Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
JCO Oncol Pract ; 19(8): 620-625, 2023 08.
Article in English | MEDLINE | ID: mdl-37319386

ABSTRACT

PURPOSE: In patients with metastatic lung adenocarcinoma, evidence-based first-line treatment decisions require analysis of tumors for genomic alterations (GAs). Optimizing the genotyping paradigm may improve the delivery of precision oncology care. Actionable GAs can be identified by analyzing tumor tissue or circulating tumor DNA using liquid biopsy. Consensus guidelines for when to use liquid biopsy have not been established. We evaluated the routine use of liquid biopsy performed simultaneously with tissue testing in patients with newly diagnosed, stage IV lung adenocarcinoma. METHODS: We performed a retrospective study comparing patients who underwent tissue genotyping alone (standard biopsy group) with patients who had simultaneous liquid and tissue genotyping (combined biopsy group). We examined the time to reach a final diagnosis, the need for repeat biopsies, and diagnostic accuracy. RESULTS: Forty two patients in the combined biopsy group and 78 in the standard biopsy group met the inclusion criteria. The standard group had a mean time to diagnosis of 33.5 days, compared with 20.6 days in the combined group (P < .001 by two-tailed t-test). In the combined group, 14 patients did not have sufficient tissue for molecular analysis (30%); however, in 11 (79%) of these patients, liquid biopsy identified a GA that eliminated the need for a second tissue biopsy. In patients who completed both tests, each test found actionable GAs missed by the other. CONCLUSION: Performing liquid biopsy simultaneously with tissue genotyping is feasible in an academic community medical center. Potential advantages of simultaneous liquid and tissue biopsies include shorter time to obtain a definitive molecular diagnosis, reduced need for a repeat biopsy, and improved detection of actionable mutations, although a sequential strategy that saves costs by beginning with a liquid biopsy may be ideal.


Subject(s)
Adenocarcinoma of Lung , Circulating Tumor DNA , Lung Neoplasms , Humans , Circulating Tumor DNA/analysis , Circulating Tumor DNA/genetics , Lung Neoplasms/genetics , Lung Neoplasms/diagnosis , Genotype , Retrospective Studies , Precision Medicine , Adenocarcinoma of Lung/genetics
2.
Diagnostics (Basel) ; 11(11)2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34829439

ABSTRACT

Efforts to control SARS-CoV-2 have been challenged by the emergence of variant strains that have important implications for clinical and epidemiological decision making. Four variants of concern (VOCs) have been designated by the Centers for Disease Control and Prevention (CDC), namely, B.1.617.2 (delta), B.1.1.7 (alpha), B.1.351 (beta), and P.1 (gamma), although the last three have been downgraded to variants being monitored (VBMs). VOCs and VBMs have shown increased transmissibility and/or disease severity, resistance to convalescent SARS-CoV-2 immunity and antibody therapeutics, and the potential to evade diagnostic detection. Methods are needed for point-of-care (POC) testing to rapidly identify these variants, protect vulnerable populations, and improve surveillance. Antigen-detection rapid diagnostic tests (Ag-RDTs) are ideal for POC use, but Ag-RDTs that recognize specific variants have not yet been implemented. Here, we describe a mAb (2E8) that is specific for the SARS-CoV-2 spike protein N501 residue. The 2E8 mAb can distinguish the delta VOC from variants with the N501Y meta-signature, which is characterized by convergent mutations that contribute to increased virulence and evasion of host immunity. Among the N501Y-containing mutants formerly designated as VOCs (alpha, beta, and gamma), a previously described mAb, CB6, can distinguish beta from alpha and gamma. When used in a sandwich ELISA, these mAbs sort these important SARS-CoV-2 variants into three diagnostic categories, namely, (1) delta, (2) alpha or gamma, and (3) beta. As delta is currently the predominant variant globally, they will be useful for POC testing to identify N501Y meta-signature variants, protect individuals in high-risk settings, and help detect epidemiological shifts among SARS-CoV-2 variants.

3.
Antibodies (Basel) ; 9(1)2020 Feb 28.
Article in English | MEDLINE | ID: mdl-32121092

ABSTRACT

Poliovirus (PV)-specific intestinal IgAs are important for cessation of PV shedding in the gastrointestinal tract following an acute infection with wild type or vaccine-derived PV strains. We sought to produce IgA monoclonal antibodies (mAbs) with PV neutralizing activity. We first performed de novo IgA discovery from primary human B cells using a hybridoma method that allows assessment of mAb binding and expression on the hybridoma surface: On-Cell mAb Screening (OCMS™). Six IgA1 mAbs were cloned by this method; three potently neutralized type 3 Sabin and wt PV strains. The hybridoma mAbs were heterogeneous, expressed in monomeric, dimeric, and aberrant forms. We also used recombinant methods to convert two high-potency anti-PV IgG mAbs into dimeric IgA1 and IgA2 mAbs. Isotype switching did not substantially change their neutralization activities. To purify the recombinant mAbs, Protein L binding was used, and one of the mAbs required a single amino acid substitution in its κ LC in order to enable protein L binding. Lastly, we used OCMS to assess IgA expression on the surface of hybridomas and transiently transfected, adherent cells. These studies have generated potent anti-PV IgA mAbs, for use in animal models, as well as additional tools for the discovery and production of human IgA mAbs.

4.
MAbs ; 11(3): 546-558, 2019 04.
Article in English | MEDLINE | ID: mdl-30794061

ABSTRACT

Hybridoma methods for monoclonal antibody (mAb) cloning are a mainstay of biomedical research, but they are hindered by the need to maintain hybridomas in oligoclonal pools during antibody screening. Here, we describe a system in which hybridomas specifically capture and display the mAbs they secrete: On-Cell mAb Screening (OCMS™). In OCMS™, mAbs displayed on the cell surface can be rapidly assayed for expression level and binding specificity using fluorescent antigens with high-content (image-based) methods or flow cytometry. OCMS™ demonstrated specific mAb binding to poliovirus and rabies virus by forming a cell surface IgG "cap", as a universal assay for anti-viral mAbs. We produced and characterized OCMS™-enabled hybridomas secreting mAbs that neutralize poliovirus and used fluorescence microscopy to identify and clone a human mAb specific for the human N-methyl-D-aspartate receptor. Lastly, we used OCMS™ to assess expression and antigen binding of a recombinant mAb produced in 293T cells. As a novel method to physically associate mAbs with the hybridomas that secrete them, OCMS™ overcomes a central challenge to hybridoma mAb screening and offers new paradigms for mAb discovery and production.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Cell Surface Display Techniques/methods , Flow Cytometry , Hybridomas/immunology , Poliovirus/immunology , Rabies virus/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , HEK293 Cells , Humans
5.
Ann Clin Transl Neurol ; 5(8): 935-951, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30128318

ABSTRACT

OBJECTIVE: Anti-NMDA receptor encephalitis (ANRE) is a potentially lethal encephalitis attributed to autoantibodies against the N-methyl-D-aspartate receptor (NMDAR). We sought to clone and characterize monoclonal antibodies (mAbs) from an ANRE patient. METHODS: We used a hybridoma method to clone two IgG mAbs from a female patient with ANRE without teratoma, and characterized their binding activities on NMDAR-transfected cell lines, cultured primary rat neurons, and mouse hippocampus. We also assessed their effects on voluntary locomotor activity in mice and binding to NMDAR in vivo. RESULTS: The mAbs are structurally distinct and arose from distinct B-cell lineages. They recognize different epitopes on the GluN1 amino terminal domain (ATD), yet both require amino acids important for post-translational modification. Both mAbs bind subsets of GluN1 on cultured rat hippocampal neurons. The 5F5 mAb binds mouse brain hippocampal tissues, and the GluN1 recognized on cultured rat neurons was substantially extra-synaptic. Antibody binding to primary hippocampal neurons induced receptor internalization. The NMDAR inhibitor MK-801 inhibited internalization without preventing mAb binding; AP5 inhibited both mAb binding and internalization. Exposure of mice to the mAbs following permeabilization of the blood brain barrier increased voluntary wheel running activity, similar to low doses of the NMDAR inhibitor, MK-801. INTERPRETATION: These mAbs recapitulate features demonstrated in previous studies of ANRE patient CSF, and exert effects on NMDAR in vitro and in vivo consistent with modulation of NMDAR activity.

6.
BMC Biotechnol ; 18(1): 41, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29945600

ABSTRACT

BACKGROUND: Anti-NMDA receptor encephalitis (ANRE) is a potentially lethal disease attributed to auto-antibodies against the N-methyl-D-aspartate receptor (NMDAR). Full recovery is possible if therapy is initiated early in the disease course. Detection of ANRE antibodies in the cerebrospinal fluid (CSF) is essential for diagnosis. The assays for ANRE-associated IgGs often rely on cells transiently transfected with NMDAR genes. A cell line that stably expresses pathogenic NMDAR epitopes could improve standardization of the assays and provide antigen that could be used in commercial solid state assay systems. RESULTS: We expressed the amino terminal domain (ATD) of the GluN1 NMDAR subunit (NR1) as a fusion protein on the outer plasma membrane of 293T cells, creating a stable cell population (293T-ATD) that is recognized by ANRE patient monoclonal antibodies in flow cytometry and immunofluorescence assays. The ATD fusion protein also contains a Myc tag and a 6XHIS tag, which provide functionality for immunoassays and antigen purification, and a TEV protease site, which allows the ATD domain to be specifically released from the cells in essentially pure form. ATD mobilized from the 293T ATD cell line maintained the pathogenic ANRE epitopes in ELISA binding assays. CSF (3/4) and sera (4/4) from ANRE patients also bound the 293T-ATD cell line, whereas normal CSF and sera did not. CONCLUSIONS: The 293T-ATD cell line is potentially adaptable to a variety of formats to identify antibodies associated with ANRE, including cell-based and soluble antigen formats, and demonstrates a useful method to produce complex proteins for research, drug discovery, and clinical diagnosis.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis/diagnosis , Antibodies, Monoclonal/immunology , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/immunology , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/immunology , Cell Line , Endopeptidases/genetics , Epitopes , HEK293 Cells , Humans , Membrane Proteins/immunology , Recombinant Proteins/immunology
7.
Toxins (Basel) ; 9(5)2017 05 19.
Article in English | MEDLINE | ID: mdl-28534855

ABSTRACT

In the paralytic disease botulism, the botulinum neurotoxin (BoNT) passes through the bloodstream to reach and inactivate neuromuscular junctions. Monoclonal antibodies (mAbs) may be useful BoNT countermeasures, as mAb combinations can rapidly clear BoNT from the blood circulation. We have previously shown that the BoNT-neutralizing potency of mAbs can be improved through red blood cell (RBC) immunoadherence. For example, a fusion protein (FP) that adheres biotinylated mAbs to the RBC surface enabled a pair of mAbs to neutralize 5000 LD50 BoNT/A in the mouse protection assay. Here, we added two mAbs to that combination, creating a 4-mAb:FP complex that neutralized 40,000 LD50 BoNT/A in vivo, and analyzed functional correlates of neutralization. The FP enhanced potency of BoNT/A immune complexes, providing the greatest magnitude of benefit to the 4-mAb combination. RBC binding of a BoNT/A complexed with 4-mAb:FP exhibited a bi-phasic clearance process in vivo. Most of the complexes were cleared within five minutes; the rest were cleared gradually over many hours. Peritoneal macrophages showed better uptake of the 4-mAb complex than the 3-mAb complex, and this was not affected by the presence of the FP. However, the addition of RBCs to the 4-mAb:FP BoNT/A doubled macrophage uptake of the complexes. Lastly, the 4-mAb:FP BoNT/A complex synergistically induced M2 macrophage polarization, as indicated by IL-10 expression, whether or not RBCs were present. RBC-targeted immunoadherence through the FP is a potent enhancer of mAb-mediated BoNT/A neutralization in vivo, and can have positive effects on BoNT/A sequestration, immune complex uptake, and macrophage activation.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antigen-Antibody Complex/immunology , Botulinum Toxins/immunology , Erythrocytes/immunology , Macrophages, Peritoneal/immunology , Animals , Female , Interleukin-10/immunology , Mice , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor-alpha/immunology
8.
Vaccine ; 35(41): 5455-5462, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28343771

ABSTRACT

Following the eradication of wild poliovirus (PV), achieving and maintaining a polio-free status will require eliminating potentially pathogenic PV strains derived from the oral attenuated vaccine. For this purpose, a combination of non-cross-resistant drugs, such as small molecules and neutralizing monoclonal antibodies (mAbs), may be ideal. We previously isolated chimpanzee and human mAbs capable of neutralizing multiple PV types (cross-neutralization). Here, we describe three additional human mAbs that neutralize types 1 and 2 PV and one mAb that neutralizes all three types. Most bind conformational epitopes and have unusually long heavy chain complementarity determining 3 domains (HC CDR3). We assessed the ability of the mAbs to neutralize A12 escape mutant PV strains, and found that the neutralizing activities of the mAbs were disrupted by different amino acid substitutions. Competitive binding studies further suggested that the specific mAb:PV interactions that enable cross-neutralization differ among mAbs and serotypes. All of the cloned mAbs bind PV in the vicinity of the "canyon", a circular depression around the 5-fold axis of symmetry through which PV recognizes its cellular receptor. We were unable to generate escape mutants to two of the mAbs, suggesting that their epitopes are important for the PV life cycle. These data indicate that PV cross-neutralization involves binding to highly conserved structures within the canyon that binds to the cellular receptor. These may be facilitated by the long HC CDR3 domains, which may adopt alternative binding configurations. We propose that the human and chimpanzee mAbs described here could have potential as anti-PV therapeutics.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Poliomyelitis/immunology , Poliovirus/immunology , Adult , Aged , Animals , Antigens, Viral/immunology , Epitopes/immunology , Humans , Middle Aged , Neutralization Tests/methods , Pan troglodytes/immunology , Pan troglodytes/virology , Poliomyelitis/prevention & control , Poliomyelitis/virology , Serogroup
9.
Mol Immunol ; 57(2): 247-54, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24184879

ABSTRACT

Immune complexes formed between monoclonal antibodies (mAbs) and toxins can neutralize toxicity in vivo by multiple mechanisms. Toxin sequestration and clearance by mAbs may be improved by enhancing their ability to bind to red blood cells (RBCs) through immune adherence. This can be achieved by converting the mAbs to heteropolymers (HPs), which are antigen-specific mAbs cross-linked to mAbs targeting the complement receptor (CR1), a protein that is expressed on the surface of RBCs in primates and mediates delivery of complement C3b-containing immune complexes to tissue macrophages. Conversion of mAbs to HPs has been shown to enhance clearance of multivalent antigens from the blood circulation, but the interaction of HPs with monovalent toxins has not been examined. Using botulinum neurotoxin (BoNT) as a model system, we studied the effect of conversion of a pair of BoNT-specific mAbs into HPs on toxin neutralization and handling in vivo. Two HPs given in combination had 166-fold greater potency than un-modified mAbs, neutralizing 5000 LD50 BoNT, when tested in transgenic mice expressing human CR1 on RBC membranes. Improvement required adherence of BoNT to the RBC in vivo and 2 HPs, rather than an HP+mAb pair. The HP pair bound BoNT to RBCs in the circulation for 2h, in comparison to BoNT-neutralizing anti-serum, which induced no detectable RBC binding. HP pairs exhibited enhanced uptake by peritoneal macrophages in vitro, compared to pairs of mAbs or mAb+HP pairs. In a post-exposure therapeutic model, HPs gave complete protection from a lethal BoNT dose up to 3h after toxin exposure. In a pre-exposure prophylaxis model, mice given HP up to 5 days prior to BoNT administration were fully protected from a lethal BoNT dose. These studies elucidate general mechanisms for the neutralization of toxins by HP pairs and demonstrate the potential utility of HPs as BoNT therapeutics.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Botulinum Toxins/immunology , Botulism/prevention & control , Erythrocytes/immunology , Macrophages/immunology , Receptors, Complement/immunology , Animals , Antigen-Antibody Complex/immunology , Botulism/immunology , Humans , Mice , Mice, Transgenic , Receptors, Complement 3b/immunology
10.
Mol Ther Methods Clin Dev ; 1: 14046, 2014.
Article in English | MEDLINE | ID: mdl-26015984

ABSTRACT

Botulinum neurotoxins are one of the most potent toxins found in nature, with broad medical applications from cosmetics to the treatment of various neuropathies. Additionally, these toxins are classified as Category A-Tier 1 agents, with human lethal doses calculated at as little as 90 ng depending upon the route of administration. Of the eight distinct botulinum neurotoxin serotypes, the most common causes of human illness are from serotypes /A, /B, and /E. Protection can be achieved by eliciting antibody responses against the receptor-binding domain of the neurotoxin. Our previous research has shown that recombinant rabies virus-based particles can effectively present heterologous antigens. Here, we describe a novel strategy using recombinant rabies virus particles that elicits a durable humoral immune response against the botulinum neurotoxin receptor binding domains from serotypes /A, /B, and /E. Following intramuscular administration of ß-propiolactone-inactivated rabies virus particles, mice elicited specific immune responses against the cognate antigen. Administration of a combination of these vectors also demonstrated antibody responses against all three serotypes based on enzyme-linked immunosorbent assay (ELISA) measurements, with minimal decay within the study timeline. Complete protection was achieved against toxin challenge from the serotypes /A and /B and partial protection for /E, indicating that a multivalent approach is feasible.

11.
Infect Immun ; 80(12): 4133-42, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22966044

ABSTRACT

Botulinum toxin is a highly potent oral and inhalation poison, which means that the toxin must have an efficient mechanism for penetration of epithelial barriers. To date, three models for toxin passage across epithelial barriers have been proposed: (i) the toxin itself undergoes binding and transcytosis; (ii) an auxiliary protein, HA35, transports toxin from the apical to the basal side of epithelial cells; and (iii) an auxiliary protein, HA35, acts on the basal side of epithelial cells to disrupt tight junctions, and this permits paracellular flux of toxin. These models were evaluated by studying toxin absorption following inhalation exposure in mice. Three types of experiments were conducted. In the first, the potency of pure neurotoxin was compared with that of progenitor toxin complex, which contains HA35. The results showed that the rate and extent of toxin absorption, as well as the potency of absorbed toxin, did not depend upon, nor were they enhanced by, the presence of HA35. In the second type of experiment, the potencies of pure neurotoxin and progenitor toxin complex were compared in the absence or presence of antibodies on the apical side of epithelial cells. Antibodies directed against the neurotoxin protected against challenge, but antibodies against HA35 did not. In the final type of experiment, the potency of pure neurotoxin and toxin complex was compared in animals pretreated to deliver antibodies to the basal side of epithelial cells. Once again, antibodies directed against the neurotoxin provided resistance to challenge, but antibodies directed against HA35 did not. Taken collectively, the data indicate that the toxin by itself is capable of crossing epithelial barriers. The data do not support any hypothesis in which HA35 is essential for toxin penetration of epithelial barriers.


Subject(s)
Botulinum Toxins, Type A/administration & dosage , Botulinum Toxins, Type A/pharmacokinetics , Botulinum Toxins/administration & dosage , Botulinum Toxins/pharmacokinetics , Epithelial Cells/metabolism , Absorption , Administration, Inhalation , Animals , Biological Transport , Botulinum Toxins/chemistry , Botulinum Toxins/poisoning , Botulinum Toxins, Type A/chemistry , Botulinum Toxins, Type A/poisoning , Disease Models, Animal , Female , Mice , Rabbits , Transcytosis
12.
Hybridoma (Larchmt) ; 30(6): 503-10, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22149274

ABSTRACT

Botulinum neurotoxins (BoNTs) are extremely potent toxins that can contaminate foods and are a public health concern. Anti-BoNT antibodies have been described that are capable of detecting BoNTs; however there still exists a need for accurate and sensitive detection capabilities for BoNTs. Herein, we describe the characterization of a panel of eight monoclonal antibodies (MAbs) generated to the non-toxic receptor-binding domain of BoNT/A (H(C)50/A) developed using a high-throughput screening approach. In two independent hybridoma fusions, two groups of four IgG MAbs were developed against recombinant H(C)50/A. Of these eight, only a single MAb, F90G5-3, bound to the whole BoNT/A protein and was characterized further. The F90G5-3 MAb slightly prolonged time to death in an in vivo mouse bioassay and was mapped by pepscan to a peptide epitope in the N-terminal subdomain of H(C)50/A (H(CN)25/A) comprising amino acid residues (985)WTLQDTQEIKQRVVF(999), an epitope that is highly immunoreactive in humans. Furthermore, we demonstrate that F90G5-3 binds BoNT/A with nanomolar efficiency. Together, our results indicate that F90G5-3 is of potential value as a diagnostic immunoreagent for BoNT/A capture assay development and bio-forensic analysis.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Antibody Specificity , Botulinum Toxins, Type A/immunology , Clostridium botulinum type A/immunology , Epitopes/immunology , Amino Acid Sequence , Animals , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibody Affinity , Antigen-Antibody Reactions , Botulinum Toxins, Type A/genetics , Cloning, Molecular , Clostridium botulinum type A/genetics , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , High-Throughput Screening Assays/methods , Humans , Hybridomas/cytology , Hybridomas/immunology , Mice , Molecular Sequence Data , Neutralization Tests , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Time Factors
13.
Vaccine ; 29(28): 4638-45, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21549784

ABSTRACT

We previously showed that rabies virus (RABV) virions are excellent vehicles for antigen presentation. Here, a reverse genetic approach was applied to generate recombinant RABV that express a chimeric protein composed of the heavy chain carboxyterminal half (HC50) of botulinum neurotoxin type A (BoNT/A) and RABV glycoprotein (G). To promote surface expression and incorporation of HC50/A into RABV virions, the RABV glycoprotein (G) ER translocation sequence, various fragments of RABV ectodomain (ED) and cytoplasmic domain were fused to HC50/A. The HC50/A chimeric proteins were expressed on the surface of cells infected with all of the recombinant RABVs, however, the highest level of surface expression was detected by utilizing 30 amino acids of the RABV G ED (HV50/A-E30). Our results also indicated that this chimeric protein was effectively incorporated into RABV virions. Immunization of mice with inactivated RABV-HC50/A-E30 virions induced a robust anti-HC50/A IgG antibody response that efficiently neutralized circulating BoNT/A in vivo, and protected mice against 1000 fold the lethal dose of BoNT/A.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Viral/immunology , Bacterial Vaccines/immunology , Botulinum Toxins, Type A/chemistry , Botulism/prevention & control , Glycoproteins/immunology , Rabies virus/genetics , Viral Envelope Proteins/immunology , Virion/genetics , Animals , Antibodies, Neutralizing/blood , Antigens, Viral/genetics , Antigens, Viral/metabolism , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/immunology , Botulinum Toxins, Type A/metabolism , Botulism/immunology , Female , Genetic Vectors/genetics , Genetic Vectors/immunology , Glycoproteins/genetics , Glycoproteins/metabolism , Immunization , Immunoglobulin G/blood , Mice , Rabies virus/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virion/metabolism
14.
J Pharmacol Exp Ther ; 338(2): 503-17, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21586604

ABSTRACT

Therapeutic antibodies are one of the major classes of medical countermeasures that can provide protection against potential bioweapons such as botulinum toxin. Although a broad array of antibodies are being evaluated for their ability to neutralize the toxin, there is little information that defines the circumstances under which these antibodies can be used. In the present study, an effort was made to quantify the temporal factors that govern therapeutic antibody use in a postchallenge scenario. Experiments were done involving inhalation administration of toxin to mice, intravenous administration to mice, and direct application to murine phrenic nerve-hemidiaphragm preparations. As part of this study, several pharmacokinetic characteristics of botulinum toxin and neutralizing antibodies were measured. The core observation that emerged from the work was that the window of opportunity within which postchallenge administration of antibodies exerted a beneficial effect increased as the challenge dose of toxin decreased. The critical factor in establishing the window of opportunity was the amount of time needed for fractional redistribution of a neuroparalytic quantum of toxin from the extraneuronal space to the intraneuronal space. This redistribution event was a dose-dependent phenomenon. It is likely that the approach used to identify the factors that govern postchallenge efficacy of antibodies against botulinum toxin can be used to assess the factors that govern postchallenge efficacy of medical countermeasures against any agent of bioterrorism or biological warfare.


Subject(s)
Antibodies, Bacterial/therapeutic use , Antibodies, Neutralizing/therapeutic use , Biological Warfare , Botulinum Toxins/toxicity , Animals , Antibodies, Bacterial/pharmacology , Antibodies, Neutralizing/pharmacology , Biological Warfare/prevention & control , Bioterrorism/prevention & control , Botulinum Toxins/antagonists & inhibitors , Dose-Response Relationship, Drug , Female , Mice , Organ Culture Techniques , Phrenic Nerve/drug effects , Phrenic Nerve/physiopathology , Protective Agents/pharmacology , Protective Agents/therapeutic use , Rabbits , Time Factors , Treatment Outcome
15.
PLoS One ; 3(8): e3023, 2008 Aug 20.
Article in English | MEDLINE | ID: mdl-18714390

ABSTRACT

BACKGROUND: Botulinum neurotoxins (BoNT) are a family of category A select bioterror agents and the most potent biological toxins known. Cloned antibody therapeutics hold considerable promise as BoNT therapeutics, but the therapeutic utility of antibodies that bind the BoNT light chain domain (LC), a metalloprotease that functions in the cytosol of cholinergic neurons, has not been thoroughly explored. METHODS AND FINDINGS: We used an optimized hybridoma method to clone a fully human antibody specific for the LC of serotype A BoNT (BoNT/A). The 4LCA antibody demonstrated potent in vivo neutralization when administered alone and collaborated with an antibody specific for the HC. In Neuro-2a neuroblastoma cells, the 4LCA antibody prevented the cleavage of the BoNT/A proteolytic target, SNAP-25. Unlike an antibody specific for the HC, the 4LCA antibody did not block entry of BoNT/A into cultured cells. Instead, it was taken up into synaptic vesicles along with BoNT/A. The 4LCA antibody also directly inhibited BoNT/A catalytic activity in vitro. CONCLUSIONS: An antibody specific for the BoNT/A LC can potently inhibit BoNT/A in vivo and in vitro, using mechanisms not previously associated with BoNT-neutralizing antibodies. Antibodies specific for BoNT LC may be valuable components of an antibody antidote for BoNT exposure.


Subject(s)
Antibodies, Monoclonal , Botulinum Antitoxin/immunology , Botulinum Toxins/antagonists & inhibitors , Botulinum Toxins/immunology , Immunoglobulin Light Chains , Antibody Specificity , Cell Line, Tumor , Cloning, Molecular , Humans , Immunoglobulin Light Chains/genetics , Kinetics , Neuroblastoma , Recombinant Proteins/immunology , Serotyping
16.
Hybridoma (Larchmt) ; 27(2): 65-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18642670

ABSTRACT

Affinity-matured human antibodies have demonstrated efficacy as countermeasures for exposure to botulinum neurotoxin (BoNT), which is the cause of the disease botulism category A select bioterror agent. Little is known, however, about the potential role of natural (un-mutated) antibodies in the protective immune response to BoNT. Here we describe the cloning of two human IgM antibodies that bind serotype A BoNT. Both are un-mutated IgM antibodies, consistent with an origin in naive B cells. One of the antibodies is able to fully neutralize a lethal dose of serotype A BoNT in vivo. These results suggest that the natural human antibody repertoire may play a role in protection from exposure to biological toxins.


Subject(s)
Antibodies, Bacterial/physiology , Botulinum Toxins, Type A/immunology , Botulinum Toxins/immunology , Clostridium botulinum/immunology , Immunoglobulin M/physiology , Amino Acid Sequence , Antibodies, Bacterial/administration & dosage , Antibodies, Bacterial/metabolism , Binding Sites, Antibody , Botulinum Toxins/antagonists & inhibitors , Botulinum Toxins/metabolism , Botulinum Toxins, Type A/antagonists & inhibitors , Botulinum Toxins, Type A/metabolism , Botulism/immunology , Botulism/prevention & control , Cell Line, Tumor , Cells, Cultured , Humans , Hybridomas , Immunoglobulin M/administration & dosage , Immunoglobulin M/metabolism , Molecular Sequence Data
17.
J Pharmacol Exp Ther ; 326(3): 856-63, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18539649

ABSTRACT

The ability of botulinum toxin to poison cholinergic nerve transmission is a dynamic phenomenon that involves not only the actions of the toxin on the body but also the actions of the body on the toxin. The former has been the subject of intense research, whereas the latter has received almost no attention. Therefore, a series of studies were performed to characterize systemic handling of botulinum toxin. The results indicated that the toxin reaches the general circulation (transcytosis across epithelial cells) without obvious changes in structure or biological activity. The general circulation acts as a holding compartment until there is adequate fractional distribution to neuromuscular junctions to produce blockade of transmission. During its transit through this compartment, the toxin 1) undergoes little biotransformation, 2) does not accumulate significantly in circulating cells, and 3) remains largely in the free state. In naive animals, the t(1/2) for toxin in the general circulation is approximately 10 h, and at any given point in time, there is little uptake in nontarget organs (liver, kidney, heart, and lung). In immunized animals, toxin clearance from the general circulation is rapid, and there is substantial accumulation of antibody-antigen complexes in liver. Thus, enhanced clearance from the circulation is a major mechanism by which active immunization can protect against poisoning.


Subject(s)
Blood Circulation/drug effects , Blood Circulation/physiology , Botulinum Toxins/pharmacokinetics , Animals , Biological Transport/drug effects , Biological Transport/physiology , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Protein Binding/drug effects , Protein Binding/physiology , Rabbits , Rats , Rats, Sprague-Dawley
18.
J Immunol Methods ; 333(1-2): 156-66, 2008 Apr 20.
Article in English | MEDLINE | ID: mdl-18313069

ABSTRACT

The affinity-matured human antibody repertoire may be ideal as a source for antibody therapeutics against infectious diseases and bioterror agents. Hybridoma methods for cloning these antibodies have many potential advantages, including convenience, high-yield antibody expression, and the ability to capture the antibodies in their native configurations. However, they have been hindered by hybridoma instability and limited accessibility of antigen-specific, class-switched human B-cells. Here, we describe an efficient, three-step method that uses human peripheral blood B-cells to produce stable hybridoma populations that are highly-enriched for affinity-matured human IgG antibodies. Peripheral blood mononuclear cells (PBMCs) are (a) selected for expression of CD27, a marker of post-germinal center B-cells, (b) cultured in vitro to promote B-cell proliferation and class-switching, and (c) fused to a genetically modified myeloma cell line. Using this strategy, we cloned 5 IgG antibodies that bind botulinum neurotoxins (BoNT), the causes of the food-borne paralytic illness, botulism, and Category A Select Bioterror agents. Two of these antibodies bind BoNT with low picomolar affinities. One (30B) is the first high-affinity human antibody to bind serotype B BoNT, and another (6A) is able to neutralize a lethal dose of serotype A BoNT in vivo in pre- and post-exposure models. This optimized hybridoma method will broadly enable access to the native human antibody repertoire.


Subject(s)
Antibodies, Bacterial/biosynthesis , Botulinum Toxins, Type A/immunology , Botulinum Toxins/immunology , Hybridomas/immunology , Immunoglobulin G/biosynthesis , Animals , Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Antibody Specificity , B-Lymphocytes/immunology , Botulinum Toxins/genetics , Botulinum Toxins, Type A/genetics , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Kinetics , Linear Models , Mice , Neutralization Tests , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , Reverse Transcriptase Polymerase Chain Reaction , Surface Plasmon Resonance , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
19.
Hybridoma (Larchmt) ; 27(1): 11-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18294071

ABSTRACT

Monoclonal antibodies have demonstrated significant potential as therapeutics for botulinum neurotoxin exposures. We previously described a hybridoma method for cloning native human antibodies that uses a murine myeloma cell line that ectopically expresses the human telomerase catalytic subunit gene (hTERT) and the murine interleukin-6 gene (mIL-6). Here we describe a heterohybridoma cell line that ectopically expresses mIL-6 and hTERT and has improved stability of hTERT expression. We fused this cell line to human peripheral blood B cells from a subject who had received the botulinum toxoid vaccine, cloning a high-affinity antibody (13A) specific for serotype A botulinum neurotoxin (BoNT/A). The 13A antibody is an affinity-matured, post-germinal center IgG(1) lambda antibody that has partial neutralization activity in vivo. 13A binds an epitope on BoNT/A that overlaps the binding epitope of an IgG antibody previously shown to fully neutralize a lethal dose of BoNT/A in vivo. The 13A antibody may be useful for diagnostic testing or for incorporation into an oligoclonal therapeutic to counteract BoNT/A exposure.


Subject(s)
Antibodies, Monoclonal/immunology , Botulinum Toxins, Type A/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/metabolism , B-Lymphocytes/immunology , Bacterial Vaccines/immunology , Botulinum Toxins, Type A/metabolism , Cell Fusion , Cell Line, Tumor , Humans , Hybridomas , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Interleukin-6/biosynthesis , Interleukin-6/immunology , Lymphoma, Non-Hodgkin , Mice , Multiple Myeloma , Protein Binding , Telomerase/biosynthesis , Telomerase/immunology
20.
Infect Immun ; 75(6): 3043-54, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17371853

ABSTRACT

Most reports dealing with vaccines against botulinum toxin have focused on the injection route of administration. This is unfortunate, because a mucosal vaccine is likely to be more efficacious for patients and pose fewer risks to health care workers and to the environment. Therefore, efforts were made to generate a mucosal vaccine that provides protection against the botulinum serotypes that typically cause human illness (serotypes A, B, and E). This work demonstrated that carboxy-terminal peptides derived from each of the three serotypes were able to bind to and penetrate human epithelial barriers in vitro, and there was no cross inhibition of membrane binding and transcytosis. The three polypeptides were then tested in vivo as a trivalent vaccine that could be administered to mice by the intranasal route. The results indicated that the mucosal vaccine evoked high secretory titers of immunoglobulin A (IgA), as well as high circulating titers of IgG and IgA, and it also evoked a high level of resistance to challenge with toxin. The immunoglobulin responses and the levels of resistance to challenge were increased by coadministration of adjuvants, such as chitosan and vitamin E. At least three mechanisms were identified to account for the antibody-induced resistance: (i) blockade of toxin absorption across epithelial cells, (ii) enhanced clearance of toxin from the circulation, and (iii) blockade of toxin action at the neuromuscular junction. These results are a compelling demonstration that a mucosal vaccine against multiple serotypes of botulinum toxin has been identified.


Subject(s)
Antibody Formation , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Botulinum Toxins/administration & dosage , Animals , Bacterial Vaccines/chemistry , Botulinum Toxins/genetics , Botulinum Toxins/immunology , Botulinum Toxins, Type A/administration & dosage , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/immunology , Cells, Cultured , Drug Administration Routes , Mice , Mucous Membrane/immunology , Serotyping
SELECTION OF CITATIONS
SEARCH DETAIL
...