Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Clin Pharmacokinet ; 40(2): 105-23, 2001.
Article in English | MEDLINE | ID: mdl-11286321

ABSTRACT

Labradimil (Cereport; also formerly referred to as RMP-7) is a 9-amino-acid peptide designed for selectivity for the bradykinin B2 receptor and a longer plasma half-life than bradykinin. It has been developed to increase the permeability of the blood-brain barrier (BBB) and is the first compound with selective bradykinin B2 receptor agonist properties to progress from concept design through to tests of efficacy in patients. In vitro studies demonstrate that labradimil has a longer half-life than bradykinin and selectively binds to bradykinin B2 receptors, initiating typical bradykinin-like second messenger systems, including increases in intracellular calcium and phosphatidylinositol turnover. Initial proof of principle studies using electron microscopy demonstrated that intravenous labradimil increases the permeability of the BBB by disengaging the tight junctions of the endothelial cells that comprise the BBB. Autoradiographic studies in rat models further demonstrated that labradimil increases the permeability of the BBB in gliomas. Intravenous or intra-arterial labradimil increases the uptake of many different radiolabelled tracers and chemotherapeutic agents into the tumour in a dose-related fashion. These effects are selective for the tumour and for the brain surrounding the tumour, and are particularly robust in tumour areas that are normally relatively impermeable. The increased chemotherapeutic concentrations are maintained for at least 90 minutes, well beyond the transient effects on the BBB. The increase in permeability with labradimil occurs rapidly but is transient, in that restoration of the BBB occurs very rapidly (2 to 5 minutes) following cessation of infusion. Even with continuous infusion of labradimil, spontaneous restoration of the barrier begins to occur within 10 to 20 minutes. Collectively, these data demonstrate that the B2 receptor system that modulates permeability of the BBB is highly sensitive and autoregulated and that careful attention to the timing of labradimil and the chemotherapeutic agent is important to achieve maximal effects. Survival studies in rodent models of both gliomas and metastatic tumours in the brain demonstrate that the enhanced uptake observed with the combination of labradimil and water-soluble chemotherapeutics enhances survival to a greater extent than achieved with chemotherapy alone. Finally, preliminary clinical trials in patients with gliomas provide confirmatory evidence that labradimil permeabilises the blood-brain tumour barrier and might, therefore, be used to increase delivery of agents such as carboplatin to tumours without the toxicity typically associated with dose escalation.


Subject(s)
Blood-Brain Barrier/drug effects , Bradykinin/agonists , Bradykinin/pharmacology , Animals , Bradykinin/analogs & derivatives , Bradykinin/pharmacokinetics , Capillary Permeability/drug effects , Central Nervous System Neoplasms/drug therapy , Dose-Response Relationship, Drug , Glioma/drug therapy , Half-Life , Humans , Rats , Receptor, Bradykinin B2 , Receptors, Bradykinin/drug effects
2.
J Pharmacol Exp Ther ; 296(2): 623-31, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160651

ABSTRACT

Delivery of chemotherapeutic agents to solid peripheral tumors is compromised because the impaired microvasculature within and surrounding tumors limits diffusion and convection of agents from the vasculature to the tumor. Using a variety of rat tumor models, we show that intravenous administration of a vasoactive bradykinin B2 receptor agonist (Cereport, or labradimil; formerly RMP-7) enhances by nearly 3 times the delivery of the chemotherapeutic agent carboplatin, as well as the larger 70-kDa marker dextran, into ectopic and orthotopic solid tumors. This effect was selective for tumor tissue, with little or no increase seen in nontumor tissues and organs. Additionally, the increased carboplatin levels observed in tumors persisted for at least 90 min (the longest time point measured). In contrast to the consistent effects with hydrophilic compounds, delivery of the lipophilic, high protein-binding chemotherapeutics paclitaxel and 1,3-bis[2-chloroethyl]-1-nitrourea (BNCU) was not enhanced. Administration of Cereport with either carboplatin or another hydrophilic chemotherapeutic agent, doxorubicin, significantly increased efficacy of both agents, manifested by suppression of tumor growth and prolonged survival in tumor-bearing rats. These data demonstrate that delivery of chemotherapeutics to tumors can be pharmacologically increased (by stimulating bradykinin B2 receptors) without increasing the systemic exposure, or therefore, the toxic liability associated with higher chemotherapeutic doses.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Bradykinin/physiology , Bradykinin/therapeutic use , Neoplasms, Experimental/blood supply , Receptors, Bradykinin/agonists , Animals , Bradykinin/administration & dosage , Bradykinin/analogs & derivatives , Cell Survival/drug effects , Female , Immunohistochemistry , Infusions, Intravenous , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Lung Neoplasms/blood supply , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Rats, Wistar , Receptor, Bradykinin B2 , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Survival Analysis , Tumor Cells, Cultured
3.
J Pharmacol Exp Ther ; 296(2): 632-41, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160652

ABSTRACT

Intravenous infusions of the bradykinin agonist Cereport (labradimil, formerly RMP-7) enhance delivery of concomitantly administered hydrophilic chemotherapeutic agents to solid tumors. The enhanced delivery produces greater in vivo efficacy of chemotherapeutic agents, manifested as suppressed tumor growth and increased survival in tumor-bearing rats. Here we elucidate the mechanisms of action involved with this unique phenomenon, at both the physical and biochemical levels. At the physical level we demonstrate that Cereport modifies the tumor vasculature in several important ways, including transient 1) reductions in interstitial fluid pressure within the tumor, 2) increases in pore size of the vasculature, and 3) increases in total vascular surface area. All three of these changes modify tumor-specific characteristics of the vasculature known to impede drug delivery to the tumor interstitium. Biochemically, we demonstrate that the activation of both of bradykinin's major signaling pathways, the nitric oxide and phospholipase A2/prostaglandin E2 are necessary events. Although pharmacologically blocking either pathway greatly reduced the effects of Cereport, stimulation of either pathway alone did not enhance delivery. However, simultaneous stimulation of both pathways (without exogenous bradykinin B2 receptor stimulation) produced a nearly 2-fold increase in delivery of carboplatin to the tumor. Thus, stimulation of endogenous bradykinin B2 receptors induces at least two parallel biochemical cascades that act synergistically to uniquely modify the tumor vasculature in ways that increase delivery and efficacy of chemotherapeutic agents.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Bradykinin/physiology , Bradykinin/therapeutic use , Neoplasms, Experimental/blood supply , Nitric Oxide/metabolism , Phospholipases A/metabolism , Prostaglandins/physiology , Regional Blood Flow/physiology , Signal Transduction/drug effects , Algorithms , Animals , Autoradiography , Biotransformation/drug effects , Blood Pressure/drug effects , Blood Vessels/drug effects , Bradykinin/analogs & derivatives , Extracellular Space/drug effects , Immunohistochemistry , Male , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Phospholipases A2 , Porosity/drug effects , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
4.
Exp Neurol ; 161(1): 234-44, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10683290

ABSTRACT

Cereport (RMP-7) enhances delivery of chemotherapeutics into brain tumors by increasing the permeability of the glioma vasculature (i.e. , the blood-brain tumor barrier; BBTB). Its effect on brain tumors has consistently been more robust than that on normal brain. The present experiments tested the hypothesis that the ability of Cereport to increase the permeability of infiltrating glioma colonies increases as the glioma colonies develop, in situ. In an initial preliminary experiment, the significant and selective effects of Cereport in tumor tissue and brain surrounding tumor were verified using [(14)C]carboplatin as a marker, 8 days after implantation of 50,000 RG2 cells. A second preliminary experiment established that the number of tumor cells initially seeded influences the growth rate of the tumor mass. Tumors seeded with 50,000 cells were larger than those seeded with 25,000 cells 3, 5, and 8 days after implantation. Next, the hypothesis that the extent of tumor growth increases Cereport's effects on the BBTB was tested by measuring the concentration of radiolabeled carboplatin in the tumor when 50,000 cells were implanted 3, 8, or 13 days prior to the experiment. While a reliable, approximately twofold increase in carboplatin concentration was seen in the 8- and 13-day-old tumors, no significant effect of Cereport was observed in the tumors that developed only 3 days, in situ. Finally, another test of the hypothesis was made by comparing Cereport's effects on 8-day-old tumors initially seeded with either 50,000 or 25,000 cells (the latter producing a smaller, more slowly developing, tumor mass). Again, significantly higher carboplatin concentrations were seen with Cereport in the 50,000 cell tumors (greater than two-fold increase), compared to the smaller, more slowly developing, 25,000 cell tumors (<30% increase). The tumor and its vasculature were characterized in additional rats implanted with RG2 cells using CD-31, laminin, and bradykinin B(2) receptor immunocytochemistry. Intense B(2) receptor staining was observed on cells within the parenchyma of normal brain and tumor but not on the vasculature of tumor or brain. An extensive network of CD-31 and laminin staining was seen within and around the tumors in all groups, indicating relatively rapid and robust changes in vascularity in response to the gliomas. However, no consistent difference in vascularity between groups was observed to account for the uptake differences seen with Cereport. Collectively, these data offer initial preclinical empirical support for the hypothesis that Cereport's effects on tumor permeability increase as the tumor grows, which we further hypothesize is likely related to features of vascular development within the tumor independent of numbers or general morphology of vessels. If a similar phenomenon is shown to occur with infiltrating colonies from spontaneously forming gliomas in humans or from newly emerging metastases in brain, these data could impact the design and conduct of future trials using approaches intended to enhance delivery of chemotherapeutics through increased permeability of the tumor vascular barrier.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Blood-Brain Barrier/drug effects , Bradykinin/analogs & derivatives , Brain Neoplasms/metabolism , Carboplatin/pharmacokinetics , Glioma/metabolism , Animals , Bradykinin/pharmacology , Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Carbon Radioisotopes/pharmacokinetics , Cell Count , Drug Delivery Systems , Glioma/blood supply , Glioma/drug therapy , Male , Neoplasm Transplantation , Neovascularization, Pathologic/physiopathology , Rats , Rats, Inbred F344
5.
Pharm Res ; 17(10): 1212-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11145226

ABSTRACT

PURPOSE: The following experiments determined whether intravenous infusions of Cereport enhance delivery of chemotherapeutics and prolong survival in rats with metastatic tumors in the brain. METHODS: Autoradiography and scintillation were used to examine uptake of the lipophilic (paclitaxel and carmustine) and the hydrophilic (carboplatin) chemotherapeutic agents, as well as the large hydrophilic marker, 70 kDa dextran. Cereport was also tested in combination with the chemotherapeutic drugs carboplatin, vinorelbine, gemcitabine and carmustine to determine if Cereport could enhance the survival benefit beyond that provided by chemotherapy alone. RESULTS: Cereport enhanced the uptake of carboplatin and dextran, but not paclitaxel or carmustine. The pattern of Cereport's uptake effect with carboplatin revealed that Cereport selectively increased the proportion of highly permeable regions. Survival was significantly enhanced when Cereport was combined with either carboplatin, vinorelbine, or gemcitabine, but not carmustine, compared to each chemotherapeutic agent alone. CONCLUSIONS: These data provide the first evidence that Cereport, or any receptor-mediated approach intended to enhance the permeability of the blood-brain tumor barrier, can increase the delivery hydrophilic drugs to metastatic tumors in the brain, increasing survival in tumor-bearing rats.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bradykinin/pharmacology , Brain Neoplasms/metabolism , Vinblastine/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Autoradiography , Blood-Brain Barrier/drug effects , Bradykinin/analogs & derivatives , Brain/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Carboplatin/pharmacokinetics , Carboplatin/pharmacology , Carmustine/pharmacokinetics , Carmustine/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Male , Rats , Rats, Inbred F344 , Vinblastine/pharmacokinetics , Vinblastine/pharmacology , Vinorelbine , Gemcitabine
6.
Neuro Oncol ; 1(4): 268-74, 1999 10.
Article in English | MEDLINE | ID: mdl-11550318

ABSTRACT

Accumulating evidence suggests that dexamethasone might decrease permeability of the blood-brain tumor barrier, further limiting the delivery of agents into brain tumors. The bradykinin B2 receptor agonist, Cereport (RMP-7), selectively increases permeability of the vasculature supplying brain tumors in both animal models and humans. The present study was conducted to characterize the effects of dexamethasone on the blood-brain tumor barrier and its potential interaction with Cereport's ability to enhance penetration of radiolabeled carboplatin. Dexamethasone (1.5 mg/kg/day, twice a day) was given to RG2 glioma-bearing rats via oral gavage for 3 consecutive days. After treatment, animals received a 15-min intracarotid infusion of Cereport (4.5 micrograms/kg) and a bolus of [14C]carboplatin. The levels of [14C]carboplatin (nCi/g) in the tumor and nontumor regions were determined at 1, 14, or 24 h after the last dose of dexamethasone. Dexamethasone, alone, significantly decreased the levels of radiolabeled carboplatin permeating the tumor (19%), although there were no significant differences between any of the time points examined. Cereport administration significantly increased levels of carboplatin in the tumor, independent of whether or not dexamethasone was given (46% with and 49% without). Although the relative effects of Cereport on tumor carboplatin levels were not affected by dexamethasone, the absolute levels achieved with Cereport were modestly reduced (44 nCi/g versus 55.5 nCi/g of [14C]carboplatin, with and without dexamethasone, respectively). Thus, while the data support the use of Cereport as adjunctive therapy in the treatment of glioma patients, they also warn that the use of dexamethasone may reduce delivery of chemotherapeutic agents to brain tumors, even when special pharmacologic measures are employed to enhance delivery.


Subject(s)
Blood-Brain Barrier/drug effects , Bradykinin/pharmacology , Brain Neoplasms/drug therapy , Carboplatin/pharmacokinetics , Dexamethasone/pharmacology , Glioma/drug therapy , Receptors, Bradykinin/agonists , Animals , Body Weight/drug effects , Bradykinin/analogs & derivatives , Brain Neoplasms/metabolism , Depression, Chemical , Drug Administration Schedule , Glioma/metabolism , Male , Neoplasm Transplantation , Organ Size/drug effects , Permeability/drug effects , Rats , Rats, Inbred F344 , Receptor, Bradykinin B2 , Tumor Cells, Cultured
7.
Brain Res ; 790(1-2): 1-13, 1998 Apr 20.
Article in English | MEDLINE | ID: mdl-9593800

ABSTRACT

Rats were subjected to transient global ischemia (four vessel occlusion) and time-related changes in the selectively vulnerable hippocampal field CA1 were characterized. The assessment included ex vivo field responses to afferent stimulation, silver staining, calpain-induced spectrin breakdown, chromatolysis, and cell death, beginning at 6 h post-ischemia and continuing until total disintegration of the pyramidal cells occurred several days later. The earliest change observed was a modest increase in the slope and amplitude of field CA1 potentials (at 6 h). The hyperresponsiveness was most apparent at higher stimulation currents and persisted unchanged at 16 h post-ischemia. Three effects became detectable within 24 h, post-ischemia: (a) an increase in concentrations of calpain-mediated, spectrin breakdown products; (b) enhanced silver staining in the deep pyramidal neurons of the field CA1 with lesser, though still apparent, staining of stratum radiatum, and (c) a decrease in amplitude and slope of field CA1 responses to afferent stimulation. Both the concentration of spectrin breakdown products and the intensity of silver staining progressively increased to a maximum at four days post ischemia, while the amplitude and slope of the field responses dropped to a very low level between 24 and 48 h. Disturbances of Nissl staining were finally evident at 48 h, with nearly complete disappearance of staining at five days post-ischemia. This study provides the first demonstration of a close and early temporal relationship between calpain proteolysis, subcellular damage to the pyramidal cells and their loss of function following global ischemia, prior to their eventual death.


Subject(s)
Brain Chemistry/physiology , Hippocampus/physiopathology , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/physiopathology , Animals , Calpain/metabolism , Cell Death/physiology , Cytoskeleton/physiology , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Hippocampus/enzymology , Neurons/cytology , Neurons/enzymology , Rats , Rats, Sprague-Dawley , Silver Staining , Spectrin/analysis , Time Factors
8.
Exp Neurol ; 142(1): 14-28, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8912895

ABSTRACT

Previous studies have shown that the bradykinin agonist, RMP-7, can safely permeabilize the blood brain barrier (BBB) by activation of constitutive B2 receptors on endothelial cells. The paper describes a series of studies using quantitative autoradiography and intracarotid infusions of RMP-7 to further elucidate the effect on BBB permeability. Because earlier studies also demonstrated that even greater effects of RMP-7 were observed in the BBB associated with brain tumors, animal models were employed so that comparisons could be made between the effects of RMP-7 within tumor, brain tissue proximal to tumor, and brain tissue distal from tumor. In the first study, the effect of RMP-7 on enhancing the BBB permeation of three compounds of different physical characteristics was directly compared ([14C]carboplatin, small, hydrophilic; [14C]dextran, large, hydrophilic; [14C]BCNU, small, lipophilic). RMP-7 increased permeability of the vascular barriers to both hydrophilic compounds, carboplatin and dextran. While the effects of RMP-7 were observed on nontumor BBB, the greatest and most consistent effects were observed on the blood brain tumor barrier. This was true for both carboplatin and dextran, with progressively less effect seen as the distance from tumor boundary increased. This topographic effect was more pronounced with the larger molecular weight compound, dextran. No effect of RMP-7 was seen in permeabilizing the BBB or the blood brain tumor barrier for the lipophilic drug, BCNU. In a second study, the generality of RMP-7's effects was established by demonstrating similar increases in permeability to carboplatin in both inbred (Fischer 344) and outbred (Wistar) rat strains, implanted with varying tumor cell lines. Finally, several additional studies were performed to gain greater insight into the dynamics involved with eventual restoration of the BBB following RMP-7 administration. In one series, it was demonstrated that the BBB begins to close nearly immediately upon withdrawal of RMP-7, with complete restoration occurring within minutes. In another series, tachyphylaxis or desensitization resulting from continuous RMP-7 infusion was studied. These studies demonstrated that 60 min of continuous RMP-7 infusion resulted in complete, spontaneous restoration of the barrier to both carboplatin and dextran. Moreover, the desensitization appears to be linked to the initial activation of the receptors in a way which suggests that obligatory desensitization may exist as part of a more complete response. These data are discussed as they relate to practical issues to enhance delivery of drugs across the BBB, as well as more fundamental issues involving the function of the BBB and its interaction with the brain.


Subject(s)
Blood-Brain Barrier/drug effects , Bradykinin/analogs & derivatives , Carboplatin/pharmacokinetics , Carcinogens/pharmacokinetics , Carmustine/pharmacokinetics , Dextrans/pharmacokinetics , Plasma Substitutes/pharmacokinetics , Animals , Autoradiography , Biomarkers , Bradykinin/agonists , Bradykinin/pharmacology , Brain Neoplasms , Capillaries/drug effects , Capillaries/metabolism , Carbon Radioisotopes/pharmacokinetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Male , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Rats, Wistar , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
9.
Cancer Res ; 56(17): 3998-4005, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8752170

ABSTRACT

Rats implanted with RG-2 gliomas were administered i.v. RMP-7 and [14C]carboplatin. Changes in the permeability of the blood-brain barrier to carboplatin were determined using quantitative autoradiography. i.v. infusions of RMP-7 induced an increase in the permeability of the vascular barrier within the tumor to carboplatin. Additionally, permeability of brain tissue proximal to, but clearly outside the tumor mass, was also increased. Progressively less uptake of [14C]carboplatin was observed as distance from the tumor border increased. The increases in permeability induced by RMP-7 occurred in a dose-related fashion. No increase in carboplatin level was observed in several nonbrain tissues, including sciatic nerve, retina, heart, lung, liver, kidney, and spleen. Finally, the permeabilizing effects of RMP-7 were shown to occur independent of histaminergic or hypotensive mechanisms. These data provide additional insight into the permeabilizing effects and mechanism of RMP-7 and offer additional support for the therapeutic utility of this novel compound as an adjunctive treatment for human gliomas.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bradykinin/analogs & derivatives , Brain Neoplasms/metabolism , Carboplatin/pharmacokinetics , Glioma/metabolism , Animals , Antineoplastic Agents/toxicity , Autoradiography , Blood-Brain Barrier , Bradykinin/pharmacology , Brain/metabolism , Capillary Permeability , Carbon Radioisotopes , Carboplatin/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Female , Histamine/physiology , Hypotension/physiopathology , Rats , Rats, Wistar
11.
Restor Neurol Neurosci ; 10(1): 49-59, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-21551853

ABSTRACT

While the function and regulation of the low affinity (p75) nerve growth factor (NGF) receptor in the central nervous system (CNS) remains a mystery, one of the more intriguing observations involves its response to injury in the adult rat striatum. Following mechanical injury to the striatum, a re-expression of striatal p75 receptors and mRNA purportedly occurs (apparently mediated by elevations in NGF), thus reversing the natural loss of these phenotypic markers that is known to occur during development. This observation has important implications for understanding both the regulation of NGF neurotrophic activity and the role of the p75 receptor, for it implies that the presence of this receptor may be required for NGF trophic activity in the CNS. In an effort to gain a greater understanding of the function and regulation of the low affinity p75 NGF receptor, we performed a series of experiments to study the injury-induced, re-expression phenomenon in the striatum. In the first experiment, we duplicated the mechanical, cannula-induced injury used in the original study. In a follow-up study, we exacerbated that injury by infusing quinolinic acid directly into the striatum. In a third study, the mechanical injury was complemented with chronic striatal infusions of NGF. In a final study, we examined striatal tissue from rats who had been protected from striatal quinolinic acid neurotoxicity by administration of NGF. In no instance was the re-expression of p75 striatal receptors observed, despite positive controls for (a) effective neural trauma, confirmed by histologic and immunocytochemical methods, (b) effective antibody staining, confirmed by appropriate basal forebrain p75 immunoreactivity, and (c) effective biological activity of exogenous NGF, confirmed by hypertrophy of choline acetyltransferase (ChAT)-positive striatal neurons and protection of ChAT-positive striatal neurons against excitotoxicity. At least two important conclusions can be drawn from these studies: (1) the presence or induction of low affinity p75 receptors is not necessary, while the presence of constitutive high affinity tropomyosin related kinase (trk) NGF receptors seem sufficient for NGF trophic activity in the CNS, and (2) the variables necessary to induce re-expression of p75 striatal receptors in adult rats have not yet been elucidated and are apparently complex.

12.
Brain Res ; 705(1-2): 125-35, 1995 Dec 24.
Article in English | MEDLINE | ID: mdl-8821743

ABSTRACT

The route taken by lanthanum (MW 139) across cerebral endothelium was delineated when the blood-brain barrier was opened by RMP-7, a novel bradykinin agonist. Balb C mice were infused through a jugular vein with LaCl3 with or without RMP-7 (5 micrograms/kg). Ten minutes later, the brains were fixed with aldehydes and processed for electron microscopy. The patency of the junctions between endothelial cells was estimated by counting the number of junctions penetrated by LaCl3. Tracer penetrated the junctions in about 25% of microvessels in vehicle infused, control mice and about 58% in the RMP-7 group, where more junctions per vessel were also penetrated. The LaCl3 then penetrated the basal lamina in about 20% of all microvessels in the RMP-7 group, versus 0.50% in the control group. From the basal lamina, the tracer entered perivascular spaces in about 13% of all microvessels in the RMP-7 group and about 0.07% in the controls. Very few endocytic pits or vesicles in the RMP-7 group were labeled, so LaCl3 did not cross endothelium by transcytosis. The increased number of tight junctions penetrated by tracer and its spread into periendothelial basal lamina and interstitial clefts indicated, therefore, a paracellular route of exudation in the RMP-7 treated animals.


Subject(s)
Blood-Brain Barrier/drug effects , Bradykinin/analogs & derivatives , Lanthanum/pharmacokinetics , Animals , Bradykinin/agonists , Bradykinin/pharmacology , Cerebrovascular Circulation/physiology , Consciousness , Endothelium/metabolism , Endothelium/ultrastructure , Hypotension/chemically induced , Injections, Intravenous , Male , Mice , Mice, Inbred BALB C , Microscopy, Electron , Sodium Chloride/pharmacology , Tight Junctions/metabolism
13.
J Cereb Blood Flow Metab ; 15(6): 969-79, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7593358

ABSTRACT

Changes in neocortex and striatum were characterized over time following focal ischemia to the brain. Rats were subjected to permanent middle cerebral artery occlusion (MCA-O) and sacrificed 1, 3, 6, 12, or 24 h later. The affected tissue was processed for tetrazolium chloride (TTC) and cresyl violet staining, as well as for Western blots to detect calpain-induced spectrin proteolysis. Significant changes in cell size and spectrin breakdown occurred within the first hour of occlusion, with further, dramatic changes in these two early markers continuing over time. Initial evidence of cell loss was noted at 1 h postocclusion in the striatum and at 3 h in the neocortex. However, even in the center of the most affected portion of the neocortex, the majority of cells appeared to be intact through 6 h. By this time, a significant TTC-defined infarct also emerged. These quantitative data indicate that calpain-induced proteolysis occurs very soon after the ischemic insult, is correlated with earliest changes in cell hypotrophy, and precedes or occurs in tandem with evidence of significant cell loss. They also demonstrate that, while some cell loss occurs earlier than previously believed, the majority of cells remains morphologically intact well beyond what is typically thought to be the window of opportunity for intervention. The results thus raise the question of how long after the ischemic event pharmaceutic intervention might be employed to salvage substantial numbers of neurons.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Calpain/physiology , Neurons/metabolism , Neurons/pathology , Animals , Benzoxazines , Brain Ischemia/therapy , Cell Count , Cerebral Arteries , Coloring Agents , Ligation , Male , Oxazines , Rats , Rats, Sprague-Dawley , Spectrin/metabolism , Tetrazolium Salts , Time Factors
14.
Neurol Res ; 17(4): 249-58, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7477738

ABSTRACT

Calpains are cytosolic, neutral proteases that normally exist in an inactive or quiescent state. They require higher than normal levels of calcium for activation which, once accomplished, lead to irreversible proteolysis of numerous cytoskeletal, membrane-associated and regulatory proteins. Because of these characteristics, calpain is gaining attention as a potentially important pathogenic variable in ischemic neuronal death. This manuscript explores this hypothesis by briefly reviewing current support for the role played by calpain in ischemic neurodegeneration, and then discussing a series of recently published studies which: 1. offer further evidence for the hypothesis, and 2. provide direct support for the idea that selective inhibition of calpain can greatly limit the neuronal damage that would normally occur following both global as well as focal brain ischemia. Thus, the data reviewed in this manuscript support the ideas that unregulated activation and proteolysis of intraneuronal calpain plays a significant role in the brain damage that occurs following an ischemic event and that delivering selective and membrane permeant calpain inhibitors to ischemic tissue may provide a powerfully effective therapeutic means of limiting neuronal damage.


Subject(s)
Calpain/physiology , Ischemic Attack, Transient/drug therapy , Nerve Degeneration/drug effects , Protease Inhibitors/therapeutic use , Animals , Calpain/antagonists & inhibitors , Cell Death , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/etiology , Neurons/pathology , Rats
15.
J Neurotrauma ; 11(6): 657-67, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7723065

ABSTRACT

Although duration of unconsciousness is commonly used as a prognostic index following traumatic brain injury (TBI), few controlled studies have statistically evaluated the relationship between unconsciousness and histologic pathology, particularly after moderate head injury. Using a pendulum-striker concussive device, a reproducible model of TBI in rats was developed. This model is uncomplicated by skull fractures, contusions, or experimenter-induced craniotomies. In the present study, the severity of the histopathology observed in this model of moderate closed-head injury at 48 h posttrauma is linearly related to the duration of unconsciousness (p < 0.0001). The pathology, assessed with a silver stain for neurodegeneration, is particularly striking if unconsciousness persists for 4 minutes or more. These data suggest that the initial period of unconsciousness may be a useful predictor of clinical brain histopathology associated with moderate closed-head injury, predicting either the degree of pathology and/or the rate it progresses if left untreated.


Subject(s)
Brain Concussion/pathology , Brain/pathology , Disease Models, Animal , Unconsciousness/pathology , Animals , Hippocampus/pathology , Male , Olfactory Bulb/pathology , Rats , Rats, Wistar , Time Factors
16.
Stroke ; 25(11): 2265-70, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7974554

ABSTRACT

BACKGROUND AND PURPOSE: This research was performed to determine whether a selective inhibitor of the calcium-dependent protease, calpain, could reduce ischemia-associated brain damage when peripherally administered after a vascular occlusion. METHODS: A variation of the rat middle cerebral artery occlusion model was used. A range of doses of AK295 (a novel calpain inhibitor synthesized for this purpose) was continuously infused through the internal carotid artery, beginning 1.25 hours from the initiation of the occlusion. Rats were killed at 21 hours, and the infarct volume was quantified. RESULTS: Postocclusion (1.25-hour) infusion of the calpain inhibitor AK295 elicited a dose-dependent neuroprotective effect after focal ischemia. The highest dose tested (3 mg/kg per hour) afforded the maximum effect, illustrated by a 32% reduction in infarct volume 21 hours after the ischemia (vehicle, 81.7 +/- 4.7 mm3; AK295, 54.9 +/- 6.9 mm3; P < .007). CONCLUSIONS: These data provide the first evidence that a peripherally administered calpain inhibitor can protect against ischemic brain damage. They offer further support for an important role of calpain proteolysis in the brain degeneration associated with cerebral ischemic events and suggest that selective calpain inhibitors provide a rational, novel, and viable means of treating such neurodegenerative problems.


Subject(s)
Brain Ischemia/pathology , Calpain/antagonists & inhibitors , Dipeptides/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Dipeptides/administration & dosage , Dose-Response Relationship, Drug , Injections, Intra-Arterial , Rats , Rats, Sprague-Dawley
17.
J Cereb Blood Flow Metab ; 14(4): 537-44, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8014200

ABSTRACT

Experiments were conducted to determine whether a potent, reversible calpain inhibitor could reduce the cortical ischemic brain damage associated with focal ischemia in the rat. AK275 (Z-Leu-Abu-CONH-CH2CH3), the active isomer of the diastereomeric mixture, CX275, was employed in conjunction with a novel method of perfusing drug directly onto the infarcted cortical surface. This protocol reduced or eliminated numerous, nonspecific pharmacokinetic, hemodynamic, and other potentially confounding variables that might complicate interpretation of any drug effect. Focal ischemia was induced using a variation of the middle cerebral artery occlusion method. These studies demonstrated a reliable and robust neuroprotective effect of AK275 over the concentration range of 10 to 200 microM (perfused supracortically at 4 microliters/h for 21 h). Moreover, a 75% reduction in infarct volume was observed when initiation of drug treatment was delayed for 3 h postocclusion. Our data further support an important role of calpain in ischemia-induced neuropathology and suggest that calpain inhibitors may provide a unique and potentially powerful means of treating stroke and other ischemic brain incidents.


Subject(s)
Brain Damage, Chronic/prevention & control , Brain Ischemia/complications , Brain Ischemia/therapy , Calpain/antagonists & inhibitors , Dipeptides/administration & dosage , Reperfusion , Animals , Body Temperature , Brain/pathology , Brain Ischemia/pathology , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Dipeptides/pharmacology , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley , Time Factors
18.
J Med Chem ; 35(22): 4143-9, 1992 Oct 30.
Article in English | MEDLINE | ID: mdl-1433217

ABSTRACT

A series of N6-(p-sulfophenyl)alkyl and N6-sulfoalkyl derivatives of adenosine was synthesized, revealing that N6-(p-sulfophenyl)adenosine (10b) is a moderately potent (Ki vs [3H]PIA in rat cortical membranes was 74nM) and A1-selective (120-fold) adenosine agonist, of exceptional aqueous solubility of > 1.5 g/mL (approximately 3 M). Compound 10b was very potent in inhibiting synaptic potentials in gerbil hippocampal slices with an IC50 of 63 nM. At a dose of 0.1 mg/kg ip in rats, 10b inhibited lipolysis (a peripheral A1 effect) by 85% after 1 h. This in vivo effect was reversed using the peripherally selective A1-antagonist 1,3-dipropyl-8-[p-(carboxyethynyl)phenyl]xanthine (BW1433). The same dose of 10b in NIH Swiss mice (ip) was nearly inactive in locomotor depression, an effect that has been shown to be centrally mediated when elicited by lower doses of other potent adenosine agonists, such as N6-cyclohexyladenosine (CHA) (Nikodijevic et al. FEBS Lett. 1990, 261, 67). HPLC studies of biodistribution of a closely related and less potent homologue, N6-[4-(p-sulfophenyl)butyl]adenosine indicated that a 25 mg/kg ip dose in mice resulted in a plasma concentration after 30 min of 0.46 micrograms/mL and no detectable drug in the brain (detection limit < 0.1% of plasma level). Although 10b at doses > 0.1 mg/kg in mice depressed locomotor activity, this depression was unlike the effects of CHA and was reversible by BW1433. These data suggest that 10b is a potent adenosine agonist in vivo and shows poor CNS penetration.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/chemical synthesis , Adenosine/metabolism , Adenosine/pharmacology , Animals , Body Temperature/drug effects , Brain/drug effects , Brain/metabolism , Evoked Potentials/drug effects , Gerbillinae , In Vitro Techniques , Lipolysis/drug effects , Male , Mice , Motor Activity/drug effects , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Purinergic/metabolism , Solubility , Structure-Activity Relationship , Sulfonic Acids/chemical synthesis , Sulfonic Acids/metabolism , Sulfonic Acids/pharmacology
19.
Health Care Superv ; 10(1): 58-62, 1991 Sep.
Article in English | MEDLINE | ID: mdl-10112258

ABSTRACT

Because the health care administrator faces unprecedented demands concerning information, well-designed and carefully selected information systems have become a critical factor in the viability and survival of health care institutions. Local area network systems are having a significant impact in this regard. Among their advantages are flexibility, dependability, low cost, and modularity. Health care facilities, have found networking to be a simple, cost-effective solution to collecting, processing, and distributing large volumes of data. The keys to a successful implementation recommended in this article should assist health care professionals once they have made the hard decision to use a local area network to process their day-to-day transactions.


Subject(s)
Hospital Information Systems , Local Area Networks , Contract Services/standards , Organization and Administration , Planning Techniques , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...