Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Trends Pharmacol Sci ; 43(3): 221-233, 2022 03.
Article in English | MEDLINE | ID: mdl-34887129

ABSTRACT

Beta cell replacement has emerged as an attractive therapeutic alternative to traditional exogenous insulin administration for management of type 1 diabetes (T1D). Beta cells deliver insulin dynamically based on individual glycometabolic requirements, providing glycemic control while significantly reducing patient burden. Although transplantation into the portal circulation is clinically available, poor engraftment, low cell survival, and immune rejection have sparked investigation of alternative strategies for beta cell transplantation. In this review, we focus on current micro- and macroencapsulation technologies for beta cell transplantation and evaluate their advantages and challenges. Specifically, we comment on recent methods to ameliorate graft hypoxia including enhanced vascularization, reduction of pericapsular fibrotic overgrowth (PFO), and oxygen supplementation. We also discuss emerging beta cell-sourcing strategies to overcome donor shortage and provide insight into potential approaches to address outstanding challenges in the field.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Islets of Langerhans , Diabetes Mellitus, Type 1/surgery , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/methods
2.
Front Behav Neurosci ; 15: 661973, 2021.
Article in English | MEDLINE | ID: mdl-34366802

ABSTRACT

Historically, many investigations into neurodegenerative diseases have focused on alterations in specific neuronal populations such as, for example, the loss of midbrain dopaminergic neurons in Parkinson's disease (PD) and loss of cholinergic transmission in Alzheimer's disease (AD). However, it has become increasingly clear that mammalian brain activities, from executive and motor functioning to memory and emotional responses, are strictly regulated by the integrity of multiple interdependent neuronal circuits. Among subcortical structures, the dopaminergic nigrostriatal and mesolimbic pathways as well as cholinergic innervation from basal forebrain and brainstem, play pivotal roles in orchestrating cognitive and non-cognitive symptoms in PD and AD. Understanding the functional interactions of these circuits and the consequent neurological changes that occur during degeneration provides new opportunities to understand the fundamental inter-workings of the human brain as well as develop new potential treatments for patients with dysfunctional neuronal circuits. Here, excerpted from a session of the European Behavioral Pharmacology Society meeting (Braga, Portugal, August 2019), we provide an update on our recent work in behavioral and cellular neuroscience that primarily focuses on interactions between cholinergic and dopaminergic systems in PD models, as well as stress in AD. These brief discussions include descriptions of (1) striatal cholinergic interneurons (CINs) and PD, (2) dopaminergic and cholinergic modulation of impulse control, and (3) the use of an implantable cell-based system for drug delivery directly the into brain and (4) the mechanisms through which day life stress, a risk factor for AD, damage protein and RNA homeostasis leading to AD neuronal malfunction.

3.
Neural Plast ; 2020: 8814028, 2020.
Article in English | MEDLINE | ID: mdl-33293946

ABSTRACT

Neuroinflammation and cytokine-dependent neurotoxicity appear to be major contributors to the neuropathology in Parkinson's disease (PD). While pharmacological advancements have been a mainstay in the treatment of PD for decades, it is becoming increasingly clear that nonpharmacological approaches including traditional and nontraditional forms of exercise and physical rehabilitation can be critical adjunctive or even primary treatment avenues. Here, we provide an overview of preclinical and clinical research detailing the biological role of proinflammatory molecules in PD and how motor rehabilitation can be used to therapeutically modulate neuroinflammation, restore neural plasticity, and improve motor function in PD.


Subject(s)
Cytokines/metabolism , Exercise Therapy , Exercise/physiology , Nerve Growth Factors/metabolism , Parkinson Disease/rehabilitation , Exercise Therapy/methods , Humans , Neuronal Plasticity/physiology , Parkinson Disease/physiopathology
4.
Expert Opin Drug Deliv ; 17(8): 1113-1118, 2020 08.
Article in English | MEDLINE | ID: mdl-32515621

ABSTRACT

INTRODUCTION: Diabetes mellitus is an ever-increasing medical condition that currently suffers 1 of 11 adults who may have lifelong commitment with insulin injections. Cell-laden hydrogels releasing insulin may provide the ultimate means of correcting diabetes. Here, we provide insights of this cell-based approach including latest preclinical and clinical progress both from academia and industry. AREA COVERED: The present article focuses on reviewing latest advances in cell-laden hydrogels both from the technological and biological perspective. The most relevant clinical results including clinical trials are also discussed. EXPERT OPINION: Current progress in technological issues (stem cells, devices, biomaterials) have contributed cell encapsulation science to have a very relevant progress in the field of diabetes treatment.


Subject(s)
Alginates/chemistry , Diabetes Mellitus/drug therapy , Insulin/administration & dosage , Humans , Hydrogels
5.
Curr Res Pharmacol Drug Discov ; 1: 19-29, 2020 Apr.
Article in English | MEDLINE | ID: mdl-34909639

ABSTRACT

Delivering glial cell line-derived neurotrophic factor (GDNF) to the brain is a potential treatment for Parkinson's Disease (PD). Here we use an implantable encapsulated cell technology that uses modified human clonal ARPE-19 â€‹cells to deliver of GDNF to the brain. In vivo studies demonstrated sustained delivery of GDNF to the rat striatum over 6 months. Anatomical benefits and behavioral efficacy were shown in 6-OHDA lesioned rats where nigral dopaminergic neurons were preserved in neuroprotection studies and dopaminergic fibers were restored in neurorecovery studies. When larger, clinical-sized devices were implanted for 3 months into the putamen of Göttingen minipigs, GDNF was widely distributed throughout the putamen and caudate producing a significant upregulation of tyrosine hydroxylase immunohistochemistry. These results are the first to provide clear evidence that implantation of encapsulated GDNF-secreting cells deliver efficacious and biologically relevant amounts of GDNF in a sustained and targeted manner that is scalable to treat the large putamen in patients with Parkinson's disease.

7.
Neural Plast ; 2019: 6286197, 2019.
Article in English | MEDLINE | ID: mdl-30984255

ABSTRACT

Methods: Human ARPE-19 cells engineered to secrete high levels of the glial cell line-derived neurotrophic factor (GDNF) were encapsulated into hollow fiber membranes. The devices were implanted into the rat striatum 1 week prior to striatal quinolinic acid injections. Animals were evaluated using a battery of validated motor tests, and histology was performed to determine the extent of GDNF diffusion and associated prevention of neuronal cell loss and behavioral deficits. Results: Encapsulated cell-based delivery of GDNF produced widespread distribution of GDNF throughout the entire implanted striatum. Stereological estimates of striatal neuron number and volume of lesion size revealed that GDNF delivery resulted in near complete neuroprotection. Conclusions: Delivery of neurotrophic molecules such as GDNF using encapsulated cells has reached a technological point where clinical evaluation is justified. Because GDNF has been effective in animal models of Parkinson's disease, stroke, epilepsy, and Huntington's disease, among other debilitating neurodegenerative diseases, encapsulated cell-based delivery of GDNF might represent one innovative means of slowing the neural degeneration seen in a myriad of currently untreatable neurological diseases.


Subject(s)
Corpus Striatum/drug effects , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Neuroprotective Agents/administration & dosage , Quinolinic Acid/toxicity , Animals , Cell Encapsulation , Cell Line , Drug Delivery Systems , Humans , LLC-PK1 Cells , Male , Neurodegenerative Diseases/drug therapy , Neurons/drug effects , Rats, Sprague-Dawley , Swine
8.
J Neurosci ; 39(11): 2144-2156, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30665947

ABSTRACT

Neurotrophic factors are candidates for treating epilepsy, but their development has been hampered by difficulties in achieving stable and targeted delivery of efficacious concentrations within the desired brain region. We have developed an encapsulated cell technology that overcomes these obstacles by providing a targeted, continuous, de novo synthesized source of high levels of neurotrophic molecules from human clonal ARPE-19 cells encapsulated into hollow fiber membranes. Here we illustrate the potential of this approach for delivering glial cell line-derived neurotrophic factor (GDNF) directly to the hippocampus of epileptic rats. In vivo studies demonstrated that bilateral intrahippocampal implants continued to secrete GDNF that produced high hippocampal GDNF tissue levels in a long-term manner. Identical implants robustly reduced seizure frequency in the pilocarpine model. Seizures were reduced rapidly, and this effect increased in magnitude over 3 months, ultimately leading to a reduction of seizures by 93%. This effect persisted even after device removal, suggesting potential disease-modifying benefits. Importantly, seizure reduction was associated with normalized changes in anxiety and improved cognitive performance. Immunohistochemical analyses revealed that the neurological benefits of GDNF were associated with the normalization of anatomical alterations accompanying chronic epilepsy, including hippocampal atrophy, cell degeneration, loss of parvalbumin-positive interneurons, and abnormal neurogenesis. These effects were associated with the activation of GDNF receptors. All in all, these results support the concept that the implantation of encapsulated GDNF-secreting cells can deliver GDNF in a sustained, targeted, and efficacious manner, paving the way for continuing preclinical evaluation and eventual clinical translation of this approach for epilepsy.SIGNIFICANCE STATEMENT Epilepsy is one of the most common neurological conditions, affecting millions of individuals of all ages. These patients experience debilitating seizures that frequently increase over time and can associate with significant cognitive decline and psychiatric disorders that are generally poorly controlled by pharmacotherapy. We have developed a clinically validated, implantable cell encapsulation system that delivers high and consistent levels of GDNF directly to the brain. In epileptic animals, this system produced a progressive and permanent reduction (>90%) in seizure frequency. These benefits were accompanied by improvements in cognitive and anxiolytic behavior and the normalization of changes in CNS anatomy that underlie chronic epilepsy. Together, these data suggest a novel means of tackling the frequently intractable neurological consequences of this devastating disorder.


Subject(s)
Epilepsy/drug therapy , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Neuroprotective Agents/administration & dosage , Seizures/drug therapy , Animals , Cell Encapsulation , Cell Line , Drug Delivery Systems/methods , Epilepsy/chemically induced , Humans , Male , Pilocarpine/administration & dosage , Rats, Sprague-Dawley , Seizures/chemically induced
9.
Prog Retin Eye Res ; 68: 67-82, 2019 01.
Article in English | MEDLINE | ID: mdl-30342088

ABSTRACT

Millions of people worldwide suffer from debilitating, progressive, and often permanent loss of vision without any viable treatment options. The complex physiological barriers of the eye contribute to the difficulty in developing novel therapies by limiting our ability to deliver therapeutics in a sustained and controlled manner; especially when attempting to deliver drugs to the posterior eye or trying to regenerate the diseased retina. Cell-based therapies offer a significant potential advancement in these situations. In particular, encapsulating, or immunoisolating, cells within implantable, semi-permeable membranes has emerged as a clinically viable means of delivering therapeutic molecules to the eye for indefinite periods of time. The optimization of encapsulation device designs is occurring together with refinements in biomaterials, genetic engineering, and stem-cell production, yielding, for the first time, the possibility of widespread therapeutic use of this technology. Here, we highlight the status of the most advanced and widely explored iteration of cell encapsulation with an eye toward translating the potential of this technological approach to the medical reality.


Subject(s)
Biological Products/administration & dosage , Cell- and Tissue-Based Therapy/methods , Drug Delivery Systems , Eye Diseases/therapy , Polymers/administration & dosage , Humans
10.
Gene Ther ; 26(3-4): 65-74, 2019 04.
Article in English | MEDLINE | ID: mdl-30464254

ABSTRACT

Temporal lobe epilepsy (TLE) is the most common type of epilepsy in adults. This neurological disorder is characterized by focal seizures originating in the temporal lobe, often with secondary generalization. A variety of pharmacological treatments exist for patients suffering from focal seizures, but systemically administered drugs offer only symptomatic relief and frequently cause unwanted side effects. Moreover, available drugs are ineffective in one third of the epilepsy patients. Thus, developing more targeted and effective treatment strategies for focal seizures, originating from, e.g., the temporal lobe, is highly warranted. In order to deliver potential anti-epileptic agents directly into the seizure focus we used encapsulated cell biodelivery (ECB), a specific type of ex vivo gene therapy. Specifically, we asked whether unilateral delivery of glial cell line-derived neurotrophic factor (GDNF), exclusively into the epileptic focus, would suppress already established spontaneous recurrent seizures (SRS) in rats. Our results show that GDNF delivered by ECB devices unilaterally into the seizure focus in the hippocampus effectively decreases the number of SRS in epileptic rats. Thus, our study demonstrates that focal unilateral delivery of neurotrophic factors, such as GDNF, using ex vivo gene therapy based on ECB devices could be an effective anti-epileptic strategy providing a bases for the development of a novel, alternative, treatment for focal epilepsies.


Subject(s)
Genetic Therapy/methods , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Seizures/therapy , Animals , Anticonvulsants/pharmacology , Brain/metabolism , Disease Models, Animal , Epilepsy/genetics , Epilepsy/physiopathology , Epilepsy/therapy , Epilepsy, Temporal Lobe/therapy , Glial Cell Line-Derived Neurotrophic Factor/genetics , Hippocampus/metabolism , Male , Neurons/metabolism , Rats , Rats, Wistar , Seizures/genetics
11.
Methods Mol Biol ; 1780: 525-547, 2018.
Article in English | MEDLINE | ID: mdl-29856034

ABSTRACT

Huntington's disease (HD) is characterized by a significant loss of striatal neurons that project to the globus pallidus and substantia nigra, together with loss of cortical projection neurons in varying regions. Mutant huntingtin is suggested to drive the pathogenesis partially by downregulating corticostriatal brain-derived neurotrophic factor (BDNF) levels and signaling. Neurotrophic factors are endogenous peptides that promote the survival and maintenance of neurons. BDNF and other neurotrophic factors have shown neuroprotective benefits in various animal models of neurodegeneration, and are interesting candidates to protect the cell populations that are destined to die in HD. In an attempt to enhance the delivery of neurotrophic factors, several methods have been established to deliver long-term neurotrophic factor gene therapy to human target tissues. This chapter discusses two alternative approaches that have been shown to have potential to deliver neurotrophic factors as a neuroprotective gene therapy for HD. The methods are (1) ex vivo approach where encapsulated cells engineered to express neurotrophic factor are inserted into brain parenchyma or ventricle, and (2) in vivo viral vector therapy, in which viral vector is injected into desired brain area to express gene of interest in the host cells.


Subject(s)
Corpus Striatum/pathology , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Huntington Disease/therapy , Nerve Growth Factors/genetics , Animals , Cell Engineering/methods , Corpus Striatum/cytology , Corpus Striatum/metabolism , Disease Models, Animal , Drug Delivery Systems/methods , Genetic Therapy/instrumentation , Genetic Therapy/trends , Genetic Vectors/genetics , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/pathology , Macaca fascicularis , Mice , Nerve Growth Factors/administration & dosage , Neurons/metabolism , Neurons/pathology , Rats , Stereotaxic Techniques/instrumentation
12.
Mol Ther Methods Clin Dev ; 9: 211-224, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29766029

ABSTRACT

Brain-derived neurotrophic factor (BDNF) may represent a therapeutic for chronic epilepsy, but evaluating its potential is complicated by difficulties in its delivery to the brain. Here, we describe the effects on epileptic seizures of encapsulated cell biodelivery (ECB) devices filled with genetically modified human cells engineered to release BDNF. These devices, implanted into the hippocampus of pilocarpine-treated rats, highly decreased the frequency of spontaneous seizures by more than 80%. These benefits were associated with improved cognitive performance, as epileptic rats treated with BDNF performed significantly better on a novel object recognition test. Importantly, long-term BDNF delivery did not alter normal behaviors such as general activity or sleep/wake patterns. Detailed immunohistochemical analyses revealed that the neurological benefits of BDNF were associated with several anatomical changes, including reduction in degenerating cells and normalization of hippocampal volume, neuronal counts (including parvalbumin-positive interneurons), and neurogenesis. In conclusion, the present data suggest that BDNF, when continuously released in the epileptic hippocampus, reduces the frequency of generalized seizures, improves cognitive performance, and reverts many histological alterations associated with chronic epilepsy. Thus, ECB device-mediated long-term supplementation of BDNF in the epileptic tissue may represent a valid therapeutic strategy against epilepsy and some of its co-morbidities.

13.
Trends Biotechnol ; 36(4): 445-456, 2018 04.
Article in English | MEDLINE | ID: mdl-29455936

ABSTRACT

Encapsulating, or immunoisolating, insulin-secreting cells within implantable, semipermeable membranes is an emerging treatment for type 1 diabetes. This approach can eliminate the need for immunosuppressive drug treatments to prevent transplant rejection and overcome the shortage of donor tissues by utilizing cells derived from allogeneic or xenogeneic sources. Encapsulation device designs are being optimized alongside the development of clinically viable, replenishable, insulin-producing stem cells, for the first time creating the possibility of widespread therapeutic use of this technology. Here, we highlight the status of the most advanced and widely explored implementations of cell encapsulation with an eye toward translating the potential of this technological approach to medical reality.


Subject(s)
Bioartificial Organs , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Pancreas, Artificial , Tissue Engineering , Animals , Clinical Trials as Topic , Humans , Insulin-Secreting Cells/chemistry , Insulin-Secreting Cells/transplantation , Materials Testing , Membranes, Artificial , Models, Animal
14.
J Comp Neurol ; 525(3): 553-573, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27490949

ABSTRACT

Central cholinergic structures within the brain of the even-toed hoofed Goettingen miniature domestic pig (Sus scrofa domesticus) were evaluated by immunohistochemical visualization of choline acetyltransferase (ChAT) and the low-affinity neurotrophin receptor, p75NTR . ChAT-immunoreactive (-ir) perikarya were seen in the olfactory tubercle, striatum, medial septal nucleus, vertical and horizontal limbs of the diagonal band of Broca, and the nucleus basalis of Meynert, medial habenular nucleus, zona incerta, neurosecretory arcuate nucleus, cranial motor nuclei III and IV, Edinger-Westphal nucleus, parabigeminal nucleus, pedunculopontine nucleus, and laterodorsal tegmental nucleus. Cholinergic ChAT-ir neurons were also found within transitional cortical areas (insular, cingulate, and piriform cortices) and hippocampus proper. ChAT-ir fibers were seen throughout the dentate gyrus and hippocampus, in the mediodorsal, laterodorsal, anteroventral, and parateanial thalamic nuclei, the fasciculus retroflexus of Meynert, basolateral and basomedial amygdaloid nuclei, anterior pretectal and interpeduncular nuclei, as well as select laminae of the superior colliculus. Double immunofluorescence demonstrated that virtually all ChAT-ir basal forebrain neurons were also p75NTR -positive. The present findings indicate that the central cholinergic system in the miniature pig is similar to other mammalian species. Therefore, the miniature pig may be an appropriate animal model for preclinical studies of neurodegenerative diseases where the cholinergic system is compromised. J. Comp. Neurol. 525:553-573, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain/metabolism , Choline O-Acetyltransferase/metabolism , Receptors, Nerve Growth Factor/metabolism , Swine, Miniature/metabolism , Animals , Brain/anatomy & histology , Female , Immunohistochemistry , Models, Animal , Swine , Swine, Miniature/anatomy & histology
15.
Trends Pharmacol Sci ; 36(8): 537-46, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26067102

ABSTRACT

Treating many chronic diseases will require a tight, minute-to-minute regulation of therapeutic molecules that is currently not achievable with most pharmaceutical therapies. For these diseases, implantable living cellular systems may be able to provide unlimited drug delivery, enabling seamless matching of treatment duration with disease longevity. Cell encapsulation is an advanced technology that achieves this goal and represents a viable therapeutic option. The advanced state of the field has allowed researchers to inch forward into therapeutic domains previously untouchable because of the myriad disparate fields that intersect biomaterials and cells. Here, we discuss the next generation of clinical trials and potential approaches, 'smart' and responsive encapsulation systems, sophisticated and multifunctional devices, and novel imaging tools, together with the future challenges in the field.


Subject(s)
Capsules/chemistry , Cell Transplantation/methods , Alginates/chemistry , Allografts/transplantation , Animals , Capsules/adverse effects , Heterografts/transplantation , Humans
17.
Adv Drug Deliv Rev ; 67-68: 131-41, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23880505

ABSTRACT

Delivering therapeutic molecules, including trophic factor proteins, across the blood brain barrier to the brain parenchyma to treat chronic neurodegenerative diseases remains one of the great challenges in biology. To be effective, delivery needs to occur in a long-term and stable manner at sufficient quantities directly to the target region in a manner that is selective but yet covers enough of the target site to be efficacious. One promising approach uses cellular implants that produce and deliver therapeutic molecules directly to the brain region of interest. Implanted cells can be precisely positioned into the desired region and can be protected from host immunological attack by encapsulating them and by surrounding them within an immunoisolatory, semipermeable capsule. In this approach, cells are enclosed within a semiporous capsule with a perm selective membrane barrier that admits oxygen and required nutrients and releases bioactive cell secretions while restricting passage of larger cytotoxic agents from the host immune defense system. Recent advances in human cell line development have increased the levels of secreted therapeutic molecules from encapsulated cells, and membrane extrusion techniques have led to the first ever clinical demonstrations of long-term survival and function of encapsulated cells in the brain parenchyma. As such, cell encapsulation is capable of providing a targeted, continuous, de novo synthesized source of very high levels of therapeutic molecules that can be distributed over significant portions of the brain.


Subject(s)
Cell Transplantation/methods , Neurodegenerative Diseases/therapy , Alzheimer Disease/therapy , Animals , Cell Transplantation/instrumentation , Humans , Huntington Disease/therapy , Nerve Growth Factor/metabolism , Parkinson Disease/therapy , Tissue Scaffolds
18.
PLoS One ; 8(10): e78490, 2013.
Article in English | MEDLINE | ID: mdl-24205243

ABSTRACT

BACKGROUND: This study aims to create a convenient reference for both clinicians and researchers so that vis-à-vis comparisons between brain disorders can be made quickly and accurately. We report here the incidence and prevalence of the major adult-onset brain disorders in the United States using a meta-analysis approach. MATERIAL AND METHODS: Epidemiological figures were collected from the most recent, reliable data available in the research literature. Population statistics were based on the most recent census from the US Census Bureau. Extrapolations were made only when necessary. The most current epidemiological studies for each disorder were chosen. All effort was made to use studies based on national cohorts. Studies reviewed were conducted between 1950 and 2009. The data of the leading studies for several neurological studies was compiled in order to obtain the most accurate extrapolations. Results were compared to commonly accepted values in order to evaluate validity. RESULTS: It was found that 6.75% of the American adult population is afflicted with brain disorders. This number was eclipsed by the 8.02% of Floridians with brain disorders, which is due to the large aged population residing in the state. CONCLUSIONS: There was a noticeable lack of epidemiological data concerning adult-onset brain disorders. Since approximately 1 out of every 7 households is affected by brain disorders, increased research into this arena is warranted.


Subject(s)
Nervous System Diseases/epidemiology , Brain Diseases/epidemiology , Data Collection , Humans , Incidence , Prevalence , United States
19.
Restor Neurol Neurosci ; 30(3): 225-36, 2012.
Article in English | MEDLINE | ID: mdl-22426041

ABSTRACT

PURPOSE: Encapsulated cell (EC) biodelivery is a promising, clinically relevant technology platform to safely target the delivery of therapeutic proteins to the central nervous system. The purpose of this study was to evaluate EC biodelivery of the novel neurotrophic factor, Meteorin, to the striatum of rats and to investigate its neuroprotective effects against quinolinic acid (QA)-induced excitotoxicity. METHODS: Meteorin-producing ARPE-19 cells were loaded into EC biodelivery devices and implanted into the striatum of rats. Two weeks after implantation, QA was injected into the ipsilateral striatum followed by assessment of neurological performance two and four weeks after QA administration. RESULTS: Implant-delivered Meteorin effectively protected against QA-induced toxicity, as manifested by both near-normal neurological performance and reduction of brain cell death. Morphological analysis of the Meteorin-treated brains showed a markedly reduced striatal lesion size. The EC biodelivery devices produced stable or even increasing levels of Meteorin throughout the study over 6 weeks. CONCLUSIONS: Stereotactically implanted EC biodelivery devices releasing Meteorin could offer a feasible strategy in the treatment of neurological diseases with an excitotoxic component such as Huntington's disease. In a broader sense, the EC biodelivery technology is a promising therapeutic protein delivery platform for the treatment of a wide range of diseases of the central nervous system.


Subject(s)
Absorbable Implants/standards , Cytoprotection/drug effects , Disease Models, Animal , Huntington Disease/drug therapy , Nerve Tissue Proteins/administration & dosage , Quinolines/toxicity , Animals , Brain Tissue Transplantation/methods , Capsules/administration & dosage , Cell Line , Cytoprotection/physiology , Humans , Huntington Disease/chemically induced , Huntington Disease/genetics , Male , Mice , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Rats , Rats, Sprague-Dawley
20.
J Control Release ; 154(3): 249-57, 2011 Sep 25.
Article in English | MEDLINE | ID: mdl-21704093

ABSTRACT

We previously engineered a macroporous, polymer-based vaccine that initially produces GM-CSF gradients to recruit local dendritic cells and subsequently presents CpG oligonucleotides, and tumor lysate to cell infiltrates to induce immune cell activation and immunity against tumor cells in peripheral tumor models. Here, we demonstrate that this system eradicates established intracranial glioma following implantation into brain tissue, whereas implantation in resection cavities obviates vaccine efficacy. Rats bearing seven-day old, intracranial glioma tumors were treated with PLG vaccines implanted into the tumor bed, resulting in retention of contralateral forelimb function (day 17) that is compromised by tumor formation in control animals, and 90% long-term survival (>100 days). Similar benefits were observed in animals receiving tumor resection plus vaccine implants into the adjacent parenchyma, but direct implantation of PLG vaccines into the resection cavity conferred no benefit. This dissociation of efficacy was likely related to GM-CSF distribution, as implantation of PLG vaccines within brain tissue produced significant GM-CSF gradients for prolonged periods, which was not detected after implantation in resection cavities. These studies demonstrate that PLG vaccine efficacy is correlated to GM-CSF gradient formation, which requires direct implantation into brain tissue, and justify further exploration of this approach for glioma treatment.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Glioma/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Polyglactin 910/chemistry , Adjuvants, Immunologic/therapeutic use , Animals , Brain/drug effects , Brain/pathology , Brain Neoplasms/pathology , Cancer Vaccines/therapeutic use , Glioma/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunotherapy , Male , Oligodeoxyribonucleotides/therapeutic use , Porosity , Prostheses and Implants , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...