Cargando…

Human Antibody Domains and Fragments Targeting Neutrophil Elastase as Candidate Therapeutics for Cancer and Inflammation-Related Diseases

Neutrophil elastase (NE) is a serine protease released during neutrophil maturation. High levels of NE are related to lung tissue damage and poor prognosis in cancer; thus, NE is a potential target for therapeutic immunotherapy for multiple lung diseases and cancers. Here, we isolate and characteriz...

Descripción completa

Detalles Bibliográficos
Autores principales: Chu, Xiaojie, Sun, Zehua, Baek, Du-San, Li, Wei, Mellors, John W., Shapiro, Steven D., Dimitrov, Dimiter S.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8539514/
https://www.ncbi.nlm.nih.gov/pubmed/34681796
http://dx.doi.org/10.3390/ijms222011136
_version_ 1784588765440770048
author Chu, Xiaojie
Sun, Zehua
Baek, Du-San
Li, Wei
Mellors, John W.
Shapiro, Steven D.
Dimitrov, Dimiter S.
author_facet Chu, Xiaojie
Sun, Zehua
Baek, Du-San
Li, Wei
Mellors, John W.
Shapiro, Steven D.
Dimitrov, Dimiter S.
author_sort Chu, Xiaojie
collection PubMed
description Neutrophil elastase (NE) is a serine protease released during neutrophil maturation. High levels of NE are related to lung tissue damage and poor prognosis in cancer; thus, NE is a potential target for therapeutic immunotherapy for multiple lung diseases and cancers. Here, we isolate and characterize two high-affinity, specific, and noncompetitive anti-NE antibodies Fab 1C10 and V(H) 1D1.43 from two large phage-displayed human Fab and V(H) libraries. After fusion with human IgG1 Fc, both of them (V(H)-Fc 1D1.43 and IgG1 1C10) inhibit NE enzymatic activity with V(H)-Fc 1D1.43 showing comparable inhibitory effects to that of the small molecule NE inhibitor SPCK and IgG1 1C10 exhibiting even higher (2.6-fold) activity than SPCK. Their epitopes, as mapped by peptide arrays combined with structural modeling, indicate different mechanisms for blocking NE activity. Both V(H)-Fc and IgG1 antibodies block NE uptake by cancer cells and fibroblast differentiation. V(H)-Fc 1D1.43 and IgG1 1C10 are promising for the antibody-based immunotherapy of cancer and inflammatory diseases.
format Online
Article
Text
id pubmed-8539514
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-85395142021-10-24 Human Antibody Domains and Fragments Targeting Neutrophil Elastase as Candidate Therapeutics for Cancer and Inflammation-Related Diseases Chu, Xiaojie Sun, Zehua Baek, Du-San Li, Wei Mellors, John W. Shapiro, Steven D. Dimitrov, Dimiter S. Int J Mol Sci Article Neutrophil elastase (NE) is a serine protease released during neutrophil maturation. High levels of NE are related to lung tissue damage and poor prognosis in cancer; thus, NE is a potential target for therapeutic immunotherapy for multiple lung diseases and cancers. Here, we isolate and characterize two high-affinity, specific, and noncompetitive anti-NE antibodies Fab 1C10 and V(H) 1D1.43 from two large phage-displayed human Fab and V(H) libraries. After fusion with human IgG1 Fc, both of them (V(H)-Fc 1D1.43 and IgG1 1C10) inhibit NE enzymatic activity with V(H)-Fc 1D1.43 showing comparable inhibitory effects to that of the small molecule NE inhibitor SPCK and IgG1 1C10 exhibiting even higher (2.6-fold) activity than SPCK. Their epitopes, as mapped by peptide arrays combined with structural modeling, indicate different mechanisms for blocking NE activity. Both V(H)-Fc and IgG1 antibodies block NE uptake by cancer cells and fibroblast differentiation. V(H)-Fc 1D1.43 and IgG1 1C10 are promising for the antibody-based immunotherapy of cancer and inflammatory diseases. MDPI 2021-10-15 /pmc/articles/PMC8539514/ /pubmed/34681796 http://dx.doi.org/10.3390/ijms222011136 Text en © 2021 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Chu, Xiaojie
Sun, Zehua
Baek, Du-San
Li, Wei
Mellors, John W.
Shapiro, Steven D.
Dimitrov, Dimiter S.
Human Antibody Domains and Fragments Targeting Neutrophil Elastase as Candidate Therapeutics for Cancer and Inflammation-Related Diseases
title Human Antibody Domains and Fragments Targeting Neutrophil Elastase as Candidate Therapeutics for Cancer and Inflammation-Related Diseases
title_full Human Antibody Domains and Fragments Targeting Neutrophil Elastase as Candidate Therapeutics for Cancer and Inflammation-Related Diseases
title_fullStr Human Antibody Domains and Fragments Targeting Neutrophil Elastase as Candidate Therapeutics for Cancer and Inflammation-Related Diseases
title_full_unstemmed Human Antibody Domains and Fragments Targeting Neutrophil Elastase as Candidate Therapeutics for Cancer and Inflammation-Related Diseases
title_short Human Antibody Domains and Fragments Targeting Neutrophil Elastase as Candidate Therapeutics for Cancer and Inflammation-Related Diseases
title_sort human antibody domains and fragments targeting neutrophil elastase as candidate therapeutics for cancer and inflammation-related diseases
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8539514/
https://www.ncbi.nlm.nih.gov/pubmed/34681796
http://dx.doi.org/10.3390/ijms222011136
work_keys_str_mv AT chuxiaojie humanantibodydomainsandfragmentstargetingneutrophilelastaseascandidatetherapeuticsforcancerandinflammationrelateddiseases
AT sunzehua humanantibodydomainsandfragmentstargetingneutrophilelastaseascandidatetherapeuticsforcancerandinflammationrelateddiseases
AT baekdusan humanantibodydomainsandfragmentstargetingneutrophilelastaseascandidatetherapeuticsforcancerandinflammationrelateddiseases
AT liwei humanantibodydomainsandfragmentstargetingneutrophilelastaseascandidatetherapeuticsforcancerandinflammationrelateddiseases
AT mellorsjohnw humanantibodydomainsandfragmentstargetingneutrophilelastaseascandidatetherapeuticsforcancerandinflammationrelateddiseases
AT shapirostevend humanantibodydomainsandfragmentstargetingneutrophilelastaseascandidatetherapeuticsforcancerandinflammationrelateddiseases
AT dimitrovdimiters humanantibodydomainsandfragmentstargetingneutrophilelastaseascandidatetherapeuticsforcancerandinflammationrelateddiseases