Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Antib Ther ; 5(4): 258-267, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36299415

ABSTRACT

Background: Interleukin (IL)25 has been implicated in tissue homeostasis at barrier surfaces and the initiation of type two inflammatory signaling in response to infection and cell injury across multiple organs. We sought to discover and engineer a high affinity neutralizing antibody and evaluate the antibody functional activity in vitro and in vivo. Methods: In this study, we generated a novel anti-IL25 antibody (22C7) and investigated the antibody's therapeutic potential for targeting IL25 in inflammation. Results: A novel anti-IL25 antibody (22C7) was generated with equivalent in vitro affinity and potency against the human and mouse orthologs of the cytokine. This translated into in vivo potency in an IL25-induced air pouch model where 22C7 inhibited the recruitment of monocytes, macrophages, neutrophils and eosinophils. Furthermore, 22C7 significantly reduced ear swelling, acanthosis and disease severity in the Aldara mouse model of psoriasiform skin inflammation. Given the therapeutic potential of IL25 targeting in inflammatory conditions, 22C7 was further engineered to generate a highly developable, fully human antibody while maintaining the affinity and potency of the parental molecule. Conclusions: The generation of 22C7, an anti-IL25 antibody with efficacy in a preclinical model of skin inflammation, raises the therapeutic potential for 22C7 use in the spectrum of IL25-mediated diseases.

2.
MAbs ; 13(1): 1850395, 2021.
Article in English | MEDLINE | ID: mdl-33459147

ABSTRACT

We report here the discovery and optimization of a novel T cell retargeting anti-GUCY2C x anti-CD3ε bispecific antibody for the treatment of solid tumors. Using a combination of hybridoma, phage display and rational design protein engineering, we have developed a fully humanized and manufacturable CD3 bispecific antibody that demonstrates favorable pharmacokinetic properties and potent in vivo efficacy. Anti-GUCY2C and anti-CD3ε antibodies derived from mouse hybridomas were first humanized into well-behaved human variable region frameworks with full retention of binding and T-cell mediated cytotoxic activity. To address potential manufacturability concerns, multiple approaches were taken in parallel to optimize and de-risk the two antibody variable regions. These approaches included structure-guided rational mutagenesis and phage display-based optimization, focusing on improving stability, reducing polyreactivity and self-association potential, removing chemical liabilities and proteolytic cleavage sites, and de-risking immunogenicity. Employing rapid library construction methods as well as automated phage display and high-throughput protein production workflows enabled efficient generation of an optimized bispecific antibody with desirable manufacturability properties, high stability, and low nonspecific binding. Proteolytic cleavage and deamidation in complementarity-determining regions were also successfully addressed. Collectively, these improvements translated to a molecule with potent single-agent in vivo efficacy in a tumor cell line adoptive transfer model and a cynomolgus monkey pharmacokinetic profile (half-life>4.5 days) suitable for clinical development. Clinical evaluation of PF-07062119 is ongoing.


Subject(s)
Antibodies, Bispecific/immunology , CD3 Complex/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Enterotoxin/immunology , Animals , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/therapeutic use , Cell Line, Tumor , Female , Humans , Hybridomas , Macaca fascicularis/immunology , Macaca fascicularis/metabolism , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/metabolism , Protein Engineering/methods , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacokinetics , Single-Chain Antibodies/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
JCI Insight ; 2(9)2017 May 04.
Article in English | MEDLINE | ID: mdl-28469079

ABSTRACT

Initial promising results with immune sera guided early human mAb approaches against Gram-negative sepsis to an LPS neutralization mechanism, but these efforts failed in human clinical trials. Emergence of multidrug resistance has renewed interest in pathogen-specific mAbs. We utilized a pair of antibodies targeting Klebsiella pneumoniae LPS, one that both neutralizes LPS/TLR4 signaling and mediates opsonophagocytic killing (OPK) (54H7) and one that only promotes OPK (KPE33), to better understand the contribution of each mechanism to mAb protection in an acutely lethal pneumonia model. Passive immunization 24 hours prior to infection with KPE33 protected against lethal infection significantly better than 54H7, while delivery of either mAb 1 hour after infection resulted in similar levels of protection. These data suggest that early neutralization of LPS-induced signaling limits protection afforded by these mAbs. LPS neutralization prevented increases in the numbers of γδT cells, a major producer of the antimicrobial cytokine IL-17A, the contribution of which was confirmed using il17a-knockout mice. We conclude that targeting LPS for OPK without LPS signaling neutralization has potential to combat Gram-negative infection by engaging host immune defenses, rather than inhibiting beneficial innate immune pathways.

4.
PLoS One ; 12(1): e0170529, 2017.
Article in English | MEDLINE | ID: mdl-28107434

ABSTRACT

Antibody therapy against antibiotics resistant Klebsiella pneumoniae infections represents a promising strategy, the success of which depends critically on the ability to identify appropriate antibody targets. Using a target-agnostic strategy, we recently discovered MrkA as a potential antibody target and vaccine antigen. Interestingly, the anti-MrkA monoclonal antibodies isolated through phage display and hybridoma platforms all recognize an overlapping epitope, which opens up important questions including whether monoclonal antibodies targeting different MrkA epitopes can be generated and if they possess different protective profiles. In this study we generated four anti-MrkA antibodies targeting different epitopes through phage library panning against recombinant MrkA protein. These anti-MrkA antibodies elicited strong in vitro and in vivo protections against a multi-drug resistant Klebsiella pneumoniae strain. Furthermore, mutational and epitope analysis suggest that the two cysteine residues may play essential roles in maintaining a MrkA structure that is highly compacted and exposes limited antibody binding/neutralizing epitopes. These results suggest the need for further in-depth understandings of the structure of MrkA, the role of MrkA in the pathogenesis of Klebsiella pneumoniae and the protective mechanism adopted by anti-MrkA antibodies to fully explore the potential of MrkA as an efficient therapeutic target and vaccine antigen.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Klebsiella pneumoniae/immunology , Animals , Drug Resistance, Multiple, Bacterial/immunology , Epitopes/immunology , Flow Cytometry , Interferometry , Klebsiella Infections/immunology , Mice , Mice, Inbred C57BL , Recombinant Proteins
5.
J Infect Dis ; 213(11): 1800-8, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26768253

ABSTRACT

The increasing incidence of Klebsiella pneumoniae infections refractory to treatment with current broad-spectrum antibiotic classes warrants the exploration of alternative approaches, such as antibody therapy and/or vaccines, for prevention and treatment. However, the lack of validated targets shared by spectrums of clinical strains poses a significant challenge. We adopted a target-agnostic approach to identify protective antibodies against K. pneumoniae Several monoclonal antibodies were isolated from phage display and hybridoma platforms by functional screening for opsonophagocytic killing activity. We further identified their common target antigen to be MrkA, a major protein in the type III fimbriae complex, and showed that these serotype-independent anti-MrkA antibodies reduced biofilm formation in vitro and conferred protection in multiple murine pneumonia models. Importantly, mice immunized with purified MrkA proteins also showed reduced bacterial burden following K. pneumoniae challenge. Taken together, these results support MrkA as a promising target for K. pneumoniae antibody therapeutics and vaccines.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Fimbriae Proteins/immunology , Klebsiella pneumoniae/immunology , Animals , Antibody Specificity , Bacterial Vaccines/immunology , Biofilms , Cytotoxicity, Immunologic , Humans , Hybridomas , Klebsiella Infections/prevention & control , Mice , Mice, Inbred C57BL , Peptide Library , Phagocytosis , Respiratory Mucosa/microbiology
6.
PLoS One ; 10(2): e0115828, 2015.
Article in English | MEDLINE | ID: mdl-25706559

ABSTRACT

Release of endogenous damage associated molecular patterns (DAMPs), including members of the S100 family, are associated with infection, cellular stress, tissue damage and cancer. The extracellular functions of this family of calcium binding proteins, particularly S100A8, S100A9 and S100A12, are being delineated. They appear to mediate their functions via receptor for advanced glycation endproducts (RAGE) or TLR4, but there remains considerable uncertainty over the relative physiological roles of these DAMPs and their pattern recognition receptors. In this study, we surveyed the capacity of S100 proteins to induce proinflammatory cytokines and cell migration, and the contribution RAGE and TLR4 to mediate these responses in vitro. Using adenoviral delivery of murine S100A9, we also examined the potential for S100A9 homodimers to trigger lung inflammation in vivo. S100A8, S100A9 and S100A12, but not the S100A8/A9 heterodimer, induced modest levels of TLR4-mediated cytokine production from human PBMC. In contrast, for most S100s including S100A9, RAGE blockade inhibited S100-mediated cell migration of THP1 cells and major leukocyte populations, whereas TLR4-blockade had no effect. Intranasal administration of murine S100A9 adenovirus induced a specific, time-dependent predominately macrophage infiltration that coincided with elevated S100A9 levels and proinflammatory cytokines in the BAL fluid. Inflammatory cytokines were markedly ablated in the TLR4-defective mice, but unexpectedly the loss of TLR4 signaling or RAGE-deficiency did not appreciably impact the S100A9-mediated lung pathology or the inflammatory cell infiltrate in the alveolar space. These data demonstrate that physiological levels of S100A9 homodimers can trigger an inflammatory response in vivo, and despite the capacity of RAGE and TLR4 blockade to inhibit responses in vitro, the response is predominately independent of both these receptors.


Subject(s)
Calgranulin B/pharmacology , Cell Movement/physiology , Signal Transduction/drug effects , Animals , Cell Line , Cell Movement/drug effects , Humans , Inflammation/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Toll-Like Receptor 4/metabolism
7.
J Leukoc Biol ; 77(5): 644-51, 2005 May.
Article in English | MEDLINE | ID: mdl-15703200

ABSTRACT

Ly-49 receptors regulate mouse natural killer cell functions. Members of the polymorphic Ly-49 multigene family recognize specific alleles of major histocompatibility complex class I (MHC I) or MHC I-like proteins. Previous studies have provided insight into the nature of Ly-49A and -C interaction with their high-affinity MHC I ligands, H-2Dd and Kb, respectively. Unlike Ly-49C, recognition of MHC I by Ly-49A is regulated in part by residues within the beta4-beta5 loop of its ectodomain. Ly-49A and -G are within the same Ly-49 subfamily, and both receptors recognize Dd. However, there have been no studies that define specific sites on Ly-49G that mediate class I MHC recognition. The Ly-49G receptors of different inbred mouse strains can differ as a result of amino acid polymorphisms within their ectodomains. In this report, we have generated a novel antibody, CK-1, which recognizes Ly-49G(B6) and a Ly-49G(B6)-like receptor, Ly-49M(nonobese diabetic), but not Ly-49G(BALB/c). By exploiting the differences within ectodomains of C57BL/6 and BALB/c Ly-49G allele products, we identified epitopes recognized by the Ly-49G-specific antibodies CK-1 and Cwy-3, whose epitopes mapped within the beta4-beta5 loop and the beta1 strand, respectively, and were nonoverlapping. Although both antibodies specifically recognized the Ly-49G(B6) ectodomain, Cwy-3 was unable to block its interaction with MHC I, and CK-1 significantly inhibited it. The importance of residues within the beta4-beta5 loop in Ly-49G recognition demonstrates that its interaction with MHC I is similar to that of Ly-49A but not Ly-49C.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Ly/immunology , Epitope Mapping , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Amino Acid Sequence , Animals , Antigens, Ly/chemistry , Antigens, Ly/genetics , COS Cells , Chlorocebus aethiops , Flow Cytometry , Genes, MHC Class I/immunology , Killer Cells, Natural/cytology , Lectins, C-Type , Lymphocyte Subsets/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Polymorphism, Genetic , Protein Structure, Tertiary , Receptors, NK Cell Lectin-Like , Sequence Alignment , Species Specificity
8.
Hybrid Hybridomics ; 21(4): 271-80, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12193280

ABSTRACT

A hybridoma, CSC-1, which secretes monoclonal antibody (MAb) specific for a cell surface molecule on African green monkey kidney cell line, BGMK, was isolated and characterized. The cell surface molecule recognized by CSC-1 is widely expressed on a variety of human cell lines. Among the hematopoietic cell lines examined, the CSC-1 marker seems to be preferentially expressed by lymphoid cell lines (e.g., Raji, CEM-SS, Jurkat, and MOLT-3). Although CSC-1 also recognizes some uncommitted myeloid and erythroleukemia cell lines (e.g., U-937 and K562), the CSC-1 marker expression on these cell lines is subjected to PMA-induced down-regulation. In addition, the PMA-induced cell surface down-regulation of the CSC-1 marker required more than 24 h. Proteolytic analysis shows that the CSC-1 marker can be categorized into proteinase K-resistant and-sensitive phenotypes. Furthermore, the CSC-1 marker exhibits a slow cell surface turnover rate after proteinase K treatment. Our data suggest that the CSC-1 cell surface molecule might be useful as a megakaryocytic or monocytic differentiation marker.


Subject(s)
Antibodies, Monoclonal , Cell Membrane/immunology , Cell Membrane/metabolism , Leukemia, Erythroblastic, Acute/immunology , Leukemia, Erythroblastic, Acute/metabolism , Animals , Antibody Specificity , Biomarkers , Cell Line , Chlorocebus aethiops , Endopeptidase K/metabolism , Epitopes/metabolism , Humans , Hybridomas/immunology , Mice , Mice, Inbred BALB C , Tetradecanoylphorbol Acetate/pharmacology
9.
Hybrid Hybridomics ; 21(6): 445-56, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12573108

ABSTRACT

A mouse IgG(1)-producing hybridoma, CSC-31, was isolated and characterized. The monoclonal antibody (MAb) was originally raised against monkey kidney cell-surface molecules. FACS analysis further showed that CSC-31 exhibited broad tissue and species reactivity. Although most human T- and B-cell lines failed to react with CSC-31, myeloid, and erythroleukemia cells lines such as THP-1 and K562 expressed the CSC-31 cell surface marker. Furthermore, in vitro differentiation of HL-60, U-937, and K562 showed that expression of the CSC-31 marker is associated with monocytic or megakaryocytic differentiation. However, up-regulation of the CSC-31 marker expression was not detected during granulocytic or erythroid differentiation. Through the in vitro differentiation of K562, it was demonstrated that up-regulation of the CSC-31 marker required novel PKCs and might be regulated by the MAPK signaling pathway. Last, limited biochemical analysis demonstrated that the CSC-31-specific epitope is sensitive to digestion by papain yet highly resistant to other proteases.


Subject(s)
Antibodies, Monoclonal/immunology , Cell Differentiation/immunology , Megakaryocytes/immunology , Monocytes/immunology , Animals , Antibodies, Monoclonal/metabolism , Antigens, Surface/immunology , Biomarkers , Cell Differentiation/physiology , Culture Media, Conditioned/metabolism , Epitopes/immunology , Humans , Megakaryocytes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Monocytes/metabolism , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...