1
|
Chang CY, Armstrong D, Knight JM, Gale TV, Hawley S, Wang M, Chang N, Corry DB, Kheradmand F. Sialidase fusion protein protects against influenza infection in a cigarette smoke-induced model of COPD. Mucosal Immunol 2025; 18:467-480. [PMID: 39837384 DOI: 10.1016/j.mucimm.2025.01.004] [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: 08/19/2024] [Revised: 11/20/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
First- and secondhand smokers are at an increased risk for influenza virus (IFV)-related respiratory failure and death. Despite approved influenza antiviral treatments, there is an unmet need for treatments that can improve outcomes in populations at risk for respiratory failure, including tobacco users with Chronic Obstructive Pulmonary Disease (COPD). Here we show that the sialidase fusion protein, DAS181, reduced viral burden, mitigated inflammation, and attenuated lung function loss, consistent with broad-spectrum anti-influenza responses in a mouse model of COPD and IFV-A infection. Treatment with DAS181 reprogramed the sialic acid-binding immunoglobulin-like lectins (Siglecs) in alveolar macrophages, increased expression of phagocytic marker CD169, and downregulated inhibitory Siglec-F and Siglec-H molecules. Upon reinfection, mice treated with DAS181 showed activated and protective memory response in the lungs. Collectively, we show that this sialidase fusion protein promotes a beneficial immunomodulatory reaction in the lungs, supporting a new IFV-A therapeutic option for at-risk smokers.
Collapse
Affiliation(s)
- Cheng-Yen Chang
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - John M Knight
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Max Wang
- Ansun Biopharma, San Diego, CA 92121, USA
| | | | - David B Corry
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030, USA; Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey, Houston, TX 77030, USA.
| | - Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030, USA; Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Kirby A, Suchý M, Duan D, Bazett M, Kalyan S, Shuhendler AJ. Tracking the fate of bacteria-derived site-specific immunomodulators by positron emission tomography. Nucl Med Biol 2024; 132-133:108908. [PMID: 38599145 DOI: 10.1016/j.nucmedbio.2024.108908] [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: 12/07/2023] [Revised: 03/08/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024]
Abstract
INTRODUCTION Site-specific immunomodulators (SSIs) are a novel class of therapeutics made from inactivated bacterial species designed to regulate the innate immune system in targeted organs. QBECO is a gut-targeted SSI that is being advanced clinically to treat and/or prevent inflammatory bowel disease, cancer, and serious infections of the gastrointestinal (GI) tract and proximal organs, and QBKPN is a lung-targeted SSI that is in clinical development for the treatment and/or prevention of chronic inflammatory lung disease, lung cancers and respiratory tract infections. While these SSIs have demonstrated both safety and proof-of-concept in preclinical and clinical studies, detailed understanding of their trafficking and biodistribution is yet to be fully characterized. METHODS QBECO and QBKPN were radiolabeled with [89Zr] and injected subcutaneously into healthy mice. The mice underwent Positron Emission Tomography (PET) imaging every day for eight days to track biodistribution of the SSIs. Tissue from the site of injection was collected and immunohistologically probed for immune cell infiltration. RESULTS Differential biodistribution of the two SSIs was seen, adhering to their site-specific targeting. QBKPN appeared to migrate from the site of injection (abdomen) to the cervical lymph nodes which are nearer to the respiratory tract and lungs. QBECO remained in the abdominal region, with lymphatic trafficking to the inguinal lymph nodes, which are nearer to GI-proximal tissues/organs. Immune infiltration at the site of injection comprised of neutrophils for both SSIs, and macrophages for only QBKPN. CONCLUSION Radiolabeling of SSIs allows for longitudinal in vivo imaging of biodistribution and trafficking. PET imaging revealed differential biodistribution of the SSIs based on the organotropism of the bacteria from which the SSI is derived. Trafficking from the site of injection to the targeted site is in part mediated via the lymphatics and involves macrophages and neutrophils.
Collapse
Affiliation(s)
- Alexia Kirby
- Department of Biology, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Mojmír Suchý
- University of Ottawa Heart Institute, Ottawa, ON, Canada; Department of Chemistry and Biomolecular Sciences, Ottawa, ON, Canada
| | - Daniel Duan
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | | | - Shirin Kalyan
- Qu Biologics Inc., Vancouver, BC, Canada; Department of Medicine, Division of Endocrinology, University of British Columbia, Vancouver, BC, Canada
| | - Adam J Shuhendler
- Department of Biology, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada; Department of Chemistry and Biomolecular Sciences, Ottawa, ON, Canada.
| |
Collapse
|
3
|
McDonald VM, Archbold G, Beyene T, Brew BK, Franklin P, Gibson PG, Harrington J, Hansbro PM, Johnston FH, Robinson PD, Sutherland M, Yates D, Zosky GR, Abramson MJ. Asthma and landscape fire smoke: A Thoracic Society of Australia and New Zealand position statement. Respirology 2023; 28:1023-1035. [PMID: 37712340 PMCID: PMC10946536 DOI: 10.1111/resp.14593] [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: 01/12/2023] [Accepted: 07/14/2023] [Indexed: 09/16/2023]
Abstract
Landscape fires are increasing in frequency and severity globally. In Australia, extreme bushfires cause a large and increasing health and socioeconomic burden for communities and governments. People with asthma are particularly vulnerable to the effects of landscape fire smoke (LFS) exposure. Here, we present a position statement from the Thoracic Society of Australia and New Zealand. Within this statement we provide a review of the impact of LFS on adults and children with asthma, highlighting the greater impact of LFS on vulnerable groups, particularly older people, pregnant women and Aboriginal and Torres Strait Islander peoples. We also highlight the development of asthma on the background of risk factors (smoking, occupation and atopy). Within this document we present advice for asthma management, smoke mitigation strategies and access to air quality information, that should be implemented during periods of LFS. We promote clinician awareness, and the implementation of public health messaging and preparation, especially for people with asthma.
Collapse
Affiliation(s)
- Vanessa M. McDonald
- College of Health, Medicine and WellbeingUniversity of NewcastleCallaghanNew South WalesAustralia
- Asthma and Breathing Research ProgramThe Hunter Medical Research Institute (HMRI)New LambtonNew South WalesAustralia
- Department of Sleep and Respiratory MedicineHunter New England Local Health DistrictNewcastleNew South WalesAustralia
| | - Gregory Archbold
- Asthma and Breathing Research ProgramThe Hunter Medical Research Institute (HMRI)New LambtonNew South WalesAustralia
| | - Tesfalidet Beyene
- College of Health, Medicine and WellbeingUniversity of NewcastleCallaghanNew South WalesAustralia
- Asthma and Breathing Research ProgramThe Hunter Medical Research Institute (HMRI)New LambtonNew South WalesAustralia
| | - Bronwyn K. Brew
- National Perinatal Epidemiology and Biostatistics Unit, Centre for Big Data Research in Health, Discipline of Women's Health, Faculty of MedicineUNSWSydneyNew South WalesAustralia
| | - Peter Franklin
- School of Population and Global HealthThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Peter G. Gibson
- College of Health, Medicine and WellbeingUniversity of NewcastleCallaghanNew South WalesAustralia
- Asthma and Breathing Research ProgramThe Hunter Medical Research Institute (HMRI)New LambtonNew South WalesAustralia
- Department of Sleep and Respiratory MedicineHunter New England Local Health DistrictNewcastleNew South WalesAustralia
| | - John Harrington
- Asthma and Breathing Research ProgramThe Hunter Medical Research Institute (HMRI)New LambtonNew South WalesAustralia
- Department of Sleep and Respiratory MedicineHunter New England Local Health DistrictNewcastleNew South WalesAustralia
| | - Philip M. Hansbro
- Centre for InflammationCentenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of ScienceSydneyNew South WalesAustralia
- Immune HealthHunter Medical Research Institute and The University of NewcastleCallaghanNew South WalesAustralia
| | - Fay H. Johnston
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - Paul D. Robinson
- Department of Respiratory and Sleep MedicineQueensland Children's HospitalBrisbaneQueenslandAustralia
- Children's Health and Environment Program, Child Health Research CentreUniversity of QueenslandBrisbaneQueenslandAustralia
| | | | - Deborah Yates
- Department of Thoracic MedicineSt Vincent's HospitalDarlinghurstNew South WalesAustralia
- School of Clinical MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | - Graeme R. Zosky
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
- Tasmanian School of MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Michael J. Abramson
- School of Public Health & Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
4
|
Human microbiota: a crucial gatekeeper in lung cancer initiation, progression, and treatment. MEDICINE IN MICROECOLOGY 2022. [DOI: 10.1016/j.medmic.2022.100055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
5
|
Guo H, Zhao L, Zhu J, Chen P, Wang H, Jiang M, Liu X, Sun H, Zhao W, Zheng Z, Li W, Chen B, Fang Q, Yang M, He Y, Yang Y. Microbes in lung cancer initiation, treatment, and outcome: Boon or bane? Semin Cancer Biol 2021; 86:1190-1206. [PMID: 34029741 DOI: 10.1016/j.semcancer.2021.05.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 01/17/2023]
Abstract
Lung cancer is the top reason for cancer-related deaths worldwide. The 5-year overall survival rate of lung cancer is approximately 20 % due to the delayed diagnosis and low response rate to regular treatments. Microbiota, both host-microbiota and alien pathogenic microbiota, have been investigated to be involved in a complicated and contradictory relationship with lung cancer initiation, treatments, and prognosis. Disorders of certain host-microbiota and pathogen infection are associated with the risk of lung cancers based on epidemiological evidence, and antibiotics (ATBs) could dramatically impair anti-cancer treatment efficacy, including chemotherapy and immunotherapy. Moreover, probiotics and microbe-mediated drugs are potential approaches to enhance regular anti-tumor treatments. Therefore, the knowledge of the complex dual effect of microbes on lung cancer is beneficial to take their essence and remove their dross. This review offers insight into the current trends and advancements in microbiota or microbial components related to lung cancer.
Collapse
Affiliation(s)
- Haoyue Guo
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China; Tongji University, No 1239 Siping Road, Shanghai, 200092, People's Republic of China
| | - Lishu Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China
| | - Junjie Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China; Tongji University, No 1239 Siping Road, Shanghai, 200092, People's Republic of China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China; Tongji University, No 1239 Siping Road, Shanghai, 200092, People's Republic of China
| | - Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China; Tongji University, No 1239 Siping Road, Shanghai, 200092, People's Republic of China
| | - Xiaogang Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Hui Sun
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China
| | - Wencheng Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China
| | - Zixuan Zheng
- Tongji University, No 1239 Siping Road, Shanghai, 200092, People's Republic of China; Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China
| | - Bin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China
| | - Qiyu Fang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China
| | - Menghang Yang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai, 200433, People's Republic of China; Tongji University, No 1239 Siping Road, Shanghai, 200092, People's Republic of China.
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Materials Science and Engineering, Tongji University, Shanghai, 201804, People's Republic of China.
| |
Collapse
|
6
|
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
|
7
|
Carvalho JL, Miranda M, Fialho AK, Castro-Faria-Neto H, Anatriello E, Keller AC, Aimbire F. Oral feeding with probiotic Lactobacillus rhamnosus attenuates cigarette smoke-induced COPD in C57Bl/6 mice: Relevance to inflammatory markers in human bronchial epithelial cells. PLoS One 2020; 15:e0225560. [PMID: 32330145 PMCID: PMC7182195 DOI: 10.1371/journal.pone.0225560] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/03/2020] [Indexed: 01/10/2023] Open
Abstract
COPD is a prevalent lung disease with significant impacts on public health. Affected airways exhibit pulmonary neutrophilia and consequent secretion of pro-inflammatory cytokines and proteases, which result in lung emphysema. Probiotics act as nonspecific modulators of the innate immune system that improve several inflammatory responses. To investigate the effect of Lactobacillus rhamnosus (Lr) on cigarette smoke (CS)-induced COPD C57Bl/6 mice were treated with Lr during the week before COPD induction and three times/week until euthanasia. For in vitro assays, murine bronchial epithelial cells as well as human bronchial epithelial cells exposed to cigarette smoke extract during 24 hours were treated with Lr 1 hour before CSE addition. Lr treatment attenuated the inflammatory response both in the airways and lung parenchyma, reducing inflammatory cells infiltration and the production of pro-inflammatory cytokines and chemokines. Also, Lr-treated mice presented with lower metalloproteases in lung tissue and lung remodeling. In parallel to the reduction in the expression of TLR2, TLR4, TLR9, STAT3, and NF-κB in lung tissue, Lr increased the levels of IL-10 as well as SOCS3 and TIMP1/2, indicating the induction of an anti-inflammatory environment. Similarly, murine bronchial epithelial cells as well as human bronchial epithelial cells (BEAS) exposed to CSE produced pro-inflammatory cytokines and chemokines, which were inhibited by Lr treatment in association with the production of anti-inflammatory molecules. Moreover, the presence of Lr also modulated the expression of COPD-associated transcription found into BALF of COPD mice group, i.e., Lr downregulated expression of NF-κB and STAT3, and inversely upregulated increased expression of SOCS3. Thus, our findings indicate that Lr modulates the balance between pro- and anti-inflammatory cytokines in human bronchial epithelial cells upon CS exposure and it can be a useful tool to improve the lung inflammatory response associated with COPD.
Collapse
Affiliation(s)
- J. L. Carvalho
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, São Paulo, Brazil
| | - M. Miranda
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, São Paulo, Brazil
| | - A. K. Fialho
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, São Paulo, Brazil
| | | | - E. Anatriello
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, São Paulo, Brazil
| | - A. C. Keller
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - F. Aimbire
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, São Paulo, Brazil
| |
Collapse
|
8
|
Distinct inactivated bacterial-based immune modulators vary in their therapeutic efficacies for treating disease based on the organ site of pathology. Sci Rep 2020; 10:5901. [PMID: 32246043 PMCID: PMC7125210 DOI: 10.1038/s41598-020-62735-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/17/2020] [Indexed: 11/14/2022] Open
Abstract
Recent developments in understanding how the functional phenotype of the innate immune system is programmed has led to paradigm-shifting views on immunomodulation. These advances have overturned two long-held dogmas: (1) only adaptive immunity confers immunological memory; and, (2) innate immunity lacks specificity. This work describes the observation that innate immune effector cells appear to be differentially recruited to specific pathological sites when mobilized by distinct inactivated bacterial-based stimuli administered subcutaneously. The studies presented suggest that the immune system, upon detecting the first signs of a potential infection by a specific pathogen, tends to direct its resources to the compartment from which that pathogen is most likely originating. The findings from this work puts forth the novel hypothesis that the immunotherapeutic efficacy of a microbial-based stimulus for innate immune mobilization depends on the correct selection of the microbial species used as the stimulant and its relationship to the organ in which the pathology is present.
Collapse
|
9
|
Shukla SD, Walters EH, Simpson JL, Keely S, Wark PA, O'Toole RF, Hansbro PM. Hypoxia‐inducible factor and bacterial infections in chronic obstructive pulmonary disease. Respirology 2019; 25:53-63. [DOI: 10.1111/resp.13722] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/25/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Shakti D. Shukla
- School of Biomedical Sciences and Pharmacy, Faculty of Health and MedicineUniversity of Newcastle Newcastle NSW Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research InstituteUniversity of Newcastle Newcastle NSW Australia
| | - E. Haydn Walters
- School of Medicine, College of Health and MedicineUniversity of Tasmania Hobart TAS Australia
| | - Jodie L. Simpson
- Priority Research Centre for Healthy Lungs, Hunter Medical Research InstituteUniversity of Newcastle Newcastle NSW Australia
- Respiratory and Sleep Medicine, Priority Research Centre for Healthy LungsUniversity of Newcastle Newcastle NSW Australia
| | - Simon Keely
- School of Biomedical Sciences and Pharmacy, Faculty of Health and MedicineUniversity of Newcastle Newcastle NSW Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research InstituteUniversity of Newcastle Newcastle NSW Australia
| | - Peter A.B. Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research InstituteUniversity of Newcastle Newcastle NSW Australia
- Respiratory and Sleep Medicine, Priority Research Centre for Healthy LungsUniversity of Newcastle Newcastle NSW Australia
| | - Ronan F. O'Toole
- School of Molecular Sciences, College of Science, Health and EngineeringLa Trobe University Melbourne VIC Australia
| | - Philip M. Hansbro
- School of Biomedical Sciences and Pharmacy, Faculty of Health and MedicineUniversity of Newcastle Newcastle NSW Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research InstituteUniversity of Newcastle Newcastle NSW Australia
- Centenary Institute and School of Life Sciences, Faculty of Science, University of Technology Sydney Sydney NSW Australia
| |
Collapse
|
10
|
Chronic Obstructive Pulmonary Disease and Lung Cancer: Underlying Pathophysiology and New Therapeutic Modalities. Drugs 2019; 78:1717-1740. [PMID: 30392114 DOI: 10.1007/s40265-018-1001-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer are major lung diseases affecting millions worldwide. Both diseases have links to cigarette smoking and exert a considerable societal burden. People suffering from COPD are at higher risk of developing lung cancer than those without, and are more susceptible to poor outcomes after diagnosis and treatment. Lung cancer and COPD are closely associated, possibly sharing common traits such as an underlying genetic predisposition, epithelial and endothelial cell plasticity, dysfunctional inflammatory mechanisms including the deposition of excessive extracellular matrix, angiogenesis, susceptibility to DNA damage and cellular mutagenesis. In fact, COPD could be the driving factor for lung cancer, providing a conducive environment that propagates its evolution. In the early stages of smoking, body defences provide a combative immune/oxidative response and DNA repair mechanisms are likely to subdue these changes to a certain extent; however, in patients with COPD with lung cancer the consequences could be devastating, potentially contributing to slower postoperative recovery after lung resection and increased resistance to radiotherapy and chemotherapy. Vital to the development of new-targeted therapies is an in-depth understanding of various molecular mechanisms that are associated with both pathologies. In this comprehensive review, we provide a detailed overview of possible underlying factors that link COPD and lung cancer, and current therapeutic advances from both human and preclinical animal models that can effectively mitigate this unholy relationship.
Collapse
|
11
|
Caramori G, Ruggeri P, Mumby S, Ieni A, Lo Bello F, Chimankar V, Donovan C, Andò F, Nucera F, Coppolino I, Tuccari G, Hansbro PM, Adcock IM. Molecular links between COPD and lung cancer: new targets for drug discovery? Expert Opin Ther Targets 2019; 23:539-553. [DOI: 10.1080/14728222.2019.1615884] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Gaetano Caramori
- Unità Operativa Complessa di Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Paolo Ruggeri
- Unità Operativa Complessa di Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Sharon Mumby
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Anatomic Pathology, University of Messina, Messina, Italy
| | - Federica Lo Bello
- Unità Operativa Complessa di Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Vrushali Chimankar
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | - Filippo Andò
- Unità Operativa Complessa di Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Francesco Nucera
- Unità Operativa Complessa di Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Irene Coppolino
- Unità Operativa Complessa di Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Giovanni Tuccari
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Anatomic Pathology, University of Messina, Messina, Italy
| | - Philip M. Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
- Faculty of Science, Ultimo, and Centenary Institute, Centre for Inflammation, University of Technology Sydney, Sydney, Australia
| | - Ian M. Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
12
|
Su Y, Luo H, Yang J. Heparin-binding EGF-like growth factor attenuates lung inflammation and injury in a murine model of pulmonary emphysema. Growth Factors 2018; 36:246-262. [PMID: 30600734 DOI: 10.1080/08977194.2018.1552270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pulmonary inflammation and progressive lung destruction are the major causes of chronic obstructive pulmonary disease (COPD), resulting in emphysema and irreversible pulmonary dysfunction. Heparin-binding EGF-like growth factor (HB-EGF), is known to play a protective role in the process of various inflammatory diseases. However, its effect on COPD is poorly understood. This study was designed to determine the effect of HB-EGF on lung inflammation and injury in a murine model of pulmonary emphysema. HB-EGF promoted percent survival and body weight, attenuated lung injury, inflammatory cells, and cytokines infiltration, and prevented lung function decline. Additionally, treatment of rHB-EGF suppressed the nuclear translocation of nuclear factor κB (NF-κB)/p65, decreased TUNEL-positive cells and the expression of caspase 3, and increased the expression of PCNA, HB-EGF, and EGF receptor (EGFR). We conclude that HB-EGF attenuates lung inflammation and injury, probably through the activation of EGFR, followed by suppression of NF-ΚB signalling, promotion of cell proliferation, and inhibition of apoptosis.
Collapse
Affiliation(s)
- Yanwei Su
- a School of Nursing, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| | - Heng Luo
- b Department of Pathology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| | - Jixin Yang
- c Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| |
Collapse
|
13
|
Sham HP, Bazett M, Bosiljcic M, Yang H, Luk B, Law HT, Morampudi V, Yu HB, Pankovich J, Sutcliffe S, Bressler B, Marshall JK, Fedorak RN, Chen J, Jones M, Gunn H, Kalyan S, Vallance BA. Immune Stimulation Using a Gut Microbe-Based Immunotherapy Reduces Disease Pathology and Improves Barrier Function in Ulcerative Colitis. Front Immunol 2018; 9:2211. [PMID: 30319652 PMCID: PMC6170651 DOI: 10.3389/fimmu.2018.02211] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/06/2018] [Indexed: 01/17/2023] Open
Abstract
Background: Current ulcerative colitis (UC) treatments are focused on symptom management primarily via immune suppression. Despite the current arsenal of immunosuppressant treatments, the majority of patients with UC still experience disease progression. Importantly, aggressive long-term inhibition of immune function comes with consequent risk, such as serious infections and malignancy. There is thus a recognized need for new, safe and effective treatment strategies for people living with UC that work upstream of managing the symptoms of the disease. The objective of this study was to evaluate a microbial-based treatment, QBECO, that functions to productively activate rather than suppress mucosal immune function as a novel approach to treat UC. Methods: Two established models of experimental colitis, namely chemically-induced DSS colitis and the spontaneous colitis that develops in Muc2 deficient mice, were used to assess whether QBECO treatment could ameliorate gastrointestinal disease. A small exploratory 16-week QBECO open-label trial was subsequently conducted to test the safety and tolerability of this approach and also to determine whether similar improvements in clinical disease and histopathology could be demonstrated in patients with moderate-to-severe UC. Results: QBECO treatment successfully reduced inflammation and promoted mucosal and histological healing in both experimental models and in UC patients. The preclinical models of colitis showed that QBECO ameliorated mucosal pathology, in part by reducing inflammatory cell infiltration, primarily that induced by neutrophils and inflammatory T cells. The most rapid and noticeable change observed in QBECO treated UC patients was a marked reduction in rectal bleeding. Conclusion: Collectively, this work demonstrates for the first time that strategically activating immune function rather than suppressing it, not only does not worsen colitis induced-damage, but may lead to an objective reduction in UC disease pathology.
Collapse
Affiliation(s)
| | | | | | - Hyungjun Yang
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute (BCCHRI), University of British Columbia, Vancouver, BC, Canada
| | - Beryl Luk
- Qu Biologics Inc., Vancouver, BC, Canada
| | - Hong T Law
- Qu Biologics Inc., Vancouver, BC, Canada.,Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute (BCCHRI), University of British Columbia, Vancouver, BC, Canada
| | - Vijay Morampudi
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute (BCCHRI), University of British Columbia, Vancouver, BC, Canada
| | - Hong B Yu
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute (BCCHRI), University of British Columbia, Vancouver, BC, Canada
| | | | | | - Brian Bressler
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - John K Marshall
- Department of Medicine and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Richard N Fedorak
- Division of Gastroenterology, University of Alberta, Edmonton, AB, Canada
| | - Jenny Chen
- Qu Biologics Inc., Vancouver, BC, Canada
| | | | - Hal Gunn
- Qu Biologics Inc., Vancouver, BC, Canada
| | - Shirin Kalyan
- Qu Biologics Inc., Vancouver, BC, Canada.,Division of Endocrinology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Bruce A Vallance
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute (BCCHRI), University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
14
|
Jing H, Liu L, Zhou J, Yao H. Inhibition of C-X-C Motif Chemokine 10 (CXCL10) Protects Mice from Cigarette Smoke-Induced Chronic Obstructive Pulmonary Disease. Med Sci Monit 2018; 24:5748-5753. [PMID: 30118441 PMCID: PMC6109363 DOI: 10.12659/msm.909864] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a type of obstructive lung disease characterized by long-term breathing problems and poor airflow. COPD can progress to persistent decline of pulmonary function. This study explored the effect of CXCL10 on COPD induced by cigarette smoke (CS) and its underlying mechanism. Material/Methods Wild-type (WT) mice were randomly assigned into 3 groups: the control group, the CS group, and the intervention group. Mice in the CS group were exposed to CS and mice in the CXCL10 group were exposed to CS and CXCL10 neutralizing antibody. At 24 h after the last CS exposure, body weight and lung functions of each mouse were recorded. Mice were then anesthetized for collecting bronchoalveolar lavage fluid (BALF) and lung tissues. Levels of interleukin-6 (IL-6), keratinocyte chemotactic factor (KC), and monocyte chemoattractant protein-1 (MCP-1) in supernatant and lung homogenate were detected by ELISA and real-time PCR (RT-PCR), respectively. For in vitro experiments, human bronchial epithelial cells 16HBE were stimulated with different concentrations of cigarette smoke extract (CSE) and CXCL10. Cell viability and levels of inflammatory cytokines in the cell supernatant were detected by Cell Counting Kit-8 (CCK-8) and ELISA assay, respectively. Results Our data showed significant weight loss and reduction of lung functions in mice in the CS group compared with those in the control group and intervention group. Increased levels of IL-6, KC, and MCP-1 in BALF and lung homogenate were observed in mice in the model group compared to those in the control group and intervention group. In vitro experiments also confirmed that CXCL10-neutralizing antibody can inhibit CSE-induced cell necrosis and activation of inflammatory cytokines. Conclusions Inhibited CXCL10 protects against COPD progression by decreasing secretion of inflammatory factors, which provides a new direction for the clinical prevention and treatment of COPD.
Collapse
Affiliation(s)
- Hongyu Jing
- Department of Respiratory Medicine, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Lingyun Liu
- Department of Andrology, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Junfeng Zhou
- Department of Dermatology and Venereology, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Hanxin Yao
- Department of Clinical Laboratory, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
15
|
Chen M, Zhang J, Chen Y, Qiu Y, Luo Z, Zhao S, Du L, Tian D. Hydrogen protects lung from hypoxia/re-oxygenation injury by reducing hydroxyl radical production and inhibiting inflammatory responses. Sci Rep 2018; 8:8004. [PMID: 29789753 PMCID: PMC5964155 DOI: 10.1038/s41598-018-26335-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/08/2018] [Indexed: 02/05/2023] Open
Abstract
Here we investigated whether hydrogen can protect the lung from chronic injury induced by hypoxia/re-oxygenation (H/R). We developed a mouse model in which H/R exposure triggered clinically typical lung injury, involving increased alveolar wall thickening, infiltration by neutrophils, consolidation, alveolar hemorrhage, increased levels of inflammatory factors and recruitment of M1 macrophages. All these processes were attenuated in the presence of H2. We found that H/R-induced injury in our mouse model was associated with production of hydroxyl radicals as well as increased levels of colony-stimulating factors and circulating leukocytes. H2 attenuated H/R-induced production of hydroxyl radicals, up-regulation of colony-stimulating factors, and recruitment of neutrophils and M1 macrophages to lung tissues. However, H2 did not substantially affect the H/R-induced increase in erythropoietin or pulmonary artery remodeling. Our results suggest that H2 ameliorates H/R-induced lung injury by inhibiting hydroxyl radical production and inflammation in lungs. It may also prevent colony-stimulating factors from mobilizing progenitors in response to H/R-induced injury.
Collapse
Affiliation(s)
- Meihong Chen
- Department of Respiratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, 511500, China
| | - Jie Zhang
- Department of Pathology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yun Chen
- Department of Respiratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, 511500, China
| | - Yan Qiu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zi Luo
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Department of Anesthesiology, Loudi Central Hospital, Loudi, Hunan, 417000, China
| | - Sixia Zhao
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Department of Anesthesiology, Xiangtan Central Hospital, Xiangtan, Hunan, 411100, China
| | - Lei Du
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dongbo Tian
- Department of Respiratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, 511500, China.
| |
Collapse
|
16
|
The Role of Regulatory T Cell in Nontypeable Haemophilus influenzae-Induced Acute Exacerbation of Chronic Obstructive Pulmonary Disease. Mediators Inflamm 2018; 2018:8387150. [PMID: 29725272 PMCID: PMC5872612 DOI: 10.1155/2018/8387150] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/08/2018] [Accepted: 02/06/2018] [Indexed: 12/20/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with irreversible persistent airflow limitation and enhanced inflammation. The episodes of acute exacerbation (AECOPD) largely depend on the colonized pathogens such as nontypeable Haemophilus influenzae (NTHi), one of the most commonly isolated bacteria. Regulatory T cells (Tregs) are critical in controlling inflammatory immune responses and maintaining tolerance; however, their role in AECOPD is poorly understood. In this study, we hypothesized a regulatory role of Tregs, as NTHi participated in the progress of COPD. Immunological pathogenesis was investigated in a murine COPD model induced by cigarette smoke (CS). NTHi was administrated through intratracheal instillation for an acute exacerbation. Weight loss and lung function decline were observed in smoke-exposed mice. Mice in experimental groups exhibited serious inflammatory responses via histological and cytokine assessment. Expression levels of Tregs and Th17 cells with specific cytokines TGF-β1 and IL-17 were detected to assess the balance of pro-/anti-inflammatory influence partially. Our findings suggested an anti-inflammatory activity of Tregs in CS-induced model. But this activity was suppressed after NTHi administration. Collectively, these data suggested that NTHi might play a necessary role in downregulating Foxp3 to impair the function of Tregs, helping development into AECOPD.
Collapse
|
17
|
Possebon L, de Souza Lima Lebron I, Furlan da Silva L, Tagliaferri Paletta J, Glad BG, Sant’Ana M, Iyomasa-Pilon MM, Ribeiro Souza H, de Souza Costa S, Pereira da Silva Rodriguesa G, Pereira MDL, de Haro Moreno A, Girol AP. Anti-inflammatory actions of herbal medicines in a model of chronic obstructive pulmonary disease induced by cigarette smoke. Biomed Pharmacother 2018; 99:591-597. [DOI: 10.1016/j.biopha.2018.01.106] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 01/18/2018] [Accepted: 01/24/2018] [Indexed: 12/20/2022] Open
|
18
|
Kim W, Lim D, Kim J. p-Coumaric Acid, a Major Active Compound of Bambusae Caulis in Taeniam, Suppresses Cigarette Smoke-Induced Pulmonary Inflammation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:407-421. [PMID: 29433391 DOI: 10.1142/s0192415x18500209] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Formula: see text]-coumaric acid ([Formula: see text]-CA) is a common compound found in medicinal herbs, including Bambusae Caulis in Taeniam (BC). It has been used to treat various diseases in China and Korea. Our previous study demonstrated that BC inhibits pulmonary and intestinal inflammation. In the present study, we used cigarette smoke (CS) to induce lung inflammation in vivo, and investigated the anti-inflammatory effects of [Formula: see text]-CA on CS-induced inflammatory mice model. Mice were treated with BC and [Formula: see text]-CA via oral injection 2[Formula: see text]h before CS exposure. The body weight and the inflammatory cells in the bronchoalveolar lavage fluid were measured. The levels of relative inflammatory factors were confirmed by enzyme-linked immunosorbent assay. The lung histological changes were examined by hematoxylin and eosin staining. Also, the protein level of nuclear factor-[Formula: see text]B (NF-[Formula: see text]B) was evaluated by Western blotting. Our results indicated that BC and [Formula: see text]-CA inhibited CS-induced lung inflammation by regulating pro-inflammatory productions such as cytokines, chemokine, protease and NF-[Formula: see text]B. Consequently, these data demonstrated that [Formula: see text]-CA inhibited pulmonary inflammation by suppressing NF-[Formula: see text]B activity, through which pro-inflammatory mediators were regulated. Therefore, [Formula: see text]-CA, which was shown to be a major component of BC, can be considered as a strong therapeutic candidate for treating pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Woogyeong Kim
- 1 Department of Korean Physiology, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dahae Lim
- 1 Department of Korean Physiology, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jinju Kim
- 1 Department of Korean Physiology, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
19
|
Bazett M, Costa AM, Bosiljcic M, Anderson RM, Alexander MP, Wong SWY, Dhanji S, Chen JM, Pankovich J, Lam S, Sutcliffe S, Gunn H, Kalyan S, Mullins DW. Harnessing innate lung anti-cancer effector functions with a novel bacterial-derived immunotherapy. Oncoimmunology 2017; 7:e1398875. [PMID: 29399400 PMCID: PMC5790356 DOI: 10.1080/2162402x.2017.1398875] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/16/2017] [Accepted: 10/24/2017] [Indexed: 02/06/2023] Open
Abstract
Acute infection is known to induce strong anti-tumor immune responses, but clinical translation has been hindered by the lack of an effective strategy to safely and consistently provoke a therapeutic response. These limitations are overcome with a novel treatment approach involving repeated subcutaneous delivery of a Klebsiella-derived investigational immunotherapeutic, QBKPN. In preclinical models of lung cancer, QBKPN administration consistently showed anti-cancer efficacy, which was dependent on Klebsiella pre-exposure, but was independent of adaptive immunity. Rather, QBKPN induced anti-tumor innate immunity that required NK cells and NKG2D engagement. QBKPN increased NK cells and macrophages in the lungs, altered macrophage polarization, and augmented the production of cytotoxic molecules. An exploratory trial in patients with non-small cell lung cancer demonstrated QBKPN was well tolerated, safe, and induced peripheral immune changes suggestive of macrophage polarization and reduction of PD-1 and PD-L1 expression on leukocytes. These data demonstrate preclinical efficacy, and clinical safety and tolerability, for this cancer immunotherapy strategy that exploits innate anti-tumor immune mechanisms.
Collapse
Affiliation(s)
- Mark Bazett
- Qu Biologics Inc., Vancouver, BC, V5 T 4T5, Canada
| | - Amanda M Costa
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | | | | | - Matthew P Alexander
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Stephanie W Y Wong
- Qu Biologics Inc., Vancouver, BC, V5 T 4T5, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Salim Dhanji
- Qu Biologics Inc., Vancouver, BC, V5 T 4T5, Canada
| | | | | | - Stephen Lam
- BC Cancer Research Center, Vancouver, BC, Canada
| | | | - Hal Gunn
- Qu Biologics Inc., Vancouver, BC, V5 T 4T5, Canada
| | - Shirin Kalyan
- Qu Biologics Inc., Vancouver, BC, V5 T 4T5, Canada.,Department of Medicine, Division of Endocrinology, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - David W Mullins
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Department of Medical Education, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| |
Collapse
|