Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Cancer Prev Res (Phila) ; 4(9): 1419-25, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21680702

ABSTRACT

The phytochemical resveratrol has undergone extensive preclinical investigation for its putative cancer chemopreventive properties. Low systemic availability of the parent compound due to rapid and extensive metabolism may confound its usefulness as a potential agent to prevent malignancies in organs remote from the site of absorption. Micronization allows increased drug absorption, thus increasing availability. Here we describe a pilot study of SRT501, micronized resveratrol, given as 5.0 g daily for 14 days, to patients with colorectal cancer and hepatic metastases scheduled to undergo hepatectomy. The purpose of the study was to assess the safety, pharmacokinetics, and pharmacodynamics of the formulation. SRT501 was found to be well tolerated. Mean plasma resveratrol levels following a single dose of SRT501 administration were 1,942 ± 1,422 ng/mL, exceeding those published for equivalent doses of nonmicronized resveratrol by 3.6-fold. Resveratrol was detectable in hepatic tissue following SRT501 administration (up to 2,287 ng/g). Cleaved caspase-3, a marker of apoptosis, significantly increased by 39% in malignant hepatic tissue following SRT501 treatment compared with tissue from the placebo-treated patients. SRT501 warrants further clinical exploration to assess its potential clinical utility.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Liver Neoplasms/drug therapy , Stilbenes/pharmacology , Stilbenes/pharmacokinetics , Aged , Antineoplastic Agents, Phytogenic/administration & dosage , Caspase 3/metabolism , Combined Modality Therapy/methods , Double-Blind Method , Female , Hepatectomy/methods , Humans , Male , Middle Aged , Neoplasm Metastasis , Pilot Projects , Placebos , Postoperative Complications , Resveratrol , Stilbenes/administration & dosage , Titanium/pharmacology
2.
Neurotox Res ; 5(7): 505-14, 2003.
Article in English | MEDLINE | ID: mdl-14715434

ABSTRACT

Anti-inflammatory treatment with the proteasome inhibitor MLN519 has been previously reported to be neuroprotective against ischemic brain injury in rats. These effects have been related to inhibition of the transcription factor NF-kappaB, which is activated through ubiquitin-proteasomal degradation. The aim of this study was to evaluate the effects of MLN519 to alter the expression of several inflammatory genes under the control of NF-kappaB. Male Sprague-Dawley rats underwent middle cerebral artery occlusion (MCAo) followed by vehicle or MLN519 (1.0 g/kg, i.v.) treatment immediately after reperfusion of blood to the brain at 2h. Gene expression was evaluated 3-72 h post-MCAo. The most striking effects of intravenous treatment with MLN519 were associated with reductions in ICAM-1 expression at 3 h followed by reductions in E-selectin (12-72 h). Less dramatic reductions were observed in IL-1Beta (3-24 h) and TNF-Alpha (24 h) with no apparent effects on IL-6 and VCAM-1 mRNA levels. Immunohistochemical analysis revealed that the genes most dramatically affected by MLN519 had highest expression in endothelial cells and leukocytes (E-selectin, ICAM-1),indicating that these cell types may be the primary targets of intravenously delivered MLN519 treatment.


Subject(s)
Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Cysteine Endopeptidases/drug effects , Infarction, Middle Cerebral Artery/pathology , Inflammation/metabolism , Middle Cerebral Artery/physiology , Multienzyme Complexes/drug effects , Neuroprotective Agents/pharmacology , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cytokines/biosynthesis , DNA/chemistry , DNA/genetics , DNA/isolation & purification , Gene Expression Regulation/drug effects , Immunohistochemistry , Infarction, Middle Cerebral Artery/drug therapy , Inflammation/genetics , Kinetics , Male , NF-kappa B/metabolism , Proteasome Endopeptidase Complex , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
3.
Br J Clin Pharmacol ; 54(3): 269-76, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12236847

ABSTRACT

AIMS: The main objective of this study was to investigate the safety, tolerability and pharmacodynamics of the novel proteasome inhibitor PS-519 in young male volunteers. Many pro-inflammatory mediators such as cytokines and cell adhesion molecules that are responsible for the development of the cerebral infarct are under the control of the transcription factor Nuclear Factor kappa-B (NF-kappaB). The activity of NF-kappaB is itself tightly regulated through the multicatalytic enzyme known as the proteasome. PS-519 is a novel and highly selective small molecule that inhibits the proteasome. An ex vivo assay of 20S proteasome activity allows monitoring of the drug effect in blood. PS-519 is protective in multiple animal models of cerebral ischaemia over a range of doses that achieve 20S inhibition of 40%-80%. METHODS: PS-519 has been administered to healthy male volunteers as single and repeated doses up to 1.6 mg m(-2). It was given as an intravenous bolus over 20-30 s in a double blind, randomized, placebo-controlled phase I study, examining vital signs, safety, tolerability and blood 20S proteasome inhibition. RESULTS: Thirty-nine subjects received single doses of 0.012 mg m-2-1.6 mg m(-2) and 28 subjects received doses of 0.5 mg m(-2)-1.6 mg m(-2) on three consecutive days. The drug was well tolerated. There was no clear treatment-emergent symptom or abnormality of laboratory tests. Proteasome inhibition in blood samples as measured by 20S assay achieved the intended maximum target level of 70-80% with 1.6 mg m(-2), and was reproducible with repeated dosing. CONCLUSIONS: This study has demonstrated that proteasome inhibition is well tolerated by healthy subjects at levels that are maximally neuroprotective in experimental conditions. Further clinical evaluation appears justified.


Subject(s)
Acetylcysteine/analogs & derivatives , Acetylcysteine/adverse effects , Multienzyme Complexes/antagonists & inhibitors , Acetylcysteine/pharmacokinetics , Bilirubin/metabolism , Blood Pressure/drug effects , Cysteine Endopeptidases , Double-Blind Method , Electrocardiography/drug effects , Heart Rate/drug effects , Humans , Leukocyte Count , Male , Neutrophils/drug effects , Proteasome Endopeptidase Complex
4.
Occup Environ Med ; 59(1): 13-7, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11836463

ABSTRACT

OBJECTIVES: To estimate exposures to cadmium (Cd) received by the United Kingdom population as a result of the dispersion of zinc Cd sulfide (ZnCdS) by the Ministry of Defence between 1953 and 1964, as a simulator of biological warfare agents. METHODS: A retrospective risk assessment study was carried out on the United Kingdom population during the period 1953-64. This determined land and air dispersion of ZnCdS over most of the United Kingdom, inhalation exposure of the United Kingdom population, soil contamination, and risks to personnel operating equipment that dispersed ZnCdS. RESULTS: About 4600 kg ZnCdS were dispersed from aircraft and ships, at times when the prevailing winds would allow large areas of the country to be covered. Cadmium released from 44 long range trials for which data are available, and extrapolated to a total of 76 trials to allow for trials with incomplete information, is about 1.2% of the estimated total release of Cd into the atmosphere over the same period. "Worst case" estimates are 10 microg Cd inhaled over 8 years, equivalent to Cd inhaled in an urban environment in 12100 days, or from smoking 100 cigarettes. A further 250 kg ZnCdS was dispersed from the land based sites, but significant soil contamination occurred only in limited areas, which were and have remained uninhabited. Of the four personnel involved in the dispersion procedures (who were probably exposed to much higher concentrations of Cd than people on the ground), none are suspected of having related illnesses. CONCLUSION: Exposure to Cd from dissemination of ZnCdS during the "cold war" should not have resulted in adverse health effects in the United Kingdom population.


Subject(s)
Cadmium/analysis , Environmental Exposure/analysis , Air Pollution/statistics & numerical data , Biological Warfare , Cadmium/adverse effects , Environmental Exposure/adverse effects , Humans , Residence Characteristics , Retrospective Studies , Risk Assessment , Risk Factors , Sulfides/adverse effects , Sulfides/analysis , United Kingdom , Zinc Compounds/adverse effects , Zinc Compounds/analysis
5.
Stroke ; 32(12): 2926-31, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11739997

ABSTRACT

BACKGROUND AND PURPOSE: The proteasome inhibitor PS-519 blocks activation of nuclear factor-kappaB, a major mediator of inflammation. We tested the hypothesis that combination treatment of recombinant human tissue plasminogen activator (rhtPA) and PS-519 extends the therapeutic window for treatment of stroke with rhtPA without increasing incidence of hemorrhagic transformation. METHODS: The middle cerebral artery (MCA) of male Wistar rats (n=56) was occluded by an embolus. After embolization, animals were randomly divided into the following groups: PS-519 treatment groups: PS-519 was given at 2, 4, or 6 hours after MCA occlusion; rhtPA treatment groups: rhtPA was given at 2 or 4 hours after MCA occlusion; combination treatment groups: PS-519 and rhtPA were given at 2, 4, or 6 hours after MCA occlusion; control group: the same volume of saline was given at 2 hours after MCA occlusion. RESULTS: Administration of PS-519 alone at 2 or 4 hours, but not 6 hours, significantly (P<0.05) reduced infarct volume and improved neurological recovery compared with the control group. Administration of rhtPA alone at 2 hours, but not 4 hours, significantly (P<0.05) reduced infarct volume and improved neurological recovery compared with the control group. Furthermore, combination treatment with rhtPA and PS-519 even at 6 hours significantly (P<0.05) reduced infarct volume, improved neurological recovery, and did not increase the incidence of hemorrhagic transformation compared with the control group or the group treated with PS-519 alone. CONCLUSIONS: Our data suggest that combination treatment with PS-519 and rhtPA extends the neuroprotective effect to at least 6 hours after embolization.


Subject(s)
Acetylcysteine/analogs & derivatives , Acetylcysteine/administration & dosage , Brain Ischemia/drug therapy , Cerebral Infarction/prevention & control , Intracranial Embolism/drug therapy , Tissue Plasminogen Activator/administration & dosage , Animals , Behavior, Animal/drug effects , Blood Pressure/drug effects , Body Weight/drug effects , Brain Ischemia/complications , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Count , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/prevention & control , Cerebral Infarction/etiology , Cerebral Infarction/pathology , Cysteine Endopeptidases , Cysteine Proteinase Inhibitors/administration & dosage , Disease Models, Animal , Drug Administration Schedule , Drug Therapy, Combination , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/adverse effects , Humans , Intracranial Embolism/complications , Intracranial Embolism/pathology , Male , Multienzyme Complexes/antagonists & inhibitors , Neurologic Examination , Peroxidase/metabolism , Proteasome Endopeptidase Complex , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Time Factors , Tissue Plasminogen Activator/adverse effects
6.
Am J Clin Pathol ; 116(5): 637-46, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11710679

ABSTRACT

The proteasome is an enzyme present in all cells, from yeast to human, and has a central role in the proteolytic degradation of the vast majority of intracellular proteins. Among the key proteins modulated by the proteasome are those involved in controlling inflammatory processes, cell cycle regulation, and gene expression. As such, agents that inhibit the proteasome have been shown to be active in numerous animal models of inflammation and cancer Two proteasome inhibitors are under clinical evaluation. PS-519 is being studied for the treatment of reperfusion injury that occurs following cerebral ischemia and myocardial infarction. The other, PS-341, has recently entered multiple phase 2 clinical trials for the treatment of multiple myeloma, chronic lymphocytic leukemia, and a variety of solid tumors. The proteasome may have an important role in the evolution of HIV-related disorders including AIDS and inflammatory disorders. Therapeutic strategies using proteasome inhibitors for the treatment of these conditions have now entered preclinical development.


Subject(s)
Acetylcysteine/analogs & derivatives , Acetylcysteine/therapeutic use , Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Cysteine Proteinase Inhibitors/therapeutic use , Multienzyme Complexes/antagonists & inhibitors , Neoplasms/drug therapy , Pyrazines/therapeutic use , Acetylcysteine/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Boronic Acids/pharmacokinetics , Bortezomib , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/enzymology , Clinical Trials, Phase I as Topic , Cysteine Endopeptidases , Cysteine Proteinase Inhibitors/pharmacokinetics , Disease Models, Animal , Humans , Neoplasms/enzymology , Proteasome Endopeptidase Complex , Pyrazines/pharmacokinetics
7.
J Cell Biochem ; 82(1): 110-22, 2001.
Article in English | MEDLINE | ID: mdl-11400168

ABSTRACT

The 26S proteasome degrades proteins that regulate transcription factor activation, cell cycle progression, and apoptosis. In cancer, this may allow for uncontrolled cell division, promoting tumor growth, and spread. We examined whether selective inhibition of the 26S proteasome with PS-341, a dipeptide boronic acid analogue, would block proliferation and induce apoptosis in human pancreatic cancer. Proteasome inhibition significantly blocked mitogen (FCS) induced proliferation of BxPC3 human pancreatic cancer cells in vitro, while arresting cell cycle progression and inducing apoptosis by 24 h. Accumulation of p21(Cip1-Waf-1), a cyclin dependent kinase (CDK) inhibitor normally degraded by the 26S proteasome, occurred by 3 h and correlated with cell cycle arrest. When BxPC3 pancreatic cancer xenografts were established in athymic nu/nu mice, weekly administration of 1 mg/kg PS-341 significantly inhibited tumor growth. Both cellular apoptosis and p21(Cip1-Waf-1) protein levels were increased in PS-341 treated xenografts. Inhibition of tumor xenograft growth was greatest (89%) when PS-341 was combined with the tumoricidal agent CPT-11. Combined CPT-11/PS-341 therapy, but not single agent therapy, yielded highly apoptotic tumors, significantly inhibited tumor cell proliferation, and blocked NF-kappaB activation indicating this systemic therapy was effective at the cancer cell level. 26S proteasome inhibition may represent a new therapeutic approach against this highly resistant and lethal malignancy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Cyclins/drug effects , NF-kappa B/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Peptide Hydrolases/drug effects , Proteasome Endopeptidase Complex , Pyrazines/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/physiology , Boronic Acids/metabolism , Bortezomib , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Cycle/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Dipeptides/metabolism , Dipeptides/pharmacology , Drug Resistance, Neoplasm , Humans , Irinotecan , Mice , Mice, Nude , Mitogens/administration & dosage , Pancreatic Neoplasms/physiopathology , Peptide Hydrolases/metabolism , Protease Inhibitors/pharmacology , Pyrazines/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
8.
Cancer Res ; 61(7): 3071-6, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306489

ABSTRACT

Human multiple myeloma (MM) is a presently incurable hematological malignancy, and novel biologically based therapies are urgently needed. Proteasome inhibitors represent a novel potential anticancer therapy. In this study, we demonstrate that the proteasome inhibitor PS-341 directly inhibits proliferation and induces apoptosis of human MM cell lines and freshly isolated patient MM cells; inhibits mitogen-activated protein kinase growth signaling in MM cells; induces apoptosis despite induction of p21 and p27 in both p53 wild-type and p53 mutant MM cells; overcomes drug resistance; adds to the anti-MM activity of dexamethasone; and overcomes the resistance to apoptosis in MM cells conferred by interleukin-6. PS-341 also inhibits the paracrine growth of human MM cells by decreasing their adherence to bone marrow stromal cells (BMSCs) and related nuclear factor kappaB-dependent induction of interleukin-6 secretion in BMSCs, as well as inhibiting proliferation and growth signaling of residual adherent MM cells. These data, therefore, demonstrate that PS-341 both acts directly on MM cells and alters cellular interactions and cytokine secretion in the BM millieu to inhibit tumor cell growth, induce apoptosis, and overcome drug resistance. Given the acceptable animal and human toxicity profile of PS-341, these studies provide the framework for clinical evaluation of PS-341 to improve outcome for patients with this universally fatal hematological malignancy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , I-kappa B Proteins , Multiple Myeloma/drug therapy , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Bone Marrow Cells/cytology , Bortezomib , Cell Adhesion/physiology , Cell Division/drug effects , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Growth Inhibitors/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , NF-KappaB Inhibitor alpha , Stromal Cells/cytology , Tumor Cells, Cultured
9.
Cancer Res ; 61(9): 3535-40, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11325813

ABSTRACT

Inducible activation of nuclear factor-kappaB (NF-kappaB) inhibits the apoptotic response to chemotherapy and irradiation. Activation of NF-kappaB via phosphorylation of an inhibitor protein IkappaB leads to degradation of IkappaB through the ubiquitin-proteasome pathway. We hypothesized that inactivation of proteasome function will inhibit inducible NF-kappaB activation, thereby increasing levels of apoptosis in response to chemotherapy and enhancing antitumor effects. To assess the effects of proteasome inhibition on chemotherapy response, human colorectal cancer cells were pretreated with the dipeptide boronic acid analogue PS-341 (1 microM) prior to exposure to SN-38, the active metabolite of the topoisomerase I inhibitor, CPT-11. Inducible activation of NF-kappaB and growth response were evaluated in vitro and in vivo. Effects on p53, p21, p27 and apoptosis were determined. Pretreatment with PS-341 inhibited activation of NF-kappaB induced by SN-38 and resulted in a significantly higher level of growth inhibition (64-75%) compared with treatment with PS-341 alone (20-30%) or SN-38 alone (24-47%; P < 0.002). Combination therapy resulted in a 94% decrease in tumor size compared with the control group and significantly improved tumoricidal response to treatment compared with all treatment groups (P = 0.02). The level of apoptosis was 80-90% in the treatment group that received combination treatment compared with treatment with single agent alone (10%). Proteasome inhibition blocks chemotherapy-induced NF-kappaB activation, leading to a dramatic augmentation of chemosensitivity and enhanced apoptosis. Combining proteasome inhibition with chemotherapy has significant potential to overcome the high incidence of chemotherapy resistance. Clinical studies are currently in development to evaluate the role of proteasome inhibition as an important adjuvant to systemic chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Enzyme Inhibitors/pharmacology , NF-kappa B/antagonists & inhibitors , Pyrazines/pharmacology , Animals , Apoptosis/drug effects , Boronic Acids/administration & dosage , Bortezomib , Camptothecin/administration & dosage , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Cysteine Endopeptidases , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/administration & dosage , Female , Humans , Irinotecan , Mice , Mice, Nude , Multienzyme Complexes/antagonists & inhibitors , Proteasome Endopeptidase Complex , Pyrazines/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Stroke ; 31(7): 1686-93, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10884474

ABSTRACT

BACKGROUND AND PURPOSE: Reperfusion brain injury after cerebral ischemia is associated with a developing inflammatory response at the site of infarction. Proteasome inhibitors block nuclear factor-kappaB activation and provide anti-inflammatory effects in several animal models of peripheral inflammation. We tested the novel proteasome inhibitor PS519 in a rat model of transient focal ischemia to establish its pharmacodynamics as a neuroprotection treatment and related effects on leukocyte infiltration. METHODS: Rats were subjected to 2 hours of focal cerebral ischemia by means of the filament method of middle cerebral artery occlusion (MCAo). After either 22 or 70 hours of reperfusion, infarct size was measured and neurological function, electroencephalographic (EEG) activity, and/or neutrophil and macrophage infiltration was quantified. PS519 was administered in a single intravenous bolus at 2 hours after MCAo. In addition, the therapeutic window for PS519 was estimated by delaying treatment for 4 or 6 hours after MCAo. RESULTS: Dose-response analysis of infarct volume at 24 hours revealed that PS519 neuroprotection approached 60%, and clinical evaluations showed significant improvements in neurological function and EEG activity. Neutrophil infiltration at 24 hours was also significantly decreased in cortical and striatal infarcted tissue of PS519-treated rats. Delaying the PS519 treatment up to 4 hours continued to result in significant neuroprotection. In the 72-hour injury model, infarction was reduced 40% by PS519, and significant improvements in neurological function and EEG recovery were again measured. Considerable reductions in both neutrophil and macrophage infiltration were evident. CONCLUSIONS: PS519 mitigates infarction and improves neurological recovery in brain-injured rats, an effect in part caused by a reduction in the leukocyte inflammatory response.


Subject(s)
Acetylcysteine/analogs & derivatives , Cysteine Endopeptidases/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Ischemic Attack, Transient/drug therapy , Macrophages/immunology , Multienzyme Complexes/metabolism , Neutrophils/immunology , Acetylcysteine/pharmacology , Animals , Cell Movement/immunology , Corpus Striatum/blood supply , Corpus Striatum/physiology , Disease Models, Animal , Electroencephalography , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/metabolism , Ischemic Attack, Transient/immunology , Ischemic Attack, Transient/metabolism , Macrophages/cytology , Male , Neuroprotective Agents/pharmacology , Neutrophils/cytology , Proteasome Endopeptidase Complex , Rats , Rats, Sprague-Dawley , Recovery of Function
11.
Invest New Drugs ; 18(2): 109-21, 2000 May.
Article in English | MEDLINE | ID: mdl-10857991

ABSTRACT

The ubiquitin proteasome pathway is a highly conserved intracellular pathway for the degradation of proteins. Many of the short-lived regulatory proteins which govern cell division, growth, activation, signaling and transcription are substrates that are temporally degraded by the proteasome. In recent years, new and selective inhibitors of the proteasome have been employed in cell culture systems to examine the anti-tumor potential of these agents. This review covers the chemistry of selected proteasome inhibitors, possible mechanisms of action in cell culture and the in vivo examination of proteasome inhibitors in murine and human xenograft tumor models in mice. One inhibitor, PS-341, has recently entered Phase I clinical trials in cancer patients with advanced disease to further test the potential of this approach.


Subject(s)
Antineoplastic Agents/pharmacology , Cysteine Endopeptidases/drug effects , Multienzyme Complexes/drug effects , Neoplasms/drug therapy , Animals , Humans , Mice , Neoplasms/pathology , Neoplasms, Experimental/drug therapy , Proteasome Endopeptidase Complex
12.
Clin Chem ; 46(5): 673-83, 2000 May.
Article in English | MEDLINE | ID: mdl-10794750

ABSTRACT

BACKGROUND: PS-341, a selective inhibitor of the proteasome, currently is under evaluation as an anticancer agent in multiple phase I clinical trials. In animal-model studies, PS-341 was rapidly removed from the vascular compartment and distributed widely, quickly approaching the limits of detection. An accurate pharmacodynamic assay has been developed as an alternative or complement to pharmacokinetic measurements. METHODS: Fluorogenic kinetic assays for both the chymotryptic and tryptic activities of the proteasome have been optimized for both whole blood and blood cells. Using the ratio of these activities and the catalytic mechanism of the proteasome, we developed a novel method of calculating percentage of inhibition, using two structurally unrelated inhibitors (PS-341 and lactacystin). RESULTS: This ratio method was demonstrated to be sensitive (detection limit of 13% inhibition with 10 microgram of cell lysate), specific to the proteasome (PS-341 provides >98% inhibition), accurate (112% analyte recovery), and precise (0% +/- 5% inhibition at 0 nmol/L PS-341 and 74.5% +/- 1.7% inhibition at 200 nmol/L PS-341). Using these assays, we found that both erythrocytes and leukocytes contain proteasome at 3 micromol/L. Pharmacodynamic results for PS-341 obtained from the whole-blood ratio method were comparable to those using leukocytes determined by another method. CONCLUSIONS: The described assay provides a reliable method for studying the pharmacodynamics of proteasome inhibitors and is now in use in concurrent phase I clinical trials with PS-341.


Subject(s)
Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Bortezomib , Cysteine Endopeptidases/blood , Humans , In Vitro Techniques , Kinetics , Male , Multienzyme Complexes/blood , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex , Pyrazines/pharmacology , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Fluorescence
13.
J Autoimmun ; 14(3): 205-11, 2000 May.
Article in English | MEDLINE | ID: mdl-10756082

ABSTRACT

PLP139-151-induced relapsing experimental autoimmune encephalomyelitis (R-EAE) in SJL mice is a Th1-mediated autoimmune demyelinating disease model for multiple sclerosis (MS) in which the primary disease relapse is mediated by T cells specific for the endogenous PLP178-191 epitope. This complex inflammatory process requires the co-ordinated expression of a wide variety of immune-related genes active at a variety of stages of the autoimmune process which are regulated, in part, by the transcription factor nuclear factor (NF)-kappaB which is activated via the ubiquitin-proteasome pathway. We asked if in vivo administration of a selective inhibitor of the ubiquitin-proteasome pathway, PS-519, which downregulates activation of NF-kappaB, could downregulate ongoing R-EAE. Administration of PS-519 during the remission phase, following acute clinical disease was effective in significantly reducing the incidence of clinical relapses, CNS histopathology, and T cell responses to both the initiating and relapse-associated PLP epitopes. The inhibition of clinical disease was dependent upon continuous administration of PS-519 in that recovery of T cell function and onset of disease relapses developed within 10-14 days of drug withdrawal. The data suggest that targeting the ubiquitin proteasome pathway, in particular NF-kappaB, may offer a novel and efficacious approach for the treatment of progressive autoimmune diseases, including MS.


Subject(s)
Cysteine Endopeptidases/immunology , Cysteine Proteinase Inhibitors/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multienzyme Complexes/immunology , Animals , Cysteine Proteinase Inhibitors/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Epitopes, T-Lymphocyte/immunology , Female , Mice , Myelin Proteolipid Protein/immunology , Peptide Fragments/immunology , Proteasome Endopeptidase Complex , Recurrence , Spinal Cord/immunology , T-Lymphocytes/immunology
15.
J Allergy Clin Immunol ; 104(2 Pt 1): 294-300, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10452747

ABSTRACT

BACKGROUND: Nuclear factor-kappaB (NF-kappaB) is a critical transcription factor required for the regulation of many genes involved in inflammatory responses to noxious stimuli. On activation, NF-kappaB induces the transcription of numerous proinflammatory cytokines, enzymes, and cellular adhesion molecules. Blockade of the proteasome with selective inhibitors attenuates the effects of NF-kappaB, leading to suppression of the inflammatory response. OBJECTIVE: We sought to determine whether proteasome inhibitors would be active in a model of asthma. METHODS: The mouse delayed-type hypersensitivity model was used to screen a panel of compounds for in vivo activity. The proteasome inhibitor, PS-519, was shown to be the most active in this model and was selected for further development. Allergen-induced pulmonary eosinophilia in Brown Norway rats was used subsequently to determine anti-inflammatory activity in an animal model. RESULTS: Direct administration of PS-519 into the lungs significantly reduced leukocyte numbers, particularly the selective increase in eosinophils. Because steroids are the mainstay anti-inflammatory therapy in asthma, and data is available to suggest their possible interaction to suppress the activation of NF-kappaB, rats were also treated by inhalation with combinations of a steroid and the proteasome inhibitor. In both the delayed-type hypersensitivity and the animal eosinophil model, low doses of proteasome inhibitors were shown to be effective when given with low doses of steroids. CONCLUSION: Taken together, the present data suggest that proteasome inhibition may represent a novel strategy for the treatment of inflammatory lung diseases such as asthma.


Subject(s)
Cysteine Endopeptidases/drug effects , Cysteine Proteinase Inhibitors/physiology , Multienzyme Complexes/drug effects , Animals , Asthma/physiopathology , Asthma/prevention & control , Dexamethasone/adverse effects , Hypersensitivity, Delayed/chemically induced , Lactones/chemistry , Lactones/pharmacology , Male , Mice , Mice, Inbred BALB C , Proteasome Endopeptidase Complex , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/physiopathology , Pulmonary Eosinophilia/prevention & control , Rats , Rats, Inbred BN , Structure-Activity Relationship
16.
Am J Physiol ; 277(2): E332-41, 1999 08.
Article in English | MEDLINE | ID: mdl-10444430

ABSTRACT

A new model of cachexia is described in which muscle protein metabolism related to the ubiquitin-proteasome pathway was investigated. Cloning of the colon-26 tumor produced a cell line, termed R-1, which induced cytokine (noninterleukin-1beta, interleukin-6 and tumor necrosis factor-alpha)-independent cachexia. Implantation of R-1 cells in mice elicited significant (20-30%) weight loss and decreased blood glucose by 70%, and adipose tissue levels declined by 95% and muscle weights decreased by 20-25%. Food intake was unaffected. The decrease in muscle weight reflected a decline in insoluble, but not soluble, muscle protein that was associated with a significant increase in net protein degradation. The rate of ubiquitin conjugation of proteins was significantly elevated in muscles of cachectic mice. Furthermore, the proteasome inhibitor lactacystin blocked the increase in protein breakdown but had no significant effect on proteolysis. Several markers of the ubiquitin-proteasome pathway, E2(14k) mRNA and E2(14k) protein and ubiquitin-protein conjugates, were not elevated. Future investigations with this new model should gain further insights into the mechanisms of cachexia and provide a background to evaluate novel and more efficacious therapies.


Subject(s)
Cachexia/etiology , Cachexia/metabolism , Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Neoplasms, Experimental/complications , Ubiquitins/metabolism , Animals , Cachexia/drug therapy , Dexamethasone/therapeutic use , Disease Models, Animal , Glucocorticoids/therapeutic use , Indomethacin/therapeutic use , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Muscle Proteins/metabolism , Muscles/metabolism , Proteasome Endopeptidase Complex
17.
Cancer Res ; 59(11): 2615-22, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10363983

ABSTRACT

The ubiquitin-proteasome pathway plays a critical role in the regulated degradation of proteins involved in cell cycle control and tumor growth. Dysregulating the degradation of such proteins should have profound effects on tumor growth and cause cells to undergo apoptosis. To test this hypothesis, we developed a novel series of proteasome inhibitors, exemplified by PS-341, which we describe here. As determined by the National Cancer Institute in vitro screen, PS-341 has substantial cytotoxicity against a broad range of human tumor cells, including prostate cancer cell lines. The PC-3 prostate cell line was, therefore, chosen to further examine the antitumor activity of PS-341. In vitro, PS-341 elicits proteasome inhibition, leading to an increase in the intracellular levels of specific proteins, including the cyclin-dependent kinase inhibitor, p21. Moreover, exposure of such cells to PS-341 caused them to accumulate in the G2-M phase of the cell cycle and subsequently undergo apoptosis, as indicated by nuclear condensation and poly(ADP-ribose) polymerase cleavage. Following weekly i.v. treatment of PS-341 to mice bearing the PC-3 tumor, a significant decrease (60%) in tumor burden was observed in vivo. Direct injection of PS-341 into the tumor also caused a substantial (70%) decrease in tumor volume with 40% of the drug-treated mice having no detectable tumors at the end of the study. Studies also revealed that i.v. administration of PS-341 resulted in a rapid and widespread distribution of PS-341, with highest levels identified in the liver and gastrointestinal tract and lowest levels in the skin and muscle. Modest levels were found in the prostate, whereas there was no apparent penetration of the central nervous system. An assay to follow the biological activity of the PS-341 was established and used to determine temporal drug activity as well as its ability to penetrate tissues. As such, PS-341 was shown to penetrate PC-3 tumors and inhibit intracellular proteasome activity 1.0 h after i.v. dosing. These data illustrate that PS-341 not only reaches its biological target but has a direct effect on its biochemical target, the proteasome. Importantly, the data show that inhibition of this target site by PS-341 results in reduced tumor growth in murine tumor models. Together, the results highlight that the proteasome is a novel biochemical target and that inhibitors such as PS-341 represent a unique class of antitumor agents. PS-341 is currently under clinical evaluation for advanced cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Protease Inhibitors/pharmacology , Algorithms , Animals , Antineoplastic Agents/pharmacokinetics , Boronic Acids/pharmacokinetics , Computer Simulation , Drug Screening Assays, Antitumor , Humans , Male , Mice , Mice, Nude , Models, Chemical , Protease Inhibitors/pharmacokinetics , Tumor Cells, Cultured/drug effects
18.
Methods Enzymol ; 300: 345-63, 1999.
Article in English | MEDLINE | ID: mdl-9919536

ABSTRACT

It is becoming increasingly apparent that NF-kappa B plays a critical role in regulating the inflammatory response. Data obtained from studies in our laboratories demonstrate that the proteasome plays an important role in the inflammatory cascade by regulating the activation of NF-kappa B. Indeed, the availability of selective and orally active proteasome inhibitors should prove useful in delineating the roles of the proteasome and NF-kappa B in other pathophysiological conditions such as cancer and heart disease.


Subject(s)
NF-kappa B/metabolism , Peptide Hydrolases/drug effects , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Arthritis/drug therapy , Boronic Acids/pharmacology , Cell Adhesion Molecules/biosynthesis , Cytokines/biosynthesis , Dipeptides/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , HeLa Cells , Humans , Hypersensitivity, Delayed/drug therapy , Jurkat Cells , Leupeptins/pharmacology , Rats , Rats, Inbred Lew , T-Lymphocytes/drug effects
19.
Proc Natl Acad Sci U S A ; 95(26): 15671-6, 1998 Dec 22.
Article in English | MEDLINE | ID: mdl-9861028

ABSTRACT

The transcription factor NF-kappaB activates a number of genes whose protein products are proinflammatory. In quiescent cells, NF-kappaB exists in a latent form and is activated via a signal-dependent proteolytic mechanism in which the inhibitory protein IkappaB is degraded by the ubiquitin-proteasome pathway. Consequently, inhibition of the proteasome suppresses activation of NF-kappaB. This suppression should therefore decrease transcription of many genes encoding proinflammatory proteins and should ultimately have an anti-inflammatory effect. To this end, a series of peptide boronic acid inhibitors of the proteasome, exemplified herein by PS-341, were developed. The proteasome is the large multimeric protease that catalyzes the final proteolytic step of the ubiquitin-proteasome pathway. PS-341, a potent, competitive inhibitor of the proteasome, readily entered cells and inhibited the activation of NF-kappaB and the subsequent transcription of genes that are regulated by NF-kappaB. Significantly, PS-341 displayed similar effects in vivo. Oral administration of PS-341 had anti-inflammatory effects in a model of Streptococcal cell wall-induced polyarthritis and liver inflammation in rats. The attenuation of inflammation in this model was associated with an inhibition of IkappaBalpha degradation and NF-kappaB-dependent gene expression. These experiments clearly demonstrate that the ubiquitin-proteasome pathway and NF-kappaB play important roles in regulating chronic inflammation and that, as predicted, proteasome inhibition has an anti-inflammatory effect.


Subject(s)
Arthritis, Experimental/physiopathology , Cell Adhesion Molecules/genetics , Cysteine Endopeptidases/metabolism , Cytokines/genetics , Endothelium, Vascular/physiology , Multienzyme Complexes/metabolism , NF-kappa B/metabolism , Streptococcus/immunology , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cell Adhesion Molecules/biosynthesis , Cell Wall/immunology , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Cytokines/biosynthesis , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Gene Expression Regulation , Humans , Joints/pathology , Joints/physiopathology , Nitric Oxide/metabolism , Proteasome Endopeptidase Complex , Rats , Rats, Inbred Lew , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitins/metabolism , Umbilical Veins
20.
J Neurooncol ; 37(3): 199-215, 1998 May.
Article in English | MEDLINE | ID: mdl-9524078

ABSTRACT

The effects of an intravenous (i.v.) injection of the bradykinin analog RMP-7 (100 ng/kg) were assessed in normal dogs and dogs with focal, radiation-induced brain lesions. A dose of 20 Gy was delivered to a point 0.75 cm from a removable interstitial 125I source; parameters relating to blood flow and permeability were quantified using computed tomography 2-8 weeks after irradiation. Blood flow-related endpoints included regional cerebral blood flow (rCBF), mean transit time of blood and vascular volume, while endpoints related to permeability included blood-to-brain transfer constant (Ki), brain-to-blood transfer constant and plasma volume. In unirradiated brain, an i.v. bolus of RMP-7 administered through the left cephalic vein induced a rapid and transient hypotension and a statistically significant increase in vascular volume; no alterations in any parameter related to permeability were observed. After irradiation, changes in rCBF after RMP-7 depended upon time after exposure, effects presumably due to changing morphology in the irradiated tissues. In the radiation lesions, significant increases in Ki were observed 5 minutes after injection of RMP-7, but those increases were not related to time after irradiation or alteration in blood flow-related parameters. Our results showed that RMP-7 selectively increased permeability in already damaged vasculature without affecting the extent or volume of radiation-induced vasogenic edema. These data suggest that RMP-7 may provide an effective means to enhance the delivery of compounds to an already compromised brain while not exacerbating the potential adverse effects of pre-existing vasogenic edema.


Subject(s)
Bradykinin/analogs & derivatives , Brain/drug effects , Brain/radiation effects , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/radiation effects , Animals , Blood Pressure/drug effects , Blood Pressure/radiation effects , Bradykinin/pharmacology , Brain/diagnostic imaging , Brain Edema/diagnostic imaging , Brain Edema/etiology , Dogs , Male , Radiation Injuries, Experimental , Reference Values , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...