. Reductions in tumor growth volume of 50-70% relative to vehicle-treated controls were observed in xenografts responsive to CEP-701 administration. Significant reductions of in vivo PDAC tumor invasiveness were likewise observed in four of six CEP-701-treated rat tracheal xenografts implanted s.c. in athymic nude mice. The antitumor efficacy of CEP-701 was observed in the absence of pronounced morbidity or toxicity in vivo. Taken together, these data suggest that CEP-701 may be effective as a potential therapeutic agent in the treatment or management of PDAC.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Carbazoles/therapeutic use , Indoles , Pancreatic Neoplasms/drug therapy , Receptor, trkA/antagonists & inhibitors , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Animals , Body Weight/drug effects , Drug Screening Assays, Antitumor , Female , Furans , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , Neoplasm Transplantation , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Rats , Time Factors , Trachea/drug effects , Trachea/pathology , Treatment Outcome , Tumor Cells, Cultured
5.
Ann N Y Acad Sci ; 880: 252-62, 1999 Jun 30.
Article in English | MEDLINE | ID: mdl-10415871

ABSTRACT

The survival rate for patients with pancreatic ductal adenocarcinoma (PDAC) is among the poorest for all cancers. The factors that contribute to this poor prognosis are lack of effective early detection, high rate of metastases and a generally refractory response to available treatment modalities. The most commonly used treatment methods--chemotherapy and radiation therapy--are mainly used for symptom palliation, with surgery being the only "curative" treatment option. The use of combinations of treatment modalities is the only therapy available to patients with locally advanced disease or that which is surgically unresectable. These options are still not sufficient to increase patient survival time significantly. The aggressive behavior and poor prognosis of this cancer is associated with an increased expression of many growth factors and their cognate receptors. We have demonstrated previously the aberrant expression of the Trk receptors (Trks A, B, and C) in PDAC specimens and human PDAC-derived cell lines and a biphasic, dose-dependent response of specific neurotrophic agents on the in vitro invasiveness of PDAC cells. Based on these data we have evaluated the therapeutic potential of inhibiting neurotrophin-Trk interactions using a selective Trk tyrosine kinase inhibitor (CEP-701) on subcutaneous (s.c.) and tracheal xenografts derived from the poorly differentiated PDAC cell line, Panc1. We demonstrate that CEP-701 administration at 10 mg/kg s.c. BID for 21 days inhibited tumor growth of the Panc1 s.c. xenografts in a statistically-significant manner (p < 0.01) compared to vehicle controls, in the absence of morbidity and mortality. A T/C value of 25% was observed for CEP-701-treated s.c. xenografts. In addition, CEP-701 administration inhibited tumor cell invasion in the s.c. tracheal xenograft model of in vivo invasiveness. Taken together, these data suggest that further studies are warranted to evaluate CEP-701 as a potential therapeutic agent in the treatment of PDAC.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Indoles , Pancreatic Neoplasms/drug therapy , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/chemistry , Carbazoles/chemistry , Enzyme Inhibitors/chemistry , Female , Furans , Humans , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasm Invasiveness , Neoplasm Transplantation , Pancreatic Neoplasms/pathology , Rats , Trachea
6.
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
7.
Int J Cancer ; 81(3): 417-27, 1999 May 05.
Article in English | MEDLINE | ID: mdl-10209957

ABSTRACT

The aggressive and highly metastatic behavior observed in pancreatic ductal adenocarcinoma (PDAC) may be due to autocrine and/or paracrine interactions (tumor/stromal) involving altered expression of peptide growth factors and their corresponding receptors. The neurotrophin (NT) growth factor family and their cognate receptors have been demonstrated to play a role in the invasiveness, chemotactic behavior and tumor cell survival of both neuronal and non-neuronal cancers. We hypothesized that aberrant expression of the NTs and/or the Trk receptors may contribute to the malignant phenotype of PDAC, specifically tumor cell invasiveness, through autocrine and/or paracrine interactions. In this study, we examined the expression of NTs, Trks and p75NGFR by immunohistochemical and in situ hybridization analyses in both normal (n=14) and neoplastic pancreas (n=47) and PDAC-derived cell lines (n=6). Further, we evaluated the effects of various NTs on the in vitro invasive and chemotactic behavior on 6 human PDAC-derived cell lines in a modified Boyden chamber assay. Brain-derived nerve growth factor (BDNF), NT-3, NT-4/5 and Trks A, B and C exhibited diffuse cytoplasmic and membranous immunostaining patterns in both the ducts and the acini of the exocrine pancreas and the islets of the endocrine pancreas of both normal and PDAC specimens. NT expression was primarily within the stromal compartment of the tumor, while Trk expression was weak or absent. We observed a 68%, 64% and 66% increase in the expression of Trks A, B and C, respectively, in the ductal elements of the PDAC samples examined compared with the normal adjacent tissue. Invasiveness of 4 of 6 PDAC cell lines was significantly inhibited (p<0.05) when the cells were incubated with 100 ng/ml NT. However, when select cell lines were incubated with lower concentrations of NT-3 and BDNF (0, 1, 5, 25 and 50 ng/ml), invasiveness was significantly stimulated (p<0.05) through the Matrigel matrix. Collectively, our data suggest the possibility that paracrine and/or autocrine NT-Trk interactions may influence the phenotype (possibly the invasive behavior) of PDAC.


Subject(s)
Adenocarcinoma/chemistry , Nerve Growth Factors/analysis , Pancreatic Ducts , Pancreatic Neoplasms/chemistry , Proto-Oncogene Proteins/analysis , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Nerve Growth Factor/analysis , 3T3 Cells , Adenocarcinoma/pathology , Animals , Brain-Derived Neurotrophic Factor/analysis , Humans , Mice , Neoplasm Invasiveness , Nerve Growth Factors/pharmacology , Neurotrophin 3 , Pancreatic Neoplasms/pathology , Receptor, Ciliary Neurotrophic Factor , Receptor, trkA , Receptor, trkC
8.
Cancer Res ; 59(3): 533-7, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9973195

ABSTRACT

Apoptosis is regulated by interaction of viral and cellular BCL-2 family antiapoptotic proteins with various pro-apoptotic proteins, several of which are also members of the BCL-2 family. Cellular protein BNIP3 is a BCL-2 family proapoptotic protein that interacts with viral antiapoptosis proteins such as adenoviruses E1B-19K and EBV-BHRF1 and cellular antiapoptosis proteins such as BCL-2 and BCL-xL. Database searches indicate that the human genome encodes an open reading frame for a protein, BNIP3alpha, that shares substantial homology with BNIP3. The BNIP3alpha open reading frame encodes a protein of 219 amino acids that contains a conserved BH3 domain and a COOH-terminal trans-membrane domain, characteristic of several BCL-2 family proapoptotic proteins. BNIP3alpha interacts with viral antiapoptosis protein E1B-19K and cellular antiapoptosis proteins BCL-2 and BCL-xL. Overexpression of BNIP3alpha in transfected cells results in apoptosis and suppresses the antiapoptosis activity of E1B-19K and BCL-xL. Like BNIP3, BNIP3alpha seems to be predominantly localized in mitochondria. These results suggest that BNIP3alpha is a structural and functional homologue of BNIP3. BNIP3 and BNIP3alpha seem to be the first examples of homologues among the various human proapoptotic proteins. Northern blot analysis reveals that BNIP3alpha is expressed ubiquitously in most human tissues. In contrast, BNIP3 is expressed well in several human tissues and less abundantly in certain tissues such as placenta and lung. These results suggest that although BNIP3 and BNIP3alpha may promote apoptosis simultaneously in most human tissues, BNIP3alpha may play a more universal role.


Subject(s)
Apoptosis/physiology , Membrane Proteins/genetics , Mitochondria/metabolism , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Adenovirus E1B Proteins/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , COS Cells , Cricetinae , Humans , Membrane Proteins/metabolism , Membrane Proteins/physiology , Molecular Sequence Data , Proto-Oncogene Proteins c-bcl-2/metabolism , Sequence Homology, Amino Acid , Subcellular Fractions/metabolism , Transfection , Tumor Cells, Cultured , bcl-X Protein
9.
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
10.
J Comp Neurol ; 398(3): 373-92, 1998 Aug 31.
Article in English | MEDLINE | ID: mdl-9714150

ABSTRACT

Cell survival, death, and stress signals are transduced from the cell surface to the cytoplasm and nucleus via a cascade of phosphorylation events involving the mitogen-activated protein kinase (MAPK) family. We compared the distribution of p42 mitogen-activated protein kinase (p42MAPK) and its activator MAPK or ERK kinase (MEK1; involved in transduction of growth and differentiation signals), with c-Jun N-terminal kinase (JNK1) and its activator MEK4 (involved in transduction of stress and death signals) in the adult rat central nervous system. All four kinases were present in the cytoplasm, dendrites, and axons of neurons. The presence of p42MAPK and JNK1 in dendrites and axons, as well as in cell bodies, suggests a role for these kinases in phosphorylation and regulation of cytoplasmic targets. A high degree of correspondence was found between the regional distribution of MEK1 and p42MAPK. Immunostaining for MEK1 and p42MAPK was intense in olfactory structures, neocortex, hippocampus, striatum, midline, and interlaminar thalamic nuclei, hypothalamus, brainstem, Purkinje cells, and spinal cord. In addition to neurons, p42MAPK was also present in oligodendrocytes. Whereas MEK4 was ubiquitously distributed, JNK1 was more selective. Immunostaining for MEK4 and JNK1 was intense in the olfactory bulb, lower cortical layers, the cholinergic basal forebrain, most nuclei of the thalamus, medial habenula, and cranial motor nuclei. The distribution of MEK1 and p42MAPK proteins only partially overlapped with that of MEK4 and JNK1. This suggests that the growth/differentiation and death/stress pathways affected by these kinases may not necessarily act to counterbalance each other in response to extracellular stimuli. The differential distribution of these kinases may control the specificity of neuronal function to extracellular signals.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Central Nervous System/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Blotting, Western , Central Nervous System/enzymology , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Signal Transduction/physiology
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.
Arch Biochem Biophys ; 349(2): 267-74, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9448714

ABSTRACT

The kinetic mechanism of the trk receptor-linked tyrosine kinase was determined using a baculovirus expressed trk kinase domain and a bacterially expressed phospholipase C-gamma/glutathione S-transferase (PLC-gamma/ GST) fusion protein as substrate. Product and dead-end inhibition studies indicate an ordered association of substrates to trkA kinase with the nucleotide ATP binding prior to the exogenous substrate PLC-gamma/GST, followed by release of the phosphorylated PLC-gamma/GST product prior to release of ADP (sequential ordered bi-bi mechanism). This is in contrast to the reported kinetic mechanisms of closely related EGF receptor and insulin receptor kinases which appear to proceed via a rapid equilibrium random mechanism. The indolocarbazole K-252a, which was previously shown to be a potent and relatively selective inhibitor of trk kinase activity, acts as a competitive inhibitor with respect to ATP. The data suggest that potent and selective kinase inhibitors can be rationally designed by exploring subtle variations surrounding the nucleotide binding sites of receptor tyrosine kinases.


Subject(s)
Carbazoles/pharmacology , Glutathione Transferase/metabolism , Isoenzymes/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Nerve Growth Factor/metabolism , Type C Phospholipases/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Cell Line , Humans , Indole Alkaloids , Kinetics , Phospholipase C gamma , Phosphorylation , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, trkA , Receptors, Nerve Growth Factor/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Spodoptera , Substrate Specificity , Transfection
13.
J Neurochem ; 70(2): 558-71, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9453550

ABSTRACT

In vitro studies indicate that p42/p44MAPK phosphorylate both nuclear and cytoplasmic proteins. However, the functional targets of p42/p44MAPK activation in vivo remain unclear. To address this question, we localized activated p42/p44MAPK in hippocampus and cortex and determined their signaling effects after electroconvulsive shock treatment (ECT) in rats. Phosphorylated p42/p44MAPK content increased in the cytoplasm of hippocampal neurons in response to ECT. Consistent with this cytoplasmic localization, inhibition of ECT-induced p42/p44MAPK activation by the extracellular signal-regulated kinase kinase inhibitor PD098059 blocked phosphorylation of the cytoplasmic protein microtubule-associated protein 2c (MAP2c), but failed to inhibit the induction of the nuclear protein c-Fos in response to ECT. In contrast to hippocampal neurons, cortical neurons exhibited an increase in amount of phosphorylated p42/p44MAPK in both the nucleus and cytoplasm after ECT. Accordingly, PD098059 blocked the induction of Fos-like immunoreactivity in the nuclei of cortical neurons as well as MAP2c phosphorylation in the cytoplasm. Our data indicate that both nuclear and cytoplasmic substrates can be activated by p42/p44MAPK in vivo. However, the functional targets of p42/p44MAPK signaling depend on the precise location of p42/p44MAPK within different subcellular compartments of brain regions. These results indicate unique functional pathways of p42/p44MAPK-mediated signal transduction within different brain regions in vivo.


Subject(s)
Brain/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Signal Transduction/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Nucleus/physiology , Cerebral Cortex/physiology , Cytoplasm/metabolism , Electroshock , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hippocampus/physiology , MAP Kinase Kinase 1 , Microtubule-Associated Proteins/metabolism , Mitogen-Activated Protein Kinase 3 , Organ Specificity , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Rats, Sprague-Dawley
14.
J Neurosci ; 18(1): 104-11, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9412490

ABSTRACT

Neurons undergoing apoptosis can be rescued by trophic factors that simultaneously increase the activity of extracellular signal-regulated kinase (ERK) and decrease c-Jun N-terminal kinase (JNK) and p38. We identified a molecule, CEP-1347 (KT7515), that rescues motoneurons undergoing apoptosis and investigated its effect on ERK1 and JNK1 activity. Cultured rat embryonic motoneurons, in the absence of trophic factor, began to die 24-48 hr after plating. During the first 24 hr ERK1 activity was unchanged, whereas JNK1 activity increased fourfold. CEP-1347 completely rescued motoneurons for at least 72 hr with an EC50 of 20 +/- 2 nM. CEP-1347 did not alter ERK1 activity but rapidly inhibited JNK1 activation. The IC50 of CEP-1347 for JNK1 activation was the same as the EC50 for motoneuron survival. Inhibition of JNK1 activation by CEP-1347 was not selective to motoneurons. CEP-1347 also inhibited JNK1 activity in Cos7 cells under conditions of ultraviolet irradiation, osmotic shock, and inhibition of glycosylation. Inhibition by CEP-1347 of the JNK1 signaling pathway appeared to be selective, because CEP-1347 did not inhibit p38-regulated mitogen-activated protein kinase-activated protein kinase-2 (MAPKAP2) activity in Cos7 cells subjected to osmotic shock. The direct molecular target of CEP-1347 was not JNK1, because CEP-1347 did not inhibit JNK1 activity in Cos7 cells cotransfected with MEKK1 and JNK1 cDNA constructs. This is the first demonstration of a small organic molecule that promotes motoneuron survival and that simultaneously inhibits the JNK1 signaling cascade.


Subject(s)
Apoptosis/drug effects , Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Motor Neurons/cytology , Protein Kinase Inhibitors , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carbazoles/chemical synthesis , Cell Survival/drug effects , Choline O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Fetus/cytology , Gene Expression , Imidazoles/pharmacology , Indole Alkaloids , MAP Kinase Kinase 4 , Motor Neurons/enzymology , Motor Neurons/ultrastructure , Neurites/physiology , Protein Kinases/genetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , p38 Mitogen-Activated Protein Kinases
15.
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
16.
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
17.
Anal Biochem ; 247(2): 376-81, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9177701

ABSTRACT

The high-affinity receptor for nerve growth factor (NGF), trkA, is a receptor-linked tyrosine kinase. The binding of NGF to trkA, depending on the context of its environment, can cause beneficial or deleterious responses in the target cells. For example, the activation of trkA in sympathetic and sensory neurons causes the subsequent survival and differentiation of these cells. On the other hand, the activation of trkA by NGF in other cells has been implicated in several pathologies including inflammation-induced hyperalgesia and several cancers. A radioactive binding assay to evaluate inhibitors of the kinase domain of trkA has been developed and validated. The assay monitors the specific binding of an inhibitor of trkA kinase activity, the indolocarbazole K-252a, to the trkA receptor. [3H]K-252a binds with high affinity to one site on the cytoplasmic kinase domain of the trkA receptor. Binding is saturable and reversible with a dissociation constant (Kd) of 1.5 nM. The binding assay has been used in competition binding experiments to determine the inhibition constants for other indolocarbazole compounds. The IC50 values for compounds obtained in the binding assay correlate very well with the IC50 values obtained in an enzyme-linked immunosorbent assay for trkA tyrosine kinase activity.


Subject(s)
Carbazoles/analysis , Enzyme Inhibitors/analysis , Proto-Oncogene Proteins/antagonists & inhibitors , Radioligand Assay/methods , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Nerve Growth Factor/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Binding, Competitive , Carbazoles/metabolism , Enzyme Inhibitors/metabolism , Evaluation Studies as Topic , Humans , In Vitro Techniques , Indole Alkaloids , Kinetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, trkA , Receptors, Nerve Growth Factor/metabolism
18.
J Neurochem ; 68(1): 88-94, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8978713

ABSTRACT

K-252b potentiates the neurotrophic effects of neurotrophin-3 (NT-3) in primary cultures of rat central cholinergic and peripheral sensory neurons and in a rat pheochromocytoma PC12 cell line. The ligand and receptor specificity, and role of the low-affinity neurotrophin receptor (p75NTR) in the potentiation response induced by K-252b, are unknown. To address the issues of ligand and receptor specificity of K-252b potentiation, we have examined neurotrophin-induced DNA synthesis ([3H]-thymidine incorporation) in NIH3T3 cells expressing trkA, trkB, or trkC. Neither NT-3 nor K-252b alone could stimulate mitogenic activity in the trkA-overexpressing clone. However, coaddition of K-252b (EC50 of approximately 2 nM) with 10-100 ng/ml NT-3 led to incorporation of [3H]thymidine in trkA expressing cells to a level induced by optimal concentrations of nerve growth factor (NGF). The K-252b- and NT-3-induced [3H]thymidine incorporation correlated with an increase in the tyrosine autophosphorylation of the trkA receptor as well as tyrosine phosphorylation of trk-associated phospholipase C-gamma 1 and SH2-containing proteins. K-252b did not potentiate submaximal doses of NGF, or maximal doses of brain-derived neurotrophic factor (BDNF) or neurotrophin-4/5 (NT-4/ 5) in trkA-expressing cells. Furthermore, K-252b did not potentiate DNA synthesis by submaximal doses of BDNF, NT-4/5, or NT-3 in trkB- or trkC-expressing NIH3T3 cells, suggesting that the potentiation profile for K-252b was specific for NT-3 in trkA-expressing cells. We found no expression of p75NTR in the trk-expressing NIH3T3 cells. This is the first demonstration that K-252b potentiates a trkA-mediated biological nonneuronal response by NT-3 that occurs independent of p75NTR and appears to be both ligand and receptor specific.


Subject(s)
Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Nerve Growth Factors/pharmacology , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/metabolism , 3T3 Cells , Animals , Drug Synergism , Humans , Indole Alkaloids , Ligands , Mice , Molecular Weight , Neurotrophin 3 , Phosphorylation , Rats , Receptor, trkA/genetics , Receptors, Nerve Growth Factor/physiology , Substrate Specificity , Transfection , Tumor Cells, Cultured
19.
Cancer Res ; 57(1): 176-82, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-8988061

ABSTRACT

To better understand the effects of p53 on the process of DNA damage-induced cell death, we examined the influence of p53 status on the rate of the onset and the overall extent of cell death induced by doxorubicin. We performed this study with Rat-1 fibroblasts, with Rat-1/myc cells which constitutively express c-Myc, and with Rat-1/myc/p53His175 cells derived from Rat-1/myc cells, which, in addition, express the full-length dominant-negative p53His175 mutant gene. The p53His175 mutant suppresses the transactivation function of endogenous p53 in these cells. In contrast to the parental Rat-1 cells, which exhibited only low levels of apoptosis within the first 24 h of treatment with 0.1 to 1 microM doxorubicin, similarly treated Rat-1/myc cells underwent massive and rapid apoptosis. Introduction of p53His175 into Rat-1/myc cells reversed this effect, indicating that Myc-accelerated doxorubicin-induced apoptosis requires functional p53. However, when the overall extent of cell death was measured using clonogenic assays, we found that greater than 90% of cells did not survive upon a 24-h pretreatment with doxorubicin at a concentration as low as 0.1 microM. Moreover, the effect of doxorubicin on all three cell lines was similar, irrespective of their p53 or c-Myc status. Taken together, our experiments indicate that: (a) constitutive expression of c-Myc accelerates the onset of doxorubicin-induced apoptosis in Rat-1 fibroblasts; (b) wild-type p53 function is necessary for this acceleration; and (c) neither overexpression of c-Myc nor the p53 status influences the overall extent of doxorubicin-induced cell death.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/physiology , Doxorubicin/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Down-Regulation , Genetic Vectors/genetics , Rats , Transfection , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/genetics
20.
Oncogene ; 13(4): 739-48, 1996 Aug 15.
Article in English | MEDLINE | ID: mdl-8761295

ABSTRACT

A fusion gene consisting of wild-type p53 linked to a modified ligand binding domain of the murine estrogen receptor has been constructed and should be a useful tool for studying controlled activation of wild-type p53 function in a variety of experimental cell systems. The protein product of this gene, p53ERTM, is expressed in cells constitutively but is not functional unless associated with tamoxifen or 4-hydroxytamoxifen. p53ERTM was introduced into p53-deficient mouse embryo fibroblasts (MEFs) expressing the E1A and T24 H-ras oncogenes. Activation of p53 in these transformed cells by the addition of tamoxifen or 4-hydroxytamoxifen resulted in apoptosis. In addition to engaging the apoptotic machinery, the tamoxifen-activated fusion protein exhibited other functions characteristic of wild-type p53, such as induction of WAF1 and MDM2 gene expression and activation of the p53-dependent spindle checkpoint in cells treated with nocodazole. Activation of p53ERTM expressed in p53-positive MEFs coexpressing E1A and ras had, at most, only a small cytotoxic effect. When three cell lines of transformed p53+/+ fibroblasts not expressing p53ERTM were tested for sensitivity to the DNA-damaging drug doxorubicin, the p53+/+ clones displayed either comparable sensitivity, or at most an increase in drug sensitivity of less than fourfold, as compared to several p53-/- cell lines. Our data show that restoration of wild-type p53 activity is sufficient to trigger apoptosis in p53-/- MEFs transformed with E1A and T24 H-ras and suggest that rare propagable clones of p53-normal MEFs expressing the E1A and T24 H-ras oncogenes have suffered compensatory alterations that compromise the ability to undergo p53-dependent apoptosis.


Subject(s)
Adenovirus E1A Proteins/genetics , Apoptosis/genetics , Genes, ras , Recombinant Fusion Proteins/genetics , Tamoxifen/analogs & derivatives , Animals , Cell Line, Transformed , Embryo, Mammalian/cytology , Estrogen Antagonists/pharmacology , Fibroblasts , Gene Expression Regulation , Mice , Receptors, Estrogen/genetics , Tamoxifen/pharmacology , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL