Cargando…

Targeting the Initiator Protease of the Classical Pathway of Complement Using Fragment-Based Drug Discovery

The initiating protease of the complement classical pathway, C1r, represents an upstream and pathway-specific intervention point for complement-related autoimmune and inflammatory diseases. Yet, C1r-targeted therapeutic development is currently underrepresented relative to other complement targets....

Descripción completa

Detalles Bibliográficos
Autores principales: Rushing, Blake R., Rohlik, Denise L., Roy, Sourav, Skaff, D. Andrew, Garcia, Brandon L.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7504721/
https://www.ncbi.nlm.nih.gov/pubmed/32899120
http://dx.doi.org/10.3390/molecules25174016
_version_ 1783584689026498560
author Rushing, Blake R.
Rohlik, Denise L.
Roy, Sourav
Skaff, D. Andrew
Garcia, Brandon L.
author_facet Rushing, Blake R.
Rohlik, Denise L.
Roy, Sourav
Skaff, D. Andrew
Garcia, Brandon L.
author_sort Rushing, Blake R.
collection PubMed
description The initiating protease of the complement classical pathway, C1r, represents an upstream and pathway-specific intervention point for complement-related autoimmune and inflammatory diseases. Yet, C1r-targeted therapeutic development is currently underrepresented relative to other complement targets. In this study, we developed a fragment-based drug discovery approach using surface plasmon resonance (SPR) and molecular modeling to identify and characterize novel C1r-binding small-molecule fragments. SPR was used to screen a 2000-compound fragment library for binding to human C1r. This led to the identification of 24 compounds that bound C1r with equilibrium dissociation constants ranging between 160–1700 µM. Two fragments, termed CMP-1611 and CMP-1696, directly inhibited classical pathway-specific complement activation in a dose-dependent manner. CMP-1611 was selective for classical pathway inhibition, while CMP-1696 also blocked the lectin pathway but not the alternative pathway. Direct binding experiments mapped the CMP-1696 binding site to the serine protease domain of C1r and molecular docking and molecular dynamics studies, combined with C1r autoactivation assays, suggest that CMP-1696 binds within the C1r active site. The group of structurally distinct fragments identified here, along with the structure–activity relationship profiling of two lead fragments, form the basis for future development of novel high-affinity C1r-binding, classical pathway-specific, small-molecule complement inhibitors.
format Online
Article
Text
id pubmed-7504721
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-75047212020-09-26 Targeting the Initiator Protease of the Classical Pathway of Complement Using Fragment-Based Drug Discovery Rushing, Blake R. Rohlik, Denise L. Roy, Sourav Skaff, D. Andrew Garcia, Brandon L. Molecules Article The initiating protease of the complement classical pathway, C1r, represents an upstream and pathway-specific intervention point for complement-related autoimmune and inflammatory diseases. Yet, C1r-targeted therapeutic development is currently underrepresented relative to other complement targets. In this study, we developed a fragment-based drug discovery approach using surface plasmon resonance (SPR) and molecular modeling to identify and characterize novel C1r-binding small-molecule fragments. SPR was used to screen a 2000-compound fragment library for binding to human C1r. This led to the identification of 24 compounds that bound C1r with equilibrium dissociation constants ranging between 160–1700 µM. Two fragments, termed CMP-1611 and CMP-1696, directly inhibited classical pathway-specific complement activation in a dose-dependent manner. CMP-1611 was selective for classical pathway inhibition, while CMP-1696 also blocked the lectin pathway but not the alternative pathway. Direct binding experiments mapped the CMP-1696 binding site to the serine protease domain of C1r and molecular docking and molecular dynamics studies, combined with C1r autoactivation assays, suggest that CMP-1696 binds within the C1r active site. The group of structurally distinct fragments identified here, along with the structure–activity relationship profiling of two lead fragments, form the basis for future development of novel high-affinity C1r-binding, classical pathway-specific, small-molecule complement inhibitors. MDPI 2020-09-03 /pmc/articles/PMC7504721/ /pubmed/32899120 http://dx.doi.org/10.3390/molecules25174016 Text en © 2020 by the authors. 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 (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Rushing, Blake R.
Rohlik, Denise L.
Roy, Sourav
Skaff, D. Andrew
Garcia, Brandon L.
Targeting the Initiator Protease of the Classical Pathway of Complement Using Fragment-Based Drug Discovery
title Targeting the Initiator Protease of the Classical Pathway of Complement Using Fragment-Based Drug Discovery
title_full Targeting the Initiator Protease of the Classical Pathway of Complement Using Fragment-Based Drug Discovery
title_fullStr Targeting the Initiator Protease of the Classical Pathway of Complement Using Fragment-Based Drug Discovery
title_full_unstemmed Targeting the Initiator Protease of the Classical Pathway of Complement Using Fragment-Based Drug Discovery
title_short Targeting the Initiator Protease of the Classical Pathway of Complement Using Fragment-Based Drug Discovery
title_sort targeting the initiator protease of the classical pathway of complement using fragment-based drug discovery
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7504721/
https://www.ncbi.nlm.nih.gov/pubmed/32899120
http://dx.doi.org/10.3390/molecules25174016
work_keys_str_mv AT rushingblaker targetingtheinitiatorproteaseoftheclassicalpathwayofcomplementusingfragmentbaseddrugdiscovery
AT rohlikdenisel targetingtheinitiatorproteaseoftheclassicalpathwayofcomplementusingfragmentbaseddrugdiscovery
AT roysourav targetingtheinitiatorproteaseoftheclassicalpathwayofcomplementusingfragmentbaseddrugdiscovery
AT skaffdandrew targetingtheinitiatorproteaseoftheclassicalpathwayofcomplementusingfragmentbaseddrugdiscovery
AT garciabrandonl targetingtheinitiatorproteaseoftheclassicalpathwayofcomplementusingfragmentbaseddrugdiscovery