1
|
Raavi, Koehler AN, Vegas AJ. At The Interface: Small-Molecule Inhibitors of Soluble Cytokines. Chem Rev 2025. [PMID: 40233276 DOI: 10.1021/acs.chemrev.4c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cytokines are crucial regulators of the immune system that orchestrate interactions between cells and, when dysregulated, contribute to the progression of chronic inflammation, cancer, and autoimmunity. Numerous biologic-based clinical agents, mostly monoclonal antibodies, have validated cytokines as important clinical targets and are now part of the standard of care for a number of diseases. These agents, while impactful, still suffer from limitations including a lack of oral bioavailability, high cost of production, and immunogenicity. Small-molecule cytokine inhibitors are attractive alternatives that can address these limitations. Although targeting cytokine-cytokine receptor complexes with small molecules has been a challenging research endeavor, multiple small-molecule inhibitors have now been identified, with a number of them undergoing clinical evaluation. In this review, we highlight the recent advancements in the discovery and development of small-molecule inhibitors targeting soluble cytokines. The strategies for identifying these novel ligands as well as the structural and mechanistic insights into their activity represent important milestones in tackling these challenging and clinically important protein-protein interactions.
Collapse
Affiliation(s)
- Raavi
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Angela N Koehler
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Arturo J Vegas
- Department of Chemistry, Boston University, Boston, Massachusetts 02115, United States
| |
Collapse
|
2
|
Shaw DJ, Waters LC, Strong SL, Schulze MSED, Greetham GM, Towrie M, Parker AW, Prosser CE, Henry AJ, Lawson ADG, Carr MD, Taylor RJ, Hunt NT, Muskett FW. Modulation of IL-17 backbone dynamics reduces receptor affinity and reveals a new inhibitory mechanism. Chem Sci 2023; 14:7524-7536. [PMID: 37449080 PMCID: PMC10337760 DOI: 10.1039/d3sc00728f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
Knowledge of protein dynamics is fundamental to the understanding of biological processes, with NMR and 2D-IR spectroscopy being two of the principal methods for studying protein dynamics. Here, we combine these two methods to gain a new understanding of the complex mechanism of a cytokine:receptor interaction. The dynamic nature of many cytokines is now being recognised as a key property in the signalling mechanism. Interleukin-17s (IL-17) are proinflammatory cytokines which, if unregulated, are associated with serious autoimmune diseases such as psoriasis, and although there are several therapeutics on the market for these conditions, small molecule therapeutics remain elusive. Previous studies, exploiting crystallographic methods alone, have been unable to explain the dramatic differences in affinity observed between IL-17 dimers and their receptors, suggesting there are factors that cannot be fully explained by the analysis of static structures alone. Here, we show that the IL-17 family of cytokines have varying degrees of flexibility which directly correlates to their receptor affinities. Small molecule inhibitors of the cytokine:receptor interaction are usually thought to function by either causing steric clashes or structural changes. However, our results, supported by other biophysical methods, provide evidence for an alternate mechanism of inhibition, in which the small molecule rigidifies the protein, causing a reduction in receptor affinity. The results presented here indicate an induced fit model of cytokine:receptor binding, with the more flexible cytokines having a higher affinity. Our approach could be applied to other systems where the inhibition of a protein-protein interaction has proved intractable, for example due to the flat, featureless nature of the interface. Targeting allosteric sites which modulate protein dynamics, opens up new avenues for novel therapeutic development.
Collapse
Affiliation(s)
- Daniel J Shaw
- Department of Chemistry and York Biomedical Research Institute, University of York Heslington York YO19 5DD UK
| | - Lorna C Waters
- Department of Molecular and Cell Biology/Leicester Institute of Structural and Chemical Biology, University of Leicester, University Road Leicester LE1 7RH UK
| | - Sarah L Strong
- Department of Molecular and Cell Biology/Leicester Institute of Structural and Chemical Biology, University of Leicester, University Road Leicester LE1 7RH UK
| | | | - Gregory M Greetham
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory Harwell Oxford Didcot Oxon OX11 0QX UK
| | - Mike Towrie
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory Harwell Oxford Didcot Oxon OX11 0QX UK
| | - Anthony W Parker
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory Harwell Oxford Didcot Oxon OX11 0QX UK
| | | | | | | | - Mark D Carr
- Department of Molecular and Cell Biology/Leicester Institute of Structural and Chemical Biology, University of Leicester, University Road Leicester LE1 7RH UK
| | | | - Neil T Hunt
- Department of Chemistry and York Biomedical Research Institute, University of York Heslington York YO19 5DD UK
| | - Frederick W Muskett
- Department of Molecular and Cell Biology/Leicester Institute of Structural and Chemical Biology, University of Leicester, University Road Leicester LE1 7RH UK
| |
Collapse
|
3
|
Zhang B, Dömling A. Small molecule modulators of IL-17A/IL-17RA: a patent review (2013-2021). Expert Opin Ther Pat 2022; 32:1161-1173. [PMID: 36350977 DOI: 10.1080/13543776.2022.2143264] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Interleukin-17A (IL-17A) is a well-established pro-inflammatory cytokine, which plays a pivotal role in immune and autoimmune diseases including psoriasis, asthma, psoriatic arthritis, and rheumatoid arthritis. Three currently approved monoclonal antibodies (mAbs) are in clinical practice for the treatment of multiple immune diseases. However, the disadvantages of the mAbs, such as non-oral administration, poor tissue penetration, lacking blood-brain barrier penetration, often long half-life times, narrow its application. Thus, intensive research is performed to discover potent small molecules, peptides, and macrocycles targeting the IL-17A/IL-17 RA protein-protein interaction (PPI) to modulate immune responses as an attractive approach for immunotherapy. AREAS COVERED Small molecules, macrocycles, and peptides targeting IL-17A/IL-17RA PPI from 2013 to 2021. EXPERT OPINION The rapid increase in the identification of small-molecule inhibitors of IL-17 should translate into a supplement of current biotherapeutics with mAbs. Potential advantages of small molecules over mAbs show room for clinical treatment improvement and new indication areas . An increasing number of patents and articles are recently published on small-molecule immunomodulators (SMIMs). Two compounds from Lilly and Leo Pharma are currently investigated in early clinical trials, followed by a Dice molecule. The outcome of these trials will influence future development of IL-17 inhibitors for treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Bidong Zhang
- Department of Drug Design, University of Groningen, Groningen, The Netherlands
| | - Alexander Dömling
- Department of Innovative Chemistry, CATRIN, Palackӯ University, Olomouc, Czech Republic
| |
Collapse
|
4
|
Abdelraheem E, Lubberink M, Wang W, Li J, Reyes Romero A, van der Straat R, Du X, Groves M, Dömling A. Multicomponent Macrocyclic IL-17a Modifier. ACS Med Chem Lett 2022; 13:1468-1471. [PMID: 36105327 PMCID: PMC9465830 DOI: 10.1021/acsmedchemlett.2c00257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/10/2022] [Indexed: 11/28/2022] Open
Abstract
![]()
IL-17a is a major inflammation target, with several approved
antibodies
in clinical use. Small-molecule IL-17a antagonists are an emerging
hot topic, with the recent advancement of three compounds into clinical
trials. Here, we describe the design, discovery, synthesis, and screening
of macrocyclic compounds that bind to IL-17a. We found that all currently
described IL-17a modifiers belong to the same pharmacophore model,
likely resulting in a similar receptor binding mode on IL-17a. A pipeline
of pharmacophore analysis, virtual screening, resynthesis, and protein
biophysics resulted in a potent IL-17a macrocyclic modifier.
Collapse
Affiliation(s)
- Eman Abdelraheem
- Department of Pharmacy, Drug Design Group, University of Groningen, A. Deusinglaan 1, Groningen 9700 AV, The Netherlands
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82524, Egypt
| | - Max Lubberink
- Department of Pharmacy, Drug Design Group, University of Groningen, A. Deusinglaan 1, Groningen 9700 AV, The Netherlands
| | - Wenja Wang
- Department of Pharmacy, Drug Design Group, University of Groningen, A. Deusinglaan 1, Groningen 9700 AV, The Netherlands
| | - Jingyao Li
- Department of Pharmacy, Drug Design Group, University of Groningen, A. Deusinglaan 1, Groningen 9700 AV, The Netherlands
| | - Atilio Reyes Romero
- Department of Pharmacy, Drug Design Group, University of Groningen, A. Deusinglaan 1, Groningen 9700 AV, The Netherlands
| | - Robin van der Straat
- Department of Pharmacy, Drug Design Group, University of Groningen, A. Deusinglaan 1, Groningen 9700 AV, The Netherlands
| | - Xiaochen Du
- Department of Pharmacy, Drug Design Group, University of Groningen, A. Deusinglaan 1, Groningen 9700 AV, The Netherlands
| | - Matthew Groves
- Department of Pharmacy, Drug Design Group, University of Groningen, A. Deusinglaan 1, Groningen 9700 AV, The Netherlands
| | - Alexander Dömling
- Department of Pharmacy, Drug Design Group, University of Groningen, A. Deusinglaan 1, Groningen 9700 AV, The Netherlands
| |
Collapse
|
5
|
Andrews MD, Dack KN, de Groot MJ, Lambert M, Sennbro CJ, Larsen M, Stahlhut M. Discovery of an Oral, Rule of 5 Compliant, Interleukin 17A Protein-Protein Interaction Modulator for the Potential Treatment of Psoriasis and Other Inflammatory Diseases. J Med Chem 2022; 65:8828-8842. [PMID: 35767390 DOI: 10.1021/acs.jmedchem.2c00422] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Interleukin 17A (IL-17A) is an interleukin cytokine whose dysregulation is implicated in autoimmune disorders such as psoriasis, and monoclonal antibodies against the IL-17A pathway are now well-established and very effective treatments. This article outlines the work that led to the identification of 23 as an oral, small-molecule protein-protein interaction modulator (PPIm) clinical development candidate. Protein crystallography provided knowledge of the key binding interactions between small-molecule ligands and the IL-17A dimer, and this helped in the multiparameter optimization toward identifying an orally bioavailable, Rule of 5 compliant PPIm of IL-17A. Overlap of early ligands led to a series of benzhydrylglycine-containing compounds that allowed the identification of dimethylpyrazole as a key substituent that gave PPIm with oral bioavailability. Exploration of the amino acid portion of the structure then led to dicyclopropylalanine as a group that gave potent and metabolically stable compounds, including the development candidate 23.
Collapse
Affiliation(s)
- Mark D Andrews
- Drug Design, LEO Pharma Research & Early Development, 2750 Ballerup, Denmark
| | - Kevin N Dack
- Drug Design, LEO Pharma Research & Early Development, 2750 Ballerup, Denmark
| | - Marcel J de Groot
- Drug Design, LEO Pharma Research & Early Development, 2750 Ballerup, Denmark
| | - Maja Lambert
- Drug Design, LEO Pharma Research & Early Development, 2750 Ballerup, Denmark
| | - Carl J Sennbro
- Drug Design, LEO Pharma Research & Early Development, 2750 Ballerup, Denmark
| | - Mogens Larsen
- Drug Design, LEO Pharma Research & Early Development, 2750 Ballerup, Denmark
| | - Martin Stahlhut
- Skin Research, LEO Pharma Research & Early Development, 2750 Ballerup, Denmark
| |
Collapse
|
6
|
Rahmawati SF, te Velde M, Kerstjens HAM, Dömling ASS, Groves MR, Gosens R. Pharmacological Rationale for Targeting IL-17 in Asthma. FRONTIERS IN ALLERGY 2021; 2:694514. [PMID: 35387016 PMCID: PMC8974835 DOI: 10.3389/falgy.2021.694514] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/07/2021] [Indexed: 01/09/2023] Open
Abstract
Asthma is a respiratory disease that currently affects around 300 million people worldwide and is defined by coughing, shortness of breath, wheezing, mucus overproduction, chest tightness, and expiratory airflow limitation. Increased levels of interleukin 17 (IL-17) have been observed in sputum, nasal and bronchial biopsies, and serum of patients with asthma compared to healthy controls. Patients with higher levels of IL-17 have a more severe asthma phenotype. Biologics are available for T helper 2 (Th2)-high asthmatics, but the Th17-high subpopulation has a relatively low response to these treatments, rendering it a rather severe asthma phenotype to treat. Several experimental models suggest that targeting the IL-17 pathway may be beneficial in asthma. Moreover, as increased activation of the Th17/IL-17 axis is correlated with reduced inhaled corticosteroids (ICS) sensitivity, targeting the IL-17 pathway might reverse ICS unresponsiveness. In this review, we present and discuss the current knowledge on the role of IL-17 in asthma and its interaction with the Th2 pathway, focusing on the rationale for therapeutic targeting of the IL-17 pathway.
Collapse
Affiliation(s)
- Siti Farah Rahmawati
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Department of Pharmacology and Clinical Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
| | - Maurice te Velde
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
| | - Huib A. M. Kerstjens
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
- Department of Pulmonary Medicine, University of Groningen and University Medical Center Groningen (UMCG), Groningen, Netherlands
| | | | | | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
- *Correspondence: Reinoud Gosens
| |
Collapse
|
7
|
Zheng J, Chen D, Xu J, Ding X, Wu Y, Shen HC, Tan X. Small molecule approaches to treat autoimmune and inflammatory diseases (Part III): Targeting cytokines and cytokine receptor complexes. Bioorg Med Chem Lett 2021; 48:128229. [PMID: 34214508 DOI: 10.1016/j.bmcl.2021.128229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 01/14/2023]
Abstract
Chronic and dysregulated cytokine signaling plays an important role in the pathogenic development of many autoimmune and inflammatory diseases. Despite intrinsic challenges in the disruption of interactions between cytokines and cytokine receptors, many first-in-class small-molecule inhibitors have been discovered over the past few years. The third part of the digest series presents recent progress in identifying such inhibitors and highlights the application of novel research tools in the fields of structural biology, computational analysis, screening methods, biophysical/biochemical assays and medicinal chemistry strategy.
Collapse
Affiliation(s)
- Jiamin Zheng
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Dongdong Chen
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Jie Xu
- Department of Immunology, Infectious Disease and Ophthalmology, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Xiao Ding
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Yao Wu
- Computer Aided Drug Design, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Hong C Shen
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Xuefei Tan
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China.
| |
Collapse
|
8
|
Koštrun S, Fajdetić A, Pešić D, Brajša K, Bencetić Mihaljević V, Jelić D, Petrinić Grba A, Elenkov I, Rupčić R, Kapić S, Ozimec Landek I, Butković K, Grgičević A, Žiher D, Čikoš A, Padovan J, Saxty G, Dack K, Bladh H, Skak-Nielsen T, Feldbaek Nielsen S, Lambert M, Stahlhut M. Macrolide Inspired Macrocycles as Modulators of the IL-17A/IL-17RA Interaction. J Med Chem 2021; 64:8354-8383. [PMID: 34100601 DOI: 10.1021/acs.jmedchem.1c00327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Interleukin 17 (IL-17) cytokines promote inflammatory pathophysiology in many autoimmune diseases, including psoriasis, multiple sclerosis, rheumatoid arthritis, and inflammatory bowel disease. Such broad involvement of IL-17 in various autoimmune diseases makes it an ideal target for drug discovery. Psoriasis is a chronic inflammatory disease characterized by numerous defective components of the immune system. Significantly higher levels of IL-17A have been noticed in lesions of psoriatic patients, if compared to non-lesion parts. Therefore, this paper is focused on the macrolide inspired macrocycles as potential IL-17A/IL-17RA modulators and covers the molecular design, synthesis, and in vitro profiling. Macrocycles are designed to diversify and enrich chemical space through different ring sizes and a variety of three-dimensional shapes. Inhibitors in the nM range were identified in both target-based and phenotypic assays. In vitro ADME as well as in vivo PK properties are reported.
Collapse
Affiliation(s)
- Sanja Koštrun
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Andrea Fajdetić
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Dijana Pešić
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Karmen Brajša
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | | | - Dubravko Jelić
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | | | - Ivaylo Elenkov
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Renata Rupčić
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Samra Kapić
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | | | | | - Ana Grgičević
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Dinko Žiher
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Ana Čikoš
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Jasna Padovan
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Gordon Saxty
- Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Kevin Dack
- LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark
| | - Haakan Bladh
- LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark
| | | | | | - Maja Lambert
- LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark
| | | |
Collapse
|
9
|
Waters LC, Veverka V, Strong SL, Muskett FW, Dedi N, Lawson ADG, Prosser CE, Taylor RJ, Henry AJ, Carr MD. Conformational dynamics in interleukin 17A and 17F functional complexes is a key determinant of receptor A affinity and specificity. Cytokine 2021; 142:155476. [PMID: 33706174 DOI: 10.1016/j.cyto.2021.155476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/11/2022]
Abstract
The proinflammatory cytokines IL-17A and IL-17F have been identified as key drivers of a range of human inflammatory diseases, such as psoriasis, which has led to several therapeutic antibodies targeted at IL-17A. The two cytokines have been shown to tightly associate as functional homo and hetero dimers, which induce signalling via the formation of a cell surface signalling complex with a single copy of both IL-17RA and IL-17RC. Striking differences in affinity have been observed for IL-17RA binding to IL-17AA, IL-17AF and IL-17FF, however, the functional significance and molecular basis for this has remained unclear. We have obtained comprehensive backbone NMR assignments for full length IL-17AA (79%), IL-17AF (93%) and IL-17FF (89%), which show that the dimers adopt almost identical backbone topologies in solution to those observed in reported crystal structures. Analysis of the line widths and intensities of assigned backbone amide NMR signals has revealed striking differences in the conformational plasticity and dynamics of IL-17AA compared to both IL-17AF and IL-17FF. Our NMR data indicate that a number of regions of IL-17AA are interconverting between at least two distinct conformations on a relatively slow timescale. Such conformational heterogeneity has previously been shown to play an important role in the formation of many high affinity protein-protein complexes. The locations of the affected IL-17AA residues essentially coincides with the regions of both IL-17A and IL-17F previously shown to undergo significant structural changes on binding to IL-17RA. Substantially less conformational exchange was revealed by the NMR data for IL-17FF and IL-17AF. We propose that the markedly different conformational dynamic properties of the distinct functional IL-17 dimers plays a key role in determining their affinities for IL-17RA, with the more dynamic and plastic nature of IL-17AA contributing to the significantly tighter affinity observed for binding to IL-17RA. In contrast, the dynamic properties are expected to have little influence on the affinity of IL-17 dimers for IL-17RC, which has recently been shown to induce only small structural changes in IL-17FF upon binding.
Collapse
Affiliation(s)
- Lorna C Waters
- Department of Molecular and Cell Biology/Leicester Institute of Structural and Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom.
| | - Vaclav Veverka
- Department of Molecular and Cell Biology/Leicester Institute of Structural and Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Sarah L Strong
- Department of Molecular and Cell Biology/Leicester Institute of Structural and Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Frederick W Muskett
- Department of Molecular and Cell Biology/Leicester Institute of Structural and Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | | | | | | | | | | | - Mark D Carr
- Department of Molecular and Cell Biology/Leicester Institute of Structural and Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom.
| |
Collapse
|
10
|
Dietrich JD, Longenecker KL, Wilson NS, Goess C, Panchal SC, Swann SL, Petros AM, Hobson AD, Ihle D, Song D, Richardson P, Comess KM, Cox PB, Dombrowski A, Sarris K, Donnelly-Roberts DL, Duignan DB, Gomtsyan A, Jung P, Krueger AC, Mathieu S, McClure A, Stoll VS, Wetter J, Mankovich JA, Hajduk PJ, Vasudevan A, Stoffel RH, Sun C. Development of Orally Efficacious Allosteric Inhibitors of TNFα via Fragment-Based Drug Design. J Med Chem 2020; 64:417-429. [PMID: 33378180 DOI: 10.1021/acs.jmedchem.0c01280] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor necrosis factor α (TNFα) is a soluble cytokine that is directly involved in systemic inflammation through the regulation of the intracellular NF-κB and MAPK signaling pathways. The development of biologic drugs that inhibit TNFα has led to improved clinical outcomes for patients with rheumatoid arthritis and other chronic autoimmune diseases; however, TNFα has proven to be difficult to drug with small molecules. Herein, we present a two-phase, fragment-based drug discovery (FBDD) effort in which we first identified isoquinoline fragments that disrupt TNFα ligand-receptor binding through an allosteric desymmetrization mechanism as observed in high-resolution crystal structures. The second phase of discovery focused on the de novo design and optimization of fragments with improved binding efficiency and drug-like properties. The 3-indolinone-based lead presented here displays oral, in vivo efficacy in a mouse glucose-6-phosphate isomerase (GPI)-induced paw swelling model comparable to that seen with a TNFα antibody.
Collapse
Affiliation(s)
- Justin D Dietrich
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Kenton L Longenecker
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Noel S Wilson
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Christian Goess
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, Massachusetts 01605, United States
| | - Sanjay C Panchal
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Steven L Swann
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Andrew M Petros
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Adrian D Hobson
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, Massachusetts 01605, United States
| | - David Ihle
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, Massachusetts 01605, United States
| | - Danying Song
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Paul Richardson
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Kenneth M Comess
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Philip B Cox
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Amanda Dombrowski
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Kathy Sarris
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Diana L Donnelly-Roberts
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - David B Duignan
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, Massachusetts 01605, United States
| | - Arthur Gomtsyan
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Paul Jung
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - A Chris Krueger
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Suzanne Mathieu
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, Massachusetts 01605, United States
| | - Andrea McClure
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Vincent S Stoll
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Jill Wetter
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - John A Mankovich
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, Massachusetts 01605, United States
| | - Philip J Hajduk
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Anil Vasudevan
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Robert H Stoffel
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, Massachusetts 01605, United States
| | - Chaohong Sun
- Research & Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| |
Collapse
|
11
|
Álvarez-Coiradas E, Munteanu CR, Díaz-Sáez L, Pazos A, Huber KVM, Loza MI, Domínguez E. Discovery of novel immunopharmacological ligands targeting the IL-17 inflammatory pathway. Int Immunopharmacol 2020; 89:107026. [PMID: 33045560 DOI: 10.1016/j.intimp.2020.107026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/02/2020] [Accepted: 09/16/2020] [Indexed: 01/25/2023]
Abstract
Interleukin 17 (IL-17) is a proinflammatory cytokine that acts as an immune checkpoint for several autoimmune diseases. Therapeutic neutralizing antibodies that target this cytokine have demonstrated clinical efficacy in psoriasis. However, biologics have limitations such as their high cost and their lack of oral bioavailability. Thus, it is necessary to expand the therapeutic options for this IL-17A/IL-17RA pathway, applying novel drug discovery methods to find effective small molecules. In this work, we combined biophysical and cell-based assays with structure-based docking to find novel ligands that target this pathway. First, a virtual screening of our chemical library of 60000 compounds was used to identify 67 potential ligands of IL-17A and IL-17RA. We developed a biophysical label-free binding assay to determine interactions with the extracellular domain of IL-17RA. Two molecules (CBG040591 and CBG060392) with quinazolinone and pyrrolidinedione chemical scaffolds, respectively, were confirmed as ligands of IL-17RA with micromolar affinity. The anti-inflammatory activity of these ligands as cytokine-release inhibitors was evaluated in human keratinocytes. Both ligands inhibited the release of chemokines mediated by IL-17A, with an IC50 of 20.9 ± 12.6 μM and 23.6 ± 11.8 μM for CCL20 and an IC50 of 26.7 ± 13.1 μM and 45.3 ± 13.0 μM for CXCL8. Hence, they blocked IL-17A proinflammatory activity, which is consistent with the inhibition of the signalling of the IL-17A receptor by ligand CBG060392. Therefore, we identified two novel immunopharmacological ligands targeting the IL-17A/IL-17RA pathway with antiinflammatory efficacy that can be promising tools for a drug discovery program for psoriasis.
Collapse
Affiliation(s)
- Elia Álvarez-Coiradas
- Biofarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Avenida de Barcelona s/n, 15782 Santiago de Compostela, Spain
| | - Cristian R Munteanu
- RNASA-IMEDIR, Computer Science Faculty, CITIC, Universidade da Coruña, A Coruña, 15007, Spain; Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), A Coruña 15006, Spain
| | - Laura Díaz-Sáez
- Structural Genomics Consortium & Target Discovery Institute, University of Oxford, Nuffield Department of Medicine, Old Road Campus, Oxford OX3 7DQ & OX3 7FZ, UK
| | - Alejandro Pazos
- RNASA-IMEDIR, Computer Science Faculty, CITIC, Universidade da Coruña, A Coruña, 15007, Spain; Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), A Coruña 15006, Spain
| | - Kilian V M Huber
- Structural Genomics Consortium & Target Discovery Institute, University of Oxford, Nuffield Department of Medicine, Old Road Campus, Oxford OX3 7DQ & OX3 7FZ, UK
| | - María Isabel Loza
- Biofarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Avenida de Barcelona s/n, 15782 Santiago de Compostela, Spain.
| | - Eduardo Domínguez
- Biofarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Avenida de Barcelona s/n, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
12
|
Chang A, Ting JP, Espada A, Broughton H, Molina-Martin M, Afshar S. A novel phage display vector for selection of target-specific peptides. Protein Eng Des Sel 2020; 33:5917485. [PMID: 33009572 DOI: 10.1093/protein/gzaa023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/28/2020] [Accepted: 09/04/2020] [Indexed: 11/14/2022] Open
Abstract
Intrinsic low display level of polypeptides on phage is a fundamental and limiting hurdle in successful isolation of target-specific binders by phage display technology. To circumvent this challenge, we optimized the copy number of peptides displayed on the phage surface using type 33 phage vector. We randomized the first 67 amino acids of the wild type PIII to identify mutants that would result in its reduced expression. Consequently, the display level was improved by 30-fold due to higher incorporation of the synthetic PIII-peptide fusion protein on the phage surface. Utilization of this novel phage vector should provide a solid basis for the discovery of therapeutic peptides.
Collapse
Affiliation(s)
- Alex Chang
- Department of Pharmacy, Santa Clara Valley Medical Center, San Jose CA 95128, USA
| | - Joey P Ting
- Protein Engineering, Eli Lilly Biotechnology Center, San Diego, CA 92121, USA
| | - Alfonso Espada
- Department of Discovery Chemistry Research & Technology, Centro de Investigacion Lilly, Av. de la Industria, 30, 28108 Alcobendas, Madrid, Spain
| | - Howard Broughton
- Department of Discovery Chemistry Research & Technology, Centro de Investigacion Lilly, Av. de la Industria, 30, 28108 Alcobendas, Madrid, Spain
| | - Manuel Molina-Martin
- Department of Discovery Chemistry Research & Technology, Centro de Investigacion Lilly, Av. de la Industria, 30, 28108 Alcobendas, Madrid, Spain
| | - Sepideh Afshar
- Protein Engineering, Eli Lilly Biotechnology Center, San Diego, CA 92121, USA
| |
Collapse
|
13
|
Espada A, Haro R, Castañon J, Sayago C, Perez-Cozar F, Cano L, Redero P, Molina-Martin M, Broughton H, Stites RE, Pascal BD, Griffin PR, Dodge JA, Chalmers MJ. A Decoupled Automation Platform for Hydrogen/Deuterium Exchange Mass Spectrometry Experiments. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:2580-2583. [PMID: 31724102 DOI: 10.1007/s13361-019-02331-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 06/10/2023]
Abstract
Hydrogen/deuterium exchange mass spectrometry (HDX-MS) is a biophysical technique well suited to the characterization of protein dynamics and protein-ligand interactions. In order to accurately define the rate of exchange, HDX experiments require the repeated measure of deuterium incorporation into the target protein across a range of time points. Accordingly, the HDX-MS experiment is well suited to automation, and a number of automated systems for HDX-MS have been developed. The most widely utilized platforms all operate an integrated design, where robotic liquid handling is interfaced directly with a mass spectrometer. With integrated designs, the exchange samples are prepared and injected into the LC-MS following a "real-time" serial workflow. Here we describe a new HDX-MS platform that is comprised of two complementary pieces of automation that disconnect the sample preparation from the LC-MS analysis. For preparation, a plate-based automation system is used to prepare samples in parallel, followed by immediate freezing and storage. A second piece of automation has been constructed to perform the thawing and LC-MS analysis of frozen samples in a serial mode and has been optimized to maximize the duty cycle of the mass spectrometer. The decoupled configuration described here reduces experiment time, significantly improves capacity, and improves the flexibility of the platform when compared with a fully integrated system.
Collapse
Affiliation(s)
- Alfonso Espada
- Centro de Investigación Lilly, SA, Avenida de la Industria 30, 28108, Alcobendas, Spain
| | - Ruben Haro
- Centro de Investigación Lilly, SA, Avenida de la Industria 30, 28108, Alcobendas, Spain
| | - Jesus Castañon
- Centro de Investigación Lilly, SA, Avenida de la Industria 30, 28108, Alcobendas, Spain
| | - Cristina Sayago
- Centro de Investigación Lilly, SA, Avenida de la Industria 30, 28108, Alcobendas, Spain
| | - Francisco Perez-Cozar
- Centro de Investigación Lilly, SA, Avenida de la Industria 30, 28108, Alcobendas, Spain
| | - Leticia Cano
- Centro de Investigación Lilly, SA, Avenida de la Industria 30, 28108, Alcobendas, Spain
| | - Pablo Redero
- Centro de Investigación Lilly, SA, Avenida de la Industria 30, 28108, Alcobendas, Spain
| | - Manuel Molina-Martin
- Centro de Investigación Lilly, SA, Avenida de la Industria 30, 28108, Alcobendas, Spain
| | - Howard Broughton
- Centro de Investigación Lilly, SA, Avenida de la Industria 30, 28108, Alcobendas, Spain
| | - Ryan E Stites
- Eli Lilly and Company, Lilly Corporate Center, Lilly Research Laboratories, Indianapolis, IN, 46285, USA
| | - Bruce D Pascal
- Omics Informatics LLC, 1050 Bishop Street #517, Honolulu, HI, 96813, USA
| | - Patrick R Griffin
- Department of Molecular Medicine, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Jeffrey A Dodge
- Eli Lilly and Company, Lilly Corporate Center, Lilly Research Laboratories, Indianapolis, IN, 46285, USA
| | - Michael J Chalmers
- Eli Lilly and Company, Lilly Corporate Center, Lilly Research Laboratories, Indianapolis, IN, 46285, USA.
| |
Collapse
|
14
|
Yang L, Li T, Li S, Wu Y, Shi X, Jin H, Liu Z, Zhao Y, Zhang L, Lee HC, Zhang L. Rational Design and Identification of Small-Molecule Allosteric Inhibitors of CD38. Chembiochem 2019; 20:2485-2493. [PMID: 31081167 DOI: 10.1002/cbic.201900169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Indexed: 11/09/2022]
Abstract
CD38 is a multi-functional signaling enzyme that catalyzes the biosynthesis of two calcium-mobilizing second messengers: cyclic ADP-ribose and nicotinic acid adenine dinucleotide phosphate. It also regulates intracellular nicotinamide adenine dinucleotide (NAD) contents, associated with multiple pathophysiological processes such as aging and cancer. As such, enzymatic inhibitors of CD38 offer great potential in drug development. Here, through virtual screening and enzymatic assays, we discovered compound LX-102, which targets CD38 on the side opposite its enzymatic pocket with a binding affinity of 7.7 μm. It inhibits the NADase activity of CD38 with an IC50 of 14.9 μm. Surface plasmon resonance (SPR) and hydrogen/deuterium exchange and mass spectrometry experiments verified that LX-102 competitively binds to the epitope of the therapeutic SAR 650984 antibody in an allosteric manner. Molecular dynamics simulation was performed to demonstrate the binding dynamics of CD38 with the allosteric ligand. In summary, we established that the cavity to which SAR 650984 binds was an allosteric site and was accessible for the rational design of small chemical modulators of CD38. The lead compound LX-102 that we identified in this study could also be a useful tool for probing CD38 functions and promoting drug discovery.
Collapse
Affiliation(s)
- Lixin Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P. R. China
| | - Ting Li
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
| | - Songlu Li
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
| | - Yang Wu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
| | - Xiaomeng Shi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P. R. China
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P. R. China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P. R. China
| | - Yongjuan Zhao
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P. R. China
| | - Hon Cheung Lee
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P. R. China
| |
Collapse
|
15
|
Sayago C, Gonzalez Valcarcel IC, Qian Y, Lee J, Alsina-Fernandez J, Fite NC, Carrillo JJ, Zhang FF, Chalmers MJ, Dodge JA, Broughton H, Espada A. Deciphering Binding Interactions of IL-23R with HDX-MS: Mapping Protein and Macrocyclic Dodecapeptide Ligands. ACS Med Chem Lett 2018; 9:912-916. [PMID: 30258540 DOI: 10.1021/acsmedchemlett.8b00255] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/01/2018] [Indexed: 11/29/2022] Open
Abstract
Molecular characterization of the binding epitope of IL-23R and its cognate cytokine IL-23 is paramount to understand the role in autoimmune diseases and to support the discovery of new inhibitors of this protein-protein interaction. Our results revealed that HDX-MS was able to identify the binding epitope of IL-23R:IL-23, which opened the way to evaluate a peptide macrocycle described in the literature as disrupter of this autoimmune target. Thus, the characterization of the interactions of this chemotype by HDX-MS in combination with computational approaches was achieved. To our knowledge, this is the first reported structural evidence regarding the site where a small compound binds to IL-23R.
Collapse
Affiliation(s)
| | | | - Yuewei Qian
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - John Lee
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Jorge Alsina-Fernandez
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Nathan C. Fite
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Juan J. Carrillo
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Feiyu F. Zhang
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Michael J. Chalmers
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Jeffrey A. Dodge
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | | | - Alfonso Espada
- Centro de Investigación Lilly S.A., 28108-Alcobendas, Spain
| |
Collapse
|
16
|
Ting JP, Tung F, Antonysamy S, Wasserman S, Jones SB, Zhang FF, Espada A, Broughton H, Chalmers MJ, Woodman ME, Bina HA, Dodge JA, Benach J, Zhang A, Groshong C, Manglicmot D, Russell M, Afshar S. Utilization of peptide phage display to investigate hotspots on IL-17A and what it means for drug discovery. PLoS One 2018; 13:e0190850. [PMID: 29329326 PMCID: PMC5766103 DOI: 10.1371/journal.pone.0190850] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022] Open
Abstract
To date, IL-17A antibodies remain the only therapeutic approach to correct the abnormal activation of the IL-17A/IL-17R signaling complex. Why is it that despite the remarkable success of IL-17 antibodies, there is no small molecule antagonist of IL-17A in the clinic? Here we offer a unique approach to address this question. In order to understand the interaction of IL-17A with its receptor, we combined peptide discovery using phage display with HDX, crystallography, and functional assays to map and characterize hot regions that contribute to most of the energetics of the IL-17A/IL-17R interaction. These functional maps are proposed to serve as a guide to aid in the development of small molecules that bind to IL-17A and block its interaction with IL-17RA.
Collapse
Affiliation(s)
- Joey P. Ting
- Department of protein Engineering, Eli Lilly Biotechnology Center, San Diego, California, United States of America
| | - Frances Tung
- Department of structural Biology, Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California, United States of America
| | - Stephen Antonysamy
- Department of structural Biology, Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California, United States of America
| | - Stephen Wasserman
- Department of structural Biology, Discovery Chemistry Research and Technologies, Eli Lilly and Company, Advanced Photon Source, Argonne, Illinois, United States of America
| | - Spencer B. Jones
- Lilly Research Laboratories, Indianapolis, Indiana, United States of America
| | - Feiyu F. Zhang
- Department of structural Biology, Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California, United States of America
| | | | | | - Michael J. Chalmers
- Lilly Research Laboratories, Indianapolis, Indiana, United States of America
| | - Michael E. Woodman
- Lilly Research Laboratories, Indianapolis, Indiana, United States of America
| | - Holly A. Bina
- Lilly Research Laboratories, Indianapolis, Indiana, United States of America
| | - Jeffrey A. Dodge
- Lilly Research Laboratories, Indianapolis, Indiana, United States of America
| | - Jordi Benach
- Department of structural Biology, Discovery Chemistry Research and Technologies, Eli Lilly and Company, Advanced Photon Source, Argonne, Illinois, United States of America
| | - Aiping Zhang
- Department of structural Biology, Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California, United States of America
| | - Christopher Groshong
- Department of structural Biology, Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California, United States of America
| | - Danalyn Manglicmot
- Department of structural Biology, Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California, United States of America
| | - Marijane Russell
- Department of structural Biology, Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California, United States of America
| | - Sepideh Afshar
- Department of protein Engineering, Eli Lilly Biotechnology Center, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
17
|
Fast CS, Vahidi S, Konermann L. Changes in Enzyme Structural Dynamics Studied by Hydrogen Exchange-Mass Spectrometry: Ligand Binding Effects or Catalytically Relevant Motions? Anal Chem 2017; 89:13326-13333. [DOI: 10.1021/acs.analchem.7b03506] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Courtney S. Fast
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Siavash Vahidi
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Lars Konermann
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| |
Collapse
|
18
|
Wang W, Groves MR, Dömling A. Artificial Macrocycles as IL-17A/IL-17RA Antagonists. MEDCHEMCOMM 2017; 9:22-26. [PMID: 29750108 DOI: 10.1039/c7md00464h] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Interleukin 17(A) is a pro-inflammatory cytokine involved in several auto-immune and inflammatory diseases. Current antagonists against IL17(A) or its receptor (IL17R) that show efficacy in clinical trials are monoclonal-antibodies (mAbs). However, recently designed artificial macrocyles are potent IL17-IL17R antagonists. Based on Co-crystal structures, a better understanding the biological activity and SAR of the macrocycles has been elucidated, demonstrating that macrocycles can compete with mAbs for difficult targets such as PPIs.
Collapse
Affiliation(s)
- Wenjia Wang
- Department of Drug Design, University of Groningen, A. Deusinglaan 1, Groningen, The Netherlands
| | - Matthew R Groves
- Department of Drug Design, University of Groningen, A. Deusinglaan 1, Groningen, The Netherlands
| | - Alexander Dömling
- Department of Drug Design, University of Groningen, A. Deusinglaan 1, Groningen, The Netherlands
| |
Collapse
|
19
|
Masson GR, Jenkins ML, Burke JE. An overview of hydrogen deuterium exchange mass spectrometry (HDX-MS) in drug discovery. Expert Opin Drug Discov 2017; 12:981-994. [PMID: 28770632 DOI: 10.1080/17460441.2017.1363734] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Hydrogen deuterium exchange mass spectrometry (HDX-MS) is a powerful methodology to study protein dynamics, protein folding, protein-protein interactions, and protein small molecule interactions. The development of novel methodologies and technical advancements in mass spectrometers has greatly expanded the accessibility and acceptance of this technique within both academia and industry. Areas covered: This review examines the theoretical basis of how amide exchange occurs, how different mass spectrometer approaches can be used for HDX-MS experiments, as well as the use of HDX-MS in drug development, specifically focusing on how HDX-MS is used to characterize bio-therapeutics, and its use in examining protein-protein and protein small molecule interactions. Expert opinion: HDX-MS has been widely accepted within the pharmaceutical industry for the characterization of bio-therapeutics as well as in the mapping of antibody drug epitopes. However, there is room for this technique to be more widely used in the drug discovery process. This is particularly true in the use of HDX-MS as a complement to other high-resolution structural approaches, as well as in the development of small molecule therapeutics that can target both active-site and allosteric binding sites.
Collapse
Affiliation(s)
- Glenn R Masson
- a Protein and Nucleic Acid Chemistry Division , MRC Laboratory of Molecular Biology , Cambridge , UK
| | - Meredith L Jenkins
- b Department of Biochemistry and Microbiology , University of Victoria , Victoria , Canada
| | - John E Burke
- b Department of Biochemistry and Microbiology , University of Victoria , Victoria , Canada
| |
Collapse
|
20
|
Finn MG. Technical Advances in Medicinal Chemistry. ACS COMBINATORIAL SCIENCE 2017; 19:277-278. [PMID: 28374995 DOI: 10.1021/acscombsci.7b00053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
21
|
Djuric SW, Meanwell NA. Journal of Medicinal Chemistry, Technological Advances: Highlights 2015–2016. J Med Chem 2016; 60:1-3. [DOI: 10.1021/acs.jmedchem.6b01600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Stevan W. Djuric
- AbbVie, R467, AP10-2, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Nicholas A. Meanwell
- Bristol-Myers Squibb Research and Development, 5 Research
Parkway, Wallingford, Connecticut 06492, United States
| |
Collapse
|