96 results on '"Sleeman MA"'
Search Results
2. Novel methodology to discern predictors of remission and patterns of disease activity over time using rheumatoid arthritis clinical trials data
- Author
-
Tom, BDM, Symmons, D, Brockbank, S, Carini, C, Cope, AP, Ehrenstein, MR, Fisher, BA, Goodyear, CS, Gozzard, N, Harris, R, Hicks, K, Hollis, S, Hughes-Morley, A, Isaacs, J, Kola, B, McInnes, IB, Mela, CM, Parker, G, Pedersen, AW, Ponchel, F, Sabin, T, Scott, DL, Scott, IC, Sleeman, MA, Taylor, PC, Tsuji, W, Zhong, Y, Hilkens, C, Anderson, A, Stocks, P, Lendrem, D, Tarn, J, Smith, G, Allen, B, Casement, J, Diboll, J, Harry, R, Simpson, G, Toward, R, Noble, H, Parke, A, Wu, W, Clarke, F, Galloway, J, Lempp, H, Ibrahim, F, Schwank, S, Molyneux, G, Lazarov, T, Geissmann, F, Donnelly, I, Gilmour, A, Virlan, AT, Porter, D, Emery, P, El-Jawhari, J, Parmar, R, McDermot, MF, Buch, M, Buckley, C, Young, SP, Jones, P, Raza, K, Filer, A, Pitzalis, C, Barnes, MR, Watson, DS, Tzanis, E, Thorborn, G, Fossati-Jimack, L, Kelly, S, Humby, F, Bombardieri, M, Rana, S, Jia, Z, Goldmann, K, Lewis, M, Altobelli, G, John, C, Martins, S, Nguyen, D, Ali, H, Ciurtin, C, Worthington, J, Bruce, IN, Sergeant, JC, Verstappen, SMM, Stirling, F, Farewell, V, Keidel, S, Cuff, C, Levesque, M, Long, A, Liu, Z, Lipsky, S, Harvey, B, Macoritto, M, Hong, F, Kaymakcalan, S, Ward, N, Talbot, S, Padhji, D, Sleeman, M, Finch, D, Herath, A, Lindholm, C, Jenkins, M, Ho, M, Marshall, C, Page, M, Edwards, H, Cuza, A, Rowe, A, Capdevila, FB, Loza, M, Curran, M, Verbeeck, D, Baker, D, Vranic, I, Mela, CT, Wright, S, Rowell, L, Vernon, E, Joseph, N, Payne, N, Rao, R, Binks, M, Belson, A, Ludbrook, V, Tipney, H, Ellis, J, Hasan, S, Didierlaurent, A, Burny, W, Haynes, A, Larminie, C, Dastros-Pitel, D, Jelinsky, S, Hodge, M, Maciejewski, M, Ziemek, D, Schulz-Knappe, P, Zucht, H-D, Budde, P, Coles, MC, Butler, JA, Read, S, and Consortium, RA-MAP
- Subjects
musculoskeletal diseases ,immune system diseases ,skin and connective tissue diseases - Abstract
Objectives: To identify predictors of remission and disease activity patterns in patients with rheumatoid arthritis (RA) using individual participant data (IPD) from clinical trials. Methods: Phase II and III clinical trials completed between 2002 and 2012 were identified by systematic literature review and contact with UK market authorisation holders. Anonymised baseline and follow-up IPD from non-biological arms were amalgamated. Multiple imputation was used to handle missing outcome and covariate information. Random effects logistic regression was used to identify predictors of remission, measured by the Disease Activity Score 28 (DAS28) at 6 months. Novel latent class mixed models characterised DAS28 over time. Results: IPD of 3290 participants from 18 trials were included. Of these participants, 92% received methotrexate (MTX). Remission rates were estimated at 8.4%(95%CI 7.4%to9.5%) overall, 17%(95%CI 14.8%to19.4%) for MTX-naïve patients with early RA and 3.2% (95% CI 2.4% to 4.3%) for those with prior MTX exposure at entry. In prior MTX-exposed patients, lower baseline DAS28 and MTX reinitiation were associated with remission. In MTX-naïve patients, being young, white, male, with better functional and mental health, lower baseline DAS28 and receiving concomitant glucocorticoids were associated with remission. Three DAS28 trajectory subpopulations were identified in MTX-naïve and MTX-exposed patients. A number of variables were associated with subpopulation membership and DAS28 levels within subpopulations. Conclusions: Predictors of remission differed between MTX-naïve and prior MTX-exposed patients at entry. Latent class mixed models supported differential non-biological therapy response, with three distinct trajectories observed in both MTX-naïve and MTX-exposed patients. Findings should be useful when designing future RA trials and interpreting results of biomarker studies.
- Published
- 2018
3. The RA-MAP Consortium: a working model for academia-industry collaboration
- Author
-
Cope, AP, Barnes, MR, Belson, A, Binks, M, Brockbank, S, Bonachela-Capdevila, F, Carini, C, Fisher, BA, Goodyear, CS, Emery, P, Ehrenstein, MR, Gozzard, N, Harris, R, Hollis, S, Keidel, S, Levesque, M, Lindholm, C, McDermott, MF, McInnes, IB, Mela, CM, Parker, G, Read, S, Pedersen, AW, Ponchel, F, Porter, D, Rao, R, Rowe, A, Schulz-Knappe, P, Sleeman, MA, Symmons, D, Taylor, PC, Tom, B, Tsuji, W, Verbeeck, D, Isaacs, JD, Consortium, Ra-Map, Tom, Brian [0000-0002-3335-9322], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Biomedical Research ,Big data ,MEDLINE ,Globe ,History, 21st Century ,Article ,Arthritis, Rheumatoid ,03 medical and health sciences ,Rheumatology ,medicine ,Genomic medicine ,Humans ,Industry ,Cooperative Behavior ,business.industry ,Research ,Genomics ,Public relations ,United Kingdom ,030104 developmental biology ,medicine.anatomical_structure ,Phenotype ,Cooperative behavior ,business ,Biomarkers - Abstract
Collaboration can be challenging; nevertheless, the emerging successes of large, multi-partner, multi-national cooperatives and research networks in the biomedical sector have sustained the appetite of academics and industry partners for developing and fostering new research consortia. This model has percolated down to national funding agencies across the globe, leading to funding for projects that aim to realise the true potential of genomic medicine in the 21st century and to reap the rewards of ‘big data’. In this Perspectives article, the experiences of the RA-MAP consortium, a group of more than 140 individuals affiliated with 21 academic and industry organizations that are focused on making genomic medicine in rheumatoid arthritis a reality are described. The challenges of multi-partner collaboration in the UK are highlighted and wide-ranging solutions are offered that might benefit large research consortia around the world.
- Published
- 2018
- Full Text
- View/download PDF
4. GM-CSF primes cardiac inflammation in a mouse model of Kawasaki disease
- Author
-
Stock, AT, Hansen, JA, Sleeman, MA, McKenzie, BS, Wicks, IP, Stock, AT, Hansen, JA, Sleeman, MA, McKenzie, BS, and Wicks, IP
- Abstract
Kawasaki disease (KD) is the leading cause of pediatric heart disease in developed countries. KD patients develop cardiac inflammation, characterized by an early infiltrate of neutrophils and monocytes that precipitates coronary arteritis. Although the early inflammatory processes are linked to cardiac pathology, the factors that regulate cardiac inflammation and immune cell recruitment to the heart remain obscure. In this study, using a mouse model of KD (induced by a cell wall Candida albicans water-soluble fraction [CAWS]), we identify an essential role for granulocyte/macrophage colony-stimulating factor (GM-CSF) in orchestrating these events. GM-CSF is rapidly produced by cardiac fibroblasts after CAWS challenge, precipitating cardiac inflammation. Mechanistically, GM-CSF acts upon the local macrophage compartment, driving the expression of inflammatory cytokines and chemokines, whereas therapeutically, GM-CSF blockade markedly reduces cardiac disease. Our findings describe a novel role for GM-CSF as an essential initiating cytokine in cardiac inflammation and implicate GM-CSF as a potential target for therapeutic intervention in KD.
- Published
- 2016
5. A1.10 The GM-CSF/CCL17 axis in the rheumatoid synovial environment
- Author
-
Kidger, SV, primary, Ellson, CD, additional, McInnes, IB, additional, Sleeman, MA, additional, Robinson, MJ, additional, and Goodyear, CS, additional
- Published
- 2016
- Full Text
- View/download PDF
6. Protein engineering and preclinical development of a GM-CSF receptor antibody for the treatment of rheumatoid arthritis
- Author
-
Minter, RR, Cohen, ES, Wang, B, Liang, M, Vainshtein, I, Rees, G, Eghobamien, L, Harrison, P, Sims, DA, Matthews, C, Wilkinson, T, Monk, P, Drinkwater, C, Fabri, L, Nash, A, McCourt, M, Jermutus, L, Roskos, L, Anderson, IK, and Sleeman, MA
- Subjects
Male ,Protein Engineering ,Research Papers ,Models, Biological ,Antibodies ,Recombinant Proteins ,Arthritis, Rheumatoid ,Inhibitory Concentration 50 ,Macaca fascicularis ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Immunoglobulin G ,Animals ,Humans ,Female ,Cell Surface Display Techniques ,Protein Binding - Abstract
For antibody therapies against receptor targets, in vivo outcomes can be difficult to predict because of target-mediated clearance or antigen 'sink' effects. The purpose of this work was to engineer an antibody to the GM-CSF receptor α (GM-CSFRα) with pharmacological properties optimized for chronic, s.c. treatment of rheumatoid arthritis (RA) patients.We used an in silico model of receptor occupancy to guide the target affinity and a combinatorial phage display approach for affinity maturation. Mechanism of action and internalization assays were performed on the optimized antibody in vitro before refining the modelling predictions of the eventual dosing in man. Finally, in vivo pharmacology studies in cynomolgus monkeys were carried out to inform the predictions and support future clinical development.Antibody potency was improved 8600-fold, and the target affinity was reached. The refined model predicted pharmacodynamic effects at doses as low as 1 mg kg(-1) and a study in cynomolgus monkeys confirmed in vivo efficacy at 1 mg kg(-1) dosing.This rational approach to antibody drug discovery enabled the isolation of a potent molecule compatible with chronic, s.c. self-administration by RA patients. We believe this general approach enables the development of optimal biopharmaceuticals.
- Published
- 2012
7. Protein engineering and preclinical development of a GM-CSF receptor antibody for the treatment of rheumatoid arthritis
- Author
-
Minter, RR, primary, Cohen, ES, additional, Wang, B, additional, Liang, M, additional, Vainshtein, I, additional, Rees, G, additional, Eghobamien, L, additional, Harrison, P, additional, Sims, DA, additional, Matthews, C, additional, Wilkinson, T, additional, Monk, P, additional, Drinkwater, C, additional, Fabri, L, additional, Nash, A, additional, McCourt, M, additional, Jermutus, L, additional, Roskos, L, additional, Anderson, IK, additional, and Sleeman, MA, additional
- Published
- 2012
- Full Text
- View/download PDF
8. Identification of a potent anti-IL-15 antibody with opposing mechanisms of action in vitro and in vivo
- Author
-
Finch, DK, primary, Midha, A, additional, Buchanan, CL, additional, Cochrane, D, additional, Craggs, RI, additional, Cruwys, S, additional, Grahames, C, additional, Kolbeck, R, additional, Lowe, DC, additional, Maltby, J, additional, Pattison, DV, additional, Vousden, KA, additional, Ward, A, additional, Sleeman, MA, additional, and Mallinder, PR, additional
- Published
- 2010
- Full Text
- View/download PDF
9. Mavrilimumab, a human monoclonal antibody targeting GM-CSF receptor-α, in subjects with rheumatoid arthritis: a randomised, double-blind, placebo-controlled, phase I, first-in-human study.
- Author
-
Burmester GR, Feist E, Sleeman MA, Wang B, White B, Magrini F, Burmester, Gerd R, Feist, Eugen, Sleeman, Matthew A, Wang, Bing, White, Barbara, and Magrini, Fabio
- Abstract
Objective: To evaluate the safety, tolerability, pharmacokinetic and pharmacodynamic profiles of mavrilimumab, a human monoclonal antibody targeting the granulocyte-macrophage colony-stimulating factor receptor-α, in subjects with rheumatoid arthritis (RA).Methods: A randomised, double-blind, placebo-controlled, dose-escalating phase I study in subjects with RA who received stable methotrexate treatment for ≥3 months before enrolment.Subjects: received single intravenous escalating doses of mavrilimumab (0.01-10.0 mg/kg) or placebo.Results: 32 subjects were enrolled in this study (1 unblinded subject at 0.01 mg/kg and another at 0.03 mg/kg were followed by five sequential double-blinded cohorts, n=6 each, treated with 0.1, 0.3, 1.0, 3.0 and 10.0 mg/kg, respectively). Adverse events were mild or moderate and were reported with similar frequency across all treatment cohorts. One subject (10.0 mg/kg) experienced moderate face and neck urticaria during infusion that resolved with symptomatic treatment. Systemic clearance of mavrilimumab approached that of endogenous IgG at doses >1.0 mg/kg; pharmacodynamic activity was confirmed in the 1.0 and 3.0 mg/kg cohorts by suppression of suppressor of cytokine signalling 3 mRNA transcripts. In exploratory analyses, reductions of acute phase reactants were observed in subjects with elevated C-reactive protein (>5 mg/l) and erythrocyte sedimentation rate (≥20.0 mm/h) at baseline. No significant change in Disease Activity Score 28-joint assessment (DAS28) was seen in any of the cohorts. In mavrilimumab-treated subjects (n=15) with baseline DAS28 >3.2, mean disease activity (DAS28) was significantly reduced at 4 weeks.Conclusion: In this first-in-human study, mavrilimumab showed preliminary evidence of pharmacodynamic activity. Importantly, the safety and pharmacokinetic profiles of mavrilimumab support further clinical studies in RA.Trial Registration Number: NCT00771420. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
10. Identification of a potent anti-IL-15 antibody with opposing mechanisms of action in vitro and in vivo.
- Author
-
Finch, DK, Midha, A, Buchanan, CL, Cochrane, D, Craggs, RI, Cruwys, S, Grahames, C, Kolbeck, R, Lowe, DC, Maltby, J, Pattison, DV, Vousden, KA, Ward, A, Sleeman, MA, Mallinder, PR, Finch, D K, Buchanan, C L, Craggs, R I, Lowe, D C, and Pattison, D V
- Subjects
INTERLEUKINS ,ANTI-antibodies ,BIOCHEMICAL mechanism of action ,LYMPHOCYTES ,CELL populations ,CELL proliferation ,INFLAMMATION ,CYTOKINES ,THERAPEUTIC use of monoclonal antibodies ,BINDING sites ,RESEARCH ,ANIMAL experimentation ,RESEARCH methodology ,CELL receptors ,MONOCLONAL antibodies ,IMMUNOLOGY technique ,CELL physiology ,MEDICAL cooperation ,EVALUATION research ,COMPARATIVE studies ,IMMUNITY ,T cells ,MICE ,CHEMICAL inhibitors - Abstract
Background and Purpose: Interleukin-15 (IL-15) is important in the activation and proliferation of lymphocytic cell populations and is implicated in inflammatory disease. We report the characterization of a novel monoclonal antibody DISC0280 which is specific for human IL-15.Experimental Approach: DISC0280 was characterized in a direct binding assay of IL-15 with IL-15 receptor α (IL-15Rα) and by its ability to alter IL-15 mediated proliferation of a range of cell lines (cytotoxic T lymphocyte line-2, M-07e, KIT225). A pharmacodynamic model injecting male C57/BL6 mice with IL-15 or IL-15/IL-15Rα, with or without DISC0280, and assessing changes in lymphocytic cell populations and serum cytokines was utilized.Key Results: DISC0280 inhibited the binding of IL-15 to IL-15Rα and also potently inhibits IL-15 dependent proliferation of cells expressing IL-15Rα, shared interleukin 2/ interleukin 15 receptor β chain (IL-15Rβ) and common gamma chain (γ(c) ). DISC0280 also inhibited the IL-15 dependent proliferation of M-07e cells that only express IL-15Rβ/γ(c) subunits. Human IL-15 injected into mice caused an increase in NK1.1(+) and CD3(+) cells in the spleen and peripheral blood and these effects were unexpectedly potentiated by giving DISC0280 with human IL-15. This increase in cells caused by DISC0280/IL-15 co-administration was greater than that observed when IL-15 was administered complexed with soluble IL-15Rα.Conclusions and Implications: The ability of DISC0280 to bind to the IL-15Rα-binding site on IL-15 allows trans-presentation of IL-15 by DISC0280 in vivo, similar to the trans-presentation by soluble IL-15Rα. DISC0280 may be therefore suitable as a clinical substitute for IL-15. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
11. Potential immunomodulatory effects of CAS+IMD monoclonal antibody cocktail in hospitalized patients with COVID-19.
- Author
-
Wang B, Golubov J, Oswald EM, Poon P, Wei Q, Lett C, Shehadeh F, Kaczynski M, Felix LO, Mishra B, Mylona EK, Wipperman MF, Chio E, Hamon SC, Hooper AT, Somersan-Karakaya S, Musser BJ, Petro CD, Hamilton JD, Sleeman MA, Kalliolias GD, Mylonakis E, and Skokos D
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal immunology, Hospitalization, Immunization, Passive methods, Immunophenotyping, Antibodies, Viral immunology, Adult, Drug Combinations, SARS-CoV-2 immunology, COVID-19 immunology, COVID-19 therapy, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Neutralizing immunology, COVID-19 Drug Treatment
- Abstract
Background: Passive administration of SARS-CoV-2 neutralizing monoclonal antibodies (mAbs), such as CAS + IMD (Casirivimab + Imdevimab) antibody cocktail demonstrated beneficial effects on clinical outcomes in hospitalized patients with COVID-19 who were seronegative at baseline and outpatients. However, little is known about their impact on the host immunophenotypes., Methods: We conducted an immunoprofiling study in 46 patients from a single site of a multi-site trial of CAS + IMD in hospitalized patients. We collected longitudinal samples during October 2020 ∼ April 2021, prior to the emergence of the Delta and Omicron variants and the use of COVID-19 vaccines. All collected samples were analyzed without exclusion and post-hoc statistical analysis was performed. We examined the dynamic interplay of CAS + IMD with host immunity applying dimensional reduction approach on plasma proteomics and high dimensional flow cytometry data., Findings: Using an unbiased clustering method, we identified unique immunophenotypes associated with acute inflammation and disease resolution. Compared to placebo group, administration of CAS + IMD accelerated the transition from an acute inflammatory immunophenotype, to a less inflammatory or "resolving" immunophenotype, as characterized by reduced tissue injury, proinflammatory markers and restored lymphocyte/monocyte imbalance independent of baseline serostatus. Moreover, CAS + IMD did not impair the magnitude or the quality of host T cell immunity against SARS-CoV-2 spike protein., Interpretation: Our results identified immunophenotypic changes indicative of a possible SARS-CoV-2 neutralizing antibodies-induced anti-inflammatory effect, without an evident impairment of cellular antiviral immunity, suggesting that further studies of Mabs effects on SAS-CoV-2 or other viral mediated inflammation are warranted., Funding: Regeneron Pharmaceuticals Inc and federal funds from the Department of Health and Human Services; Administration for Strategic Preparedness and Response; Biomedical Advanced Research and Development Authority, under OT number: HHSO100201700020C., Competing Interests: Declaration of interests B.W., J.G., P.P, Q.W., C.L., M.F.W., E.C., S.C.H., A.T.H., S.K., G.D.K., B.J.M., C.D.P., J.D.H., M.A.S., D.S. are employees of Regeneron Pharmaceuticals, Inc. E.M.O is former employee of Regeneron. E.M. (Eleftherios Mylonakis) received fundings from Regeneron Pharmaceuticals, SciClone Pharmaceuticals, Pfizer, Chemic Labs/KODA Therapeutics, Cidara, and Leidos Biomedical Research Inc./NCI, NIH/NIAID, NIH/NIGMS, and BARDA., (Copyright © 2024 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. A therapeutic strategy to target distinct sources of IgE and durably reverse allergy.
- Author
-
Limnander A, Kaur N, Asrat S, Tasker C, Boyapati A, Ben LH, Janczy J, Pedraza P, Abreu P, Chen WC, Godin S, Daniel BJ, Chin H, DeVeaux M, Rodriguez Lorenc K, Sirulnik A, Harari O, Stahl N, Sleeman MA, Murphy AJ, Yancopoulos GD, and Orengo JM
- Subjects
- Mice, Humans, Animals, Macaca fascicularis, Plasma Cells, Allergens, Immunoglobulin E, Anaphylaxis
- Abstract
Immunoglobulin E (IgE) is a key driver of type 1 hypersensitivity reactions and allergic disorders, which are globally increasing in number and severity. Although eliminating pathogenic IgE may be a powerful way to treat allergy, no therapeutic strategy reported to date can fully ablate IgE production. Interleukin-4 receptor α (IL-4Rα) signaling is required for IgE class switching, and IL-4Rα blockade gradually reduces, but does not eliminate, IgE. The persistence of IgE after IL-4Rα blockade may be due to long-lived IgE
+ plasma cells that maintain serological memory to allergens and thus may be susceptible to plasma cell-targeted therapeutics. We demonstrate that transient administration of a B cell maturation antigen x CD3 (BCMAxCD3) bispecific antibody markedly depletes IgE, as well as other immunoglobulins, by ablating long-lived plasma cells, although IgE and other immunoglobulins rapidly rebound after treatment. Concomitant IL-4Rα blockade specifically and durably prevents the reemergence of IgE by blocking IgE class switching while allowing the restoration of other immunoglobulins. Moreover, this combination treatment prevented anaphylaxis in mice. Together with additional cynomolgus monkey and human data, our studies demonstrate that allergic memory is primarily maintained by both non-IgE+ memory B cells that require class switching and long-lived IgE+ plasma cells. Our combination approach to durably eliminate pathogenic IgE has potential to benefit allergy in humans while preserving antibody-mediated immunity.- Published
- 2023
- Full Text
- View/download PDF
13. VSIG4 interaction with heparan sulfates inhibits VSIG4-complement binding.
- Author
-
Ebstein SY, Rafique A, Zhou Y, Krasco A, Montalvo-Ortiz W, Yu L, Custodio L, Adam RC, Bloch N, Lee K, Adewale F, Vergata D, Luz A, Coquery S, Daniel B, Ullman E, Franklin MC, Hermann A, Huang T, Olson W, Davis S, Murphy AJ, Sleeman MA, Wei J, and Skokos D
- Subjects
- Receptors, Complement genetics, Receptors, Complement metabolism, Cell Membrane metabolism, Sulfates, Heparitin Sulfate metabolism, Glycosaminoglycans metabolism
- Abstract
V-set and immunoglobulin domain-containing 4 (VSIG4) is a complement receptor of the immunoglobulin superfamily that is specifically expressed on tissue resident macrophages, and its many reported functions and binding partners suggest a complex role in immune function. VSIG4 is reported to have a role in immune surveillance as well as in modulating diverse disease phenotypes such as infections, autoimmune conditions, and cancer. However, the mechanism(s) governing VSIG4's complex, context-dependent role in immune regulation remains elusive. Here, we identify cell surface and soluble glycosaminoglycans, specifically heparan sulfates, as novel binding partners of VSIG4. We demonstrate that genetic deletion of heparan sulfate synthesis enzymes or cleavage of cell-surface heparan sulfates reduced VSIG4 binding to the cell surface. Furthermore, binding studies demonstrate that VSIG4 interacts directly with heparan sulfates, with a preference for highly sulfated moieties and longer glycosaminoglycan chains. To assess the impact on VSIG4 biology, we show that heparan sulfates compete with known VSIG4 binding partners C3b and iC3b. Furthermore, mutagenesis studies indicate that this competition occurs through overlapping binding epitopes for heparan sulfates and complement on VSIG4. Together these data suggest a novel role for heparan sulfates in VSIG4-dependent immune modulation., (© The Author(s) 2023. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
14. TRAPnSeq allows high-throughput profiling of antigen-specific antibody-secreting cells.
- Author
-
Asrat S, Devlin JC, Vecchione A, Klotz B, Setliff I, Srivastava D, Limnander A, Rafique A, Adler C, Porter S, Murphy AJ, Atwal GS, Sleeman MA, Lim WK, and Orengo JM
- Subjects
- Humans, Animals, Mice, B-Lymphocytes, Antibodies genetics, Receptors, Antigen, B-Cell genetics, Antibody-Producing Cells, Antigens
- Abstract
Following activation by cognate antigen, B cells undergo fine-tuning of their antigen receptors and may ultimately differentiate into antibody-secreting cells (ASCs). While antigen-specific B cells that express surface receptors (B cell receptors [BCRs]) can be readily cloned and sequenced following flow sorting, antigen-specific ASCs that lack surface BCRs cannot be easily profiled. Here, we report an approach, TRAPnSeq (antigen specificity mapping through immunoglobulin [Ig] secretion TRAP and Sequencing), that allows capture of secreted antibodies on the surface of ASCs, which in turn enables high-throughput screening of single ASCs against large antigen panels. This approach incorporates flow cytometry, standard microfluidic platforms, and DNA-barcoding technologies to characterize antigen-specific ASCs through single-cell V(D)J, RNA, and antigen barcode sequencing. We show the utility of TRAPnSeq by profiling antigen-specific IgG and IgE ASCs from both mice and humans and highlight its capacity to accelerate therapeutic antibody discovery from ASCs., Competing Interests: This study was sponsored by Regeneron Pharmaceuticals, Inc. All authors are current or former employees of Regeneron and may hold stock options in the company. S.A., J.C.D., A.V., B.K., I.S., G.S.A., M.A.S., W.K.L., and J.M.O. are inventors on a pending US patent application on TRAPnSeq (“Methods of Mapping Antigen Specificity to Antibody-Secreting Cells”). A.J.M. is an inventor on a pending US patent application (#16/363,774; “Humanized Rodents for Testing Therapeutic Agents”)., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
15. Autoantibodies are associated with disease progression in idiopathic pulmonary fibrosis.
- Author
-
Koether K, Besnard V, Sandig H, Carruthers A, Miranda E, Grootenboer-Mignot S, Taillé C, Chevret S, Valeyre D, Nunes H, Israel-Biet D, Lim WK, Cottin V, Corkill D, Dobson C, Groves M, Ferraro F, Guenzi E, Huang L, Sulikowski M, Mailleux A, Murray LA, Mustelin T, Strickland I, Sleeman MA, and Crestani B
- Subjects
- Humans, Lung metabolism, Disease Progression, Fibroblasts metabolism, Autoantibodies, Idiopathic Pulmonary Fibrosis drug therapy
- Abstract
Several reports have highlighted a potential role of autoreactive B-cells and autoantibodies that correlates with increased disease severity in patients with idiopathic pulmonary fibrosis (IPF). Here we show that patients with IPF have an altered B-cell phenotype and that those subjects who have autoantibodies against the intermediate filament protein periplakin (PPL) have a significantly worse outcome in terms of progression-free survival. Using a mouse model of lung fibrosis, we demonstrate that introducing antibodies targeting the endogenous protein PPL (mimicking naturally occurring autoantibodies seen in patients) directly in the lung increases lung injury, inflammation, collagen and fibronectin expression through direct activation of follicular dendritic cells, which in turn activates and drives proliferation of fibroblasts. This fibrocyte population was also observed in fibrotic foci of patients with IPF and was increased in peripheral blood of IPF patients compared to aged-matched controls. This study reiterates the complex and heterogeneous nature of IPF, identifying new pathways that may prove suitable for therapeutic intervention., Competing Interests: Conflict of interest: C. Taillé reports grants from GSK, Sanofi and AstraZeneca, advisory board participation and consulting fees from GSK, Sanofi, AstraZeneca and Novartis, lecture honoraria from GSK, Sanofi, AstraZeneca, Stallergenes and Novartis, and travel support from GSK and AstraZeneca, outside the submitted work. D. Valeyre reports travel support from ERS, ATS and CPLF, and advisory board participation with Roche and Boehringer Ingelheim, outside the submitted work. H. Nunes reports consulting fees, lecture honoraria and travel support from Boehringer Ingelheim and Roche/Genentech, and advisory board participation with Galapagos, outside the submitted work. W.K. Lim reports options and stock as part of employee compensation from Regeneron Pharmaceuticals, outside the submitted work. V. Cottin reports grants from Boehringer Ingelheim, consulting fees from Boehringer Ingelheim and Roche/Promedior, lecture honoraria from Boehringer Ingelheim, Roche/Promedior, Sanofi and AstraZeneca, travel support from Boehringer Ingelheim, Roche/Promedior and AstraZeneca, and advisory board participation with Actelion, Bayer/MSD, Roche/Promedior, Sanofi, Celgene/BMS, Galapagos, Galecto, Shionogi, Fibrogen, RedX and PureTech, outside the submitted work. D. Corkill reports support for the present manuscript from AstraZeneca/MedImmune, as a full-time employee. T. Mustelin reports grants from NIH, and consulting fees from Miro Bio, Kiniksa, Cugene, QiLu and ROME, outside the submitted work. M.A. Sleeman reports support for the present manuscript from MedImmune/AstraZeneca, and during the research was a full-time employee of MedImmune/AstraZeneca. B. Crestani reports support for the present manuscript from MedImmune/AstraZeneca; B. Crestani also reports grants from Boehringer Ingelheim, Roche, BMS and Translate Bio, consulting fees from Apellis, Boehringer Ingelheim, Roche, BMS, Translate Bio, Sanofi and Novartis, lecture honoraria from AstraZeneca, Boehringer Ingelheim, Roche, BMS and Sanofi, travel support from AstraZeneca, BMS, Boehringer Ingelheim, Roche and Novartis, and is the Vice President of the Fondation du Souffle, outside the submitted work. All other authors have nothing to disclose., (Copyright ©The authors 2023. For reproduction rights and permissions contact permissions@ersnet.org.)
- Published
- 2023
- Full Text
- View/download PDF
16. IL-4 and IL-13, not eosinophils, drive type 2 airway inflammation, remodeling and lung function decline.
- Author
-
Scott G, Asrat S, Allinne J, Keat Lim W, Nagashima K, Birchard D, Srivatsan S, Ajithdoss DK, Oyejide A, Ben LH, Walls J, Le Floc'h A, Yancopoulos GD, Murphy AJ, Sleeman MA, and Orengo JM
- Subjects
- Animals, Humans, Mice, Chemokines metabolism, Cytokines metabolism, Endothelial Cells metabolism, Inflammation metabolism, Interleukin-13 metabolism, Lung metabolism, Interleukin-4 pharmacology, Asthma metabolism, Pneumonia metabolism
- Abstract
Rationale: Type 2 (T2) asthma is characterized by airflow limitations and elevated levels of blood and sputum eosinophils, fractional exhaled nitric oxide, IgE, and periostin. While eosinophils are associated with exacerbations, the contribution of eosinophils to lung inflammation, remodeling and function remains largely hypothetical., Objectives: To determine the effect of T2 cytokines IL-4, IL-13 and IL-5 on eosinophil biology and compare the impact of depleting just eosinophils versus inhibiting all aspects of T2 inflammation on airway inflammation., Methods: Human eosinophils or endothelial cells stimulated with IL-4, IL-13 or IL-5 were assessed for gene changes or chemokine release.Mice exposed to house dust mite extract received anti-IL-4Rα (dupilumab), anti-IL-5 or control antibodies and were assessed for changes in lung histological and inflammatory endpoints., Measurements and Main Results: IL-4 or IL-13 stimulation of human eosinophils and endothelial cells induced gene expression changes related to granulocyte migration; whereas, IL-5 induced changes reflecting granulocyte differentiation.In a mouse model, blocking IL-4Rα improved lung function by impacting multiple effectors of inflammation and remodeling, except peripheral eosinophil counts, thereby disconnecting blood eosinophils from airway inflammation, remodeling and function. Blocking IL-5 globally reduced eosinophil counts but did not impact inflammatory or functional measures of lung pathology. Whole lung transcriptome analysis revealed that IL-5 or IL-4Rα blockade impacted eosinophil associated genes, whereas IL-4Rα blockade also impacted genes associated with multiple cells, cytokines and chemokines, mucus production, cell:cell adhesion and vascular permeability., Conclusions: Eosinophils are not the sole contributor to asthma pathophysiology or lung function decline and emphasizes the need to block additional mediators to modify lung inflammation and impact lung function., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: All authors reports financial support was provided by Regeneron Pharmaceuticals, Inc. and Sanofi-Aventis France SA. All authors reports a relationship with Regeneron Pharmaceuticals Inc that includes: current or former employment and equity or stock or stock options. Andrew Murphy has patent #HUMANIZED IL-4 AND IL-4Ra ANIMALS issued to Regeneron Pharmaceuticals., (Copyright © 2022 Regeneron Pharmaceuticals. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Blocking common γ chain cytokine signaling ameliorates T cell-mediated pathogenesis in disease models.
- Author
-
Le Floc'h A, Nagashima K, Birchard D, Scott G, Ben LH, Ajithdoss D, Gayvert K, Romero Hernandez A, Herbin O, Tay A, Farrales P, Korgaonkar CK, Pan H, Shah S, Kamat V, Chatterjee I, Popke J, Oyejide A, Lim WK, Kim JH, Huang T, Franklin M, Olson W, Norton T, Perlee L, Yancopoulos GD, Murphy AJ, Sleeman MA, and Orengo JM
- Subjects
- Animals, Mice, Antibodies, Monoclonal metabolism, Cytokines metabolism, Signal Transduction, Primates, Anemia, Aplastic metabolism, Graft vs Host Disease metabolism, T-Lymphocytes metabolism, T-Lymphocytes pathology, Interleukin Receptor Common gamma Subunit antagonists & inhibitors, Interleukin Receptor Common gamma Subunit metabolism
- Abstract
The common γ chain (γc; IL-2RG) is a subunit of the interleukin (IL) receptors for the γc cytokines IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. The lack of appropriate neutralizing antibodies recognizing IL-2RG has made it difficult to thoroughly interrogate the role of γc cytokines in inflammatory and autoimmune disease settings. Here, we generated a γc cytokine receptor antibody, REGN7257, to determine whether γc cytokines might be targeted for T cell-mediated disease prevention and treatment. Biochemical, structural, and in vitro analysis showed that REGN7257 binds with high affinity to IL-2RG and potently blocks signaling of all γc cytokines. In nonhuman primates, REGN7257 efficiently suppressed T cells without affecting granulocytes, platelets, or red blood cells. Using REGN7257, we showed that γc cytokines drive T cell-mediated disease in mouse models of graft-versus-host disease (GVHD) and multiple sclerosis by affecting multiple aspects of the pathogenic response. We found that our xenogeneic GVHD mouse model recapitulates hallmarks of acute and chronic GVHD, with T cell expansion/infiltration into tissues and liver fibrosis, as well as hallmarks of immune aplastic anemia, with bone marrow aplasia and peripheral cytopenia. Our findings indicate that γc cytokines contribute to GVHD and aplastic anemia pathology by promoting these characteristic features. By demonstrating that broad inhibition of γc cytokine signaling with REGN7257 protects from immune-mediated disorders, our data provide evidence of γc cytokines as key drivers of pathogenic T cell responses, offering a potential strategy for the management of T cell-mediated diseases.
- Published
- 2023
- Full Text
- View/download PDF
18. Antibodies as drugs-a Keystone Symposia report.
- Author
-
Cable J, Saphire EO, Hayday AC, Wiltshire TD, Mousa JJ, Humphreys DP, Breij ECW, Bruhns P, Broketa M, Furuya G, Hauser BM, Mahévas M, Carfi A, Cantaert T, Kwong PD, Tripathi P, Davis JH, Brewis N, Keyt BA, Fennemann FL, Dussupt V, Sivasubramanian A, Kim PM, Rawi R, Richardson E, Leventhal D, Wolters RM, Geuijen CAW, Sleeman MA, Pengo N, and Donnellan FR
- Subjects
- Humans, Immunotherapy, Antibodies, Bispecific therapeutic use
- Abstract
Therapeutic antibodies have broad indications across diverse disease states, such as oncology, autoimmune diseases, and infectious diseases. New research continues to identify antibodies with therapeutic potential as well as methods to improve upon endogenous antibodies and to design antibodies de novo. On April 27-30, 2022, experts in antibody research across academia and industry met for the Keystone symposium "Antibodies as Drugs" to present the state-of-the-art in antibody therapeutics, repertoires and deep learning, bispecific antibodies, and engineering., (© 2022 New York Academy of Sciences.)
- Published
- 2023
- Full Text
- View/download PDF
19. Comparison of the Inflammatory Circuits in Psoriasis Vulgaris, Non‒Pustular Palmoplantar Psoriasis, and Palmoplantar Pustular Psoriasis.
- Author
-
Wang CQ, Haxhinasto S, Garcet S, Kunjravia N, Cueto I, Gonzalez J, Rambhia D, Harari O, Sleeman MA, Hamilton JD, Lim WK, Freudenberg J, Kalliolias GD, Thakker P, Bissonnette R, and Krueger JG
- Subjects
- Humans, Skin pathology, Inflammation pathology, Psoriasis, Skin Diseases pathology
- Abstract
Palmoplantar pustular psoriasis (PPPP) and non‒pustular palmoplantar psoriasis (NPPP) are localized, debilitating forms of psoriasis. The inflammatory circuits involved in PPPP and NPPP are not well-understood. To compare the cellular and immunological features that differentiate PPPP and NPPP, skin biopsies were collected from a total of 30 participants with PPPP, NPPP, and psoriasis vulgaris (PV) and from 10 healthy participants. A subset consented to a second biopsy after 3 additional weeks off medication. Histologic staining of lesional and nonlesional skin showed higher neutrophil counts in PPPP than in NPPP and PV and higher CD8
+ T-cell counts in NPPP. RNA sequencing and transcriptional analysis of skin biopsies showed enhanced IFN-γ pathway activation in NPPP lesions but stronger signatures of IL-17 pathway and neutrophil-related genes (e.g., IL36A) in PPPP lesional skin. Serum analysis on the Olink platform detected higher concentrations of T helper type 1, IFN-γ‒inducible chemokines in NPPP, and higher neutrophil-associated cytokines in PPPP. Taken together, this evidence suggests more pronounced T helper 1‒mediated inflammation in NPPP than in PV and PPPP and stronger neutrophil-associated activity in PPPP than in NPPP and PV. These data support targeting inflammatory pathways associated with neutrophilic inflammation (e.g., IL-36 signaling) for therapeutic development in PPPP., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
20. Cancer cell-derived type I interferons instruct tumor monocyte polarization.
- Author
-
Kwart D, He J, Srivatsan S, Lett C, Golubov J, Oswald EM, Poon P, Ye X, Waite J, Zaretsky AG, Haxhinasto S, Au-Yeung E, Gupta NT, Chiu J, Adler C, Cherravuru S, Malahias E, Negron N, Lanza K, Coppola A, Ni M, Song H, Wei Y, Atwal GS, Macdonald L, Oristian NS, Poueymirou W, Jankovic V, Fury M, Lowy I, Murphy AJ, Sleeman MA, Wang B, and Skokos D
- Subjects
- Humans, Mice, Animals, Monocytes, Interferon Type I, Neoplasms
- Abstract
Monocytes are highly plastic immune cells that modulate antitumor immunity. Therefore, identifying factors that regulate tumor monocyte functions is critical for developing effective immunotherapies. Here, we determine that endogenous cancer cell-derived type I interferons (IFNs) control monocyte functional polarization. Guided by single-cell transcriptomic profiling of human and mouse tumors, we devise a strategy to distinguish and separate immunostimulatory from immunosuppressive tumor monocytes by surface CD88 and Sca-1 expression. Leveraging this approach, we show that cGAS-STING-regulated cancer cell-derived IFNs polarize immunostimulatory monocytes associated with anti-PD-1 immunotherapy response in mice. We also demonstrate that immunosuppressive monocytes convert into immunostimulatory monocytes upon cancer cell-intrinsic cGAS-STING activation. Consistently, we find that human cancer cells can produce type I IFNs that polarize monocytes, and our immunostimulatory monocyte gene signature is enriched in patient tumors that respond to anti-PD-1 immunotherapy. Our work exposes a role for cancer cell-derived IFNs in licensing monocyte functions that influence immunotherapy outcomes., Competing Interests: Declaration of interests All authors were employees of Regeneron Pharmaceuticals at the time of their contributions. E.O. is now an employee of Bristol Myers Squibb, X.Y. is now an employee of Novartis, and E.M. is now an employee of Calico., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
21. CD22-targeted CD28 bispecific antibody enhances antitumor efficacy of odronextamab in refractory diffuse large B cell lymphoma models.
- Author
-
Wei J, Montalvo-Ortiz W, Yu L, Krasco A, Olson K, Rizvi S, Fiaschi N, Coetzee S, Wang F, Ullman E, Ahmed HS, Herlihy E, Lee K, Havel L, Potocky T, Ebstein S, Frleta D, Khatri A, Godin S, Hamon S, Brouwer-Visser J, Gorenc T, MacDonald D, Hermann A, Chaudhry A, Sirulnik A, Olson W, Lin J, Thurston G, Lowy I, Murphy AJ, Smith E, Jankovic V, Sleeman MA, and Skokos D
- Subjects
- Animals, Humans, CD28 Antigens, CD8-Positive T-Lymphocytes, Antigens, CD19, Neoplasm Recurrence, Local drug therapy, Sialic Acid Binding Ig-like Lectin 2 therapeutic use, Antibodies, Bispecific pharmacology, Antibodies, Bispecific therapeutic use, Lymphoma, Non-Hodgkin drug therapy, Lymphoma, Large B-Cell, Diffuse drug therapy, Antineoplastic Agents pharmacology
- Abstract
Although many patients with diffuse large B cell lymphoma (DLBCL) may achieve a complete response to frontline chemoimmunotherapy, patients with relapsed/refractory disease typically have poor outcomes. Odronextamab, a CD20xCD3 bispecific antibody that provides "signal 1" through the activation of the T cell receptor/CD3 complex, has exhibited early, promising activity for patients with highly refractory DLBCL in phase 1 trials. However, not all patients achieve complete responses, and many relapse, thus representing a high unmet medical need. Here, we investigated whether adding a costimulatory "signal 2" by engaging CD28 receptors on T cells could augment odronextamab activity. We demonstrate that REGN5837, a bispecific antibody that cross-links CD22-expressing tumor cells with CD28-expressing T cells, enhances odronextamab by potentiating T cell activation and cytolytic function. In preclinical DLBCL studies using human immune system-reconstituted animals, REGN5837 promotes the antitumor activity of odronextamab and induces intratumoral expansion of reprogrammable T cells while skewing away from a dysfunctional state. Although REGN5837 monotherapy shows limited activity and no toxicity in primate studies, it augments T cell activation when dosed in combination with odronextamab. In addition, analysis of non-Hodgkin lymphoma clinical samples reveals an increase in CD28
+ CD8+ T cells after odronextamab treatment, demonstrating the presence of a population that could potentially be targeted by REGN5837. Collectively, our data demonstrate that REGN5837 can markedly enhance the antitumor activity of odronextamab in preclinical NHL models, and the combination of these two bispecific antibodies may provide a chemotherapy-free approach for the treatment of DLBCL.- Published
- 2022
- Full Text
- View/download PDF
22. Biologics as novel therapeutics for the treatment of allergy: Challenges and opportunities.
- Author
-
Atanasio A, Orengo JM, Sleeman MA, and Stahl N
- Abstract
Over the last 4 decades there has been a significant global increase in the incidence and prevalence of IgE-mediated allergy. Although much progress has been made in the management of allergy via patient education, pharmacotherapy and immunomodulatory treatment regimens, significant unmet need remains. Advancements in our knowledge base surrounding the type 2 immune response, production of IgE and maintenance of immunological memory has led the field to explore targeted intervention of allergic pathways using monoclonal antibodies (mAbs). Intervention at various stages of the allergic cascade offers the opportunity to prevent initiation and/or maintenance of the type 2 immune response and effectively provide therapeutic benefit to patients. Furthermore, a better understanding of the protective mechanisms involved in allergen specific immunotherapy (AIT) has led us to appreciate the interplay of immunoglobulins in the allergic response, specifically the benefit in shifting the IgG:IgE ratio in favor of functionally relevant blocking IgG. Thus, treatments that lower IgE or boost IgG with the ability to outcompete IgE binding to allergen also present a favorable approach in the treatment of allergy. In this short review we discuss and highlight recent advances in the use of biologics to treat severe allergy, highlighting the key challenges but also the significant opportunities and advances to date., Competing Interests: All authors are employees of Regeneron Pharmaceuticals and hold company stock or stock options., (© 2022 Atanasio, Orengo, Sleeman and Stahl.)
- Published
- 2022
- Full Text
- View/download PDF
23. Diffuse alveolar damage patterns reflect the immunological and molecular heterogeneity in fatal COVID-19.
- Author
-
Erjefält JS, de Souza Xavier Costa N, Jönsson J, Cozzolino O, Dantas KC, Clausson CM, Siddhuraj P, Lindö C, Alyamani M, Lombardi SCFS, Mendroni Júnior A, Antonangelo L, Faria CS, Duarte-Neto AN, de Almeida Monteiro RA, Rebello Pinho JR, Gomes-Gouvêa MS, Verciano Pereira R, Monteiro JS, Setubal JC, de Oliveira EP, Theodoro Filho J, Sanden C, Orengo JM, Sleeman MA, da Silva LFF, Saldiva PHN, Dolhnikoff M, and Mauad T
- Subjects
- Cytokines, Humans, Lung pathology, SARS-CoV-2, COVID-19
- Abstract
Background: Severe COVID-19 lung disease exhibits a high degree of spatial and temporal heterogeneity, with different histological features coexisting within a single individual. It is important to capture the disease complexity to support patient management and treatment strategies. We provide spatially decoded analyses on the immunopathology of diffuse alveolar damage (DAD) patterns and factors that modulate immune and structural changes in fatal COVID-19., Methods: We spatially quantified the immune and structural cells in exudative, intermediate, and advanced DAD through multiplex immunohistochemistry in autopsy lung tissue of 18 COVID-19 patients. Cytokine profiling, viral, bacteria, and fungi detection, and transcriptome analyses were performed., Findings: Spatial DAD progression was associated with expansion of immune cells, macrophages, CD8+ T cells, fibroblasts, and (lymph)angiogenesis. Viral load correlated positively with exudative DAD and negatively with disease/hospital length. In all cases, enteric bacteria were isolated, and Candida parapsilosis in eight cases. Cytokines correlated mainly with macrophages and CD8+T cells. Pro-coagulation and acute repair were enriched pathways in exudative DAD whereas intermediate/advanced DAD had a molecular profile of elevated humoral and innate immune responses and extracellular matrix production., Interpretation: Unraveling the spatial and molecular immunopathology of COVID-19 cases exposes the responses to SARS-CoV-2-induced exudative DAD and subsequent immune-modulatory and remodeling changes in proliferative/advanced DAD that occur side-by-side together with secondary infections in the lungs. These complex features have important implications for disease management and the development of novel treatments., Funding: CNPq, Bill and Melinda Gates Foundation, HC-Convida, FAPESP, Regeneron Pharmaceuticals, and the Swedish Heart & Lung Foundation., Competing Interests: Declaration of interests J.E. is the founder of Medetect AB, Lund, Sweden. J.J., C.S. and C.L. are employees at Medetect AB. M.A.S. and J.O. are employees and shareholders at Regeneron Pharmaceuticals. All other co-authors have no conflict of interest to declare with the subject of the manuscript., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. A single-cell map of dynamic chromatin landscapes of immune cells in renal cell carcinoma.
- Author
-
Kourtis N, Wang Q, Wang B, Oswald E, Adler C, Cherravuru S, Malahias E, Zhang L, Golubov J, Wei Q, Lemus S, Ni M, Ding Y, Wei Y, Atwal GS, Thurston G, Macdonald LE, Murphy AJ, Dhanik A, Sleeman MA, Tykodi SS, and Skokos D
- Subjects
- CD8-Positive T-Lymphocytes, Chromatin genetics, Humans, NF-kappa B, Carcinoma, Renal Cell genetics, Kidney Neoplasms genetics
- Abstract
A complete chart of the chromatin regulatory elements of immune cells in patients with cancer and their dynamic behavior is necessary to understand the developmental fates and guide therapeutic strategies. Here, we map the single-cell chromatin landscape of immune cells from blood, normal tumor-adjacent kidney tissue and malignant tissue from patients with early-stage clear cell renal cell carcinoma (ccRCC). We catalog the T cell states dictated by tissue-specific and developmental-stage-specific chromatin accessibility patterns, infer key chromatin regulators and observe rewiring of regulatory networks in the progression to dysfunction in CD8
+ T cells. Unexpectedly, among the transcription factors orchestrating the path to dysfunction, NF-κB is associated with a pro-apoptotic program in late stages of dysfunction in tumor-infiltrating CD8+ T cells. Importantly, this epigenomic profiling stratified ccRCC patients based on a NF-κB-driven pro-apoptotic signature. This study provides a rich resource for understanding the functional states and regulatory dynamics of immune cells in ccRCC., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
25. Targeting immunodominant Bet v 1 epitopes with monoclonal antibodies prevents the birch allergic response.
- Author
-
Atanasio A, Franklin MC, Kamat V, Hernandez AR, Badithe A, Ben LH, Jones J, Bautista J, Yancopoulos GD, Olson W, Murphy AJ, Sleeman MA, and Orengo JM
- Subjects
- Animals, Basophils drug effects, Basophils immunology, Humans, Immunoglobulin E immunology, Mast Cells drug effects, Mast Cells immunology, Mice, Inbred BALB C, Rhinitis, Allergic, Seasonal blood, Rhinitis, Allergic, Seasonal immunology, Mice, Allergens immunology, Antibodies, Monoclonal pharmacology, Antigens, Plant immunology, Immunodominant Epitopes immunology, Immunoglobulin G pharmacology, Passive Cutaneous Anaphylaxis immunology
- Abstract
Background: Blocking the major cat allergen, Fel d 1, with mAbs was effective in preventing an acute cat allergic response., Objectives: This study sought to extend the allergen-specific antibody approach and demonstrate that a combination of mAbs targeting Bet v 1, the immunodominant and most abundant allergenic protein in birch pollen, can prevent the birch allergic response., Methods: Bet v 1-specific mAbs, REGN5713, REGN5714, and REGN5715, were isolated using the VelocImmune platform. Surface plasmon resonance, x-ray crystallography, and cryo-electron microscopy determined binding kinetics and structural data. Inhibition of IgE-binding, basophil activation, and mast cell degranulation were assessed via blocking ELISA, flow cytometry, and the passive cutaneous anaphylaxis mouse model., Results: REGN5713, REGN5714, and REGN5715 bind with high affinity and noncompetitively to Bet v 1. A cocktail of all 3 antibodies, REGN5713/14/15, blocks IgE binding to Bet v 1 and inhibits Bet v 1- and birch pollen extract-induced basophil activation ex vivo and mast cell degranulation in vivo. Crystal structures of the complex of Bet v 1 with immunoglobulin antigen-binding fragments of REGN5713 or REGN5715 show distinct interaction sites on Bet v 1. Cryo-electron microscopy reveals a planar and roughly symmetrical complex formed by REGN5713/14/15 bound to Bet v 1., Conclusions: These data confirm the immunodominance of Bet v 1 in birch allergy and demonstrate blockade of the birch allergic response with REGN5713/14/15. Structural analyses show simultaneous binding of REGN5713, REGN5714, and REGN5715 with substantial areas of Bet v 1 exposed, suggesting that targeting specific epitopes is sufficient to block the allergic response., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
26. Humanization of T cell-mediated immunity in mice.
- Author
-
Moore MJ, Zhong M, Hansen J, Gartner H, Grant C, Huang M, Harris FM, Tu N, Bowerman NA, Edelmann KH, Barry T, Herbin O, Tay CS, DiLillo DJ, Decker CE, Levenkova N, Shevchuk J, Dhanik A, Meagher KA, Karr A, Roos J, Lee WY, Suh D, Eckersdorff M, Meagher TC, Koss M, Esau L, Sleeman MA, Babb R, Chen G, Kyratsous CA, Poueymirou WT, McWhirter JR, Voronina VA, Guo C, Gurer C, Yancopoulos GD, Murphy AJ, and Macdonald LE
- Subjects
- Animals, Humans, Mice, Mice, Inbred C57BL, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell, alpha-beta immunology, T-Lymphocytes immunology
- Abstract
Despite the enormous promise of T cell therapies, the isolation and study of human T cell receptors (TCRs) of dedicated specificity remains a major challenge. To overcome this limitation, we generated mice with a genetically humanized system of T cell immunity. We used VelociGene technology to replace the murine TCRαβ variable regions, along with regions encoding the extracellular domains of co-receptors CD4 and CD8, and major histocompatibility complex (MHC) class I and II, with corresponding human sequences. The resulting “VelociT” mice have normal myeloid and lymphoid immune cell populations, including thymic and peripheral αβ T cell subsets comparable with wild-type mice. VelociT mice expressed a diverse TCR repertoire, mounted functional T cell responses to lymphocytic choriomeningitis virus infection, and could develop experimental autoimmune encephalomyelitis. Immunization of VelociT mice with human tumor-associated peptide antigens generated robust, antigen-specific responses and led to identification of a TCR against tumor antigen New York esophageal squamous cell carcinoma-1 with potent antitumor activity. These studies demonstrate that VelociT mice mount clinically relevant T cell responses to both MHC-I– and MHC-II–restricted antigens, providing a powerful new model for analyzing T cell function in human disease. Moreover, VelociT mice are a new platform for de novo discovery of therapeutic human TCRs.
- Published
- 2021
- Full Text
- View/download PDF
27. Chronic pharmacological antagonism of the GM-CSF receptor in mice does not replicate the pulmonary alveolar proteinosis phenotype but does alter lung surfactant turnover.
- Author
-
Corkill DJ, Hunt AN, Hinrichs MJ, White N, Rebelatto M, Roskos L, Nys J, Scott A, Robinson MJ, Ryan P, Postle AD, and Sleeman MA
- Subjects
- Animals, Antibodies, Monoclonal, Humanized pharmacology, Arthritis, Rheumatoid therapy, Autoantibodies chemistry, Bronchoalveolar Lavage Fluid, COVID-19 immunology, Choline analogs & derivatives, Female, Granulocyte-Macrophage Colony-Stimulating Factor chemistry, Inflammation, Interleukin-6 metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Pulmonary Alveolar Proteinosis genetics, SARS-CoV-2 immunology, Surface-Active Agents, Arthritis, Rheumatoid metabolism, COVID-19 therapy, Pulmonary Alveolar Proteinosis immunology, Pulmonary Surfactants metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor antagonists & inhibitors, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism
- Abstract
Granulocyte macrophage colony stimulating factor (GM-CSF) is a key participant in, and a clinical target for, the treatment of inflammatory diseases including rheumatoid arthritis (RA). Therapeutic inhibition of GM-CSF signalling using monoclonal antibodies to the α-subunit of the GM-CSF receptor (GMCSFRα) has shown clear benefit in patients with RA, giant cell arteritis (GCAs) and some efficacy in severe SARS-CoV-2 infection. However, GM-CSF autoantibodies are associated with the development of pulmonary alveolar proteinosis (PAP), a rare lung disease characterised by alveolar macrophage (AM) dysfunction and the accumulation of surfactant lipids. We assessed how the anti-GMCSFRα approach might impact surfactant turnover in the airway. Female C57BL/6J mice received a mouse-GMCSFRα blocking antibody (CAM-3003) twice per week for up to 24 weeks. A parallel, comparator cohort of the mouse PAP model, GM-CSF receptor β subunit (GMCSFRβ) knock-out (KO), was maintained up to 16 weeks. We assessed lung tissue histopathology alongside lung phosphatidylcholine (PC) metabolism using stable isotope lipidomics. GMCSFRβ KO mice reproduced the histopathological and biochemical features of PAP, accumulating surfactant PC in both broncho-alveolar lavage fluid (BALF) and lavaged lung tissue. The incorporation pattern of methyl-D9-choline showed impaired catabolism and not enhanced synthesis. In contrast, chronic supra-pharmacological CAM-3003 exposure (100 mg/kg) over 24 weeks did not elicit a histopathological PAP phenotype despite some changes in lung PC catabolism. Lack of significant impairment of AM catabolic function supports clinical observations that therapeutic antibodies to this pathway have not been associated with PAP in clinical trials., (© 2021 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2021
- Full Text
- View/download PDF
28. Butyrophilin-like 2 regulates site-specific adaptations of intestinal γδ intraepithelial lymphocytes.
- Author
-
Panea C, Zhang R, VanValkenburgh J, Ni M, Adler C, Wei Y, Ochoa F, Schmahl J, Tang Y, Siao CJ, Poueymirou W, Espert J, Lim WK, Atwal GS, Murphy AJ, Sleeman MA, Hovhannisyan Z, and Haxhinasto S
- Subjects
- Animals, Butyrophilins metabolism, Female, Mice, Mice, Inbred C57BL, Butyrophilins genetics, Ileum immunology, Immunity, Innate genetics, Immunity, Mucosal genetics, Intraepithelial Lymphocytes metabolism
- Abstract
Tissue-resident γδ intraepithelial lymphocytes (IELs) orchestrate innate and adaptive immune responses to maintain intestinal epithelial barrier integrity. Epithelia-specific butyrophilin-like (Btnl) molecules induce perinatal development of distinct Vγ TCR
+ IELs, however, the mechanisms that control γδ IEL maintenance within discrete intestinal segments are unclear. Here, we show that Btnl2 suppressed homeostatic proliferation of γδ IELs preferentially in the ileum. High throughput transcriptomic characterization of site-specific Btnl2-KO γδ IELs reveals that Btnl2 regulated the antimicrobial response module of ileal γδ IELs. Btnl2 deficiency shapes the TCR specificities and TCRγ/δ repertoire diversity of ileal γδ IELs. During DSS-induced colitis, Btnl2-KO mice exhibit increased inflammation and delayed mucosal repair in the colon. Collectively, these data suggest that Btnl2 fine-tunes γδ IEL frequencies and TCR specificities in response to site-specific homeostatic and inflammatory cues. Hence, Btnl-mediated targeting of γδ IEL development and maintenance may help dissect their immunological functions in intestinal diseases with segment-specific manifestations., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
29. Sequence of αPD-1 relative to local tumor irradiation determines the induction of abscopal antitumor immune responses.
- Author
-
Wei J, Montalvo-Ortiz W, Yu L, Krasco A, Ebstein S, Cortez C, Lowy I, Murphy AJ, Sleeman MA, and Skokos D
- Subjects
- Animals, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor transplantation, Disease Models, Animal, Drug Administration Schedule, Drug Screening Assays, Antitumor, Female, Humans, Mice, Neoplasms immunology, Primary Cell Culture, Radiosurgery, Time Factors, Tumor Microenvironment drug effects, Tumor Microenvironment immunology, Tumor Microenvironment radiation effects, Chemoradiotherapy methods, Immune Checkpoint Inhibitors administration & dosage, Neoplasms therapy, Programmed Cell Death 1 Receptor antagonists & inhibitors
- Abstract
Although radiotherapy has been used for over a century to locally control tumor growth, alone it rarely induces an abscopal response or systemic antitumor immunity capable of inhibiting distal tumors outside of the irradiation field. Results from recent studies suggest that combining immune checkpoint blockades to radiotherapy may enhance abscopal activity. However, the treatment conditions and underlying immune mechanisms that consistently drive an abscopal response during radiation therapy combinations remain unknown. Here, we analyzed the antitumor responses at primary and distal tumor sites, demonstrating that the timing of αPD-1 antibody administration relative to radiotherapy determined the potency of the induced abscopal response. Blockade of the PD-1 pathway after local tumor irradiation resulted in the expansion of polyfunctional intratumoral CD8
+ T cells, a decrease in intratumoral dysfunctional CD8+ T cells, expansion of reprogrammable CD8+ T cells, and induction of potent abscopal responses. However, administration of αPD-1 before irradiation almost completely abrogated systemic immunity, which associated with increased radiosensitivity and death of CD8+ T cells. The subsequent reduction of polyfunctional effector CD8+ T cells at the irradiated tumor site generated a suboptimal systemic antitumor response and the loss of abscopal responses. Therefore, this report maximizes the potential synergy between radiotherapy and αPD-1 immunotherapy, information that will benefit clinical combinations of radiotherapy and immune checkpoint blockade., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2021
- Full Text
- View/download PDF
30. Immunohistochemical Characterization of the IL-13:IL-4 Receptor α Axis in the Skin of Adult Patients with Moderate to Severe Atopic Dermatitis and Healthy Controls.
- Author
-
Miranda E, Roberts J, Novick S, Lapointe JM, Bruijnzeel-Koomen C, Thijs J, Sleeman MA, May RD, Hijnen D, and Strickland I
- Subjects
- Adult, Aged, Biomarkers analysis, Biomarkers metabolism, Biopsy, Case-Control Studies, Dermatitis, Atopic immunology, Dermatitis, Atopic pathology, Female, Healthy Volunteers, Humans, Immunohistochemistry, Interleukin-13 metabolism, Interleukin-4 Receptor alpha Subunit metabolism, Langerhans Cells immunology, Langerhans Cells metabolism, Male, Melanocytes immunology, Melanocytes metabolism, Middle Aged, Severity of Illness Index, Skin cytology, Skin immunology, Young Adult, Dermatitis, Atopic diagnosis, Interleukin-13 analysis, Interleukin-4 Receptor alpha Subunit analysis, Skin pathology
- Published
- 2021
- Full Text
- View/download PDF
31. Enhanced IL-36R signaling promotes barrier impairment and inflammation in skin and intestine.
- Author
-
Hovhannisyan Z, Liu N, Khalil-Aguero S, Panea C, VanValkenburgh J, Zhang R, Lim WK, Bai Y, Fury W, Huang T, Garnova E, Fairhurst J, Kim J, Aryal S, Ajithdoss D, Oyejide A, Del Pilar Molina-Portela M, E H, Poueymirou W, Oristian NS, Brydges S, Liu X, Olson W, Yancopoulos G, Murphy AJ, Sleeman MA, and Haxhinasto S
- Subjects
- Animals, Biomarkers, Dermatitis pathology, Disease Models, Animal, Disease Susceptibility, Gastroenteritis pathology, Humans, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Mice, Skin metabolism, Skin pathology, Dermatitis etiology, Dermatitis metabolism, Gastroenteritis etiology, Gastroenteritis metabolism, Receptors, Interleukin-1 metabolism, Signal Transduction
- Abstract
Deficiency in interleukin-36R (IL-36R) antagonist caused by loss-of-function mutations in IL-36RN leads to DITRA (deficiency of IL-36 receptor antagonist), a rare inflammatory human disease that belongs to a subgroup of generalized pustular psoriasis (GPP). We report a functional genetic mouse model of DITRA with enhanced IL-36R signaling analogous to that observed in patients with DITRA, which provides new insight into our understanding of the IL-36 family of molecules in regulating barrier integrity across multiple tissues. Humanized DITRA-like mice displayed increased skin inflammation in a preclinical model of psoriasis, and in vivo blockade of IL-36R pathway using anti-human IL-36R antibody ameliorated imiquimod-induced skin pathology as both prophylactic and therapeutic treatments. Deeper characterization of the humanized DITRA-like mice revealed that deregulated IL-36R signaling promoted tissue pathology during intestinal injury and led to impairment in mucosal restoration in the repair phase of chronic dextran sulfate sodium (DSS)-induced colitis. Blockade of IL-36R pathway significantly ameliorated DSS-induced intestinal inflammation and rescued the inability of DITRA-like mice to recover from mucosal damage in vivo. Our results indicate a central role for IL-36 in regulating proinflammatory responses in the skin and epithelial barrier function in the intestine, suggesting a new therapeutic potential for targeting the IL-36R axis in psoriasis and at the later stages of intestinal pathology in inflammatory bowel disease., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
32. Absence of central tolerance in Aire-deficient mice synergizes with immune-checkpoint inhibition to enhance antitumor responses.
- Author
-
Benitez AA, Khalil-Agüero S, Nandakumar A, Gupta NT, Zhang W, Atwal GS, Murphy AJ, Sleeman MA, and Haxhinasto S
- Subjects
- Animals, CD8-Positive T-Lymphocytes immunology, Melanoma, Experimental immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Neoplasm Transplantation, T-Lymphocytes immunology, Tumor Escape immunology, AIRE Protein, Immune Checkpoint Inhibitors immunology, Immune Tolerance immunology, Neoplasms, Experimental immunology, Polyendocrinopathies, Autoimmune immunology, Transcription Factors deficiency
- Abstract
The endogenous anti-tumor responses are limited in part by the absence of tumor-reactive T cells, an inevitable consequence of thymic central tolerance mechanisms ensuring prevention of autoimmunity. Here we show that tumor rejection induced by immune checkpoint blockade is significantly enhanced in Aire-deficient mice, the epitome of central tolerance breakdown. The observed synergy in tumor rejection extended to different tumor models, was accompanied by increased numbers of activated T cells expressing high levels of Gzma, Gzmb, Perforin, Cxcr3, and increased intratumoural levels of Cxcl9 and Cxcl10 compared to wild-type mice. Consistent with Aire's central role in T cell repertoire selection, single cell TCR sequencing unveiled expansion of several clones with high tumor reactivity. The data suggest that breakdown in central tolerance synergizes with immune checkpoint blockade in enhancing anti-tumor immunity and may serve as a model to unmask novel anti-tumor therapies including anti-tumor TCRs, normally purged during central tolerance.
- Published
- 2020
- Full Text
- View/download PDF
33. Tumor-targeted CD28 bispecific antibodies enhance the antitumor efficacy of PD-1 immunotherapy.
- Author
-
Waite JC, Wang B, Haber L, Hermann A, Ullman E, Ye X, Dudgeon D, Slim R, Ajithdoss DK, Godin SJ, Ramos I, Wu Q, Oswald E, Poon P, Golubov J, Grote D, Stella J, Pawashe A, Finney J, Herlihy E, Ahmed H, Kamat V, Dorvilliers A, Navarro E, Xiao J, Kim J, Yang SN, Warsaw J, Lett C, Canova L, Schulenburg T, Foster R, Krueger P, Garnova E, Rafique A, Babb R, Chen G, Stokes Oristian N, Siao CJ, Daly C, Gurer C, Martin J, Macdonald L, MacDonald D, Poueymirou W, Smith E, Lowy I, Thurston G, Olson W, Lin JC, Sleeman MA, Yancopoulos GD, Murphy AJ, and Skokos D
- Subjects
- Animals, CD28 Antigens, Humans, Immunotherapy, Mice, Programmed Cell Death 1 Receptor, Antibodies, Bispecific therapeutic use, Neoplasms drug therapy
- Abstract
Monoclonal antibodies that block the programmed cell death 1 (PD-1) checkpoint have revolutionized cancer immunotherapy. However, many major tumor types remain unresponsive to anti-PD-1 therapy, and even among responsive tumor types, most of the patients do not develop durable antitumor immunity. It has been shown that bispecific antibodies activate T cells by cross-linking the TCR/CD3 complex with a tumor-specific antigen (TSA). The class of TSAxCD3 bispecific antibodies have generated exciting results in early clinical trials. We have recently described another class of "costimulatory bispecifics" that cross-link a TSA to CD28 (TSAxCD28) and cooperate with TSAxCD3 bispecifics. Here, we demonstrate that these TSAxCD28 bispecifics (one specific for prostate cancer and the other for epithelial tumors) can also synergize with the broader anti-PD-1 approach and endow responsiveness-as well as long-term immune memory-against tumors that otherwise do not respond to anti-PD-1 alone. Unlike CD28 superagonists, which broadly activate T cells and induce cytokine storm, TSAxCD28 bispecifics display little or no toxicity when used alone or in combination with a PD-1 blocker in genetically humanized immunocompetent mouse models or in primates and thus may provide a well-tolerated and "off the shelf" combination approach with PD-1 immunotherapy that can markedly enhance antitumor efficacy., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
34. Endogenous retroviral proteins provide an immunodominant but not requisite antigen in a murine immunotherapy tumor model.
- Author
-
Ye X, Waite JC, Dhanik A, Gupta N, Zhong M, Adler C, Malahias E, Ni M, Wei Y, Gurer C, Zhang W, Macdonald LE, Murphy AJ, Sleeman MA, and Skokos D
- Subjects
- Animals, Immunotherapy, Lymphocyte Activation, Mice, Receptors, Antigen, T-Cell genetics, Endogenous Retroviruses, Neoplasms therapy
- Abstract
Clinical observations suggest that responses to cancer immunotherapy are correlated with intra-tumoral T cell receptor (TCR) clonality, tumor mutation burden (TMB) and host HLA genotype, highlighting the importance of host T cell recognition of tumor antigens. However, the dynamic interplay between T cell activation state and changes in TCR repertoire in driving the identification of potential immunodominant antigen(s) remains largely unexplored. Here, we performed single-cell RNA-sequencing on CD8
+ tumor-infiltrating T cells (TILs) using the murine colorectal tumor model MC38 to identify unique TCR sequences and validate their tumor reactivity. We found that the majority of clonally expanded TILs are tumor-reactive and their TCR repertoire is unique amongst individual MC38 tumor-bearing mice. Our query identified that multiple expanded TCR clones recognized the retroviral epitope p15E as an immunodominant antigen. In addition, we found that the endogenous retroviral genome encoding for p15E is highly expressed in MC38 tumors, but not in normal tissues, due to epigenetic derepression. Further, we demonstrated that the p15E-specific TILs exhibit an activated phenotype and an increase in frequency upon treatment with anti-41BB and anti-PD-1 combination immunotherapy. Importantly, we showed that although p15E-specific TILs are not required to mount a primary anti-tumor response, they contributed to the development of strong immune memory. Overall our results revealed that endogenous retroviral antigens expressed by tumor cells may represent an important and underappreciated category of tumor antigens that could be readily targeted in the clinic., (© 2020 Regeneron Pharmaceuticals. Published with license by Taylor & Francis Group, LLC.)- Published
- 2020
- Full Text
- View/download PDF
35. Dual blockade of IL-4 and IL-13 with dupilumab, an IL-4Rα antibody, is required to broadly inhibit type 2 inflammation.
- Author
-
Le Floc'h A, Allinne J, Nagashima K, Scott G, Birchard D, Asrat S, Bai Y, Lim WK, Martin J, Huang T, Potocky TB, Kim JH, Rafique A, Papadopoulos NJ, Stahl N, Yancopoulos GD, Murphy AJ, Sleeman MA, and Orengo JM
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Inflammation, Interleukin-4, Mice, Interleukin-13
- Abstract
Background: Dupilumab, a fully human monoclonal antibody that binds IL-4Rα and inhibits signaling of both IL-4 and IL-13, has shown efficacy across multiple diseases with underlying type 2 signatures and is approved for treatment of asthma, atopic dermatitis, and chronic sinusitis with nasal polyposis. We sought to provide a comprehensive analysis of the redundant and distinct roles of IL-4 and IL-13 in type 2 inflammation and report dupilumab mechanisms of action., Methods: Using primary cell assays and a mouse model of house dust mite-induced asthma, we compared IL-4 vs IL-13 vs IL-4Rα blockers., Results: Intranasal administration of either IL-4 or IL-13 confers an asthma-like phenotype in mice by inducing immune cell lung infiltration, including eosinophils, increasing cytokine/chemokine expression and mucus production, thus demonstrating redundant functions of these cytokines. We further teased out their respective contributions using human in vitro culture systems. Then, in a mouse asthma model by comparing in head-to-head studies, either IL-4 or IL-13 inhibition to dual IL-4/IL-13 inhibition, we demonstrate that blockade of both IL-4 and IL-13 is required to broadly block type 2 inflammation, which translates to protection from allergen-induced lung function impairment. Notably, only dual IL-4/IL-13 blockade prevented eosinophil infiltration into lung tissue without affecting circulating eosinophils, demonstrating that tissue, but not circulating eosinophils, contributes to disease pathology., Conclusions: Overall, these data support IL-4 and IL-13 as key drivers of type 2 inflammation and help provide insight into the therapeutic mechanism of dupilumab, a dual IL-4/IL-13 blocker, in multiple type 2 diseases., (© 2019 The Authors. Allergy published by John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
36. FPR-1 is an important regulator of neutrophil recruitment and a tissue-specific driver of pulmonary fibrosis.
- Author
-
Leslie J, Millar BJ, Del Carpio Pons A, Burgoyne RA, Frost JD, Barksby BS, Luli S, Scott J, Simpson AJ, Gauldie J, Murray LA, Finch DK, Carruthers AM, Ferguson J, Sleeman MA, Rider D, Howarth R, Fox C, Oakley F, Fisher AJ, Mann DA, and Borthwick LA
- Subjects
- Animals, Bleomycin toxicity, Humans, Ligands, Mice, Inbred C57BL, Mice, Knockout, Pulmonary Fibrosis chemically induced, Pulmonary Fibrosis pathology, Receptors, Formyl Peptide genetics, Receptors, Formyl Peptide metabolism, Neutrophil Infiltration physiology, Pulmonary Fibrosis physiopathology, Receptors, Formyl Peptide physiology
- Abstract
Neutrophils are the most abundant inflammatory cells at the earliest stages of wound healing and play important roles in wound repair and fibrosis. Formyl peptide receptor 1 (FPR-1) is abundantly expressed on neutrophils and has been shown to regulate their function, yet the importance of FPR-1 in fibrosis remains ill defined. FPR-1-deficient (fpr1-/-) mice were protected from bleomycin-induced pulmonary fibrosis but developed renal and hepatic fibrosis normally. Mechanistically, we observed a failure to effectively recruit neutrophils to the lungs of fpr1-/- mice, whereas neutrophil recruitment was unaffected in the liver and kidney. Using an adoptive transfer model we demonstrated that the defect in neutrophil recruitment to the lung was intrinsic to the fpr1-/- neutrophils, as C57BL/6 neutrophils were recruited normally to the damaged lung in fpr1-/- mice. Finally, C57BL/6 mice in which neutrophils had been depleted were protected from pulmonary fibrosis. In conclusion, FPR-1 and FPR-1 ligands are required for effective neutrophil recruitment to the damaged lung. Failure to recruit neutrophils or depletion of neutrophils protects from pulmonary fibrosis.
- Published
- 2020
- Full Text
- View/download PDF
37. Chronic allergen exposure drives accumulation of long-lived IgE plasma cells in the bone marrow, giving rise to serological memory.
- Author
-
Asrat S, Kaur N, Liu X, Ben LH, Kajimura D, Murphy AJ, Sleeman MA, Limnander A, and Orengo JM
- Subjects
- Anaphylaxis immunology, Animals, Bone Marrow Cells immunology, Environmental Exposure, Humans, Mast Cells immunology, Mice, Allergens immunology, Immunoglobulin E blood, Immunologic Memory, Plasma Cells immunology, Pyroglyphidae immunology
- Abstract
Immunoglobulin E (IgE) plays an important role in allergic diseases. Nevertheless, the source of IgE serological memory remains controversial. We reexamined the mechanism of serological memory in allergy using a dual reporter system to track IgE
+ plasma cells in mice. Short-term allergen exposure resulted in the generation of IgE+ plasma cells that resided mainly in secondary lymphoid organs and produced IgE that was unable to degranulate mast cells. In contrast, chronic allergen exposure led to the generation of long-lived IgE+ plasma cells that were primarily derived from sequential class switching of IgG1, accumulated in the bone marrow, and produced IgE capable of inducing anaphylaxis. IgE+ plasma cells were found in the bone marrow of human allergic, but not nonallergic donors, and allergen-specific IgE produced by these cells was able to induce mast cell degranulation when transferred to mice. These data demonstrate that long-lived IgE+ bone marrow plasma cells arise during chronic allergen exposure and establish serological memory in both mice and humans., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2020
- Full Text
- View/download PDF
38. A class of costimulatory CD28-bispecific antibodies that enhance the antitumor activity of CD3-bispecific antibodies.
- Author
-
Skokos D, Waite JC, Haber L, Crawford A, Hermann A, Ullman E, Slim R, Godin S, Ajithdoss D, Ye X, Wang B, Wu Q, Ramos I, Pawashe A, Canova L, Vazzana K, Ram P, Herlihy E, Ahmed H, Oswald E, Golubov J, Poon P, Havel L, Chiu D, Lazo M, Provoncha K, Yu K, Kim J, Warsaw JJ, Stokes Oristian N, Siao CJ, Dudgeon D, Huang T, Potocky T, Martin J, MacDonald D, Oyejide A, Rafique A, Poueymirou W, Kirshner JR, Smith E, Olson W, Lin J, Thurston G, Sleeman MA, Murphy AJ, and Yancopoulos GD
- Subjects
- Animals, Antigens, Neoplasm immunology, Cell Line, Tumor, Cell Proliferation, Cytokines metabolism, Cytotoxicity, Immunologic, Female, HEK293 Cells, Humans, Immunological Synapses metabolism, Lymphocyte Activation immunology, Macaca fascicularis, Mice, Neoplasms pathology, Receptors, Antigen, T-Cell metabolism, T-Lymphocytes immunology, Xenograft Model Antitumor Assays, Antibodies, Bispecific immunology, CD28 Antigens immunology, CD3 Complex immunology, Neoplasms immunology
- Abstract
T cell activation is initiated upon binding of the T cell receptor (TCR)/CD3 complex to peptide-major histocompatibility complexes ("signal 1"); activation is enhanced by engagement of a second "costimulatory" receptor, such as the CD28 receptor on T cells binding to its cognate ligand(s) on the target cell ("signal 2"). CD3-based bispecific antibodies act by replacing conventional signal 1, linking T cells to tumor cells by binding a tumor-specific antigen (TSA) with one arm of the bispecific and bridging to TCR/CD3 with the other. Although some of these so-called TSAxCD3 bispecifics have demonstrated promising antitumor efficacy in patients with cancer, their activity remains to be optimized. Here, we introduce a class of bispecific antibodies that mimic signal 2 by bridging TSA to the costimulatory CD28 receptor on T cells. We term these TSAxCD28 bispecifics and describe two such bispecific antibodies: one specific for ovarian and the other for prostate cancer antigens. Unlike CD28 superagonists, which broadly activate T cells and resulted in profound toxicity in early clinical trials, these TSAxCD28 bispecifics show limited activity and no toxicity when used alone in genetically humanized immunocompetent mouse models or in primates. However, when combined with TSAxCD3 bispecifics, they enhance the artificial synapse between a T cell and its target cell, potentiate T cell activation, and markedly improve antitumor activity of CD3 bispecifics in a variety of xenogeneic and syngeneic tumor models. Combining this class of CD28-costimulatory bispecific antibodies with the emerging class of TSAxCD3 bispecifics may provide well-tolerated, off-the-shelf antibody therapies with robust antitumor efficacy., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
39. IL-33 blockade affects mediators of persistence and exacerbation in a model of chronic airway inflammation.
- Author
-
Allinne J, Scott G, Lim WK, Birchard D, Erjefält JS, Sandén C, Ben LH, Agrawal A, Kaur N, Kim JH, Kamat V, Fury W, Huang T, Stahl N, Yancopoulos GD, Murphy AJ, Sleeman MA, and Orengo JM
- Subjects
- Animals, Asthma chemically induced, Asthma pathology, Asthma therapy, Humans, Inflammation drug therapy, Inflammation immunology, Inflammation pathology, Interleukin-33 antagonists & inhibitors, Lung pathology, Mice, Mice, Transgenic, Th2 Cells immunology, Th2 Cells pathology, Airway Remodeling immunology, Asthma immunology, Interleukin-33 immunology, Lung immunology, Pyroglyphidae immunology, Signal Transduction immunology
- Abstract
Background: Severe inflammatory airway diseases are associated with inflammation that does not resolve, leading to structural changes and an overall environment primed for exacerbations., Objective: We sought to identify and inhibit pathways that perpetuate this heightened inflammatory state because this could lead to therapies that allow for a more quiescent lung that is less predisposed to symptoms and exacerbations., Methods: Using prolonged exposure to house dust mite in mice, we developed a mouse model of persistent and exacerbating airway disease characterized by a mixed inflammatory phenotype., Results: We show that lung IL-33 drives inflammation and remodeling beyond the type 2 response classically associated with IL-33 signaling. IL-33 blockade with an IL-33 neutralizing antibody normalized established inflammation and improved remodeling of both the lung epithelium and lung parenchyma. Specifically, IL-33 blockade normalized persisting and exacerbating inflammatory end points, including eosinophilic, neutrophilic, and ST2
+ CD4+ T-cell infiltration. Importantly, we identified a key role for IL-33 in driving lung remodeling because anti-IL-33 also re-established the presence of ciliated cells over mucus-producing cells and decreased myofibroblast numbers, even in the context of continuous allergen exposure, resulting in improved lung function., Conclusion: Overall, this study shows that increased IL-33 levels drive a self-perpetuating amplification loop that maintains the lung in a state of lasting inflammation and remodeled tissue primed for exacerbations. Thus IL-33 blockade might ameliorate symptoms and prevent exacerbations by quelling persistent inflammation and airway remodeling., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
40. A stromal cell niche sustains ILC2-mediated type-2 conditioning in adipose tissue.
- Author
-
Rana BMJ, Jou E, Barlow JL, Rodriguez-Rodriguez N, Walker JA, Knox C, Jolin HE, Hardman CS, Sivasubramaniam M, Szeto A, Cohen ES, Scott IC, Sleeman MA, Chidomere CI, Cruz Migoni S, Caamano J, Jorgensen HF, Carobbio S, Vidal-Puig A, and McKenzie ANJ
- Subjects
- Animals, Cell Proliferation, Eosinophils metabolism, Interleukin-33, Interleukin-5 biosynthesis, Mice, Inbred BALB C, Mice, Inbred C57BL, Stromal Cells cytology, Adipose Tissue, White cytology, Immunity, Innate, Lymphocytes cytology, Lymphocytes immunology
- Abstract
Group-2 innate lymphoid cells (ILC2), type-2 cytokines, and eosinophils have all been implicated in sustaining adipose tissue homeostasis. However, the interplay between the stroma and adipose-resident immune cells is less well understood. We identify that white adipose tissue-resident multipotent stromal cells (WAT-MSCs) can act as a reservoir for IL-33, especially after cell stress, but also provide additional signals for sustaining ILC2. Indeed, we demonstrate that WAT-MSCs also support ICAM-1-mediated proliferation and activation of LFA-1-expressing ILC2s. Consequently, ILC2-derived IL-4 and IL-13 feed back to induce eotaxin secretion from WAT-MSCs, supporting eosinophil recruitment. Thus, MSCs provide a niche for multifaceted dialogue with ILC2 to sustain a type-2 immune environment in WAT., (© 2019 Rana et al.)
- Published
- 2019
- Full Text
- View/download PDF
41. Neutrophil GM-CSF receptor dynamics in acute lung injury.
- Author
-
De Alessandris S, Ferguson GJ, Dodd AJ, Juss JK, Devaprasad A, Piper S, Wyatt O, Killick H, Corkill DJ, Cohen ES, Pandit A, Radstake TRDJ, Simmonds R, Condliffe AM, Sleeman MA, Cowburn AS, Finch DK, and Chilvers ER
- Subjects
- Acute Lung Injury chemically induced, Acute Lung Injury genetics, Acute Lung Injury pathology, Adult, Animals, Cell Line, Tumor, Cytokine Receptor Common beta Subunit genetics, Cytokine Receptor Common beta Subunit immunology, Disease Models, Animal, Female, Humans, Lipopolysaccharides toxicity, Male, Mice, Mice, Inbred BALB C, Mice, Transgenic, Neutrophils pathology, Pulmonary Alveoli pathology, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Time Factors, Acute Lung Injury immunology, Gene Expression Regulation immunology, Neutrophils immunology, Pulmonary Alveoli immunology, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor immunology
- Abstract
GM-CSF is important in regulating acute, persistent neutrophilic inflammation in certain settings, including lung injury. Ligand binding induces rapid internalization of the GM-CSF receptor (GM-CSFRα) complex, a process essential for signaling. Whereas GM-CSF controls many aspects of neutrophil biology, regulation of GM-CSFRα expression is poorly understood, particularly the role of GM-CSFRα in ligand clearance and whether signaling is sustained despite major down-regulation of GM-CSFRα surface expression. We established a quantitative assay of GM-CSFRα surface expression and used this, together with selective anti-GM-CSFR antibodies, to define GM-CSFRα kinetics in human neutrophils, and in murine blood and alveolar neutrophils in a lung injury model. Despite rapid sustained ligand-induced GM-CSFRα loss from the neutrophil surface, which persisted even following ligand removal, pro-survival effects of GM-CSF required ongoing ligand-receptor interaction. Neutrophils recruited to the lungs following LPS challenge showed initially high mGM-CSFRα expression, which along with mGM-CSFRβ declined over 24 hr; this was associated with a transient increase in bronchoalveolar lavage fluid (BALF) mGM-CSF concentration. Treating mice in an LPS challenge model with CAM-3003, an anti-mGM-CSFRα mAb, inhibited inflammatory cell influx into the lung and maintained the level of BALF mGM-CSF. Consistent with neutrophil consumption of GM-CSF, human neutrophils depleted exogenous GM-CSF, independent of protease activity. These data show that loss of membrane GM-CSFRα following GM-CSF exposure does not preclude sustained GM-CSF/GM-CSFRα signaling and that this receptor plays a key role in ligand clearance. Hence neutrophilic activation via GM-CSFR may play an important role in neutrophilic lung inflammation even in the absence of high GM-CSF levels or GM-CSFRα expression., (©2018 The Authors. Society for Leukocyte Biology Published by Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
42. Peripheral lymph nodes contain migratory and resident innate lymphoid cell populations.
- Author
-
Dutton EE, Gajdasik DW, Willis C, Fiancette R, Bishop EL, Camelo A, Sleeman MA, Coccia M, Didierlaurent AM, Tomura M, Pilataxi F, Morehouse CA, Carlesso G, and Withers DR
- Subjects
- Animals, Flow Cytometry, Interferon-gamma metabolism, L-Selectin metabolism, Lymph Nodes immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Receptors, CCR7 metabolism, Sphingosine-1-Phosphate Receptors genetics, Transcriptome, Cell Movement immunology, Immunity, Innate immunology, Killer Cells, Natural immunology, Lymph Nodes cytology, Th1 Cells immunology
- Abstract
Tissue residency is considered a defining feature of the innate lymphoid cell (ILC) populations located within mucosal and adipose tissues. ILCs are also present within all lymphoid tissues, but whether ILCs migrate between lymphoid and nonlymphoid sites and in what context is poorly understood. To determine whether migratory ILCs exist within peripheral lymph nodes (LNs), we labeled all cells within the brachial LN (bLN) of transgenic mice expressing a photoconvertible fluorescent protein by direct exposure to light. Tracking of cellular changes in the labeled LN revealed the gradual migration of new ILCs into the tissue, balanced by egress of ILCs dependent on sphingosine-1-phosphate receptors. Most of the migratory ILCs were ILC1s, entering LNs directly from the circulation in a CD62L- and CCR7-dependent manner and thus behaving like conventional natural killer (cNK) cells. Upon egress, both ILC1s and cNK cells were found to recirculate through peripheral LNs. A distinct population of migratory ILC2s were detected in the LN, but most of the ILC3s were tissue resident. Functionally, both migratory and resident ILC1s within LNs were able to rapidly produce IFN-γ to support the generation of robust T
H 1 T cell responses after immunization. Thus, migratory and resident ILC populations exist within peripheral LNs, with ILC1s, akin to cNK cells, able to traffic into these tissues where they can contribute to the initiation of adaptive immunity., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
43. Discovery and characterisation of an antibody that selectively modulates the inhibitory activity of plasminogen activator inhibitor-1.
- Author
-
Vousden KA, Lundqvist T, Popovic B, Naiman B, Carruthers AM, Newton P, Johnson DJD, Pomowski A, Wilkinson T, Dufner P, de Mendez I, Mallinder PR, Murray C, Strain M, Connor J, Murray LA, Sleeman MA, Lowe DC, Huntington JA, and Vaughan TJ
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Neutralizing chemistry, Antibody Specificity, Humans, Mice, Models, Molecular, Plasminogen Activator Inhibitor 1 chemistry, Protein Conformation, Rats, Antibodies, Neutralizing immunology, Plasminogen Activator Inhibitor 1 immunology
- Abstract
Plasminogen activator inhibitor-1 (PAI-1) is a serine protease inhibitor (serpin) that regulates fibrinolysis, cell adhesion and cell motility via its interactions with plasminogen activators and vitronectin. PAI-1 has been shown to play a role in a number of diverse pathologies including cardiovascular diseases, obesity and cancer and is therefore an attractive therapeutic target. However the multiple patho-physiological roles of PAI-1, and understanding the relative contributions of these in any one disease setting, make the development of therapeutically relevant molecules challenging. Here we describe the identification and characterisation of fully human antibody MEDI-579, which binds with high affinity and specificity to the active form of human PAI-1. MEDI-579 specifically inhibits serine protease interactions with PAI-1 while conserving vitronectin binding. Crystallographic analysis reveals that this specificity is achieved through direct binding of MEDI-579 Fab to the reactive centre loop (RCL) of PAI-1 and at the same exosite used by both tissue and urokinase plasminogen activators (tPA and uPA). We propose that MEDI-579 acts by directly competing with proteases for RCL binding and as such is able to modulate the interaction of PAI-1 with tPA and uPA in a way not previously described for a human PAI-1 inhibitor.
- Published
- 2019
- Full Text
- View/download PDF
44. Combination cancer immunotherapy targeting PD-1 and GITR can rescue CD8 + T cell dysfunction and maintain memory phenotype .
- Author
-
Wang B, Zhang W, Jankovic V, Golubov J, Poon P, Oswald EM, Gurer C, Wei J, Ramos I, Wu Q, Waite J, Ni M, Adler C, Wei Y, Macdonald L, Rowlands T, Brydges S, Siao J, Poueymirou W, MacDonald D, Yancopoulos GD, Sleeman MA, Murphy AJ, and Skokos D
- Subjects
- Animals, Antigens, Differentiation, T-Lymphocyte immunology, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes pathology, Female, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Neoplasms immunology, Phenotype, CD8-Positive T-Lymphocytes immunology, Glucocorticoid-Induced TNFR-Related Protein immunology, Immunologic Memory immunology, Immunotherapy, Neoplasms therapy, Programmed Cell Death 1 Receptor immunology
- Abstract
Most patients with cancer do not develop durable antitumor responses after programmed cell death protein 1 (PD-1) or programmed cell death ligand 1(PD-L1) checkpoint inhibition monotherapy because of an ephemeral reversal of T cell dysfunction and failure to promote long-lasting immunological T cell memory. Activating costimulatory pathways to induce stronger T cell activation may improve the efficacy of checkpoint inhibition and lead to durable antitumor responses. We performed single-cell RNA sequencing of more than 2000 tumor-infiltrating CD8
+ T cells in mice receiving both PD-1 and GITR (glucocorticoid-induced tumor necrosis factor receptor-related protein) antibodies and found that this combination synergistically enhanced the effector function of expanded CD8+ T cells by restoring the balance of key homeostatic regulators CD226 and T cell immunoreceptor with Ig and ITIM domains (TIGIT), leading to a robust survival benefit. Combination therapy decreased CD8+ T cell dysfunction and induced a highly proliferative precursor effector memory T cell phenotype in a CD226-dependent manner. PD-1 inhibition rescued CD226 activity by preventing PD-1-Src homology region 2 (SHP2) dephosphophorylation of the CD226 intracellular domain, whereas GITR agonism decreased TIGIT expression. Unmasking the molecular pathways driving durable antitumor responses will be essential to the development of rational approaches to optimizing cancer immunotherapy., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2018
- Full Text
- View/download PDF
45. Serum biomarker profiles suggest that atopic dermatitis is a systemic disease.
- Author
-
Thijs JL, Strickland I, Bruijnzeel-Koomen CAFM, Nierkens S, Giovannone B, Knol EF, Csomor E, Sellman BR, Mustelin T, Sleeman MA, de Bruin-Weller MS, Herath A, Drylewicz J, May RD, and Hijnen D
- Subjects
- Adult, Animals, Asthma blood, Female, Humans, Inflammation blood, Male, Mice, Mice, Transgenic, Biomarkers blood, Dermatitis, Atopic blood
- Published
- 2018
- Full Text
- View/download PDF
46. Identification of periplakin as a major regulator of lung injury and repair in mice.
- Author
-
Besnard V, Dagher R, Madjer T, Joannes A, Jaillet M, Kolb M, Bonniaud P, Murray LA, Sleeman MA, and Crestani B
- Subjects
- Alveolar Epithelial Cells drug effects, Alveolar Epithelial Cells immunology, Animals, Bleomycin administration & dosage, Bleomycin toxicity, Cytokines immunology, Cytokines metabolism, Disease Models, Animal, Down-Regulation, Humans, Idiopathic Pulmonary Fibrosis chemically induced, Idiopathic Pulmonary Fibrosis immunology, Lung Injury chemically induced, Lung Injury immunology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Plakins genetics, Respiratory Mucosa cytology, Respiratory Mucosa drug effects, Respiratory Mucosa immunology, Signal Transduction immunology, Alveolar Epithelial Cells pathology, Idiopathic Pulmonary Fibrosis pathology, Lung Injury pathology, Plakins metabolism, Respiratory Mucosa pathology
- Abstract
Periplakin is a component of the desmosomes that acts as a cytolinker between intermediate filament scaffolding and the desmosomal plaque. Periplakin is strongly expressed by epithelial cells in the lung and is a target antigen for autoimmunity in idiopathic pulmonary fibrosis. The aim of this study was to determine the role of periplakin during lung injury and remodeling in a mouse model of lung fibrosis induced by bleomycin. We found that periplakin expression was downregulated in the whole lung and in alveolar epithelial cells following bleomycin-induced injury. Deletion of the Ppl gene in mice improved survival and reduced lung fibrosis development after bleomycin-induced injury. Notably, Ppl deletion promoted an antiinflammatory alveolar environment linked to profound changes in type 2 alveolar epithelial cells, including overexpression of antiinflammatory cytokines, decreased expression of profibrotic mediators, and altered cell signaling with a reduced response to TGF-β1. These results identify periplakin as a previously unidentified regulator of the response to injury in the lung.
- Published
- 2018
- Full Text
- View/download PDF
47. Central inhibition of granulocyte-macrophage colony-stimulating factor is analgesic in experimental neuropathic pain.
- Author
-
Nicol LSC, Thornton P, Hatcher JP, Glover CP, Webster CI, Burrell M, Hammett K, Jones CA, Sleeman MA, Billinton A, and Chessell I
- Subjects
- Analgesics therapeutic use, Animals, Antibodies therapeutic use, Brain cytology, CD11b Antigen metabolism, Calcium-Binding Proteins metabolism, Cells, Cultured, Cytokines metabolism, Disease Models, Animal, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Glial Fibrillary Acidic Protein metabolism, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Granulocyte-Macrophage Colony-Stimulating Factor immunology, Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Lipopolysaccharides pharmacology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microfilament Proteins metabolism, Neuralgia pathology, Neuroglia drug effects, Signal Transduction drug effects, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Neuralgia drug therapy, Neuralgia metabolism
- Abstract
With less than 50% of patients responding to the current standard of care and poor efficacy and selectivity of current treatments, neuropathic pain continues to be an area of considerable unmet medical need. Biological therapeutics such as monoclonal antibodies (mAbs) provide better intrinsic selectivity; however, delivery to the central nervous system (CNS) remains a challenge. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is well described in inflammation-induced pain, and early-phase clinical trials evaluating its antagonism have exemplified its importance as a peripheral pain target. Here, we investigate the role of this cytokine in a murine model of traumatic nerve injury and show that deletion of the GM-CSF receptor or treatment with an antagonizing mAb alleviates pain. We also demonstrate enhanced analgesic efficacy using an engineered construct that has greater capacity to penetrate the CNS. Despite observing GM-CSF receptor expression in microglia and astrocytes, the gliosis response in the dorsal horn was not altered in nerve injured knockout mice compared with wild-type littermate controls as evaluated by ionized calcium binding adapter molecule 1 (Iba1) and glial fibrillary acidic protein, respectively. Functional analysis of glial cells revealed that pretreatment with GM-CSF potentiated lipopolysaccharide-induced release of proinflammatory cytokines. In summary, our data indicate that GM-CSF is a proinflammatory cytokine that contributes to nociceptive signalling through driving spinal glial cell secretion of proinflammatory mediators. In addition, we report a successful approach to accessing CNS pain targets, providing promise for central compartment delivery of analgesics.
- Published
- 2018
- Full Text
- View/download PDF
48. Corrigendum: The RA-MAP Consortium: a working model for academia-industry collaboration.
- Author
-
Cope AP, Barnes MR, Belson A, Binks M, Brockbank S, Bonachela-Capdevila F, Carini C, Fisher BA, Goodyear CS, Emery P, Ehrenstein MR, Gozzard N, Harris R, Hollis S, Keidel S, Levesque M, Lindholm C, McDermott MF, McInnes IB, Mela CM, Parker G, Read S, Pedersen AW, Ponchel F, Porter D, Rao R, Rowe A, Schulze-Knappe P, Sleeman MA, Symmons D, Taylor PC, Tom B, Tsuji W, Verbeeck D, and Isaacs JD
- Abstract
This corrects the article DOI: 10.1038/nrrheum.2017.200.
- Published
- 2018
- Full Text
- View/download PDF
49. The RA-MAP Consortium: a working model for academia-industry collaboration.
- Author
-
Cope AP, Barnes MR, Belson A, Binks M, Brockbank S, Bonachela-Capdevila F, Carini C, Fisher BA, Goodyear CS, Emery P, Ehrenstein MR, Gozzard N, Harris R, Hollis S, Keidel S, Levesque M, Lindholm C, McDermott MF, McInnes IB, Mela CM, Parker G, Read S, Pedersen AW, Ponchel F, Porter D, Rao R, Rowe A, Schulz-Knappe P, Sleeman MA, Symmons D, Taylor PC, Tom B, Tsuji W, Verbeeck D, and Isaacs JD
- Subjects
- Arthritis, Rheumatoid therapy, Biomarkers, Genomics history, History, 21st Century, Humans, Phenotype, United Kingdom epidemiology, Arthritis, Rheumatoid genetics, Biomedical Research organization & administration, Cooperative Behavior, Genomics methods, Industry organization & administration, Research organization & administration
- Abstract
Collaboration can be challenging; nevertheless, the emerging successes of large, multi-partner, multi-national cooperatives and research networks in the biomedical sector have sustained the appetite of academics and industry partners for developing and fostering new research consortia. This model has percolated down to national funding agencies across the globe, leading to funding for projects that aim to realise the true potential of genomic medicine in the 21st century and to reap the rewards of 'big data'. In this Perspectives article, the experiences of the RA-MAP consortium, a group of more than 140 individuals affiliated with 21 academic and industry organizations that are focused on making genomic medicine in rheumatoid arthritis a reality are described. The challenges of multi-partner collaboration in the UK are highlighted and wide-ranging solutions are offered that might benefit large research consortia around the world.
- Published
- 2018
- Full Text
- View/download PDF
50. EASI p-EASI: Utilizing a combination of serum biomarkers offers an objective measurement tool for disease severity in atopic dermatitis patients.
- Author
-
Thijs JL, Drylewicz J, Fiechter R, Strickland I, Sleeman MA, Herath A, May RD, Bruijnzeel-Koomen CAFM, Knol EF, Giovannone B, de Bruin-Weller MS, Nierkens S, and Hijnen DJ
- Subjects
- Biomarkers blood, Humans, Interleukin-22, Chemokine CCL17 blood, Dermatitis, Atopic blood, Interleukins blood, Receptors, Interleukin-2 blood, Severity of Illness Index
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.