1
|
Cunnion K, Goss J, Hair P, Dell L, Roberson D, Thienel U, Müller M, Carstensen-Aurèche S, Badorrek P, Holz O, Hohlfeld JM. RLS-0071, a novel anti-inflammatory agent, significantly reduced inflammatory biomarkers in a randomised human evaluation of mechanisms and safety study. ERJ Open Res 2024; 10:01006-2023. [PMID: 39286057 PMCID: PMC11403592 DOI: 10.1183/23120541.01006-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/23/2024] [Indexed: 09/19/2024] Open
Abstract
Background This study was a randomised, double-blind, placebo-controlled study intended to establish the translatability of the RLS-0071 mechanisms of action from animal disease models to humans by inhibiting neutrophil-mediated inflammation at the tissue level and major inflammatory biomarkers. We hypothesised that RLS-0071 inhibits a temporary neutrophil-mediated inflammation in the lungs induced by inhalation of low-dose lipopolysaccharide (LPS) in healthy participants. Methods Participants were randomised to one of three arms to receive inhaled LPS followed by three doses of either low-dose (10 mg·kg-1) or high-dose (120 mg·kg-1 loading dose followed by two doses of 40 mg·kg-1) RLS-0071 i.v. or placebo (saline) every 8 h. Biomarkers evaluating inflammatory responses, with absolute neutrophil counts in induced sputum as the primary end-point, were collected before and at 6 and 24 h after LPS challenge. Results Active treatment with RLS-0071 showed a similar safety profile to participants receiving placebo. RLS-0071 significantly decreased the numbers of neutrophils in sputum at 6 h post LPS by approximately half (p=0.04). Neutrophil effectors myeloperoxidase, neutrophil elastase and interleukin-1β in sputum were also significantly decreased at 6 h for RLS-0071 compared with placebo. Several biomarkers showed trends suggesting sustained decreases for RLS-0071 versus placebo at 24 h. Conclusion This clinical trial demonstrated that RLS-0071 was safe and well tolerated and modulated neutrophil-mediated inflammation in humans after inhaled LPS challenge, consistent with results from prior animal model studies.
Collapse
Affiliation(s)
- Kenji Cunnion
- Department of Research and Medical Affairs, ReAlta Life Sciences, Norfolk, VA, USA
- Children's Hospital of The King's Daughters, Norfolk, VA, USA
- Children's Specialty Group, Norfolk, VA, USA
- Dept Paediatrics, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Jessica Goss
- Department of Research and Medical Affairs, ReAlta Life Sciences, Norfolk, VA, USA
| | - Pamela Hair
- Department of Research and Medical Affairs, ReAlta Life Sciences, Norfolk, VA, USA
| | - Linda Dell
- Department of Research and Medical Affairs, ReAlta Life Sciences, Norfolk, VA, USA
| | - Destrey Roberson
- Department of Research and Medical Affairs, ReAlta Life Sciences, Norfolk, VA, USA
| | - Ulrich Thienel
- Department of Research and Medical Affairs, ReAlta Life Sciences, Norfolk, VA, USA
| | - Meike Müller
- Division of Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
- German Centre for Lung Research (DZL, BREATH), Hannover, Germany
| | - Saskia Carstensen-Aurèche
- Division of Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
- German Centre for Lung Research (DZL, BREATH), Hannover, Germany
| | - Philipp Badorrek
- Division of Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - Olaf Holz
- Division of Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
- German Centre for Lung Research (DZL, BREATH), Hannover, Germany
| | - Jens M Hohlfeld
- Division of Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
- German Centre for Lung Research (DZL, BREATH), Hannover, Germany
- Department of Respiratory Medicine and Infectious Disease, Hannover Medical School, Hannover, Germany
| |
Collapse
|
2
|
Daae HL, Graff P, Foss OAH, Kofoed VC, Afanou AK, Frederiksen MW, Madsen AM. A cross-sectional study on occupational exposure to microorganisms, endotoxin, hydrogen sulfide, and dust during work at drilling waste treatment plants. Ann Work Expo Health 2024; 68:58-77. [PMID: 37995292 PMCID: PMC10773208 DOI: 10.1093/annweh/wxad069] [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: 08/02/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023] Open
Abstract
This cross-sectional study aims to obtain knowledge about workers' exposure to airborne dust, bacterial and fungal species, endotoxin, biofilm formation, and hydrogen sulfide (H2S) in drilling waste treatment plants. In total, 408 full-shift personal samples, 66 work areas, 40 drilling waste, and reference (outdoor air and seawater) samples were analyzed. Some workers were exposed to high levels of endotoxin (207 EU/m3), bacteria (3.8 × 104 colony forming units (CFU)/m3 and 9.8 × 104 DNA copies/m3), or fungi (1.4 × 107 CFU/m3 and 3,600 copies/m3). The exposure levels to endotoxin, bacteria, and peaks of H2S were dependent on the treatment technique. All types of drilling waste contained large concentrations of bacteria compared to the seawater references. Elevated concentrations of airborne bacteria were found close to drilling waste basins. In total, 116, 146, and 112 different bacterial species were found in workers' exposure, work areas, and the drilling waste, respectively. An overlap in bacterial species found in the drilling waste and air (personal and work area) samples was found. Of the bacterial species found, 49 are classified as human pathogens such as Escherichia coli, Enterobacter cloacae, and Klebsiella oxytoca. In total, 44 fungal species were found in the working environment, and 6 of these are classified as human pathogens such as Aspergillus fumigatus. In conclusion, across the drilling waste treatment plants, human pathogens were present in the drilling waste, and workers' exposure was affected by the drilling waste treated at the plants with elevated exposure to endotoxin and bacteria. Elevated exposure was related to working as apprentices or chemical engineers, and working with cleaning, or slop water, and working in the daytime.
Collapse
Affiliation(s)
- Hanne Line Daae
- National Institute of Occupational Health (STAMI), Department of Chemistry and Toxicology, Gydas vei 8, Majorstuen, 0363 Oslo, Norway
| | - Pål Graff
- National Institute of Occupational Health (STAMI), Department of Chemistry and Toxicology, Gydas vei 8, Majorstuen, 0363 Oslo, Norway
| | - Oda Astrid Haarr Foss
- National Institute of Occupational Health (STAMI), Department of Chemistry and Toxicology, Gydas vei 8, Majorstuen, 0363 Oslo, Norway
| | - Victor Carp Kofoed
- National Research Centre for the Working Environment, Department of Chemistry and Microbiology, Lersø Parkallé 105, 2100 Copenhagen Ø, Denmark
| | - Anani Komlavi Afanou
- National Institute of Occupational Health (STAMI), Department of Chemistry and Toxicology, Gydas vei 8, Majorstuen, 0363 Oslo, Norway
| | - Margit Wagtberg Frederiksen
- National Research Centre for the Working Environment, Department of Chemistry and Microbiology, Lersø Parkallé 105, 2100 Copenhagen Ø, Denmark
| | - Anne Mette Madsen
- National Research Centre for the Working Environment, Department of Chemistry and Microbiology, Lersø Parkallé 105, 2100 Copenhagen Ø, Denmark
| |
Collapse
|
3
|
Larsson P, Holz O, Koster G, Postle A, Olin AC, Hohlfeld JM. Exhaled breath particles as a novel tool to study lipid composition of epithelial lining fluid from the distal lung. BMC Pulm Med 2023; 23:423. [PMID: 37924084 PMCID: PMC10623716 DOI: 10.1186/s12890-023-02718-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/19/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Surfactant phospholipid (PL) composition plays an important role in lung diseases. We compared the PL composition of non-invasively collected exhaled breath particles (PEx) with bronchoalveolar lavage (BAL) and induced sputum (ISP) at baseline and following endotoxin (LPS) challenges. METHODS PEx and BAL were collected from ten healthy nonsmoking participants before and after segmental LPS challenge. Four weeks later, PEx and ISP were sampled in the week before and after a whole lung LPS inhalation challenge. PL composition was analysed using mass spectrometry. RESULTS The overall PL composition of BAL, ISP and PEx was similar, with PC(32:0) and PC(34:1) representing the largest fractions in all three sample types (baseline PC(32:0) geometric mean mol%: 52.1, 56.9, and 51.7, PC(34:1) mol%: 11.7, 11.9 and 11.4, respectively). Despite this similarity, PEx PL composition was more closely related to BAL than to ISP. For most lipids comparable inter-individual differences in BAL, ISP, and PEx were found. PL composition of PEx was repeatable. The most pronounced increase following segmental LPS challenge was detected for SM(d34:1) in BAL (0.24 to 0.52 mol%) and following inhalation LPS challenge in ISP (0.45 to 0.68 mol%). An increase of SM(d34:1) following segmental LPS challenge was also detectable in PEx (0.099 to 0.103 mol%). The inhalation challenge did not change PL composition of PEx. CONCLUSION Our data supports the peripheral origin of PEx. The lack of PL changes in PEx after inhalation challenge might to be due to the overall weaker response of inhaled LPS which primarily affects the larger airways. Compared with BAL, which always contains lining fluid from both peripheral lung and central airways, PEx analysis might add value as a selective and non-invasive method to investigate peripheral airway PL composition. TRIAL REGISTRATION NCT03044327, first posted 07/02/2017.
Collapse
Affiliation(s)
- Per Larsson
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olaf Holz
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, 30625, Hannover, Germany.
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany.
| | - Grielof Koster
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Anthony Postle
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Anna-Carin Olin
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jens M Hohlfeld
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, 30625, Hannover, Germany
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Hannover Medical School, Department of Respiratory Medicine, Hannover, Germany
| |
Collapse
|
4
|
Gress C, Vogel-Claussen J, Badorrek P, Müller M, Hohl K, Konietzke M, Litzenburger T, Seibold W, Gupta A, Hohlfeld JM. The effect of bradykinin 1 receptor antagonist BI 1026706 on pulmonary inflammation after segmental lipopolysaccharide challenge in healthy smokers. Pulm Pharmacol Ther 2023; 82:102246. [PMID: 37562641 DOI: 10.1016/j.pupt.2023.102246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/25/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Bradykinin 1 receptor (B1R) signalling pathways may be involved in the inflammatory pathophysiology of chronic obstructive pulmonary disease (COPD). B1R signalling is induced by inflammatory stimuli or tissue injury and leads to activation and increased migration of pro-inflammatory cells. Lipopolysaccharide (LPS) lung challenge in man is an experimental method of exploring inflammation in the lung whereby interference in these pathways can help to assess pharmacologic interventions in COPD. BI 1026706, a potent B1R antagonist, was hypothesized to reduce the inflammatory activity after segmental lipopolysaccharide (LPS) challenge in humans due to decreased pulmonary cell influx. METHODS In a monocentric, randomized, double-blind, placebo-controlled, parallel-group, phase I trial, 57 healthy, smoking subjects were treated for 28 days with either oral BI 1026706 100 mg bid or placebo. At day 21, turbo-inversion recovery magnitude magnetic resonance imaging (TIRM MRI) was performed. On the last day of treatment, pre-challenge bronchoalveolar lavage fluid (BAL) and biopsies were sampled, followed by segmental LPS challenge (40 endotoxin units/kg body weight) and saline control instillation in different lung lobes. Twenty-four hours later, TIRM MRI was performed, then BAL and biopsies were collected from the challenged segments. In BAL samples, cells were differentiated for neutrophil numbers as the primary endpoint. Other endpoints included assessment of safety, biomarkers in BAL (e.g. interleukin-8 [IL-8], albumin and total protein), B1R expression in lung biopsies and TIRM score by MRI as a measure for the extent of pulmonary oedema. RESULTS After LPS, but not after saline, high numbers of inflammatory cells, predominantly neutrophils were observed in the airways. IL-8, albumin and total protein were also increased in BAL samples after LPS challenge as compared with saline control. There were no significant differences in cells or other biomarkers from BAL in volunteers treated with BI 1026706 compared with those treated with placebo. Unexpectedly, neutrophil numbers in BAL were 30% higher and MRI-derived extent of oedema was significantly higher with BI 1026706 treatment compared with placebo, 24 h after LPS challenge. Adverse events were mainly mild to moderate and not different between treatment groups. CONCLUSIONS Treatment with BI 1026706 for four weeks was safe and well-tolerated in healthy smoking subjects. BI 1026706 100 mg bid did not provide evidence for anti-inflammatory effects in the human bronchial LPS challenge model. TRIAL REGISTRATION The study was registered on January 14, 2016 at ClinicalTrials.gov (NCT02657408).
Collapse
Affiliation(s)
- Christina Gress
- Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany; German Center for Lung Research (BREATH), Hannover, Germany
| | - Jens Vogel-Claussen
- German Center for Lung Research (BREATH), Hannover, Germany; Department of Diagnostic and Interventional Radiology, Hannover Medical School, Germany
| | - Philipp Badorrek
- Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Meike Müller
- Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany; German Center for Lung Research (BREATH), Hannover, Germany
| | - Kathrin Hohl
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | | | | | - Abhya Gupta
- Boehringer Ingelheim International GmbH, Biberach, Germany
| | - Jens M Hohlfeld
- Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany; German Center for Lung Research (BREATH), Hannover, Germany; Department of Respiratory Medicine, Hannover Medical School, Germany.
| |
Collapse
|
5
|
Resiliac J, Rohlfing M, Santoro J, Hussain SRA, Grayson MH. Low-Dose Lipopolysaccharide Protects from Lethal Paramyxovirus Infection in a Macrophage- and TLR4-Dependent Process. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:348-355. [PMID: 36480273 PMCID: PMC9851983 DOI: 10.4049/jimmunol.2200604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/14/2022] [Indexed: 01/03/2023]
Abstract
Respiratory diseases are a major public health burden and a leading cause of death and disability in the world. Understanding antiviral immune responses is crucial to alleviate morbidity and mortality associated with these respiratory viral infections. Previous data from human and animal studies suggested that pre-existing atopy may provide some protection against severe disease from a respiratory viral infection. However, the mechanism(s) of protection is not understood. Low-dose LPS has been shown to drive an atopic phenotype in mice. In addition, LPS has been shown in vitro to have an antiviral effect. We examined the effect of LPS treatment on mortality to the murine parainfluenza virus Sendai virus. Low-dose LPS treatment 24 h before inoculation with a normally lethal dose of Sendai virus greatly reduced death. This protection was associated with a reduced viral titer and reduced inflammatory cytokine production in the airways. The administration of LPS was associated with a marked increase in lung neutrophils and macrophages. Depletion of neutrophils failed to reverse the protective effect of LPS; however, depletion of macrophages reversed the protective effect of LPS. Further, we demonstrate that the protective effect of LPS depends on type I IFN and TLR4-MyD88 signaling. Together, these studies demonstrate pretreatment with low-dose LPS provides a survival advantage against a severe respiratory viral infection through a macrophage-, TLR4-, and MyD88-dependent pathway.
Collapse
Affiliation(s)
- Jenny Resiliac
- The Ohio State University College of Medicine, Biomedical Sciences Graduate Program, Columbus, Ohio
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Michelle Rohlfing
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Jennifer Santoro
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Syed-Rehan A. Hussain
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Mitchell H. Grayson
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital and The Ohio State University, Columbus, OH
| |
Collapse
|
6
|
Haase LM, Birk T, Poland CA, Holz O, Müller M, Bachand AM, Mundt KA. Cross-sectional Study of Workers Employed at a Copper Smelter-Effects of Long-term Exposures to Copper on Lung Function and Chronic Inflammation. J Occup Environ Med 2022; 64:e550-e558. [PMID: 35902212 PMCID: PMC9426729 DOI: 10.1097/jom.0000000000002610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The aim of the study was to assess the effect of exposure to copper-containing dust on lung function and inflammatory endpoints among workers of a German copper plant, effects rarely studied before. METHODS One hundred four copper-exposed smelter workers and 70 referent workers from the precious metal and lead facilities were included, with different metal exposures in both groups due to the different process materials. Body plethysmography, exhaled nitric oxide (FeNO) measurements, and blood sampling were conducted in all workers. Smoking status and the use of respiratory protective equipment were considered. In a subgroup of 40 nonsmoking volunteers (28 copper-exposed and 12 referents), sputum biomarkers were assessed. RESULTS Median lung function values of both copper-exposed and the referent groups were within reference ranges of "healthy" individuals, and statistical differences between the groups were mostly not evident. Similarly, differences in blood and sputum biomarkers were too small to be biologically relevant. CONCLUSION The results suggest the absence of the detectable effects of copper-containing dust exposure on lung function or chronic inflammation within the investigated cohort.
Collapse
|
7
|
Holz O, Müller M, Carstensen S, Olin AC, Hohlfeld JM. Inflammatory cytokines can be monitored in exhaled breath particles following segmental and inhalation endotoxin challenge in healthy volunteers. Sci Rep 2022; 12:5620. [PMID: 35379863 PMCID: PMC8979977 DOI: 10.1038/s41598-022-09399-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/22/2022] [Indexed: 11/10/2022] Open
Abstract
Particles in exhaled air (PEx) are generated when collapsed small airways reopen during breathing. PEx can be noninvasively collected by particle impaction, allowing the analysis of undiluted epithelial lining fluid (ELF). We used the endotoxin (LPS) challenge model to proof the concept that PEx can be used to monitor inflammatory changes in the lung. In this pilot study PEx were collected from ten healthy nonsmoking subjects using the PExA® instrument twice before and twice after a segmental LPS challenge (5, 21 h). Following a 4-week washout period, PEx were collected during the week before and 5 h after a whole lung LPS inhalation challenge. PEx biomarkers were compared to blood, bronchoalveolar lavage (BAL) following segmental challenge and induced sputum (ISP) following inhalation challenge. A clear LPS-induced inflammatory response was detectable in BAL fluid, ISP and blood. Albumin and surfactant-protein D were detectable in all PEx samples. While most baseline cytokines were close to or below the detection limit, the median (IQR) IL-6 and IL-8 concentrations in PEx increased significantly after segmental (0.04 (0.03; 0.06) fg/ng PEx; 0.10 (0.08; 0.17) fg/ng PEx) and inhalation LPS challenge (0.19 (0.15; 0.23) fg/ng PEx; 0.32 (0.23; 0.42) fg/ng PEx). Using a highly sensitive analysis platform, we were able to detect a cytokine response in PEx during the early phase of LPS-induced inflammation. This will broaden the spectrum of applications for this noninvasive method to monitor inflammatory processes in the lung, including its use in clinical trials for respiratory drug development.Trial registration: The study has been registered on 07.02.2017 at Clinicaltrials.gov (NCT03044327).
Collapse
Affiliation(s)
- Olaf Holz
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625, Hannover, Germany. .,German Center for Lung Research (BREATH), Hannover, Germany.
| | - Meike Müller
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625, Hannover, Germany
| | - Saskia Carstensen
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625, Hannover, Germany
| | - Anna-Carin Olin
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
| | - Jens M Hohlfeld
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625, Hannover, Germany.,German Center for Lung Research (BREATH), Hannover, Germany.,Department of Respiratory Medicine, Hannover Medical School (MHH), Hannover, Germany
| |
Collapse
|
8
|
Holz O, van Vorstenbosch R, Günther F, Schuchardt S, Trinkmann F, van Schooten FJ, Smolinska A, Hohlfeld J. Changes of breath volatile organic compounds in healthy volunteers following segmental and inhalation endotoxin challenge. J Breath Res 2022; 16. [PMID: 35366648 DOI: 10.1088/1752-7163/ac6359] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 04/01/2022] [Indexed: 11/11/2022]
Abstract
Background It is still unclear how airway inflammation affects the breath volatile organic compounds (VOC) profile in exhaled air. We therefore analyzed breath following well-defined pulmonary endotoxin (lipopolysaccharide, LPS) challenges. Methods Breath was collected from 10 healthy non-smoking subjects at eight time points before and after segmental and whole lung LPS inhalation challenge. Four Tenax-TA® adsorption tubes were simultaneously loaded from an aluminum reservoir cylinder and independently analyzed by two research groups using gas chromatography - mass spectrometry. Airway inflammation was assessed in bronchoalveolar lavage (BAL) and in sputum after segmental and inhaled LPS challenge, respectively. Results Segmental LPS challenge significantly increased the median (interquartile range, IQR) percentage of neutrophils in BAL from 3.0 (4.2) % to 64.0 (7.3) %. The inhalation challenge increased sputum neutrophils from 33.9 (26.8) % to 78.3 (13.5) %. We observed increases in breath aldehydes at both time points after segmental and inhaled LPS challenge. These results were confirmed by an independent laboratory. The longitudinal breath analysis also revealed distinct VOC patterns related to environmental exposures, clinical procedures, and to metabolic changes after food intake. Conclusions Changes in breath aldehydes suggest a relationship to LPS induced inflammation compatible with lipid peroxidation processes within the lung. Findings from our longitudinal data highlight the need for future studies to better consider the potential impact of the multiple VOCs from detergents, hygiene or lifestyle products a subject is continuously exposed to. We suspect that this very individual "owncloud" exposure is contributing to an increased variability of breath aldehydes, which might limit a use as inflammatory markers in daily clinical practice.
Collapse
Affiliation(s)
- Olaf Holz
- Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Feodor-Lynen-Str. 15, Hannover, 30625, GERMANY
| | | | - Frank Günther
- Bio- and Environmental Analytics, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Feodor-Lynen-Str. 15, Hannover, 30625, GERMANY
| | - Sven Schuchardt
- Bio- and Environmental Analytics, Fraunhofer-Institut fur Toxikologie und Experimentelle Medizin, Feodor-Lynen-Str. 15, Hannover, Niedersachsen, 30625, GERMANY
| | - Frederik Trinkmann
- Pneumology and Critical Care Medicine, Thoraxklinik at University Hospital Heidelberg, Röntgenstraße 1, Heidelberg, 69126, GERMANY
| | - Frederik Jan van Schooten
- Department of Toxicology, University of Maastricht, Universiteitssingel 50, THE NETHERLANDS, Maastricht, 6229 ER, NETHERLANDS
| | - Agnieszka Smolinska
- Toxicology Department, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, NETHERLANDS
| | - Jens Hohlfeld
- ITEM, Fraunhofer Institute for Toxicology and Experimental Medicine, Nikolai-Fuchs-Str. 1, Hannover, 30625, GERMANY
| |
Collapse
|
9
|
Hannon G, Prina-Mello A. Endotoxin contamination of engineered nanomaterials: Overcoming the hurdles associated with endotoxin testing. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1738. [PMID: 34254460 DOI: 10.1002/wnan.1738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 12/30/2022]
Abstract
Nanomaterials are highly susceptible to endotoxin contamination due their large surface-to-volume ratios and endotoxins propensity to associate readily to hydrophobic and cationic surfaces. Additionally, the stability of endotoxin ensures it cannot be removed efficiently through conventional sterilization techniques such as autoclaving and ionizing radiation. In recent times, the true significance of this hurdle has come to light with multiple reports from the United States Nanotechnology Characterization Laboratory, in particular, along with our own experiences of endotoxin testing from multiple Horizon 2020-funded projects which highlight the importance of this issue for the clinical translation of nanomaterials. Herein, we provide an overview on the topic of endotoxin contamination of nanomaterials intended for biomedical applications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Gary Hannon
- Nanomedicine and Molecular Imaging Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Dublin, Ireland.,Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Adriele Prina-Mello
- Nanomedicine and Molecular Imaging Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Dublin, Ireland.,Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
10
|
Carstensen S, Holz O, Hohlfeld JM, Müller M. Quantitative analysis of endotoxin-induced inflammation in human lung cells by Chipcytometry. Cytometry A 2021; 99:967-976. [PMID: 33860615 DOI: 10.1002/cyto.a.24352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/05/2021] [Accepted: 04/08/2021] [Indexed: 11/07/2022]
Abstract
Chipcytometry is a tool that uses iterative staining cycles with multiple antibodies for a detailed characterization of cells. Cell recognition is based on morphological features. Cells fixed on microfluidic chips can be stored and shipped enabling a centralized analysis, which is important for assessments in multi-center clinical trials. The method was initially implemented for the analysis of cells from peripheral blood. We adapted it to more heterogeneous human lung cells from bronchoalveolar lavage (BAL) fluid and induced sputum (IS). We aimed to assess the performance of Chipcytometry to detect and quantify the endotoxin induced inflammatory response in healthy subjects. BAL and IS samples of 10 healthy subjects were collected prior to and following segmental and inhaled endotoxin challenge. Samples were analyzed by Chipcytometry and were compared with flow cytometry, and differential cell count (DCC). Chipcytometry clearly detected the endotoxin induced inflammatory response which was characterized by a massive increase of neutrophils (BAL: 2.5% to 54.7%; IS: 40.5% to 71.1%) and monocytes (BAL: 7.7% to 24.7%; IS: 8.0% to 14.5%). While some differences between detection methods exist, the overall results were comparable. The ability of Chipcytometry to verify fluorescent signals with morphological features improved the precision of rare cell analysis such as of induced sputum lymphocytes. In conclusion, Chipcytometry enables the quantitative analysis of cells from BAL fluid and IS. Advantages over DCC and flow cytometry include the storage of cells on chips, the ability for re-analysis and the mapping of surface marker binding to morphological information. It therefore appears to be a promising method for use in clinical respiratory drug development.
Collapse
Affiliation(s)
- Saskia Carstensen
- Clinical Airway Research, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Olaf Holz
- Clinical Airway Research, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
- German Center for Lung Research (BREATH), Hannover, Germany
| | - Jens M Hohlfeld
- Clinical Airway Research, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
- German Center for Lung Research (BREATH), Hannover, Germany
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Meike Müller
- Clinical Airway Research, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| |
Collapse
|
11
|
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
|
12
|
Andreas S, Bothner U, de la Hoz A, Kloer I, Trampisch M, Alter P. A Post Hoc Holter ECG Analysis of Olodaterol and Formoterol in Moderate-to-Very-Severe COPD. Int J Chron Obstruct Pulmon Dis 2020; 15:1955-1965. [PMID: 32848381 PMCID: PMC7428408 DOI: 10.2147/copd.s246353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022] Open
Abstract
Background Patients with chronic obstructive pulmonary disease (COPD) are at risk of developing cardiac arrhythmias and elevated heart rate. A theoretical mechanistic association based on the interaction of long-acting β2-agonists (LABAs) with adrenoreceptors in the heart and vasculature is assumed as a potential class-related risk. Therefore, we performed a pooled analysis of Holter electrocardiogram (ECG) data from four 48-week, randomized, double-blind, placebo-controlled, parallel-group, Phase III clinical trials evaluating olodaterol (5 μg or 10 μg) or formoterol (12 µg) versus placebo. Methods We analyzed Holter ECG data from a representative subset of 775 patients with Global Initiative for Chronic Obstructive Lung Disease stage 2–4 COPD from four studies (1222.11–14) assessing olodaterol (5 μg and 10 μg) and formoterol (12 µg) versus placebo. Results No statistically significant (P>0.3) or clinically relevant differences in the shift from baseline of premature supraventricular or ventricular beats were observed among the active treatment and the placebo groups. Minor and transient differences were observed in the adjusted mean heart rate from baseline during treatment in all groups. There was a numerically small but statistically significant increase for formoterol at Week 24, olodaterol 5 μg at Weeks 12 and 40, and olodaterol 10 μg at Week 40 (all less than 3.0 beats per minute). Mean heart rates returned to a statistically non-significant change at Week 48 for all treatment groups. No increase in major adverse cardiovascular events was observed. Conclusion Treatment with olodaterol or formoterol is not associated with arrhythmias or a persistent increase in heart rate as assessed by Holter ECG in patients with COPD. Trial Registration ClinicalTrials.gov identifiers: NCT00782210 (1222.11); NCT00782509 (1222.12); NCT00793624 (1222.13); NCT00796653 (1222.14).
Collapse
Affiliation(s)
- Stefan Andreas
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,LungClinic Immenhausen, Immenhausen, Germany, Member of the German Center for Lung Research (DZL)
| | - Ulrich Bothner
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Alberto de la Hoz
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Isabel Kloer
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | | | - Peter Alter
- Department of Medicine, Pulmonary and Critical Care Medicine, Philipps University of Marburg (UMR), Marburg, Germany, Member of the German Center for Lung Research (DZL)
| |
Collapse
|
13
|
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
|
14
|
Viegas C, Caetano LA, Cox J, Korkalainen M, Haines SR, Dannemiller KC, Viegas S, Reponen T. The effects of waste sorting in environmental microbiome, THP-1 cell viability and inflammatory responses. ENVIRONMENTAL RESEARCH 2020; 185:109450. [PMID: 32244107 DOI: 10.1016/j.envres.2020.109450] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/28/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023]
Abstract
Workers in the waste sorting industry are exposed to diverse bioaerosols. Characterization of these bioaerosols is necessary to more accurately assess the health risks of exposure. The use of high-throughput DNA sequencing for improved analysis of microbial composition of bioaerosols, in combination with their in vitro study in relevant cell cultures, represents an important opportunity to find answers on the biological effects of bioaerosols. This study aimed to characterize by high-throughput sequencing the biodiversity present in complex aerosol mixtures retained in forklift air conditioning filters of a waste-sorting industry and its effects on cytotoxicity and secretion of proinflammatory cytokines in vitro using human macrophages derived from monocytic THP-1 cells. Seventeen filters from the filtration system from forklifts operating in one waste sorting facility and one control filter (similar filter without prior use) were analyzed using high-throughput sequencing and toxicological tests in vitro. A trend of positive correlation was seen between the number of bacterial and fungal OTUs (r = 0.47, p = 0.06). Seven filters (39%) exhibited low or moderate cytotoxicity (p < 0.05). The highest cytotoxic responses had a reduction in cell viability between 17 and 22%. Filter samples evoked proinflammatory responses, especially the production of TNFα. No significant correlation was found between fungal richness and inflammatory responses in vitro. The data obtained stress the need of thorough exposure assessment in waste-sorting industry and to take immunomodulatory properties into consideration for bioaerosols hazard characterization. The broad spectrum of microbial contamination detected in this study demonstrates that adequate monitoring of bioaerosol exposure is necessary to evaluate and minimize risks. The combined techniques can support the implementation of effective environmental monitoring programs of public and occupational health importance.
Collapse
Affiliation(s)
- C Viegas
- H&TRC- Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Portugal; NOVA National School of Public Health, Public Health Research Centre, Universidade NOVA de Lisboa, Portugal; Comprehensive Health Research Center (CHRC), Portugal.
| | - L A Caetano
- H&TRC- Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Portugal; Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 649-003, Lisbon, Portugal
| | - J Cox
- Department of Environmental Health, University of Cincinnati, P.O. Box 670056, Cincinnati, OH, 45242, USA
| | - M Korkalainen
- Finnish Institute for Health and Welfare (THL), Environmental Health, P.O. Box 95, FIN-70701, Kuopio, Finland
| | - S R Haines
- Department of Civil, Environmental, and Geodetic Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA; Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA; Environmental Science Graduate Program, The Ohio State University, Columbus, OH, USA
| | - K C Dannemiller
- Department of Civil, Environmental, and Geodetic Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA; Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA
| | - S Viegas
- H&TRC- Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Portugal; NOVA National School of Public Health, Public Health Research Centre, Universidade NOVA de Lisboa, Portugal; Comprehensive Health Research Center (CHRC), Portugal
| | - T Reponen
- Department of Environmental Health, University of Cincinnati, P.O. Box 670056, Cincinnati, OH, 45242, USA
| |
Collapse
|
15
|
Kern AL, Biller H, Klimeš F, Voskrebenzev A, Gutberlet M, Renne J, Müller M, Holz O, Wacker F, Hohlfeld JM, Vogel-Claussen J. Noninvasive Monitoring of the Response of Human Lungs to Low-Dose Lipopolysaccharide Inhalation Challenge Using MRI: A Feasibility Study. J Magn Reson Imaging 2019; 51:1669-1676. [PMID: 31729119 DOI: 10.1002/jmri.27000] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Development of antiinflammatory drugs for lung diseases demands novel methods for noninvasive assessment of inflammatory processes in the lung. PURPOSE To investigate the feasibility of hyperpolarized 129 Xe MRI, 1 H T1 time mapping, and dynamic contrast-enhanced (DCE) perfusion MRI for monitoring the response of human lungs to low-dose inhaled lipopolysaccharide (LPS) challenge compared to inflammatory cell counts from induced-sputum analysis. STUDY TYPE Prospective feasibility study. POPULATION Ten healthy volunteers underwent MRI before and 6 hours after inhaled LPS challenge with subsequent induced-sputum collection. FIELD STRENGTH/SEQUENCES 1.5T/hyperpolarized 129 Xe MRI: Interleaved multiecho imaging of dissolved and gas phase, ventilation imaging, dissolved-phase spectroscopy, and chemical shift saturation recovery spectroscopy. 1 H MRI: Inversion recovery fast low-angle shot imaging for T1 mapping, time-resolved angiography with stochastic trajectories for DCE MRI. ASSESSMENT Dissolved-phase ratios of 129 Xe in red blood cells (RBC), tissue/plasma (TP) and gas phase (GP), ventilation defect percentage, septal wall thickness, surface-to-volume ratio, capillary transit time, lineshape parameters in dissolved-phase spectroscopy, 1 H T1 time, blood volume, flow, and mean transit time were determined and compared to cell counts. STATISTICAL TESTS Wilcoxon signed-rank test, Pearson correlation. RESULTS The percentage of neutrophils in sputum was markedly increased after LPS inhalation compared to baseline, P = 0.002. The group median RBC-TP ratio was significantly reduced from 0.40 to 0.31, P = 0.004, and 1 H T1 was significantly elevated from 1157.6 msec to 1187.8 msec after LPS challenge, P = 0.027. DCE MRI exhibited no significant changes in blood volume, P = 0.64, flow, P = 0.17, and mean transit time, P = 0.11. DATA CONCLUSION Hyperpolarized 129 Xe dissolved-phase MRI and 1 H T1 mapping may provide biomarkers for noninvasive assessment of the response of human lungs to LPS inhalation. By its specificity to the alveolar region, hyperpolarized 129 Xe MRI together with 1 H T1 mapping adds value to sputum analysis. LEVEL OF EVIDENCE 1 Technical Efficacy Stage: 2 J. Magn. Reson. Imaging 2020;51:1669-1676.
Collapse
Affiliation(s)
- Agilo L Kern
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Heike Biller
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Filip Klimeš
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Andreas Voskrebenzev
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Marcel Gutberlet
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Julius Renne
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Meike Müller
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Olaf Holz
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Frank Wacker
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Jens M Hohlfeld
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany.,Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Jens Vogel-Claussen
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
16
|
Sood AK, Burbank AJ, Lawson M, Zhou H, Wells HB, Peden DB, Hernandez ML. Systemic inflammatory response to inhaled endotoxin does not correlate with airway response. Respir Res 2019; 20:252. [PMID: 31718667 PMCID: PMC6852787 DOI: 10.1186/s12931-019-1227-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/31/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Endotoxin is a component of particulate matter linked to respiratory disease. Our group has shown that experimental endotoxin inhalation challenge reproducibly triggers neutrophilic inflammation in the airways and in peripheral blood. Sputum induction is currently the only available method for assessing airway neutrophilia but is laborious and time-consuming. This analysis examined the correlation between systemic and airway inflammatory responses to endotoxin to determine if peripheral blood could serve as a surrogate marker for neutrophilic airway inflammation. METHODS We conducted a retrospective study of 124 inhaled endotoxin challenges conducted at our center using 20,000 endotoxin units (EU) of Clinical Center Reference Endotoxin (CCRE). Venipuncture and induced sputum samples were obtained at baseline and 6 hours after completion of endotoxin challenge. The relationship between change in sputum neutrophils (post-challenge - baseline) and change in peripheral blood neutrophils (post-challenge - baseline) was assessed using Spearman's correlation analyses. RESULTS Inhaled endotoxin induced a significant increase in mean sputum percent neutrophils and peripheral blood absolute neutrophil counts in healthy adults with or without mild asthma, but no significant correlation was found between airway and systemic neutrophilia (r = 0.13, p = 0.18). Stratification by degree of airway neutrophil response and by atopic or asthmatic status did not change the results. CONCLUSIONS Inhalation challenge with endotoxin safely and effectively induces airway neutrophilic inflammation in most individuals. Increases in endotoxin-induced peripheral blood neutrophils do not correlate well with airway responses and should not be used as a surrogate marker of airway inflammation.
Collapse
Affiliation(s)
- Amika K Sood
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, 104 Mason Farm Rd #CB7310, Chapel Hill, NC, 27599-7310, USA.,Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, University of North Carolina, 030 MacNider Hall CB#7231, 333 S Columbia St, Chapel Hill, NC, 27599, USA
| | - Allison J Burbank
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, 104 Mason Farm Rd #CB7310, Chapel Hill, NC, 27599-7310, USA. .,Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, University of North Carolina, 030 MacNider Hall CB#7231, 333 S Columbia St, Chapel Hill, NC, 27599, USA.
| | - Michael Lawson
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, 3101 McGavran-Greenberg Hall CB#7420, 135 Dauer Drive, Chapel Hill, NC, 27599, USA
| | - Haibo Zhou
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, 104 Mason Farm Rd #CB7310, Chapel Hill, NC, 27599-7310, USA.,Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, 3101 McGavran-Greenberg Hall CB#7420, 135 Dauer Drive, Chapel Hill, NC, 27599, USA
| | - Heather B Wells
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, 104 Mason Farm Rd #CB7310, Chapel Hill, NC, 27599-7310, USA
| | - David B Peden
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, 104 Mason Farm Rd #CB7310, Chapel Hill, NC, 27599-7310, USA.,Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, University of North Carolina, 030 MacNider Hall CB#7231, 333 S Columbia St, Chapel Hill, NC, 27599, USA
| | - Michelle L Hernandez
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, 104 Mason Farm Rd #CB7310, Chapel Hill, NC, 27599-7310, USA.,Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, University of North Carolina, 030 MacNider Hall CB#7231, 333 S Columbia St, Chapel Hill, NC, 27599, USA
| |
Collapse
|
17
|
Hadjigol S, Netto KG, Maltby S, Tay HL, Nguyen TH, Hansbro NG, Eyers F, Hansbro PM, Yang M, Foster PS. Lipopolysaccharide induces steroid-resistant exacerbations in a mouse model of allergic airway disease collectively through IL-13 and pulmonary macrophage activation. Clin Exp Allergy 2019; 50:82-94. [PMID: 31579973 DOI: 10.1111/cea.13505] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/26/2019] [Accepted: 09/15/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Acute exacerbations of asthma represent a major burden of disease and are often caused by respiratory infections. Viral infections are recognized as significant triggers of exacerbations; however, less is understood about the how microbial bioproducts such as the endotoxin (lipopolysaccharide (LPS)) trigger episodes. Indeed, increased levels of LPS have been linked to asthma onset, severity and steroid resistance. OBJECTIVE The goal of this study was to identify mechanisms underlying bacterial-induced exacerbations by employing LPS as a surrogate for infection. METHODS We developed a mouse model of LPS-induced exacerbation on the background of pre-existing type-2 allergic airway disease (AAD). RESULTS LPS-induced exacerbation was characterized by steroid-resistant airway hyperresponsiveness (AHR) and an exaggerated inflammatory response distinguished by increased numbers of infiltrating neutrophils/macrophages and elevated production of lung inflammatory cytokines, including TNFα, IFNγ, IL-27 and MCP-1. Expression of the type-2 associated inflammatory factors such as IL-5 and IL-13 were elevated in AAD but not altered by LPS exposure. Furthermore, AHR and airway inflammation were no longer suppressed by corticosteroid (dexamethasone) treatment after LPS exposure. Depletion of pulmonary macrophages by administration of 2-chloroadenosine into the lungs suppressed AHR and reduced IL-13, TNFα and IFNγ expression. Blocking IL-13 function, through either IL-13-deficiency or administration of specific blocking antibodies, also suppressed AHR and airway inflammation. CONCLUSIONS & CLINICAL RELEVANCE We present evidence that IL-13 and innate immune pathways (in particular pulmonary macrophages) contribute to LPS-induced exacerbation of pre-existing AAD and provide insight into the complex molecular processes potentially underlying microbial-induced exacerbations.
Collapse
Affiliation(s)
- Sara Hadjigol
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Keilah G Netto
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Steven Maltby
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Hock L Tay
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Thi H Nguyen
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Fiona Eyers
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
18
|
Winkler C, Hochdörfer T, Israelsson E, Hasselberg A, Cavallin A, Thörn K, Muthas D, Shojaee S, Lüer K, Müller M, Mjösberg J, Vaarala O, Hohlfeld J, Pardali K. Activation of group 2 innate lymphoid cells after allergen challenge in asthmatic patients. J Allergy Clin Immunol 2019; 144:61-69.e7. [PMID: 30731124 DOI: 10.1016/j.jaci.2019.01.027] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 01/13/2019] [Accepted: 01/22/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Group 2 innate lymphoid cells (ILC2s) are effective producers of IL-5 and IL-13 during allergic inflammation and bridge the innate and adaptive immune responses. ILC2 numbers are increased in asthmatic patients compared with healthy control subjects. Thus far, human data describing their phenotype during acute allergic inflammation in the lung are incomplete. OBJECTIVES This study aims to characterize and compare blood- and lung-derived ILC2s before and after segmental allergen challenge in patients with mild-to-moderate asthma with high blood eosinophil counts (≥300 cells/μL). METHODS ILC2s were isolated from blood and bronchoalveolar lavage (BAL) fluid before and after segmental allergen challenge. Cells were sorted by means of flow cytometry, cultured and analyzed for cytokine release or migration, and sequenced for RNA expression. RESULTS ILC2s were nearly absent in the alveolar space under baseline conditions, but numbers increased significantly after allergen challenge (P < .05), whereas at the same time, ILC2 numbers in blood were reduced (P < .05). Prostaglandin D2 and CXCL12 levels in BAL fluid correlated with decreased ILC2 numbers in blood (P = .004, respective P = .024). After allergen challenge, several genes promoting type 2 inflammation were expressed at greater levels in BAL fluid compared with blood ILC2s, whereas blood ILC2s remain unactivated. CONCLUSION ILC2s accumulate at the site of allergic inflammation and are recruited from the blood. Their transcriptional and functional activation pattern promotes type 2 inflammation.
Collapse
Affiliation(s)
- Carla Winkler
- Respiratory, Inflammation and Autoimmunity, Biotech IMED Unit, AstraZeneca, Gothenburg, Sweden.
| | - Thomas Hochdörfer
- Respiratory, Inflammation and Autoimmunity, Biotech IMED Unit, AstraZeneca, Gothenburg, Sweden
| | - Elisabeth Israelsson
- Respiratory, Inflammation and Autoimmunity, Biotech IMED Unit, AstraZeneca, Gothenburg, Sweden
| | - Annemarie Hasselberg
- Respiratory, Inflammation and Autoimmunity, Biotech IMED Unit, AstraZeneca, Gothenburg, Sweden
| | - Anders Cavallin
- Respiratory, Inflammation and Autoimmunity, Biotech IMED Unit, AstraZeneca, Gothenburg, Sweden
| | - Kristofer Thörn
- Respiratory, Inflammation and Autoimmunity, Biotech IMED Unit, AstraZeneca, Gothenburg, Sweden
| | - Daniel Muthas
- Respiratory, Inflammation and Autoimmunity, Biotech IMED Unit, AstraZeneca, Gothenburg, Sweden
| | - Shervin Shojaee
- Discovery Science, Biotech IMED Unit, AstraZeneca, Gothenburg, Sweden
| | - Katrin Lüer
- Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Meike Müller
- Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Jenny Mjösberg
- Center for Infectious Diseases, Karolinska Institute, Stockholm, Sweden; Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Outi Vaarala
- Respiratory, Inflammation and Autoimmunity, Biotech IMED Unit, AstraZeneca, Gothenburg, Sweden
| | - Jens Hohlfeld
- Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany; Member of the German Center for Lung Research (BREATH), Hannover, Germany; Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Katerina Pardali
- Respiratory, Inflammation and Autoimmunity, Biotech IMED Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
19
|
Mulvanny A, Jackson N, Pattwell C, Wolosianka S, Southworth T, Singh D. The dose response of inhaled LPS challenge in healthy subjects. EUR J INFLAMM 2018. [DOI: 10.1177/2058739218784820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Lipopolysaccharide (LPS) inhalation causes neutrophilic airway inflammation. We used LPS produced to Good Manufacturing Practice (GMP) standards to characterise the dose response. A total of 15 healthy non-smoking subjects inhaled 5-, 15- and 50-µg LPS. Whole blood cell counts and serum C-reactive protein (CRP) were measured at baseline and up to 24 h post challenge. Sputum was induced at baseline and 6 h post challenge for cell counts and quantification of myeloperoxidase (MPO), interleukin (IL)-1β, IL-6, IL-8 and tumour necrosis factor α (TNF-α) in supernatants. LPS inhalation was well tolerated. Blood neutrophil counts increased at 6 h post LPS with all doses. Serum CRP significantly increased with 15- and 50-µg LPS. All LPS doses significantly increased sputum neutrophil percentage ( P < 0.001). IL-1β, IL-6 and TNF-α were significantly increased in sputum supernatant following challenge with 50-µg LPS, there was no change in MPO or IL-8. The 50-µg LPS was well tolerated and produced a robust inflammatory response. This study supports the use of 50-µg GMP-grade LPS as a suitable challenge agent in clinical trials.
Collapse
Affiliation(s)
- Alex Mulvanny
- The Medicines Evaluation Unit, Manchester, UK
- The University of Manchester; Division of Infection, Immunity & Respiratory Medicine; Manchester Academic Health Science Centre; Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | | | | | | | - Thomas Southworth
- The Medicines Evaluation Unit, Manchester, UK
- The University of Manchester; Division of Infection, Immunity & Respiratory Medicine; Manchester Academic Health Science Centre; Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Dave Singh
- The Medicines Evaluation Unit, Manchester, UK
- The University of Manchester; Division of Infection, Immunity & Respiratory Medicine; Manchester Academic Health Science Centre; Manchester University NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
20
|
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
|
21
|
Viegas S, Caetano LA, Korkalainen M, Faria T, Pacífico C, Carolino E, Quintal Gomes A, Viegas C. Cytotoxic and Inflammatory Potential of Air Samples from Occupational Settings with Exposure to Organic Dust. TOXICS 2017; 5:E8. [PMID: 29051440 PMCID: PMC5606674 DOI: 10.3390/toxics5010008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/13/2017] [Accepted: 02/21/2017] [Indexed: 12/27/2022]
Abstract
Organic dust and related microbial exposures are the main inducers of several respiratory symptoms. Occupational exposure to organic dust is very common and has been reported in diverse settings. In vitro tests using relevant cell cultures can be very useful for characterizing the toxicity of complex mixtures present in the air of occupational environments such as organic dust. In this study, the cell viability and the inflammatory response, as measured by the production of pro-inflammatory cytokines tumor necrosis factor-α (TNFα) and interleukin-1 β (IL-1β), were determined in human macrophages derived from THP-1 monocytic cells. These cells were exposed to air samples from five occupational settings known to possess high levels of contamination of organic dust: poultry and swine feed industries, waste sorting, poultry production and slaughterhouses. Additionally, fungi and particle contamination of those settings was studied to better characterize the organic dust composition. All air samples collected from the assessed workplaces caused both cytotoxic and pro-inflammatory effects. The highest responses were observed in the feed industry, particularly in swine feed production. This study emphasizes the importance of measuring the organic dust/mixture effects in occupational settings and suggests that differences in the organic dust content may result in differences in health effects for exposed workers.
Collapse
Affiliation(s)
- Susana Viegas
- Environment and Health Research Group, Escola Superior de Tecnologia da Saúde de Lisboa, ESTeSL, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal.
- Centro de Investigação em Saúde Pública, Escola Nacional de Saúde Pública, Universidade NOVA de Lisboa, 1600-560 Lisbon, Portugal.
| | - Liliana Aranha Caetano
- Environment and Health Research Group, Escola Superior de Tecnologia da Saúde de Lisboa, ESTeSL, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal.
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 649-003 Lisbon, Portugal.
| | - Merja Korkalainen
- National Institute for Health and Welfare (THL), Department of Health Security, Chemicals and Health Unit, P.O. Box 95, FIN-70701 Kuopio, Finland.
| | - Tiago Faria
- Environment and Health Research Group, Escola Superior de Tecnologia da Saúde de Lisboa, ESTeSL, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal.
| | - Cátia Pacífico
- Environment and Health Research Group, Escola Superior de Tecnologia da Saúde de Lisboa, ESTeSL, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal.
| | - Elisabete Carolino
- Environment and Health Research Group, Escola Superior de Tecnologia da Saúde de Lisboa, ESTeSL, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal.
| | - Anita Quintal Gomes
- Environment and Health Research Group, Escola Superior de Tecnologia da Saúde de Lisboa, ESTeSL, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal.
- Institute of Molecular Medicine, Faculty of Medicine. University of Lisbon, 649-028 Lisbon, Portugal.
| | - Carla Viegas
- Environment and Health Research Group, Escola Superior de Tecnologia da Saúde de Lisboa, ESTeSL, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal.
- Centro de Investigação em Saúde Pública, Escola Nacional de Saúde Pública, Universidade NOVA de Lisboa, 1600-560 Lisbon, Portugal.
| |
Collapse
|
22
|
Doyen V, Pilcer G, Dinh PHD, Corazza F, Bernard A, Bergmann P, Lefevre N, Amighi K, Michel O. Inflammation induced by inhaled lipopolysaccharide depends on particle size in healthy volunteers. Br J Clin Pharmacol 2016; 82:1371-1381. [PMID: 27331367 DOI: 10.1111/bcp.13052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 05/24/2016] [Accepted: 06/20/2016] [Indexed: 12/29/2022] Open
Abstract
AIMS In drug development, the anti-inflammatory properties of new molecules in the lung are currently tested using the inhaled lipopolysaccharide (LPS) model. The total and regional lung bioavailability of inhaled particles depends significantly on their size. The objective of the present study was to compare inflammatory responses in healthy volunteers after the inhalation of LPS of varying droplet size. METHODS Three nebulizers were characterized by different droplet size distributions [mean mass median aerodynamic diameters: Microcirrus (2.0 μm), MB2 (3.2 μm) and Pari (7.9 μm)]. Participants inhaled three boluses of a 20 μg (technetium 99 m-labelled) solution of LPS, randomly delivered by each nebulizer. We measured the lung deposition of the nebulized LPS by gamma-scintigraphy, while blood and sputum biomarkers were evaluated before and after challenges. RESULTS MB2 and Pari achieved greater lung deposition than Microcirrus [171.5 (±72.9) and 217.6 (±97.8) counts pixel-1 , respectively, vs. 67.9 (±20.6) counts pixel-1 ; P < 0.01]. MB2 and Pari caused higher levels of blood C-reactive protein and more total cells and neutrophils in sputum compared with Microcirrus (P < 0.05). C-reactive protein levels correlated positively with lung deposition (P < 0.01). CONCLUSIONS Inhalation of large droplets of LPS gave rise to greater lung deposition and induced a more pronounced systemic and bronchial inflammatory response than small droplets. The systemic inflammatory response correlated with lung deposition. NCT01081392.
Collapse
Affiliation(s)
- Virginie Doyen
- Clinic of Immuno-allergology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium.
| | - Gabrielle Pilcer
- Laboratory of Pharmaceutics and Biopharmaceutics, Faculty of Pharmacy, ULB, Brussels, Belgium
| | - Phong Huy Duc Dinh
- Clinic of Immuno-allergology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | - Francis Corazza
- Laboratory of Immunology, CHU Brugmann, ULB, Brussels, Belgium
| | - Alfred Bernard
- Unit of Toxicology and Applied Pharmacology, Faculty of Medicine, ULB, Brussels, Belgium
| | - Pierre Bergmann
- Nuclear medicine department, CHU Brugmann, ULB, Brussels, Belgium
| | - Nicolas Lefevre
- Clinic of Pulmonology, Allergology and Cystic Fibrosis, Hôpital Universitaire des Enfants Reine Fabiola, ULB, Brussels, Belgium
| | - Karim Amighi
- Laboratory of Pharmaceutics and Biopharmaceutics, Faculty of Pharmacy, ULB, Brussels, Belgium
| | - Olivier Michel
- Clinic of Immuno-allergology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
23
|
Kobernick AK, Peden DB, Zhou H, Zhou Q, Dillon MA, Alexis NE. Reproducibility of the inflammatory response to inhaled endotoxin in healthy volunteers. J Allergy Clin Immunol 2016; 138:1205-1207. [PMID: 27297997 DOI: 10.1016/j.jaci.2016.04.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/24/2016] [Accepted: 04/06/2016] [Indexed: 11/20/2022]
Affiliation(s)
- Aaron K Kobernick
- Center for Environmental Medicine, Asthma and Lung Biology, Division of Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC.
| | - David B Peden
- Center for Environmental Medicine, Asthma and Lung Biology, Division of Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Haibo Zhou
- Division of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC
| | - Qinging Zhou
- Division of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC
| | | | - Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, Division of Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC
| |
Collapse
|
24
|
Motwani MP, Flint JD, De Maeyer RP, Fullerton JN, Smith AM, Marks DJ, Gilroy DW. Novel translational model of resolving inflammation triggered by UV-killed E. coli. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016; 2:154-65. [PMID: 27499924 PMCID: PMC4958736 DOI: 10.1002/cjp2.43] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/05/2016] [Indexed: 11/23/2022]
Abstract
Whilst numerous studies investigating the aetiology of inflammatory diseases have been performed in rodents, the applicability of these data to human pathophysiology is frequently debated. Regardless of the strengths and weaknesses of rodent models in biomedical research, there is a need to develop models of experimental inflammation in humans. Here, we describe a self‐resolving acute inflammatory response triggered by the intradermal injection of UV‐killed Escherichia coli into the forearm of healthy volunteers. Cells and exudates were harvested from onset to resolution by applying negative pressure over the inflamed site. Onset was characterized by high blood flow, neutrophilia and peak levels of pro‐inflammatory cytokines, whilst resolution showed a decline in blood blow, reduction in neutrophils, increase in monocytes/macrophages and waning of classic pro‐inflammatory cytokine levels. An anti‐inflammatory effect, defined as suppression of onset phase events, was demonstrated by administering naproxen, a conventional non‐steroidal anti‐inflammatory drug. In summary, this model of resolving acute inflammation is minimally invasive, highly tractable and allows simultaneous investigation of the vascular response, cellular trafficking and chemical mediator profile of onset and resolution phases of acute inflammation in humans. It can serve as a translational platform to provide mechanistic insights and to test the clinical efficacy of novel anti‐inflammatory and pro‐resolving drugs, and also as a tool in patients to explore inherent defects in resolution pathways.
Collapse
Affiliation(s)
- Madhur P Motwani
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| | - Julia D Flint
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| | - Roel Ph De Maeyer
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| | - James N Fullerton
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| | - Andrew M Smith
- Microbial Diseases, Eastman Dental Institute, University College London London WC1X 8LD UK
| | - Daniel Jb Marks
- Centre for Molecular Medicine, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| | - Derek W Gilroy
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| |
Collapse
|
25
|
Bennett WD, Alexis NE, Almond M, Herbst M, Zeman KL, Peden DB. Effect of inhaled endotoxin on mucociliary clearance and airway inflammation in mild smokers and nonsmokers. J Aerosol Med Pulm Drug Deliv 2015; 27:459-65. [PMID: 24568613 DOI: 10.1089/jamp.2013.1089] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND In healthy nonsmokers, inhaled endotoxin [lipopolysaccharide (LPS)] challenge induces airway neutrophilia and modifies innate immune responses, but the effect on mucociliary clearance (MCC), a key host defense response, is unknown. Although smokers are chronically exposed to LPS through inhaled tobacco smoke, the acute effect of inhaled LPS on both MCC and airway inflammation is also unknown. The purpose of this study was to determine the effect of inhaled LPS on MCC in nonsmokers and mild smokers with normal pulmonary function. METHODS We performed an open-label inhalational challenge with 20,000 endotoxin units in healthy adult nonsmokers (n=18) and young adult, mild smokers (n=12). At 4 hr post LPS challenge, we measured MCC over a period of 2 hr, followed by sputum induction to assess markers of airway inflammation. RESULTS No significant changes in spirometry occurred in either group following LPS challenge. Following LPS, MCC was significantly (p<0.05) slowed in nonsmokers, but not in smokers [MCC=10±9% (challenge) vs. 15±8% (baseline), MCC=14±9% (challenge) vs. 16±10% (baseline), respectively]. Both groups showed a significant (p<0.05) increase in sputum neutrophils 6 hr post LPS challenge versus baseline. Although there was no correlation between the increased neutrophilia and depressed MCC post LPS in the nonsmokers, baseline neutrophil concentration predicted the LPS-induced decrease in MCC in the nonsmokers, i.e., lower baseline neutrophil concentration was associated with greater depression in MCC with LPS challenge (p<0.05). CONCLUSIONS These data show that a mild exposure to endotoxin acutely slows MCC in healthy nonsmokers. MCC in mild smokers is unaffected by mild endotoxin challenge, likely due to preexisting effects of cigarette smoke on their airway epithelium.
Collapse
Affiliation(s)
- William D Bennett
- 1 Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina (UNC) at Chapel Hill , NC
| | | | | | | | | | | |
Collapse
|
26
|
Holz O, Biller H, Mueller M, Kane K, Rosano M, Hanrahan J, Hava DL, Hohlfeld JM. Efficacy and safety of inhaled calcium lactate PUR118 in the ozone challenge model--a clinical trial. BMC Pharmacol Toxicol 2015; 16:21. [PMID: 26265479 PMCID: PMC4533952 DOI: 10.1186/s40360-015-0021-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 07/24/2015] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The ozone challenge model can be used to assess the efficacy of anti-inflammatory compounds in early phases of clinical drug development. PUR118, a calcium salt based formulation engineered in the iSPERSE(TM) dry powder delivery technology, is a novel anti-inflammatory drug for COPD. Here we evaluated the efficacy and safety of three doses of PUR118 in attenuating ozone-induced airway inflammation in healthy volunteers. METHODS In a single-blind, phase 1B proof of concept study, 24 subjects were enrolled to sequentially receive three doses of PUR118 (5.5 mg, n = 18; 11.0 mg, n = 18; 2.8 mg, n = 16). Each dose was inhaled 3 times (1, 13, 25 h, preceded by 2 puffs salbutamol) before the ozone exposure (250 ppb, 3 h intermittent exercise). Sputum was induced 3 h after the end of exposure. RESULTS Sputum neutrophils, sputum CD14+ cells, as well as concentrations of IL1B, IL6, IL8, MMP9, and TNFA in sputum supernatant significantly increased after ozone exposure (n = 24). The percentage of sputum neutrophils (n = 12 who completed all treatments) did not change following treatment with different doses of PUR118. The high dose treatment group (n = 16) showed a decrease in the percentage and number of sputum macrophages (p ≤ 0.05) as well as a decrease in blood neutrophils (p = 0.04), and an increase in blood CD14 + cells (p = 0.04) compared to baseline. All dosages of PUR118 were safe and well tolerated. CONCLUSION Ozone challenge resulted in the expected and significant increase of sputum inflammatory parameters. Treatment with multiple rising doses of PUR118 was safe and three applications within 25 h prior to the ozone challenge had small effects on ozone-induced airway inflammation. TRIAL REGISTRATION ClinicalTrials.gov: NCT01690949. Registered 12 September 2012.
Collapse
Affiliation(s)
- Olaf Holz
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, 30625, Hannover, Germany.
- Member of the German Center for Lung Research (BREATH), Hannover, Germany.
| | - H Biller
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, 30625, Hannover, Germany
| | - M Mueller
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, 30625, Hannover, Germany
| | - K Kane
- Pulmatrix, Inc., Lexington, MA, USA
| | - M Rosano
- Pulmatrix, Inc., Lexington, MA, USA
| | | | - D L Hava
- Pulmatrix, Inc., Lexington, MA, USA
| | - J M Hohlfeld
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, 30625, Hannover, Germany
- Member of the German Center for Lung Research (BREATH), Hannover, Germany
| |
Collapse
|
27
|
Krug N, Hohlfeld JM, Kirsten AM, Kornmann O, Beeh KM, Kappeler D, Korn S, Ignatenko S, Timmer W, Rogon C, Zeitvogel J, Zhang N, Bille J, Homburg U, Turowska A, Bachert C, Werfel T, Buhl R, Renz J, Garn H, Renz H. Allergen-induced asthmatic responses modified by a GATA3-specific DNAzyme. N Engl J Med 2015; 372:1987-95. [PMID: 25981191 DOI: 10.1056/nejmoa1411776] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The most prevalent phenotype of asthma is characterized by eosinophil-dominated inflammation that is driven by a type 2 helper T cell (Th2). Therapeutic targeting of GATA3, an important transcription factor of the Th2 pathway, may be beneficial. We evaluated the safety and efficacy of SB010, a novel DNA enzyme (DNAzyme) that is able to cleave and inactivate GATA3 messenger RNA (mRNA). METHODS We conducted a randomized, double-blind, placebo-controlled, multicenter clinical trial of SB010 involving patients who had allergic asthma with sputum eosinophilia and who also had biphasic early and late asthmatic responses after laboratory-based allergen provocation. A total of 40 patients could be evaluated; 21 were assigned to receive 10 mg of SB010, and 19 were assigned to receive placebo, with each study drug administered by means of inhalation once daily for 28 days. An allergen challenge was performed before and after the 28-day period. The primary end point was the late asthmatic response as quantified by the change in the area under the curve (AUC) for forced expiratory volume in 1 second (FEV1). RESULTS After 28 days, SB010 attenuated the mean late asthmatic response by 34%, as compared with the baseline response, according to the AUC for FEV1, whereas placebo was associated with a 1% increase in the AUC for FEV1 (P=0.02). The early asthmatic response with SB010 was attenuated by 11% as measured by the AUC for FEV1, whereas the early response with placebo was increased by 10% (P=0.03). Inhibition of the late asthmatic response by SB010 was associated with attenuation of allergen-induced sputum eosinophilia and with lower levels of tryptase in sputum and lower plasma levels of interleukin-5. Allergen-induced levels of fractional exhaled nitric oxide and airway hyperresponsiveness to methacholine were not affected by either SB010 or placebo. CONCLUSIONS Treatment with SB010 significantly attenuated both late and early asthmatic responses after allergen provocation in patients with allergic asthma. Biomarker analysis showed an attenuation of Th2-regulated inflammatory responses. (Funded by Sterna Biologicals and the German Federal Ministry of Education and Research; ClinicalTrials.gov number, NCT01743768.).
Collapse
Affiliation(s)
- Norbert Krug
- From the Fraunhofer Institute for Toxicology and Experimental Medicine (N.K., J.M.H.) and the Department of Dermatology and Allergy, Hannover Medical School (J.Z., T.W.), Hannover, Pulmonary Research Institute at Lung Clinic Grosshansdorf, Grosshansdorf (A.-M.K.), Institut für klinische Forschung Pneumologie, Clinical Research Center Respiratory Medicine, Frankfurt (O.K.), Insaf Respiratory Research Institute, Wiesbaden (K.M.B.), Inamed, Gauting (D.K., W.T.), Pulmonary Department, Medical Clinic, University Hospital Mainz, Mainz (S.K., R.B.), Charité Research Organization, Berlin (S.I.), FGK Clinical Research, Munich (C.R.), Sterna Biologicals (J.B., U.H., A.T., J.R.) and Institute of Laboratory Medicine, Philipps University Marburg, a member of Universities Giessen and Marburg Lung Center (H.G., H.R.), Marburg - all in Germany; Upper Airways Research Laboratory, University Hospital Ghent, Ghent, Belgium (N.Z., C.B.); and the Division of Ear, Nose, and Throat Diseases, Clintec, Karolinska Institute, Stockholm (N.Z., C.B.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Zielen S, Trischler J, Schubert R. Lipopolysaccharide challenge: immunological effects and safety in humans. Expert Rev Clin Immunol 2015; 11:409-18. [DOI: 10.1586/1744666x.2015.1012158] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
29
|
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
|
30
|
Disease models of chronic inflammatory airway disease: applications and requirements for clinical trials. Curr Opin Pulm Med 2014; 20:37-45. [PMID: 24231465 DOI: 10.1097/mcp.0000000000000013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review will discuss methodologies and applicability of key inflammatory models of respiratory disease in proof of concept or proof of efficacy clinical studies. In close relationship with these models, induced sputum and inflammatory cell counts will be addressed for phenotype-directed drug development. Additionally, important regulatory aspects regarding noninvestigational medicinal products used in bronchial challenges or clinical inflammatory models of respiratory disease will be highlighted. RECENT FINDINGS The recognition of an ever increasing number of phenotypes and endotypes within conditions such as asthma and chronic obstructive pulmonary disease urges phenotyping of study populations already in early clinical phases of drug development. Apart from the choice of a relevant disease model, recent studies show that especially targeted therapies need to be tested in well defined disease subsets for adequate efficacy assessment. Noninvasive biomarkers, especially sputum inflammatory cell counts, aid phenotyping and are useful outcome measures for novel, targeted therapies. SUMMARY Disease phenotyping becomes increasingly important for efficient and cost-effective drug development and subsequent disease management. Inflammatory models of respiratory disease combined with sputum biomarkers are important tools in this approach.
Collapse
|
31
|
Michel O, Doyen V, Leroy B, Bopp B, Dinh DHP, Corazza F, Wattiez R. Expression of calgranulin A/B heterodimer after acute inhalation of endotoxin: proteomic approach and validation. BMC Pulm Med 2013; 13:65. [PMID: 24237763 PMCID: PMC4225611 DOI: 10.1186/1471-2466-13-65] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 11/11/2013] [Indexed: 12/04/2022] Open
Abstract
Background The acute inhalation of endotoxin mimicks several aspects of the inflammation related to chronic obstructive pulmonary disease (COPD). The aim of the current study was to identify and to validate biomarkers of endotoxin-induced airways’ inflammation. Methods The cellular count in the induced-sputum, was measured before and after an inhalation of 20 mcg endotoxin, in 8 healthy volunteers. A proteomic analysis was applied to identify the more relevant proteins expression, before measurement by ELISA. The amplitude and the repeatability of the markers were evaluated among another population of 12 healthy subjects. Results There was a significant rise of viable cells (p <0.01), macrophages (p <0.05), and neutrophils (p <0.02) 24 hours after endotoxin inhalation, and of neutrophils (p <0.02) and lymphocytes (p <0.05) at 6 hours. Among the highest amplitude responses, the two dimensional electrophoretic separation shown proteolytic activity and overexpression of protein spots. By MALDI-TOF mass spectrometry, the last were identified as calgranulin A and B. The expression of the bioactive A/B heterodimeric complex was confirmed by ELISA both in the sputum (p <0.01) and at the blood level (p <0.01). The intra-subject repeatability of the sputum calgranulin A/B was highly significant (p <0.0001). Conclusion In healthy subjects, the inhalation of endotoxin induced expression of sputum calgranulin A/B that could be a biomarker of the endotoxin response/exposure.
Collapse
Affiliation(s)
- Olivier Michel
- Clinic of Immuno-allergology, CHU Brugmann (ULB), pl Van Gehuchten 4, B-1020 Brussels, Belgium.
| | | | | | | | | | | | | |
Collapse
|