Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 16(8): 3534-3543, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31199881

RESUMEN

Erythropoietin (EPO) is a potential therapeutic for Alzheimer's disease (AD); however, limited blood-brain barrier (BBB) penetration reduces its applicability as a CNS therapeutic. Antibodies against the BBB transferrin receptor (TfRMAbs) act as molecular Trojan horses for brain drug delivery, and a fusion protein of EPO and TfRMAb, designated TfRMAb-EPO, is protective in a mouse model of AD. TfRMAbs have Fc effector function side effects, and removal of the Fc N-linked glycosylation site by substituting Asn with Gly reduces the Fc effector function. However, the effect of such Fc mutations on the pharmacokinetics (PK) of plasma clearance of TfRMAb-based fusion proteins, such as TfRMAb-EPO, is unknown. To examine this, the plasma PK of TfRMAb-EPO (wild-type), which expresses the mouse IgG1 constant heavy chain region and includes the Asn residue at position 292, was compared to the mutant TfRMAb-N292G-EPO, in which the Asn residue at position 292 is mutated to Gly. Plasma PK was compared following IV, IP, and SQ administration for doses between 0.3 and 3 mg/kg in adult male C57 mice. The results show a profound increase in clearance (6- to 8-fold) of the TfRMAb-N292G-EPO compared with the wild-type TfRMAb-EPO following IV administration. The clearance of both the wild-type and mutant TfRMAb-EPO fusion proteins followed nonlinear PK, and a 10-fold increase in dose resulted in a 7- to 11-fold decrease in plasma clearance. Following IP and SQ administration, the Cmax values of the TfRMAb-N292G-EPO mutant were profoundly (37- to 114-fold) reduced compared with the wild-type TfRMAb-EPO, owing to comparable increases in plasma clearance of the mutant fusion protein. The wild-type TfRMAb fusion protein was associated with reticulocyte suppression, and the N292G mutation mitigated this suppression of reticulocytes. Overall, the beneficial suppression of effector function via the N292G mutation may be offset by the deleterious effect this mutation has on the plasma levels of the TfRMAb-EPO fusion protein, especially following SQ administration, which is the preferred route of administration in humans for chronic neurodegenerative diseases including AD.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Barrera Hematoencefálica/metabolismo , Eritropoyetina/farmacocinética , Inmunoconjugados/farmacocinética , Receptores de Transferrina/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacocinética , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular , Células Endoteliales , Eritropoyetina/administración & dosificación , Eritropoyetina/genética , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/genética , Inmunoconjugados/inmunología , Regiones Constantes de Inmunoglobulina/administración & dosificación , Regiones Constantes de Inmunoglobulina/genética , Regiones Constantes de Inmunoglobulina/inmunología , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inyecciones Intravenosas , Inyecciones Subcutáneas , Masculino , Ratones , Mutación , Receptores de Transferrina/inmunología , Receptores de Transferrina/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Reticulocitos/efectos de los fármacos
2.
Mol Pharm ; 15(2): 602-608, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29251941

RESUMEN

Mucopolysaccharidosis Type IIIA (MPSIIIA), also known as Sanfilippo A syndrome, is an inherited neurodegenerative disease caused by mutations in the lysosomal enzyme, N-sulfoglucosamine sulfohydrolase (SGSH), also known as sulfamidase. Mutations in the SGSH enzyme, the only mammalian heparan N-sulfatase, cause accumulation of lysosomal inclusion bodies in brain cells comprising heparan sulfate (HS) glycosaminoglycans (GAGs). Treatment of MPSIIIA with intravenous recombinant SGSH is not possible because this large molecule does not cross the blood-brain barrier (BBB). BBB penetration by SGSH was enabled in the present study by re-engineering this enzyme as an IgG-SGSH fusion protein, where the IgG domain is a chimeric monoclonal antibody (mAb) against the mouse transferrin receptor (TfR), designated the cTfRMAb. The IgG domain of the fusion protein acts as a molecular Trojan horse to deliver the enzyme into brain via transport on the endogenous BBB TfR. The cTfRMAb-SGSH fusion protein bound to the mouse TfR with high affinity, ED50 = 0.74 ± 0.07 nM, and retained high SGSH enzyme activity, 10 043 ± 1003 units/mg protein, which is comparable to recombinant human SGSH. Male and female MPSIIIA mice, null for the SGSH enzyme, were treated for 6 weeks with thrice-weekly intraperitoneal injections of vehicle, 5 mg/kg of the cTfRMAb alone, or 5 mg/kg of the cTfRMAb-SGSH fusion protein, starting at the age of 2 weeks, and were euthanized 1 week after the last injection. Brain and liver HS, as determined by liquid chromatography-mass spectrometry, were elevated 30-fold and 36-fold, respectively, in the MPSIIIA mouse. Treatment of the mice with the cTfRMAb-SGSH fusion protein caused a 70% and 85% reduction in brain and liver HS, respectively. The reduction in brain HS was associated with a 28% increase in latency on the rotarod test of motor activity in male mice. The mice exhibited no injection related reactions, and only a low titer end of study antidrug antibody response was observed. In conclusion, substantial reductions in brain pathologic GAGs in a murine model of MPSIIIA are produced by chronic systemic administration of an IgG-SGSH fusion protein engineered to penetrate the BBB via receptor-mediated transport.


Asunto(s)
Encéfalo/efectos de los fármacos , Heparitina Sulfato/análisis , Hidrolasas/uso terapéutico , Inmunoglobulina G/uso terapéutico , Mucopolisacaridosis III/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Encéfalo/irrigación sanguínea , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Heparitina Sulfato/metabolismo , Humanos , Hidrolasas/genética , Hidrolasas/inmunología , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Mucopolisacaridosis III/patología , Receptores de Transferrina/inmunología , Receptores de Transferrina/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico
3.
Mol Pharm ; 15(11): 5207-5216, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30226787

RESUMEN

A monoclonal antibody (mAb) against the blood-brain barrier (BBB) transferrin receptor (TfR) is a potential agent for delivery of biologic drugs to the brain across the BBB. However, to date, no TfRMAb has been tested with chronic dosing in a primate model. A humanized TfRMAb against the human (h) TfR1, which cross reacts with the primate TfR, was genetically engineered with high affinity (ED50 = 0.18 ± 0.04 nM) for the human TfR type 1 (TfR1). For acute dosing, the hTfRMAb was tritiated and injected intravenously (IV) in the Rhesus monkey, which confirmed rapid delivery of the humanized hTfRMAb into both brain parenchyma, via transport across the BBB, and into cerebrospinal fluid (CSF), via transport across the choroid plexus. For chronic dosing, a total of 8 adult Rhesus monkeys (4 males, 4 females) were treated twice weekly for 4 weeks with 0, 3, 10, or 30 mg/kg of the humanized hTfRMAb via a 60 min IV infusion for a total of 8 doses prior to euthanasia and microscopic examination of brain and peripheral organs. A pharmacokinetics analysis showed the plasma clearance of the hTfRMAb in the primate was nonlinear, and plasma clearance was increased over 20-fold with chronic treatment of the low dose, 3 mg/kg, of the antibody. Chronic treatment of the primates with the 30 mg/kg dose caused anemia associated with suppressed blood reticulocytes. Immunohistochemistry of terminal brain tissue showed microglia activation, based on enhanced IBA1 immuno-staining, in conjunction with astrogliosis, based on increased GFAP immuno-staining. Moderate axonal/myelin degeneration was observed in the sciatic nerve. Further studies need to be conducted to determine if this neuropathology is induced by the antibody effector function, or is an intrinsic property of targeting the TfR in brain. The results indicate that chronic treatment of Rhesus monkeys with a humanized hTfRMAb may have a narrow therapeutic index, with associated toxicity related to microglial activation and astrogliosis of the brain.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Barrera Hematoencefálica/metabolismo , Portadores de Fármacos/farmacocinética , Enfermedades del Sistema Nervioso/inducido químicamente , Receptores de Transferrina/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/genética , Antígenos CD/genética , Axones/efectos de los fármacos , Axones/patología , Barrera Hematoencefálica/patología , Portadores de Fármacos/administración & dosificación , Femenino , Inyecciones Intravenosas , Macaca mulatta , Masculino , Modelos Animales , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Enfermedades del Sistema Nervioso/patología , Ingeniería de Proteínas , Receptores de Transferrina/genética , Pruebas de Toxicidad Crónica
4.
Mol Pharm ; 15(11): 4963-4973, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30252487

RESUMEN

Erythropoietin (EPO), a glycoprotein cytokine essential to hematopoiesis, has neuroprotective effects in rodent models of Alzheimer's disease (AD). However, high therapeutic doses or invasive routes of administration of EPO are required to achieve effective brain concentrations due to low blood-brain barrier (BBB) penetrability, and high EPO doses result in hematopoietic side effects. These obstacles can be overcome by engineering a BBB-penetrable analog of EPO, which is rapidly cleared from the blood, by fusing EPO to a chimeric monoclonal antibody targeting the transferrin receptor (cTfRMAb), which acts as a molecular Trojan horse to ferry the EPO into the brain via the transvascular route. In the current study, we investigated the effects of the BBB-penetrable analog of EPO on AD pathology in a double transgenic mouse model of AD. Five and a half month old male APPswe/PSEN1dE9 (APP/PS1) transgenic mice were treated with saline ( n = 10) or the BBB-penetrable EPO ( n = 10) 3 days/week intraperitoneally for 8 weeks, compared to same-aged C57BL/6J wild-type mice treated with saline ( n = 8) with identical regiment. At 9 weeks following treatment initiation, exploration and spatial memory were assessed with the open-field and Y-maze test, mice were sacrificed, and brains were evaluated for Aß peptide load, synaptic loss, BBB disruption, microglial activation, and microhemorrhages. APP/PS1 mice treated with the BBB-penetrable cTfRMAb-EPO fusion protein had significantly lower cortical and hippocampal Aß peptide number ( p < 0.05) and immune-positive area ( p < 0.05), a decrease in hippocampal synaptic loss ( p < 0.05) and cortical microglial activation ( p < 0.001), and improved spatial memory ( p < 0.05) compared with APP/PS1 saline controls. BBB-penetrating EPO was not associated with microhemorrhage development. The cTfRMAb-EPO fusion protein offers therapeutic benefits by targeting multiple targets of AD pathogenesis and progression (Aß load, synaptic loss, microglial activation) and improving spatial memory in the APP/PS1 mouse model of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Eritropoyetina/administración & dosificación , Inmunoconjugados/administración & dosificación , Receptores de Transferrina/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Células CHO , Cricetulus , Modelos Animales de Enfermedad , Eritropoyetina/genética , Eritropoyetina/farmacocinética , Humanos , Inmunoconjugados/genética , Inmunoconjugados/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Permeabilidad , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacocinética , Resultado del Tratamiento
5.
Mol Pharm ; 14(4): 1271-1277, 2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-28279069

RESUMEN

Mucopolysaccharidosis type I (MPSI) is caused by mutations in the gene encoding the lysosomal enzyme, α-l-iduronidase (IDUA), and patients with MPSI are currently treated with IDUA enzyme replacement therapy (ERT). However, the majority of MPSI patients have severe CNS involvement, and conventional ERT does not treat the brain. The failure of ERT to treat the brain is believed to be due to the lack of IDUA transport through the blood-brain barrier (BBB). However, BBB transport of IDUA has not been directly measured, to date. BBB transport of IDUA may be enhanced by fusion of the enzyme to a monoclonal antibody (mAb) against the human insulin receptor (HIR). The HIRMAb binds the insulin receptor on the BBB to trigger transport into the brain and acts as a molecular Trojan horse to deliver IDUA to brain cells. Therefore, the purpose of the present investigation was to compare, side-by-side, the BBB transport of IDUA alone and the HIRMAb-IDUA fusion protein in the Rhesus monkey in vivo. Each protein was radio-iodinated by conjugation with the [125I]-Bolton-Hunter reagent and injected intravenously (IV) in the primate. The uptake by brain, and peripheral organs, was measured by whole body autoradiography. The results show there is no transport of IDUA alone into the brain, but that the brain uptake of the HIRMAb-IDUA fusion protein is high, 1.2% injected dose/brain. There is comparable uptake of the IDUA and the HIRMAb-IDUA fusion protein by peripheral organs, where uptake is primarily controlled by the mannose 6-phosphate receptor. The work suggests that treatment of MPSI with the HIRMAb-IDUA fusion protein will be as effective as IDUA in peripheral organs, but offer the benefit of treatment of the central nervous system in MPSI.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antígenos CD/metabolismo , Encéfalo/metabolismo , Iduronidasa/metabolismo , Receptor de Insulina/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Humanos , Macaca mulatta , Mucopolisacaridosis I/metabolismo , Receptor IGF Tipo 2/metabolismo , Succinimidas/metabolismo
6.
Mol Pharm ; 14(7): 2340-2349, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28514851

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Barrera Hematoencefálica/metabolismo , Receptores de Transferrina/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/uso terapéutico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Crioultramicrotomía , Ensayo de Inmunoadsorción Enzimática , Humanos , Masculino , Ratones , Ratones Transgénicos , Microscopía Fluorescente
7.
Mol Pharm ; 13(9): 3241-6, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27513815

RESUMEN

Brain penetration of recombinant protein drugs is possible following the re-engineering of the drug as an IgG fusion protein. The IgG domain is a monoclonal antibody (mAb) against an endogenous blood-brain barrier (BBB) receptor transporter, such as the insulin receptor. One such mAb targets the human insulin receptor (HIR) and is active in Rhesus monkeys. Prior work has measured the plasma pharmacokinetics of HIRMAb-derived fusion proteins following intravenous (IV) infusion. However, an alternative method of administration for chronic treatment of brain disease is the subcutaneous (SQ) route. The extent to which an antibody against the insulin receptor undergoes systemic distribution and clearance is unknown. Therefore, in the present study, the rate of plasma clearance of the HIRMAb is measured in Rhesus monkeys following IV or SQ administration of 3, 10, and 30 mg/kg doses of the antibody. The HIRMAb is readily absorbed into the systemic circulation following SQ injection with a 42% plasma bioavailability. The rate of plasma clearance of the antibody, 0.04-0.06 mL/min/kg, is the same following either IV or SQ administration. Owing to the slow rate of plasma clearance of the antibody, high concentrations of the HIRMAb are sustained in plasma for days after the SQ injection. The plasma concentration of the HIRMAb exceeds 0.8 mg/mL, which is 9% of the entire plasma IgG pool in the primate, after the SQ injection of the high dose, 30 mg/kg, of the antibody. In summary, the pharmacokinetics of plasma clearance of the HIRMAb are such that HIRMAb-derived fusion proteins can be developed as protein therapeutics for the brain with chronic SQ administration on a weekly or twice-weekly regimen.


Asunto(s)
Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/metabolismo , Receptor de Insulina/antagonistas & inhibidores , Receptor de Insulina/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoglobulina G/metabolismo , Macaca mulatta , Masculino , Proteínas Recombinantes de Fusión/metabolismo
8.
Mol Pharm ; 13(4): 1385-92, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-26910785

RESUMEN

Mucopolysaccharidosis Type IIIB (MPSIIIB) is caused by mutations in the gene encoding the lysosomal enzyme, α-N-acetylglucosaminidase (NAGLU). MPSIIIB presents with severe disease of the central nervous system, but intravenous NAGLU enzyme replacement therapy has not been developed because the NAGLU enzyme does not cross the blood-brain barrier (BBB). A BBB-penetrating form of the enzyme was produced by re-engineering NAGLU as an IgG-enzyme fusion protein, where the IgG domain is a monoclonal antibody (mAb) against the human insulin receptor (HIR). The HIRMAb traverses the BBB via transport on the endogenous insulin receptor and acts as a molecular Trojan horse to ferry the fused NAGLU across the BBB from blood. The NAGLU was fused to the carboxyl terminus of each heavy chain of the HIRMAb via an extended 31-amino acid linker, and the fusion protein is designated HIRMAb-LL-NAGLU. The fusion protein retains high affinity binding to the HIR, and on a molar basis has an enzyme activity equal to that of recombinant human NAGLU. Treatment of MPSIIIB fibroblasts with the fusion protein normalizes intracellular NAGLU enzyme activity and reduces sulfate incorporation into intracellular glycosoaminoglycan. The fusion protein is targeted to the lysosomal compartment of the cells as shown by confocal microscopy. The fusion protein was radiolabeled with the [(125)I]-Bolton-Hunter reagent and injected intravenously in the adult Rhesus monkey. The fusion protein was rapidly cleared from plasma by all major peripheral organs. The high brain uptake of the fusion protein, 1% injected dose/brain, enables normalization of brain NAGLU enzyme activity with a therapeutic dose of 1 mg/kg. The HIRMAb-LL-NAGLU fusion protein is a new treatment of the brain in MPSIIIB, which can be administered by noninvasive intravenous infusion.


Asunto(s)
Acetilglucosaminidasa/metabolismo , Anticuerpos Monoclonales/metabolismo , Barrera Hematoencefálica/metabolismo , Fibroblastos/metabolismo , Glicosaminoglicanos/metabolismo , Receptor de Insulina/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Acetilglucosaminidasa/genética , Animales , Anticuerpos Monoclonales/genética , Transporte Biológico/fisiología , Humanos , Macaca mulatta , Proteínas Recombinantes de Fusión/genética
10.
Biotechnol Bioeng ; 111(11): 2317-25, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24889100

RESUMEN

Mucopolysaccharidosis (MPS) Type II is caused by mutations in the gene encoding the lysosomal enzyme, iduronate 2-sulfatase (IDS). The majority of MPSII cases affect the brain. However, enzyme replacement therapy with recombinant IDS does not treat the brain, because IDS is a large molecule drug that does not cross the blood-brain barrier (BBB). To enable BBB penetration, IDS has been re-engineered as an IgG-IDS fusion protein, where the IgG domain is a monoclonal antibody (MAb) against the human insulin receptor (HIR). The HIRMAb crosses the BBB via receptor-mediated transport on the endogenous BBB insulin receptor, and the HIRMAb domain of the fusion protein acts as a molecular Trojan horse to ferry the fused IDS into brain from blood. The present study reports on the first safety pharmacology and pharmacokinetics study of the HIRMAb-IDS fusion protein. Juvenile male Rhesus monkeys were infused intravenously (IV) weekly for 26 weeks with 0, 3, 10, or 30 mg/kg of the HIRMAb-IDS fusion protein. The plasma clearance of the fusion protein followed a linear pharmacokinetics profile, which was equivalent either with measurements of the plasma concentration of immunoreactive HIRMAb-IDS fusion protein, or with assays of plasma IDS enzyme activity. Anti-drug antibody (ADA) titers were monitored monthly, and the ADA response was primarily directed against the variable region of the HIRMAb domain of the fusion protein. No infusion related reactions or clinical signs of immune response were observed during the course of the study. A battery of safety pharmacology, clinical chemistry, and tissue histopathology showed no signs of adverse events, and demonstrate the safety profile of chronic treatment of primates with 3-30 mg/kg weekly IV infusion doses of the HIRMAb-IDS fusion protein.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Antígenos CD/inmunología , Glicoproteínas/efectos adversos , Glicoproteínas/farmacocinética , Receptor de Insulina/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Glicoproteínas/administración & dosificación , Glicoproteínas/genética , Infusiones Intravenosas , Macaca mulatta , Tasa de Depuración Metabólica , Plasma/química , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacocinética
11.
Mol Pharm ; 11(8): 2928-34, 2014 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-24949884

RESUMEN

Mutations in the lysosomal enzyme, N-sulfoglucosamine sulfohydrolase (SGSH), also called sulfamidase, cause accumulation of lysosomal inclusion bodies in the brain of children born with mucopolysaccharidosis type IIIA, also called Sanfilippo type A syndrome. Enzyme replacement therapy with recombinant SGSH does not treat the brain because the enzyme is a large molecule drug that does not cross the blood-brain barrier (BBB). A BBB-penetrating form of SGSH was produced by re-engineering the enzyme as an IgG fusion protein, where the IgG domain is a monoclonal antibody (mAb) against the human insulin receptor (HIR). The HIRMAb domain of the HIRMAb-SGSH fusion protein acts as a molecular Trojan horse to ferry the fused enzyme across the BBB. The HIRMAb-SGSH was produced in stably transfected host cells and purified to homogeneity by protein A chromatography. The fusion protein reacted with antibodies against either human IgG or SGSH on Western blotting. High affinity binding to the HIR was retained following SGSH fusion to the HIRMAb, with an EC50 of 0.33 ± 0.05 nM in an HIR binding ELISA. The SGSH enzyme activity of the HIRMAb-SGSH fusion protein was 4712 ± 388 units/mg protein based on a two-step fluorometric enzyme assay. The HIRMAb-SGSH was taken up by lysosomes in MPSIIIA fibroblasts, and treatment of these cells with the fusion protein caused an 83% reduction in sulfate incorporation into lysosomal glycosoaminoglycans. The HIRMAb-SGSH fusion protein was radiolabeled with the [(125)I]-Bolton-Hunter reagent and injected intravenously in the Rhesus monkey. The brain uptake of the fusion protein was high, ∼1% injected dose/brain. Calculations, based on this level of brain uptake, suggest normalization of brain SGSH enzyme activity is possible following administration of therapeutic doses of the fusion protein. These studies describe a novel IgG-SGSH fusion protein that is a new noninvasive treatment of the brain in MPS type IIIA.


Asunto(s)
Anticuerpos Monoclonales/química , Glicosaminoglicanos/química , Hidrolasas/química , Mucopolisacaridosis III/tratamiento farmacológico , Receptor de Insulina/química , Animales , Barrera Hematoencefálica/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Terapia de Reemplazo Enzimático/métodos , Fibroblastos/metabolismo , Hidrolasas/genética , Inmunoglobulina G/química , Lisosomas/química , Macaca mulatta , Masculino , Microscopía Confocal , Mucopolisacaridosis III/metabolismo , Mutación , Permeabilidad , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química
12.
Bioconjug Chem ; 24(10): 1741-9, 2013 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-24059813

RESUMEN

Recombinant proteins are large molecule drugs that do not cross the blood-brain barrier (BBB). However, BBB-penetration of protein therapeutics is enabled by re-engineering the recombinant protein as IgG fusion proteins. The IgG domain is a monoclonal antibody (mAb) against an endogenous BBB receptor-mediated transport system, such as the human insulin receptor (HIR), and acts as a molecular Trojan horse to ferry the fused protein across the BBB. In the present study, a recombinant lysosomal enzyme, iduronate 2-sulfatase (IDS), is fused to the HIRMAb, and BBB penetration of the IDS alone vs the HIRMAb-IDS fusion protein is compared in the Rhesus monkey. Recombinant IDS and the HIRMAb-IDS fusion protein were radiolabeled with indirect iodination with the [(125)I]-Bolton-Hunter reagent and with direct iodination with Iodogen/[(125)I]-idodine. IDS and the HIRMAb-IDS fusion protein have comparable plasma pharmacokinetics and uptake by peripheral organs. IDS does not cross the BBB. The HIRMAb-IDS fusion protein crosses the BBB and the brain uptake is 1% of injected dose/brain. Brain imaging shows HIRMAb-IDS penetration to all parts of brain, and immunoprecipitation of brain radioactivity shows intact fusion protein in brain. The use of BBB molecular Trojan horses enables brain imaging of recombinant proteins that are re-engineered for BBB transport.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Barrera Hematoencefálica/metabolismo , Iduronato Sulfatasa/farmacocinética , Proteínas Recombinantes de Fusión/farmacocinética , Animales , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/inmunología , Barrera Hematoencefálica/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Caballos , Humanos , Iduronato Sulfatasa/análisis , Radioisótopos de Yodo/análisis , Macaca mulatta , Radiografía , Receptor de Insulina/inmunología , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/inmunología , Succinimidas/análisis
13.
Bioconjug Chem ; 24(1): 97-104, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23249376

RESUMEN

The chronic administration of recombinant fusion proteins in preclinical animal models may generate an immune response and the formation of antidrug antibodies (ADA). Such ADAs could alter the plasma pharmacokinetics of the fusion protein, and mask any underlying toxicity of the recombinant fusion protein. In the present study, a model IgG-enzyme fusion protein was evaluated with chronic dosing of rhesus monkeys. The IgG domain of the fusion protein is a genetically engineered monoclonal antibody (mAb) against the human insulin receptor (HIR), which is shown to cross-react with the primate insulin receptor. The enzyme domain of the fusion protein is human iduronidase (IDUA), the lysosomal enzyme mutated in Mucopolysaccharidosis Type I (MPSI). MPSI affects the brain, but enzyme replacement therapy is not effective for the brain, because IDUA does not cross the blood-brain barrier (BBB). The HIRMAb domain of the fusion protein acts as a molecular Trojan horse to deliver the IDUA across the BBB. The HIRMAb-IDUA fusion protein was administered to rhesus monkeys with weekly intravenous infusions of 3-30 mg/kg for 6 months, and the pharmacokinetics, immune response, and tissue toxicology were assessed. The pharmacokinetics of plasma clearance of the fusion protein was determined with measurements of plasma IDUA enzyme activity. ADAs formed during the course of the 6 months of treatment, as determined by a sandwich ELISA. However, the plasma clearance of the fusion protein at the start and end of the 6-month study was comparable at all drug doses. Fusion protein administration for 6 months showed no evidence of chronic tissue toxicity. These studies demonstrate that the immune response produced with chronic treatment of primates with an IgG-enzyme fusion protein has no effect on the pharmacokinetics of plasma clearance of the fusion protein.


Asunto(s)
Formación de Anticuerpos , Iduronidasa/inmunología , Inmunoglobulina G/inmunología , Macaca mulatta/inmunología , Receptor de Insulina/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/toxicidad , Humanos , Iduronidasa/administración & dosificación , Iduronidasa/farmacocinética , Iduronidasa/toxicidad , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/toxicidad , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/toxicidad
14.
Biotechnol Bioeng ; 110(5): 1456-65, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23192358

RESUMEN

Metachromatic leukodystrophy (MLD) is a lysosomal storage disorder of the brain caused by mutations in the gene encoding the lysosomal sulfatase, arylsulfatase A (ASA). It is not possible to treat the brain in MLD with recombinant ASA, because the enzyme does not cross the blood-brain barrier (BBB). In the present investigation, a BBB-penetrating IgG-ASA fusion protein is engineered and expressed, where the ASA monomer is fused to the carboxyl terminus of each heavy chain of an engineered monoclonal antibody (MAb) against the human insulin receptor (HIR). The HIRMAb crosses the BBB via receptor-mediated transport on the endogenous BBB insulin receptor, and acts as a molecular Trojan horse to ferry the ASA into brain from blood. The HIRMAb-ASA is expressed in stably transfected Chinese hamster ovary cells grown in serum free medium, and purified by protein A affinity chromatography. The fusion protein retains high affinity binding to the HIR, EC50 = 0.34 ± 0.11 nM, and retains high ASA enzyme activity, 20 ± 1 units/mg. The HIRMAb-ASA fusion protein is endocytosed and triaged to the lysosomal compartment in MLD fibroblasts. The fusion protein was radio-labeled with the Bolton-Hunter reagent, and the [(125) I]-HIRMAb-ASA rapidly penetrates the brain in the Rhesus monkey following intravenous administration. Film and emulsion autoradiography of primate brain shows global distribution of the fusion protein throughout the monkey brain. These studies describe a new biological entity that is designed to treat the brain of humans with MLD following non-invasive, intravenous infusion of an IgG-ASA fusion protein.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antígenos CD/metabolismo , Encéfalo/metabolismo , Cerebrósido Sulfatasa/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Receptor de Insulina/metabolismo , Proteínas Recombinantes de Fusión/farmacocinética , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Barrera Hematoencefálica/metabolismo , Química Encefálica , Células CHO , Cerebrósido Sulfatasa/química , Cerebrósido Sulfatasa/genética , Cricetinae , Cricetulus , Humanos , Radioisótopos de Yodo/química , Radioisótopos de Yodo/farmacocinética , Leucodistrofia Metacromática , Macaca mulatta , Masculino , Unión Proteica , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Distribución Tisular
15.
Mol Pharm ; 10(9): 3507-13, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23924247

RESUMEN

Anti-amyloid antibodies (AAA) are under development as new therapeutics that disaggregate the amyloid plaque in brain in Alzheimer's disease (AD). However, the AAAs are large molecule drugs that do not cross the blood-brain barrier (BBB), in the absence of BBB disruption. In the present study, an AAA was re-engineered for receptor-mediated transport across the BBB via the endogenous BBB transferrin receptor (TfR). A single chain Fv (ScFv) antibody form of an AAA was fused to the carboxyl terminus of each heavy chain of a chimeric monoclonal antibody (mAb) against the mouse TfR, and this produced a tetravalent bispecific antibody designated the cTfRMAb-ScFv fusion protein. Unlike a conventional AAA, which has a plasma half-time of weeks, the cTfRMAb-ScFv fusion protein is cleared from plasma in mice with a mean residence time of about 3 h. Therefore, a novel protocol was developed for the treatment of one year old presenilin (PS)-1/amyloid precursor protein (APP) AD double transgenic PSAPP mice, which were administered daily subcutaneous (sc) injections of 5 mg/kg of the cTfRMAb-ScFv fusion protein for 12 consecutive weeks. At the end of the treatment, brain amyloid plaques were quantified with confocal microscopy using both Thioflavin-S staining and immunostaining with the 6E10 antibody against Abeta amyloid fibrils. Fusion protein treatment caused a 57% and 61% reduction in amyloid plaque in the cortex and hippocampus, respectively. No increase in plasma immunoreactive Abeta amyloid peptide, and no cerebral microhemorrhage, was observed. Chronic daily sc treatment of the mice with the fusion protein caused no immune reactions and only a low titer antidrug antibody response. In conclusion, re-engineering AAAs for receptor-mediated BBB transport allows for reduction in brain amyloid plaque without cerebral microhemorrhage following daily sc treatment for 12 weeks.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/inmunología , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/uso terapéutico , Encéfalo/metabolismo , Placa Amiloide/metabolismo , Receptores de Transferrina/inmunología , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Ratones , Ratones Transgénicos
16.
Mol Pharm ; 10(4): 1425-31, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23410508

RESUMEN

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.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/química , Receptores del Factor de Necrosis Tumoral/administración & dosificación , Receptores del Factor de Necrosis Tumoral/química , Animales , Anticuerpos Monoclonales/química , Área Bajo la Curva , Barrera Hematoencefálica , Diseño de Fármacos , Etanercept , Inflamación , Infusiones Intravenosas , Infusiones Parenterales , Infusiones Subcutáneas , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Transferrina/química , Proteínas Recombinantes de Fusión/metabolismo
17.
Bioconjug Chem ; 23(6): 1318-21, 2012 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-22624578

RESUMEN

The Aß amyloid peptide of Alzheimer's disease (AD) is a potentially large molecule radiopharmaceutical for imaging the brain amyloid burden, should the peptide be made transportable across the blood-brain barrier (BBB). Peptides can be made BBB-penetrating with the combined use of Trojan horse and avidin-biotin technologies. The peptide is monobiotinylated and attached to a fusion protein of avidin (AV) and a genetically engineered monoclonal antibody (mAb) against the human insulin receptor (HIR). The fusion protein is designated HIRMAb-AV, and is produced by stably transfected mammalian host cells grown in biotin free medium. The HIRMAb domain of the fusion protein acts as a molecular Trojan horse, which crosses the BBB via transport on the endogenous insulin receptor. The avidin domain of the fusion protein creates a high-affinity linker between the HIRMAb and the biotinylated peptide radiopharmaceutical. The 4 kDa Aß(1-40) amyloid peptide of AD was N-biotinylated and radiolabeled with (125)I. The amyloid plaque binding of the [(125)I]-N-biotinyl-Aß(1-40) peptide, either without or with conjugation to the HIRMAb-AV fusion protein, was tested with film autoradiography and tissue sections of autopsy AD brain. The study shows the biotinyl-Aß(1-40) peptide still binds to amyloid plaque in AD brain to the same extent when the peptide radiopharmaceutical is either free or conjugated to the HIRMAb-AV fusion protein. The study supports further evaluation of antibody-targeted peptide radiopharmaceuticals as large molecule neuro-imaging agents that penetrate the BBB.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides , Avidina , Inmunoglobulina G , Fragmentos de Péptidos , Placa Amiloide/diagnóstico por imagen , Radiofármacos , Proteínas Recombinantes de Fusión , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Autorradiografía , Avidina/química , Biotinilación , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Humanos , Inmunoglobulina G/química , Radioisótopos de Yodo/química , Modelos Moleculares , Fragmentos de Péptidos/química , Placa Amiloide/patología , Radiografía , Radiofármacos/química , Proteínas Recombinantes de Fusión/química
18.
Drug Metab Dispos ; 40(10): 2021-5, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22822036

RESUMEN

Hurler's syndrome, or mucopolysaccharidosis type I, is a lysosomal storage disorder caused by mutations in the gene encoding the lysosomal enzyme iduronidase (IDUA). The disease affects both peripheral tissues and the central nervous system (CNS). Recombinant IDUA treatment does not affect the CNS, because IDUA does not cross the blood-brain barrier (BBB). To enable BBB penetration, human IDUA was re-engineered as an IgG-IDUA fusion protein, where the IgG domain is a genetically engineered monoclonal antibody (MAb) against the human insulin receptor (HIR). The HIRMAb penetrates the brain from the blood via transport on the endogenous BBB insulin receptor and acts as a molecular Trojan horse to deliver the fused IDUA to the brain. Before human testing, the HIRMAb-IDUA fusion protein was evaluated in a 6-month weekly dosing toxicology study at doses of 0, 3, 9, and 30 mg/kg/week of the fusion protein administered to 40 rhesus monkeys. The focus of the present study is the effect of chronic high dose administration of this fusion protein on plasma glucose and long-term glycemic control. The results show that the HIRMAb has weak insulin agonist activity and causes hypoglycemia at the high dose, 30 mg/kg, after intravenous infusion in normal saline. When dextrose is added to the saline infusion solution, no hypoglycemia is observed at any dose. An intravenous glucose tolerance test performed at the end of the 6 months of chronic treatment showed no change in glucose tolerance at any dose of the HIRMAb-IDUA fusion protein.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Glucemia/efectos de los fármacos , Iduronidasa/administración & dosificación , Receptor de Insulina/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Anticuerpos Monoclonales/toxicidad , Barrera Hematoencefálica/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Terapia de Reemplazo Enzimático , Femenino , Glucosa/administración & dosificación , Prueba de Tolerancia a la Glucosa , Humanos , Hipoglucemia/sangre , Hipoglucemia/inducido químicamente , Hipoglucemia/prevención & control , Iduronidasa/toxicidad , Infusiones Intravenosas , Macaca mulatta , Masculino , Mucopolisacaridosis I/sangre , Mucopolisacaridosis I/tratamiento farmacológico , Mucopolisacaridosis I/enzimología , Receptor de Insulina/inmunología , Proteínas Recombinantes de Fusión/toxicidad , Factores de Tiempo
19.
Drug Metab Dispos ; 40(2): 329-35, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22065691

RESUMEN

Mucopolysaccharidosis (MPS) type II (Hunter's syndrome) is caused by mutations in the iduronate 2-sulfatase (IDS) fusion protein. MPS-II affects the brain, and enzyme replacement therapy is not effective in the brain, because the enzyme does not cross the blood-brain barrier. To treat mouse models of MPS-II with brain-penetrating IDS, the lysosomal enzyme was reengineered as an IgG-IDS fusion protein. The mature human IDS was fused to the carboxyl terminus of both heavy chains of the chimeric monoclonal antibody (MAb) against the mouse transferrin receptor (TfR), and the fusion protein is designated cTfRMAb-IDS. The purity and identity of the fusion protein was confirmed by electrophoresis and Western blotting with antibodies to mouse IgG and human IDS. The EC50 of binding of the cTfRMAb-IDS fusion protein to the mouse TfR (0.85 ± 0.15 nM) was comparable to the EC50 of binding of the cTfRMAb (0.78 ± 0.05 nM). The IDS enzyme activity of the cTfRMAb-IDS fusion protein was 126 ± 1 nmol · h⁻¹ · µg⁻¹ protein. After intravenous injection in the mouse, the cTfRMAb-IDS fusion protein was rapidly removed from plasma and distributed to tissues, including brain and spinal cord. The uptake of the fusion protein by brain or spinal cord was 1.3 ± 0.1 and 2.2 ± 0.2% injected dose/g, respectively, which is 100-fold greater than the brain uptake of IDS alone. This work shows that a lysosomal sulfatase can be reengineered as an IgG-enzyme fusion protein that rapidly penetrates the brain after intravenous administration.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Barrera Hematoencefálica/metabolismo , Diseño de Fármacos , Glicoproteínas/metabolismo , Inmunoglobulina G/metabolismo , Receptores de Transferrina/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacocinética , Animales , Anticuerpos Monoclonales/genética , Afinidad de Anticuerpos , Encéfalo/metabolismo , Vectores Genéticos , Glicoproteínas/genética , Humanos , Inmunoglobulina G/genética , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Permeabilidad , Ingeniería de Proteínas , Receptores de Transferrina/metabolismo , Proteínas Recombinantes de Fusión/sangre , Proteínas Recombinantes de Fusión/metabolismo , Médula Espinal/metabolismo , Sulfatasas/genética , Sulfatasas/metabolismo , Distribución Tisular
20.
Pharmaceutics ; 14(7)2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35890374

RESUMEN

The treatment of neurological disorders with large-molecule biotherapeutics requires that the therapeutic drug be transported across the blood-brain barrier (BBB). However, recombinant biotherapeutics, such as neurotrophins, enzymes, decoy receptors, and monoclonal antibodies (MAb), do not cross the BBB. These biotherapeutics can be re-engineered as brain-penetrating bifunctional IgG fusion proteins. These recombinant proteins comprise two domains, the transport domain and the therapeutic domain, respectively. The transport domain is an MAb that acts as a molecular Trojan horse by targeting a BBB-specific endogenous receptor that induces receptor-mediated transcytosis into the brain, such as the human insulin receptor (HIR) or the transferrin receptor (TfR). The therapeutic domain of the IgG fusion protein exerts its pharmacological effect in the brain once across the BBB. A generation of bifunctional IgG fusion proteins has been engineered using genetically engineered MAbs directed to either the BBB HIR or TfR as the transport domain. These IgG fusion proteins were validated in animal models of lysosomal storage disorders; acute brain conditions, such as stroke; or chronic neurodegeneration, such as Parkinson's disease and Alzheimer's disease. Human phase I-III clinical trials were also completed for Hurler MPSI and Hunter MPSII using brain-penetrating IgG-iduronidase and -iduronate-2-sulfatase fusion protein, respectively.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA