Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
1.
Neurobiol Dis ; 78: 162-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25841760

ABSTRACT

Substantial interest persists for developing neurotrophic factors to treat neurodegenerative diseases. At the same time, significant progress has been made in implementing gene therapy as a means to provide long-term expression of bioactive neurotrophic factors to targeted sites in the brain. Nonetheless, to date, no double-blind clinical trial has achieved positive results on its primary endpoint despite robust benefits achieved in animal models. A major issue with advancing the field is the paucity of information regarding the expression and effects of neurotrophic factors in human neurodegenerative brain, relative to the well-characterized responses in animal models. To help fill this information void, we examined post-mortem brain tissue from four patients with nigrostriatal degeneration who had participated in clinical trials testing gene delivery of neurturin to the putamen of patients. Each had died of unrelated causes ranging from 1.5-to-3-months (2 Parkinson's disease patients), to 4+-years (1 Parkinson's disease and 1 multiple-system atrophy-parkinsonian type patient) following gene therapy. Quantitative and immunohistochemical evaluation of neurturin, alpha-synuclein, tyrosine hydroxylase (TH) and an oligodendroglia marker (Olig 2) were performed in each brain. Comparable volumes-of-expression of neurturin were seen in the putamen in all cases (~15-22%; mean=18.5%). TH-signal in the putamen was extremely sparse in the shorter-term cases. A 6-fold increase was seen in longer-term cases, but was far less than achieved in animal models of nigrostriatal degeneration with similar or even far less NRTN exposure. Less than 1% of substantia nigra (SN) neurons stained for neurturin in the shorter-term cases. A 15-fold increase was seen in the longer-term cases, but neurturin was still only detected in ~5% of nigral cells. These data provide unique insight into the functional status of advanced, chronic nigrostriatal degeneration in human brain and the response of these neurons to neurotrophic factor stimulation. They demonstrate mild but persistent expression of gene-mediated neurturin over 4-years, with an apparent, time-related amplification of its transport and biological effects, albeit quite weak, and provide unique information to help plan and design future trials.


Subject(s)
Corpus Striatum/metabolism , Neurodegenerative Diseases/metabolism , Neurturin/metabolism , Substantia Nigra/metabolism , alpha-Synuclein/metabolism , Aged , Basic Helix-Loop-Helix Transcription Factors , Dependovirus , Genetic Therapy , Genetic Vectors , Humans , Middle Aged , Nerve Tissue Proteins , Neural Pathways/metabolism , Neural Pathways/pathology , Neural Pathways/virology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/therapy , Neurodegenerative Diseases/virology , Neurons/metabolism , Neurturin/genetics , Oligodendrocyte Transcription Factor 2 , Tyrosine 3-Monooxygenase/metabolism
2.
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
3.
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
4.
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
5.
J Comp Neurol ; 427(1): 1-18, 2000 Nov 06.
Article in English | MEDLINE | ID: mdl-11042588

ABSTRACT

Bradykinin has long been known to exist in the central nervous system and has been hypothesized to mediate specific functions. Despite an increasing understanding of the functions of bradykinin, little is known about the cell types expressing the bradykinin receptor within the brain. The present investigation employed a monoclonal antibody directed against the 15-amino-acid portion of the C-terminal of the human bradykinin B2 receptor to establish the cellular distribution of bradykinin B2 receptor immunoreactivity in the rat brain. Bradykinin B2 receptor immunoreactivity was ubiquitously and selectively observed in neurons, including those within the olfactory bulb, cerebral cortex, hippocampus, basal forebrain, basal ganglia, thalamus, hypothalamus, cerebellum, and brainstem nuclei. Bradykinin B2 receptor immunoreactivity was also present in the circumventricular organs including choroid plexus, subfornical organ, median eminence, and area postrema. Double-labeling experiments colocalizing the bradykinin B2 receptor with the neuronal marker NeuN or the astrocytic marker glial fibrillary acidic protein revealed that virtually 100% of the bradykinin B2 receptor-immunoreactive positive cells were neurons. The widespread distribution of bradykinin B2 receptor immunoreactivity in neuronal compartments suggests a greater than previously appreciated role for this peptide in neuronal function.


Subject(s)
Bradykinin/metabolism , Brain/metabolism , Rats, Inbred F344/metabolism , Receptors, Bradykinin/metabolism , Amygdala/cytology , Amygdala/metabolism , Animals , Antibody Specificity/immunology , Basal Ganglia/cytology , Basal Ganglia/metabolism , Basal Nucleus of Meynert/cytology , Basal Nucleus of Meynert/metabolism , Brain/cytology , Brain Stem/cytology , Brain Stem/metabolism , Cerebellum/cytology , Cerebellum/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Diencephalon/cytology , Diencephalon/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Humans , Immunohistochemistry , Male , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Rats , Rats, Inbred F344/anatomy & histology , Receptor, Bradykinin B2 , Septal Nuclei/cytology , Septal Nuclei/metabolism
6.
Pharm Res ; 17(7): 767-75, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10990193

ABSTRACT

PURPOSE: Implantation of biodegradable polymers provides a powerful method to deliver high, sustained concentrations of chemotherapeutics to brain tumors. The present studies examined the ability of injectable polymeric microspheres, formulated to release carboplatin or BCNU for 2-3 weeks, to enhance survival in a rodent model of surgically-resected glioma. METHODS: Rat glioma (RG2) cells were implanted into the cortex of rats and allowed to grow for 10 days prior to surgical resection. Rats were given either surgical resection only, bolus injection (100 microg) or microspheres containing 10, 50, or 100 microg of carboplatin or BCNU. The microspheres were implanted, via hypodermic injection, either directly into the surgical cavity or into the tissue along the perimeter of the cavity. RESULTS: The order of survival among treatment groups was: no resection < resection only < bolus chemotherapy < sustained release chemotherapy. Carboplatin and BCNU did not differ in this respect and in each case, the enhanced survival achieved with sustained release was dose-related. However, the enhanced survival achieved with carboplatin was substantially greater when the microspheres were implanted into the perimeter wall of the resection cavity, compared to implantation into the cavity itself. The enhanced survival produced by carboplatin implants along the resection perimeter was associated with a significant attenuation of regrowth of the tumor. Finally, in a separate study in non-tumor brain, atomic absorption spectrophotometry revealed that while the microspheres produced significantly prolonged tissue levels of carboplatin relative to a bolus injection, carboplatin diffusion was limited to brain tissue extending primarily 0.5 mm from the injection site. CONCLUSIONS: These data demonstrate: (1) that sustained delivery of chemotherapy is superior to equipotent bolus doses following tumor resection, and (2) that direct injection of sustained release microspheres into the tissue surrounding a growing tumor mass may provide superior effects over injections into the surgical cavity. They also suggest that successful implementation of this approach in humans may require measures or circumstances that improve upon the limited spatial drug diffusion from the implantation site.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Carmustine/administration & dosage , Glioma/drug therapy , Polymers/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Carboplatin/administration & dosage , Carboplatin/pharmacokinetics , Delayed-Action Preparations , Glioma/metabolism , Glioma/mortality , Male , Microspheres , Polymers/pharmacokinetics , Rats , Rats, Inbred F344
7.
Pharm Res ; 17(7): 776-81, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10990194

ABSTRACT

PURPOSE: Delivery of chemotherapeutics using implantable, biodegradable polymers provides a potentially powerful method of treating brain tumors. The present studies examined the ability of injectable microspheres, formulated to release carboplatin or BCNU for 2-3 weeks, to enhance survival in a rodent model of deep, inoperable glioma. METHODS: Rat glioma (RG2) cells were implanted into the striatum of rats. In a first experiment, the tumors were allowed to grow for 3 days, followed by either no treatment, bolus chemotherapy (100 microg), or implantation of microspheres containing 10, 50, or 100 microg of carboplatin. The microspheres were implanted, via hypodermic injection, directly into the center of the small, 3-day-old tumors. In a second experiment, tumors grew for 8 days prior to treatment with either carboplatin- or BCNU-loaded microspheres. The microspheres were then injected either directly into the center of these larger tumors or into three sites along the perimeter of the tumor. Separate sets of animals received bolus chemotherapy (100 microg) into either the tumor center or around the tumor perimeter. RESULTS: Injection of carboplatin-loaded microspheres into the center of the small 3 day old, tumors produced dose-related increases in survival. When injections of carboplatin- or BCNU-loaded microspheres were made into the center of the larger, 8-day-old tumors, survival was not enhanced. However, when the microspheres were injected along the perimeter of the larger tumors, sustained-release chemotherapy did significantly prolong survival. Bolus chemotherapy was less effective than sustained release chemotherapy. CONCLUSIONS: Together, these data: (1) demonstrate that sustained delivery of chemotherapy in or near the tumor site is superior to equipotent bolus doses in inoperable tumors, (2) demonstrate that injection of sustained release microspheres into the tissue surrounding a growing tumor may provide superior effects over injections directly into the tumor mass, and (3) suggest that this approach may provide a useful means of selectively delivering chemotherapeutics to tumors or portions of tumors that cannot otherwise be treated with conventional surgical approaches.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Carboplatin/administration & dosage , Carmustine/administration & dosage , Corpus Striatum/drug effects , Glioma/drug therapy , Polymers/administration & dosage , Animals , Brain Neoplasms/mortality , Delayed-Action Preparations , Glioma/mortality , Male , Microspheres , Rats , Rats, Inbred F344
8.
Neurosurgery ; 47(1): 189-97; discussion 197-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10917362

ABSTRACT

OBJECTIVE: Cereport (Alkermes, Inc., Cambridge, MA), or, as it has been previously called, RMP-7 (receptor-mediated permeabilizer-7), is a bradykinin analog that has been shown to produce a transient, pharmacologically mediated opening of the blood-brain barrier. The purpose of the present study was to determine whether the efficacy of boron neutron capture therapy (BNCT) could be enhanced by means of intracarotid (i.c.) infusion of Cereport, in combination with intravenous (i.v.) injection or i.c. infusion of boronophenylalanine (BPA) in the F98 rat glioma model. METHODS: For biodistribution studies, Fischer rats bearing intracerebral implants of the F98 glioma received i.v. or i.c. injections of 300 or 500 mg/kg body weight (b.w.) of BPA with or without i.c. infusion of 1.5 microg/kg b.w. of Cereport. For therapy studies, BNCT was initiated 14 days after intracerebral implantation of 10(3) F98 cells. The i.v. or i.c. injection of BPA (500 mg/kg b.w.) was given with or without Cereport, and the animals were irradiated 2.5 hours later at the Brookhaven Medical Research Reactor with a collimated beam of thermal neutrons delivered to the head. RESULTS: At a BPA dose of 500 mg/kg b.w., tumor boron concentrations (mean +/- standard deviation) were 55.7 +/- 9.6 microg/g with Cereport versus 33.6 +/- 3.9 microg/g without Cereport at 2.5 hours after i.c. infusion of BPA, and concentrations were 29.4 +/- 9.9 microg/g with Cereport versus 15.4 +/- 3.5 microg/g without Cereport (P < 0.05) after i.v. injection of BPA. After i.c. administration of BPA and Cereport, the tumor-to-blood ratio was 5.4 +/- 0.6, and the tumor-to-brain ratio was 5.2 +/- 2.4. After BNCT with BPA at a dose of 500 mg/kg, the survival time was 50 +/- 16 days for i.c. administration of BPA with Cereport versus 40 +/- 6 days without Cereport (P = 0.05), 38 +/- 4 days for i.v. administration of BPA with Cereport versus 34 +/- 3 days without Cereport (P = 0.02), 28 +/- 5 days for irradiated controls, and 23 +/- 3 days for untreated controls. Compared with untreated controls, there was a 117% increase in lifespan in rats that received an i.c. infusion of Cereport and then BPA, and an 86% increase in lifespan in rats that received i.c. administration of BPA without Cereport. CONCLUSION: These studies have established that i.c. administration of Cereport can not only increase tumor uptake of BPA, but also enhance the efficacy of BNCT.


Subject(s)
Blood-Brain Barrier/drug effects , Boron Compounds/therapeutic use , Boron Neutron Capture Therapy , Bradykinin/analogs & derivatives , Brain Neoplasms/mortality , Brain Neoplasms/radiotherapy , Glioma/mortality , Glioma/radiotherapy , Phenylalanine/analogs & derivatives , Radiation-Sensitizing Agents/therapeutic use , Animals , Boron Compounds/pharmacokinetics , Bradykinin/pharmacology , Phenylalanine/pharmacokinetics , Phenylalanine/therapeutic use , Radiation-Sensitizing Agents/pharmacokinetics , Rats , Rats, Inbred F344 , Survival Rate , Tissue Distribution
9.
Exp Neurol ; 163(2): 495-529, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10833325

ABSTRACT

Life's Journey If life is indeed a journey, then poetry must be the map that reveals all its topographic possibilitiesellipsis while science is the compass that keeps us from getting lost. -R. T. Bartus, Simple Words for Complex Lives, (c) 1998 In the nearly 20 years since the cholinergic hypothesis was initially formulated, significant progress has been achieved. Initial palliative treatments for Alzheimer's disease (AD) have proven beneficial and have gained FDA approval, the use of animal models for studying AD and other neurodegenerative diseases has achieved wider acceptance, and important insight into the potential causes and pathogenic variables associated with various neurodegenerative diseases continues to increase. This paper reviews the current status of the cholinergic hypothesis in the context of continuing efforts to improve upon existing treatments for AD and explores the role that animal models might continue to play. Using the benefit of hindsight, particular emphasis is placed on an analysis of the approaches, strategies, and assumptions regarding animal models that proved useful in developing the initial treatments and those that did not. Additionally, contemporary issues of AD are discussed within the context of the cholinergic hypothesis, with particular attention given to how they may impact the further refinement of animal models, and the development of even more effective treatments. Finally, arguments are presented that, despite the deserved enthusiasm and optimism for identifying means of halting the pathogenesis of AD, a clear need for more effective palliative treatments will continue, long after successful pathogenic treatments are available. This review, therefore, focuses on issues and experiences intended to: (a) facilitate further development and use of animal models for AD and other neurodegenerative diseases, and (b) accelerate the identification of newer, even more effective treatments.


Subject(s)
Alzheimer Disease/therapy , Cholinergic Agents , Cognition , Disease Models, Animal , Memory Disorders/therapy , Age Factors , Animals , Cholinergic Agents/pharmacology , Cholinergic Agents/therapeutic use , Cognition/drug effects , Cognition/physiology , Forecasting , Humans
10.
J Pharmacol Exp Ther ; 293(3): 903-11, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10869391

ABSTRACT

Several experiments studied the effects of i.v. infusions of the bradykinin agonist, Cereport (RMP-7), on permeability of the blood-brain tumor barrier in rat gliomas. First, the ability of Cereport to increase uptake of two poorly blood-brain barrier-penetrating drugs (lypophilic paclitaxel and hydrophilic carboplatin) was directly compared to provide new information regarding the scope of delivery effects achieved with Cereport. Next, the increased uptake of platinum into tumor and brain surrounding tumor was shown to closely parallel that of radiolabeled carboplatin, confirming that delivery of a biologically active moiety is increased with Cereport. This study also demonstrated that the elevated tumor levels of platinum persisted for at least 2 h. The enhanced carboplatin uptake was then examined using a novel, high spatial resolution analysis of autoradiography. This revealed that the effects of Cereport were not uniform throughout the tumor, because it especially modified those areas normally impermeable to carboplatin. Finally, a range of i.v. Cereport doses (3.0 and 9.0 microg/kg) was tested in combination with carboplatin to determine whether increased survival might be achieved and to define the relationship between Cereport dose, plasma levels, uptake of carboplatin, and enhanced survival. Survival was enhanced only by the high dose of Cereport; the high dose also produced robust increases in carboplatin uptake and plasma concentrations of Cereport estimated to achieve the K(i), whereas the low dose did not. These data offer fundamental information regarding the effects of Cereport on delivery of chemotherapeutic agents to brain tumors and provide new insight into receptor-mediated permeability of the blood-brain tumor barrier.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bradykinin/analogs & derivatives , Brain Neoplasms/drug therapy , Carboplatin/pharmacokinetics , Glioma/drug therapy , Animals , Bradykinin/administration & dosage , Bradykinin/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Glioma/metabolism , Glioma/mortality , Infusions, Intravenous , Male , Paclitaxel/pharmacokinetics , Platinum/pharmacokinetics , Rats , Rats, Inbred F344
11.
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
12.
J Cereb Blood Flow Metab ; 20(1): 66-73, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10616794

ABSTRACT

Administration of the selective calpain inhibitor AK295 has been shown to attenuate motor and cognitive dysfunction after brain trauma in rats. To explore mechanisms underlying the behavioral efficacy of posttraumatic calpain inhibition, we investigated histologic consequences of AK295 administration. Anesthetized Sprague-Dawley rats received lateral fluid percussion brain injury of moderate severity (2.2 to 2.4 atm) or served as uninjured controls. At 15 minutes after injury, animals were randomly assigned to receive a 48-hour infusion of either 2 mmol/L AK295 (120 to 140 mg/kg) or saline via the carotid artery. At 48 hours and 1 week after injury, spectrin fragments generated specifically by calpain were detected by Western blotting and immunohistochemistry, respectively, in saline-treated, brain-injured animals. Interestingly, equivalent spectrin breakdown was observed in AK295-treated animals when cortical and hippocampal regions were evaluated. Similarly, administration of the calpain inhibitor did not attenuate cortical lesion size or the numbers of apoptotic cells in the cortex, subcortical white matter, or hippocampus, as verified by terminal deoxynucleotidyl transferase-mediated biotinylated deoxyuridine triphosphate nick-end labeling and morphology, at 48 hours after injury. These data suggest that an overt reduction in spectrin proteolysis, cortical lesion, or apoptotic cell death at 48 hours or 1 week is not required for behavioral improvements associated with calpain inhibition by AK295 after experimental brain injury in rats.


Subject(s)
Behavior, Animal/drug effects , Brain Injuries/metabolism , Brain Injuries/psychology , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Animals , Apoptosis/drug effects , Brain Injuries/physiopathology , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Male , Peptide Hydrolases/metabolism , Rats , Rats, Sprague-Dawley , Spectrin/metabolism
13.
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
14.
Alcohol Clin Exp Res ; 23(11): 1761-71, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10591592

ABSTRACT

BACKGROUND: This study was planned to determine the feasibility of using a slow release naloxone preparation to treat alcoholism, because compliance with medication is a significant problem in alcoholics. METHODS: Experiments were performed in alcohol-preferring P rats maintained either on continuous access or on limited access (1 hr/day) to alcohol with water and food provided ad libitum. Naloxone (Nx) was administered either by twice daily subcutaneous injections or by slow release (1.1 mg/kg/hr) osmotic minipump. In limited access experiments, Nx was injected immediately before access to alcohol. RESULTS: An initial experiment estimated the dose-effect curve for Nx subcutaneous suppression on alcohol intake. Nx (2.5-20 mg/kg) had a stronger effect during the first 2 hr after injection (ED50 = 2.1 mg/kg); however, the effect was more modest on 24-hr consumption. Similar results were found with chronic Nx treatment. Low doses of Nx (0.5 and 2.0 mg/kg) injected immediately before limited access to alcohol produced almost complete suppression of alcohol intake for at least 14 consecutive days. However, 14 days of treatment with 26 mg/kg/day by minipump or injection produced an initial 50% suppression of 24-hr alcohol intake with the gradual development of tolerance. An acute challenge with Nx immediately after the pumps were scheduled to be empty provided additional evidence of tolerance development in chronically Nx-treated rats. Brain micro-opiate receptors, estimated autoradiographically by using the ligand [3H][D-Ala2,N-Me-Phe4, Gly-ol5][tyrosyl-3,5-3H]-enkephalin, showed that rats chronically exposed to Nx and showing tolerance to Nx suppression of drinking exhibited 17% to 250% increases in [3H][D-Ala2,N-Me-Phe4, Gly-ol5][tyrosyl-3,5-3H]-enkephalin binding. CONCLUSIONS: High doses of Nx are required to suppress continuous access alcohol consumption in P rats, and tolerance develops to the ethanol consumption-suppressing effect of Nx that may be related to increases in micro-opiate receptors.


Subject(s)
Alcohol Drinking/drug therapy , Drug Tolerance/physiology , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Receptors, Opioid, mu/metabolism , Alcohol Drinking/metabolism , Analgesics, Opioid/pharmacology , Animals , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Feasibility Studies , Male , Rats , Receptors, Opioid, mu/drug effects
16.
Pharm Res ; 16(9): 1360-5, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10496650

ABSTRACT

PURPOSE: To study Cereport (RMP-7, bradykinin B2 agonist) effects on human brain microvascular endothelial cell (HBMEC) monolayer permeability. METHODS: HBMEC grown on transwell membranes were exposed to Cereport. The monolayer permeability was determined with [14C]-inulin (MW. 5,200) and [3H]-dextran (MW. 70,000). RESULTS: Cereport increased the HBMEC permeability to [14C]-inulin, but not to [3H]-dextran. The effect was transient, maximal at 15 min (i.e., 79.3% increase), and polarized to the basolateral membrane. An inverted U, dose-response curve was observed with active concentrations of Cereport from 0.01 to 0.5 nmol/L, the plateau maximal effect between 0.5 and 10 nmol/L, and loss of activity at the highest concentration, i.e., 20 nmol/L. Cyclic AMP-specific phosphodiesterase 3 (PDE3) inhibitor rolipram (10 micromol/L) abolished Cereport effects, while cGMP-specific PDE5 inhibitor, zaniprast (50 micromol/L) enhanced by 31% (p < 0.05) the effect of 0.1 nmolL Cereport. Unlabeled Cereport displaced [125I]-bradykinin and/or [125I]-Cereport from the basolateral side. There was no specific Cereport binding to the apical side. CONCLUSIONS: Cereport exerts specific time, dose and size dependent actions on HMBEC monolayer that are restricted to the basolateral membrane. Its effects can be further modulated through changes in cAMP and cGMP second messenger systems.


Subject(s)
Blood-Brain Barrier/drug effects , Bradykinin/analogs & derivatives , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Inulin/pharmacokinetics , Binding, Competitive , Bradykinin/metabolism , Bradykinin/pharmacology , Carbon Radioisotopes , Cells, Cultured , Cerebral Cortex/blood supply , Dextrans/pharmacokinetics , Humans , Iodine Radioisotopes , Microcirculation/drug effects , Phosphodiesterase Inhibitors/pharmacology , Purinones/pharmacology , Receptor, Bradykinin B2 , Receptors, Bradykinin/physiology , Rolipram/pharmacology , Tritium
17.
Nat Med ; 5(8): 881-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10426310

ABSTRACT

The occurrence of multiple tumors in an organ heralds a rapidly fatal course. Although intravascular administration may deliver oncolytic viruses/vectors to each of these tumors, its efficiency is impeded by an antiviral activity present in complement-depleted plasma of rodents and humans. Here, this activity was shown to interact with complement in a calcium-dependent fashion, and antibody neutralization studies indicated preimmune IgM has a contributing role. Short-term exposure to cyclophosphamide (CPA) partially suppressed this activity in rodents and humans. At longer time points, cyclophosphamide also abrogated neutralizing antibody responses. Cyclophosphamide treatment of rats with large single or multiple intracerebral tumors substantially increased viral survival and propagation, leading to neoplastic regression.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , Immunosuppression Therapy , Viruses/immunology , Animals , Antibodies, Viral/blood , Antibody Formation/drug effects , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Brain Neoplasms/virology , Complement System Proteins/immunology , Cyclophosphamide/pharmacology , Female , Glioma/mortality , Glioma/therapy , Glioma/virology , Humans , Immunity, Innate/drug effects , Immunoglobulin M/blood , Male , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Rats, Nude , Survival Rate , Time Factors , Tumor Cells, Cultured , Viruses/isolation & purification
18.
Br J Cancer ; 80(7): 964-70, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10362103

ABSTRACT

Cereport (RMP-7) is a selective bradykinin B2 receptor agonist which increases the permeability of the 'blood-brain tumour barrier' (BBTB) to increase delivery of chemotherapeutic agents to brain tumours. A series of experiments was performed in an RG2 rodent model of glioma to evaluate and refine intravenous (i.v.) parameters to optimize Cereport's clinical utility. The first experiment demonstrated that while carboplatin levels were increased by twofold when given as a bolus during the Cereport infusion, no increase in carboplatin levels were seen when Cereport and carboplatin were simultaneously co-infused for 15 min. A subsequent experiment established that a major factor responsible for the lack of an effect with the co-infusion paradigm was tachyphylaxis to Cereport during the 15 min infusion, for a progressively diminished response to Cereport occurred over that time frame, as plasma levels of carboplatin were rising. A final experiment adjusted the timing of the Cereport and carboplatin infusions so that higher plasma carboplatin levels were achieved prior to initiating the Cereport infusion. Significant uptake effects were achieved when the carboplatin infusion preceded the Cereport infusion by 10 min (i.e. 5 min overlap in the delivery of the two agents). Collectively, these data provide the first systematic evaluation of dosing parameters involving receptor-mediated changes in BBTB permeability and provide new information regarding the pharmacodynamics and potential clinical use of Cereport.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bradykinin/analogs & derivatives , Brain Neoplasms/drug therapy , Carboplatin/administration & dosage , Carboplatin/pharmacokinetics , Glioma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Blood-Brain Barrier/drug effects , Bradykinin/administration & dosage , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Synergism , Male , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Tachyphylaxis
19.
Cell Transplant ; 8(1): 47-58, 1999.
Article in English | MEDLINE | ID: mdl-10338275

ABSTRACT

The delivery of therapeutic molecules to the brain has been limited in part due to the presence of the blood-brain barrier. One potential solution is the implantation of biodegradable polymers with sustained release of drugs. Poly (DL-lactide-co-glycolide) (PLG) is a bioerodible polymer with a long and successful history of use as a suture material. More recently, PLG has been investigated for localized and sustained delivery of molecules into both peripheral sites and the brain. Despite its well-defined safety profile for parenteral applications, little information exists concerning the safety of PLG when implanted into the brain. To further characterize the biocompatibility of PLG in the brain, we examined the gliotic response following implants of PLG into the brains of rats. As a control, each animal received an injection of the suspension medium into the contralateral hemisphere. Following implantation, PLG was well tolerated. GFAP-positive astrocytes were observed throughout the cerebral cortex and striatum on both the implanted and control sides, with the reaction being greatest within the heavily myelinated fiber tracts of the corpus callosum. Quantitative analyses revealed that this reaction occurred within 1 h postsurgery, reached its peak at 1 week following surgery, and then decreased markedly by 1 month postsurgery. A minimal gliotic reaction was still present 1 year postsurgery but was localized to the needle tract. No differences in GFAP reactivity were seen between the polymer-implanted and control sides at any time point. Histological analysis determined that the majority of the PLG disappeared between 1 and 4 weeks. A set of parallel studies in which PLG samples were retrieved from the brain at various time points corroborated these findings and determined that the majority of PLG degraded within 2 weeks following implantation. Together, these results demonstrate that PLG is well tolerated following implantation into the CNS and that the astrocytic response to PLG is largely a consequence of the mechanical trauma that occurs during surgery. The biocompatibility of PLG implanted into the CNS provides further support for its use in a wide range of new therapeutic applications for sustained and localized drug delivery to the brain.


Subject(s)
Biocompatible Materials/administration & dosage , Brain/surgery , Delayed-Action Preparations/administration & dosage , Lactic Acid/administration & dosage , Polyglycolic Acid/administration & dosage , Polymers/administration & dosage , Animals , Corpus Striatum/surgery , Glial Fibrillary Acidic Protein/isolation & purification , Injections, Intraventricular , Lactic Acid/metabolism , Male , Microspheres , Polyglycolic Acid/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution
20.
Exp Neurol ; 155(2): 315-26, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10072307

ABSTRACT

Many forms of acute brain injury are associated with a secondary, glutamate- and calcium-dependent cascade which greatly exacerbates the final damage. The calcium-dependent protease, calpain, has been implicated as an important variable in this pathogenic process. The present studies tested (i) if similar secondary degeneration occurs following surgical ablation of a discrete area within rat visual cortex, (ii) if activation of calpain contributes to the secondary degeneration by spreading into areas adjacent to the ablation, and (iii) if blocking calpain's proteolytic effects reduces the secondary degeneration attendant to the lesion. Antibodies selective for a protein fragment specifically generated by calpain were used to map areas in which the protease had been activated. Labeling was present 5 min after surgery in a narrow zone surrounding the ablated region. The volume of the immunopositive staining increased twofold within 24 h and fivefold by 48 h, at which time it was equivalent in size to the original lesion. This staining pattern significantly decreased in size at 5 days postsurgery. Application of calpain inhibitors to the ablation site immediately after surgery reduced the spread of calpain activation by approximately 80%. Following cortical ablation, the volume of the lateral geniculate nucleus ipsilateral to the cortical ablation shrank by 46 +/- 3% in control rats but only by 31 +/- 5% in animals given the calpain inhibitors. These results establish that (i) a secondary degenerative cascade is unleashed following discrete cortical surgery which expands into brain areas clearly outside the initial perturbation site, (ii) the gradual expansion of calpain activation contributes to the underlying secondary pathology, and (iii) blocking calpain activity substantially reduces atrophy in areas anatomically connected, but physically distal to the damaged zone. The possible utility of topical applications of calpain inhibitors, or analogously acting drugs, in minimizing the secondary effects of brain surgery is discussed.


Subject(s)
Calcium/physiology , Calpain/antagonists & inhibitors , Cerebral Cortex/pathology , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Animals , Antibodies , Calpain/immunology , Cerebral Cortex/injuries , Geniculate Bodies/pathology , Immunohistochemistry , Male , Nerve Degeneration/pathology , Rats , Rats, Sprague-Dawley , Spectrin/immunology , Spectrin/metabolism , Visual Cortex/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...