1
|
Abusara OH, Hammad AM, Debas R, Al-Shalabi E, Waleed M, Scott Hall F. The inflammation and oxidative status of rat lung tissue following smoke/vapor exposure via E-cigarette, cigarette, and waterpipe. Gene 2025; 935:149066. [PMID: 39491601 DOI: 10.1016/j.gene.2024.149066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Tobacco smoking is a major worldwide health issue that contributes to millions of deaths annually. Electronic cigarettes (E-cigarettes) are also harmful. Smoke/vapor from E-cigarettes and tobacco products consists of free radicals and other toxic substances. Tissue damage in smokers, such as lungs, is highly observed and is linked to oxidative damage and inflammation. METHODS The inflammation and oxidative status of rat lung tissues was examined following whole-body smoke/vapor exposure via E-cigarette, cigarette, and waterpipe for 2 h daily, 5 days per week for 8 weeks. RESULTS Lung tissue damage was higher in cigarettes and waterpipe groups compared to the E-cigarette group. Collectively, there was a significant increase (p < 0.05) in the mRNA expression of pro-inflammatory mediators (TNF-α, NF-κB, IL-1β) with the exception of IL-1β in the E-cigarettes group. As for the anti-inflammatory mediators (Nrf2 and IL-10), a significant reduction (p < 0.05) of mRNA expression was observed with the exception of Nrf2 in the E-cigarette group. As for IL-6, there was a significant increase in its mRNA expression (p < 0.05) in the cigarette and waterpipe groups. There was also a significant decrease (p < 0.05) in the antioxidant activity of all antioxidants tested (GPx, SOD, and CAT) in all groups with the exception of SOD in the cigarette group. CONCLUSION Smoke/vapor administered via E-cigarette, cigarette, and waterpipe elicits inflammation and oxidative stress in rat lungs that is accompanied by histopathological changes.
Collapse
Affiliation(s)
- Osama H Abusara
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Alaa M Hammad
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan.
| | - Rasha Debas
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Eveen Al-Shalabi
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Mohammed Waleed
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
2
|
Kim BK, Yang WJ, Seong YS, Choi YJ, Park HJ, Byun MK, Chang YS, Cho JH, Kim CY. Comparative Assessment of Acute Pulmonary Effects Induced by Heat-Not-Burn Tobacco Aerosol Inhalation in a Murine Model. Int J Mol Sci 2025; 26:1135. [PMID: 39940903 PMCID: PMC11817633 DOI: 10.3390/ijms26031135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Tobacco smoking remains a major global health concern, causing preventable deaths and economic strain. Although new tobacco products such as heat-not-burn (HnB) are safer alternatives to traditional cigarettes, research on their associated risks remains limited. This study aimed to investigate the effects of HnB smoke exposure on the lungs compared to those of traditional cigarettes and the combined use of HnB and cigarettes using experiments with a mouse model. We quantitatively analyzed changes in the levels of 92 blood plasma proteins using the proximity extension assay method and observed significant changes in their levels in mice exposed to different smoke conditions; specifically, the levels of certain proteins, including Ccl20, Cxcl1, and Pdgfb, increased in the HnB smoke-exposed group, suggesting activation of nicotine pathways. Comparative analysis with traditional cigarette smoke-exposed mice further highlighted similarities and differences in their protein expression profiles. This study contributes to an improved understanding of the biological mechanisms underlying the harmful effects of alternative nicotine delivery systems and identifies potential biomarkers associated with the harmful effects of HnB smoke exposure. However, the precise impact of nicotine on the immune system may be influenced by various factors, necessitating further research.
Collapse
Affiliation(s)
- Beong Ki Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Dongguk University College of Medicine, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea;
| | - Won Jin Yang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Ye Seul Seong
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul 06273, Republic of Korea;
| | - Yong Jun Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Hye Jung Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Min Kwang Byun
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Yoon Soo Chang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Jae Hwa Cho
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Chi Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University College of Medicine, Korea University Ansan Hospital, Ansan 15355, Republic of Korea
| |
Collapse
|
3
|
Ghazi S, Song MA, El-Hellani A. A scoping review of the toxicity and health impact of IQOS. Tob Induc Dis 2024; 22:TID-22-97. [PMID: 38832049 PMCID: PMC11145630 DOI: 10.18332/tid/188867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/05/2024] Open
Abstract
This work aims to summarize the current evidence on the toxicity and health impact of IQOS, taking into consideration the data source. On 1 June 2022, we searched PubMed, Web of Science, and Scopus databases using the terms: 'heated tobacco product', 'heat-not-burn', 'IQOS', and 'tobacco heating system'. The search was time-restricted to update a previous search conducted on 8 November 2021, on IQOS data from 2010-2021. The data source [independent, Philip Morris International (PMI), or other manufacturers] was retrieved from relevant sections of each publication. Publications were categorized into two general categories: 1) Toxicity assessments included in vitro, in vivo, and systems toxicology studies; and 2) The impact on human health included clinical studies assessing biomarkers of exposure and biomarkers of health effects. Generally, independent studies used classical in vitro and in vivo approaches, but PMI studies combined these with modeling of gene expression (i.e. systems toxicology). Toxicity assessment and health impact studies covered pulmonary, cardiovascular, and other systemic toxicity. PMI studies overall showed reduced toxicity and health risks of IQOS compared to cigarettes, but independent data did not always conform with this conclusion. This review highlights some discrepancies in IQOS risk assessment regarding methods, depth, and breadth of data collection, as well as conclusions based on the data source.
Collapse
Affiliation(s)
- Sarah Ghazi
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus OH, United States
| | - Min-Ae Song
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus OH, United States
- Center for Tobacco Research, The Ohio State University Comprehensive Cancer Center, Columbus OH, United States
| | - Ahmad El-Hellani
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus OH, United States
- Center for Tobacco Research, The Ohio State University Comprehensive Cancer Center, Columbus OH, United States
| |
Collapse
|
4
|
Chandy M, Hill T, Jimenez-Tellez N, Wu JC, Sarles SE, Hensel E, Wang Q, Rahman I, Conklin DJ. Addressing Cardiovascular Toxicity Risk of Electronic Nicotine Delivery Systems in the Twenty-First Century: "What Are the Tools Needed for the Job?" and "Do We Have Them?". Cardiovasc Toxicol 2024; 24:435-471. [PMID: 38555547 PMCID: PMC11485265 DOI: 10.1007/s12012-024-09850-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/19/2024] [Indexed: 04/02/2024]
Abstract
Cigarette smoking is positively and robustly associated with cardiovascular disease (CVD), including hypertension, atherosclerosis, cardiac arrhythmias, stroke, thromboembolism, myocardial infarctions, and heart failure. However, after more than a decade of ENDS presence in the U.S. marketplace, uncertainty persists regarding the long-term health consequences of ENDS use for CVD. New approach methods (NAMs) in the field of toxicology are being developed to enhance rapid prediction of human health hazards. Recent technical advances can now consider impact of biological factors such as sex and race/ethnicity, permitting application of NAMs findings to health equity and environmental justice issues. This has been the case for hazard assessments of drugs and environmental chemicals in areas such as cardiovascular, respiratory, and developmental toxicity. Despite these advances, a shortage of widely accepted methodologies to predict the impact of ENDS use on human health slows the application of regulatory oversight and the protection of public health. Minimizing the time between the emergence of risk (e.g., ENDS use) and the administration of well-founded regulatory policy requires thoughtful consideration of the currently available sources of data, their applicability to the prediction of health outcomes, and whether these available data streams are enough to support an actionable decision. This challenge forms the basis of this white paper on how best to reveal potential toxicities of ENDS use in the human cardiovascular system-a primary target of conventional tobacco smoking. We identify current approaches used to evaluate the impacts of tobacco on cardiovascular health, in particular emerging techniques that replace, reduce, and refine slower and more costly animal models with NAMs platforms that can be applied to tobacco regulatory science. The limitations of these emerging platforms are addressed, and systems biology approaches to close the knowledge gap between traditional models and NAMs are proposed. It is hoped that these suggestions and their adoption within the greater scientific community will result in fresh data streams that will support and enhance the scientific evaluation and subsequent decision-making of tobacco regulatory agencies worldwide.
Collapse
Affiliation(s)
- Mark Chandy
- Robarts Research Institute, Western University, London, N6A 5K8, Canada
| | - Thomas Hill
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Nerea Jimenez-Tellez
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - S Emma Sarles
- Biomedical and Chemical Engineering PhD Program, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Edward Hensel
- Department of Mechanical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Daniel J Conklin
- Division of Environmental Medicine, Department of Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, 580 S. Preston St., Delia Baxter, Rm. 404E, Louisville, KY, 40202, USA.
| |
Collapse
|
5
|
Kurachi T, Chuman S, Suzuki T, Kubota T, Ishikawa S. Effects of aerosols from heated tobacco products with flavors on the discoloration of bovine tooth enamel. Clin Exp Dent Res 2023; 9:1069-1077. [PMID: 37438937 PMCID: PMC10728517 DOI: 10.1002/cre2.764] [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: 11/25/2022] [Revised: 03/31/2023] [Accepted: 06/22/2023] [Indexed: 07/14/2023] Open
Abstract
OBJECTIVES This in vitro study assessed the potential of tooth discoloration by aerosols generated from three heated tobacco products (HTPs) with different specifications: in-direct heating tobacco system platform 1.0a (IT1.0a), in-direct heating tobacco system platform 2.0a (IT2.0a), and direct heating tobacco system platform 3.0a (DT3.0a). In addition, three flavor types (regular, menthol, and berry menthol) were selected for each HTP to characterize the effect of flavor types on tooth discoloration. MATERIAL AND METHODS Six bovine tooth samples were exposed directly to aerosols generated from one pack of each HTP: 350 puffs for IT1.0a, 325 puffs for IT2.0a, and 220 puffs for DT3.0a. Six bovine tooth samples were also exposed to air (350 puffs) and smoke generated from one pack of cigarettes (160 puffs) as negative and positive controls, respectively. The color of each tooth sample was measured before and after exposure. The overall color changes were assessed using overall color differences (ΔE) calculated according to the Commission International de I'Eclairage color system. A one-way analysis of variance followed by Tukey's post hoc test was used to compare ΔE among bovine tooth samples exposed to air, cigarette smoke, and aerosols generated from each HTP. RESULTS ΔE values for tooth samples exposed to air and aerosols generated from the three HTPs (IT1.0a, IT2.0a, and DT3.0a) were significantly lower than ΔE value for tooth samples exposed to cigarette smoke. ΔE values obtained with DT3.0a were significantly higher than those obtained with air-exposed control samples. However, ΔE values obtained with IT1.0a and IT2.0a were not significantly different from that obtained with air-exposed control samples. No HTPs showed significant differences in ΔE values among the three flavor types. CONCLUSIONS This study showed that HTP aerosols reduce tooth discoloration potential compared with cigarette smoke, regardless of flavor types, and the tooth discoloration potential of the product may depend on product specifications.
Collapse
Affiliation(s)
- Takeshi Kurachi
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc.YokohamaKanagawaJapan
| | - Shinnosuke Chuman
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc.YokohamaKanagawaJapan
| | - Takuya Suzuki
- Scientific and Regulatory Affairs, JT International SAGenevaSwitzerland
| | | | - Shinkichi Ishikawa
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc.YokohamaKanagawaJapan
| |
Collapse
|
6
|
Popp W, Reese L, Scotti E. Heated Tobacco Products and Chronic Obstructive Pulmonary Disease: A Narrative Review of Peer-Reviewed Publications. EUROPEAN MEDICAL JOURNAL 2023. [DOI: 10.33590/emj/10309781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
An estimated 65 million people worldwide have moderate or severe chronic obstructive pulmonary disease (COPD), an umbrella term used to describe a group of progressive lung diseases that obstruct airflow such as emphysema and chronic bronchitis. Smoking contributes to an estimated 90% of COPD cases, as the harmful chemicals produced during tobacco combustion damage the lungs and airways. Although smoking cessation is the only intervention shown to improve COPD prognosis in smokers, many patients who try to quit continue to smoke. The continued use of conventional cigarettes exacerbates COPD symptoms, and globally more than 3 million people die from the disease every year. The last two decades have seen the introduction of combustion-free nicotine delivery alternatives that produce significantly lower levels of the harmful components in cigarette smoke, and researchers have begun to assess the impact of switching from cigarettes to these products. Several studies have examined how patients with COPD use e-cigarettes as assistance for quitting, but few have examined how heated tobacco products (HTP) may reduce risk. This narrative review summarises results from pre-clinical, clinical, and real-world evidence studies showing possible harm reduction benefits for patients with COPD who switch to HTPs rather than continuing to smoke cigarettes. Epidemiological studies, real-world data analyses, and randomised clinical trials must be conducted to determine whether switching from cigarettes to HTPs can improve health outcomes in patients with COPD who would otherwise continue to smoke combustible cigarettes.
Collapse
Affiliation(s)
- Wolfgang Popp
- Ordinationszentrum and Privatklinik Döbling, Vienna, Austria
| | - Lindsay Reese
- Philip Morris International R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Elena Scotti
- Philip Morris International R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| |
Collapse
|
7
|
Ohashi K, Hayashida A, Nozawa A, Matsumura K, Ito S. Human vasculature-on-a-chip with macrophage-mediated endothelial activation: The biological effect of aerosol from heated tobacco products on monocyte adhesion. Toxicol In Vitro 2023; 89:105582. [PMID: 36933580 DOI: 10.1016/j.tiv.2023.105582] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/28/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Heated tobacco products (HTPs) are expected to have the potential to reduce risks of smoking-associated cardiovascular disease (CVD). However, mechanism-based investigations of the effect of HTPs on atherosclerosis remain insufficient and further studies under human-relevant situations are desired for deeper understanding of the reduced risk potential of HTPs. In this study, we first developed an in vitro model of monocyte adhesion by considering macrophage-derived proinflammatory cytokine-mediated endothelial activation using an organ-on-a-chip (OoC), which provided great opportunities to mimic major aspects of human physiology. Then biological activities of aerosol from three different types of HTPs in terms of monocyte adhesion were compared with that of cigarette smoke (CS). Our model showed that the effective concentration ranges of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were close to the actual condition in CVD pathogenesis. The model also showed that monocyte adhesion was less induced by each HTP aerosol than CS, which may be caused by less proinflammatory cytokine secretion. In summary, our vasculature-on-a-chip model assessed the difference in biological effects between cigarettes and HTPs, and suggested a reduced risk potential of HTPs for atherosclerosis.
Collapse
Affiliation(s)
- Kazuhiro Ohashi
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama 227-8512, Kanagawa, Japan.
| | - Ayaka Hayashida
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama 227-8512, Kanagawa, Japan.
| | - Atsuko Nozawa
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama 227-8512, Kanagawa, Japan.
| | - Kazushi Matsumura
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama 227-8512, Kanagawa, Japan.
| | - Shigeaki Ito
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama 227-8512, Kanagawa, Japan.
| |
Collapse
|
8
|
Wong ET, Luettich K, Cammack L, Chua CS, Sciuscio D, Merg C, Corciulo M, Piault R, Ashutosh K, Smith C, Leroy P, Moine F, Glabasnia A, Diana P, Chia C, Tung CK, Ivanov N, Hoeng J, Peitsch M, Lee KM, Vanscheeuwijck P. Assessment of inhalation toxicity of cigarette smoke and aerosols from flavor mixtures: 5-week study in A/J mice. J Appl Toxicol 2022; 42:1701-1722. [PMID: 35543240 PMCID: PMC9545811 DOI: 10.1002/jat.4338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/20/2022] [Accepted: 05/05/2022] [Indexed: 11/10/2022]
Abstract
Most flavors used in e-liquids are generally recognized as safe for oral consumption, but their potential effects when inhaled are not well characterized. In vivo inhalation studies of flavor ingredients in e-liquids are scarce. A structure-based grouping approach was used to select 38 flavor group representatives (FGR) on the basis of known and in silico-predicted toxicological data. These FGRs were combined to create prototype e-liquid formulations and tested against cigarette smoke (CS) in a 5-week inhalation study. Female A/J mice were whole-body exposed for 6 h/day, 5 days/week, for 5 weeks to air, mainstream CS, or aerosols from (1) test formulations containing propylene glycol (PG), vegetable glycerol (VG), nicotine (N; 2% w/w), and flavor (F) mixtures at low (4.6% w/w), medium (9.3% w/w), or high (18.6% w/w) concentration or (2) base formulation (PG/VG/N). Male A/J mice were exposed to air, PG/VG/N, or PG/VG/N/F-high under the same exposure regimen. There were no significant mortality or in-life clinical findings in the treatment groups, with only transient weight loss during the early exposure adaptation period. While exposure to flavor aerosols did not cause notable lung inflammation, it caused only minimal adaptive changes in the larynx and nasal epithelia. In contrast, exposure to CS resulted in lung inflammation and moderate-to-severe changes in the epithelia of the nose, larynx, and trachea. In summary, the study evaluates an approach for assessing the inhalation toxicity potential of flavor mixtures, thereby informing the selection of flavor exposure concentrations (up to 18.6%) for a future chronic inhalation study.
Collapse
Affiliation(s)
- Ee Tsin Wong
- PMI R&DPhilip Morris International Research Laboratories Pte LtdSingapore
| | | | - Lydia Cammack
- PMI R&DPhilip Morris International Research Laboratories Pte LtdSingapore
| | - Chin Suan Chua
- PMI R&DPhilip Morris International Research Laboratories Pte LtdSingapore
| | | | - Celine Merg
- PMI R&DPhilip Morris Products S.ANeuchâtelSwitzerland
| | | | - Romain Piault
- PMI R&DPhilip Morris Products S.ANeuchâtelSwitzerland
| | | | | | - Patrice Leroy
- PMI R&DPhilip Morris Products S.ANeuchâtelSwitzerland
| | - Fabian Moine
- PMI R&DPhilip Morris Products S.ANeuchâtelSwitzerland
| | | | | | - Cecilia Chia
- PMI R&DPhilip Morris International Research Laboratories Pte LtdSingapore
| | - Ching Keong Tung
- PMI R&DPhilip Morris International Research Laboratories Pte LtdSingapore
| | | | - Julia Hoeng
- PMI R&DPhilip Morris Products S.ANeuchâtelSwitzerland
| | | | | | | |
Collapse
|
9
|
Staal YCM, Bil W, Bokkers BGH, Soeteman-Hernández LG, Stephens WE, Talhout R. Challenges in Predicting the Change in the Cumulative Exposure of New Tobacco and Related Products Based on Emissions and Toxicity Dose-Response Data. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10528. [PMID: 36078242 PMCID: PMC9518537 DOI: 10.3390/ijerph191710528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Many novel tobacco products have been developed in recent years. Although many may emit lower levels of several toxicants, their risk in the long term remains unclear. We previously published a method for the exposure assessment of mixtures that can be used to compare the changes in cumulative exposure to carcinogens among tobacco products. While further developing this method by including more carcinogens or to explore its application to non-cancer endpoints, we encountered a lack of data that are required for better-substantiated conclusions regarding differences in exposure between products. In this special communication, we argue the case for more data on adverse health effects, as well as more data on the composition of the emissions from tobacco products. Such information can be used to identify significant changes in relevance to health using the cumulative exposure method with different products and to substantiate regulatory decisions.
Collapse
Affiliation(s)
- Yvonne C. M. Staal
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| | - Wieneke Bil
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| | - Bas G. H. Bokkers
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| | - Lya G. Soeteman-Hernández
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| | - W. Edryd Stephens
- School of Earth & Environmental Sciences, University of St Andrews, St Andrews KY16 9AJ, UK
| | - Reinskje Talhout
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| |
Collapse
|
10
|
Wang Y, Ryu R, Seo JM, Lee JJ. Effects of conventional and heated tobacco product smoking on discoloration of artificial denture teeth. J Prosthet Dent 2022; 128:206-210. [PMID: 33608106 DOI: 10.1016/j.prosdent.2020.05.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 11/18/2022]
Abstract
STATEMENT OF PROBLEM Cigarette smoke can cause discoloration of artificial denture teeth. However, studies on the effects of heated tobacco product smoke on artificial denture teeth are lacking. PURPOSE The purpose of this in vitro study was to evaluate the effects of conventional cigarette and heated tobacco product smoke on the color stability of artificial denture teeth. MATERIAL AND METHODS Ninety maxillary central incisor denture teeth (Endura Anterior HC5 A3; Shofu) were randomly divided into 3 groups (n=30). Teeth in the control group were exposed to air; those in group CC were exposed to conventional cigarette (Marlboro Medium; Philip Morris) smoke, and those in group HT were exposed to heated tobacco product (IQOS 2.4 plus holder, Marlboro Heets Silver; Philip Morris) smoke. Before the experiment, the shade of the artificial denture teeth was evaluated in accordance with the Commission International de I'Eclairage (CIELab) color system by using a spectrophotometer (Shadepilot; DeguDent GmbH). The average CIELab value was estimated by scanning the entire labial surface of each specimen. To simulate smoking, standard conditions described by the Coresta Recommended Method N°22 were used-the puff duration was 2 seconds, with a 60-second interval between puffs. For each cigarette, 6 puffs and 6 intervals were simulated across 372 seconds. A total of 105 cigarettes were used based on a smoking simulation of 15 cigarettes each day for 7 days. The teeth in the control group were stored in fresh air in the smoke chamber for the same period as those in the experimental groups. After the experiment, L∗, a∗, and b∗ values were measured, and ΔE was calculated to evaluate the color change. All statistical analyses were performed with a statistical software program using a paired t test to determine discoloration after exposure to cigarette smoke. One-way ANOVA and the Tukey test were used to evaluate the significant differences between groups (α=.05). RESULTS Lightness was significantly lower in the CC and HT groups (P<.001). All CIELab values showed statistically significant differences in the CC group. The greatest color change was observed in the CC group (ΔE=6.93 ±0.59), whereas the HT group showed a clinically imperceptible color change (ΔE=0.79 ±0.21). Discoloration was minimal in the CC group (ΔE=0.34 ±0.13). CONCLUSIONS Conventional cigarette and heated tobacco product smoke can change the color of denture teeth. Heated tobacco product smoke causes less discoloration of denture teeth.
Collapse
Affiliation(s)
- Yuankun Wang
- Postgraduate student, Department of Prosthodontics, Institute of Oral Bio-Science, School of Dentistry, Jeonbuk National University, Jeonju, Republic of Korea
| | - Ri Ryu
- Resident and Postgraduate student, Department of Prosthodontics, Institute of Oral Bio-Science, School of Dentistry, Jeonbuk National University, Jeonju, Republic of Korea
| | - Jae-Min Seo
- Associate Professor, Department of Prosthodontics, Institute of Oral Bio-Science, School of Dentistry, Jeonbuk National University and Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Jung-Jin Lee
- Assistant Professor, Department of Prosthodontics, Institute of Oral Bio-Science, School of Dentistry, Jeonbuk National University and Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea.
| |
Collapse
|
11
|
Eden MJ, Farra YM, Matz J, Bellini C, Oakes JM. Pharmacological and physiological response in Apoe -/- mice exposed to cigarette smoke or e-cigarette aerosols. Inhal Toxicol 2022; 34:260-274. [PMID: 35793285 DOI: 10.1080/08958378.2022.2086948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Electronic cigarettes (e-cigs) are popular nicotine delivery devices, yet the health effects remain unclear. To determine equivalent biomarkers, we characterized the immediate response in Apoe-/- mice exposed to tank/box-mod e-cig (e-cigtank), pod e-cig (e-cigpod), or cig smoke. MATERIALS AND METHODS Reproducible puff profiles were generated for each aerosol and delivered to Apoe-/- mice via a nose-only exposure system. Serum cotinine levels were quantified at various time points through ELISA and utilized to model cotinine pharmacokinetics. In addition, particle size measurements and mouse respiratory function were characterized to calculate particle dosimetry. RESULTS AND DISCUSSION Cig and e-cigtank particles were lognormally distributed with similar count median diameters (cig: 178 ± 2, e-cigtank: 200 ± 34nm), while e-cigpod particles were bimodally distributed and smaller (116 ± 13 and 13.3 ± 0.4 nm). Minute volumes decreased with cig exposure (5.4 ± 2.7 mL/min) compared to baseline (90.8 ± 11.6 mL/min), and less so with e-cigtank (45.2 ± 9.2 mL/min) and e-cigpod exposures (58.6 ± 6.8 mL/min), due to periods of apnea in the cig exposed groups. Cotinine was absorbed and eliminated most rapidly in the e-cigpod group (tmax = 14.5; t1/2' = 51.9 min), whereas cotinine was absorbed (cig: 50.4, e-cigtank: 40.1 min) and eliminated (cig: 104.6, e-cigtank: 94.1 min) similarly in the cig and e-cigtank groups. For exposure times which equate the area under the cotinine-concentration curve, ∼6.4× (e-cigtank) and 4.6× (e-cigpod) more nicotine deposited in e-cig compared to cig exposed mice. CONCLUSIONS This study provides a basis for incorporating cotinine pharmacokinetics into preclinical exposure studies, allowing for longitudinal studies of structural and functional changes due to exposure.
Collapse
Affiliation(s)
- Matthew J Eden
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Yasmeen M Farra
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Jacqueline Matz
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Chiara Bellini
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Jessica M Oakes
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
12
|
Uguna CN, Snape CE. Should IQOS Emissions Be Considered as Smoke and Harmful to Health? A Review of the Chemical Evidence. ACS OMEGA 2022; 7:22111-22124. [PMID: 35811880 PMCID: PMC9260752 DOI: 10.1021/acsomega.2c01527] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
The chemical evidence that IQOS emissions fit the definition of both an aerosol and smoke, and that IQOS and potentially other heated tobacco products (HTPs) pose some harmful health threats from the range of compounds released even at somewhat lower concentrations is reviewed. Further, we address the yields of harmful and potentially harmful compounds (HPHCs), including polycyclic aromatic hydrocarbons (PAHs), and the constituents of IQOS emission that are diagnostic of pyrolysis to provide information on the temperatures reached in IQOS tobacco sticks. The HPHCs present in IQOS emissions are the same as in conventional cigarette smoke (CCs), analogous to emissions from earlier generation of HTPs classed as smoke. However, Philip Morris International (PMI) studies have to some degree underestimated IQOS aerosol HPHC yields, which are a factor of between 3.2 and 3.6 higher when expressed on a tobacco rather than an IQOS stick basis compared to the reference 3R4F cigarette. Further, IQOS emissions contain carbon particles, which fit definition of both aerosol and smoke. Continual reheating of deposited tar in the IQOS device will occur with real-life use, likely leading to generation of even higher concentrations of HPHCs and particulate matter. Despite IQOS not exceeding 350 °C, local hot spots could exist, causing formation of species (phenol/cresols, PAHs). It is recommended that the impact of repeated use to determine the levels of black carbon (insoluble organic matter) in the particulate matter, and the extent to which compounds in IQOS emissions are formed by pyrolysis need to be assessed rigorously. To address whether uneven temperature profiles in heat sticks can lead to potential hot spots that could, for example, lead to PAH formation, it is recommended that pyrolysis studies on tobacco and other constituents of HTPs are required in conjunction with more effort on heating tobacco blends under controlled temperature/time conditions.
Collapse
|
13
|
Crowley LN, Le BL, Cicalo C, Brown J, Li Y, Kim YJ, Lee JH, Pan JH, Lennon SL, Han BK, Kim JK. Acrolein, an environmental toxicant and its applications to in vivo and in vitro atherosclerosis models: An update. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 93:103890. [PMID: 35613694 DOI: 10.1016/j.etap.2022.103890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/14/2022] [Accepted: 05/18/2022] [Indexed: 06/03/2023]
Abstract
Cardiovascular disease, the foremost cause of death worldwide, is an overarching disease term that encompasses a number of disorders involving the heart and circulatory system, including atherosclerosis. Atherosclerosis is a primary cause of cardiovascular diseases and is caused by buildup of plaque and narrowing of blood vessels. Epidemiological studies have suggested that environmental pollutants are implicated in atherosclerosis disease progression. Among many environmental pollutants, acrolein (Acr) is an abundant reactive aldehyde and is ubiquitously present in cigarette smoke as well as food products (e.g., overheated oils and wine). Despite its ubiquitous presence and potential impact on the etiology of cardiovascular disease, a limited consensus has been made in regard to Acr exposure conditions to induce atherosclerosis in vivo. This mini-review summarizes in vivo atherosclerosis models using Acr to investigate biochemical and phenotypic changes related to atherosclerosis and in vitro mechanistic studies involving Acr and atherosclerosis.
Collapse
Affiliation(s)
- Liana N Crowley
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716, USA
| | - Brandy L Le
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716, USA
| | - Cara Cicalo
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716, USA
| | - Justin Brown
- Department of Medical and Molecular Sciences, University of Delaware, Newark, DE 19716, USA
| | - Yali Li
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716, USA
| | - Young Jun Kim
- Department of Food and Biotechnology, Korea University, Sejong 30019, South Korea
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, Sejong 30019, South Korea
| | - Jeong Hoon Pan
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716, USA
| | - Shannon L Lennon
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE 19713, USA
| | - Bok Kyung Han
- Department of Food and Biotechnology, Korea University, Sejong 30019, South Korea
| | - Jae Kyeom Kim
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
14
|
Giebe S, Hofmann A, Brux M, Lowe F, Breheny D, Morawietz H, Brunssen C. Comparative study of the effects of cigarette smoke versus next generation tobacco and nicotine product extracts on endothelial function. Redox Biol 2021; 47:102150. [PMID: 34601427 PMCID: PMC8531844 DOI: 10.1016/j.redox.2021.102150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 11/29/2022] Open
Abstract
Tobacco smoking and hemodynamic forces are key stimuli for the development of endothelial dysfunction. As an alternative to smoking, next generation tobacco and nicotine products (NGP) are now widely used. However, little is known about their potential pro-inflammatory and atherogenic effects on the endothelium. In this study, we analyzed key parameters of endothelial function after exposure to aqueous smoke extracts (AqE) of a heated tobacco product (HTP), an electronic cigarette (e-cig), a conventional cigarette (3R4F) and pure nicotine. All experiments were performed under atheroprotective high laminar or atherogenic low flow with primary human endothelial cells. Treatment with 3R4F, but not alternative smoking products, reduced endothelial cell viability and wound healing capability via the PI3K/AKT/eNOS(NOS3) pathway. Laminar flow delayed detrimental effects on cell viability by 3R4F treatment. 3R4F stimulation led to activation of NRF2 antioxidant defense system at nicotine concentrations ≥0.56 μg/ml and increased expression of its target genes HMOX1 and NQO1. Treatment with HTP revealed an induction of HMOX1 and NQO1 at dosages with ≥1.68 μg/ml nicotine, whereas e-cig and nicotine exposure had no impact. Analyses of pro-inflammatory genes revealed an increased ICAM1 expression under 3R4F treatment. 3R4F reduced VCAM1 expression in a dose-dependent manner; HTP treatment had similar but milder effects; e-cig and nicotine treatment had no impact. SELE expression was induced by 3R4F under static conditions. High laminar flow prevented this upregulation. Stimulation with laminar flow led to downregulation of CCL2 (MCP-1). From this downregulated level, only 3R4F increased CCL2 expression at higher concentrations. Finally, under static conditions, all components increased adhesion of monocytes to endothelial cells. Interestingly, only stimulation with 3R4F revealed increased monocyte adhesion under atherosclerosis-prone low flow. In conclusion, all product categories activated anti-oxidative or pro-inflammatory patterns. NGP responses were typically lower than in 3R4F exposed cells. Also, 3R4F stimulation led to an impaired endothelial wound healing and induced a pro-inflammatory phenotype compared to NGP treatment.
Collapse
Affiliation(s)
- Sindy Giebe
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Anja Hofmann
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Melanie Brux
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Frazer Lowe
- Group Research & Development, British American Tobacco, Southampton, United Kingdom
| | - Damien Breheny
- Group Research & Development, British American Tobacco, Southampton, United Kingdom
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
15
|
Wong ET, Luettich K, Krishnan S, Wong SK, Lim WT, Yeo D, Büttner A, Leroy P, Vuillaume G, Boué S, Hoeng J, Vanscheeuwijck P, Peitsch MC. Reduced Chronic Toxicity and Carcinogenicity in A/J Mice in Response to Life-Time Exposure to Aerosol From a Heated Tobacco Product Compared With Cigarette Smoke. Toxicol Sci 2021; 178:44-70. [PMID: 32780830 PMCID: PMC7657344 DOI: 10.1093/toxsci/kfaa131] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We conducted an inhalation study, in accordance with Organisation for Economic Co-operation and Development Test Guideline 453, exposing A/J mice to tobacco heating system (THS) 2.2 aerosol or 3R4F reference cigarette smoke (CS) for up to 18 months to evaluate chronic toxicity and carcinogenicity. All exposed mice showed lower thymus and spleen weight, blood lymphocyte counts, and serum lipid concentrations than sham mice, most likely because of stress and/or nicotine effects. Unlike THS 2.2 aerosol-exposed mice, CS-exposed mice showed increased heart weight, changes in red blood cell profiles and serum liver function parameters. Similarly, increased pulmonary inflammation, altered lung function, and emphysematous changes were observed only in CS-exposed mice. Histopathological changes in other respiratory tract organs were significantly lower in the THS 2.2 aerosol-exposed groups than in the CS-exposed group. Chronic exposure to THS 2.2 aerosol also did not increase the incidence or multiplicity of bronchioloalveolar adenomas or carcinomas relative to sham, whereas CS exposure did. Male THS 2.2 aerosol-exposed mice had a lower survival rate than sham mice, related to an increased incidence of urogenital issues that appears to be related to congenital factors rather than test item exposure. The lower impact of THS 2.2 aerosol exposure on tumor development and chronic toxicity is consistent with the significantly reduced levels of harmful and potentially harmful constituents in THS 2.2 aerosol relative to CS. The totality of the evidence from this study further supports the risk reduction potential of THS 2.2 for lung diseases in comparison with cigarettes.
Collapse
Affiliation(s)
- Ee Tsin Wong
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Karsta Luettich
- Department of Life Sciences, Systems Toxicology, PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | - Subash Krishnan
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Sin Kei Wong
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Wei Ting Lim
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Demetrius Yeo
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | | | - Patrice Leroy
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Grégory Vuillaume
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Stéphanie Boué
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Julia Hoeng
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Patrick Vanscheeuwijck
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Manuel C Peitsch
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| |
Collapse
|
16
|
Titz B, Sewer A, Luettich K, Wong ET, Guedj E, Nury C, Schneider T, Xiang Y, Trivedi K, Vuillaume G, Leroy P, Büttner A, Martin F, Ivanov NV, Vanscheeuwijck P, Hoeng J, Peitsch MC. Respiratory Effects of Exposure to Aerosol From the Candidate Modified-Risk Tobacco Product THS 2.2 in an 18-Month Systems Toxicology Study With A/J Mice. Toxicol Sci 2021; 178:138-158. [PMID: 32780831 PMCID: PMC7657339 DOI: 10.1093/toxsci/kfaa132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Smoking cessation is the most effective measure for reducing the risk of smoking-related diseases. However, switching to less harmful products (modified-risk tobacco products [MRTP]) can be an alternative to help reduce the risk for adult smokers who would otherwise continue to smoke. In an 18-month chronic carcinogenicity/toxicity study in A/J mice (OECD Test Guideline 453), we assessed the aerosol of Tobacco Heating System 2.2 (THS 2.2), a candidate MRTP based on the heat-not-burn principle, compared with 3R4F cigarette smoke (CS). To capture toxicity- and disease-relevant mechanisms, we complemented standard toxicology endpoints with in-depth systems toxicology analyses. In this part of our publication series, we report on integrative assessment of the apical and molecular exposure effects on the respiratory tract (nose, larynx, and lungs). Across the respiratory tract, we found changes in inflammatory response following 3R4F CS exposure (eg, antimicrobial peptide response in the nose), with both shared and distinct oxidative and xenobiotic responses. Compared with 3R4F CS, THS 2.2 aerosol exerted far fewer effects on respiratory tract histology, including adaptive tissue changes in nasal and laryngeal epithelium and inflammation and emphysematous changes in the lungs. Integrative analysis of molecular changes confirmed the substantially lower impact of THS 2.2 aerosol than 3R4F CS on toxicologically and disease-relevant molecular processes such as inflammation, oxidative stress responses, and xenobiotic metabolism. In summary, this work exemplifies how apical and molecular endpoints can be combined effectively for toxicology assessment and further supports findings on the reduced respiratory health risks of THS 2.2 aerosol.
Collapse
Affiliation(s)
- Bjoern Titz
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | - Alain Sewer
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | - Karsta Luettich
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | - Ee Tsin Wong
- Philip Morris International Research Laboratories Pte. Ltd, Singapore 117406
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | - Catherine Nury
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | | | - Yang Xiang
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | - Keyur Trivedi
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | | | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | | | - Florian Martin
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | | | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| |
Collapse
|
17
|
Battey JND, Szostak J, Phillips B, Teng C, Tung CK, Lim WT, Yeo YS, Ouadi S, Baumer K, Thomas J, Martinis J, Sierro N, Ivanov NV, Vanscheeuwijck P, Peitsch MC, Hoeng J. Impact of 6-Month Exposure to Aerosols From Potential Modified Risk Tobacco Products Relative to Cigarette Smoke on the Rodent Gastrointestinal Tract. Front Microbiol 2021; 12:587745. [PMID: 34276574 PMCID: PMC8283309 DOI: 10.3389/fmicb.2021.587745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 06/10/2021] [Indexed: 01/14/2023] Open
Abstract
Cigarette smoking causes adverse health effects that might occur shortly after smoking initiation and lead to the development of inflammation and cardiorespiratory disease. Emerging studies have demonstrated the role of the intestinal microbiome in disease pathogenesis. The intestinal microbiome is susceptible to the influence of environmental factors such as smoking, and recent studies have indicated microbiome changes in smokers. Candidate modified risk tobacco products (CMRTP) are being developed to provide substitute products to lower smoking-related health risks in smokers who are unable or unwilling to quit. In this study, the ApoE–/– mouse model was used to investigate the impact of cigarette smoke (CS) from the reference cigarette 3R4F and aerosols from two CMRTPs based on the heat-not-burn principle [carbon-heated tobacco product 1.2 (CHTP 1.2) and tobacco heating system 2.2 (THS 2.2)] on the intestinal microbiome over a 6-month period. The effect of cessation or switching to CHTP 1.2 after 3 months of CS exposure was also assessed. Next-generation sequencing was used to evaluate the impact of CMRTP aerosols in comparison to CS on microbiome composition and gene expression in the digestive tract of mice. Our analyses highlighted significant gene dysregulation in response to 3R4F exposure at 4 and 6 months. The findings showed an increase in the abundance of Akkermansiaceae upon CS exposure, which was reversed upon cessation. Cessation resulted in a significant decrease in Akkemansiaceae abundance, whereas switching to CHTP 1.2 resulted in an increase in Lactobacillaceae abundance. These microbial changes could be important for understanding the effect of CS on gut function and its relevance to disease pathogenesis via the microbiome.
Collapse
Affiliation(s)
| | - Justyna Szostak
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Blaine Phillips
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore, Singapore
| | - Charles Teng
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore, Singapore
| | - Ching Keong Tung
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore, Singapore
| | - Wei Ting Lim
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore, Singapore
| | - Ying Shan Yeo
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore, Singapore
| | - Sonia Ouadi
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Karine Baumer
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Jerome Thomas
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Jacopo Martinis
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Nicolas Sierro
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | | | | | | | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| |
Collapse
|
18
|
Wong ET, Szostak J, Titz B, Lee T, Wong SK, Lavrynenko O, Merg C, Corciulo M, Simicevic J, Auberson M, Peric D, Dulize R, Bornand D, Loh GJ, Lee KM, Zhang J, Miller JH, Schlage WK, Guedj E, Schneider T, Phillips B, Leroy P, Choukrallah MA, Sierro N, Buettner A, Xiang Y, Kuczaj A, Ivanov NV, Luettich K, Vanscheeuwijck P, Peitsch MC, Hoeng J. A 6-month inhalation toxicology study in Apoe -/- mice demonstrates substantially lower effects of e-vapor aerosol compared with cigarette smoke in the respiratory tract. Arch Toxicol 2021; 95:1805-1829. [PMID: 33963423 PMCID: PMC8113187 DOI: 10.1007/s00204-021-03020-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/03/2021] [Indexed: 11/26/2022]
Abstract
Cigarette smoking is the major cause of chronic obstructive pulmonary disease. Considerable attention has been paid to the reduced harm potential of nicotine-containing inhalable products such as electronic cigarettes (e-cigarettes). We investigated the effects of mainstream cigarette smoke (CS) and e-vapor aerosols (containing nicotine and flavor) generated by a capillary aerosol generator on emphysematous changes, lung function, and molecular alterations in the respiratory system of female Apoe-/- mice. Mice were exposed daily (3 h/day, 5 days/week) for 6 months to aerosols from three different e-vapor formulations-(1) carrier (propylene glycol and vegetable glycerol), (2) base (carrier and nicotine), or (3) test (base and flavor)-or to CS from 3R4F reference cigarettes. The CS and base/test aerosol concentrations were matched at 35 µg nicotine/L. CS exposure, but not e-vapor exposure, led to impairment of lung function (pressure-volume loop area, A and K parameters, quasi-static elastance and compliance) and caused marked lung inflammation and emphysematous changes, which were confirmed histopathologically and morphometrically. CS exposure caused lung transcriptome (activation of oxidative stress and inflammatory responses), lipidome, and proteome dysregulation and changes in DNA methylation; in contrast, these effects were substantially reduced in response to the e-vapor aerosol exposure. Compared with sham, aerosol exposure (carrier, base, and test) caused a slight impact on lung inflammation and epithelia irritation. Our results demonstrated that, in comparison with CS, e-vapor aerosols induced substantially lower biological and pathological changes in the respiratory tract associated with chronic inflammation and emphysema.
Collapse
Affiliation(s)
- Ee Tsin Wong
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore, 117406, Singapore
| | - Justyna Szostak
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland.
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Tom Lee
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore, 117406, Singapore
| | - Sin Kei Wong
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore, 117406, Singapore
| | - Oksana Lavrynenko
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Celine Merg
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Maica Corciulo
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Jovan Simicevic
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Mehdi Auberson
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Dariusz Peric
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Remi Dulize
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - David Bornand
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Guo Jie Loh
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore, 117406, Singapore
| | | | - Jingjie Zhang
- Altria Client Services LLC, 601 East Jackson Street, Richmond, VA, 23219, USA
| | - John H Miller
- Altria Client Services LLC, 601 East Jackson Street, Richmond, VA, 23219, USA
| | | | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Thomas Schneider
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Blaine Phillips
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore, 117406, Singapore
| | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | | | - Nicolas Sierro
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | | | - Yang Xiang
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Arkadiusz Kuczaj
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Karsta Luettich
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | | | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| |
Collapse
|
19
|
Kogel U, Wong ET, Szostak J, Tan WT, Lucci F, Leroy P, Titz B, Xiang Y, Low T, Wong SK, Guedj E, Ivanov NV, Schlage WK, Peitsch MC, Kuczaj A, Vanscheeuwijck P, Hoeng J. Impact of whole-body versus nose-only inhalation exposure systems on systemic, respiratory, and cardiovascular endpoints in a 2-month cigarette smoke exposure study in the ApoE -/- mouse model. J Appl Toxicol 2021; 41:1598-1619. [PMID: 33825214 PMCID: PMC8519037 DOI: 10.1002/jat.4149] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 11/09/2022]
Abstract
Cigarette smoking is one major modifiable risk factor in the development and progression of chronic obstructive pulmonary disease and cardiovascular disease. To characterize and compare cigarette smoke (CS)-induced disease endpoints after exposure in either whole-body (WB) or nose-only (NO) exposure systems, we exposed apolipoprotein E-deficient mice to filtered air (Sham) or to the same total particulate matter (TPM) concentration of mainstream smoke from 3R4F reference cigarettes in NO or WB exposure chambers (EC) for 2 months. At matching TPM concentrations, we observed similar concentrations of carbon monoxide, acetaldehyde, and acrolein, but higher concentrations of nicotine and formaldehyde in NOEC than in WBEC. In both exposure systems, CS exposure led to the expected adaptive changes in nasal epithelia, altered lung function, lung inflammation, and pronounced changes in the nasal epithelial transcriptome and lung proteome. Exposure in the NOEC caused generally more severe histopathological changes in the nasal epithelia and a higher stress response as indicated by body weight decrease and lower blood lymphocyte counts compared with WB exposed mice. Erythropoiesis, and increases in total plasma triglyceride levels and atherosclerotic plaque area were observed only in CS-exposed mice in the WBEC group but not in the NOEC group. Although the composition of CS in the breathing zone is not completely comparable in the two exposure systems, the CS-induced respiratory disease endpoints were largely confirmed in both systems, with a higher magnitude of severity after NO exposure. CS-accelerated atherosclerosis and other pro-atherosclerotic factors were only significant in WBEC.
Collapse
Affiliation(s)
- Ulrike Kogel
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Ee Tsin Wong
- Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Justyna Szostak
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Wei Teck Tan
- Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Francesco Lucci
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Patrice Leroy
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Bjoern Titz
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Yang Xiang
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Tiffany Low
- Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Sin Kei Wong
- Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Emmanuel Guedj
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Nikolai V Ivanov
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Walter K Schlage
- Biology Consultant, Max-Baermann-Str. 21, Bergisch Gladbach, Germany
| | - Manuel C Peitsch
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Arkadiusz Kuczaj
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Patrick Vanscheeuwijck
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Julia Hoeng
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchatel, Switzerland
| |
Collapse
|
20
|
Talikka M, Belcastro V, Boué S, Marescotti D, Hoeng J, Peitsch MC. Applying Systems Toxicology Methods to Drug Safety. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11522-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
21
|
Immunomodulatory Effects of Hydrolyzed Seawater Pearl Tablet (HSPT) on Th1/Th2 Functionality in a Mice Model of Chronic Obstructive Pulmonary Disease (COPD) Induced by Cigarette Smoke. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:5931652. [PMID: 33281913 PMCID: PMC7688355 DOI: 10.1155/2020/5931652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/09/2020] [Accepted: 10/30/2020] [Indexed: 11/17/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is predicted to become the third leading cause of death around the world. The present study is designed to investigate whether hydrolyzed seawater pearl tablet (HSPT) has immunoregulatory effects on the Th1/Th2 functionality in cigarette smoke-induced COPD model mice. The determination of the amino acid composition of HSPT was carried out by high-performance liquid chromatography (HPLC) with precolumn phenylisothiocyanate (PITC) derivatization. COPD model mice were constructed by cigarette smoking (CS) treatment and HSPT was administered. HSPT inhibited the infiltration of inflammation in the airway of the lung, reduced influx of eosinophils (EOSs), lymphocytes (LYMs), neutrophils (NEUs), and macrophages (MACs) in the bronchoalveolar lavage fluid (BALF), decreased the levels of IFN-γ, IL-2, IL-4, and IL-10 in the serum and lung, and decreased the expression of aforementioned cytokines in the spleen and lung in CS-treated mice. Besides, HSPT also had the ability to reduce the amount of CD3+CD4+ T cells and modulate the Th1/Th2 balance. Taken together, this study supports the consensus that CS is a critical factor to induce and aggravate COPD. HSPT could regulate the balance of Th1/Th2 in CS-induced COPD model mice, indicating its effects on inhibiting the development of COPD.
Collapse
|
22
|
van der Plas A, Pouly S, Blanc N, Haziza C, de La Bourdonnaye G, Titz B, Hoeng J, Ivanov NV, Taranu B, Heremans A. Impact of switching to a heat-not-burn tobacco product on CYP1A2 activity. Toxicol Rep 2020; 7:1480-1486. [PMID: 33204648 PMCID: PMC7649435 DOI: 10.1016/j.toxrep.2020.10.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/18/2020] [Accepted: 10/23/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Cigarette smoking induces cytochrome P450 1A2 (CYP1A2) expression and activity, while smoking cessation normalizes the levels of this enzyme. The aim of this publication is to summarize the data on CYP1A2 gene expression and activity in preclinical and clinical studies on the Tobacco Heating System (THS), currently marketed as IQOS® with HEETs®, and to summarize the potential effects on CYP1A2 to be expected upon switching to reduced-risk products (RRPs). METHODS We summarized PMI's preclinical and clinical data on the effects of switching from cigarette smoking to THS. RESULTS Data from four preclinical mouse and rat studies showed that, upon either cessation of cigarette smoke exposure or switching to THS exposure, the upregulation of CYP1A2 observed with exposure to cigarette smoke reverted close to fresh-air levels. Data from four clinical studies yielded similar results on CYP1A2 activity within a time frame of five days. Furthermore, the effects of switching to THS were similar to those seen after smoking cessation. CONCLUSIONS Because smoking cessation and switching to either electronic cigarettes or THS seem to have similar effects on CYP1A2 activity, the same measures taken for patients treated with narrow therapeutic index drugs that are metabolized by CYP1A2 and who quit smoking should be recommended for those switching to RRPs.
Collapse
Affiliation(s)
- Angela van der Plas
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Sandrine Pouly
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Nicolas Blanc
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Christelle Haziza
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | | | - Bjorn Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Nikolai V. Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Brindusa Taranu
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Annie Heremans
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| |
Collapse
|
23
|
Schlage WK, Titz B, Iskandar A, Poussin C, Van der Toorn M, Wong ET, Pratte P, Maeder S, Schaller JP, Pospisil P, Boue S, Vuillaume G, Leroy P, Martin F, Ivanov NV, Peitsch MC, Hoeng J. Comparing the preclinical risk profile of inhalable candidate and potential candidate modified risk tobacco products: A bridging use case. Toxicol Rep 2020; 7:1187-1206. [PMID: 32995294 PMCID: PMC7502378 DOI: 10.1016/j.toxrep.2020.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/24/2020] [Accepted: 09/01/2020] [Indexed: 11/25/2022] Open
Abstract
Heated tobacco products tested for reduced exposure and reduced risk properties. Bridging opportunities for nonclinical results from two heated tobacco products. Similarly reduced impact on apical and molecular endpoints relative to cigarettes. Evidence evaluated along a “causal chain of events leading to disease” (CELSD). Representative assays along CELSD could support nonclinical substantial equivalence.
Cigarette smoking causes major preventable diseases, morbidity, and mortality worldwide. Smoking cessation and prevention of smoking initiation are the preferred means for reducing these risks. Less harmful tobacco products, termed modified-risk tobacco products (MRTP), are being developed as a potential alternative for current adult smokers who would otherwise continue smoking. According to a regulatory framework issued by the US Food and Drug Administration, a manufacturer must provide comprehensive scientific evidence that the product significantly reduces harm and the risk of tobacco-related diseases, in order to obtain marketing authorization for a new MRTP. For new tobacco products similar to an already approved predicate product, the FDA has foreseen a simplified procedure for assessing “substantial equivalence”. In this article, we present a use case that bridges the nonclinical evidence from previous studies demonstrating the relatively reduced harm potential of two heat-not-burn products based on different tobacco heating principles. The nonclinical evidence was collected along a “causal chain of events leading to disease” (CELSD) to systematically follow the consequences of reduced exposure to toxicants (relative to cigarette smoke) through increasing levels of biological complexity up to disease manifestation in animal models of human disease. This approach leverages the principles of systems biology and toxicology as a basis for further extrapolation to human studies. The experimental results demonstrate a similarly reduced impact of both products on apical and molecular endpoints, no novel effects not seen with cigarette smoke exposure, and an effect of switching from cigarettes to either MRTP that is comparable to that of complete smoking cessation. Ideally, a subset of representative assays from the presented sequence along the CELSD could be sufficient for predicting similarity or substantial equivalence in the nonclinical impact of novel products; this would require further validation, for which the present use case could serve as a starting point.
Collapse
Key Words
- BIF, biological impact factor
- CELSD, causal chain of events leading to disease
- CHTP, carbon heated tobacco product
- CS, cigarette smoke
- CVD, cardiovascular disease
- GVP, gas/vapor phase
- HPHC, harmful and potentially harmful constituents
- MRTP, modified risk tobacco product
- Modified risk tobacco product (MRTP)
- NPA, network perturbation amplitude
- PMI, Philip Morris International
- RBIF, relative BIF
- Substantial equivalence
- Systems toxicology
- THS, Tobacco Heating System
- TPM, total particulate matter
- Tobacco harm reduction
Collapse
Affiliation(s)
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Anita Iskandar
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Carine Poussin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Marco Van der Toorn
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Ee Tsin Wong
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Pascal Pratte
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Serge Maeder
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Jean-Pierre Schaller
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Pavel Pospisil
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Stephanie Boue
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Grégory Vuillaume
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Florian Martin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| |
Collapse
|
24
|
Haziza C, de La Bourdonnaye G, Donelli A, Skiada D, Poux V, Weitkunat R, Baker G, Picavet P, Lüdicke F. Favorable Changes in Biomarkers of Potential Harm to Reduce the Adverse Health Effects of Smoking in Smokers Switching to the Menthol Tobacco Heating System 2.2 for 3 Months (Part 2). Nicotine Tob Res 2020; 22:549-559. [PMID: 31125079 PMCID: PMC7164580 DOI: 10.1093/ntr/ntz084] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 05/23/2019] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Tobacco Heating System (THS) 2.2, a candidate modified-risk tobacco product, aims at offering an alternative to cigarettes for smokers while substantially reducing the exposure to harmful and potentially harmful constituents found in cigarette smoke. METHODS One hundred and sixty healthy adult US smokers participated in this randomized, three-arm parallel group, controlled clinical study. Subjects were randomized in a 2:1:1 ratio to menthol Tobacco Heating System 2.2 (mTHS), menthol cigarette, or smoking abstinence for 5 days in confinement and 86 subsequent ambulatory days. Endpoints included biomarkers of exposure to harmful and potentially harmful constituents (reported in our co-publication, Part 1) and biomarkers of potential harm (BOPH). RESULTS Compliance (protocol and allocated product exposure) was 51% and 18% in the mTHS and smoking abstinence arms, respectively, on day 90. Nonetheless, favorable changes in BOPHs of lipid metabolism (total cholesterol and high- and low-density cholesterol), endothelial dysfunction (soluble intercellular adhesion molecule-1), oxidative stress (8-epi-prostaglandin F2α), and cardiovascular risk factors (eg, high-sensitivity C-reactive protein) were observed in the mTHS group. Favorable effects in other BOPHs, including ones related to platelet activation (11-dehydrothromboxane B2) and metabolic syndrome (glucose), were more pronounced in normal weight subjects. CONCLUSIONS The results suggest that the reduced exposure demonstrated when switching to mTHS is associated with overall improvements in BOPHs, which are indicative of pathomechanistic pathways underlying the development of smoking-related diseases, with some stronger effects in normal weight subjects. IMPLICATIONS Switching to mTHS was associated with favorable changes for some BOPHs indicative of biological pathway alterations (eg, oxidative stress and endothelial dysfunction). The results suggest that switching to mTHS has the potential to reduce the adverse health effects of smoking and ultimately the risk of smoking-related diseases. Switching to mTHS for 90 days led to reductions in a number of biomarkers of exposure in smokers, relative to those who continued smoking cigarettes, which were close to those observed when stopping smoking (reported in our co-publication, Part 1). Initial findings suggest reduced levels of 8-epi-prostaglandin F2α and intercellular adhesion molecule 1, when switching to mTHS for 90 days. These changes are comparable to what is observed upon smoking cessation. In normal weight subjects, additional favorable changes were seen in 11-dehydrothromboxane B2, fibrinogen, homocysteine, hs-CRP, percentage of predicted forced expiratory volume in 1 second, systolic blood pressure, diastolic blood pressure, glucose, high-density lipoprotein, apolipoprotein A1, and triglycerides. TRIAL REGISTRATION NCT01989156.
Collapse
Affiliation(s)
- Christelle Haziza
- PMI Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | | | - Andrea Donelli
- PMI Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Dimitra Skiada
- PMI Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Valerie Poux
- PMI Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Rolf Weitkunat
- PMI Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Gizelle Baker
- PMI Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Patrick Picavet
- PMI Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Frank Lüdicke
- PMI Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| |
Collapse
|
25
|
Reumann MK, Schaefer J, Titz B, Aspera-Werz RH, Wong ET, Szostak J, Häussling V, Ehnert S, Leroy P, Tan WT, Kuczaj A, Audretsch C, Springer F, Badke A, Augat P, Quentanilla-Fend L, Martella M, Lee KM, Peitsch MC, Hoeng J, Nussler AK. E-vapor aerosols do not compromise bone integrity relative to cigarette smoke after 6-month inhalation in an ApoE -/- mouse model. Arch Toxicol 2020; 94:2163-2177. [PMID: 32409933 PMCID: PMC7303066 DOI: 10.1007/s00204-020-02769-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Cigarette smoke (CS) exposure is one of the leading risk factors for human health. Nicotine-containing inhalable products, such as e-cigarettes, can effectively support tobacco harm reduction approaches. However, there are limited comparative data on the effects of the aerosols generated from electronic vapor products (e-vapor) and CS on bone. Here, we report the effects of e-vapor aerosols and CS on bone morphology, structure, and strength in a 6-month inhalation study. Eight-week-old ApoE-/- mice were exposed to aerosols from three different e-vapor formulations-CARRIER (propylene glycol and vegetable glycerol), BASE (CARRIER and nicotine), TEST (BASE and flavor)-to CS from 3R4F reference cigarettes at matched nicotine concentrations (35 µg/L) or to fresh air (Sham) (N = 10 per group). Tibiae were analyzed for bone morphology by µCT imaging, biomechanics by three-point bending, and by histological analysis. CS inhalation caused a significant decrease in cortical and total bone volume fraction and bone density relative to e-vapor aerosols. Additionally, CS exposure caused a decrease in ultimate load and stiffness. In contrast, bone structural and biomechanical parameters were not significantly affected by e-vapor aerosol or Sham exposure. At the dissection time point, there was no significant difference in body weight or tibia bone weight or length among the groups. Histological findings revealed microcracks in cortical bone areas among all exposed groups compared to Sham control. In conclusion, because of the bone-preserving effect of e-vapor aerosols relative to CS exposure, e-vapor products could potentially constitute less harmful alternatives to cigarettes in situations in which bone health is of importance.
Collapse
Affiliation(s)
- Marie K Reumann
- Siegfried Weller Research Institute, BG Unfallklinik, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany
- Department of Trauma and Reconstructive Surgery, BG Unfallklinik, Eberhard Karls University, Tuebingen, Germany
| | - Jenny Schaefer
- Siegfried Weller Research Institute, BG Unfallklinik, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany
| | - Bjoern Titz
- PMI R&D, Phillip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchatel, Switzerland
| | - Romina H Aspera-Werz
- Siegfried Weller Research Institute, BG Unfallklinik, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany
| | - Ee Tsin Wong
- PMI R&D, Phillip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore
| | - Justyna Szostak
- PMI R&D, Phillip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchatel, Switzerland
| | - Victor Häussling
- Siegfried Weller Research Institute, BG Unfallklinik, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany
| | - Sabrina Ehnert
- Siegfried Weller Research Institute, BG Unfallklinik, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany
| | - Patrice Leroy
- PMI R&D, Phillip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchatel, Switzerland
| | - Wei Teck Tan
- PMI R&D, Phillip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore
| | - Arkadiusz Kuczaj
- PMI R&D, Phillip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchatel, Switzerland
| | - Christof Audretsch
- Department of Trauma and Reconstructive Surgery, BG Unfallklinik, Eberhard Karls University, Tuebingen, Germany
| | - Fabian Springer
- Department of Radiology, BG Unfallklinik Tuebingen and University Hospital Tuebingen, Tuebingen, Germany
- Institute for Biomechanics, BG Unfallklinik Murnau, Germany & Paracelsus Medical University, Salzburg, Austria
| | - Andreas Badke
- Department of Trauma and Reconstructive Surgery, BG Unfallklinik, Eberhard Karls University, Tuebingen, Germany
| | - Peter Augat
- Institute for Biomechanics, BG Unfallklinik Murnau, Germany & Paracelsus Medical University, Salzburg, Austria
| | | | - Manuela Martella
- Department of Pathology, Eberhard Karls University, Tuebingen, Germany
| | - K Monica Lee
- Altria Client Services LLC, 6601 West Broad Street, Richmond, VA, USA
| | - Manuel C Peitsch
- PMI R&D, Phillip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchatel, Switzerland
| | - Julia Hoeng
- PMI R&D, Phillip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchatel, Switzerland
| | - Andreas K Nussler
- Siegfried Weller Research Institute, BG Unfallklinik, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany.
| |
Collapse
|
26
|
Titz B, Szostak J, Sewer A, Phillips B, Nury C, Schneider T, Dijon S, Lavrynenko O, Elamin A, Guedj E, Tsin Wong E, Lebrun S, Vuillaume G, Kondylis A, Gubian S, Cano S, Leroy P, Keppler B, Ivanov NV, Vanscheeuwijck P, Martin F, Peitsch MC, Hoeng J. Multi-omics systems toxicology study of mouse lung assessing the effects of aerosols from two heat-not-burn tobacco products and cigarette smoke. Comput Struct Biotechnol J 2020; 18:1056-1073. [PMID: 32419906 PMCID: PMC7218232 DOI: 10.1016/j.csbj.2020.04.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 04/19/2020] [Indexed: 12/15/2022] Open
Abstract
Multi-omics systems toxicology study, comprising five omics data modalities. Multi-Omics Factor Analysis and multi-modality functional network interpretation. Cigarettes smoke (CS) induced complex immunoregulatory interactions across molecular layers. Aerosols from two heat-not-burn tobacco products had less impact on lungs than CS.
Cigarette smoke (CS) causes adverse health effects and, for smoker who do not quit, modified risk tobacco products (MRTPs) can be an alternative to reduce the risk of developing smoking-related diseases. Standard toxicological endpoints can lack sensitivity, with systems toxicology approaches yielding broader insights into toxicological mechanisms. In a 6-month systems toxicology study on ApoE−/− mice, we conducted an integrative multi-omics analysis to assess the effects of aerosols from the Carbon Heated Tobacco Product (CHTP) 1.2 and Tobacco Heating System (THS) 2.2—a potential and a candidate MRTP based on the heat-not-burn (HnB) principle—compared with CS at matched nicotine concentrations. Molecular exposure effects in the lungs were measured by mRNA/microRNA transcriptomics, proteomics, metabolomics, and lipidomics. Integrative data analysis included Multi-Omics Factor Analysis and multi-modality functional network interpretation. Across all five data modalities, CS exposure was associated with an increased inflammatory and oxidative stress response, and lipid/surfactant alterations. Upon HnB aerosol exposure these effects were much more limited or absent, with reversal of CS-induced effects upon cessation and switching to CHTP 1.2. Functional network analysis revealed CS-induced complex immunoregulatory interactions across the investigated molecular layers (e.g., itaconate, quinolinate, and miR-146) and highlighted the engagement of the heme–Hmox–bilirubin oxidative stress axis by CS. This work exemplifies how multi-omics approaches can be leveraged within systems toxicology studies and the generated multi-omics data set can facilitate the development of analysis methods and can yield further insights into the effects of toxicological exposures on the lung of mice.
Collapse
Key Words
- CHTP, Carbon Heated Tobacco Product
- COPD, chronic obstructive pulmonary disease
- CS, cigarette smoke
- Cigarette smoking
- Inhalation toxicology
- LC, liquid chromatography
- MOFA, Multi-Omics Factor Analysis
- MS, mass spectrometry
- Modified risk tobacco product (MRTP)
- Multi-omics
- PCSF, prize-collecting Steiner forest
- ROS, reactive oxygen species
- Systems toxicology
- THS, Tobacco Heating System
- cMRTP, candidate modified risk tobacco product
- sGCCA, sparse generalized canonical correlation analysis
Collapse
Affiliation(s)
- Bjoern Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Justyna Szostak
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Alain Sewer
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Blaine Phillips
- Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Catherine Nury
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Thomas Schneider
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Sophie Dijon
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Oksana Lavrynenko
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Ashraf Elamin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Ee Tsin Wong
- Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Stefan Lebrun
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Grégory Vuillaume
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Athanasios Kondylis
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Sylvain Gubian
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Stephane Cano
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | | | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | | | - Florian Martin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| |
Collapse
|
27
|
Ekroos K, Lavrynenko O, Titz B, Pater C, Hoeng J, Ivanov NV. Lipid-based biomarkers for CVD, COPD, and aging - A translational perspective. Prog Lipid Res 2020; 78:101030. [PMID: 32339553 DOI: 10.1016/j.plipres.2020.101030] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/23/2020] [Accepted: 03/16/2020] [Indexed: 12/11/2022]
Abstract
For many diseases, there is an unmet need for new or better biomarkers for improved disease risk assessment and monitoring, as available markers lack sufficient specificity. Lipids are drawing major interest as potential candidates for filling these gaps. This has recently been demonstrated by the identification of selective ceramides for prediction of cardiovascular mortality, enabling improved risk assessment of cardiovascular disease compared with conventional clinical markers. In this review, we discuss current lipid biomarker findings and the possible connection between cardiovascular disease, chronic obstructive pulmonary disease, and aging. Moreover, we discuss how to overcome the current roadblocks facing lipid biomarker research. We stress the need for improved quantification, standardization of methodologies, and establishment of initial reference values to allow for an efficient transfer path of research findings into the clinical landscape, and, ultimately, to put newly identified biomarkers into practical use.
Collapse
Affiliation(s)
- Kim Ekroos
- Lipidomics Consulting Ltd., Irisviksvägen 31D, 02230 Esbo, Finland.
| | - Oksana Lavrynenko
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| | - Calin Pater
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| |
Collapse
|
28
|
Ito S, Matsumura K, Ishimori K, Ishikawa S. In vitro long-term repeated exposure and exposure switching of a novel tobacco vapor product in a human organotypic culture of bronchial epithelial cells. J Appl Toxicol 2020; 40:1248-1258. [PMID: 32319113 PMCID: PMC7496418 DOI: 10.1002/jat.3982] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 12/16/2022]
Abstract
Next‐generation tobacco products and nicotine delivery systems such as heat‐not‐burn tobacco products and electronic cigarettes, the usage of which is expected to have a beneficial impact on public health, have gained popularity over the past decade. However, the risks associated with the long‐term use of such products are still incompletely understood. Although the risks of these products should be clarified through epidemiological studies, such studies are normally performed based on each product category, not product‐by‐product. Therefore, investigation of the risk on a product‐by‐product basis is important to provide specific scientific evidence. In the current study, we performed the 40‐day repeated exposure of in vitro human bronchial epithelial tissues to cigarette smoke (CS) or vapor from our proprietary novel tobacco vapor product (NTV). In addition, tissue samples exposed to CS were switched to NTV or CS exposure was stopped at 20 days to reflect a situation where smokers switched to NTV or ceased to smoke. All tissue samples were assessed in terms of toxicity, inflammation and transcriptomic alterations. Tissue samples switched to NTV and the cessation of exposure samples showed recovery from CS‐induced damage although there was a time‐course difference. Moreover, repeated exposure to NTV produced negligible effects on the tissue samples while CS produced cumulative effects. Our results suggest that the use of NTV, including switching to NTV from cigarette smoking, has fewer effects on bronchial epithelial tissues than continuing smoking. We carried out the 40‐day repeated exposure of in vitro bronchial epithelial tissues to cigarette smoke (CS) or vapor from novel tobacco vapor product (NTV) and intermediate switching from CS exposure to NTV exposure. Long‐term exposure to NTV resulted in negligible effect on the tissues. Moreover, the tissues that intermediately switched to NTV exposure showed recovery from CS‐induced damage similar to exposure cessation. These results implied that exposure to NTV showed few effects on bronchial epithelial tissues.
Collapse
Affiliation(s)
- Shigeaki Ito
- Scientific Product Assessment Center, Japan Tobacco Inc., Yokohama, Japan
| | - Kazushi Matsumura
- Scientific Product Assessment Center, Japan Tobacco Inc., Yokohama, Japan
| | - Kanae Ishimori
- Scientific Product Assessment Center, Japan Tobacco Inc., Yokohama, Japan
| | - Shinkichi Ishikawa
- Scientific Product Assessment Center, Japan Tobacco Inc., Yokohama, Japan
| |
Collapse
|
29
|
Choukrallah MA, Hoeng J, Peitsch MC, Martin F. Lung transcriptomic clock predicts premature aging in cigarette smoke-exposed mice. BMC Genomics 2020; 21:291. [PMID: 32272900 PMCID: PMC7147004 DOI: 10.1186/s12864-020-6712-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 03/31/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lung aging is characterized by a number of structural alterations including fibrosis, chronic inflammation and the alteration of inflammatory cell composition. Chronic exposure to cigarette smoke (CS) is known to induce similar alterations and may contribute to premature lung aging. Additionally, aging and CS exposure are associated with transcriptional alterations in the lung. The current work aims to explore the interaction between age- and CS- associated transcriptomic perturbations and develop a transcriptomic clock able to predict the biological age and the impact of external factors on lung aging. RESULTS Our investigations revealed a substantial overlap between transcriptomic response to CS exposure and age-related transcriptomic alterations in the murine lung. Of particular interest is the strong upregulation of immunoglobulin genes with increased age and in response to CS exposure, indicating an important implication of B-cells in lung inflammation associated with aging and smoking. Furthermore, we used a machine learning approach based on Lasso regression to build a transcriptomic age model that can accurately predict chronological age in untreated mice and the deviations associated with certain exposures. Interestingly, CS-exposed-mice were predicted to be prematurely aged in contrast to mice exposed to fresh air or to heated tobacco products (HTPs). The accelerated aging rate associated with CS was reversed upon smoking cessation or switching to HTPs. Additionally, our model was able to predict premature aging associated with thoracic irradiation from an independent public dataset. CONCLUSIONS Aging and CS exposure share common transcriptional alteration patterns in the murine lung. The massive upregulation of B-cell restricted genes during these processes shed light on the contribution of cell composition and particularly immune cells to the measured transcriptomic signal. Through machine learning approach, we show that gene expression changes can be used to accurately monitor the biological age and the modulations associated with certain exposures. Our findings also suggest that the premature lung aging is reversible upon the reduction of harmful exposures.
Collapse
Affiliation(s)
| | - Julia Hoeng
- Philip Morris International R&D, Quai Jeanrenaud 5, 2003, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- Philip Morris International R&D, Quai Jeanrenaud 5, 2003, Neuchâtel, Switzerland
| | - Florian Martin
- Philip Morris International R&D, Quai Jeanrenaud 5, 2003, Neuchâtel, Switzerland
| |
Collapse
|
30
|
Szostak J, Wong ET, Titz B, Lee T, Wong SK, Low T, Lee KM, Zhang J, Kumar A, Schlage WK, Guedj E, Phillips B, Leroy P, Buettner A, Xiang Y, Martin F, Sewer A, Kuczaj A, Ivanov NV, Luettich K, Vanscheeuwijck P, Peitsch MC, Hoeng J. A 6-month systems toxicology inhalation study in ApoE -/- mice demonstrates reduced cardiovascular effects of E-vapor aerosols compared with cigarette smoke. Am J Physiol Heart Circ Physiol 2020; 318:H604-H631. [PMID: 31975625 DOI: 10.1152/ajpheart.00613.2019] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Smoking cigarettes is harmful to the cardiovascular system. Considerable attention has been paid to the reduced harm potential of alternative nicotine-containing inhalable products such as e-cigarettes. We investigated the effects of E-vapor aerosols or cigarette smoke (CS) on atherosclerosis progression, cardiovascular function, and molecular changes in the heart and aorta of female apolipoprotein E-deficient (ApoE-/-) mice. The mice were exposed to aerosols from three different E-vapor formulations: 1) carrier (propylene glycol and vegetable glycerol), 2) base (carrier and nicotine), or 3) test (base and flavor) or to CS from 3R4F reference cigarettes for up to 6 mo. Concentrations of CS and base or test aerosols were matched at 35 µg nicotine/L. Exposure to CS, compared with sham-exposed fresh air controls, accelerated atherosclerotic plaque formation, whereas no such effect was seen for any of the three E-vapor aerosols. Molecular changes indicated disease mechanisms related to oxidative stress and inflammation in general, plus changes in calcium regulation, and altered cytoskeletal organization and microtubule dynamics in the left ventricle. While ejection fraction, fractional shortening, cardiac output, and isovolumic contraction time remained unchanged following E-vapor aerosols exposure, the nicotine-containing base and test aerosols caused an increase in isovolumic relaxation time similar to CS. A nicotine-related increase in pulse wave velocity and arterial stiffness was also observed, but it was significantly lower for base and test aerosols than for CS. These results demonstrate that in comparison with CS, E-vapor aerosols induce substantially lower biological responses associated with smoking-related cardiovascular diseases.NEW & NOTEWORTHY Analysis of key urinary oxidative stress markers and proinflammatory cytokines showed an absence of oxidative stress and inflammation in the animals exposed to E-vapor aerosols. Conversely, animals exposed to conventional cigarette smoke had high urinary levels of these markers. When compared with conventional cigarette smoke, E-vapor aerosols induced smaller atherosclerotic plaque surface area and volume. Systolic and diastolic cardiac function, as well as endothelial function, were further significantly less affected by electronic cigarette aerosols than conventional cigarette smoke. Molecular analysis demonstrated that E-vapor aerosols induce significantly smaller transcriptomic dysregulation in the heart and aorta compared with conventional cigarette smoke.
Collapse
Affiliation(s)
- Justyna Szostak
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Ee Tsin Wong
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Bjoern Titz
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Tom Lee
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Sin Kei Wong
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Tiffany Low
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | | | | | | | | | - Emmanuel Guedj
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Blaine Phillips
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Patrice Leroy
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | | | - Yang Xiang
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Florian Martin
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Alain Sewer
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Arkadiusz Kuczaj
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Nikolai V Ivanov
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Karsta Luettich
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Patrick Vanscheeuwijck
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Julia Hoeng
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| |
Collapse
|
31
|
Boué S, Goedertier D, Hoeng J, Kuczaj A, Majeed S, Mathis C, May A, Phillips B, Peitsch MC, Radtke F, Schlage WK, Tan WT, Vanscheeuwijck P. State-of-the-art methods and devices for the generation, exposure, and collection of aerosols from heat-not-burn tobacco products. TOXICOLOGY RESEARCH AND APPLICATION 2020. [DOI: 10.1177/2397847319897869] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tobacco harm reduction is increasingly recognized as a promising approach to accelerate the decline in smoking prevalence and smoking-related population harm. Potential modified risk tobacco products (MRTPs) must undergo a rigorous premarket toxicological risk assessment. The ability to reproducibly generate, collect, and use aerosols is critical for the characterization, and preclinical assessment of aerosol-based candidate MRTPs (cMRTPs), such as noncombusted cigarettes, also referred to as heated tobacco products, tobacco heating products, or heat-not-burn (HNB) tobacco products. HNB tobacco products generate a nicotine-containing aerosol by heating tobacco instead of burning it. The aerosols generated by HNB products are qualitatively and quantitatively highly different from cigarette smoke (CS). This constitutes technical and experimental challenges comparing the toxicity of HNB aerosols with CS. The methods and experimental setups that have been developed for the study of CS cannot be directly transposed to the study of HNB aerosols. Significant research efforts are dedicated to the development, characterization, and validation of experimental setups and methods suitable for HNB aerosols. They are described in this review, with a particular focus on the Tobacco Heating System version 2.2. This is intended to support further studies, the objective evaluation and verification of existing evidence, and the development of scientifically substantiated HNB MRTPs.
Collapse
Affiliation(s)
- Stéphanie Boué
- Philip Morris International (PMI) Research & Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Didier Goedertier
- Philip Morris International (PMI) Research & Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Julia Hoeng
- Philip Morris International (PMI) Research & Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Arkadiusz Kuczaj
- Philip Morris International (PMI) Research & Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Shoaib Majeed
- Philip Morris International (PMI) Research & Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Carole Mathis
- Philip Morris International (PMI) Research & Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Anne May
- Consultants in Science, Epalinges, Switzerland
| | - Blaine Phillips
- Philip Morris International (PMI) Research & Development, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore
| | - Manuel C Peitsch
- Philip Morris International (PMI) Research & Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Falk Radtke
- Philip Morris International (PMI) Research & Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | | | - Wei Teck Tan
- Philip Morris International (PMI) Research & Development, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore
| | - Patrick Vanscheeuwijck
- Philip Morris International (PMI) Research & Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| |
Collapse
|
32
|
Szostak J, Titz B, Schlage WK, Guedj E, Sewer A, Phillips B, Leroy P, Buettner A, Neau L, Trivedi K, Martin F, Ivanov NV, Vanscheeuwijck P, Peitsch MC, Hoeng J. Structural, functional, and molecular impact on the cardiovascular system in ApoE -/- mice exposed to aerosol from candidate modified risk tobacco products, Carbon Heated Tobacco Product 1.2 and Tobacco Heating System 2.2, compared with cigarette smoke. Chem Biol Interact 2020; 315:108887. [PMID: 31705857 DOI: 10.1016/j.cbi.2019.108887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/26/2022]
Abstract
AIM To investigate the molecular, structural, and functional impact of aerosols from candidate modified risk tobacco products (cMRTP), the Carbon Heated Tobacco Product (CHTP) 1.2 and Tobacco Heating System (THS) 2.2, compared with that of mainstream cigarette smoke (CS) on the cardiovascular system of ApoE-/- mice. METHODS Female ApoE-/- mice were exposed to aerosols from THS 2.2 and CHTP 1.2 or to CS from the 3R4F reference cigarette for up to 6 months at matching nicotine concentrations. A Cessation and a Switching group (3 months exposure to 3R4F CS followed by filtered air or CHTP 1.2 for 3 months) were included. Cardiovascular effects were investigated by echocardiographic, histopathological, immunohistochemical, and transcriptomics analyses. RESULTS Continuous exposure to cMRTP aerosols did not affect atherosclerosis progression, heart function, left ventricular (LV) structure, or the cardiovascular transcriptome. Exposure to 3R4F CS triggered atherosclerosis progression, reduced systolic ejection fraction and fractional shortening, caused heart LV hypertrophy, and initiated significant dysregulation in the transcriptomes of the heart ventricle and thoracic aorta. Importantly, the structural, functional, and molecular changes caused by 3R4F CS were improved in the smoking cessation and switching groups. CONCLUSION Exposure to cMRTP aerosols lacked most of the CS exposure-related functional, structural, and molecular effects. Smoking cessation or switching to CHTP 1.2 aerosol caused similar recovery from the 3R4F CS effects in the ApoE-/- model, with no further acceleration of plaque progression beyond the aging-related rate.
Collapse
Affiliation(s)
- Justyna Szostak
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Walter K Schlage
- Biology Consultant, Max-Baermann-Str. 21, 51429, Bergisch Gladbach, Germany.
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Alain Sewer
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Blaine Phillips
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | | | - Laurent Neau
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Keyur Trivedi
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Florian Martin
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Patrick Vanscheeuwijck
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| |
Collapse
|
33
|
Reyes-Caballero H, Park B, Loube J, Sanchez I, Vinayachandran V, Choi Y, Woo J, Edwards J, Brinkman MC, Sussan T, Mitzner W, Biswal S. Immune modulation by chronic exposure to waterpipe smoke and immediate-early gene regulation in murine lungs. Tob Control 2019; 29:s80-s89. [PMID: 31852817 DOI: 10.1136/tobaccocontrol-2019-054965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/22/2019] [Accepted: 08/24/2019] [Indexed: 01/02/2023]
Abstract
OBJECTIVE We investigated the effects of chronic waterpipe (WP) smoke on pulmonary function and immune response in a murine model using a research-grade WP and the effects of acute exposure on the regulation of immediate-early genes (IEGs). METHODS WP smoke was generated using three WP smoke puffing regimens based on the Beirut regimen. WP smoke samples generated under these puffing regimens were quantified for nicotine concentration. Mice were chronically exposed for 6 months followed by assessment of pulmonary function and airway inflammation. Transcriptomic analysis using RNAseq was conducted after acute exposure to characterise the IEG response. These biomarkers were then compared with those generated after exposure to dry smoke (without water added to the WP bowl). RESULTS We determined that nicotine composition in WP smoke ranged from 0.4 to 2.5 mg per puffing session. The lung immune response was sensitive to the incremental severity of chronic exposure, with modest decreases in airway inflammatory cells and chemokine levels compared with air-exposed controls. Pulmonary function was unmodified by chronic WP exposure. Acute WP exposure was found to activate the immune response and identified known and novel IEG as potential biomarkers of WP exposure. CONCLUSION Chronic exposure to WP smoke leads to immune suppression without significant changes to pulmonary function. Transcriptomic analysis of the lung after acute exposure to WP smoke showed activation of the immune response and revealed IEGs that are common to WP and dry smoke, as well as pools of IEGs unique to each exposure, identifying potential biomarkers specific to WP exposure.
Collapse
Affiliation(s)
- Hermes Reyes-Caballero
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bongsoo Park
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jeffrey Loube
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ian Sanchez
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vinesh Vinayachandran
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Youngshim Choi
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Juhyung Woo
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Justin Edwards
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Thomas Sussan
- Toxicology Directorate, US Army Public Health Command, Aberdeen Proving Ground, Maryland, USA
| | - Wayne Mitzner
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shyam Biswal
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
34
|
Mallock N, Pieper E, Hutzler C, Henkler-Stephani F, Luch A. Heated Tobacco Products: A Review of Current Knowledge and Initial Assessments. Front Public Health 2019; 7:287. [PMID: 31649912 PMCID: PMC6795920 DOI: 10.3389/fpubh.2019.00287] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/20/2019] [Indexed: 01/29/2023] Open
Abstract
The health risks of tobacco smoking have been documented in numerous studies and smoking rates have declined in developed countries over the last 50 years. Today, we know that cigarette smoking is the major cause of preventable deaths due to tobacco smoke induced diseases. As a consequence of an increased awareness of smoking-related health risks, heated tobacco products (HTPs) are marketed as reduced toxicant alternatives to conventional tobacco products. Manufacturers claim that levels of toxicants and hazardous compounds are significantly reduced, implying that inhalation of the modified aerosol is less harmful compared to conventional cigarettes. In this manuscript, previous assessments of HTPs are briefly summarized, including a short discussion on challenges with the adaption of standard analytical methods used for tobacco smoke. The reliability of analytical data is important for risk assessment approaches that are based on reduced toxicant exposure. In order to assess a putative reduction of health risks, an integrated study design is required that should include clinical studies and epidemiology data. One manufacturer applied for a classification as a Modified Risk Tobacco Product (MRTP) in the United States, based on extensive toxicological studies that have also been published. However, data are not yet sufficient for a reliable assessment or recognition of putatively reduced health risks. Challenges regarding a classification in Europe are also discussed briefly in this review.
Collapse
Affiliation(s)
- Nadja Mallock
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Elke Pieper
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Christoph Hutzler
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Frank Henkler-Stephani
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Andreas Luch
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| |
Collapse
|
35
|
Martin F, Gubian S, Talikka M, Hoeng J, Peitsch MC. NPA: an R package for computing network perturbation amplitudes using gene expression data and two-layer networks. BMC Bioinformatics 2019; 20:451. [PMID: 31481014 PMCID: PMC6724309 DOI: 10.1186/s12859-019-3016-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
Background High-throughput gene expression technologies provide complex datasets reflecting mechanisms perturbed in an experiment, typically in a treatment versus control design. Analysis of these information-rich data can be guided based on a priori knowledge, such as networks of related proteins or genes. Assessing the response of a specific mechanism and investigating its biological basis is extremely important in systems toxicology; as compounds or treatment need to be assessed with respect to a predefined set of key mechanisms that could lead to toxicity. Two-layer networks are suitable for this task, and a robust computational methodology specifically addressing those needs was previously published. The NPA package (https://github.com/philipmorrisintl/NPA) implements the algorithm, and a data package of eight two-layer networks representing key mechanisms, such as xenobiotic metabolism, apoptosis, or epithelial immune innate activation, is provided. Results Gene expression data from an animal study are analyzed using the package and its network models. The functionalities are implemented using R6 classes, making the use of the package seamless and intuitive. The various network responses are analyzed using the leading node analysis, and an overall perturbation, called the Biological Impact Factor, is computed. Conclusions The NPA package implements the published network perturbation amplitude methodology and provides a set of two-layer networks encoded in the Biological Expression Language.
Collapse
Affiliation(s)
- Florian Martin
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Sylvain Gubian
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Marja Talikka
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| |
Collapse
|
36
|
Hoeng J, Maeder S, Vanscheeuwijck P, Peitsch MC. Assessing the lung cancer risk reduction potential of candidate modified risk tobacco products. Intern Emerg Med 2019; 14:821-834. [PMID: 30767158 PMCID: PMC6722152 DOI: 10.1007/s11739-019-02045-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/30/2019] [Indexed: 12/19/2022]
Abstract
Smoking is the major cause of lung cancer. While the risk of lung cancer increases with the number of cigarettes smoked and the duration of smoking, it also decreases upon smoking cessation. The development of candidate modified risk tobacco products (cMRTP) is aimed at providing smokers who will not quit with alternatives to cigarettes that present less risk of harm and smoking-related disease. It is necessary to assess the risk reduction potential of cMRTPs, including their potential to reduce the risk of lung cancer. Assessing the lung cancer risk reduction potential of cMRTPs is hampered by (i) the absence of clinical risk markers that are predictive of future lung cancer development, (ii) the latency of lung cancer manifestation (decades of smoking), and (iii) the slow reduction in excess risk upon cessation and a fortiori upon switching to a cMRTP. It is, therefore, likely that only long-term epidemiology will provide definitive answers to this question and allow to first verify that a cMRTP reduces the risk of lung cancer and if it does, to quantify the reduction in excess lung cancer risk associated with a cMRTP. For this to be possible, the cMRTP would need to be available in the market and used exclusively by a large portion of current smokers. Here, we propose that a mechanism-based approach represents a solid alternative to show in a pre-market setting that switching to a cMRTP is likely to significantly reduce the risk of lung cancer. This approach is based on the causal chain of events that leads from smoking to disease and leverages both non-clinical and clinical studies as well as the principles of systems toxicology. We also discuss several important challenges inherent to the assessment of cMRTPs as well as key aspects regarding product use behavior.
Collapse
Affiliation(s)
- Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Serge Maeder
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | | | - Manuel C. Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| |
Collapse
|
37
|
Lüdicke F, Picavet P, Baker G, Haziza C, Poux V, Lama N, Weitkunat R. Effects of Switching to the Tobacco Heating System 2.2 Menthol, Smoking Abstinence, or Continued Cigarette Smoking on Biomarkers of Exposure: A Randomized, Controlled, Open-Label, Multicenter Study in Sequential Confinement and Ambulatory Settings (Part 1). Nicotine Tob Res 2019; 20:161-172. [PMID: 28177489 PMCID: PMC5896533 DOI: 10.1093/ntr/ntw287] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 10/25/2016] [Indexed: 12/02/2022]
Abstract
Introduction The menthol Tobacco Heating System 2.2 (mTHS) is a newly developed candidate modified-risk tobacco product intended to reduce exposure to the harmful and potentially harmful constituents (HPHCs) of conventional cigarette (CC) smoke. This study examined the impact of switching to mTHS on biomarkers of exposure to HPHCs relative to menthol CCs (mCCs) and smoking abstinence (SA). Methods In this three-arm, parallel-group study, 160 Japanese adult smokers (23–65 years; smoking ≥10 mCCs per day) were randomized to mTHS (n = 78), mCC (n = 42), or SA (n = 40) for 5 days in confinement and 85 days in ambulatory settings. Endpoints included biomarkers of exposure to HPHCs, human puffing topography, safety, and subjective effects of smoking measures. Results After 5 days of product use, the concentrations of carboxyhemoglobin, 3-hydroxypropylmercapturic acid, monohydroxybutenyl mercapturic acid, and S-phenylmercapturic acid were 55%, 49%, 87%, and 89% lower (p < .001), respectively, in the mTHS group than in the mCC group. Other biomarkers of exposure (measured as secondary endpoints) were 50%–94% lower in the mTHS group than in the mCC group on day 5. These reductions in the mTHS group were maintained at day 90, similar to the SA group. Switching to mTHS was associated with changes in human puffing topography (shorter puff intervals and more frequent puffs). The urge-to-smoke and smoking satisfaction levels on day 90 were similar in the mTHS and the mCC groups. Conclusion Switching from mCCs to mTHS significantly reduced exposure to HPHCs relative to continuing smoking mCCs with concentrations similar to those observed following SA in Japanese adult smokers. Implications This randomized study compared the impact of switching to a modified-risk tobacco product candidate mTHS on biomarkers of exposure to HPHCs of cigarette smoke relative to continuing smoking cigarettes or abstaining from smoking in sequential confinement and ambulatory settings. The study showed that switching to mTHS was associated with significant biomarker reductions within 5 days in confinement, these reductions being maintained throughout the ambulatory setting up to day 90. The results provide evidence that switching to mTHS reduces real-life exposure to HPHCs in adult smokers.
Collapse
Affiliation(s)
- Frank Lüdicke
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Patrick Picavet
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Gizelle Baker
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Christelle Haziza
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Valerie Poux
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Nicola Lama
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Rolf Weitkunat
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| |
Collapse
|
38
|
Talikka M, Belcastro V, Gubian S, Martin F, Peitsch MC, Hoeng J. Systems toxicology meta-analysis—From aerosol exposure to nanotoxicology. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
39
|
Boué S, Schlage WK, Page D, Hoeng J, Peitsch MC. Toxicological assessment of Tobacco Heating System 2.2: Findings from an independent peer review. Regul Toxicol Pharmacol 2019; 104:115-127. [PMID: 30878573 DOI: 10.1016/j.yrtph.2019.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/12/2019] [Accepted: 03/09/2019] [Indexed: 01/09/2023]
Abstract
Offering safer alternatives to cigarettes, such as e-cigarettes and heated tobacco products, to smokers who are not willing to quit could reduce the harm caused by smoking. Extensive and rigorous scientific studies are conducted to assess the relative risk of such potentially modified risk tobacco products compared with that of smoking cigarettes. In addition to the peer review of publications reporting individual studies, we aimed to gauge the plausibility of the evidence to the scientific community and appreciate likely necessary additions prior to regulatory submission. Therefore, we sponsored a two-tier peer review organized by an independent third party who identified, recruited, and managed 7 panels of 5-12 experts whose identity remains unknown to us. The reviewers had access to all publications and raw data from preclinical and clinical studies via a web portal. The reviewers were asked questions regarding study design, methods, quality of data, and interpretation of results to judge the validity of the conclusions regarding the relative effects of the Tobacco Heating System 2.2 compared with cigarettes. Once their conclusions were submitted, the experts had the opportunity to participate in an anonymized online debate with their fellow panel members. We present here the results obtained from this innovative peer review effort which revealed supportive or very supportive of the study methods and results, and support the robustness of the studies and validity of the conclusions.
Collapse
Affiliation(s)
- Stéphanie Boué
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Walter K Schlage
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - David Page
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| |
Collapse
|
40
|
Queloz S, Etter JF. An online survey of users of tobacco vaporizers, reasons and modes of utilization, perceived advantages and perceived risks. BMC Public Health 2019; 19:642. [PMID: 31133009 PMCID: PMC6537171 DOI: 10.1186/s12889-019-6957-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/08/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Tobacco vaporizers heat tobacco without burning it, to produce an inhalable aerosol. Various models have recently appeared on the market, mostly manufactured by the tobacco industry, but few of the studies published on tobacco vaporizers are independent from the manufacturers. The goals of this study were to explore who uses tobacco vaporizers, how these products are used, reasons for utilization, perceived advantages and risks. METHODS Online questionnaire collected from October 2016 to January 2018 in self-selected visitors aged > 18 to an anti-addiction website. RESULTS We obtained 170 valid responses, of whom 104 were using tobacco vaporizers. For homogeneity, we included only the 102 users of the Brand 1 tobacco vaporizer in our analysis, as there were only two users of other vaporizers. Among these 102 vaporizer users, about half were current cigarette smokers (57%), the rest were former cigarette smokers. The median age was 41, and the median duration of utilization was 9 months. Most (88%) used the vaporizer daily, 8% were occasional users and 4% were past users. Among current smokers, 80% were currently trying to reduce their cigarette consumption and 29% were trying to quit. The vaporizer was used mainly to replace cigarettes (94%), because it was perceived to be less toxic than cigarettes (89%), to help stop smoking or to avoid starting smoking again (72%), or to reduce cigarette consumption (71%). Current smokers who were daily or occasional vaporizer users reported smoking a median of 8.0 cigarettes per day, compared with 20.0 per day before they started to use the vaporizer (p < .0001, Wilcoxon signed-rank test). CONCLUSIONS In this online sample of early adopters, Brand 1 was by far the most frequently used tobacco vaporizer. It was used by current or former smokers only, mainly to replace cigarettes, and satisfaction ratings were good. Users considered the tobacco vaporizer to be less toxic than cigarette smoke and perceived it to be helpful for reducing or stopping smoking.
Collapse
Affiliation(s)
- Sébastien Queloz
- Institute of Global Health, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-François Etter
- Institute of Global Health, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
41
|
Phillips B, Szostak J, Titz B, Schlage WK, Guedj E, Leroy P, Vuillaume G, Martin F, Buettner A, Elamin A, Sewer A, Sierro N, Choukrallah MA, Schneider T, Ivanov NV, Teng C, Tung CK, Lim WT, Yeo YS, Vanscheeuwijck P, Peitsch MC, Hoeng J. A six-month systems toxicology inhalation/cessation study in ApoE -/- mice to investigate cardiovascular and respiratory exposure effects of modified risk tobacco products, CHTP 1.2 and THS 2.2, compared with conventional cigarettes. Food Chem Toxicol 2019; 126:113-141. [PMID: 30763686 DOI: 10.1016/j.fct.2019.02.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/24/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023]
Abstract
Smoking is one of the major modifiable risk factors in the development and progression of chronic obstructive pulmonary disease (COPD) and cardiovascular disease (CVD). Modified-risk tobacco products (MRTP) are being developed to provide substitute products for smokers who are unable or unwilling to quit, to lessen the smoking-related health risks. In this study, the ApoE-/- mouse model was used to investigate the impact of cigarette smoke (CS) from the reference cigarette 3R4F, or aerosol from two potential MRTPs based on the heat-not-burn principle, carbon heated tobacco product 1.2 (CHTP1.2) and tobacco heating system 2.2 (THS 2.2), on the cardiorespiratory system over a 6-month period. In addition, cessation or switching to CHTP1.2 after 3 months of CS exposure was assessed. A systems toxicology approach combining physiology, histology and molecular measurements was used to evaluate the impact of MRTP aerosols in comparison to CS. CHTP1.2 and THS2.2 aerosols, compared with CS, demonstrated lower impact on the cardiorespiratory system, including low to absent lung inflammation and emphysematous changes, and reduced atherosclerotic plaque formation. Molecular analyses confirmed the lower engagement of pathological mechanisms by MRTP aerosols than CS. Both cessation and switching to CHTP1.2 reduced the observed CS effects to almost sham exposure levels.
Collapse
Affiliation(s)
- Blaine Phillips
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Justyna Szostak
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | | | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Gregory Vuillaume
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Florian Martin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | | | - Ashraf Elamin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Alain Sewer
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Nicolas Sierro
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | | | - Thomas Schneider
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Charles Teng
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Ching Keong Tung
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Wei Ting Lim
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Ying Shan Yeo
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Patrick Vanscheeuwijck
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| |
Collapse
|
42
|
Lee KM, Hoeng J, Harbo S, Kogel U, Gardner W, Oldham M, Benson E, Talikka M, Kondylis A, Martin F, Titz B, Ansari S, Trivedi K, Guedj E, Elamin A, Ivanov NV, Vanscheeuwijck P, Peitsch MC, McKinney WJ. Biological changes in C57BL/6 mice following 3 weeks of inhalation exposure to cigarette smoke or e-vapor aerosols. Inhal Toxicol 2019; 30:553-567. [DOI: 10.1080/08958378.2019.1576807] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A, Neuchâtel, Switzerland
| | | | - Ulrike Kogel
- PMI R&D, Philip Morris Products S.A, Neuchâtel, Switzerland
| | | | | | | | - Marja Talikka
- PMI R&D, Philip Morris Products S.A, Neuchâtel, Switzerland
| | | | - Florian Martin
- PMI R&D, Philip Morris Products S.A, Neuchâtel, Switzerland
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A, Neuchâtel, Switzerland
| | - Sam Ansari
- PMI R&D, Philip Morris Products S.A, Neuchâtel, Switzerland
| | - Keyur Trivedi
- PMI R&D, Philip Morris Products S.A, Neuchâtel, Switzerland
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A, Neuchâtel, Switzerland
| | - Ashraf Elamin
- PMI R&D, Philip Morris Products S.A, Neuchâtel, Switzerland
| | | | | | | | | |
Collapse
|
43
|
Choukrallah MA, Sierro N, Martin F, Baumer K, Thomas J, Ouadi S, Hoeng J, Peitsch MC, Ivanov NV. Tobacco Heating System 2.2 has a limited impact on DNA methylation of candidate enhancers in mouse lung compared with cigarette smoke. Food Chem Toxicol 2019; 123:501-510. [PMID: 30414959 DOI: 10.1016/j.fct.2018.11.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/19/2018] [Accepted: 11/07/2018] [Indexed: 01/17/2023]
Abstract
Cigarette smoke (CS) exposure has been shown to correlate with changes in DNA methylation levels, however, the impact of CS on DNA methylation at genome-wide scale is missing. Here, we used whole-genome bisulfite sequencing to assess the effects of CS extract and aerosol from the Tobacco Heating System (THS) 2.2, a candidate modified risk tobacco product, on DNA methylation in lung and liver tissues from apolipoprotein E-deficient mice during an eight-month period of exposure. We found that in lung tissue, CS mainly induced hypermethylation of candidate enhancers at late time points, while promoters were less affected. This effect was strongly reduced upon cessation or switching to THS 2.2. By contrast, chronic exposure to THS 2.2 had a limited effect on DNA methylation at both promoters and enhancers. We also identified members of the Ets and Fox families of transcription factors as potential players in the epigenetic response to CS exposure in lung tissue. In contrast to the lung, DNA methylation in the liver was largely insensitive to all investigated exposures. In summary, our investigations indicate that CS-related DNA methylation alterations are tissue-specific, occur mainly at enhancers and are strongly reduced upon smoking cessation or switching to THS2.2.
Collapse
Affiliation(s)
| | - Nicolas Sierro
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Florian Martin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Karine Baumer
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Jerome Thomas
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Sonia Ouadi
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| |
Collapse
|
44
|
Dautzenberg B, Dautzenberg MD. [Systematic analysis of the scientific literature on heated tobacco]. Rev Mal Respir 2019; 36:82-103. [PMID: 30429092 DOI: 10.1016/j.rmr.2018.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/26/2018] [Indexed: 10/27/2022]
Abstract
INTRODUCTION The tobacco industry (TI) reports that heated tobacco reduces risk of tobacco use and will replace cigarettes. An analysis of the scientific literature was conducted in order to enlighten professionals and decision-makers. METHOD After a Medline query in February 2018, a systematic analysis was conducted. RESULTS Of the 100 papers published in 2008-2018, 75 have authors affiliated or linked to TI. Emissions contain gases, droplets and solid particles, so are smokes. The main products are: THS2.2 (Iqos®) which heats mini-cigarettes at 340°C, the THP1.0 (Glo®) which heats at 240°C sticks delivering about half as much nicotine, Ploom® which uses reconstituted tobacco microcapsules heated at 180°C. Under the experimental conditions, there is a reduction of toxic emissions and biological effects, but the expected risk reduction is not demonstrated. Symptoms related to passive smoking are described. The 4 epidemiological articles report that heated tobacco is used in 10 to 45% of cases by non-smokers and demonstrate the effectiveness of TI promotion campaigns. Thus, the THS2.2 is more a gateway to smoking (20%) than an exit door (11%); moreover, it is not expected risk reduction among the 69% who are mixed users. CONCLUSIONS While reducing emissions is documented, reducing the risk to the smoker who switches to heated-tobacco remains to be demonstrated. On the other hand, the worsening of the global tobacco risk related to the promotion of the products by the TI is anticipated, justifying that the authorities take the appropriate measures to control the promotion of heated tobacco.
Collapse
Affiliation(s)
- B Dautzenberg
- Service de pharmacologie, Pitié-Salpêtrière, AP-HP, 75013 Paris, France; Consultation de médecine, hôpital Marmottan, 75017 Paris, France; Consultation de tabacologie, institut Arthur-Vernes, 75006 Paris, France; Paris sans tabac, 14, avenue Bosquet, 75007 Paris, France.
| | | |
Collapse
|
45
|
Nabavizadeh P, Liu J, Havel CM, Ibrahim S, Derakhshandeh R, Jacob Iii P, Springer ML. Vascular endothelial function is impaired by aerosol from a single IQOS HeatStick to the same extent as by cigarette smoke. Tob Control 2018; 27:s13-s19. [PMID: 30206183 PMCID: PMC6202192 DOI: 10.1136/tobaccocontrol-2018-054325] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/10/2018] [Accepted: 05/11/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Heated tobacco products (also called 'heat-not-burn' products) heat tobacco at temperatures below that of combustion, causing nicotine and other compounds to aerosolise. One such product, IQOS from Philip Morris International, is being marketed internationally with claims of harm reduction. We sought to determine whether exposure to IQOS aerosol impairs arterial flow-mediated dilation (FMD), a measure of vascular endothelial function that is impaired by tobacco smoke. METHODS We exposed anaesthetised rats (n=8/group) via nose cone to IQOS aerosol from single HeatSticks, mainstream smoke from single Marlboro Red cigarettes or clean air for a series of consecutive 30 s cycles over 1.5-5 min. Each cycle consisted of 15 or 5 s of exposure followed by removal from the nose cone. We measured pre-exposure and postexposure FMD, and postexposure serum nicotine and cotinine. RESULTS FMD was impaired comparably by ten 15 s exposures and ten 5 s exposures to IQOS aerosol and to cigarette smoke, but not by clean air. Serum nicotine levels were similar to plasma levels after humans have smoked one cigarette, confirming that exposure conditions had real-world relevance. Postexposure nicotine levels were ~4.5-fold higher in rats exposed to IQOS than to cigarettes, despite nicotine being measured in the IQOS aerosol at ~63% the amount measured in smoke. When IQOS exposure was briefer, leading to comparable serum nicotine levels to the cigarette group, FMD was still comparably impaired. CONCLUSIONS Acute exposures to IQOS aerosol impairs FMD in rats. IQOS use does not necessarily avoid the adverse cardiovascular effects of smoking cigarettes.
Collapse
Affiliation(s)
- Pooneh Nabavizadeh
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Jiangtao Liu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Christopher M Havel
- Division of Clinical Pharmacology and Experimental Therapeutics, University of California, San Francisco, San Francisco, California, USA
| | - Sharina Ibrahim
- Division of Cardiology, University of California, San Francisco, San Francisco, California, USA
| | - Ronak Derakhshandeh
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Peyton Jacob Iii
- Division of Clinical Pharmacology and Experimental Therapeutics, University of California, San Francisco, San Francisco, California, USA
- Center for Tobacco Control Research and Education, University of California, San Francisco, San Francisco, California, USA
| | - Matthew L Springer
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
- Division of Cardiology, University of California, San Francisco, San Francisco, California, USA
- Center for Tobacco Control Research and Education, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
46
|
Poussin C, Laurent A, Kondylis A, Marescotti D, van der Toorn M, Guedj E, Goedertier D, Acali S, Pak C, Dulize R, Baumer K, Peric D, Maluenda E, Bornand D, Suarez IG, Schlage WK, Ivanov NV, Peitsch MC, Hoeng J. In vitro systems toxicology-based assessment of the potential modified risk tobacco product CHTP 1.2 for vascular inflammation- and cytotoxicity-associated mechanisms promoting adhesion of monocytic cells to human coronary arterial endothelial cells. Food Chem Toxicol 2018; 120:390-406. [PMID: 30026091 DOI: 10.1016/j.fct.2018.07.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/29/2018] [Accepted: 07/13/2018] [Indexed: 12/24/2022]
Abstract
Cigarette smoking causes cardiovascular diseases. Heating tobacco instead of burning it reduces the amount of toxic compounds in the aerosol and may exert a reduced impact on health compared with cigarette smoke. Aqueous extract from the aerosol of a potential modified risk tobacco product, the Carbon Heated Tobacco Product (CHTP) 1.2, was compared in vitro with aqueous extract from the smoke of a 3R4F reference cigarette for its impact on the adhesion of monocytic cells to artery endothelial cells. Human coronary artery endothelial cells (HCAEC) were treated for 4 h with conditioned media from human monocytic Mono Mac 6 (MM6) cells exposed to CHTP1.2 or 3R4F extracts for 2 h or directly with those extracts freshly generated. In vitro monocyte-endothelial cell adhesion was measured concomitantly with inflammatory, oxidative stress, cytotoxicity, and death markers. Furthermore, transcriptomics analyses enabled to quantify the level of perturbation in HCAECs, and provide biological interpretation for the underlying molecular changes following exposure to 3R4F or CHTP1.2 extract. Our systems toxicology study demonstrated that approximately 10-15-fold higher concentrations of the CHTP 1.2 aerosol extract were needed to elicit similar effects as the 3R4F smoke extract on cardiovascular disease-relevant inflammation and cytotoxicity-related mechanisms and markers investigated in vitro.
Collapse
Affiliation(s)
- Carine Poussin
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| | - Alexandra Laurent
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Athanasios Kondylis
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Diego Marescotti
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Marco van der Toorn
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Didier Goedertier
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Stefano Acali
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Claudius Pak
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Rémi Dulize
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Karine Baumer
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Dariusz Peric
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Elodie Maluenda
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - David Bornand
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Ignacio Gonzalez Suarez
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Walter K Schlage
- Biology Consultant, Max-Baermann-Str. 21, 51429 Bergisch Gladbach, Germany
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Part of Philip Morris International Group of Companies, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| |
Collapse
|
47
|
Ansari SM, Lama N, Blanc N, Bosilkovska M, Donelli A, Picavet P, Baker G, Haziza C, Lüdicke F. Evaluation of Biological and Functional Changes in Healthy Smokers Switching to the Tobacco Heating System 2.2 Versus Continued Tobacco Smoking: Protocol for a Randomized, Controlled, Multicenter Study. JMIR Res Protoc 2018; 7:e11294. [PMID: 30143474 PMCID: PMC6128954 DOI: 10.2196/11294] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/25/2018] [Accepted: 07/25/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Tobacco harm reduction, substituting less harmful tobacco products for combustible cigarettes, is a complementary approach for smokers who would otherwise continue to smoke. The Philip Morris International (PMI) Tobacco Heating System (THS) 2.2 is a novel tobacco product with the potential to reduce the risk of harm in smokers compared to continued smoking of combustible cigarettes. It heats tobacco electrically in a controlled manner, never allowing the temperature to exceed 350°C, thereby preventing the combustion process from taking place and producing substantially lower levels of toxicants while providing nicotine, taste, ritual, and a sensory experience that closely parallels combustible cigarettes. Previous clinical studies have demonstrated reduced exposure to the toxicants (approaching the levels observed after quitting) for smokers who switched to THS 2.2, for three months. For adult smokers who would otherwise continue smoking combustible cigarettes, switching to THS 2.2 may represent an alternative way to reduce the risk of tobacco-related diseases. OBJECTIVE This study aimed to further substantiate the harm reduction potential of THS 2.2 by demonstrating favorable changes in a set of 8 coprimary endpoints, representative of pathomechanistic pathways (ie, inflammation, oxidative stress, lipid metabolism, respiratory function, and genotoxicity), linked to smoking-related diseases, in smokers switching from combustible cigarettes to THS 2.2. METHODS This study was a randomized, controlled, two-arm parallel group, multicenter ambulatory US study conducted in healthy adult smokers switching from combustible cigarettes to THS 2.2 compared with smokers continuing to smoke combustible cigarettes for six months. Subjects had a smoking history of at least ten years and did not intend to quit within the next six months. RESULTS Enrollment started in March 2015 and the trial was completed in September 2016. In total, 984 subjects were randomized (combustible cigarettes, n=483; THS 2.2, n=477), and 803 completed the study. The results are expected to be available in a subsequent publication in 2019. CONCLUSIONS In this paper, we describe the rationale and design for this clinical study that focused on the evaluation of THS 2.2's potential to reduce the risk of smoking-related diseases compared with that of combustible cigarettes. This study will provide insights regarding favorable changes in biological and functional endpoints informed by effects known to be seen upon smoking cessation. TRIAL REGISTRATION ClinicalTrials.gov NCT02396381; http://clinicaltrials.gov/ct2/show/NCT02396381 (Archived by WebCite at http://www.webcitation.org/71PCRdagP). REGISTERED REPORT IDENTIFIER RR1-10.2196/11294.
Collapse
Affiliation(s)
- S Michael Ansari
- Philip Morris International Research & Development, Neuchâtel, Switzerland
| | - Nicola Lama
- Philip Morris International Research & Development, Neuchâtel, Switzerland
| | - Nicolas Blanc
- Philip Morris International Research & Development, Neuchâtel, Switzerland
| | - Marija Bosilkovska
- Philip Morris International Research & Development, Neuchâtel, Switzerland
| | - Andrea Donelli
- Philip Morris International Research & Development, Neuchâtel, Switzerland
| | - Patrick Picavet
- Philip Morris International Research & Development, Neuchâtel, Switzerland
| | - Gizelle Baker
- Philip Morris International Research & Development, Neuchâtel, Switzerland
| | - Christelle Haziza
- Philip Morris International Research & Development, Neuchâtel, Switzerland
| | - Frank Lüdicke
- Philip Morris International Research & Development, Neuchâtel, Switzerland
| |
Collapse
|
48
|
Phillips BW, Schlage WK, Titz B, Kogel U, Sciuscio D, Martin F, Leroy P, Vuillaume G, Krishnan S, Lee T, Veljkovic E, Elamin A, Merg C, Ivanov NV, Peitsch MC, Hoeng J, Vanscheeuwijck P. A 90-day OECD TG 413 rat inhalation study with systems toxicology endpoints demonstrates reduced exposure effects of the aerosol from the carbon heated tobacco product version 1.2 (CHTP1.2) compared with cigarette smoke. I. Inhalation exposure, clinical pathology and histopathology. Food Chem Toxicol 2018; 116:388-413. [PMID: 29654848 DOI: 10.1016/j.fct.2018.04.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/28/2018] [Accepted: 04/07/2018] [Indexed: 01/25/2023]
Abstract
Within the framework of a systems toxicology approach, the inhalation toxicity of aerosol from a novel tobacco-heating potentially modified risk tobacco product (MRTP), the carbon-heated tobacco product (CHTP) 1.2, was characterized and compared with that of mainstream smoke (CS) from the 3R4F reference cigarette in a 90-day nose-only rat inhalation study in general accordance with OECD TG 413. CHTP1.2 is a heat-not-burn product using a carbon heat source to produce an aerosol that contains nicotine and tobacco flavor. At equal or twice the nicotine concentration in the test atmospheres, inhalation of CHTP1.2 aerosol led to a significantly lower exposure to harmful constituents and induced less respiratory tract irritation, systemic, and pathological effects compared with CS. Nasal epithelial changes were less pronounced in the CHTP1.2- than in the CS-exposed groups and reverted in the nicotine concentration-matched group after a recovery period. Lung inflammation was minimal in the CHTP1.2-treated groups compared with the moderate extent seen in the 3R4F groups. Many other toxicological endpoints evaluated did not show CHTP1.2 aerosol exposure-related effects, and no effects not seen for 3R4F were observed. These observations were consistent with findings from previous studies in which rats were exposed to MRTP aerosols containing similar nicotine concentrations.
Collapse
Affiliation(s)
- Blaine W Phillips
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Walter K Schlage
- Biology Consultant, Max-Baermann-Str. 21, 51429, Bergisch Gladbach, Germany
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Ulrike Kogel
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Davide Sciuscio
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Florian Martin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Gregory Vuillaume
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Subash Krishnan
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Tom Lee
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Emilija Veljkovic
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Ashraf Elamin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Celine Merg
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland
| | - Patrick Vanscheeuwijck
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland.
| |
Collapse
|
49
|
Florence JM, Krupa A, Booshehri LM, Gajewski AL, Kurdowska AK. Disrupting the Btk Pathway Suppresses COPD-Like Lung Alterations in Atherosclerosis Prone ApoE -/- Mice Following Regular Exposure to Cigarette Smoke. Int J Mol Sci 2018; 19:ijms19020343. [PMID: 29364178 PMCID: PMC5855565 DOI: 10.3390/ijms19020343] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/19/2018] [Accepted: 01/21/2018] [Indexed: 01/08/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with severe chronic inflammation that promotes irreversible tissue destruction. Moreover, the most broadly accepted cause of COPD is exposure to cigarette smoke. There is no effective cure and significantly, the mechanism behind the development and progression of this disease remains unknown. Our laboratory has demonstrated that Bruton’s tyrosine kinase (Btk) is a critical regulator of pro-inflammatory processes in the lungs and that Btk controls expression of matrix metalloproteinase-9 (MMP-9) in the alveolar compartment. For this study apolipoprotein E null (ApoE−/−) mice were exposed to SHS to facilitate study in a COPD/atherosclerosis comorbidity model. We applied two types of treatments, animals received either a pharmacological inhibitor of Btk or MMP-9 specific siRNA to minimize MMP-9 expression in endothelial cells or neutrophils. We have shown that these treatments had a protective effect in the lung. We have noted a decrease in alveolar changes related to SHS induced inflammation in treated animals. In summary, we are presenting a novel concept in the field of COPD, i.e., that Btk may be a new drug target for this disease. Moreover, cell specific targeting of MMP-9 may also benefit patients affected by this disease.
Collapse
Affiliation(s)
- Jon M Florence
- Department of Cellular and Molecular Biology, University of Texas Health Science Center, Tyler, TX 75708, USA.
| | - Agnieszka Krupa
- Department of Cellular and Molecular Biology, University of Texas Health Science Center, Tyler, TX 75708, USA.
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland.
| | - Laela M Booshehri
- Department of Cellular and Molecular Biology, University of Texas Health Science Center, Tyler, TX 75708, USA.
| | - Adrian L Gajewski
- Department of Cellular and Molecular Biology, University of Texas Health Science Center, Tyler, TX 75708, USA.
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland.
| | - Anna K Kurdowska
- Department of Cellular and Molecular Biology, University of Texas Health Science Center, Tyler, TX 75708, USA.
| |
Collapse
|
50
|
Lüdicke F, Picavet P, Baker G, Haziza C, Poux V, Lama N, Weitkunat R. Effects of Switching to the Menthol Tobacco Heating System 2.2, Smoking Abstinence, or Continued Cigarette Smoking on Clinically Relevant Risk Markers: A Randomized, Controlled, Open-Label, Multicenter Study in Sequential Confinement and Ambulatory Settings (Part 2). Nicotine Tob Res 2018; 20:173-182. [PMID: 28177498 PMCID: PMC5896432 DOI: 10.1093/ntr/ntx028] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 02/06/2017] [Indexed: 11/13/2022]
Abstract
Introduction Modified-risk tobacco products are expected to reduce exposure to harmful and potentially harmful constituents of cigarette smoke, and ultimately reduce the health burden of smoking-related diseases. Clinically relevant risk markers of smoking-related diseases inform about the risk profile of new tobacco products in the absence of in-market epidemiological data. The menthol Tobacco Heating System 2.2 (mTHS) is a modified-risk tobacco product in development as an alternative to cigarettes (conventional cigarettes [CCs]). Methods In this parallel-group study, Japanese adult smokers (23-65 years; ≥10 mCCs/day) were randomized to mTHS, menthol CCs (mCC), or smoking abstinence (SA) for 5 days in confinement and 85 days in ambulatory settings. Endpoints included biomarkers of exposure to harmful and potentially harmful constituents and clinically relevant risk markers of smoking-related diseases. Results One-hundred and sixty participants were randomized to the mTHS (n = 78), mCC (n = 42), and SA (n = 40) groups. Switching to the mTHS was associated with reductions in biomarkers of exposure compared with continuing mCCs. Reductions in 8-epi-prostaglandin F2α (biomarker of oxidative stress), 11-dehydro-thromboxane B2 (biomarker of platelet activation), soluble intracellular adhesion molecule-1 (biomarker of endothelial function), and an increase in high-density lipoprotein cholesterol (biomarker of lipid metabolism) and forced expiratory volume in 1 second (biomarker of lung function) occurred in the mTHS group compared with the mCC group. The changes in the mTHS group approached those in the SA group. Conclusions Switching from mCCs to mTHS was associated with improvements in clinically relevant risk markers linked to mechanistic pathways involved in smoking-related diseases. Implications In this three-way randomized study, switching from menthol cigarettes to mTHS for 5 days in confinement and 85 days in ambulatory settings was associated with reductions in biomarkers of exposure to cigarette smoke, and changes were observed in clinically relevant biomarkers of oxidative stress (8-epi-prostaglandin F2α), platelet activity (11-dehydro-thromboxane B2), endothelial function (soluble intracellular adhesion molecule-1), lipid metabolism (high-density lipoprotein cholesterol) and lung function (forced expiratory volume in 1 second), similar to the SA group. The results suggest that switching to the mTHS has the potential to reduce the adverse health effects of conventional cigarettes.
Collapse
Affiliation(s)
- Frank Lüdicke
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Patrick Picavet
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Gizelle Baker
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Christelle Haziza
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Valerie Poux
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Nicola Lama
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| | - Rolf Weitkunat
- Philip Morris Products S.A., PMI Research and Development, Neuchâtel, Switzerland
| |
Collapse
|