1
|
Neyton LPA, Patel RK, Sarma A, Willmore A, Haller SC, Kangelaris KN, Eckalbar WL, Erle DJ, Krummel MF, Hendrickson CM, Woodruff PG, Langelier CR, Calfee CS, Fragiadakis GK. Distinct pulmonary and systemic effects of dexamethasone in severe COVID-19. Nat Commun 2024; 15:5483. [PMID: 38942804 PMCID: PMC11213873 DOI: 10.1038/s41467-024-49756-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 06/18/2024] [Indexed: 06/30/2024] Open
Abstract
Dexamethasone is the standard of care for critically ill patients with COVID-19, but the mechanisms by which it decreases mortality and its immunological effects in this setting are not understood. Here we perform bulk and single-cell RNA sequencing of samples from the lower respiratory tract and blood, and assess plasma cytokine profiling to study the effects of dexamethasone on both systemic and pulmonary immune cell compartments. In blood samples, dexamethasone is associated with decreased expression of genes associated with T cell activation, including TNFSFR4 and IL21R. We also identify decreased expression of several immune pathways, including major histocompatibility complex-II signaling, selectin P ligand signaling, and T cell recruitment by intercellular adhesion molecule and integrin activation, suggesting these are potential mechanisms of the therapeutic benefit of steroids in COVID-19. We identify additional compartment- and cell- specific differences in the effect of dexamethasone that are reproducible in publicly available datasets, including steroid-resistant interferon pathway expression in the respiratory tract, which may be additional therapeutic targets. In summary, we demonstrate compartment-specific effects of dexamethasone in critically ill COVID-19 patients, providing mechanistic insights with potential therapeutic relevance. Our results highlight the importance of studying compartmentalized inflammation in critically ill patients.
Collapse
Affiliation(s)
- Lucile P A Neyton
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Ravi K Patel
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - Aartik Sarma
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Andrew Willmore
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Sidney C Haller
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | | | - Walter L Eckalbar
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - David J Erle
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Lung Biology Center, University of California, San Francisco, CA, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Carolyn M Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Charles R Langelier
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
| | - Carolyn S Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Anesthesia, University of California, San Francisco, CA, USA
| | - Gabriela K Fragiadakis
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA.
- Division of Rheumatology, University of California, San Francisco, CA, USA.
| |
Collapse
|
2
|
Buters TP, Hameeteman P, Jansen IME, van Hindevoort FC, Ten Voorde W, Grievink HW, Schoonakker M, de Kam ML, Gilroy DW, Feiss G, Rissmann R, Jansen MAA, Burggraaf J, Moerland M. Clinical, cellular and molecular effects of corticosteroids on the response to intradermal lipopolysaccharide administration in healthy volunteers. Clin Pharmacol Ther 2021; 111:964-971. [PMID: 34935141 PMCID: PMC9305467 DOI: 10.1002/cpt.2516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/12/2021] [Indexed: 11/12/2022]
Abstract
The intradermal lipopolysaccharide (LPS) challenge in healthy volunteers has proven to be a valuable tool to study local inflammation in vivo. In the current study the inhibitory effects of oral and topical corticosteroid treatment on intradermal LPS responses were evaluated to benchmark the challenge for future investigational drugs. Twenty‐four healthy male volunteers received a two‐and‐a‐half‐day twice daily (b.i.d.) pretreatment with topical clobetasol propionate 0.05% and six healthy volunteers received a two‐and‐a‐half‐day b.i.d. pretreatment with oral prednisolone at 0.25 mg/kg body weight per administration. Participants received one injection regimen of either 0, 2, or 4 intradermal LPS injections (5 ng LPS in 50 µL 0.9% sodium chloride solution). The LPS response was evaluated by noninvasive (perfusion, skin temperature, and erythema) and invasive assessments (cellular and cytokine responses) in suction blister exudate. Both corticosteroids significantly suppressed the clinical inflammatory response (erythema P = 0.0001 for clobetasol and P = 0.0016 for prednisolone; heat P = 0.0245 for clobetasol, perfusion P < 0.0001 for clobetasol and P = 0.0036 for prednisolone). Clobetasol also significantly reduced the number of monocytes subsets, dendritic cells, natural killer cells, and T cells in blister exudate. A similar effect was observed for prednisolone. No relevant corticosteroid effects were observed on the cytokine response to LPS. We successfully demonstrated that the anti‐inflammatory effects of corticosteroids can be detected using our intradermal LPS challenge model, validating it for evaluation of future investigational drugs, as an initial assessment of the anti‐inflammatory effects of such compounds in a minimally invasive manner.
Collapse
Affiliation(s)
- Thomas P Buters
- Centre for Human Drug Research, Leiden, the Netherlands.,Leiden University Medical Centre, Leiden, the Netherlands
| | | | | | | | - Wouter Ten Voorde
- Centre for Human Drug Research, Leiden, the Netherlands.,Leiden University Medical Centre, Leiden, the Netherlands
| | | | | | | | - Derek W Gilroy
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, University College London, London, UK
| | - Gary Feiss
- Cutanea Life Sciences, Wayne, Pennsylvania, USA
| | - Robert Rissmann
- Centre for Human Drug Research, Leiden, the Netherlands.,Leiden University Medical Centre, Leiden, the Netherlands.,Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Jacobus Burggraaf
- Centre for Human Drug Research, Leiden, the Netherlands.,Leiden University Medical Centre, Leiden, the Netherlands.,Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Matthijs Moerland
- Centre for Human Drug Research, Leiden, the Netherlands.,Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
3
|
Mole S, Harry A, Fowler A, Hotee S, Warburton J, Waite S, Beerahee M, Behm DJ, Badorrek P, Müller M, Faulenbach C, Lazaar AL, Hohlfeld JM. Investigating the effect of TRPV4 inhibition on pulmonary-vascular barrier permeability following segmental endotoxin challenge. Pulm Pharmacol Ther 2020; 64:101977. [PMID: 33189900 DOI: 10.1016/j.pupt.2020.101977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/20/2020] [Accepted: 11/10/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Acute Respiratory Distress Syndrome (ARDS) is associated with increased pulmonary-vascular permeability. In the lung, transient receptor potential vanilloid 4 (TRPV4), a Ca2+-permeable cation channel, is a regulator of endothelial permeability and pulmonary edema. We performed a Phase I, placebo-controlled, double-blind, randomized, parallel group, proof-of-mechanism study to investigate the effects of TRPV4 channel blocker, GSK2798745, on pulmonary-vascular barrier permeability using a model of lipopolysaccharide (LPS)-induced lung inflammation. METHODS Healthy participants were randomized 1:1 to receive 2 single doses of GSK2798745 or placebo, 12 h apart. Two hours after the first dose, participants underwent bronchoscopy and segmental LPS instillation. Total protein concentration and neutrophil counts were measured in bronchoalveolar lavage (BAL) samples collected before and 24 h after LPS challenge, as markers of barrier permeability and inflammation, respectively. The primary endpoint was baseline adjusted total protein concentration in BAL at 24 h after LPS challenge. A Bayesian framework was used to estimate the posterior probability of any percentage reduction (GSK2798745 relative to placebo). Safety endpoints included the incidence of adverse events (AEs), vital signs, 12-lead electrocardiogram, clinical laboratory and haematological evaluations, and spirometry. RESULTS Forty-seven participants were dosed and 45 completed the study (22 on GSK2798745 and 23 on placebo). Overall, GSK2798745 was well tolerated. Small reductions in mean baseline adjusted BAL total protein (~9%) and neutrophils (~7%) in the LPS-challenged segment were observed in the GSK2798745 group compared with the placebo group; however, the reductions did not meet pre-specified success criteria of at least a 95% posterior probability that the percentage reduction in the mean 24-h post LPS BAL total protein level (GSK2798745 relative to placebo) exceeded zero. Median plasma concentrations of GSK2798745 were predicted to inhibit TRPV4 on lung vascular endothelial cells by ~70-85% during the 24 h after LPS challenge; median urea-corrected BAL concentrations of GSK2798745 were 3.0- to 8.7-fold higher than those in plasma. CONCLUSIONS GSK2798745 did not affect segmental LPS-induced elevation of BAL total protein or neutrophils, despite blood and lung exposures that were predicted to be efficacious. CLINICALTRIALS. GOV IDENTIFIER NCT03511105.
Collapse
Affiliation(s)
- Sarah Mole
- GlaxoSmithKline, Gunnells Wood Road, Stevenage, UK.
| | - Anya Harry
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA, 19426, USA
| | - Andy Fowler
- GlaxoSmithKline, Stockley Park, West Uxbridge, Middlesex, UB11 1BT, UK
| | - Sarah Hotee
- GlaxoSmithKline, Gunnells Wood Road, Stevenage, UK
| | | | - Sarah Waite
- GlaxoSmithKline, Stockley Park, West Uxbridge, Middlesex, UB11 1BT, UK
| | | | - David J Behm
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA, 19426, USA
| | - Philipp Badorrek
- Fraunhofer-Institut Fuer Toxikologie und Experimentelle Medizin [ITEM], Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Meike Müller
- Fraunhofer-Institut Fuer Toxikologie und Experimentelle Medizin [ITEM], Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Cornelia Faulenbach
- Fraunhofer-Institut Fuer Toxikologie und Experimentelle Medizin [ITEM], Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Aili L Lazaar
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA, 19426, USA
| | - Jens M Hohlfeld
- Fraunhofer-Institut Fuer Toxikologie und Experimentelle Medizin [ITEM], Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany; Hannover Medical School and German Centre for Lung Research, Medizinische Hochschule Hannover OE6876, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
4
|
Brooks D, Barr LC, Wiscombe S, McAuley DF, Simpson AJ, Rostron AJ. Human lipopolysaccharide models provide mechanistic and therapeutic insights into systemic and pulmonary inflammation. Eur Respir J 2020; 56:13993003.01298-2019. [PMID: 32299854 DOI: 10.1183/13993003.01298-2019] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 03/18/2020] [Indexed: 02/07/2023]
Abstract
Inflammation is a key feature in the pathogenesis of sepsis and acute respiratory distress syndrome (ARDS). Sepsis and ARDS continue to be associated with high mortality. A key contributory factor is the rudimentary understanding of the early events in pulmonary and systemic inflammation in humans, which are difficult to study in clinical practice, as they precede the patient's presentation to medical services. Lipopolysaccharide (LPS), a constituent of the outer membrane of Gram-negative bacteria, is a trigger of inflammation and the dysregulated host response in sepsis. Human LPS models deliver a small quantity of LPS to healthy volunteers, triggering an inflammatory response and providing a window to study early inflammation in humans. This allows biological/mechanistic insights to be made and new therapeutic strategies to be tested in a controlled, reproducible environment from a defined point in time. We review the use of human LPS models, focussing on the underlying mechanistic insights that have been gained by studying the response to intravenous and pulmonary LPS challenge. We discuss variables that may influence the response to LPS before considering factors that should be considered when designing future human LPS studies.
Collapse
Affiliation(s)
- Daniel Brooks
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Laura C Barr
- Dept of Respiratory Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Sarah Wiscombe
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Daniel F McAuley
- School of Medicine, Dentistry and Biomedical Sciences, Institute for Health Sciences, Wellcome-Wolfson Institute for Experimental Medicine, Belfast, UK
| | - A John Simpson
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Anthony J Rostron
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
5
|
Liu FC, Yu HP, Lin CY, Elzoghby AO, Hwang TL, Fang JY. Use of cilomilast-loaded phosphatiosomes to suppress neutrophilic inflammation for attenuating acute lung injury: the effect of nanovesicular surface charge. J Nanobiotechnology 2018; 16:35. [PMID: 29602314 PMCID: PMC5877390 DOI: 10.1186/s12951-018-0364-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/26/2018] [Indexed: 01/04/2023] Open
Abstract
Background Cilomilast is a phosphodiesterase 4 (PDE4) inhibitor for treating inflammatory lung diseases. This agent has a narrow therapeutic index with significant adverse effects on the nervous system. This study was conducted to entrap cilomilast into PEGylated phosphatidylcholine-rich niosomes (phosphatiosomes) to improve pulmonary delivery via the strong affinity to pulmonary surfactant film. Neutrophils were used as a cell model to test the anti-inflammatory activity of phosphatiosomes. In an in vivo approach, mice were given lipopolysaccharide to produce acute lung injury. The surface charge in phosphatiosomes that influenced the anti-inflammatory potency is discussed in this study. Results The average diameter of the phosphatiosomes was about 100 nm. The zeta potential of anionic and cationic nanovesicles was − 35 and 32 mV, respectively. Cilomilast in both its free and nanocapsulated forms inhibited superoxide anion production but not elastase release in activated neutrophils. Cationic phosphatiosomes mitigated calcium mobilization far more effectively than the free drug. In vivo biodistribution evaluated by organ imaging demonstrated a 2-fold ameliorated lung uptake after dye encapsulation into the phosphatiosomes. The lung/brain distribution ratio increased from 3 to 11 after nanocarrier loading. The intravenous nanocarriers deactivated the neutrophils in ALI, resulting in the elimination of hemorrhage and alveolar wall damage. Only cationic phosphatiosomes could significantly suppress IL-1β and TNF-α in the inflamed lung tissue. Conclusions These results suggest that phosphatiosomes should further be investigated as a potential nanocarrier for the treatment of pulmonary inflammation.
Collapse
Affiliation(s)
- Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Huang-Ping Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Cheng-Yu Lin
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, 259 Wen-Hwa 1st Road, Kweishan, Taoyuan, 333, Taiwan
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.,Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Tsong-Long Hwang
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan. .,Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan. .,Cell Pharmacology Laboratory, Graduate Institute of Natural Products, Chang Gung University, 259 Wen-Hwa 1st Road, Kweishan, Taoyuan, 333, Taiwan. .,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan, Taoyuan, Taiwan.
| | - Jia-You Fang
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan. .,Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, 259 Wen-Hwa 1st Road, Kweishan, Taoyuan, 333, Taiwan. .,Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan. .,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan, Taoyuan, Taiwan.
| |
Collapse
|
6
|
Patel NR, Cunoosamy DM, Fagerås M, Taib Z, Asimus S, Hegelund-Myrbäck T, Lundin S, Pardali K, Kurian N, Ersdal E, Kristensson C, Korsback K, Palmér R, Brown MN, Greenaway S, Siew L, Clarke GW, Rennard SI, Make BJ, Wise RA, Jansson P. The development of AZD7624 for prevention of exacerbations in COPD: a randomized controlled trial. Int J Chron Obstruct Pulmon Dis 2018; 13:1009-1019. [PMID: 29628759 PMCID: PMC5877500 DOI: 10.2147/copd.s150576] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background p38 mitogen-activated protein kinase (MAPK) plays a central role in the regulation and activation of pro-inflammatory mediators. COPD patients have increased levels of activated p38 MAPK, which correlate with increased lung function impairment, alveolar wall inflammation, and COPD exacerbations. Objectives These studies aimed to assess the effect of p38 inhibition with AZD7624 in healthy volunteers and patients with COPD. The principal hypothesis was that decreasing lung inflammation via inhibition of p38α would reduce exacerbations and improve quality of life for COPD patients at high risk for acute exacerbations. Methods The p38 isoform most relevant to lung inflammation was assessed using an in situ proximity ligation assay in severe COPD patients and donor controls. Volunteers aged 18–55 years were randomized into the lipopolysaccharide (LPS) challenge study, which investigated the effect of a single dose of AZD7624 vs placebo on inflammatory biomarkers. The Proof of Principle study randomized patients aged 40–85 years with a diagnosis of COPD for >1 year to AZD7624 or placebo to assess the effect of p38 inhibition in decreasing the rate of exacerbations. Results The p38 isoform most relevant to lung inflammation was p38α, and AZD7624 specifically inhibited p38α and p38β isoforms in human alveolar macrophages. Thirty volunteers were randomized in the LPS challenge study. AZD7624 reduced the increase from baseline in sputum neutrophils and TNF-α by 56.6% and 85.4%, respectively (p<0.001). In the 213 patients randomized into the Proof of Principle study, there was no statistically significant difference between AZD7624 and placebo when comparing the number of days to the first moderate or severe exacerbation or early dropout. Conclusion Although p38α is upregulated in the lungs of COPD patients, AZD7624, an isoform-specific inhaled p38 MAPK inhibitor, failed to show any benefit in patients with COPD.
Collapse
Affiliation(s)
- Naimish R Patel
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden.,Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Hospital, Boston, MA
| | - Danen M Cunoosamy
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Malin Fagerås
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Ziad Taib
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Sara Asimus
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | | | - Sofia Lundin
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Katerina Pardali
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Nisha Kurian
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Eva Ersdal
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | | | - Katarina Korsback
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Robert Palmér
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Mary N Brown
- Innovative Medicines and Early Development, AstraZeneca, Boston, MA, USA
| | | | - Leonard Siew
- Quintiles Drug Research Unit at Guy's Hospital, London
| | - Graham W Clarke
- Quintiles Drug Research Unit at Guy's Hospital, London.,Department of Cardiothoracic Pharmacology, NHLI, Imperial College London, London, UK
| | - Stephen I Rennard
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska, Omaha, NE, USA.,Clinical Discovery Unit, Innovative Medicines and Early Development, AstraZeneca, Cambridge, UK
| | - Barry J Make
- Division of Pulmonary Sciences and Critical Care Medicine, National Jewish Health, University of Colorado, Denver, CO
| | - Robert A Wise
- Division of Pulmonary and Critical Care, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Paul Jansson
- Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
7
|
Lexmond AJ, Singh D, Frijlink HW, Clarke GW, Page CP, Forbes B, van den Berge M. Realising the potential of various inhaled airway challenge agents through improved delivery to the lungs. Pulm Pharmacol Ther 2018; 49:27-35. [PMID: 29331645 DOI: 10.1016/j.pupt.2018.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 10/18/2022]
Abstract
Inhaled airway challenges provoke bronchoconstriction in susceptible subjects and are a pivotal tool in the diagnosis and monitoring of obstructive lung diseases, both in the clinic and in the development of new respiratory medicines. This article reviews the main challenge agents that are in use today (methacholine, mannitol, adenosine, allergens, endotoxin) and emphasises the importance of controlling how these agents are administered. There is a danger that the optimal value of these challenge agents may not be realised due to suboptimal inhaled delivery; thus considerations for effective and reproducible challenge delivery are provided. This article seeks to increase awareness of the importance of precise delivery of inhaled agents used to challenge the airways for diagnosis and research, and is intended as a stepping stone towards much-needed standardisation and harmonisation in the administration of inhaled airway challenge agents.
Collapse
Affiliation(s)
- Anne J Lexmond
- King's College London, Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, 150 Stamford Street, London SE1 9NH, United Kingdom; University of Groningen, Department of Pharmaceutical Technology and Biopharmacy, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Dave Singh
- University of Manchester, Medicines Evaluation Unit, University Hospital of South Manchester Foundation Trust, The Langley Building, Southmoor Road, Wythenshawe, Manchester M23 9QZ, United Kingdom
| | - Henderik W Frijlink
- University of Groningen, Department of Pharmaceutical Technology and Biopharmacy, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Graham W Clarke
- hVIVO, Queen Mary BioEnterprises Innovation Centre, 42 New Road, London E1 2AX, United Kingdom; Imperial College, Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Guy Scadding Building, Cale Street, London SW3 6LY, United Kingdom
| | - Clive P Page
- King's College London, Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Ben Forbes
- King's College London, Institute of Pharmaceutical Science, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Department of Pulmonary Diseases, Hanzeplein 1, 9700 RB Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Hanzeplein 1, 9700 RB Groningen, The Netherlands
| |
Collapse
|
8
|
Singh D. Evaluation of New Drugs for Asthma and COPD: Endpoints, Biomarkers and Clinical Trial Design. Handb Exp Pharmacol 2017; 237:243-264. [PMID: 27838852 DOI: 10.1007/164_2016_70] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There remains a considerable need to develop novel therapies for patients with asthma and chronic obstructive pulmonary disease (COPD). The greatest challenge at the moment is measuring the effects of novel anti-inflammatory drugs, as these drugs often cause only small effects on lung function. Measurements that demonstrate the pharmacological and clinical effects of these drugs are needed. Furthermore, we now recognise that only subgroups of patients are likely to respond to these novel drugs, so using biomarkers to determine the clinical phenotype most suitable for such therapies is important. An endotype is a subtype of a (clinical) condition defined by a distinct pathophysiological mechanism. An endotype-driven approach may be more helpful in drug development, enabling drugs to be targeted specifically towards specific biological mechanisms rather than clinical characteristics. This requires the development of biomarkers to define endotypes and/or to measure drug effects. This newer approach should continue alongside efforts to optimise the measurement of clinical endpoints, including patient-reported outcome measurements, required by drug regulatory authorities.
Collapse
Affiliation(s)
- Dave Singh
- Medicines Evaluation Unit, University of Manchester, University Hospital of South Manchester Foundations Trust, Langley Building, Southmoor Road, Wythenshawe, Manchester, M23 9Q2, UK.
| |
Collapse
|
9
|
Bhagwat SS, Larsen AK, Seternes OM, Bang BE. Mixed exposure to bacterial lipopolysaccharide and seafood proteases augments inflammatory signalling in an airway epithelial cell model (A549). Toxicol Ind Health 2016; 32:1866-1874. [PMID: 26149191 DOI: 10.1177/0748233715590914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Seafood industry workers exhibit increased prevalence of respiratory symptoms due to exposure to bioaerosols containing a mixture of bioactive agents. In this study, a human pulmonary epithelial cell model (A549) was exposed to mixtures of bacterial lipopolysaccharide (LPS) and protease-activated receptor-2 (PAR-2) agonists H-Ser-Leu-Ile-Gly-Lys-Val-NH2 (SLIGKV-NH2), purified salmon ( Salmo salar) trypsin or purified king crab ( Paralithodes camtschaticus) trypsin. The inflammatory response was measured based on nuclear factor-kappa B (NF-κB) activation of transcription in a luciferase reporter gene assay and interleukin 8 (IL-8) secretion in an enzyme-linked immunosorbent assay. We observed that mixtures of SLIGKV-NH2 or trypsins with LPS augmented the activation of NF-κB and secretion of IL-8. The effect on IL-8 secretion was synergistic when both trypsins and LPS were used in the lower concentration range. The results demonstrate that exposure to mixtures of agents that are relevant to seafood industry workplaces may lead to increased inflammatory signalling compared with exposure to the individual agents alone. Furthermore, the results indicate that synergism may occur with the combined exposure to seafood trypsins and LPS and is most likely to occur when exposure to either agent is low.
Collapse
Affiliation(s)
- Sampada S Bhagwat
- 1 Department of Occupational and Environmental Medicine, University Hospital of North Norway, Tromsø, Norway.,2 Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Anett K Larsen
- 3 Department of Arctic and Marine Biology, Faculty of Biosciences, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ole-Morten Seternes
- 4 Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Berit E Bang
- 1 Department of Occupational and Environmental Medicine, University Hospital of North Norway, Tromsø, Norway.,2 Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
10
|
Gupta V, Banyard A, Mullan A, Sriskantharajah S, Southworth T, Singh D. Characterization of the inflammatory response to inhaled lipopolysaccharide in mild to moderate chronic obstructive pulmonary disease. Br J Clin Pharmacol 2016; 79:767-76. [PMID: 25377849 DOI: 10.1111/bcp.12546] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 10/28/2014] [Indexed: 01/10/2023] Open
Abstract
AIMS Lipopolysaccharide (LPS) inhalation causes increased airway and systemic inflammation. We investigated LPS inhalation in patients with chronic obstructive pulmonary disease (COPD) as a model of bacterial exacerbations. We studied safety, changes in sputum and systemic biomarkers. We have also investigated interleukin (IL)-17 concentrations in this model. METHODS Twelve COPD patients inhaled 5 μg LPS. Safety was monitored over 24 h. Sputum was induced at baseline, 6 and 24 h for cells and IL-8, IL-17, neutrophil elastase, monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1β (MIP-1β) in supernatants. Serum was collected at baseline, 4, 8 and 24 h for IL-6, C-reactive protein (CRP) and Clara cell protein (CC-16) concentrations. Peripheral blood mononuclear cells (PBMCs) were isolated at baseline and 4 h for systemic IL-17 analysis. RESULTS LPS 5 μg was well tolerated. The greatest FEV1 change was 11.7% (mean) at 1 h (95% CI 5.1-18.2%). There was a large range in maximal fall (2.5-37.7%). Total sputum cell count and neutrophil count significantly increased 6 and 24 h post-LPS. There was no change in sputum supernatant mediators. IL-6, CRP and CC-16 increased post-inhalation, with different temporal patterns. CD4+ and CD8+ cell associated IL-17 significantly increased at 4 h. CONCLUSIONS Inhaled LPS in COPD patients safely causes increased airway and systemic inflammation. This may be a model for studying COPD exacerbations.
Collapse
Affiliation(s)
- Vandana Gupta
- Medicines Evaluation Unit, University Hospital of South Manchester Foundation Trust, University of Manchester, Southmoor Rd, Manchester, UK
| | | | | | | | | | | |
Collapse
|
11
|
Bartko J, Stiebellehner L, Derhaschnig U, Schoergenhofer C, Schwameis M, Prosch H, Jilma B. Dissociation between systemic and pulmonary anti-inflammatory effects of dexamethasone in humans. Br J Clin Pharmacol 2016; 81:865-77. [PMID: 26647918 PMCID: PMC4834593 DOI: 10.1111/bcp.12857] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/23/2015] [Accepted: 12/01/2015] [Indexed: 12/14/2022] Open
Abstract
Aims The local pulmonary inflammatory response has a different temporal and qualitative profile compared with the systemic inflammatory response. Although glucocorticoids substantially downregulate the systemic release of acute‐phase mediators, it is not clear whether they have comparable inhibitory effects in the human lung compartment. Therefore, we compared the anti‐inflammatory effects of a pure glucocorticoid agonist, dexamethasone, on bronchoalveolar lavage and blood cytokine concentrations in response to bronchially instilled endotoxin. Methods In this randomized, double‐blind and placebo‐controlled trial, 24 volunteers received dexamethasone or placebo and had endotoxin instilled into a lung segment and saline instilled into a contralateral segment, followed by bronchoalveolar lavage. Results Bronchially instilled endotoxin induced a local and systemic inflammatory response. Dexamethasone strongly blunted the systemic interleukin (IL) 6 and C‐reactive protein release. In sharp contrast, dexamethasone left the local release of acute‐phase mediators in the lungs virtually unchanged: bronchoalveolar lavage levels of IL‐6 were only 18% lower and levels of IL‐8 were even higher with dexamethasone compared with placebo, although the differences between treatments were not statistically significant (P = 0.07 and P = 0.08, respectively). However, dexamethasone had inhibitory effects on pulmonary protein extravasation and neutrophil migration. Conclusions The present study demonstrated a remarkable dissociation between the systemic anti‐inflammatory effects of glucocorticoids and its protective effects on capillary leak on the one hand and surprisingly low anti‐inflammatory effects in the lungs on the other.
Collapse
Affiliation(s)
- Johann Bartko
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Ulla Derhaschnig
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Michael Schwameis
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Prosch
- Department of Radiology, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Singh D, Siew L, Christensen J, Plumb J, Clarke GW, Greenaway S, Perros-Huguet C, Clarke N, Kilty I, Tan L. Oral and inhaled p38 MAPK inhibitors: effects on inhaled LPS challenge in healthy subjects. Eur J Clin Pharmacol 2015; 71:1175-84. [PMID: 26265232 PMCID: PMC4564450 DOI: 10.1007/s00228-015-1920-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/28/2015] [Indexed: 11/12/2022]
Abstract
Background Inhaled LPS causes neutrophilic airway inflammation in healthy subjects. We compared the effects of p38 MAPK inhibitors and fluticasone propionate on the LPS response. Methods Three randomised, double-blind, placebo-controlled, single dose crossover studies were performed. Active treatments were the oral p38 MAPK inhibitor PH-797804 30 mg (study 1), PH-797804 30 mg and the inhaled p38 MAPK inhibitor PF-03715455 20 mg (study 2) and inhaled fluticasone propionate 500 μg (study 3). The primary endpoint was sputum neutrophil percentage. Results Sputum neutrophil percentage post-LPS challenge was significantly inhibited (15.1 and 15.3 % reduction) by PH-797804 compared to placebo in studies 1 and 2 (p = 0.0096 and 0.0001, respectively), and by PF-03715455 (8.0 % reduction, p = 0.031); fluticasone propionate had no effect. PH-797804 significantly inhibited the increase in inflammatory mediators (IL-6, MCP-1, MIP1β and CC16) in sputum supernatant, while PF-03715455 had no effect. PH-797804 and PF-03715455 both inhibited IL-6, MCP-1, MIP1β, CC16 and CRP levels in plasma, with PH-797804 having greater effects. Fluticasone propionate had no effect on sputum supernatant or plasma biomarkers. Conclusions PH-797804 had the greatest impact on neutrophilic airway inflammation. Oral administration of p38 MAPK inhibitors may optimise pulmonary anti-inflammatory effects. Electronic supplementary material The online version of this article (doi:10.1007/s00228-015-1920-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dave Singh
- University Of Manchester, Medicines Evaluation Unit, University Hospital of South Manchester Foundation Trust, Manchester, M23 9QZ, UK.
| | - Leonard Siew
- Quintiles Drug Research Unit, Respiratory and Inflammation Early Clinical Development, Quintiles Ltd, London, SE1 1YR, UK
| | | | - Jonathan Plumb
- University Of Manchester, Medicines Evaluation Unit, University Hospital of South Manchester Foundation Trust, Manchester, M23 9QZ, UK
| | - Graham W Clarke
- Quintiles Drug Research Unit, Respiratory and Inflammation Early Clinical Development, Quintiles Ltd, London, SE1 1YR, UK.,Department of Cardiothoracic Pharmacology, Imperial College, National Heart and Lung Institute, London, UK
| | - Steve Greenaway
- Quintiles Drug Research Unit, Respiratory and Inflammation Early Clinical Development, Quintiles Ltd, London, SE1 1YR, UK
| | | | - Nick Clarke
- Pfizer Inc. Inflammation and Remodeling Unit, Cambridge, MA, USA
| | - Iain Kilty
- Pfizer Inc. Inflammation and Remodeling Unit, Cambridge, MA, USA
| | - Lisa Tan
- Pfizer Inc. Inflammation and Remodeling Unit, Cambridge, MA, USA
| |
Collapse
|
13
|
Michel O, Dinh PHD, Doyen V, Corazza F. Anti-TNF inhibits the airways neutrophilic inflammation induced by inhaled endotoxin in human. BMC Pharmacol Toxicol 2014; 15:60. [PMID: 25371053 PMCID: PMC4236481 DOI: 10.1186/2050-6511-15-60] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 10/14/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Inhaled endotoxin induces airways'neutrophilia, in human. TNF-a being a key cytokine in the response to endotoxin, the effect of anti-TNF on the endotoxin-induced neutrophilic response was evaluated among healthy volunteers. METHODS Among a population of 30 healthy subjects, an induced-sputum was collected 2 weeks before, and 24 hours after an inhalation of 20 mcg endotoxin (E. coli 026:B6). Then, the subjects were randomized into 3 parallel groups treated with control, oral methylprednisolone 20 mg/day during 7 days or anti-TNF (adalimumab, Humira®, Abbott) 40 mg s.c.. One week later, an induced-sputum was sampled, 24 hours after an inhalation of endotoxin. RESULTS After endotoxin inhalation, the number of total cells, neutrophils and macrophages was significantly increased (p <0.001). Compared to the response to endotoxin among the control group, anti-TNF inhibited the endotoxin-induced neutrophil influx, both in relative (51.3 (±6.4)% versus 26.2 (±5.3)%, p <0.002) and in absolute values (1321 (443-3935) cells/mcL versus 247 (68-906) cells/mcL, p <0.02). The endotoxin-induced neutrophilic response was not significantly modified among the control group and oral corticosteroid group. CONCLUSIONS While oral corticosteroid had no effect, anti-TNF inhibited the neutrophil influx in sputum, induced by inhalation of endotoxin, in human subject. The endotoxin model could be an early predictor of clinical efficacy of novel therapeutics. TRIAL REGISTRATION ClinicalTrials.gov NCT02252809 (EudraCT2008-005526-37).
Collapse
Affiliation(s)
- Olivier Michel
- Clinic of Allergology and Immunology, CHU Brugmann (Université Libre de Bruxelles - ULB), 4 pl Van Gehuchten, B -1020, Brussels, Belgium.
| | | | | | | |
Collapse
|
14
|
Barr LC, Brittan M, Morris AC, McAuley DF, McCormack C, Fletcher AM, Richardson H, Connell M, Patel D, Wallace WAH, Rossi AG, Davidson DJ, Manson L, Turner M, Hirani N, Walsh TS, Anderson NH, Dhaliwal K, Simpson AJ. A randomized controlled trial of peripheral blood mononuclear cell depletion in experimental human lung inflammation. Am J Respir Crit Care Med 2013; 188:449-55. [PMID: 23627345 DOI: 10.1164/rccm.201212-2334oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
RATIONALE Depletion of monocytes reduces LPS-induced lung inflammation in mice, suggesting monocytes as potential therapeutic targets in acute lung injury. OBJECTIVES To investigate whether depletion of circulating blood monocytes has beneficial effects on markers of systemic and pulmonary inflammation in a human model of acute lung inflammation. METHODS A total of 30 healthy volunteers were enrolled in a randomized controlled trial. Volunteers inhaled LPS at baseline, and were randomized to receive active mononuclear cell depletion by leukapheresis, or sham leukapheresis, in a double-blind fashion (15 volunteers per group). Serial blood counts were measured, bronchoalveolar lavage (BAL) was performed at 9 hours, and [(18)F]fluorodeoxyglucose positron emission tomography at 24 hours. The primary endpoint was the increment in circulating neutrophils at 8 hours. MEASUREMENTS AND MAIN RESULTS As expected, inhalation of LPS induced neutrophilia and an up-regulation of inflammatory mediators in the blood and lungs of all volunteers. There was no significant difference between the depletion and sham groups in the mean increment in blood neutrophil count at 8 hours (6.16 × 10(9)/L and 6.15 × 10(9)/L, respectively; P = 1.00). Furthermore, there were no significant differences in BAL neutrophils or protein, positron emission tomography-derived measures of global lung inflammation, or cytokine levels in plasma or BAL supernatant between the study groups. No serious adverse events occurred, and no symptoms were significantly different between the groups. CONCLUSIONS These findings do not support a role for circulating human monocytes in the early recruitment of neutrophils during LPS-mediated acute lung inflammation in humans.
Collapse
Affiliation(s)
- Laura C Barr
- University of Edinburgh/Medical Research Council Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Aul R, Armstrong J, Duvoix A, Lomas D, Hayes B, Miller BE, Jagger C, Singh D. Inhaled LPS challenges in smokers: a study of pulmonary and systemic effects. Br J Clin Pharmacol 2013; 74:1023-32. [PMID: 22469312 DOI: 10.1111/j.1365-2125.2012.04287.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
AIMS Lipopolysaccharide (LPS) is a TLR4 agonist which activates NFκB dependent cytokine production. We investigated LPS inhalation in healthy smokers as a model of COPD bacterial exacerbations. We studied safety, reproducibility, the translocation of the NFκB subunit p65 in sputum cells and changes in systemic biomarkers of inflammation. METHODS Twelve smokers inhaled 5 and 30 µg LPS and safety was monitored over 24 h. IL-6, CRP, CCl-18, SP-D, CC-16 and β-defensin 2 were measured in serum samples collected at baseline, 4, 8 and 24 h. Sputum was induced at baseline, 6 and 24 h for cell counts and p65 expression. Repeated challenges were performed after a 2 week interval in 10 smokers. RESULTS LPS inhalation was well tolerated. Significant increases occurred in sputum neutrophil counts with both doses, with a maximum increase of 21.5% at 6 h after 30 µg which was reproducible, r(i ) (intraclass correlation coefficient) = 0.88. LPS increased sputum cell nuclear p65 translocation and phospho-p65 expression. All of the serum biomarkers increased following challenge but with different temporal patterns. DISCUSSION Inhaled LPS challenge in smokers causes pulmonary and systemic inflammation that involves NFκB activation. This appears to be a suitable model for studying bacterial exacerbations of COPD.
Collapse
Affiliation(s)
- Raminder Aul
- University of Manchester, Medicines Evaluation Unit, University Hospital of South Manchester, Manchester, UK
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Janssen O, Schaumann F, Holz O, Lavae-Mokhtari B, Welker L, Winkler C, Biller H, Krug N, Hohlfeld JM. Low-dose endotoxin inhalation in healthy volunteers--a challenge model for early clinical drug development. BMC Pulm Med 2013; 13:19. [PMID: 23537365 PMCID: PMC3635929 DOI: 10.1186/1471-2466-13-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 03/22/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Inhalation of endotoxin (LPS) induces a predominantly neutrophilic airway inflammation and has been used as model to test the anti-inflammatory activity of novel drugs. In the past, a dose exceeding 15-50 μg was generally needed to induce a sufficient inflammatory response. For human studies, regulatory authorities in some countries now request the use of GMP-grade LPS, which is of limited availability. It was therefore the aim of this study to test the effect and reproducibility of a low-dose LPS challenge (20,000 E.U.; 2 μg) using a flow- and volume-controlled inhalation technique to increase LPS deposition. METHODS Two to four weeks after a baseline sputum induction, 12 non-smoking healthy volunteers inhaled LPS on three occasions, separated by at least 4 weeks. To modulate the inflammatory effect of LPS, a 5-day PDE4 inhibitor (Roflumilast) treatment preceded the last challenge. Six hours after each LPS inhalation, sputum induction was performed. RESULTS The low-dose LPS inhalation was well tolerated and increased the mean percentage of sputum neutrophils from 25% to 72%. After the second LPS challenge, 62% neutrophils and an increased percentage of monocytes were observed. The LPS induced influx of neutrophils and the cumulative inflammatory response compared with baseline were reproducible. Treatment with Roflumilast for 5 days did not have a significant effect on sputum composition. CONCLUSION The controlled inhalation of 2 μg GMP-grade LPS is sufficient to induce a significant neutrophilic airway inflammation in healthy volunteers. Repeated low-dose LPS challenges potentially result in a small shift of the neutrophil/monocyte ratio; however, the cumulative response is reproducible, enabling the use of this model for "proof-of-concept" studies for anti-inflammatory compounds during early drug development.
Collapse
Affiliation(s)
- Ole Janssen
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
van der Merwe R, Molfino NA. Challenge models to assess new therapies in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2012; 7:597-605. [PMID: 23055710 PMCID: PMC3459659 DOI: 10.2147/copd.s30664] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity and mortality. Current therapies confer partial benefits either by incompletely improving airflow limitation or by reducing acute exacerbations, hence new therapies are desirable. In the absence of robust early predictors of clinical efficacy, the potential success of novel therapeutic agents in COPD will not entirely be known until the drugs enter relatively large and costly clinical trials. New predictive models in humans, and new study designs are being sought to allow for confirmation of pharmacodynamic and potentially clinically meaningful effects in early development. This review focuses on human challenge models with lipopolysaccharide endotoxin, ozone, and rhinovirus, in the early clinical development phases of novel therapeutic agents for the treatment and reduction of exacerbations in COPD.
Collapse
|
18
|
Seehase S, Lauenstein HD, Schlumbohm C, Switalla S, Neuhaus V, Förster C, Fieguth HG, Pfennig O, Fuchs E, Kaup FJ, Bleyer M, Hohlfeld JM, Braun A, Sewald K, Knauf S. LPS-induced lung inflammation in marmoset monkeys - an acute model for anti-inflammatory drug testing. PLoS One 2012; 7:e43709. [PMID: 22952743 PMCID: PMC3429492 DOI: 10.1371/journal.pone.0043709] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 07/25/2012] [Indexed: 01/18/2023] Open
Abstract
Increasing incidence and substantial morbidity and mortality of respiratory diseases requires the development of new human-specific anti-inflammatory and disease-modifying therapeutics. Therefore, new predictive animal models that closely reflect human lung pathology are needed. In the current study, a tiered acute lipopolysaccharide (LPS)-induced inflammation model was established in marmoset monkeys (Callithrix jacchus) to reflect crucial features of inflammatory lung diseases. Firstly, in an ex vivo approach marmoset and, for the purposes of comparison, human precision-cut lung slices (PCLS) were stimulated with LPS in the presence or absence of the phosphodiesterase-4 (PDE4) inhibitor roflumilast. Pro-inflammatory cytokines including tumor necrosis factor-alpha (TNF-α) and macrophage inflammatory protein-1 beta (MIP-1β) were measured. The corticosteroid dexamethasone was used as treatment control. Secondly, in an in vivo approach marmosets were pre-treated with roflumilast or dexamethasone and unilaterally challenged with LPS. Ipsilateral bronchoalveolar lavage (BAL) was conducted 18 hours after LPS challenge. BAL fluid was processed and analyzed for neutrophils, TNF-α, and MIP-1β. TNF-α release in marmoset PCLS correlated significantly with human PCLS. Roflumilast treatment significantly reduced TNF-α secretion ex vivo in both species, with comparable half maximal inhibitory concentration (IC(50)). LPS instillation into marmoset lungs caused a profound inflammation as shown by neutrophilic influx and increased TNF-α and MIP-1β levels in BAL fluid. This inflammatory response was significantly suppressed by roflumilast and dexamethasone. The close similarity of marmoset and human lungs regarding LPS-induced inflammation and the significant anti-inflammatory effect of approved pharmaceuticals assess the suitability of marmoset monkeys to serve as a promising model for studying anti-inflammatory drugs.
Collapse
Affiliation(s)
- Sophie Seehase
- Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Pathology Unit, German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany
| | - Hans-Dieter Lauenstein
- Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Pathology Unit, German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany
| | | | - Simone Switalla
- Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Vanessa Neuhaus
- Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Christine Förster
- Institute of Pathology, Klinikum Region Hannover Klinikum Nordstadt, Hannover, Germany
| | - Hans-Gerd Fieguth
- Division of Thoracic Surgery, Klinikum Region Hannover Klinikum Oststadt-Heidehaus, Hannover, Germany
| | - Olaf Pfennig
- Institute of Pathology, Klinikum Region Hannover Klinikum Nordstadt, Hannover, Germany
| | | | - Franz-Josef Kaup
- Pathology Unit, German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany
| | - Martina Bleyer
- Pathology Unit, German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany
| | - Jens M. Hohlfeld
- Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Armin Braun
- Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Katherina Sewald
- Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Sascha Knauf
- Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Pathology Unit, German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany
| |
Collapse
|
19
|
Aul R, Patel S, Summerhill S, Kilty I, Plumb J, Singh D. LPS challenge in healthy subjects: An investigation of neutrophil chemotaxis mechanisms involving CXCR1 and CXCR2. Int Immunopharmacol 2012; 13:225-31. [DOI: 10.1016/j.intimp.2012.04.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 04/16/2012] [Accepted: 04/17/2012] [Indexed: 01/28/2023]
|
20
|
Lee YJ, Eun JR. Cilostazol Decreases Ethanol-Mediated TNFalpha Expression in RAW264.7 Murine Macrophage and in Liver from Binge Drinking Mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2012; 16:131-8. [PMID: 22563259 PMCID: PMC3339289 DOI: 10.4196/kjpp.2012.16.2.131] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 03/15/2012] [Accepted: 04/03/2012] [Indexed: 01/18/2023]
Abstract
Alcoholic hepatitis is a leading cause of liver failure in which the increased production of tumor necrosis factor α (TNFα) plays a critical role in progression of alcoholic liver disease. In the present study, we investigated the effects of cilostazol, a selective inhibitor of type III phosphodiesterase on ethanol-mediated TNFα production in vitro and in vivo, and the effect of cilostazol was compared with that of pentoxifylline, which is currently used in clinical trial. RAW264.7 murine macrophages were pretreated with ethanol in the presence or absence of cilostazol then, stimulated with lipopolysacchride (LPS). Cilostazol significantly suppressed the level of LPS-stimulated TNFα mRNA and protein with a similar degree to that by pentoxifylline. Cilostazol increased the basal AMP-activated protein kinase (AMPK) activity as well as normalized the decreased AMPK by LPS. AICAR, an AMPK activator and db-cAMP also significantly decreased TNFα production in RAW264.7 cells, but cilostazol did not affect the levels of intracellular cAMP and reactive oxygen species (ROS) production. The in vivo effect of cilostazol was examined using ethanol binge drinking (6 g/kg) mice model. TNFα mRNA and protein decreased in liver from ethanol gavaged mice compared to that from control mice. Pretreatment of mice with cilostazol or pentoxifylline further reduced the TNFα production in liver. These results demonstrated that cilostazol effectively decrease the ethanol-mediated TNFα production both in murine macrophage and in liver from binge drinking mice and AMPK may be responsible for the inhibition of TNFα production by cilostazol.
Collapse
Affiliation(s)
- Youn Ju Lee
- Department of Pharmacology, School of Medicine, Catholic University of Daegu, Daegu 705-718, Korea
| | | |
Collapse
|
21
|
Page CP, Spina D. Selective PDE inhibitors as novel treatments for respiratory diseases. Curr Opin Pharmacol 2012; 12:275-86. [PMID: 22497841 DOI: 10.1016/j.coph.2012.02.016] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 02/23/2012] [Indexed: 02/05/2023]
Abstract
Phosphodiesterases (PDEs) are a family of enzymes which catalyse the metabolism of the intracellular cyclic nucleotides, c-AMP and c-GMP that are expressed in a variety of cell types and in the context of respiratory diseases, It is now recognised that the use of PDE3, PDE4 and mixed PDE3/4 inhibitors can provide clinical benefit to patients with asthma or chronic obstructive pulmonary disease (COPD). The orally active PDE4 inhibitor Roflumilast-n-oxide has been approved for treatment of severe exacerbations of COPD as add-on therapy to standard drugs. This review discusses the involvement of PDEs in airway diseases and various strategies that are currently being pursued to improve efficacy and reduce side-effects of PDE4 inhibitors, including delivery via the inhaled route, mixed PDE inhibitors and/or antisense biologicals targeted towards PDE4.
Collapse
Affiliation(s)
- Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, Franklin Wilkins Building, King's College London, London SE1 9NH, UK.
| | | |
Collapse
|
22
|
Cooper PJ, Rodrigues LC, Barreto ML. Influence of poverty and infection on asthma in Latin America. Curr Opin Allergy Clin Immunol 2012; 12:171-8. [PMID: 22391754 PMCID: PMC7612855 DOI: 10.1097/aci.0b013e3283510967] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE OF REVIEW Asthma in Latin America is a growing public health problem and seems to be most prevalent and cause most morbidity among poor urban populations. This article will review the findings of recent human studies of the associations of asthma prevalence in Latin America with factors associated with poverty and inequality including childhood infections, stress, environment, nutrition and diet. RECENT FINDINGS Most asthma in childhood in Latin America is nonatopic and has been associated with exposures related to environmental dirt, diet and psychosocial distress. These factors are strongly linked to poverty and inequality. Interestingly, infections with bacterial, viral and parasitic pathogens in childhood appear to attenuate atopy in childhood but have no effect on asthma symptoms. There are biologically plausible mechanisms by which dirt exposures (e.g. endotoxin and other microbial products and nonmicrobial irritants), diet and obesity and psychosocial stress may cause airways inflammation. SUMMARY Most childhood asthma in Latin America is nonatopic for which important risk factors are those of poverty including poor hygiene (i.e. dirt), poor diet and obesity and psychosocial stress. There is evidence that exposures to infections in early childhood reduce atopy but not asthma. Research is needed to identify causes of nonatopic asthma that may be suitable for primary prevention or other public health intervention strategies for asthma in Latin America.
Collapse
Affiliation(s)
- Philip J Cooper
- Centro de Investigaciones FEPIS, Quinindé, Esmeraldas Province, Ecuador.
| | | | | |
Collapse
|
23
|
Virtala R, Ekman AK, Jansson L, Westin U, Cardell LO. Airway inflammation evaluated in a human nasal lipopolysaccharide challenge model by investigating the effect of a CXCR2 inhibitor. Clin Exp Allergy 2011; 42:590-6. [DOI: 10.1111/j.1365-2222.2011.03921.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 10/07/2011] [Accepted: 10/13/2011] [Indexed: 12/18/2022]
Affiliation(s)
| | - A.-K. Ekman
- Division of ENT Diseases; CLINTEC; Karolinska Institutet; Karolinska University Hospital; Stockholm Sweden
| | | | - U. Westin
- Department of Otorhinolaryngology; Skåne University Hospital; Lund University; Malmö Sweden
| | - L. O. Cardell
- Division of ENT Diseases; CLINTEC; Karolinska Institutet; Karolinska University Hospital; Stockholm Sweden
| |
Collapse
|
24
|
Korsgren M, Linden M, Entwistle N, Cook J, Wollmer P, Andersson M, Larsson B, Greiff L. Inhalation of LPS induces inflammatory airway responses mimicking characteristics of chronic obstructive pulmonary disease. Clin Physiol Funct Imaging 2011; 32:71-9. [PMID: 22152082 DOI: 10.1111/j.1475-097x.2011.01058.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIM Inhalation of lipopolysaccharide (LPS) produces both systemic and pulmonary inflammatory responses. The aim of this study was to further characterize the response to LPS in order to develop a human model suitable for early testing of drug candidates developed for the treatment for chronic obstructive pulmonary disease (COPD). MATERIALS Blood and induced sputum were obtained 4, 24 and 48 h following inhalation of saline and LPS (5 and 50 μg). Blood was analysed for C-reactive protein (CRP), α(1)-antitrypsin and neutrophils/leucocytes, and sputum was analysed for biomarkers of neutrophil inflammation and remodelling activities, i.e. neutrophil elastase (NE) protein/activity and α(1)-antitrypsin. Levels of tumour necrosis factor-α (TNFα) were measured in both blood and sputum. Urine was collected 0-24 and 24-48 h postchallenge, and desmosine, a biomarker of elastin degradation, was measured. RESULTS Lipopolysaccharide inhalation induced dose-dependent flu-like symptoms and increases in plasma CRP and α(1)-antitrypsin as well as increases in blood neutrophil/leucocyte numbers. Furthermore, LPS produced increases in sputum TNFα and sputum NE activity. Urine levels of desmosine were unaffected by the LPS challenge. All subjects recovered 48 h postchallenge, and indices of inflammatory activity were significantly lower at this observation point cf 24 h postchallenge. CONCLUSION Inhalation of LPS in healthy volunteers can be used as a safe and stable model of neutrophil inflammation. Blood/plasma and sputum indices can be employed to monitor the response to LPS. We suggest that this model may be used for initial human studies of novel COPD-active drugs.
Collapse
Affiliation(s)
- Magnus Korsgren
- Department of Clinical Pharmacology, Lund University Hospital, Lund, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Press NJ, Banner KH. PDE4 inhibitors - a review of the current field. PROGRESS IN MEDICINAL CHEMISTRY 2009; 47:37-74. [PMID: 19328289 DOI: 10.1016/s0079-6468(08)00202-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Neil J Press
- Novartis Institutes for Biomedical Research, Horsham, West Sussex RH12 5AB, UK
| | | |
Collapse
|
26
|
Roflumilast attenuates pulmonary inflammation upon segmental endotoxin challenge in healthy subjects: A randomized placebo-controlled trial. Pulm Pharmacol Ther 2008; 21:616-23. [DOI: 10.1016/j.pupt.2008.02.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Revised: 02/10/2008] [Accepted: 02/13/2008] [Indexed: 11/17/2022]
|
27
|
Alexis NE, Brickey WJ, Lay JC, Wang Y, Roubey RAS, Ting JPY, Peden DB. Development of an inhaled endotoxin challenge protocol for characterizing evoked cell surface phenotype and genomic responses of airway cells in allergic individuals. Ann Allergy Asthma Immunol 2008; 100:206-15. [PMID: 18426139 DOI: 10.1016/s1081-1206(10)60444-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Environmental exposure to endotoxin is a known cause of exacerbation of asthma. Inhaled endotoxin protocols have been used to evaluate airway cell surface phenotypes associated with antigen presentation and innate immunity in healthy volunteers, but not in allergic volunteers. OBJECTIVES To establish the safety of challenge with low-dose endotoxin (10,000 endotoxin units) (lipopolysaccharide [LPS]) inhalation in allergic individuals, to measure airway cell surface phenotypes associated with antigen presentation and innate immunity in induced sputum (IS) after LPS challenge, and to conduct gene expression profiling in IS cells to determine which host genetic networks are modified by LPS inhalation. METHODS Induced sputum was obtained before and 6 hours after LPS inhalation in 10 allergic volunteers (8 with asthma and 2 with rhinitis). Flow cytometry was used to examine cell surface phenotypes on IS cells. Genomic expression was analyzed on a subset of IS samples (n = 10) using microarray and ingenuity pathway analysis. RESULTS A total of 10,000 endotoxin units of LPS induced significant up-regulation of membrane CD14, CD11b, CD16, HLA-DR, CD86, and Fcepsilon receptor 1 on sputum phagocytes and increased expression of genes that influence antigen-presenting surface molecules (HLA-DR, chemokine ligand 2 or monocyte chemoattractant protein 1, v-rel reticuloendotheliosis viral oncogene homolog, prostaglandin-endoperoxide synthase 2 or cyclooxygenase 2, and transforming growth factor beta), immune activation (CD14, interleukin 1beta, and regulated upon activation, normal T cell expressed and secreted), and inflammation (intracellular adhesion molecule 1 and inhibitory kappaBalpha). Gene profiles for nuclear factor kappaB, interleukin 1, and tumor necrosis factor pathways were also significantly affected. CONCLUSIONS Low-dose inhaled endotoxin challenge is safe in allergic individuals with mild to moderate disease. It enhances airway cell surface phenotypes and expression of genes associated with antigen presentation, innate immunity, and inflammation. Microarray with ingenuity pathway analysis can be successfully applied to sputum cells to characterize genetic responses to inhaled exacerbants.
Collapse
Affiliation(s)
- Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| | | | | | | | | | | | | |
Collapse
|