Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
JCI Insight ; 7(24)2022 12 22.
Article in English | MEDLINE | ID: mdl-36346674

ABSTRACT

Antisense oligonucleotides (ASOs) have emerged as one of the most innovative new genetic drug modalities. However, their high molecular weight limits their bioavailability for otherwise-treatable neurological disorders. We investigated conjugation of ASOs to an antibody against the murine transferrin receptor, 8D3130, and evaluated it via systemic administration in mouse models of the neurodegenerative disease spinal muscular atrophy (SMA). SMA, like several other neurological and neuromuscular diseases, is treatable with single-stranded ASOs that modulate splicing of the survival motor neuron 2 (SMN2) gene. Administration of 8D3130-ASO conjugate resulted in elevated levels of bioavailability to the brain. Additionally, 8D3130-ASO yielded therapeutic levels of SMN2 splicing in the central nervous system of adult human SMN2-transgenic (hSMN2-transgenic) mice, which resulted in extended survival of a severely affected SMA mouse model. Systemic delivery of nucleic acid therapies with brain-targeting antibodies offers powerful translational potential for future treatments of neuromuscular and neurodegenerative diseases.


Subject(s)
Muscular Atrophy, Spinal , Neurodegenerative Diseases , Mice , Animals , Humans , Oligonucleotides/pharmacology , Oligonucleotides/therapeutic use , Neurodegenerative Diseases/drug therapy , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Central Nervous System , Oligonucleotides, Antisense/therapeutic use , Mice, Transgenic , Disease Models, Animal
2.
ACS Synth Biol ; 11(7): 2229-2237, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35797032

ABSTRACT

Rapid and flexible plasmid construct generation at scale is one of the most limiting first steps in drug discovery projects. These hurdles can partly be overcome by adopting modular DNA design principles, automated sequence fragmentation, and plasmid assembly. To this end we have designed a robust, multimodule golden gate based cloning platform for construct generation with a wide range of applications. The assembly efficiency of the system was validated by splitting sfGFP and sfCherry3C cassettes and expressing them in E. coli followed by fluorometric assessment. To minimize timelines and cost for complex constructs, we developed a software tool named FRAGLER (FRAGment recycLER) that performs codon optimization, multiple sequence alignment, and automated generation of fragments for recycling. To highlight the flexibility and robustness of the platform, we (i) generated plasmids for SarsCoV2 protein reagents, (ii) automated and parallelized assemblies, and (iii) built modular libraries of chimeric antigen receptors (CARs) variants. Applying the new assembly framework, we have greatly streamlined plasmid construction and increased our capacity for rapid generation of complex plasmids.


Subject(s)
COVID-19 , Escherichia coli , Cloning, Molecular , DNA/genetics , Escherichia coli/genetics , Genetic Vectors , Humans , Plasmids/genetics , RNA, Viral , SARS-CoV-2 , Synthetic Biology
3.
ACS Synth Biol ; 11(4): 1613-1626, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35389220

ABSTRACT

Next-generation DNA vectors for cancer immunotherapies and vaccine development require promoters eliciting predefined transcriptional activities specific to target cell types, such as dendritic cells (DCs), which underpin immune response. In this study, we describe the de novo design of DC-specific synthetic promoters via in silico assembly of cis-transcription factor response elements (TFREs) that harness the DC transcriptional landscape. Using computational genome mining approaches, candidate TFREs were identified within promoter sequences of highly expressed DC-specific genes or those exhibiting an upregulated expression during DC maturation. Individual TFREs were then screened in vitro in a target DC line and off-target cell lines derived from skeletal muscle, fibroblast, epithelial, and endothelial cells using homotypic (TFRE repeats in series) reporter constructs. Based on these data, a library of heterotypic promoter assemblies varying in the TFRE composition, copy number, and sequential arrangement was constructed and tested in vitro to identify DC-specific promoters. Analysis of the transcriptional activity and specificity of these promoters unraveled underlying design rules, primarily TFRE composition, which govern the DC-specific synthetic promoter activity. Using these design rules, a second library of exclusively DC-specific promoters exhibiting varied transcriptional activities was generated. All DC-specific synthetic promoter assemblies exhibited >5-fold activity in the target DC line relative to off-target cell lines, with transcriptional activities ranging from 8 to 67% of the nonspecific human cytomegalovirus (hCMV-IE1) promoter. We show that bioinformatic analysis of a mammalian cell transcriptional landscape is an effective strategy for de novo design of cell-type-specific synthetic promoters with precisely controllable transcriptional activities.


Subject(s)
Computational Biology , Endothelial Cells , Animals , Dendritic Cells/metabolism , Endothelial Cells/metabolism , Gene Library , Humans , Mammals/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/genetics
4.
ACS Chem Neurosci ; 12(19): 3708-3718, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34505762

ABSTRACT

Alzheimer's disease is associated with the deposition of extracellular senile plaques, made primarily of amyloid-ß (Aß), particularly peptides Aß1-42 and Aß1-40. Neprilysin, or neutral endopeptidase (NEP), catalyzes proteolysis of the amyloid peptides (Aß) and is recognized as one of the major regulators of the levels of these peptides in the brain, preventing Aß accumulation and plaque formation. Here, we used a combination of techniques to elucidate the mechanism of Aß binding and cleavage by NEP. Our findings indicate that the Aß31-X cleavage products remain bound to the neprilysin active site, reducing proteolytic activity. Interestingly, it was already shown that this Aß31-35 sequence is also critical for recognition of Aß peptides by other targets, such as the serpin-enzyme complex receptor in neuronal cells.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Amyloid , Humans , Neprilysin , Plaque, Amyloid
5.
MAbs ; 13(1): 1874121, 2021.
Article in English | MEDLINE | ID: mdl-33499723

ABSTRACT

Receptor-mediated transcytosis (RMT) is used to enhance the delivery of monoclonal antibodies (mAb) into the central nervous system (CNS). While the binding to endogenous receptors on the brain capillary endothelial cells (BCECs) may facilitate the uptake of mAbs in the brain, a strong affinity for the receptor may hinder the efficiency of transcytosis. To quantitatively investigate the effect of binding affinity on the pharmacokinetics (PK) of anti-transferrin receptor (TfR) mAbs in different regions of the rat brain, we conducted a microdialysis study to directly measure the concentration of free mAbs at different sites of interest. Our results confirmed that bivalent anti-TfR mAb with an optimal dissociation constant (KD) value (76 nM) among four affinity variants can have up to 10-fold higher transcytosed free mAb exposure in the brain interstitial fluid (bISF) compared to lower and higher affinity mAbs (5 and 174 nM). This bell-shaped relationship between KD values and the increased brain exposure of mAbs was also visible when using whole-brain PK data. However, we found that mAb concentrations in postvascular brain supernatant (obtained after capillary depletion) were almost always higher than the concentrations measured in bISF using microdialysis. We also observed that the increase in mAb area under the concentration curve in CSF compartments was less significant, which highlights the challenge in using CSF measurement as a surrogate for estimating the efficiency of RMT delivery. Our results also suggest that the determination of mAb concentrations in the brain using microdialysis may be necessary to accurately measure the PK of CNS-targeted antibodies at the site-of-actions in the brain.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibody Affinity/immunology , Brain/metabolism , Microdialysis/methods , Receptors, Transferrin/immunology , Animals , Antibodies, Monoclonal/cerebrospinal fluid , Antibodies, Monoclonal/immunology , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/blood , Area Under Curve , Biological Transport , Blood-Brain Barrier/metabolism , Brain/cytology , CHO Cells , Cricetinae , Cricetulus , Endothelial Cells/metabolism , Humans , Male , Rats, Sprague-Dawley , Transcytosis , Trastuzumab/administration & dosage , Trastuzumab/blood
6.
Pain ; 160(9): 1989-2003, 2019 09.
Article in English | MEDLINE | ID: mdl-31045747

ABSTRACT

P2X4 is a ligand-gated ion channel implicated in neuropathic pain. Drug discovery efforts targeting P2X4 have been unsuccessful largely because of the difficulty in engineering specificity and selectivity. Here, we describe for the first time the generation of a panel of diverse monoclonal antibodies (mAbs) to human and mouse P2X4, capable of both positive and negative modulation of channel function. The affinity-optimised anti-P2X4 mAb IgG#151-LO showed exquisite selectivity for human P2X4 and induced potent and complete block of P2X4 currents. Site-directed mutagenesis of P2X4 revealed the head domain as a key interaction site for inhibitory mAbs. Inhibition of spinal P2X4 either by intrathecal delivery of an anti-P2X4 mAb or by systemic delivery of an anti-P2X4 bispecific mAb with enhanced blood-spinal cord barrier permeability produced long-lasting (>7 days) analgesia in a mouse model of neuropathic pain. We therefore propose that inhibitory mAbs binding the head domain of P2X4 have therapeutic potential for the treatment of neuropathic pain.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Neuralgia/metabolism , Neuralgia/prevention & control , Receptors, Purinergic P2X4/metabolism , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Injections, Spinal , Mice , Mice, Inbred C57BL , Protein Binding/physiology , Purinergic P2X Receptor Antagonists/administration & dosage , Purinergic P2X Receptor Antagonists/metabolism , Rats , Rats, Sprague-Dawley
7.
J Cereb Blood Flow Metab ; 39(10): 2074-2088, 2019 10.
Article in English | MEDLINE | ID: mdl-29845881

ABSTRACT

Delivery of biologic drugs across the blood-brain barrier is becoming a reality. However, the solutions often involve the assembly of complex multi-specific antibody molecules. Here we utilize a simple 12 amino-acid peptide originating from the melanotransferrin (MTf) protein that has shown improved brain delivery properties. 3D confocal fluorescence microscopic analysis demonstrated brain parenchymal localisation of a fluorescently labelled antibody (NIP228) when chemically conjugated to either the MTf peptide or full-length MTf protein. Measurement of plasma kinetics demonstrated the MTf peptide fusions had very similar kinetics to an unmodified NIP228 control antibody, whereas the fusion to MTf protein had significantly reduced plasma exposure most likely due to a higher tissue distribution in the periphery. Brain exposure for the MTf peptide fusions was significantly increased for the duration of the study, exceeding that of the fusions to full length MTf protein. Using a neuropathic pain model, we have demonstrated that fusions to interleukin-1 receptor antagonist (IL-1RA) are able to induce significant and durable analgesia following peripheral administration. These data demonstrate that recombinant and chemically conjugated MTf-based brain delivery vectors can deliver therapeutic levels of drug to the central nervous system.


Subject(s)
Drug Carriers/metabolism , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Membrane Glycoproteins/metabolism , Neuralgia/drug therapy , Peptides/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Drug Carriers/chemistry , Humans , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Interleukin 1 Receptor Antagonist Protein/pharmacokinetics , Male , Membrane Glycoproteins/chemistry , Mice, Inbred C57BL , Neuralgia/metabolism , Peptides/chemistry
8.
Mol Pharm ; 15(4): 1420-1431, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29485883

ABSTRACT

The blood-brain barrier (BBB) is a formidable obstacle for brain delivery of therapeutic antibodies. However, antibodies against the transferrin receptor (TfR), enriched in brain endothelial cells, have been developed as delivery carriers of therapeutic cargoes into the brain via a receptor-mediated transcytosis pathway. In vitro and in vivo studies demonstrated that either a low-affinity or monovalent binding of these antibodies to the TfR improves their release on the abluminal side of the BBB and target engagement in brain parenchyma. However, these studies have been performed with mouse-selective TfR antibodies that recognize different TfR epitopes and have varied binding characteristics. In this study, we evaluated serum pharmacokinetics and brain and CSF exposure of the rat TfR-binding antibody OX26 affinity variants, having KDs of 5 nM, 76 nM, 108 nM, and 174 nM, all binding the same epitope in bivalent format. Pharmacodynamic responses were tested in the Hargreaves chronic pain model after conjugation of OX26 affinity variants with the analgesic and antiepileptic peptide, galanin. OX26 variants with affinities of 76 nM and 108 nM showed enhanced brain and cerebrospinal fluid (CSF) exposure and higher potency in the Hargreaves model, compared to a 5 nM affinity variant; lowering affinity to 174 nM resulted in prolonged serum pharmacokinetics, but reduced brain and CSF exposure. The study demonstrates that binding affinity optimization of TfR-binding antibodies could improve their brain and CSF exposure even in the absence of monovalent TfR engagement.


Subject(s)
Antibodies, Monoclonal/chemistry , Brain/drug effects , Galanin/chemistry , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibody Affinity/physiology , Bioengineering/methods , Blood-Brain Barrier/metabolism , Brain/metabolism , Cerebrospinal Fluid/metabolism , Galanin/metabolism , Male , Protein Transport/physiology , Rats , Rats, Sprague-Dawley
9.
Pain ; 159(3): 550-559, 2018 03.
Article in English | MEDLINE | ID: mdl-29351125

ABSTRACT

With less than 50% of patients responding to the current standard of care and poor efficacy and selectivity of current treatments, neuropathic pain continues to be an area of considerable unmet medical need. Biological therapeutics such as monoclonal antibodies (mAbs) provide better intrinsic selectivity; however, delivery to the central nervous system (CNS) remains a challenge. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is well described in inflammation-induced pain, and early-phase clinical trials evaluating its antagonism have exemplified its importance as a peripheral pain target. Here, we investigate the role of this cytokine in a murine model of traumatic nerve injury and show that deletion of the GM-CSF receptor or treatment with an antagonizing mAb alleviates pain. We also demonstrate enhanced analgesic efficacy using an engineered construct that has greater capacity to penetrate the CNS. Despite observing GM-CSF receptor expression in microglia and astrocytes, the gliosis response in the dorsal horn was not altered in nerve injured knockout mice compared with wild-type littermate controls as evaluated by ionized calcium binding adapter molecule 1 (Iba1) and glial fibrillary acidic protein, respectively. Functional analysis of glial cells revealed that pretreatment with GM-CSF potentiated lipopolysaccharide-induced release of proinflammatory cytokines. In summary, our data indicate that GM-CSF is a proinflammatory cytokine that contributes to nociceptive signalling through driving spinal glial cell secretion of proinflammatory mediators. In addition, we report a successful approach to accessing CNS pain targets, providing promise for central compartment delivery of analgesics.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Analgesics/therapeutic use , Animals , Antibodies/therapeutic use , Brain/cytology , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Neuralgia/pathology , Neuroglia/drug effects , Signal Transduction/drug effects
10.
Pain ; 158(4): 660-668, 2017 04.
Article in English | MEDLINE | ID: mdl-28009628

ABSTRACT

Neuropathic pain is a major unmet medical need, with only 30% to 35% of patients responding to the current standard of care. The discovery and development of novel therapeutics to address this unmet need have been hampered by poor target engagement, the selectivity of novel molecules, and limited access to the relevant compartments. Biological therapeutics, either monoclonal antibodies (mAbs) or peptides, offer a solution to the challenge of specificity as the intrinsic selectivity of these kinds of molecules is significantly higher than traditional medicinal chemistry-derived approaches. The interleukin-1 receptor system within the spinal cord has been implicated in the amplification of pain signals, and its central antagonism provides relief of neuropathic pain. Targeting the IL-1 system in the spinal cord with biological drugs, however, raises the even greater challenge of delivery to the central compartment. Targeting the transferrin receptor with monoclonal antibodies has proved successful in traversing the endothelial cell-derived blood-brain barrier and delivering proteins to the central nervous system. In this study, we describe a novel construct exemplifying an engineered solution to overcome these challenges. We have generated a novel anti-transferrin receptor-interleukin-1 receptor antagonist fusion that transports to the central nervous system and delivers efficacy in a model of nerve ligation-induced hypersensitivity. Approaches such as these provide promise for novel and selective analgesics that target the central compartment.


Subject(s)
Antibodies/therapeutic use , Central Nervous System/drug effects , Hyperalgesia/etiology , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Receptors, Transferrin/immunology , Sciatica/complications , Animals , Antibodies/pharmacology , Central Nervous System/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Recombinant Fusion Proteins/pharmacology , Time Factors
11.
Drug Discov Today Technol ; 20: 49-52, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27986223

ABSTRACT

Delivery of large molecule drugs across the blood brain barrier is increasingly being seen as an achievable goal. Several technologies have been described where following peripheral administration the molecules can be detected in the brain. Foremost amongst these technologies are antibodies against the transferrin receptor. Following a burst of publications in the very early twenty first century, excitement seemed to wane as contrary data started to emerge. Over the last few years antibodies against transferrin receptor have again started to raise hopes of successful drug delivery to the central nervous system, as protein engineering techniques have allowed a more detailed understanding of the antibody properties necessary for successful transport across the blood brain barrier.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Delivery Systems , Receptors, Transferrin/metabolism , Animals , Antibodies/immunology , Humans , Pharmaceutical Preparations/metabolism , Receptors, Transferrin/immunology
12.
PLoS One ; 11(7): e0158114, 2016.
Article in English | MEDLINE | ID: mdl-27437944

ABSTRACT

Neprilysin (NEP) is an endogenous protease that degrades a wide range of peptides including amyloid beta (Aß), the main pathological component of Alzheimer's disease (AD). We have engineered NEP as a potential therapeutic for AD but found in pre-clinical safety testing that this variant increased prothrombin time (PT) and activated partial thromboplastin time (APTT). The objective of the current study was to investigate the effect of wild type NEP and the engineered variant on coagulation and define the mechanism by which this effect is mediated. PT and APTT were measured in cynomolgus monkeys and rats dosed with a human serum albumin fusion with an engineered variant of NEP (HSA-NEPv) as well as in control plasma spiked with wild type or variant enzyme. The coagulation factor targeted by NEP was determined using in vitro prothrombinase, calibrated automated thrombogram (CAT) and fibrin formation assays as well as N-terminal sequencing of fibrinogen treated with the enzyme. We demonstrate that HSA-NEP wild type and HSA-NEPv unexpectedly impaired coagulation, increasing PT and APTT in plasma samples and abolishing fibrin formation from fibrinogen. This effect was mediated through cleavage of the N-termini of the Aα- and Bß-chains of fibrinogen thereby significantly impairing initiation of fibrin formation by thrombin. Fibrinogen has therefore been identified for the first time as a substrate for NEP wild type suggesting that the enzyme may have a role in regulating fibrin formation. Reductions in NEP levels observed in AD and cerebral amyloid angiopathy may contribute to neurovascular degeneration observed in these conditions.


Subject(s)
Alzheimer Disease/drug therapy , Cerebral Amyloid Angiopathy/drug therapy , Neprilysin/administration & dosage , Serum Albumin/genetics , Alzheimer Disease/blood , Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Animals , Blood Coagulation/drug effects , Cerebral Amyloid Angiopathy/blood , Cerebral Amyloid Angiopathy/genetics , Fibrin/drug effects , Fibrin/metabolism , Fibrinogen/antagonists & inhibitors , Humans , Macaca fascicularis , Neprilysin/adverse effects , Neprilysin/genetics , Partial Thromboplastin Time , Proteolysis/drug effects , Prothrombin Time , Rats , Serum Albumin/administration & dosage , Serum Albumin/adverse effects , Thromboplastin/genetics
13.
FASEB J ; 30(5): 1927-40, 2016 05.
Article in English | MEDLINE | ID: mdl-26839377

ABSTRACT

Receptor mediated transcytosis harnessing the cellular uptake and transport of natural ligands across the blood-brain barrier (BBB) has been identified as a means for antibody delivery to the CNS. In this study, we characterized bispecific antibodies in which a BBB-crossing antibody fragment FC5 was used as a BBB carrier. Cargo antibodies were either a high-affinity, selective antibody antagonist of the metabotropic glutamate receptor-1 (BBB-mGluR1), a widely abundant CNS target, or an IgG that does not bind the CNS target (BBB-NiP). Both BBB-NiP and BBB-mGluR1 demonstrated a similar 20-fold enhanced rate of transcytosis across an in vitro BBB model compared with mGluR1 IgG fused to a control antibody fragment. All 3 bispecific antibodies exhibited identical pharmacokinetics in vivo Comparative assessment of BBB-NiP and BBB-mGluR1 revealed that, whereas their serum pharmacokinetics and BBB penetration were identical, their central disposition (brain levels) and elimination (cerebrospinal fluid levels) were widely different, due to central target-mediated removal of the mGluR1-engaging antibody. Central mGluR1 target engagement after systemic administration was demonstrated by a dose-dependent inhibition of mGluR-1-mediated thermal hyperalgesia and by colocalization of the antibody with thalamic neurons involved in mGluR1-mediated pain processing. We demonstrate the feasibility of targeting central G-protein-coupled receptors using a BBB-crossing bispecific antibody approach and emerging principles that govern brain distribution and disposition of these antibodies. These data will be important for designing safe and selective CNS antibody therapeutics.-Webster, C. I., Caram-Salas, N., Haqqani, A. S., Thom, G., Brown, L., Rennie, K., Yogi, A., Costain, W., Brunette, E., Stanimirovic, D. B. Brain penetration, target engagement, and disposition of the blood-brain barrier-crossing bispecific antibody antagonist of metabotropic glutamate receptor type 1.


Subject(s)
Antibodies, Bispecific/pharmacology , Brain/metabolism , Pain/drug therapy , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Analgesics , Animals , Biological Products/metabolism , Biological Transport , Blood-Brain Barrier/metabolism , Brain/drug effects , Camelidae , Cell Membrane , HEK293 Cells , Hot Temperature/adverse effects , Humans , Immunoconjugates/metabolism , Immunoglobulin G/immunology , Pain/etiology , Protein Engineering/methods , Rats , Receptors, Metabotropic Glutamate/metabolism
14.
MAbs ; 8(2): 253-63, 2016.
Article in English | MEDLINE | ID: mdl-26821574

ABSTRACT

The immunogenicity of clinically administered antibodies has clinical implications for the patients receiving them, ranging from mild consequences, such as increased clearance of the drug from the circulation, to life-threatening effects. The emergence of methods to engineer variable regions resulting in the generation of humanised and fully human antibodies as therapeutics has reduced the potential for adverse immunogenicity. However, due to differences in sequence referred to as allotypic variation, antibody constant regions are not homogeneous within the human population, even within sub-classes of the same immunoglobulin isotype. For therapeutically administered antibodies, the potential exists for an immune response from the patient to the antibody if the allotype of patient and antibody do not match. Allotypic distribution in the human population varies within and across ethnic groups making the choice of allotype for a therapeutic antibody difficult. This study investigated the potential of human IgG1 allotypes to stimulate responses in human CD4(+) T cells from donors matched for homologous and heterologous IgG1 allotypes. Allotypic variants of the therapeutic monoclonal antibody trastuzumab were administered to genetically defined allotypic matched and mismatched donor T cells. No significant responses were observed in the mismatched T cells. To investigate the lack of T-cell responses in relation to mismatched allotypes, HLA-DR agretopes were identified via MHC associated peptide proteomics (MAPPs). As expected, many HLA-DR restricted peptides were presented. However, there were no peptides presented from the sequence regions containing the allotypic variations. Taken together, the results from the T-cell assay and MAPPs assay indicate that the allotypic differences in human IgG1 do not represent a significant risk for induction of immunogenicity.


Subject(s)
Blood Donors , CD4-Positive T-Lymphocytes/immunology , HLA-DR Antigens/immunology , Immunoglobulin G/immunology , Immunoglobulin Gm Allotypes/immunology , Female , Humans
16.
PLoS One ; 9(8): e104001, 2014.
Article in English | MEDLINE | ID: mdl-25089527

ABSTRACT

Neprilysin is a transmembrane zinc metallopeptidase that degrades a wide range of peptide substrates. It has received attention as a potential therapy for Alzheimer's disease due to its ability to degrade the peptide amyloid beta. However, its broad range of peptide substrates has the potential to limit its therapeutic use due to degradation of additional peptides substrates that tightly regulate many physiological processes. We sought to generate a soluble version of the ectodomain of neprilysin with improved activity and specificity towards amyloid beta as a potential therapeutic for Alzheimer's disease. Extensive amino acid substitutions were performed at positions surrounding the active site and inner surface of the enzyme and variants screened for activity on amyloid beta 1-40, 1-42 and a variety of other physiologically relevant peptides. We identified several mutations that modulated and improved both enzyme selectivity and intrinsic activity. Neprilysin variant G399V/G714K displayed an approximately 20-fold improved activity on amyloid beta 1-40 and up to a 3,200-fold reduction in activity on other peptides. Along with the altered peptide substrate specificity, the mutant enzyme produced a markedly altered series of amyloid beta cleavage products compared to the wild-type enzyme. Crystallisation of the mutant enzyme revealed that the amino acid substitutions result in alteration of the shape and size of the pocket containing the active site compared to the wild-type enzyme. The mutant enzyme offers the potential for the more efficient degradation of amyloid beta in vivo as a therapeutic for the treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/chemistry , Neprilysin/chemistry , Peptide Fragments/chemistry , Peptides/chemistry , Protein Engineering , Recombinant Proteins/chemistry , Alzheimer Disease/drug therapy , Amino Acid Sequence , Amino Acid Substitution , Gene Expression , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Neprilysin/genetics , Peptides/genetics , Protein Structure, Tertiary , Proteolysis , Recombinant Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Structure-Activity Relationship
17.
Brain ; 137(Pt 2): 553-64, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24259408

ABSTRACT

Alzheimer's disease is characterized by the accumulation of amyloid deposits in the brain and the progressive loss of cognitive functions. Although the precise role of amyloid-ß in disease progression remains somewhat controversial, many efforts to halt or reverse disease progression have focussed on reducing its synthesis or enhancing its removal. It is believed that brain and peripheral soluble amyloid-ß are in equilibrium and it has previously been hypothesized that a reduction in peripheral amyloid-ß can lower brain amyloid-ß, thereby reducing formation of plaques predominantly composed of insoluble amyloid-ß; the so-called peripheral sink hypothesis. Here we describe the use of an amyloid-ß degrading enzyme, the endogenous metallopeptidase neprilysin, which is fused to albumin to extend plasma half-life and has been engineered to confer increased amyloid-ß degradation activity. We used this molecule to investigate the effect of degradation of peripheral amyloid-ß on amyloid-ß levels in the brain and cerebrospinal fluid after repeated intravenous dosing for up to 4 months in Tg2576 transgenic mice, and 1 month in rats and monkeys. This molecule proved highly effective at degradation of amyloid-ß in the periphery but did not alter brain or cerebrospinal fluid amyloid-ß levels, suggesting that the peripheral sink hypothesis is not valid and is the first time that this has been demonstrated in non-human primates.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Brain/drug effects , Brain/metabolism , Neprilysin/administration & dosage , Animals , Female , Humans , Injections, Intravenous , Macaca fascicularis , Male , Mice , Mice, Transgenic , Random Allocation , Rats , Rats, Sprague-Dawley
18.
BMC Neurosci ; 14: 59, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23773766

ABSTRACT

BACKGROUND: Modelling the blood-CNS barriers of the brain and spinal cord in vitro continues to provide a considerable challenge for research studying the passage of large and small molecules in and out of the central nervous system, both within the context of basic biology and for pharmaceutical drug discovery. Although there has been considerable success over the previous two decades in establishing useful in vitro primary endothelial cell cultures from the blood-CNS barriers, no model fully mimics the high electrical resistance, low paracellular permeability and selective influx/efflux characteristics of the in vivo situation. Furthermore, such primary-derived cultures are typically labour-intensive and generate low yields of cells, limiting scope for experimental work. We thus aimed to establish protocols for the high yield isolation and culture of endothelial cells from both rat brain and spinal cord. Our aim was to optimise in vitro conditions for inducing phenotypic characteristics in these cells that were reminiscent of the in vivo situation, such that they developed into tight endothelial barriers suitable for performing investigative biology and permeability studies. METHODS: Brain and spinal cord tissue was taken from the same rats and used to specifically isolate endothelial cells to reconstitute as in vitro blood-CNS barrier models. Isolated endothelial cells were cultured to expand the cellular yield and then passaged onto cell culture inserts for further investigation. Cell culture conditions were optimised using commercially available reagents and the resulting barrier-forming endothelial monolayers were characterised by functional permeability experiments and in vitro phenotyping by immunocytochemistry and western blotting. RESULTS: Using a combination of modified handling techniques and cell culture conditions, we have established and optimised a protocol for the in vitro culture of brain and, for the first time in rat, spinal cord endothelial cells. High yields of both CNS endothelial cell types can be obtained, and these can be passaged onto large numbers of cell culture inserts for in vitro permeability studies. The passaged brain and spinal cord endothelial cells are pure and express endothelial markers, tight junction proteins and intracellular transport machinery. Further, both models exhibit tight, functional barrier characteristics that are discriminating against large and small molecules in permeability assays and show functional expression of the pharmaceutically important P-gp efflux transporter. CONCLUSIONS: Our techniques allow the provision of high yields of robust sister cultures of endothelial cells that accurately model the blood-CNS barriers in vitro. These models are ideally suited for use in studying the biology of the blood-brain barrier and blood-spinal cord barrier in vitro and for pre-clinical drug discovery.


Subject(s)
Blood-Brain Barrier/cytology , Blood-Brain Barrier/physiology , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Models, Biological , Spinal Cord/cytology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Brain/anatomy & histology , Cells, Cultured , Chromatography, Liquid , Claudin-5/metabolism , Coculture Techniques , Dextrans/metabolism , Electric Impedance , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Isoquinolines/metabolism , Male , Mass Spectrometry , Neuroglia/physiology , Permeability , Rats , Rats, Wistar
19.
MAbs ; 5(3): 406-17, 2013.
Article in English | MEDLINE | ID: mdl-23567207

ABSTRACT

Antibodies have become the fastest growing class of biological therapeutics, in part due to their exquisite specificity and ability to modulate protein-protein interactions with a high biological potency. The relatively large size and bivalency of antibodies, however, limits their use as therapeutics in certain circumstances. Antibody fragments, such as single-chain variable fragments and antigen binding-fragments, have emerged as viable alternatives, but without further modifications these monovalent formats have reduced terminal serum half-lives because of their small size and lack of an Fc domain, which is required for FcRn-mediated recycling. Using rational engineering of the IgG4 Fc domain to disrupt key interactions at the CH3-CH3 interface, we identified a number of point mutations that abolish Fc dimerization and created half-antibodies, a novel monovalent antibody format that retains a monomeric Fc domain. Introduction of these mutations into an IgG1 framework also led to the creation of half-antibodies. These half-antibodies were shown to be soluble, thermodynamically stable and monomeric, characteristics that are favorable for use as therapeutic proteins. Despite significantly reduced FcRn binding in vitro, which suggests that avidity gains in a dimeric Fc are critical to optimal FcRn binding, this format demonstrated an increased terminal serum half-life compared with that expected for most alternative antibody fragments.


Subject(s)
Histocompatibility Antigens Class I/genetics , Immunoglobulin G/genetics , Mutation/genetics , Receptors, Fc/genetics , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/genetics , Animals , Half-Life , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/chemistry , Mice , Mice, Inbred BALB C , Protein Binding/drug effects , Protein Engineering , Protein Stability/drug effects , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/genetics , Receptors, Fc/chemistry , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...