Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Alzheimers Dement (N Y) ; 5: 627-636, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31660425

RESUMO

INTRODUCTION: Low blood-brain barrier (BBB) penetration and hematopoietic side effects limit the therapeutic development of erythropoietin (EPO) for Alzheimer's disease (AD). A fusion protein of EPO and a chimeric monoclonal antibody targeting the mouse transferrin receptor (cTfRMAb) has been engineered. The latter drives EPO into the brain via receptor-mediated transcytosis across the BBB and increases its peripheral clearance to reduce hematopoietic side effects of EPO. Our previous work shows the protective effects of this BBB-penetrating EPO in AD mice but hematologic effects have not been studied. Herein, we investigate the hematologic safety and therapeutic effects of chronic cTfRMAb-EPO dosing, in comparison to recombinant human EPO (rhu-EPO), in AD mice. METHODS: Male APPswe PSEN1dE9 (APP/PS1) mice (9.5 months) were treated with saline (n = 11), and equimolar doses of cTfRMAb-EPO (3 mg/kg, n = 7), or rhu-EPO (0.6 mg/kg, n = 9) 2 days/week subcutaneously for 6 weeks, compared to saline-treated wild-type mice (n = 10). At 6 weeks, exploration and memory were assessed, and mice were sacrificed at 8 weeks. Spleens were weighed, and brains were evaluated for amyloid beta (Aß) load and synaptophysin. Blood was collected at 4, 6 and 8 weeks for a complete blood count and white blood cells differential. RESULTS: cTfRMAb-EPO transiently increased reticulocyte counts after 4 weeks, followed by normalization of reticulocytes at 6 and 8 weeks. rhu-EPO transiently increased red blood cell count, hemoglobin and hematocrit, and significantly decreased mean corpuscular volume and reticulocytes at 4 weeks, which remained low at 6 weeks. At 8 weeks, a significant decline in red blood cell indices was observed with rhu-EPO treatment. Exploration and cognitive deficits were significantly worse in APP/PS1-rhu-EPO mice. Both cTfRMAb-EPO and rhu-EPO decreased 6E10-positive brain Aß load; however, cTfRMAb-EPO and not rhu-EPO selectively reduced brain Aß1-42 and elevated synaptophysin expression. DISCUSSION: Chronic treatment with cTfRMAb-EPO results in better hematologic safety, behavioral, and therapeutic indices compared with rhu-EPO, supporting the development of this BBB-penetrable EPO analog for AD.

2.
Mol Pharm ; 14(7): 2340-2349, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28514851

RESUMO

Tumor necrosis factor alpha (TNF-α) driven processes are involved at multiple stages of Alzheimer's disease (AD) pathophysiology and disease progression. Biologic TNF-α inhibitors (TNFIs) are the most potent class of TNFIs but cannot be developed for AD since these macromolecules do not cross the blood-brain barrier (BBB). A BBB-penetrating TNFI was engineered by the fusion of the extracellular domain of the type II human TNF receptor (TNFR) to a chimeric monoclonal antibody (mAb) against the mouse transferrin receptor (TfR), designated as the cTfRMAb-TNFR fusion protein. The cTfRMAb domain functions as a molecular Trojan horse, binding to the mouse TfR and ferrying the biologic TNFI across the BBB via receptor-mediated transcytosis. The aim of the study was to examine the effect of this BBB-penetrating biologic TNFI in a mouse model of AD. Six-month-old APPswe, PSEN 1dE9 (APP/PS1) transgenic mice were treated with saline (n = 13), the cTfRMAb-TNFR fusion protein (n = 12), or etanercept (non-BBB-penetrating biologic TNFI; n = 11) 3 days per week intraperitoneally. After 12 weeks of treatment, recognition memory was assessed using the novel object recognition task, mice were sacrificed, and brains were assessed for amyloid beta (Aß) load, neuroinflammation, BBB damage, and cerebral microhemorrhages. The cTfRMAb-TNFR fusion protein caused a significant reduction in brain Aß burden (both Aß peptide and plaque), neuroinflammatory marker ICAM-1, and a BBB disruption marker, parenchymal IgG, and improved recognition memory in the APP/PS1 mice. Fusion protein treatment resulted in low antidrug-antibody formation with no signs of either immune reaction or cerebral microhemorrhage development with chronic 12-week treatment. Chronic treatment with the cTfRMAb-TNFR fusion protein, a BBB-penetrating biologic TNFI, offers therapeutic benefits by targeting Aß pathology, neuroinflammation, and BBB-disruption, overall improving recognition memory in a transgenic mouse model of AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Anticorpos Monoclonais/uso terapêutico , Barreira Hematoencefálica/metabolismo , Receptores da Transferrina/antagonistas & inibidores , Proteínas Recombinantes de Fusão/uso terapêutico , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Crioultramicrotomia , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência
3.
Mol Pharm ; 10(4): 1425-31, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23410508

RESUMO

Tumor necrosis factor (TNF)-α is a proinflammatory cytokine active in the brain. Etanercept, the TNF decoy receptor (TNFR), does not cross the blood-brain barrier (BBB). The TNFR was re-engineered for BBB penetration as a fusion protein with a chimeric monoclonal antibody (mAb) against the mouse transferrin receptor (TfR), and this fusion protein is designated cTfRMAb-TNFR. The cTfRMAb domain of the fusion protein acts as a molecular Trojan horse and mediates transport via the endogenous BBB TfR. To support future chronic treatment of mouse models of neural disease with daily administration of the cTfRMAb-TNFR fusion protein, a series of pharmacokinetics and brain uptake studies in the mouse was performed. The cTfRMAb-TNFR fusion protein was radiolabeled and injected into mice via the intravenous, intraperitoneal (IP), or subcutaneous (SQ) routes of administration at doses ranging from 0.35 to 10 mg/kg. The distribution of the fusion protein into plasma following the IP or SQ routes was enhanced by increasing the injection dose from 3 to 10 mg/kg. The fusion protein demonstrated long circulation times with high metabolic stability following the IP or SQ routes of injection. The IP or SQ routes produced concentrations of the cTfRMAb-TNFR fusion protein in the brain that exceed by 20- to 50-fold the concentration of TNFα in pathologic conditions of the brain. The SQ injection is the preferred route of administration, as the level of cTfRMAb fusion protein produced in the brain is comparable to that generated with intravenous injection, and at a much lower plasma area under the concentration curve of the fusion protein as compared to IP administration.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Imunoglobulina G/administração & dosagem , Imunoglobulina G/química , Receptores do Fator de Necrose Tumoral/administração & dosagem , Receptores do Fator de Necrose Tumoral/química , Animais , Anticorpos Monoclonais/química , Área Sob a Curva , Barreira Hematoencefálica , Desenho de Fármacos , Etanercepte , Inflamação , Infusões Intravenosas , Infusões Parenterais , Infusões Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores da Transferrina/química , Proteínas Recombinantes de Fusão/metabolismo
4.
Brain Res ; 1507: 91-6, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23428543

RESUMO

Stroke therapy may be optimized by combination therapy with both a neuroprotective neurotrophin and an anti-inflammatory agent. In the present work, the model neurotrophin is glial cell line-derived neurotrophic factor (GDNF), and the model anti-inflammatory agent is the type II tumor necrosis factor receptor (TNFR) decoy receptor. Both the GDNF and the TNFR are large molecules that do not cross the blood-brain barrier (BBB), which is intact in the early hours after stroke when neural rescue is still possible. The GDNF and the TNFR decoy receptor were re-engineered for BBB transport as IgG fusion proteins, wherein the GDNF or the TNFR are fused to the heavy chain of a chimeric monoclonal antibody (MAb) against the mouse transferrin receptor (TfR), and these fusion proteins are designated cTfRMAb-GDNF and cTfRMAb-TNFR, respectively. Mice were treated intravenously with (a) saline, (b) GDNF alone, (c) the cTfRMAb-GDNF fusion protein alone, or (d) the combined cTfRMAb-GDNF and cTfRMAb-TNFR fusion proteins, following a 1-h reversible middle cerebral artery occlusion (MCAO). The cTfRMAb-GDNF fusion protein alone caused a significant 25% and 30% reduction in hemispheric and cortical stroke volumes. Combined treatment with the cTfRMAb-GDNF and cTfRMAb-TNFR fusion proteins caused a significant 54%, 69% and 30% reduction in hemispheric, cortical and subcortical stroke volumes. Conversely, intravenous GDNF had no therapeutic effect. In conclusion, combination treatment with BBB penetrating IgG-GDNF and IgG-TNFR fusion proteins enhances the therapeutic effect of single treatment with the IgG-GDNF fusion protein following delayed intravenous administration in acute stroke.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/uso terapêutico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Barreira Hematoencefálica , Quimioterapia Combinada , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Imunoglobulina G/genética , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Acidente Vascular Cerebral/patologia
5.
J Cereb Blood Flow Metab ; 33(1): 13-21, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23072749

RESUMO

The brain is in many ways an immunologically and pharmacologically privileged site. The blood-brain barrier (BBB) of the cerebrovascular endothelium and its participation in the complex structure of the neurovascular unit (NVU) restrict access of immune cells and immune mediators to the central nervous system (CNS). In pathologic conditions, very well-organized immunologic responses can develop within the CNS, raising important questions about the real nature and the intrinsic and extrinsic regulation of this immune privilege. We assess the interactions of immune cells and immune mediators with the BBB and NVU in neurologic disease, cerebrovascular disease, and intracerebral tumors. The goals of this review are to outline key scientific advances and the status of the science central to both the neuroinflammation and CNS barriers fields, and highlight the opportunities and priorities in advancing brain barriers research in the context of the larger immunology and neuroscience disciplines. This review article was developed from reports presented at the 2011 Annual Blood-Brain Barrier Consortium Meeting.


Assuntos
Barreira Hematoencefálica/imunologia , Doenças do Sistema Nervoso Central/imunologia , Inflamação Neurogênica/imunologia , Animais , Endotélio Vascular/imunologia , Humanos , Neuroimagem , Neuroimunomodulação
6.
J Cereb Blood Flow Metab ; 32(10): 1933-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22714051

RESUMO

Tumor necrosis factor (TNF)-α is produced in brain in response to acute cerebral ischemia, and promotes neuronal apoptosis. Biologic TNF inhibitors (TNFIs), such as the etanercept, cannot be developed as new stroke treatments because these large molecule drugs do not cross the blood-brain barrier (BBB). A BBB-penetrating biologic TNFI was engineered by fusion of the type II human TNF receptor (TNFR) to each heavy chain of a genetically engineered chimeric monoclonal antibody (MAb) against the mouse transferrin receptor (TfR), designated as cTfRMAb-TNFR fusion protein. The cTfRMAb domain of the fusion protein acts as a molecular Trojan horse to deliver the fused TNFR across the BBB. Etanercept or the cTfRMAb-TNFR fusion protein (1 mg/kg) was administered intravenously in adult mice subjected to 1-hour reversible middle cerebral artery occlusion up to 90 minutes after the occlusion. Neuroprotection was assessed at 24 hours or 7 days after occlusion. The cTfRMAb-TNFR fusion protein treatment caused a significant 45%, 48%, 42%, and 54% reduction in hemispheric, cortical, and subcortical stroke volumes, and neural deficit, respectively. Intravenous etanercept had no therapeutic effect. Biologic TNFIs can be reengineered for BBB penetration, and the IgG-TNFR fusion protein is therapeutic after delayed intravenous administration in experimental stroke.


Assuntos
Encéfalo/patologia , Cadeias gama de Imunoglobulina/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Receptores do Fator de Necrose Tumoral/uso terapêutico , Acidente Vascular Cerebral/prevenção & controle , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Administração Intravenosa , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Encéfalo/efeitos dos fármacos , Células CHO , Cricetinae , Humanos , Cadeias gama de Imunoglobulina/administração & dosagem , Cadeias gama de Imunoglobulina/imunologia , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/prevenção & controle , Camundongos , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/imunologia , Receptores da Transferrina/imunologia , Receptores do Fator de Necrose Tumoral/administração & dosagem , Receptores do Fator de Necrose Tumoral/imunologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Acidente Vascular Cerebral/patologia
7.
J Pharmacol Exp Ther ; 339(2): 618-23, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21831964

RESUMO

Biologic tumor necrosis factor (TNF)-α inhibitors do not cross the blood-brain barrier (BBB). A BBB-penetrating TNF-α inhibitor was engineered by fusion of the extracellular domain of the type II human TNF receptor (TNFR) to the carboxyl terminus of the heavy chain of a mouse/rat chimeric monoclonal antibody (MAb) against the mouse transferrin receptor (TfR), and this fusion protein is designated cTfRMAb-TNFR. The cTfRMAb-TNFR fusion protein and etanercept bound human TNF-α with high affinity and K(D) values of 374 ± 77 and 280 ± 80 pM, respectively. Neuroprotection in brain in vivo after intravenous administration of the fusion protein was examined in a mouse model of Parkinson's disease. Mice were also treated with saline or a non-BBB-penetrating TNF decoy receptor, etanercept. After intracerebral injection of the nigral-striatal toxin, 6-hydroxydopamine, mice were treated every other day for 3 weeks. Treatment with the cTfRMAb-TNFR fusion protein caused an 83% decrease in apomorphine-induced rotation, a 67% decrease in amphetamine-induced rotation, a 82% increase in vibrissae-elicited forelimb placing, and a 130% increase in striatal tyrosine hydroxylase (TH) enzyme activity. In contrast, chronic treatment with etanercept, which does not cross the BBB, had no effect on neurobehavior or striatal TH enzyme activity. A bridging enzyme-linked immunosorbent assay specific for the cTfRMAb-TNFR fusion protein showed that the immune response generated in the mice was low titer. In conclusion, a biologic TNF inhibitor is neuroprotective after intravenous administration in a mouse model of neurodegeneration, providing that the TNF decoy receptor is reengineered to cross the BBB.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Barreira Hematoencefálica/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Transtornos Parkinsonianos/tratamento farmacológico , Receptores do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Comportamento Animal/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/metabolismo , Células CHO , Grupos Controle , Corpo Estriado/efeitos dos fármacos , Cricetinae , Etanercepte , Humanos , Imunoglobulina G/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacocinética , Transtornos Parkinsonianos/imunologia , Ratos , Receptores da Transferrina/imunologia , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico , Substância Negra/efeitos dos fármacos , Receptores Chamariz do Fator de Necrose Tumoral/farmacocinética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Tirosina 3-Mono-Oxigenase/biossíntese
8.
Bioconjug Chem ; 22(8): 1611-8, 2011 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-21707084

RESUMO

The genetic engineering, host cell expression, purity, identity, and in vivo brain drug targeting properties are described for a new IgG-fusion protein, designated the cTfRMAb-AV fusion protein. Avidin (AV) is fused to the carboxyl terminus of the heavy chain of the genetically engineered chimeric monoclonal antibody (mAb) against the mouse transferrin receptor (TfR). The TfRMAb binds the endogenous TfR on the blood-brain barrier (BBB), which triggers transport into brain from blood. The cTfRMAb-AV fusion protein is produced in stably transfected Chinese hamster ovary cells, which are grown in serum free medium under conditions of biotin starvation. Following affinity purification, the purity and identity of the cTfRMAb-AV fusion protein were verified by electrophoresis and Western blotting. The affinity of the cTfRMAb for the murine TfR is high, K(I) = 4.6 ± 0.5 nM, despite fusion of avidin to the antibody heavy chain. The model peptide radiopharmaceutical used in this study is the Aß(1-40) amyloid peptide of Alzheimer's disease (AD), which in a brain-penetrating form could be used to image the amyloid plaque in brain in AD. The BBB transport and brain uptake of the [(125)I]-Aß(1-40) peptide was measured in mice injected intravenously (IV) with the peptide either free or conjugated to the cTfRMAb-AV fusion protein. The brain uptake of the free Aß(1-40) peptide was very low, 0.1% of injected dose (ID)/gram brain following i.v. injection, and is comparable to the brain uptake of a brain blood volume marker. However, the brain uptake of the Aß(1-40) peptide was high, 2.1 ± 0.2% ID/gram brain, following attachment of the biotinylated peptide to the cTfRMAb-AV fusion protein. Capillary depletion analysis showed the peptide penetrated the brain parenchyma from blood. The cTfRMAb-AV fusion protein is a new drug delivery system that can target to mouse brain monobiotinylated peptide or antisense radiopharmaceuticals.


Assuntos
Encéfalo/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Peptídeos/administração & dosagem , Compostos Radiofarmacêuticos/administração & dosagem , Proteínas Recombinantes de Fusão/uso terapêutico , Peptídeos beta-Amiloides/farmacocinética , Animais , Anticorpos Monoclonais , Avidina , Barreira Hematoencefálica/química , Barreira Hematoencefálica/metabolismo , Imunoglobulina G , Camundongos , Compostos Radiofarmacêuticos/farmacocinética , Receptores da Transferrina/imunologia
9.
Drug Metab Dispos ; 39(1): 71-6, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20884844

RESUMO

Biologic tumor necrosis factor inhibitors (TNFIs) include TNF decoy receptors (TNFRs). TNFα plays a pathologic role in both acute and chronic brain disease. However, biologic TNFIs cannot be developed as brain therapeutics because these large molecule drugs do not cross the blood-brain barrier (BBB). To enable penetration of the brain via receptor-mediated transport, the human TNFR type II was re-engineered as an IgG fusion protein, where the IgG part is a chimeric monoclonal antibody (MAb) against the mouse transferrin receptor (TfR), and this fusion protein is designated cTfRMAb-TNFR. The cTfRMAb part of the fusion protein acts as a molecular Trojan horse to ferry the TNFR across the BBB via transport on the endogenous BBB TfR. cTfRMAb-TNFR was expressed by stably transfected Chinese hamster ovary cells and purified by affinity chromatography to homogeneity on electrophoretic gels. The fusion protein reacted with antibodies to both mouse IgG and the human TNFR and bound TNFα with high affinity (K(d) = 96 ± 34 pM). cTfRMAb-TNFR was rapidly transported into mouse brain in vivo after intravenous administration, and the brain uptake of the fusion protein was 2.8 ± 0.5% of injected dose per gram of brain, which is >45-fold higher than the brain uptake of an IgG that does not recognize the mouse TfR. This new IgG-TNFR fusion protein can be tested in mouse models of brain diseases in which TNFα plays a pathologic role.


Assuntos
Barreira Hematoencefálica/metabolismo , Receptores da Transferrina/metabolismo , Receptores Chamariz do Fator de Necrose Tumoral/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Encéfalo/metabolismo , Encefalopatias/metabolismo , Células CHO , Cricetinae , Cricetulus , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores da Transferrina/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
10.
J Biotechnol ; 146(1-2): 84-91, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20100527

RESUMO

Decoy receptors, such as the human tumor necrosis factor receptor (TNFR), are potential new therapies for brain disorders. However, decoy receptors are large molecule drugs that are not transported across the blood-brain barrier (BBB). To enable BBB transport of a TNFR decoy receptor, the human TNFR-II extracellular domain was re-engineered as a fusion protein with a chimeric monoclonal antibody (MAb) against the human insulin receptor (HIR). The HIRMAb acts as a molecular Trojan horse to ferry the TNFR therapeutic decoy receptor across the BBB. The HIRMAb-TNFR fusion protein was expressed in stably transfected CHO cells, and was analyzed with electrophoresis, Western blotting, size exclusion chromatography, and binding assays for the HIR and TNFalpha. The HIRMAb-TNFR fusion protein was radio-labeled by trititation, in parallel with the radio-iodination of recombinant TNFR:Fc fusion protein, and the proteins were co-injected in the adult Rhesus monkey. The TNFR:Fc fusion protein did not cross the primate BBB in vivo, but the uptake of the HIRMAb-TNFR fusion protein was high and 3% of the injected dose was taken up by the primate brain. The TNFR was selectively targeted to brain, relative to peripheral organs, following fusion to the HIRMAb. This study demonstrates that decoy receptors may be re-engineered as IgG fusion proteins with a BBB molecular Trojan horse that selectively targets the brain, and enables penetration of the BBB in vivo. IgG-decoy receptor fusion proteins represent a new class of human neurotherapeutics.


Assuntos
Anticorpos Monoclonais/metabolismo , Antígenos CD/imunologia , Encéfalo/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Receptor de Insulina/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Encéfalo/efeitos dos fármacos , Química Encefálica , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Feminino , Vetores Genéticos , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Cadeias gama de Imunoglobulina/imunologia , Cadeias gama de Imunoglobulina/metabolismo , Macaca mulatta , Camundongos , Engenharia de Proteínas/métodos , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacocinética
11.
Mol Pharm ; 6(5): 1536-43, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19624167

RESUMO

The tumor necrosis factor-alpha receptor (TNFR) extracellular domain (ECD) is a decoy receptor that could be developed as a neurotherapeutic for stroke, brain injury, or chronic neurodegeneration. However, the TNFR ECD is a large molecule therapeutic that does not cross the blood-brain barrier (BBB). Human TNFR ECD was re-engineered by fusion of the receptor protein to the carboxyl terminus of the chimeric monoclonal antibody (mAb) to the human insulin receptor (HIR). The HIRMAb-TNFR fusion protein is bifunctional, and binds both the HIR, to trigger receptor-mediated transport across the BBB, and TNFalpha, to sequester this cytotoxic cytokine. COS cells were dual transfected with the heavy chain (HC) and light chain fusion protein expression plasmids, and the HC of the fusion protein was immunoreactive with antibodies to both human IgG and TNFR. The HIRMAb-TNFR fusion protein bound to the extracellular domain of the HIR with an affinity comparable to the HIRMAb, and bound TNFalpha with a K(D) of 0.34 +/- 0.17 nM. Both the TNFR:Fc fusion protein and the HIRMAb-TNFR fusion protein blocked the cytotoxic actions of TNFalpha on human cells in a bioassay. In conclusion, these studies describe the re-engineering of the TNFR ECD to make this decoy receptor transportable across the human BBB.


Assuntos
Barreira Hematoencefálica , Sistemas de Liberação de Medicamentos , Cadeias gama de Imunoglobulina/química , Receptores do Fator de Necrose Tumoral/química , Animais , Sequência de Bases , Bioensaio , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiologia , Encefalopatias/terapia , Células COS , Linhagem Celular , Chlorocebus aethiops , Primers do DNA/genética , Humanos , Cadeias gama de Imunoglobulina/genética , Cadeias gama de Imunoglobulina/metabolismo , Cadeias gama de Imunoglobulina/uso terapêutico , Técnicas In Vitro , Cinética , Estrutura Terciária de Proteína , Ensaio Radioligante , Receptor de Insulina/metabolismo , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores do Fator de Necrose Tumoral/uso terapêutico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Transfecção , Fator de Necrose Tumoral alfa/metabolismo
12.
Mol Pharm ; 6(3): 747-51, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19093871

RESUMO

Delivery of short interfering RNA (siRNA) to cells in culture, and in vivo, is possible with combined use of a receptor-specific monoclonal antibody (mAb) and avidin-biotin technology. In the present studies, the luciferase gene is transiently expressed in human 293 epithelial cells. The siRNA delivery system is composed of the siRNA, monobiotinylated on the 3'-terminus of the sense strand, and a conjugate of streptavidin (SA) and a mAb to the human insulin receptor (HIR). Exposure of cells to 3'-biotinyl-siRNA bound to the HIRMAb/SA conjugate, but not to unconjugated SA, avidin, or the HIRMAb, causes a >90% reduction in luciferase gene expression. The receptor-targeted siRNA effect is maximal at 48 h after delivery of the siRNA to the cells, and the effect is lost by 7 days after a single application of the targeted siRNA in culture. The KI of the receptor-targeted siRNA inhibition of gene expression is 30.5 +/- 11.7 nM, and significant inhibition is observed with siRNA concentrations as low as 3 nM. In conclusion, the combination of a receptor-specific targeting ligand, such as the HIRMAb, and avidin-biotin technology allows for high affinity capture of the monobiotinylated siRNA by the targeting mAb. The siRNA is effectively delivered to the cytosol of cells, and knockdown of gene expression with the HIRMAb/SA delivery system is comparable to RNA interference effects obtained with cationic polyplexes. Whereas the use of cationic polyplexes in vivo is problematic, the bond between the targeting mAb and the siRNA is stable with avidin-biotin technology, and RNAi effects at distant sites such as brain are observed in vivo following an intravenous administration of the targeted siRNA.


Assuntos
Anticorpos Monoclonais/química , Antígenos CD/imunologia , Avidina/química , Biotina/química , RNA Interferente Pequeno/química , RNA Interferente Pequeno/fisiologia , Receptor de Insulina/imunologia , Animais , Anticorpos Monoclonais/imunologia , Linhagem Celular , Humanos , Luciferases/genética , Camundongos , Interferência de RNA/fisiologia , Estreptavidina/química , Fatores de Tempo
13.
Bioconjug Chem ; 19(3): 731-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18278853

RESUMO

The genetic engineering, expression, and validation of a fusion protein of avidin (AV) and a chimeric monoclonal antibody (mAb) to the human insulin receptor (HIR) is described. The 15 kDa avidin monomer was fused to the carboxyl terminus of the heavy chain of the HIRMAb. The fusion protein heavy chain reacted with antibodies specific for human IgG and avidin, and had the same affinity for binding to the HIR extracellular domain as the original chimeric HIRMAb. The fusion protein qualitatively bound biotinylated ligands, but was secreted fully saturated with biotin by COS cells, owing to the high level of biotin in tissue culture medium. Chinese hamster ovary (CHO) cells were permanently transfected with a tandem vector expressing the fusion protein genes, and high expressing cell lines were isolated by methotrexate amplification and dilutional cloning. The product expressed by CHO cells had high binding to the HIR, and migrated as a homogeneous species in size exclusion HPLC and native polyacrylamide gel electrophoresis. The CHO cells were adapted to a 4 week culture in biotin depleted medium, and the HIRMAb-AV fusion protein expressed under these conditions had 1 unoccupied biotin binding site per molecule, based on a [3H]-biotin ultrafiltration assay. The HIRMAb-AV increased biotin uptake by human cells >15-fold, and mediated the endocytosis of fluorescein-biotin, as demonstrated by confocal microscopy. In summary, the HIRMAb-AV fusion protein is a new drug targeting system for humans that can be adapted to monobiotinylated drugs or nucleic acids.


Assuntos
Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Biotina/química , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/farmacologia , Receptores de Droga/efeitos dos fármacos , Animais , Anticorpos Monoclonais/biossíntese , Avidina/química , Biotina/farmacocinética , Western Blotting , Células CHO , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Cromatografia Líquida de Alta Pressão , Cricetinae , Cricetulus , DNA Complementar/biossíntese , DNA Complementar/genética , Diálise , Sistemas de Liberação de Medicamentos , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Engenharia Genética , Humanos , Microscopia Confocal , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Soroalbumina Bovina/química , Ultrafiltração
14.
J Cereb Blood Flow Metab ; 28(2): 291-301, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17667996

RESUMO

A novel organic anion transporter selectively expressed at the blood-brain barrier (BBB), originally designated BBB-specific anion transporter type 1 (BSAT1), and now classified as Slco1c1, has been cloned from a BBB genomics program as a partial cDNA; this study describes the cloning and expression of the full-length cDNA from a rat brain capillary cDNA library. Northern analysis revealed the selective expression of the transporter at the BBB, and the transporter was expressed after permanent transfection of human 293 cells with cDNA encoding either the full length or open reading frame mRNA. The full-length transporter cDNA was 2.6 kb, and the mRNA was highly expressed at the rat brain microvasculature, but not in kidney, liver, heart, or lung, or in glial cells or brain glial tumors. Blood-brain barrier-specific anion transporter type 1 expression in 293 cells was poor after the transfection of the full-length cDNA, whereas transporter expression in 293 cells was high after transfection of the open reading frame. The transporter showed asymmetric kinetic properties in comparison of the influx and efflux of model substrates, thyroxine (T4), triiodothyronine (T3), and estradiol-glucuronide (E2G). Thyroxine and T3 inhibited the influx of E2G, but E2G did not inhibit thyroxine influx, and T3 only weakly inhibited the influx of T4. Extracellular E2G stimulated the transefflux of intracellular T4. Blood-brain barrier-specific anion transporter type 1 is a novel organic anion transporter that is a sodium-independent exchanger that may participate in the active efflux of iodothyronines and steroid conjugates at the BBB.


Assuntos
Barreira Hematoencefálica/fisiologia , Proteínas de Transporte de Cátions Orgânicos/genética , Regiões 5' não Traduzidas/genética , Actinas/biossíntese , Actinas/genética , Animais , Sequência de Bases , Sítios de Ligação/efeitos dos fármacos , Northern Blotting , Linhagem Celular , Clonagem Molecular , Reagentes de Ligações Cruzadas , DNA Complementar/biossíntese , DNA Complementar/genética , Genômica , Humanos , Hibridização In Situ , Dados de Sequência Molecular , Poli A/biossíntese , Poli A/genética , RNA/biossíntese , RNA/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tiroxina/metabolismo , Transfecção
15.
Pharm Res ; 24(12): 2309-16, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17926121

RESUMO

PURPOSE: The effective delivery of short interfering RNA (siRNA) to brain following intravenous administration requires the development of a delivery system for transport of the siRNA across the brain capillary endothelial wall, which forms the blood-brain barrier in vivo. METHODS: siRNA was delivered to brain in vivo with the combined use of a receptor-specific monoclonal antibody delivery system, and avidin-biotin technology. The siRNA was mono-biotinylated on either terminus of the sense strand, in parallel with the production of a conjugate of the targeting MAb and streptavidin. RESULTS: Rat glial cells (C6 or RG-2) were permanently transfected with the luciferase gene, and implanted in the brain of adult rats. Following the formation of intra-cranial tumors, the rats were treated with a single intravenous injection of 270 microg/kg of biotinylated siRNA attached to a transferrin receptor antibody via a biotin-streptavidin linker. The intravenous administration of the siRNA caused a 69-81% decrease in luciferase gene expression in the intracranial brain cancer in vivo. CONCLUSIONS: Brain delivery of siRNA following intravenous administration is possible with siRNAs that are targeted to brain with the combined use of receptor specific antibody delivery systems and avidin-biotin technology.


Assuntos
Anticorpos Monoclonais/metabolismo , Biotina/metabolismo , Neoplasias Encefálicas/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores da Transferrina/imunologia , Estreptavidina/metabolismo , Animais , Biotinilação , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Genes Reporter , Humanos , Luciferases/genética , Masculino , RNA Interferente Pequeno/administração & dosagem , Ratos , Ratos Endogâmicos F344 , Receptor de Insulina/imunologia , Fatores de Tempo , Transfecção
16.
Pharm Res ; 24(9): 1772-87, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17554608

RESUMO

The development of gene- and RNA interference (RNAi)-based therapeutics represents a challenge for the drug delivery field. The global brain distribution of DNA genes, as well as the targeting of specific regions of the brain, is even more complicated because conventional delivery systems, i.e. viruses, have poor diffusion in brain when injected in situ and do not cross the blood-brain barrier (BBB), which is only permeable to lipophilic molecules of less than 400 Da. Recent advances in the "Trojan Horse Liposome" (THL) technology applied to the transvascular non-viral gene therapy of brain disorders presents a promising solution to the DNA/RNAi delivery obstacle. The THL is comprised of immunoliposomes carrying either a gene for protein replacement or small hairpin RNA (shRNA) expression plasmids for RNAi effect, respectively. The THL is engineered with known lipids containing polyethyleneglycol (PEG), which stabilizes its structure in vivo in circulation. The tissue target specificity of THL is given by conjugation of approximately 1% of the PEG residues to peptidomimetic monoclonal antibodies (MAb) that bind to specific endogenous receptors (i.e. insulin and transferrin receptors) located on both the BBB and the brain cellular membranes, respectively. These MAbs mediate (a) receptor-mediated transcytosis of the THL complex through the BBB, (b) endocytosis into brain cells and (c) transport to the brain cell nuclear compartment. The present review presents an overview of the THL technology and its current application to gene therapy and RNAi, including experimental models of Parkinson's disease and brain tumors.


Assuntos
Barreira Hematoencefálica , Encéfalo/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Animais , Transporte Biológico , Genes Reporter , Humanos , Lipossomos , Interferência de RNA , RNA Interferente Pequeno/genética
17.
Biotechnol Bioeng ; 96(2): 381-91, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16937408

RESUMO

A murine monoclonal antibody (MAb) to the human insulin receptor (HIR) has been engineered for use as a brain drug delivery system for transport across the human blood-brain barrier (BBB). The HIRMAb was humanized by complementarity determining region (CDR) grafting on the framework regions (FR) of the human B43 IgG heavy chain and the human REI kappa light chain. A problem encountered in the humanization process was the poor secretion of the CDR-grafted HIRMAb by myeloma cells. This problem was solved by the production of human/mouse hybrids of the engineered heavy chain variable region (VH), which led to the replacement of five amino acids in the FR3 of the VH with original murine amino acids. No replacement of FR amino acids in the light chain variable region (VL) was required. The affinity of the humanized HIRMAb for the HIR was decreased 27% relative to the murine HIRMAb. The humanized HIRMAb avidly bound to the HIR of isolated human brain capillaries, which are used as an in vitro model system of the human BBB. The HIRMAb cross reacts with the HIR of Old World primates such as the Rhesus monkey. The humanized HIRMAb was radiolabeled with 125-iodine, and injected intravenously into an adult, anesthetized Rhesus monkey. Brain scanning showed the humanized HIRMAb was rapidly transported into all parts of the primate brain after intravenous administration. The humanized HIRMAb may be used as a brain drug and gene delivery system for the targeting of large molecule therapeutics across the BBB in humans.


Assuntos
Anticorpos Monoclonais/farmacocinética , Barreira Hematoencefálica/metabolismo , Portadores de Fármacos/farmacocinética , Sistemas de Liberação de Medicamentos , Receptor de Insulina/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/genética , Sequência de Bases , Linhagem Celular Tumoral , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Leves de Imunoglobulina/genética , Região Variável de Imunoglobulina/genética , Macaca mulatta , Camundongos , Dados de Sequência Molecular , Engenharia de Proteínas/métodos , Receptor de Insulina/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética
18.
Biochim Biophys Acta ; 1715(2): 104-10, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16125134

RESUMO

The large neutral amino acid transporter type 1, LAT1, is the principal neutral amino acid transporter expressed at the blood-brain barrier (BBB). Owing to the high affinity (low Km) of the LAT1 isoform, BBB amino acid transport in vivo is very sensitive to transport competition effects induced by hyperaminoacidemias, such as phenylketonuria. The low Km of LAT1 is a function of specific amino acid residues, and the transporter is comprised of 12 phylogenetically conserved cysteine (Cys) residues. LAT1 is highly sensitive to inhibition by inorganic mercury, but the specific cysteine residue(s) of LAT1 that account for the mercury sensitivity is not known. LAT1 forms a heterodimer with the 4F2hc heavy chain, which are joined by a disulfide bond between Cys160 of LAT1 and Cys110 of 4F2hc. The present studies use site-directed mutagenesis to convert each of the 12 cysteines of LAT1 and each of the 2 cysteines of 4F2hc into serine residues. Mutation of the cysteine residues of the 4F2hc heavy chain of the hetero-dimeric transporter did not affect transporter activity. The wild type LAT1 was inhibited by HgCl2 with a Ki of 0.56+/-0.11 microM. The inhibitory effect of HgCl2 for all 12 LAT1 Cys mutants was examined. However, except for the C439S mutant, the inhibition by HgCl2 for 11 of the 12 Cys mutants was comparable to the wild type transporter. Mutation of only 2 of the 12 cysteine residues of the LAT1 light chain, Cys88 and Cys439, altered amino acid transport. The Vmax was decreased 50% for the C88S mutant. A kinetic analysis of the C439S mutant could not be performed because transporter activity was not significantly above background. Confocal microscopy showed the C439S LAT1 mutant was not effectively transferred to the oocyte plasma membrane. These studies show that the Cys439 residue of LAT1 plays a significant role in either folding or insertion of the transporter protein in the plasma membrane.


Assuntos
Cisteína/genética , Transportador 1 de Aminoácidos Neutros Grandes/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Primers do DNA , Cinética , Transportador 1 de Aminoácidos Neutros Grandes/química , Microscopia Confocal , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oócitos/química , Coelhos , Xenopus laevis
19.
NeuroRx ; 2(1): 139-50, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15717065

RESUMO

The human epidermal growth factor receptor (EGFR) plays an oncogenic role in solid cancer, including brain primary and metastatic cancers. Transvascular nonviral gene therapy in combination with EGFR-RNA interference (RNAi) represents a new therapeutic approach to silencing oncogenic genes in solid cancers. This is achieved with pegylated immunoliposomes (PIL) carrying short hairpin RNA expression plasmids driven by the U6 RNA polymerase promoter and directed to target EGFR expression by RNAi. The PIL is comprised of a mixture of known lipids containing polyethyleneglycol (PEG), which stabilizes the PIL structure in vivo in circulation. The tissue target specificity of PILs is given by conjugation of approximately 1% of the PEG residues to monoclonal antibodies (mAbs) that bind to specific endogenous receptors (i.e., insulin and transferrin receptors) located in the brain vascular endothelium, which forms the blood brain barrier (BBB), and brain cellular membranes, respectively. These mAbs are known to induce 1) receptor-mediated transcytosis of the PIL complex through the BBB and 2) transport to the brain cell nuclear compartment. Treatment of an experimental human brain tumor model in scid mice is possible with weekly intravenous RNAi gene therapy causing reduced tumor expression of EGFR and 88% increase in survival time of these mice with advanced intracranial brain cancer. The availability of additional RNAi tumor targets may improve the therapeutic efficacy of this new anticancer drug. The accessibility to chimeric and/or humanized mAbs directed to human BBB and brain cell specific-receptors may accelerate the application of this technology to the treatment of human tumors.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Interferência de RNA , Animais , Receptores ErbB/efeitos dos fármacos , Expressão Gênica , Vetores Genéticos , Humanos , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/uso terapêutico
20.
J Nucl Med ; 45(10): 1766-75, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15471847

RESUMO

UNLABELLED: Imaging endogenous gene expression with sequence-specific antisense radiopharmaceuticals is possible if the antisense agent is enabled to traverse the biologic membrane barriers that separate the blood compartment from messenger RNA (mRNA) molecules in the cytoplasm of the target cell. The present studies were designed to image endogenous gene expression in brain cancer using peptide nucleic acid (PNA) antisense agents that were modified to allow for (a) chelation of the 111In radionuclide and (b) attachment to a brain targeting system, which delivers the PNA across both the blood-brain barrier (BBB) and the tumor cell membrane. METHODS: PNAs were designed that were antisense to either the rat glial fibrillary acidic protein (GFAP) mRNA or the rat caveolin-1alpha (CAV) mRNA. The PNA contained an amino-terminal diethylenetriaminepentaacetic acid moiety to chelate 111In and a carboxyl-terminal epsilon-biotinyl lysine residue, which enabled attachment to the delivery system. The latter comprised streptavidin (SA) and the murine OX26 monoclonal antibody to the rat transferrin receptor (TfR), which were joined by a thiol-ether linker. Control PNAs were not conjugated to SA-OX26. Brain tumors developed after the intracerebral injection of rat RG2 glial cells in adult Fischer CD344 rats. GFAP and CAV gene expression in the tumor in vivo was monitored by confocal microscopy and Northern blotting with GFAP and CAV complementary DNAs. RESULTS: If the PNA was not targeted to the TfR, then no imaging of any brain structures was possible, owing to the absence of PNA transport across the BBB. Conjugation of the 111In-GFAP-PNA to the SA-OX26 delivery system did not image brain cancer, owing to the downregulation of the GFAP mRNA in brain glial tumors. In contrast, brain cancer was selectively imaged with the 111In-CAV-PNA conjugated to SA-OX26 owing to upregulation of CAV gene expression in brain cancer. CONCLUSION: Imaging endogenous gene expression in vivo with PNA antisense radiopharmaceuticals is possible if drug-targeting technology is used. Attachment of the PNA antisense agent to the targeting ligand enables the antisense radiopharmaceutical to traverse biologic membrane barriers and access intracellular target mRNA molecules.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Perfilação da Expressão Gênica/métodos , Proteínas de Neoplasias/metabolismo , Ácidos Nucleicos Peptídicos/farmacocinética , Animais , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , DNA Antissenso/administração & dosagem , Técnicas de Transferência de Genes , Glioma/diagnóstico por imagem , Glioma/metabolismo , Radioisótopos de Índio/farmacocinética , Proteínas de Neoplasias/genética , Ácidos Nucleicos Peptídicos/genética , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA