Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Infect Dis ; 228(5): 555-563, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37062677

RESUMEN

Emerging variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) possess mutations that prevent antibody therapeutics from maintaining antiviral binding and neutralizing efficacy. Monoclonal antibodies (mAbs) shown to neutralize Wuhan-Hu-1 SARS-CoV-2 (ancestral) strain have reduced potency against newer variants. Plasma-derived polyclonal hyperimmune drugs have improved neutralization breadth compared with mAbs, but lower titers against SARS-CoV-2 require higher dosages for treatment. We previously developed a highly diverse, recombinant polyclonal antibody therapeutic anti-SARS-CoV-2 immunoglobulin hyperimmune (rCIG). rCIG was compared with plasma-derived or mAb standards and showed improved neutralization of SARS-CoV-2 across World Health Organization variants; however, its potency was reduced against some variants relative to ancestral, particularly omicron. Omicron-specific antibody sequences were enriched from yeast expressing rCIG-scFv and exhibited increased binding and neutralization to omicron BA.2 while maintaining ancestral strain binding and neutralization. Polyclonal antibody libraries such as rCIG can be utilized to develop antibody therapeutics against present and future SARS-CoV-2 threats.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2/genética , Anticuerpos Monoclonales/uso terapéutico , Antivirales , Saccharomyces cerevisiae , Anticuerpos Neutralizantes/uso terapéutico , Glicoproteína de la Espiga del Coronavirus/genética , Anticuerpos Antivirales/uso terapéutico
2.
Pathogens ; 11(7)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35890050

RESUMEN

Conventionally, hyperimmune globulin drugs manufactured from pooled immunoglobulins from vaccinated or convalescent donors have been used in treating infections where no treatment is available. This is especially important where multi-epitope neutralization is required to prevent the development of immune-evading viral mutants that can emerge upon treatment with monoclonal antibodies. Using microfluidics, flow sorting, and a targeted integration cell line, a first-in-class recombinant hyperimmune globulin therapeutic against SARS-CoV-2 (GIGA-2050) was generated. Using processes similar to conventional monoclonal antibody manufacturing, GIGA-2050, comprising 12,500 antibodies, was scaled-up for clinical manufacturing and multiple development/tox lots were assessed for consistency. Antibody sequence diversity, cell growth, productivity, and product quality were assessed across different manufacturing sites and production scales. GIGA-2050 was purified and tested for good laboratory procedures (GLP) toxicology, pharmacokinetics, and in vivo efficacy against natural SARS-CoV-2 infection in mice. The GIGA-2050 master cell bank was highly stable, producing material at consistent yield and product quality up to >70 generations. Good manufacturing practices (GMP) and development batches of GIGA-2050 showed consistent product quality, impurity clearance, potency, and protection in an in vivo efficacy model. Nonhuman primate toxicology and pharmacokinetics studies suggest that GIGA-2050 is safe and has a half-life similar to other recombinant human IgG1 antibodies. These results supported a successful investigational new drug application for GIGA-2050. This study demonstrates that a new class of drugs, recombinant hyperimmune globulins, can be manufactured consistently at the clinical scale and presents a new approach to treating infectious diseases that targets multiple epitopes of a virus.

3.
Nat Biotechnol ; 39(8): 989-999, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33859400

RESUMEN

Plasma-derived polyclonal antibody therapeutics, such as intravenous immunoglobulin, have multiple drawbacks, including low potency, impurities, insufficient supply and batch-to-batch variation. Here we describe a microfluidics and molecular genomics strategy for capturing diverse mammalian antibody repertoires to create recombinant multivalent hyperimmune globulins. Our method generates of diverse mixtures of thousands of recombinant antibodies, enriched for specificity and activity against therapeutic targets. Each hyperimmune globulin product comprised thousands to tens of thousands of antibodies derived from convalescent or vaccinated human donors or from immunized mice. Using this approach, we generated hyperimmune globulins with potent neutralizing activity against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in under 3 months, Fc-engineered hyperimmune globulins specific for Zika virus that lacked antibody-dependent enhancement of disease, and hyperimmune globulins specific for lung pathogens present in patients with primary immune deficiency. To address the limitations of rabbit-derived anti-thymocyte globulin, we generated a recombinant human version and demonstrated its efficacy in mice against graft-versus-host disease.


Asunto(s)
Linfocitos B/inmunología , COVID-19/terapia , Globulinas/biosíntesis , SARS-CoV-2/inmunología , Animales , Anticuerpos Antivirales/inmunología , Células CHO , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Globulinas/inmunología , Humanos , Inmunización Pasiva , Ratones , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Virus Zika/inmunología , Sueroterapia para COVID-19
4.
MAbs ; 12(1): 1803646, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32744131

RESUMEN

IN VITRO: affinity maturation of therapeutic monoclonal antibodies is commonly applied to achieve desired properties, such as improved binding kinetics and affinity. Currently there are no universally accepted protocols for generation of variegated antibody libraries or selection thereof. Here, we performed affinity maturation using a yeast-based single-chain variable fragment (scFv) expression system to compare two mutagenesis methods: random mutagenesis across the entire V(D)J region by error-prone PCR, and a novel combinatorial mutagenesis process limited to the complementarity-determining regions (CDRs). We applied both methods of mutagenesis to four human antibodies against well-known immuno-oncology target proteins. Detailed sequence analysis showed an even mutational distribution across the entire length of the scFv for the error-prone PCR method and an almost exclusive targeting of the CDRs for the combinatorial method. Though there were distinct mutagenesis profiles for each target antibody and mutagenesis method, we found that both methods improved scFv affinity with similar efficiency. When a subset of the affinity-matured antibodies was expressed as full-length immunoglobulin, the measured affinity constants were mostly comparable to those of the respective scFv, but the full-length antibodies were inferior to their scFv counterparts for one of the targets. Furthermore, we found that improved affinity for the full-length antibody did not always translate into enhanced binding to cell-surface expressed antigen or improved immune checkpoint blocking ability, suggesting that screening with full-length antibody or antigen-binding fragment formats might be advantageous and the subject of a future study.


Asunto(s)
Afinidad de Anticuerpos/genética , Mutagénesis , Anticuerpos de Cadena Única , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/genética , Humanos , Reacción en Cadena de la Polimerasa , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética
5.
Nat Biotechnol ; 38(5): 609-619, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32393905

RESUMEN

T cells engineered to express antigen-specific T cell receptors (TCRs) are potent therapies for viral infections and cancer. However, efficient identification of clinical candidate TCRs is complicated by the size and complexity of T cell repertoires and the challenges of working with primary T cells. Here we present a high-throughput method to identify TCRs with high functional avidity from diverse human T cell repertoires. The approach used massively parallel microfluidics to generate libraries of natively paired, full-length TCRαß clones, from millions of primary T cells, which were then expressed in Jurkat cells. The TCRαß-Jurkat libraries enabled repeated screening and panning for antigen-reactive TCRs using peptide major histocompatibility complex binding and cellular activation. We captured more than 2.9 million natively paired TCRαß clonotypes from six healthy human donors and identified rare (<0.001% frequency) viral-antigen-reactive TCRs. We also mined a tumor-infiltrating lymphocyte sample from a patient with melanoma and identified several tumor-specific TCRs, which, after expression in primary T cells, led to tumor cell killing.


Asunto(s)
Antígenos/análisis , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T/citología , Ingeniería Celular , Biblioteca de Genes , Humanos , Células Jurkat , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Linfocitos T/inmunología , Virus/inmunología
6.
J Biol Chem ; 294(15): 5790-5804, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30796163

RESUMEN

T-cell receptors (TCR) have considerable potential as therapeutics and antibody-like reagents to monitor disease progression and vaccine efficacy. Whereas antibodies recognize only secreted and surface-bound proteins, TCRs recognize otherwise inaccessible disease-associated intracellular proteins when they are presented as processed peptides bound to major histocompatibility complexes (pMHC). TCRs have been primarily explored for cancer therapy applications but could also target infectious diseases such as cytomegalovirus (CMV). However, TCRs are more difficult to express and engineer than antibodies, and advanced methods are needed to enable their widespread use. Here, we engineered the human CMV-specific TCR RA14 for high-affinity and robust soluble expression. To achieve this, we adapted our previously reported mammalian display system to present TCR extracellular domains and used this to screen CDR3 libraries for clones with increased pMHC affinity. After three rounds of selection, characterized clones retained peptide specificity and activation when expressed on the surface of human Jurkat T cells. We obtained high yields of soluble, monomeric protein by fusing the TCR extracellular domains to antibody hinge and Fc constant regions, adding a stabilizing disulfide bond between the constant domains and disrupting predicted glycosylation sites. One variant exhibited 50 nm affinity for its cognate pMHC, as measured by surface plasmon resonance, and specifically stained cells presenting this pMHC. Our work has identified a human TCR with high affinity for the immunodominant CMV peptide and offers a new strategy to rapidly engineer soluble TCRs for biomedical applications.


Asunto(s)
Citomegalovirus/inmunología , Expresión Génica , Biblioteca de Genes , Ingeniería de Proteínas , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Células CHO , Cricetulus , Citomegalovirus/genética , Humanos , Regiones Constantes de Inmunoglobulina/genética , Regiones Constantes de Inmunoglobulina/inmunología , Células Jurkat , Ratones , Dominios Proteicos , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Solubilidad
7.
Curr Opin Chem Eng ; 19: 131-141, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29911002

RESUMEN

Serum therapy fell out of favor 80 years ago, but antibodies against infectious diseases are now experiencing a renaissance. With the evolution of antibiotic-resistant bacteria, the emergence of new pathogens, and a growing population of immunocompromised individuals coupled with improvements in antibody manufacturing and biological efficacy, antibodies are an increasingly attractive therapeutic option. In this review, we highlight successful clinical strategies and discuss recent applications of advanced antibody engineering approaches to combat infectious diseases. Case studies include antibody mixtures to neutralize Staphylococcus aureus; bispecific antibodies promoting Pseudomonas aeruginosa clearance; antibody-antibiotic conjugates to eradicate S. aureus from protected intracellular niches; and novel anti-RSV antibodies with extended serum half-life. These new designs are powerful strategies for targeting infectious diseases due to their abilities to target multiple antigens and induce novel clearance mechanisms.

8.
J Vet Med Sci ; 79(1): 60-64, 2017 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-27666464

RESUMEN

The baboon model of Bordetella pertussis infection is the newest and most clinically accurate model of the human disease to date. However, among the 15 experimentally infected baboons in this study, a subset of baboons did not exhibit the expected high bacterial colonization levels or increase in white blood cell count. Moreover, cultures of nasopharyngeal wash samples from several baboons suggested B. bronchiseptica coinfection. Analysis of serum antibodies recognizing filamentous hemagglutinin, pertussis toxin and B. pertussis lipo-oligosaccharide indicated that several baboons had likely been previously exposed to Bordetella species and that prior exposure correlated with partial protection from B. pertussis infection. Notably, all animals with a baseline Fha titer of 5 IU/ml or below exhibited symptoms typical of the model, suggesting this value can be used as inclusion criteria for animals prior to study enrollment. While B. pertussis infection is endemic to human populations and B. bronchiseptica is common in wild small mammals, this study illustrates that baboons can readily harbor both organisms. Awareness of Bordetella species that share antigens capable of generating protective immune responses and tracking of prior exposure to those species is required for successful use of the baboon model of pertussis.


Asunto(s)
Infecciones por Bordetella/inmunología , Bordetella bronchiseptica/inmunología , Bordetella pertussis/inmunología , Tos Ferina/inmunología , Adhesinas Bacterianas/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Infecciones por Bordetella/microbiología , Bordetella bronchiseptica/aislamiento & purificación , Coinfección , Modelos Animales de Enfermedad , Papio , Tos Ferina/microbiología
9.
Clin Vaccine Immunol ; 23(11): 851-862, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27581436

RESUMEN

Bispecific antibodies are a rapidly growing class of therapeutic molecules, originally developed for the treatment of cancer but recently explored for the treatment of autoimmune and infectious diseases. Bordetella pertussis is a reemerging pathogen, and several of the key symptoms of infection are caused by the pertussis toxin (PTx). Two humanized antibodies, hu1B7 and hu11E6, bind distinct epitopes on PTx and, when coadministered, mitigate disease severity in murine and baboon models of infection. Here we describe the generation of a bispecific human IgG1 molecule combining the hu1B7 and hu11E6 binding sites via a knobs-in-holes design. The bispecific antibody showed binding activity equivalent to that of the antibody mixture in a competition enzyme-linked immunosorbent assay (ELISA). A CHO cell neutralization assay provided preliminary evidence for synergy between the two antibodies, while a murine model of PTx-induced leukocytosis definitively showed synergistic neutralization. Notably, the bispecific antibody retained the synergy observed for the antibody mixture, supporting the conclusion that synergy is due to simultaneous blockade of both the catalytic and receptor binding activities of pertussis toxin. These data suggest that a hu1B7/hu11E6 bispecific antibody is a viable alternative to an antibody mixture for pertussis treatment.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales Humanizados/inmunología , Epítopos/inmunología , Inmunoglobulina G/inmunología , Toxina del Pertussis/inmunología , Tos Ferina/terapia , Animales , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/metabolismo , Anticuerpos Monoclonales Humanizados/uso terapéutico , Sitios de Unión de Anticuerpos , Bordetella pertussis/inmunología , Células CHO , Cricetinae , Cricetulus , Sinergismo Farmacológico , Ensayo de Inmunoadsorción Enzimática , Epítopos/química , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/aislamiento & purificación , Inmunoglobulina G/metabolismo , Ratones , Pruebas de Neutralización , Tos Ferina/inmunología , Tos Ferina/prevención & control
10.
Sci Transl Med ; 7(316): 316ra195, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26631634

RESUMEN

Despite widespread vaccination, pertussis rates are rising in industrialized countries and remain high worldwide. With no specific therapeutics to treat disease, pertussis continues to cause considerable infant morbidity and mortality. The pertussis toxin is a major contributor to disease, responsible for local and systemic effects including leukocytosis and immunosuppression. We humanized two murine monoclonal antibodies that neutralize pertussis toxin and expressed them as human immunoglobulin G1 molecules with no loss of affinity or in vitro neutralization activity. When administered prophylactically to mice as a binary cocktail, antibody treatment completely mitigated the Bordetella pertussis-induced rise in white blood cell counts and decreased bacterial colonization. When administered therapeutically to baboons, antibody-treated, but not untreated control animals, experienced a blunted rise in white blood cell counts and accelerated bacterial clearance rates. These preliminary findings support further investigation into the use of these antibodies to treat human neonatal pertussis in conjunction with antibiotics and supportive care.


Asunto(s)
Anticuerpos Monoclonales Humanizados/química , Toxina del Pertussis/química , Tos Ferina/terapia , Animales , Bordetella pertussis , Células CHO , Cricetulus , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulina G/química , Región Variable de Inmunoglobulina , Lactante , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Papio , Pronóstico , Vacunación
11.
J Biol Chem ; 290(44): 26457-70, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26296891

RESUMEN

Knowledge of the fine location of neutralizing and non-neutralizing epitopes on human pathogens affords a better understanding of the structural basis of antibody efficacy, which will expedite rational design of vaccines, prophylactics, and therapeutics. However, full utilization of the wealth of information from single cell techniques and antibody repertoire sequencing awaits the development of a high throughput, inexpensive method to map the conformational epitopes for antibody-antigen interactions. Here we show such an approach that combines comprehensive mutagenesis, cell surface display, and DNA deep sequencing. We develop analytical equations to identify epitope positions and show the method effectiveness by mapping the fine epitope for different antibodies targeting TNF, pertussis toxin, and the cancer target TROP2. In all three cases, the experimentally determined conformational epitope was consistent with previous experimental datasets, confirming the reliability of the experimental pipeline. Once the comprehensive library is generated, fine conformational epitope maps can be prepared at a rate of four per day.


Asunto(s)
Anticuerpos/química , Antígenos de Neoplasias/química , Moléculas de Adhesión Celular/química , Mapeo Epitopo/métodos , Epítopos/química , Factor de Necrosis Tumoral alfa/química , Antígenos de Neoplasias/genética , Moléculas de Adhesión Celular/genética , Epítopos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutagénesis , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA