1
|
Seluk L, Davis AE, Rhoads S, Wechsler ME. Novel asthma treatments: Advancing beyond approved novel step-up therapies for asthma. Ann Allergy Asthma Immunol 2025; 134:9-18. [PMID: 39393433 DOI: 10.1016/j.anai.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/13/2024]
Abstract
Over the past 2 decades, the management of severe asthma has shifted from relying on inhaled corticosteroids and bronchodilators to more precise, targeted approaches. Monoclonal antibodies designed to address specific molecular pathways in asthma have transformed care for patients with severe asthma. Because therapy targeting IgE became the first biologic developed for allergic asthma in 2003, monoclonal antibodies targeting interleukin (IL)-5, IL-5 receptor, IL-4/-13 receptor, and thymic stromal lymphopoietin have been approved for treating difficult-to-treat asthma, improving symptoms, reducing exacerbations, and reducing oral corticosteroid dosing. Despite these advances, many patients continue to experience asthma exacerbations and symptoms and fail to achieve remission. To address this, pharmaceutical companies and researchers are exploring novel therapies targeting different aspects of asthma pathophysiology, including cytokines, enzymes, and cellular pathways. Innovative treatments such as inhaled biologics, ultra-long-acting biologics, and combination biologics are in development. New molecular targets, such as Bruton tyrosine kinase, OX-40 ligand, and Janus kinase, offer promise for addressing unmet needs in asthma care. Although many therapies have failed to get approval for use because of a lack of efficacy, trial design, or toxicity, these experiments still provide insights into asthma's underlying mechanisms. The future of asthma management looks promising, with emerging therapies aiming to improve patient outcomes. The challenge will lie in identifying the right therapy for each patient and developing personalized treatment strategies.
Collapse
Affiliation(s)
- Lior Seluk
- Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, Colorado
| | - Andrea E Davis
- Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, Colorado; Division of Pulmonary Sciences and Critical Care Medicine, The Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sarah Rhoads
- Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, Colorado; Division of Pulmonary Sciences and Critical Care Medicine, The Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael E Wechsler
- Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, Colorado.
| |
Collapse
|
2
|
Desai B, Adrish M, Mohan A, Lugogo NL. Biologics in Asthma: Emerging Biologics. Immunol Allergy Clin North Am 2024; 44:751-763. [PMID: 39389722 DOI: 10.1016/j.iac.2024.07.008] [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] [Indexed: 10/12/2024]
Abstract
Advances in our understanding of asthma pathophysiology have led to the advent of multiple targeted asthma therapies such as biologics. However, partial response to biologics occurs, indicating residual disease activity in some patients. Hence, there exists a need for new therapies that focus on novel pathways, alongside perhaps evaluation of combination biologic therapies and modulators of downstream cytokine activation. Therefore, although our current focus is on biologics; it is critical to take a more holistic approach including consideration for nonbiologic therapies that have the potential to significantly advance asthma care.
Collapse
Affiliation(s)
- Brinda Desai
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Muhammad Adrish
- Department of Pulmonary & Critical Care, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Arjun Mohan
- Department of Medicine, University of Michigan, 300 North Ingalls Street, Suite 2d21, Ann Arbor, MI 48109, USA
| | - Njira L Lugogo
- Department of Medicine, University of Michigan, 300 North Ingalls Street, Suite 2c40, Ann Arbor, MI 48109, USA.
| |
Collapse
|
3
|
Rzewińska A, Szlęk J, Dąbrowski D, Juszczyk E, Mróz K, Räikkönen H, Siven M, Wieczorek M, Dorożyński P. Development of a Formulation and In Vitro Evaluation of a Pulmonary Drug Delivery System for a Novel Janus Kinase (JAK) Inhibitor, CPL409116. Pharmaceutics 2024; 16:1157. [PMID: 39339194 PMCID: PMC11435004 DOI: 10.3390/pharmaceutics16091157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/13/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The pursuit of targeted therapies for cytokine-dependent diseases has led to the discovery of Janus kinase (JAK) inhibitors, a promising class of drugs. Among them, CPL409116, a selective dual JAK and rho-associated protein kinase inhibitor (ROCK), has demonstrated potential for treating conditions such as pulmonary fibrosis exacerbated by the COVID-19 pandemic. This study investigated the feasibility of delivering CPL409116 via inhalation, with the aim of minimizing the systemic adverse effects associated with oral administration. Two micronization methods, jet milling and spray drying, were assessed for CPL409116, with spray drying chosen for its ability to produce an amorphous form of the compound. Moreover, parameters such as the mixing energy, drug load, and force control agent significantly influenced the fine particle fraction (FPF), a critical parameter for pulmonary drug delivery. This study provides insights into optimizing the formulation parameters to enhance the delivery efficiency of CPL409116 to the lungs, offering potential for improved therapeutic outcomes in cytokine-dependent pulmonary diseases.
Collapse
Affiliation(s)
- Aleksandra Rzewińska
- Finished Dosage Forms Department, Research and Development Center, Celon Pharma S.A., Marymoncka 15, 05-052 Kazuń Nowy, Poland
- Department of Drug Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warszawa, Poland
| | - Jakub Szlęk
- Chair and Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Damian Dąbrowski
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warszawa, Poland
| | - Ewelina Juszczyk
- Finished Dosage Forms Department, Research and Development Center, Celon Pharma S.A., Marymoncka 15, 05-052 Kazuń Nowy, Poland
| | - Katarzyna Mróz
- Finished Dosage Forms Department, Research and Development Center, Celon Pharma S.A., Marymoncka 15, 05-052 Kazuń Nowy, Poland
| | - Heikki Räikkönen
- Faculty of Pharmacy, University of Helsinki, Viikinkaari 5, 00014 Helsinki, Finland
| | - Mia Siven
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014 Helsinki, Finland
- Helsinki Institute of Sustainability Science HELSUS, University of Helsinki, 00014 Helsinki, Finland
| | - Maciej Wieczorek
- Research and Development Center, Celon Pharma S.A., Marymoncka 15, 05-052 Kazuń Nowy, Poland
| | - Przemysław Dorożyński
- Department of Drug Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warszawa, Poland
- Department of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
4
|
Dierick BJH, Eikholt AA, van de Hei SJ, Muris JWM, Kerstjens HAM, van Boven JFM. Reshaping respiratory care: potential advances in inhaled pharmacotherapy in asthma. Expert Opin Pharmacother 2024; 25:1507-1516. [PMID: 39099418 DOI: 10.1080/14656566.2024.2389258] [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: 05/31/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/06/2024]
Abstract
INTRODUCTION Asthma is a common disease with a global burden of 358 million patients. Despite improvements in pharmacological and non-pharmacological treatments, many patients still do not achieve complete asthma control. Therefore, innovative pharmacotherapy is important. AREAS COVERED Following a semi-structured search in Pubmed, an overview of advances in inhaled asthma therapy is provided, looking at innovations in digital inhalers, eco-friendly inhalers and novel inhaled biologic therapies, antibiotics and vaccines, as well as other potential novel asthma therapy targets. EXPERT OPINION Digital inhalers, sending reminders and monitoring inhalation technique electronically, can support medication adherence and improve asthma control. To reduce the global warming potential of traditional aerosols used in pressurized metered-dose inhalers (HFA-134a, HFA-227ea), greener alternatives are under development (HFA-152a, HFO-1234ze) that are expected to be available by 2025. Current pharmacological advances in asthma therapy are mainly achieved by novel biologicals (anti-IgE, anti-IL5, anti-IL4/13, and anti-TSLP) targeting specific severe asthma phenotypes. While injection is the usual administration route for biologics and vaccines used in asthma, inhalation is an option being explored, although several (mainly formulation) challenges need to be overcome. Other potential novel future inhaled asthma therapies include anti-IL-33/ST2 biologicals and JAK inhibitors, all still requiring more clinical evidence.
Collapse
Affiliation(s)
- Boudewijn J H Dierick
- Department of Clinical Pharmacy & Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Primary and Long-term Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Amber A Eikholt
- Department of Clinical Pharmacy & Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Susanne J van de Hei
- Department of Clinical Pharmacy & Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Jean W M Muris
- Department of General Practice, Research Institute CAPHRI, Maastricht University, Maastricht, The Netherlands
| | - Huib A M Kerstjens
- Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Pulmonary Diseases and Tuberculosis, University of Groningen University Medical Center Groningen, Groningen, The Netherlands
| | - Job F M van Boven
- Department of Clinical Pharmacy & Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| |
Collapse
|
5
|
Xiao X, Lei Y, Yao T, Huang T, Yan P, Cao L, Cao Y. PM 10 exposure induces bronchial hyperresponsiveness by upreguating acetylcholine muscarinic 3 receptor. Toxicol Appl Pharmacol 2024; 490:117035. [PMID: 39019094 DOI: 10.1016/j.taap.2024.117035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
Exposure to particulate matter (PM10) can induce respiratory diseases that are closely related to bronchial hyperresponsiveness. However, the involved mechanism remains to be fully elucidated. This study aimed to demonstrate the effects of PM10 on the acetylcholine muscarinic 3 receptor (CHRM3) expression and the role of the ERK1/2 pathway in rat bronchial smooth muscle. A whole-body PM10 exposure system was used to stimulate bronchial hyperresponsiveness in rats for 2 and 4 months, accompanied by MEK1/2 inhibitor U0126 injection. The whole-body plethysmography system and myography were used to detect the pulmonary and bronchoconstrictor function, respectively. The mRNA and protein levels were determined by Western blotting, qPCR, and immunofluorescence. Enzyme-linked immunosorbent assay was used to detect the inflammatory cytokines. Compared with the filtered air group, 4 months of PM10 exposure significantly increased CHRM3-mediated pulmonary function and bronchial constriction, elevated CHRM3 mRNA and protein expression levels on bronchial smooth muscle, then induced bronchial hyperreactivity. Additionally, 4 months of PM10 exposure caused an increase in ERK1/2 phosphorylation and increased the secretion of inflammatory factors in bronchoalveolar lavage fluid. Treatment with the MEK1/2 inhibitor, U0126 inhibited the PM10 exposure-induced phosphorylation of the ERK1/2 pathway, thereby reducing the PM10 exposure-induced upregulation of CHRM3 in bronchial smooth muscle and CHRM3-mediated bronchoconstriction. U0126 could rescue PM10 exposure-induced pathological changes in the bronchus. In conclusion, PM10 exposure can induce bronchial hyperresponsiveness in rats by upregulating CHRM3, and the ERK1/2 pathway may be involved in this process. These findings could reveal a potential therapeutic target for air pollution induced respiratory diseases.
Collapse
Affiliation(s)
- Xue Xiao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Yali Lei
- Shanghai Environmental Monitoring Center, Shanghai 200232, China
| | - Tong Yao
- Precision Medical Institute, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West 5th Road, 710004, China
| | - Tingting Huang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Pingping Yan
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Lei Cao
- Precision Medical Institute, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West 5th Road, 710004, China.
| | - Yongxiao Cao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
6
|
Cugudda A, La Manna S, Marasco D. Are peptidomimetics the compounds of choice for developing new modulators of the JAK-STAT pathway? Front Immunol 2024; 15:1406886. [PMID: 38983855 PMCID: PMC11232365 DOI: 10.3389/fimmu.2024.1406886] [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: 03/25/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024] Open
Abstract
Protein-protein interactions (PPIs) play critical roles in a wide range of biological processes including the dysregulation of cellular pathways leading to the loss of cell function, which in turn leads to diseases. The dysfunction of several signaling pathways is linked to the insurgence of pathological processes such as inflammation, cancer development and neurodegeneration. Thus, there is an urgent need for novel chemical modulators of dysregulated PPIs to drive progress in targeted therapies. Several PPIs have been targeted by bioactive compounds, and, often, to properly cover interacting protein regions and improve the biological activities of modulators, a particular focus concerns the employment of macrocycles as proteomimetics. Indeed, for their physicochemical properties, they occupy an intermediate space between small organic molecules and macromolecular proteins and are prominent in the drug discovery process. Peptide macrocycles can modulate fundamental biological mechanisms and here we will focus on peptidomimetics active on the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathways.
Collapse
Affiliation(s)
| | | | - Daniela Marasco
- Department of Pharmacy, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
7
|
Rinderknecht CH, Ning M, Wu C, Wilson MS, Gampe C. Designing inhaled small molecule drugs for severe respiratory diseases: an overview of the challenges and opportunities. Expert Opin Drug Discov 2024; 19:493-506. [PMID: 38407117 DOI: 10.1080/17460441.2024.2319049] [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: 11/27/2023] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
INTRODUCTION Inhaled drugs offer advantages for the treatment of respiratory diseases over oral drugs by delivering the drug directly to the lung, thus improving the therapeutic index. There is an unmet medical need for novel therapies for lung diseases, exacerbated by a multitude of challenges for the design of inhaled small molecule drugs. AREAS COVERED The authors review the challenges and opportunities for the design of inhaled drugs for respiratory diseases with a focus on new target discovery, medicinal chemistry, and pharmacokinetic, pharmacodynamic, and toxicological evaluation of drug candidates. EXPERT OPINION Inhaled drug discovery is facing multiple unique challenges. Novel biological targets are scarce, as is the guidance for medicinal chemistry teams to design compounds with inhalation-compatible features. It is exceedingly difficult to establish a PK/PD relationship given the complexity of pulmonary PK and the impact of physical properties of the drug substance on PK. PK, PD and toxicology studies are technically challenging and require large amounts of drug substance. Despite the current challenges, the authors foresee that the design of inhaled drugs will be facilitated in the future by our increasing understanding of pathobiology, emerging medicinal chemistry guidelines, advances in drug formulation, PBPK models, and in vitro toxicology assays.
Collapse
Affiliation(s)
| | - Miaoran Ning
- Drug Metabolism and Pharmacokinetics, gRED, Genentech, South San Francisco, CA, USA
| | - Connie Wu
- Development Sciences Safety Assessment, Genentech, South San Francisco, CA, USA
| | - Mark S Wilson
- Discovery Immunology, gRED, Genentech, South San Francisco, CA, USA
| | - Christian Gampe
- Discovery Chemistry, gRED, Genentech, South San Francisco, CA, USA
| |
Collapse
|
8
|
Ullas S, Sinclair C. Applications of Flow Cytometry in Drug Discovery and Translational Research. Int J Mol Sci 2024; 25:3851. [PMID: 38612661 PMCID: PMC11011675 DOI: 10.3390/ijms25073851] [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: 02/21/2024] [Revised: 03/23/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Flow cytometry is a mainstay technique in cell biology research, where it is used for phenotypic analysis of mixed cell populations. Quantitative approaches have unlocked a deeper value of flow cytometry in drug discovery research. As the number of drug modalities and druggable mechanisms increases, there is an increasing drive to identify meaningful biomarkers, evaluate the relationship between pharmacokinetics and pharmacodynamics (PK/PD), and translate these insights into the evaluation of patients enrolled in early clinical trials. In this review, we discuss emerging roles for flow cytometry in the translational setting that supports the transition and evaluation of novel compounds in the clinic.
Collapse
Affiliation(s)
| | - Charles Sinclair
- Flagship Pioneering, 140 First Street, Cambridge, MA 02141, USA;
| |
Collapse
|
9
|
Bonelli M, Kerschbaumer A, Kastrati K, Ghoreschi K, Gadina M, Heinz LX, Smolen JS, Aletaha D, O'Shea J, Laurence A. Selectivity, efficacy and safety of JAKinibs: new evidence for a still evolving story. Ann Rheum Dis 2024; 83:139-160. [PMID: 37923366 PMCID: PMC10850682 DOI: 10.1136/ard-2023-223850] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/18/2023] [Indexed: 11/07/2023]
Abstract
Fundamental insight gained over the last decades led to the discovery of cytokines as pivotal drivers of inflammatory diseases such as rheumatoid arthritis, psoriasis/psoriasis arthritis, inflammatory bowel diseases, atopic dermatitis and spondylarthritis. A deeper understanding of the pro-inflammatory and anti-inflammatory effects of various cytokines has prompted new cytokine-targeting therapies, which revolutionised the treatment options in the last years for patients with inflammatory disorders. Disease-associated immune responses typically involve a complex interplay of multiple cytokines. Therefore, blockade of one single cytokine does not necessarily lead to a persistent remission in all patients with inflammatory disorders and fostered new therapeutic strategies targeting intracellular pathways shared by multiple cytokines. By inhibiting JAK-STAT signalling pathways common to families of cytokines, JAK-inhibitors (JAKinibs) have created a new paradigm for the treatment of inflammatory diseases. Multiple agents have been approved for various disorders and more are being investigated for several new indications. Second-generation selective JAKinibs have been devised with the aim to achieve an increased selectivity and a possible reduced risk of side effects. In the current review, we will summarise the current body of evidence of pan versus selective JAKinibs and the most recent insights on new side effects and indications, including COVID-19.
Collapse
Affiliation(s)
- Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Kastriot Kastrati
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Massimo Gadina
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Leonhard X Heinz
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - John O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Arian Laurence
- Translational Gastroenterology Unit, Department of Haematology, University College Hospital, UCLH Hospitals NHS Trust, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
Wu Z, Luo Z, Sun W, Shi Y, Ding Q. Integrating Network Pharmacology and Experimental Validation to Elucidate the Mechanism of Jiegeng Decoction in Improving Allergic Asthma. ACS OMEGA 2023; 8:48081-48090. [PMID: 38144091 PMCID: PMC10733997 DOI: 10.1021/acsomega.3c06914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/26/2023]
Abstract
Allergic asthma is a prevalent form of asthma that is characterized primarily by airway inflammation. Jiegeng decoction (JGT) is a traditional Chinese herbal formula known for its anti-inflammatory properties and has been used to treat respiratory diseases for centuries. This study aimed to investigate the biological effects and mechanisms of action of JGT in improving allergic asthma. An experimental allergic asthma mouse model was established using ovalbumin. The results showed that JGT significantly improved inflammation cell infiltration in the lung tissue of allergic asthmatic mice and the inflammatory environment of Th2 cells in the bronchoalveolar lavage fluid while also reducing serum IgE levels. Subsequently, 38 components of JGT were identified through liquid chromatography-mass spectrometry. Network pharmacology revealed that regulating inflammation and immune responses is the primary biological process by which JGT improves allergic asthma, with Th2 cell differentiation and the JAK-STAT signaling pathway being the key mechanisms of action. Finally, qPCR, flow cytometry, and Western blotting were used to validate that JGT inhibited Th2 cell differentiation by blocking the JAK1-STAT6 signaling pathway in CD4+ T cells, ultimately improving allergic asthma. This study provides a novel perspective on the therapeutic potential of JGT in the treatment of allergic asthma.
Collapse
Affiliation(s)
- Zhihai Wu
- School
of Life Sciences, Beijing University of
Chinese Medicine, Beijing 100029, China
| | - Zhiqiang Luo
- National
Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di
Herbs, Beijing 100700, China
- State
Key Laboratory of Dao-di Herbs, National Resource Center for Chinese
Materia Medica, China Academy of Chinese
Medical Sciences, Beijing 100700, China
| | - Wen Sun
- School
of Life Sciences, Beijing University of
Chinese Medicine, Beijing 100029, China
| | - Yuanyuan Shi
- School
of Life Sciences, Beijing University of
Chinese Medicine, Beijing 100029, China
- Shenzhen
Research Institute, Beijing University of
Chinese Medicine, Shenzhen 518118, China
| | - Qi Ding
- Shenzhen
Research Institute, Beijing University of
Chinese Medicine, Shenzhen 518118, China
| |
Collapse
|
11
|
Lin Y, Wu Y, Ma F, Shan C, Ma J, Li W, Pan H, Miao X, Liu J, Wang X, Ni Z. Exploration of the mechanism of Qi-Xian decoction in asthmatic mice using metabolomics combined with network pharmacology. Front Mol Biosci 2023; 10:1263962. [PMID: 38155957 PMCID: PMC10753777 DOI: 10.3389/fmolb.2023.1263962] [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: 07/20/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
Introduction: Qi-Xian Decoction (QXD), a traditional Chinese medicine (TCM) formula consisting of eight herbs, has been clinically used to treat asthma. However, the underlying mechanisms have not been completely elucidated. This study aimed to combine metabolomics and network pharmacology to reveal the mechanism of action of QXD in asthma treatment. Methods: An ovalbumin (OVA)-induced asthma mouse model was constructed to evaluate the therapeutic effects of QXD. Serum metabolomics and network pharmacology were combined to study the mechanism of anti-asthma action as well as the potential target, and related biological functions were validated. Results: The QXD treatment has demonstrated significant protective effects in OVA-induced asthmatic mice, as evidenced by its ability to inhibit inflammation, IgE, mucus overproduction, and airway hyperreactivity (AHR). Metabolomic analysis has revealed a total of 140 differential metabolites associated with QXD treatment. In addition, network pharmacology has identified 126 genes that are linked to the effects of QXD, including TNF, IL-6, IL1β, STAT3, MMP9, EGFR, JUN, CCL2, TLR4, MAPK3 and MAPK8. Through comprehensive gene-metabolite interaction network analysis, seven key metabolites have been identified and associated with the potential anti-asthmatic effect of QXD, with palmitic acid (PA) being the most notable among them. In vitro validation studies have confirmed the gene-metabolite interaction involving PA, IL-6, and MAPK8. Furthermore, our research has demonstrated that QXD treatment can effectively inhibit PA-promoted IL-6 expression in MH-S cells and reduce PA concentration in OVA-induced asthmatic mice. Conclusion: The regulation of metabolic pathways by QXD was found to be associated with its anti-asthmatic action, which provides insight into the mechanism of QXD in treating asthma.
Collapse
Affiliation(s)
- Yuhua Lin
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Wu
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fuqi Ma
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cuiting Shan
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jialu Ma
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenguan Li
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huayang Pan
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiayi Miao
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinjin Liu
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiongbiao Wang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenhua Ni
- Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Castelo-Soccio L, Kim H, Gadina M, Schwartzberg PL, Laurence A, O'Shea JJ. Protein kinases: drug targets for immunological disorders. Nat Rev Immunol 2023; 23:787-806. [PMID: 37188939 PMCID: PMC10184645 DOI: 10.1038/s41577-023-00877-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 05/17/2023]
Abstract
Protein kinases play a major role in cellular activation processes, including signal transduction by diverse immunoreceptors. Given their roles in cell growth and death and in the production of inflammatory mediators, targeting kinases has proven to be an effective treatment strategy, initially as anticancer therapies, but shortly thereafter in immune-mediated diseases. Herein, we provide an overview of the status of small molecule inhibitors specifically generated to target protein kinases relevant to immune cell function, with an emphasis on those approved for the treatment of immune-mediated diseases. The development of inhibitors of Janus kinases that target cytokine receptor signalling has been a particularly active area, with Janus kinase inhibitors being approved for the treatment of multiple autoimmune and allergic diseases as well as COVID-19. In addition, TEC family kinase inhibitors (including Bruton's tyrosine kinase inhibitors) targeting antigen receptor signalling have been approved for haematological malignancies and graft versus host disease. This experience provides multiple important lessons regarding the importance (or not) of selectivity and the limits to which genetic information informs efficacy and safety. Many new agents are being generated, along with new approaches for targeting kinases.
Collapse
Affiliation(s)
- Leslie Castelo-Soccio
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hanna Kim
- Juvenile Myositis Pathogenesis and Therapeutics Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Massimo Gadina
- Translational Immunology Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela L Schwartzberg
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arian Laurence
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK.
- University College London Hospitals NHS Foundation Trust, London, UK.
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
13
|
Nilsson M, Berggren K, Berglund S, Cerboni S, Collins M, Dahl G, Elmqvist D, Grimster NP, Hendrickx R, Johansson JR, Kettle JG, Lepistö M, Rhedin M, Smailagic A, Su Q, Wennberg T, Wu A, Österlund T, Naessens T, Mitra S. Discovery of the Potent and Selective Inhaled Janus Kinase 1 Inhibitor AZD4604 and Its Preclinical Characterization. J Med Chem 2023; 66:13400-13415. [PMID: 37738648 DOI: 10.1021/acs.jmedchem.3c00554] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
JAK-STAT cytokines are critical in regulating immunity. Persistent activation of JAK-STAT signaling pathways by cytokines drives chronic inflammatory diseases such as asthma. Herein, we report on the discovery of a highly JAK1-selective, ATP-competitive series of inhibitors having a 1000-fold selectivity over other JAK family members and the approach used to identify compounds suitable for inhaled administration. Ultimately, compound 16 was selected as the clinical candidate, and upon dry powder inhalation, we could demonstrate a high local concentration in the lung as well as low plasma concentrations, suggesting no systemic JAK1 target engagement. Compound 16 has progressed into clinical trials. Using 16, we found JAK1 inhibition to be more efficacious than JAK3 inhibition in IL-4-driven Th2 asthma.
Collapse
Affiliation(s)
- Magnus Nilsson
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Kristina Berggren
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Susanne Berglund
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Silvia Cerboni
- Bioscience, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Mia Collins
- Bioscience, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Göran Dahl
- Structure and Biophysics, Research and Early Development, Discovery Science, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - David Elmqvist
- Early Product Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Neil P Grimster
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Ramon Hendrickx
- DMPK, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Johan R Johansson
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Jason G Kettle
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Matti Lepistö
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Magdalena Rhedin
- Bioscience, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Amir Smailagic
- Bioscience, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Qibin Su
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Tiiu Wennberg
- Bioscience, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Allan Wu
- Discovery Sciences, R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Torben Österlund
- Mechanistic Biology & Profiling, Research and Early Development, Discovery Science, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Thomas Naessens
- Bioscience, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Suman Mitra
- Bioscience, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| |
Collapse
|
14
|
Mattsson J, Israelsson E, Björhall K, Yrlid LF, Thörn K, Thorén A, Toledo EA, Jinton L, Öberg L, Wingren C, Tapani S, Jackson SG, Skogberg G, Lundqvist AJ, Hendrickx R, Cavallin A, Österlund T, Grimster NP, Nilsson M, Åstrand A. Selective Janus kinase 1 inhibition resolves inflammation and restores hair growth offering a viable treatment option for alopecia areata. SKIN HEALTH AND DISEASE 2023; 3:e209. [PMID: 37275428 PMCID: PMC10233092 DOI: 10.1002/ski2.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 01/30/2023]
Abstract
Background Janus Kinase (JAK) inhibition has recently demonstrated therapeutic efficacy in both restoring hair growth and resolving inflammation in Alopecia Areata (AA). These effects are dose dependent and mainly efficacious at ranges close to a questionable risk profile. Objectives We explored the possibility to separate the beneficial and adverse effects of JAK inhibition by selectively inhibiting JAK1 and thereby avoiding side effects associated with JAK2 blockade. Methods The C3H/HeJ mouse model of AA was used to demonstrate therapeutic efficacy in vivo with different regimens of a selection of JAK inhibitors in regards to systemic versus local drug exposure. Human peripheral blood lymphocytes were stimulated in vitro to demonstrate translation to the human situation. Results We demonstrate that selective inhibition of JAK1 produces fast resolution of inflammation and complete restoration of hair growth in the C3H/HeJ mouse model of AA. Furthermore, we show that topical treatment does not restore hair growth and that treatment needs to be extended well beyond that of restored hair growth in order to reach treatment-free remission. For translatability to human disease, we show that cytokines involved in AA pathogenesis are similarly inhibited by selective JAK1 and pan-JAK inhibition in stimulated human peripheral lymphocytes and specifically in CD8+ T cells. Conclusion This study demonstrates that systemic exposure is required for efficacy in AA and we propose that a selective JAK1 inhibitor will offer a treatment option with a superior safety profile to pan-JAK inhibitors for these patients.
Collapse
Affiliation(s)
- Johan Mattsson
- Bioscience, Research and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Elisabeth Israelsson
- Translational Science and Experimental MedicineResearch and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Karin Björhall
- Bioscience, Research and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Linda Fahlén Yrlid
- Bioscience, Research and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Kristoffer Thörn
- Translational Science and Experimental MedicineResearch and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Anna Thorén
- Animal Science and TechnologiesClinical Pharmacology & Safety SciencesBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Emelie Andersén Toledo
- Animal Science and TechnologiesClinical Pharmacology & Safety SciencesBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Lisa Jinton
- Bioscience, Research and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Lisa Öberg
- Translational Science and Experimental MedicineResearch and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Cecilia Wingren
- Bioscience, Research and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Sofia Tapani
- Early Biometrics & Statistical InnovationData Science & AIBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Sonya G. Jackson
- Bioscience, Research and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Gabriel Skogberg
- Bioscience, Research and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Anders J. Lundqvist
- Drug Metabolism & PharmacokineticsResearch and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Ramon Hendrickx
- Drug Metabolism & PharmacokineticsResearch and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Anders Cavallin
- Bioscience, Research and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Torben Österlund
- The Discovery Sciences UnitBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | | | - Magnus Nilsson
- Medicinal ChemistryBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Annika Åstrand
- Bioscience, Research and Early DevelopmentRespiratory & Immunology (R&I)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| |
Collapse
|
15
|
McClean N, Hasday JD, Shapiro P. Progress in the development of kinase inhibitors for treating asthma and COPD. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 98:145-178. [PMID: 37524486 DOI: 10.1016/bs.apha.2023.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Current therapies to mitigate inflammatory responses involved in airway remodeling and associated pathological features of asthma and chronic obstructive pulmonary disease (COPD) are limited and largely ineffective. Inflammation and the release of cytokines and growth factors activate kinase signaling pathways that mediate changes in airway mesenchymal cells such as airway smooth muscle cells and lung fibroblasts. Proliferative and secretory changes in mesenchymal cells exacerbate the inflammatory response and promote airway remodeling, which is often characterized by increased airway smooth muscle mass, airway hyperreactivity, increased mucus secretion, and lung fibrosis. Thus, inhibition of relevant kinases has been viewed as a potential therapeutic approach to mitigate the debilitating and, thus far, irreversible airway remodeling that occurs in asthma and COPD. Despite FDA approval of several kinase inhibitors for the treatment of proliferative disorders, such as cancer and inflammation associated with rheumatoid arthritis and ulcerative colitis, none of these drugs have been approved to treat asthma or COPD. This review will provide a brief overview of the role kinases play in the pathology of asthma and COPD and an update on the status of kinase inhibitors currently in clinical trials for the treatment of obstructive pulmonary disease. In addition, potential issues associated with the current kinase inhibitors, which have limited their success as therapeutic agents in treating asthma or COPD, and alternative approaches to target kinase functions will be discussed.
Collapse
Affiliation(s)
- Nathaniel McClean
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Jeffery D Hasday
- Department of Medicine, Division of Pulmonary Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States.
| |
Collapse
|