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










Publication year range
1.
J AOAC Int ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38775648

ABSTRACT

BACKGROUND: Bovine lactoferrin is increasingly being used as an ingredient in infant formula manufacture to enhance nutritional efficacy through the provision of growth, immunoprotective and antimicrobial factors to the neonate. OBJECTIVE: To evaluate method reproducibility of AOAC 2021.07 Official First Action method for compliance with the performance requirements described in Standard Method Performance Requirement (SMPR®) 2020.005. METHOD: Eight laboratories participated in the analysis of blind-duplicate samples of seven nutritional products. Samples were diluted in buffer, and an optical biosensor immunoassay was used in a direct assay format to quantitate bovine lactoferrin by its interaction with an immobilized anti-lactoferrin antibody. Quantitation was accomplished by the external standard technique with interpolation from a 4-parameter calibration regression. RESULTS: After outliers were removed, precision as reproducibility was found to be within limits set in SMPR 2020.005 (≤ 9%) for six out of seven samples and all had acceptable HorRatR values ranging from 1.0 to 2.1. Additionally, comparison with an alternative independent Stakeholder Panel on Infant Formula and Adult Nutritionals (SPIFAN) First Action method (heparin clean-up LC UV), showed negligible difference between results. CONCLUSIONS: The method described is suitable for the quantification of intact, undenatured bovine lactoferrin in powdered infant formulas. The SPIFAN Expert Review Panel evaluated the method and accompanying validation data from this multi-laboratory testing study in July 2023 and recommended Official Method 2021.07 for adoption as a Final Action Official Method. HIGHLIGHTS: A multi-laboratory validation study of an automated optical biosensor immunoassay for the determination of intact, undenatured bovine lactoferrin is described.

2.
J R Soc Interface ; 20(208): 20230337, 2023 11.
Article in English | MEDLINE | ID: mdl-37935360

ABSTRACT

Red edge excitation shift (REES) spectroscopy relies on the unique emission profiles of fluorophore-solvent interactions to profile protein molecular dynamics. Recently, we reported the use of REES to compare the stability of 32 polymorphic IgG antibodies natively containing tryptophan reporter fluorophores. Here, we expand on this work to investigate the sensitivity of REES to variations in tryptophan content using a subset of IgG3 antibodies containing arginine to tryptophan polymorphisms. Structural analysis revealed that the additional tryptophan residues were situated in highly solvated environments. Subsequently, REES showed clear differences in fluorescence emission profiles when compared with the unmutated variants, thereby limiting direct comparison of their structural dynamics. These findings highlight the exquisite sensitivity of REES to minor variations in protein structure and tryptophan composition.


Subject(s)
Proteins , Tryptophan , Tryptophan/chemistry , Spectrometry, Fluorescence/methods
3.
Clin Exp Immunol ; 214(3): 289-295, 2023 12 13.
Article in English | MEDLINE | ID: mdl-37565297

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for coronavirus disease of 2019 (COVID-19), has caused havoc around the world. While several COVID-19 vaccines and drugs have been authorized for use, these antiviral drugs remain beyond the reach of most low- and middle-income countries. Rapid viral evolution is reducing the efficacy of vaccines and monoclonal antibodies and contributing to the deaths of some fully vaccinated persons. Others with normal immunity may have chosen not to be vaccinated and remain at risk if they contract the infection. Vaccines may not protect some immunodeficient patients from SARS-CoV-2, who are also at increased risk of chronic COVID-19 infection, a dangerous stalemate between the virus and a suboptimal immune response. Intra-host viral evolution could rapidly lead to the selection and dominance of vaccine and monoclonal antibody-resistant clades of SARS-CoV-2. There is thus an urgent need to develop new treatments for COVID-19. The NZACE2-Patari project, comprising modified soluble angiotensin-converting enzyme 2 (ACE2) molecules, seeks to intercept and block SARS-CoV-2 infection of the respiratory mucosa. In vitro data presented here show that soluble wild-type ACE2 molecules retain the ability to effectively block the Spike (S) glycoprotein of SARS-CoV-2 variants including the ancestral Wuhan, delta (B.1.617.2) and omicron (B.1.1.529) strains. This therapeutic strategy may prove effective if implemented early during the nasal phase of the infection and may act synergistically with other antiviral drugs such as Paxlovid to further mitigate disease severity.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Angiotensin-Converting Enzyme 2 , COVID-19 Vaccines , Peptidyl-Dipeptidase A , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Patient Acuity
4.
Biotechnol Rep (Amst) ; 38: e00791, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36915646

ABSTRACT

Antigen-specific polyclonal immunoglobulins derived from the serum, colostrum, or milk of immunized ruminant animals have potential as scalable therapeutics for the control of viral diseases including COVID-19. Here we show that the immunization of sheep with fusions of the SARS-CoV-2 receptor binding domain (RBD) to ovine IgG2a Fc domains promotes significantly higher levels of antigen-specific antibodies compared to native RBD or full-length spike antigens. This antibody population contained elevated levels of neutralizing antibodies that suppressed binding between the RBD and hACE2 receptors in vitro. A second immune-stimulating fusion candidate, Granulocyte-macrophage colony-stimulating factor (GM-CSF), induced high neutralizing responses in select animals but narrowly missed achieving significance. We further demonstrated that the antibodies induced by these fusion antigens were transferred into colostrum/milk and possessed cross-neutralizing activity against diverse SARS-CoV-2 variants. Our findings highlight a new pathway for recombinant antigen design in ruminant animals with applications in immune milk production and animal health.

5.
Protein Sci ; 32(3): e4589, 2023 03.
Article in English | MEDLINE | ID: mdl-36759959

ABSTRACT

The constant regions of clinical monoclonal antibodies are derived from a select number of allotypes found in IgG subclasses. Despite a long-term acknowledgment that this diversity may impact both antibody function and developability, there is a lack of data on the stability of variants carrying these mutations. Here, we generated a panel of IgG1, IgG2, and IgG3 antibodies with 32 unique constant region alleles and performed a systematic comparison of stability using red edge excitation shift (REES). This technique exploits the fluorescent properties of tryptophan residues to measure antibody structural dynamics which predict flexibility and the propensity to unfold. Our REES measurements revealed broad stability differences between subclasses with IgG3 possessing the poorest overall stability. Further interrogation of differences between variants within each subclass enabled the high-resolution profiling of individual allotype stabilities. Crucially, these observed differences were not found to be linked to N297-linked glycan heterogeneity. Our work demonstrates diverse stabilities (and dynamics) for a range of naturally occurring constant domain alleles and the utility of REES as a method for rapid and sensitive antibody stability profiling, requiring only laboratory spectrophotometry equipment.


Subject(s)
Antibodies, Monoclonal , Immunoglobulin G , Immunoglobulin G/chemistry
6.
Front Microbiol ; 14: 1119834, 2023.
Article in English | MEDLINE | ID: mdl-36819065

ABSTRACT

Neisseria gonorrhoeae (also known as gonococcus) has been causing gonorrhoea in humans since ancient Egyptian times. Today, global gonorrhoea infections are rising at an alarming rate, in concert with an increasing number of antimicrobial-resistant strains. The gonococcus has concurrently evolved several intricate mechanisms that promote pathogenesis by evading both host immunity and defeating common therapeutic interventions. Central to these adaptations is the ability of the gonococcus to manipulate various host microenvironments upon infection. For example, the gonococcus can survive within neutrophils through direct regulation of both the oxidative burst response and maturation of the phagosome; a concerning trait given the important role neutrophils have in defending against invading pathogens. Hence, a detailed understanding of how N. gonorrhoeae exploits the human host to establish and maintain infection is crucial for combating this pathogen. This review summarizes the mechanisms behind host manipulation, with a central focus on the exploitation of host epithelial cell signaling to promote colonization and invasion of the epithelial lining, the modulation of the host immune response to evade both innate and adaptive defenses, and the manipulation of host cell death pathways to both assist colonization and combat antimicrobial activities of innate immune cells. Collectively, these pathways act in concert to enable N. gonorrhoeae to colonize and invade a wide array of host tissues, both establishing and disseminating gonococcal infection.

7.
Cell ; 185(21): 4008-4022.e14, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36150393

ABSTRACT

The continual evolution of SARS-CoV-2 and the emergence of variants that show resistance to vaccines and neutralizing antibodies threaten to prolong the COVID-19 pandemic. Selection and emergence of SARS-CoV-2 variants are driven in part by mutations within the viral spike protein and in particular the ACE2 receptor-binding domain (RBD), a primary target site for neutralizing antibodies. Here, we develop deep mutational learning (DML), a machine-learning-guided protein engineering technology, which is used to investigate a massive sequence space of combinatorial mutations, representing billions of RBD variants, by accurately predicting their impact on ACE2 binding and antibody escape. A highly diverse landscape of possible SARS-CoV-2 variants is identified that could emerge from a multitude of evolutionary trajectories. DML may be used for predictive profiling on current and prospective variants, including highly mutated variants such as Omicron, thus guiding the development of therapeutic antibody treatments and vaccines for COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/genetics , Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , Humans , Mutation , Pandemics , Protein Binding , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics
8.
J Anim Sci ; 100(8)2022 Aug 01.
Article in English | MEDLINE | ID: mdl-35678245

ABSTRACT

Passive transfer of colostral immunoglobulins from the cow to the calf is essential for calf health. The objective of this study was to determine if prepartum administration of a vaccine stimulates increased concentrations of colostral immunoglobulins of dairy cows beyond what is explained by vaccine-specific immunoglobulins. A prospective cohort study was conducted on a spring-calving commercial dairy farm that had a policy of only vaccinating cows with even ear tag numbers with a calf diarrhea vaccine, whereas cows with odd ear tag numbers were left unvaccinated. Cows in the vaccinated group (even ear tag numbers, n = 204) received a sensitizer and booster vaccination with a vaccine against bovine rotavirus (serotypes G6 and G10), bovine coronavirus, and E. coli having the K99 pili adherence factor. A sensitizer was given because the study vaccine was different from the vaccine previously used. Cows in the control group (odd ear tag numbers, n = 194) received a 2-mL subcutaneous sterile saline solution. Both groups received two treatments at a 3-wk interval, completing the treatments approximately 2 wk prior to the planned start of calving. During the calving period, technicians separated calves from cows immediately after parturition and prior to suckling, and cows were completely milked out within 6 h of parturition. Vaccine-specific, total, and nonvaccine-specific (total minus vaccine-specific) concentrations of immunoglobulin classes A, G1, G2a, and M (IgA, IgG1, IgG2a, and IgM, respectively) were quantified by mass spectrometry for 20 colostrum samples from each treatment group. Predicted mean non-vaccine-specific colostral IgM concentrations were 8.76 (95% CI = 7.18-10.67) and 5.78 (95% CI = 4.74-7.05) mg/mL for vaccinated and control cows, respectively (P = 0.005). Predicted mean non-vaccine-specific colostral IgG1 concentrations were 106.08 (95% CI = 92.07-120.08) and 95.30 (95% CI = 81.30-109.31) mg/mL among vaccinated and control cows, respectively; however, these means were not significantly different (P = 0.278). It is thus possible that the vaccine, in addition to specifically managing infectious calf diarrhea, may also have non-specific benefits by improving colostrum quality through increased non-vaccine-specific colostrum IgM concentrations. Further research is necessary to determine the mechanism for these preliminary findings, whether the effect may occur in other immunoglobulin classes, and what impacts it may have on calf health outcomes.


Unlike human babies, calves do not receive protective immune proteins (immunoglobulins) from the mother before birth, so a sufficient volume of immunoglobulin-rich colostrum of adequate quality must be consumed within hours of birth. It can be a challenge to meet this requirement for all dairy calves. Prior to calving, cows can be vaccinated with a vaccine against specific infectious causes of calf diarrhea to stimulate elevated concentrations of specific immunoglobulins in their colostrum, which is consumed by their calves to protect them until their own immune systems develop. We enrolled cows that were either vaccinated or not with a calf diarrhea vaccine and, using novel laboratory techniques, measured concentrations of immunoglobulin classes A, G, and M in their colostrum. As expected, vaccinated cows had elevated concentrations of vaccine-specific immunoglobulins in their colostrum. However, they also had elevated non-vaccine-specific concentrations of immunoglobulin M. The vaccine may therefore have stimulated a nonspecific increase in colostral immunoglobulin M concentrations. Further research is necessary to confirm the preliminary findings of the present study and determine the mechanism for this apparent nonspecific increase in colostral immunoglobulin M concentrations, whether it may occur in other immunoglobulin classes, and whether it may benefit calf health and growth.


Subject(s)
Colostrum , Vaccines , Animals , Animals, Newborn , Cattle , Colostrum/chemistry , Diarrhea/prevention & control , Diarrhea/veterinary , Escherichia coli , Female , Humans , Immunoglobulin G/analysis , Immunoglobulin M , Pregnancy , Prospective Studies
9.
Trends Pharmacol Sci ; 43(2): 123-135, 2022 02.
Article in English | MEDLINE | ID: mdl-34895944

ABSTRACT

The biophysical and functional properties of monoclonal antibody (mAb) drug candidates are often improved by protein engineering methods to increase the probability of clinical efficacy. One emerging method is deep mutational scanning (DMS) which combines the power of exhaustive protein mutagenesis and functional screening with deep sequencing and bioinformatics. The application of DMS has yielded significant improvements to the affinity, specificity, and stability of several preclinical antibodies alongside novel applications such as introducing multi-specific binding properties. DMS has also been applied directly on target antigens to precisely map antibody-binding epitopes and notably to profile the mutational escape potential of viral targets (e.g., SARS-CoV-2 variants). Finally, DMS combined with machine learning is enabling advances in the computational screening and engineering of therapeutic antibodies.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Viral , Humans , Spike Glycoprotein, Coronavirus
10.
Front Immunol ; 11: 2016, 2020.
Article in English | MEDLINE | ID: mdl-32973808

ABSTRACT

Polymorphic diversity in antibody constant domains has long been defined by allotypic motifs that cross react with the sera of other individuals. Improvements in sequencing technologies have led to the discovery of a large number of new allelic sequences that underlie this diversity. Many of the point mutations lie outside traditional allotypic motifs suggesting they do not elicit immunogenic responses. As antibodies play an important role in immune defense and biotechnology, understanding how this newly resolved diversity influences the function of antibodies is important. This review investigates the current known diversity of antibody alleles at a protein level for each antibody isotype as well as the kappa and lambda light chains. We focus on evidence emerging for how these mutations perturb antibody interactions with antigens and Fc receptors that are critical for function, as well as the influence this might have on the use of antibodies as therapeutics and reagents.


Subject(s)
Immunoglobulin Allotypes/immunology , Immunoglobulin Constant Regions/genetics , Alleles , Animals , Antibody Formation , Antibody-Dependent Cell Cytotoxicity , Cross Reactions , Genetic Variation , Humans , Receptors, Fc/metabolism
11.
Front Immunol ; 10: 2630, 2019.
Article in English | MEDLINE | ID: mdl-31798579

ABSTRACT

Immune cell therapies based on the integration of synthetic antigen receptors comprise a powerful strategy for the treatment of diverse diseases, most notably T cells engineered to express chimeric antigen receptors (CAR) for targeted cancer therapy. In addition to T lymphocytes, B lymphocytes may also represent valuable immune cells that can be engineered for therapeutic purposes such as protein replacement therapy or recombinant antibody production. In this article, we report a promising concept for the molecular design, optimization, and genomic integration of a novel class of synthetic antigen receptors, chimeric B cell receptors (CBCR). We initially optimized CBCR expression and detection by modifying the extracellular surface tag, the transmembrane regions and intracellular signaling domains. For this purpose, we stably integrated a series of CBCR variants using CRISPR-Cas9 into immortalized B cell hybridomas. Subsequently, we developed a reliable and consistent pipeline to precisely introduce cassettes of several kb size into the genome of primary murine B cells also using CRISPR-Cas9 induced HDR. Finally, we were able to show the robust surface expression and antigen recognition of a synthetic CBCR in primary B cells. We anticipate CBCRs and our approach for engineering primary B cells will be a valuable tool for the advancement of future B cell- based immune cell therapies.


Subject(s)
B-Lymphocytes , Gene Editing/methods , Protein Engineering/methods , Receptors, Antigen, B-Cell/genetics , Receptors, Artificial/genetics , Animals , CRISPR-Cas Systems , Mice , Receptors, Antigen, B-Cell/immunology , Receptors, Artificial/immunology
12.
Nucleic Acids Res ; 46(14): 7436-7449, 2018 08 21.
Article in English | MEDLINE | ID: mdl-29931269

ABSTRACT

Antibody engineering is often performed to improve therapeutic properties by directed evolution, usually by high-throughput screening of phage or yeast display libraries. Engineering antibodies in mammalian cells offer advantages associated with expression in their final therapeutic format (full-length glycosylated IgG); however, the inability to express large and diverse libraries severely limits their potential throughput. To address this limitation, we have developed homology-directed mutagenesis (HDM), a novel method which extends the concept of CRISPR/Cas9-mediated homology-directed repair (HDR). HDM leverages oligonucleotides with degenerate codons to generate site-directed mutagenesis libraries in mammalian cells. By improving HDR to a robust efficiency of 15-35% and combining mammalian display screening with next-generation sequencing, we validated this approach can be used for key applications in antibody engineering at high-throughput: rational library construction, novel variant discovery, affinity maturation and deep mutational scanning (DMS). We anticipate that HDM will be a valuable tool for engineering and optimizing antibodies in mammalian cells, and eventually enable directed evolution of other complex proteins and cellular therapeutics.


Subject(s)
Antibodies/immunology , CRISPR-Cas Systems , Mutagenesis, Site-Directed , Protein Engineering/methods , Amino Acid Sequence , Animals , Antibodies/genetics , Antibodies/metabolism , Antibody Affinity/genetics , Antibody Affinity/immunology , Base Sequence , Cell Line , DNA Breaks, Double-Stranded , High-Throughput Nucleotide Sequencing/methods , Humans , Hybridomas , Oligonucleotides/genetics , Oligonucleotides/metabolism , Recombinational DNA Repair
13.
Sci Rep ; 7: 45775, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28374766

ABSTRACT

The development of programmable nucleases has enabled the application of new genome engineering strategies for cellular immunotherapy. While targeted nucleases have mostly been used to knock-out or knock-in genes in immune cells, the scarless exchange of entire immunogenomic alleles would be of great interest. In particular, reprogramming the polymorphic MHC locus could enable the creation of matched donors for allogeneic cellular transplantation. Here we show a proof-of-concept for reprogramming MHC-specificity by performing CRISPR-Cas9-assisted cassette exchange. Using murine antigen presenting cell lines (RAW264.7 macrophages), we demonstrate that the generation of Cas9-induced double-stranded breaks flanking the native MHC-I H2-Kd locus led to exchange of an orthogonal H2-Kb allele. MHC surface expression allowed for easy selection of reprogrammed cells by flow cytometry, thus obviating the need for additional selection markers. MHC-reprogrammed cells were fully functional as they could present H2-Kd-restricted peptide and activate cognate T cells. Finally, we investigated the role of various donor template formats on exchange efficiency, discovering that templates that underwent in situ linearization resulted in the highest MHC-reprogramming efficiency. These findings highlight a potential new approach for the correcting of MHC mismatches in cellular transplantation.


Subject(s)
CRISPR-Cas Systems , Major Histocompatibility Complex/genetics , Animals , Cell Line , Cell Line, Tumor , DNA Breaks, Double-Stranded , Fibroblasts , Flow Cytometry , H-2 Antigens/genetics , H-2 Antigens/immunology , Macrophages/immunology , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred BALB C
14.
Nat Commun ; 7: 12535, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27531490

ABSTRACT

Hybridomas, fusions of primary mouse B cells and myelomas, are stable, rapidly-proliferating cell lines widely utilized for antibody screening and production. Antibody specificity of a hybridoma clone is determined by the immunoglobulin sequence of the primary B cell. Here we report a platform for rapid reprogramming of hybridoma antibody specificity by immunogenomic engineering. Here we use CRISPR-Cas9 to generate double-stranded breaks in immunoglobulin loci, enabling deletion of the native variable light chain and replacement of the endogenous variable heavy chain with a fluorescent reporter protein (mRuby). New antibody genes are introduced by Cas9-targeting of mRuby for replacement with a donor construct encoding a light chain and a variable heavy chain, resulting in full-length antibody expression. Since hybridomas surface express and secrete antibodies, reprogrammed cells are isolated using flow cytometry and cell culture supernatant is used for antibody production. Plug-and-(dis)play hybridomas can be reprogrammed with only a single transfection and screening step.


Subject(s)
Genetic Engineering/methods , Hybridomas/immunology , Animals , Antibodies/metabolism , Antibody Specificity , Antigens/metabolism , Cell Line , Gene Editing , Mice , Reproducibility of Results
15.
Chimia (Aarau) ; 70(6): 439-42, 2016.
Article in English | MEDLINE | ID: mdl-27363374

ABSTRACT

The simplicity of the CRISPR/Cas9 technology has been transformative in making targeted genome editing accessible for laboratories around the world. However, due to the sheer volume of literature generated in the past five years, determining the best format and delivery method of CRISPR/Cas9 components can be challenging. Here, we provide a brief overview of the progress that has been made in the ex vivo genome editing of mammalian cells and summarize the key advances made for improving efficiency and delivery of CRISPR/Cas9 in DNA, RNA, and protein form. In particular, we highlight the delivery of Cas9 components to human cells for advanced genome editing applications such as large gene insertion.


Subject(s)
Cell Engineering , Clustered Regularly Interspaced Short Palindromic Repeats , Animals , DNA/genetics , In Vitro Techniques , Mammals , RNA, Messenger/genetics , Ribonucleoproteins/administration & dosage
16.
Chem Biol ; 21(12): 1603-9, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25500223

ABSTRACT

All clinically approved antibodies are of the IgG isotype and mediate the clearance of target cells via binding to Fcγ receptors and complement (C1q). Even though IgA can elicit powerful cytotoxic action via FcαRI receptor binding, IgA antibodies have not been amenable to therapeutic development. Here, we report the engineering of a "cross-isotype" antibody, IgGA, which combines the effector functions of both IgG and IgA. IgGA binds to FcαRI with an affinity comparable to that of IgA, and to the activating Fcγ receptors, FcγRI and FcγRIIa, with high affinity, and displays increased binding to C1q compared to IgG. Unlike trastuzumab-IgG, trastuzumab-IgGA potently activates both neutrophils and macrophages to kill Her2(+) cancer cells. Furthermore, IgGA mediates greater complement-dependent cytotoxicity than IgG1 or IgA antibodies. The multitude of IgGA effector functions could be important for therapeutic purposes and highlights the concept of engineering antibodies that combine effector functions from multiple antibody isotypes.


Subject(s)
Cross Reactions , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Protein Engineering , Receptors, Fc/metabolism , Amino Acid Sequence , Cell Line, Tumor , Complement System Proteins/metabolism , Humans , Immunoglobulin A/chemistry , Immunoglobulin A/genetics , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Models, Molecular , Molecular Sequence Data , Neutrophils/immunology , Protein Structure, Tertiary , Receptors, IgG/metabolism
17.
Blood ; 124(22): 3241-9, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25232058

ABSTRACT

The efficacy of most therapeutic monoclonal antibodies (mAbs) targeting tumor antigens results primarily from their ability to elicit potent cytotoxicity through effector-mediated functions. We have engineered the fragment crystallizable (Fc) region of the immunoglobulin G (IgG) mAb, HuM195, targeting the leukemic antigen CD33, by introducing the triple mutation Ser293Asp/Ala330Leu/Ile332Glu (DLE), and developed Time-lapse Imaging Microscopy in Nanowell Grids to analyze antibody-dependent cell-mediated cytotoxicity kinetics of thousands of individual natural killer (NK) cells and mAb-coated target cells. We demonstrate that the DLE-HuM195 antibody increases both the quality and the quantity of NK cell-mediated antibody-dependent cytotoxicity by endowing more NK cells to participate in cytotoxicity via accrued CD16-mediated signaling and by increasing serial killing of target cells. NK cells encountering targets coated with DLE-HuM195 induce rapid target cell apoptosis by promoting simultaneous conjugates to multiple target cells and induce apoptosis in twice the number of target cells within the same period as the wild-type mAb. Enhanced target killing was also associated with increased frequency of NK cells undergoing apoptosis, but this effect was donor-dependent. Antibody-based therapies targeting tumor antigens will benefit from a better understanding of cell-mediated tumor elimination, and our work opens further opportunities for the therapeutic targeting of CD33 in the treatment of acute myeloid leukemia.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Killer Cells, Natural/immunology , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Genetic Engineering , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Mutagenesis , Primary Cell Culture , Time-Lapse Imaging
18.
Proc Natl Acad Sci U S A ; 111(13): 4928-33, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24639495

ABSTRACT

The adaptive immune system confers protection by generating a diverse repertoire of antibody receptors that are rapidly expanded and contracted in response to specific targets. Next-generation DNA sequencing now provides the opportunity to survey this complex and vast repertoire. In the present work, we describe a set of tools for the analysis of antibody repertoires and their application to elucidating the dynamics of the response to viral vaccination in human volunteers. By analyzing data from 38 separate blood samples across 2 y, we found that the use of the germ-line library of V and J segments is conserved between individuals over time. Surprisingly, there appeared to be no correlation between the use level of a particular VJ combination and degree of expansion. We found the antibody RNA repertoire in each volunteer to be highly dynamic, with each individual displaying qualitatively different response dynamics. By using combinatorial phage display, we screened selected VH genes paired with their corresponding VL library for affinity against the vaccine antigens. Altogether, this work presents an additional set of tools for profiling the human antibody repertoire and demonstrates characterization of the fast repertoire dynamics through time in multiple individuals responding to an immune challenge.


Subject(s)
Antibodies/immunology , Immunity/immunology , Viral Vaccines/immunology , Clone Cells , Genetic Vectors , Healthy Volunteers , Humans , Immunoglobulin Variable Region/genetics , Male , Mutation/genetics , Reproducibility of Results , Time Factors , V(D)J Recombination/genetics , Vaccination
19.
ACS Chem Biol ; 8(2): 368-75, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23030766

ABSTRACT

Glycans anchored to residue N297 of the antibody IgG Fc domain are critical in mediating binding toward FcγRs to direct both adaptive and innate immune responses. However, using a full length bacterial IgG display system, we have isolated aglycosylated Fc domains with mutations that confer up to a 160-fold increase in the affinity toward the low affinity FcγRIIa-R131 allele as well as high selectivity against binding to the remarkably homologous human inhibitory receptor, FcγRIIb. The mutant Fc domain (AglycoT-Fc1004) contained a total of 5 amino acid substitutions that conferred an activating to inhibitory ratio of 25 (A/I ratio; FcyRIIa-R131:FcγRIIb). Incorporation of this engineered Fc into trastuzumab, an anti-Her2 antibody, resulted in a 75% increase in tumor cell phagocytosis by macrophages compared to that of the parental glycosylated trastuzumab with both medium and low Her2-expressing cancer cells. A mathematical model has been developed to help explain how receptor affinity and the A/I ratio relate to improved antibody dependent cell-mediated phagocytosis. Our model provides guidelines for the future engineering of Fc domains with enhanced effector function.


Subject(s)
Immunoglobulin G/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Phagocytosis , Protein Engineering , Receptor, ErbB-2/metabolism , Receptors, IgG/metabolism , Cell Line, Tumor , Glycosylation , Humans , Substrate Specificity
20.
Curr Opin Biotechnol ; 22(6): 858-67, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21420850

ABSTRACT

In recent years a number of aglycosylated therapeutic antibodies have entered the clinic. The clinical evaluation of these antibodies has served to dispel concerns that the absence of the ubiquitous N297 glycan in the Fc of IgG might result in immunogenicity, poor in vivo stability or unfavorable pharmacokinetics. Importantly, recent studies have now demonstrated that aglycosylated antibodies can be engineered to display novel effector functions and mechanisms of action that do not appear to be possible with their glycosylated counterparts. Moreover, the ability to manufacture aglycosylated antibodies in lower eukaryotes or in bacteria provides significant bioprocessing advantages in terms of shorter bioprocess development and running times and by completely bypassing the problems associated with the glycan heterogeneity of conventional antibodies. These advantages are poised to catapult aglycosylated antibodies to the forefront of protein therapeutics.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/metabolism , Biotechnology , Immunoglobulin G/metabolism , Immunoglobulin G/therapeutic use , Immunotherapy , Animals , Antibodies, Monoclonal/therapeutic use , Bacteria/genetics , Glycosylation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...