1
|
Luccioli S, Seabol L. Anaphylaxis in children: Latest insights. Allergy Asthma Proc 2025; 46:168-184. [PMID: 40380371 DOI: 10.2500/aap.2025.46.250005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2025]
Abstract
Background: The diagnosis and management of anaphylaxis in pediatric populations can be a particularly formidable challenge due to its variable definitions and atypical symptom presentation, which can often masquerade as other conditions. This complexity often leads to delays in early recognition and timely intervention. Most pediatric anaphylaxis guidelines emphasize the importance of identifying and avoiding triggers, ensuring accurate dosing and prompt administration of epinephrine to prevent severe complications. There is also growing scientific interest in strategies to intervene early in food allergy development to prevent allergies and protect infants and children from severe allergic reactions. Objective: This report aimed to review key aspects of the pathophysiology, epidemiology, management, and prevention of anaphylaxis in the pediatric population. Also, approved treatment modalities and future research to treat and prevent anaphylactic reactions are discussed. Methods: A review of the medical literature was conducted by using terms that included anaphylaxis, severe allergic reaction, pediatric, prevalence, desensitization, and immunotherapy. Results: Food allergies remain the leading trigger of pediatric anaphylaxis, followed by Hymenoptera venom, whereas drug allergies are less common in children compared with adults. A review of the literature underscores the importance of recognizing early signs and symptoms of anaphylaxis, particularly in preverbal infants, of identifying and eliminating key triggers and of prompt epinephrine administration in the immediate management of pediatric anaphylaxis. Advances in oral immunotherapy and other treatments (e.g., biologics) provide new management options. Notably, anti-immunoglobulin E therapy with omalizumab has shown substantial protection against reactions to accidental food exposure in children as young as 1 year old and with food allergy. Conclusion: This report explores critical aspects of anaphylaxis that affect allergic diseases in infants and children. Gaining a deeper understanding of age-specific triggers and the diverse symptoms of anaphylaxis will significantly enhance diagnosis, treatment, and prevention strategies, ultimately improving the timeliness of interventions. Recent approvals of novel therapies for food allergies, along with promising developments for future treatment and prevention of anaphylaxis in pediatric populations, hold exciting potential for better management of these conditions.
Collapse
Affiliation(s)
- Stefano Luccioli
- From the Division of Rheumatology, Immunology and Allergy, Department of Medicine, Georgetown School of Medicine, Washington, D.C. and
| | - Liliana Seabol
- Department of Medicine, Georgetown School of Medicine, Washington, D.C
| |
Collapse
|
2
|
Alchoueiry M, Mansour H, Khabibullin D, Han T, Chaturantabut S, Bzeih W, Tang Y, Williams JF, Hirsch MS, Priolo C, Sellers WR, Henske EP. Targeting KIT With Antibody-Drug Conjugates in Chromophobe Renal Cell Carcinoma. Clin Genitourin Cancer 2025; 23:102359. [PMID: 40408838 DOI: 10.1016/j.clgc.2025.102359] [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: 04/01/2025] [Accepted: 04/12/2025] [Indexed: 05/25/2025]
Abstract
INTRODUCTION Chromophobe renal cell carcinoma (ChRCC) is the third most common type of RCC. There are no proven therapies for patients with metastatic ChRCC, with a median survival of 27 months. KIT (CD117) is a membrane-associated tyrosine kinase receptor. Antibody-drug conjugates (ADC) targeting KIT were previously found to be safe and effective in preclinical models of KIT-positive cancers but have not been tested in ChRCC. RESULTS In The Cancer Genome Atlas, KIT mRNA expression is higher in ChRCC than any other tumor type with the mean expression 12 times higher than matched normal kidney. Of the 15 metastatic ChRCC specimens stained for KIT at our institution, 87% were positive. In single-cell RNA sequencing data, KIT and SCF, the KIT ligand, are co-expressed in ChRCC tumor cells. We found that KIT mRNA expression is significantly higher in ChRCC-derived cells compared to clear cell renal cell carcinoma (ccRCC)-derived cells and normal kidney cells. Western blot analysis confirmed KIT expression in 5 ChRCC cell lines. Despite high KIT expression, knockdown of KIT or treatment with KIT targeting tyrosine kinase inhibitors did not decrease ChRCC cell proliferation. LOP628, a KIT ADC, decreased the viability of the ChRCC-derived cells by ∼60% with no effect on ccRCC cells. CONCLUSION Together, these data demonstrate that KIT is a viable therapeutic target for antibody-drug conjugates in ChRCC, providing a foundation for further investigation into KIT-targeted therapies.
Collapse
Affiliation(s)
- Michel Alchoueiry
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hadi Mansour
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Damir Khabibullin
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Tiegang Han
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Wafaa Bzeih
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Yan Tang
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jessica F Williams
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Carmen Priolo
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Elizabeth P Henske
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
3
|
Miyamoto K, Miller RM, Voors‐Pette C, Oosterhaven JAF, van den Dobbelsteen M, Mihara K, Geldof M, Sato Y, Matsuda N, Kirita S, Sawa M, Arimura A. Safety, pharmacokinetics, and pharmacodynamics of sofnobrutinib, a novel non-covalent BTK inhibitor, in healthy subjects: First-in-human phase I study. Clin Transl Sci 2024; 17:e70060. [PMID: 39523516 PMCID: PMC11551066 DOI: 10.1111/cts.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/08/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
Bruton's tyrosine kinase (BTK) is a potential therapeutic target for allergic and autoimmune diseases. This first-in-human phase I study evaluated safety, pharmacokinetic, and pharmacodynamic profiles of sofnobrutinib (formerly AS-0871), a highly selective, orally available, non-covalent BTK inhibitor, in healthy adult subjects. Single ascending doses (SAD; 5-900 mg) and multiple ascending doses (MAD; 50-300 mg twice daily [b.i.d.] for 14 days [morning dose only on Day 14]) of sofnobrutinib were tested. In the entire study, all adverse events (AEs) were mild or moderate, and no apparent dose-proportional trend in severity or frequency was observed. No serious treatment-emergent AEs, cardiac arrythmias, or bleeding-related AEs were reported. In the SAD part, sofnobrutinib exhibited approximately dose-dependent systemic exposures up to 900 mg with rapid absorption (median time to maximum concentration of 2.50-4.00 h) and gradual decline (mean half-lives of 3.7-9.0 h). In the MAD part, sofnobrutinib showed low accumulation after multiple dosing (mean accumulation ratios of ≤1.54) and reached a steady state on ≤Day 7. Single dosing of sofnobrutinib rapidly and dose-dependently suppressed basophil and B-cell activations in ex vivo whole blood assays. Multiple dosing of sofnobrutinib achieved 50.8%-79.4%, 67.6%-93.6%, and 90.1%-98.0% inhibition of basophil activation during the dosing interval of 50, 150, and 300 mg b.i.d., respectively. Based on pharmacokinetic-pharmacodynamic analysis, half-maximal inhibitory concentration (IC50) of sofnobrutinib for basophil activation was 54.06 and 57.01 ng/mL in the SAD and MAD parts, respectively. Similarly, IC50 for B-cell activation was 187.21 ng/mL. These data support further investigation of sofnobrutinib in allergic and autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Akinori Arimura
- CarnaBio USA, Inc.South San FranciscoCaliforniaUSA
- Carna Biosciences, Inc.KobeJapan
| |
Collapse
|
4
|
Ghelli C, Costanzo G, Canonica GW, Heffler E, Paoletti G. New evidence in food allergies treatment. Curr Opin Allergy Clin Immunol 2024; 24:251-256. [PMID: 38814736 DOI: 10.1097/aci.0000000000000999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
PURPOSE OF REVIEW To acknowledge, the newly available treatments for food allergy described in the latest scientific literature, such as oral immunotherapy (OIT), biologics and the combination of them in managing patients with IgE-mediated food allergies. RECENT FINDINGS Recent studies suggest that OIT and biologics, alone or together, can have a role as disease-modifying treatments for food allergies. The FDA has recently approved omalizumab as a treatment for food allergy. Other biologics are currently under evaluation and further studies are needed to assess the efficacy and safety of these therapies. SUMMARY The allergology scenario is rapidly evolving, the recent introduction and approval of new therapeutic strategies such as biotechnological drugs and allergen immunotherapy is changing the therapeutic paradigm: we are witnessing a shift from a strategy based on avoiding the trigger and reversing an allergic reaction already in progress, to one that aims to modify the natural history of the disease by acting on the immunological mechanisms that determine it. This approach is consistent with the modern perspective of a personalized patient-tailored medicine. In this opinion review, we will provide a brief analysis of current and future therapeutic options for IgE-mediated food allergy, focusing on OIT, biologics and their combination.
Collapse
Affiliation(s)
- Chiara Ghelli
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | | | | | | |
Collapse
|
5
|
Lin EV, Suresh RV, Dispenza MC. Bruton's tyrosine kinase inhibition for the treatment of allergic disorders. Ann Allergy Asthma Immunol 2024; 133:33-42. [PMID: 38492772 PMCID: PMC11222055 DOI: 10.1016/j.anai.2024.03.002] [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: 01/29/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
IgE signaling through its high-affinity receptor FcεRI is central to the pathogenesis of numerous allergic disorders. Oral inhibitors of Bruton's tyrosine kinase (BTKis), which are currently Food and Drug Administration-approved for treating B cell malignancies, broadly inhibit the FcεRI pathway in human mast cells and basophils, and therefore may be effective allergen-independent therapies for a variety of allergic diseases. The application of these drugs to the allergy space was previously limited by the low kinase selectivity and subsequent toxicities of early-generation compounds. Fortunately, next-generation, highly selective BTKis in clinical development appear to have more favorable risk-benefit profiles, and their likelihood of being Food and Drug Administration-approved for an allergy indication is increasing. Recent clinical trials have indicated the remarkable and rapid efficacy of the second-generation BTKi acalabrutinib in preventing clinical reactivity to peanut ingestion in adults with peanut allergy. In addition, next-generation BTKis including remibrutinib effectively reduce disease activity in patients with antihistamine-refractory chronic spontaneous urticaria. Finally, several BTKis are currently under investigation in early clinical trials for atopic dermatitis and asthma. In this review, we summarize recent data supporting the use of these drugs as novel therapies in food allergy, anaphylaxis, urticaria, and other allergic disorders. We also discuss safety data derived from trials using both short-term and chronic dosing of BTKis.
Collapse
Affiliation(s)
- Erica V Lin
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ragha V Suresh
- Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland
| | - Melanie C Dispenza
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
6
|
De Bondt M, Renders J, Struyf S, Hellings N. Inhibitors of Bruton's tyrosine kinase as emerging therapeutic strategy in autoimmune diseases. Autoimmun Rev 2024; 23:103532. [PMID: 38521213 DOI: 10.1016/j.autrev.2024.103532] [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: 01/21/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
Bruton's tyrosine kinase (BTK) is a cytoplasmic, non-receptor signal transducer, initially identified as an essential signaling molecule for B cells, with genetic mutations resulting in a disorder characterized by disturbed B cell and antibody development. Subsequent research revealed the critical role of BTK in the functionality of monocytes, macrophages and neutrophils. Various immune cells, among which B cells and neutrophils, rely on BTK activity for diverse signaling pathways downstream of multiple receptors, which makes this kinase an ideal target to treat hematological malignancies and autoimmune diseases. First-generation BTK inhibitors are already on the market to treat hematological disorders. It has been demonstrated that B cells and myeloid cells play a significant role in the pathogenesis of different autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus and primary Sjögren's syndrome. Consequently, second-generation BTK inhibitors are currently being developed to treat these disorders. Despite the acknowledged involvement of BTK in various cell types, the focus on B cells often overshadows its impact on innate immune cells. Among these cell types, neutrophils are often underestimated in the pathogenesis of autoimmune diseases. In this narrative review, the function of BTK in different immune cell subsets is discussed, after which an overview is provided of different upcoming BTK inhibitors tested for treatment of autoimmune diseases. Special attention is paid to BTK inhibition and its effect on neutrophil biology.
Collapse
Affiliation(s)
- Mirre De Bondt
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Belgium, Herestraat 49, box 1042, 3000 Leuven; Neuro Immune Connections & Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Agoralaan building C, 3095 Diepenbeek, Belgium
| | - Janne Renders
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Belgium, Herestraat 49, box 1042, 3000 Leuven
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Belgium, Herestraat 49, box 1042, 3000 Leuven
| | - Niels Hellings
- Neuro Immune Connections & Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Agoralaan building C, 3095 Diepenbeek, Belgium.
| |
Collapse
|
7
|
Bernstein JA, Maurer M, Saini SS. BTK signaling-a crucial link in the pathophysiology of chronic spontaneous urticaria. J Allergy Clin Immunol 2024; 153:1229-1240. [PMID: 38141832 DOI: 10.1016/j.jaci.2023.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Chronic spontaneous urticaria (CSU) is an inflammatory skin disorder that manifests with itchy wheals, angioedema, or both for more than 6 weeks. Mast cells and basophils are the key pathogenic drivers of CSU; their activation results in histamine and cytokine release with subsequent dermal inflammation. Two overlapping mechanisms of mast cell and basophil activation have been proposed in CSU: type I autoimmunity, also called autoallergy, which is mediated via IgE against various autoallergens, and type IIb autoimmunity, which is mediated predominantly via IgG directed against the IgE receptor FcεRI or FcεRI-bound IgE. Both mechanisms involve cross-linking of FcεRI and activation of downstream signaling pathways, and they may co-occur in the same patient. In addition, B-cell receptor signaling has been postulated to play a key role in CSU by generating autoreactive B cells and autoantibody production. A cornerstone of FcεRI and B-cell receptor signaling is Bruton tyrosine kinase (BTK), making BTK inhibition a clear therapeutic target in CSU. The potential application of early-generation BTK inhibitors, including ibrutinib, in allergic and autoimmune diseases is limited owing to their unfavorable benefit-risk profile. However, novel BTK inhibitors with improved selectivity and safety profiles have been developed and are under clinical investigation in autoimmune diseases, including CSU. In phase 2 trials, the BTK inhibitors remibrutinib and fenebrutinib have demonstrated rapid and sustained improvements in CSU disease activity. With phase 3 studies of remibrutinib ongoing, it is hoped that BTK inhibitors will present an effective, well-tolerated option for patients with antihistamine-refractory CSU, a phenotype that presents a considerable clinical challenge.
Collapse
Affiliation(s)
- Jonathan A Bernstein
- Department of Internal Medicine, Allergy and Immunology Section, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marcus Maurer
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Sarbjit S Saini
- Johns Hopkins Asthma and Allergy Center, Division of Allergy and Clinical Immunology, The Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
8
|
Suresh RV, Dunnam C, Vaidya D, Wood RA, Bochner BS, MacGlashan DW, Dispenza MC. A phase II study of Bruton's tyrosine kinase inhibition for the prevention of anaphylaxis. J Clin Invest 2023; 133:e172335. [PMID: 37384412 PMCID: PMC10425211 DOI: 10.1172/jci172335] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/28/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUNDIgE-mediated anaphylaxis is a potentially fatal systemic allergic reaction for which there are no currently FDA-approved preventative therapies. Bruton's tyrosine kinase (BTK) is an essential enzyme for IgE-mediated signaling pathways and is an ideal pharmacologic target to prevent allergic reactions. In this open-label trial, we evaluated the safety and efficacy of acalabrutinib, a BTK inhibitor that is FDA approved to treat some B cell malignancies, in preventing clinical reactivity to peanut in adults with peanut allergy.METHODSAfter undergoing graded oral peanut challenge to establish their baseline level of clinical reactivity, 10 patients had a 6-week rest period, then received 4 standard doses of 100 mg acalabrutinib twice daily and underwent repeat food challenge. The primary endpoint was the change in patients' threshold dose of peanut protein to elicit an objective clinical reaction.RESULTSAt baseline, patients tolerated a median of 29 mg of peanut protein before objective clinical reaction. During subsequent food challenge on acalabrutinib, patients' median tolerated dose significantly increased to 4,044 mg (range 444-4,044 mg). 7 patients tolerated the maximum protocol amount (4,044 mg) of peanut protein with no clinical reaction, and the other 3 patients' peanut tolerance increased between 32- and 217-fold. 3 patients experienced a total of 4 adverse events that were considered to be possibly related to acalabrutinib; all events were transient and nonserious.CONCLUSIONAcalabrutinib pretreatment achieved clinically relevant increases in patients' tolerance to their food allergen, thereby supporting the need for larger, placebo-controlled trials.TRIAL REGISTRATIONClinicalTrials.gov NCT05038904FUNDINGAstraZeneca Pharmaceuticals, the Johns Hopkins Institute for Clinical and Translational Research, the Ludwig Family Foundation, and NIH grants AI143965 and AI106043.
Collapse
Affiliation(s)
- Ragha V. Suresh
- Division of Allergy and Clinical Immunology, Department of Medicine
| | - Collin Dunnam
- Division of Allergy and Clinical Immunology, Department of Medicine
| | - Dhananjay Vaidya
- Division of General Internal Medicine, Department of Medicine, and
| | - Robert A. Wood
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bruce S. Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | |
Collapse
|
9
|
Dispenza MC, Metcalfe DD, Olivera A. Research Advances in Mast Cell Biology and Their Translation Into Novel Therapies for Anaphylaxis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:2032-2042. [PMID: 36958519 PMCID: PMC10330051 DOI: 10.1016/j.jaip.2023.03.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/25/2023]
Abstract
Anaphylaxis is an acute, potentially life-threatening systemic allergic reaction for which there are no known reliable preventative therapies. Its primary cell mediator, the mast cell, has several pathophysiologic roles and functions in IgE-mediated reactions that continue to be poorly understood. Recent advances in the understanding of allergic mechanisms have identified novel targets for inhibiting mast cell function and activation. The prevention of anaphylaxis is within reach with new drugs that could modulate immune tolerance, mast cell proliferation and differentiation, and IgE regulation and production. Several US Food and Drug Administration-approved drugs for chronic urticaria, mastocytosis, and cancer are also being repurposed to prevent anaphylaxis. New therapeutics have not only shown promise in potential efficacy for preventing IgE-mediated reactions, but in some cases, they are able to inform us about mast cell mechanisms in vivo. This review summarizes the most recent advances in the treatment of anaphylaxis that have arisen from new pharmacologic tools and our current understanding of mast cell biology.
Collapse
Affiliation(s)
- Melanie C Dispenza
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Md.
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergy Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Ana Olivera
- Mast Cell Biology Section, Laboratory of Allergy Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| |
Collapse
|
10
|
Suresh RV, Dunnam C, Vaidya D, Wood RA, Bochner BS, MacGlashan DW, Dispenza MC. Bruton's tyrosine kinase inhibition for the prevention of anaphylaxis: an open-label, phase 2 trial. RESEARCH SQUARE 2023:rs.3.rs-2757218. [PMID: 37066249 PMCID: PMC10104202 DOI: 10.21203/rs.3.rs-2757218/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
IgE-mediated anaphylaxis is a potentially fatal systemic allergic reaction for which there are no known preventative therapies. Bruton's tyrosine kinase (BTK) is an essential enzyme for IgE-mediated signaling pathways, and is an ideal pharmacologic target to prevent allergic reactions. In this open-label trial (NCT05038904), we evaluated the safety and efficacy of acalabrutinib, a BTK inhibitor that is FDA-approved to treat some B cell malignancies, in preventing clinical reactivity to peanut in adults with IgE-mediated peanut allergy. After undergoing a graded oral peanut challenge to establish their baseline level of clinical reactivity, all patients then received four standard doses of 100 mg acalabrutinib twice daily and underwent repeat food challenge. The primary endpoint was the change in patients' threshold dose of peanut protein to elicit an objective clinical reaction. At baseline, patients tolerated a median of 29 mg of peanut protein before objective clinical reaction. During subsequent food challenge on acalabrutinib, patients' median tolerated dose significantly increased to 4,044 mg (range, 444 - 4,044 mg). 7 of 10 patients tolerated the maximum protocol amount (4,044 mg) of peanut protein with no objective clinical reaction, and the other 3 patients' peanut tolerance increased between 32- and 217-fold. Three patients experienced a total of 4 adverse events that were considered by the investigators to be possibly related to acalabrutinib; all events were transient and nonserious. These results demonstrate that acalabrutinib pretreatment can achieve clinically-relevant increases in patients' tolerance to their food allergen, thereby supporting the need for larger, placebo-controlled trials.
Collapse
Affiliation(s)
- Ragha V Suresh
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Allergy and Clinical Immunology, Baltimore, MD
| | - Collin Dunnam
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Allergy and Clinical Immunology, Baltimore, MD
| | - Dhananjay Vaidya
- Johns Hopkins University School of Medicine, Department of Medicine, Division of General Internal Medicine, Baltimore, MD
| | - Robert A Wood
- Johns Hopkins University School of Medicine, Department of Pediatrics, Division of Allergy, Immunology, and Rheumatology, Baltimore, MD
| | - Bruce S Bochner
- Northwestern University Feinberg School of Medicine, Department of Medicine, Division of Allergy and Immunology, Chicago, IL
| | - Donald W MacGlashan
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Allergy and Clinical Immunology, Baltimore, MD
| | - Melanie C Dispenza
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Allergy and Clinical Immunology, Baltimore, MD
| |
Collapse
|
11
|
Novel biologics for treatment of chronic spontaneous urticaria. J Allergy Clin Immunol 2022; 150:1256-1259. [PMID: 36180286 DOI: 10.1016/j.jaci.2022.06.027] [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: 04/19/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 12/14/2022]
Abstract
Chronic idiopathic/spontaneous urticaria (CIU/CSU) causes significant impairments in quality of life and is often unresponsive to antihistamines. Although the anti-IgE mAb omalizumab has been an important addition to the therapeutic armamentarium for the management of patients with CSU, there are still a significant percentage of patients who do not respond to the combination of antihistamines and omalizumab. As a result, additional treatments are needed. With the expanding knowledge of the pathogenesis of CSU and the role of mast cells, novel therapeutic agents targeting unique pathways important in CSU are in development. This review focuses on the rationale behind, and results of, novel therapies trialed in CSU.
Collapse
|
12
|
Burchett JR, Dailey JM, Kee SA, Pryor DT, Kotha A, Kankaria RA, Straus DB, Ryan JJ. Targeting Mast Cells in Allergic Disease: Current Therapies and Drug Repurposing. Cells 2022; 11:3031. [PMID: 36230993 PMCID: PMC9564111 DOI: 10.3390/cells11193031] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
The incidence of allergic disease has grown tremendously in the past three generations. While current treatments are effective for some, there is considerable unmet need. Mast cells are critical effectors of allergic inflammation. Their secreted mediators and the receptors for these mediators have long been the target of allergy therapy. Recent drugs have moved a step earlier in mast cell activation, blocking IgE, IL-4, and IL-13 interactions with their receptors. In this review, we summarize the latest therapies targeting mast cells as well as new drugs in clinical trials. In addition, we offer support for repurposing FDA-approved drugs to target mast cells in new ways. With a multitude of highly selective drugs available for cancer, autoimmunity, and metabolic disorders, drug repurposing offers optimism for the future of allergy therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - John J. Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
13
|
Purvis GSD, Aranda‐Tavio H, Channon KM, Greaves DR. Bruton's TK regulates myeloid cell recruitment during acute inflammation. Br J Pharmacol 2022; 179:2754-2770. [PMID: 34897650 PMCID: PMC9361009 DOI: 10.1111/bph.15778] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/27/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Bruton's TK (BTK) is a non-receptor kinase best known for its role in B lymphocyte development that is critical for proliferation and survival of leukaemic cells in B-cell malignancies. However, BTK is expressed in myeloid cells, particularly neutrophils, monocytes and macrophages where its inhibition has been reported to cause anti-inflammatory properties. EXPERIMENTAL APPROACH We explored the role of BTK on migration of myeloid cells (neutrophils, monocytes and macrophages), in vitro using chemotaxis assays and in vivo using zymosan-induced peritonitis as model systems. KEY RESULTS Using the zymosan-induced peritonitis model of sterile inflammation, we demonstrated that acute inhibition of BTK prior to zymosan challenge reduced phosphorylation of BTK in circulating neutrophils and monocytes. Moreover, pharmacological inhibition of BTK with ibrutinib specifically inhibited neutrophil and Ly6Chi monocytes, but not Ly6Clo monocyte recruitment to the peritoneum. X-linked immunodeficient (XID) mice, which have a point mutation in the Btk gene, had reduced neutrophil and monocyte recruitment to the peritoneum following zymosan challenge. Pharmacological or genetic inhibition of BTK signalling substantially reduced human monocyte and murine macrophage chemotaxis, to a range of clinically relevant chemoattractants (C5a and CCL2). We also demonstrated that inhibition of BTK in tissue resident macrophages significantly decreases chemokine secretion by reducing NF-κB activity and Akt signalling. CONCLUSION AND IMPLICATIONS Our work has identified a new role of BTK in regulating myeloid cell recruitment via two mechanisms, reducing monocyte/macrophages' ability to undergo chemotaxis and reducing chemokine secretion, via reduced NF-κB and Akt activity in tissue resident macrophages.
Collapse
Affiliation(s)
- Gareth S. D. Purvis
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- BHF Centre of Research ExcellenceUniversity of OxfordOxfordUK
| | | | - Keith M. Channon
- BHF Centre of Research ExcellenceUniversity of OxfordOxfordUK
- Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
- Department of Cardiovascular Medicine, Radcliffe Department of MedicineJohn Radcliffe HospitalOxfordUK
| | - David R. Greaves
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- BHF Centre of Research ExcellenceUniversity of OxfordOxfordUK
| |
Collapse
|
14
|
Stehlin F, Mahdi-Aljedani R, Canton L, Monzambani-Banderet V, Miauton A, Girard C, Kammermann K, Meylan S, Ribi C, Harr T, Yerly D, Muller YD. Intradermal Testing With COVID-19 mRNA Vaccines Predicts Tolerance. FRONTIERS IN ALLERGY 2022; 3:818049. [PMID: 36238929 PMCID: PMC9552867 DOI: 10.3389/falgy.2022.818049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/14/2022] [Indexed: 11/26/2022] Open
Abstract
Background The newly developed mRNA-based COVID-19 vaccines can provoke anaphylaxis, possibly induced by polyethylene glycol (PEG) contained in the vaccine. The management of persons with a history of PEG allergy or with a suspected allergic reaction after the first dose remains to be defined. Methods In this real-life study, we defined two cohorts of individuals: one pre-vaccination including 187 individuals with high-risk profiles for developing anaphylaxis and a second post-vaccination including 87 individuals with suspected allergic reactions after the COVID-19 mRNA vaccine. Upon negative skin test with an mRNA vaccine, a two-step (10–90%) vaccination protocol was performed. Positive skin tests were confirmed with the basophil activation test (BAT). Results Among 604,267 doses of vaccine, 87 suspected allergic reactions (5 after the booster) were reported to our division for further investigations: 18/87 (21%) were consistent with anaphylaxis, 78/87 (90%) were female, and 47/87 (54%) received the BNT162b2 mRNA vaccine. Vaccine skin tests were negative in 96% and 76% of the pre- and post-vaccination cohorts, respectively. A two-step vaccination was tolerated in 232/236 (98%) of individuals with negative tests. Four individuals experienced isolated asthmatic reactions during the two-step challenge. Vaccine-positive skin tests were consistently confirmed by BAT; CD63 and CD203c expression was selectively inhibited with ibrutinib, suggesting an IgE-dependent mechanism. Conclusion Sensitization to SARS-CoV-2 mRNA vaccines can be detected with intradermal testing. Significantly more individuals were sensitized to mRNA vaccines in the post-vaccination cohort. A two-step 10–90%-vaccination protocol can be safely administered upon negative skin testing.
Collapse
Affiliation(s)
- Florian Stehlin
- Division of Immunology and Allergy, University Hospital of Lausanne, Lausanne, Switzerland
| | - Rima Mahdi-Aljedani
- Division of Immunology and Allergy, University Hospital of Lausanne, Lausanne, Switzerland
| | - Loris Canton
- Division of Immunology and Allergy, University Hospital of Lausanne, Lausanne, Switzerland
| | | | - Alix Miauton
- Tropical, Travel and Vaccination Clinic, Center for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
| | - Cedric Girard
- Division of Pharmacy, University Hospital of Lausanne, Lausanne, Switzerland
| | - Kevin Kammermann
- Adverse Drug Reactions - Analysis & Consulting (ADR-AC) GmbH, Bern, Switzerland
| | - Sylvain Meylan
- Infectious Diseases Service, University Hospital Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Camillo Ribi
- Division of Immunology and Allergy, University Hospital of Lausanne, Lausanne, Switzerland
| | - Thomas Harr
- Division of Immunology and Allergy, University Hospital of Geneva, Geneva, Switzerland
| | - Daniel Yerly
- Adverse Drug Reactions - Analysis & Consulting (ADR-AC) GmbH, Bern, Switzerland
| | - Yannick D. Muller
- Division of Immunology and Allergy, University Hospital of Lausanne, Lausanne, Switzerland
- *Correspondence: Yannick D. Muller
| |
Collapse
|
15
|
Levi-Schaffer F, Gibbs BF, Hallgren J, Pucillo C, Redegeld F, Siebenhaar F, Vitte J, Mezouar S, Michel M, Puzzovio PG, Maurer M. Selected recent advances in understanding the role of human mast cells in health and disease. J Allergy Clin Immunol 2022; 149:1833-1844. [PMID: 35276243 DOI: 10.1016/j.jaci.2022.01.030] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/12/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022]
Abstract
Mast cells are highly granular tissue-resident cells and key drivers of inflammation, particularly in allergies as well as in other inflammatory diseases. Most mast cell research was initially conducted in rodents but has increasingly shifted to the human system, with the advancement of research technologies and methodologies. Today we can analyze primary human cells including rare subpopulations, we can produce and maintain mast cells isolated from human tissues, and there are several human mast cell lines. These tools have substantially facilitated our understanding of their role and function in different organs in both health and disease. We can now define more clearly where human mast cells originate from, how they develop, which mediators they store, produce de novo, and release, how they are activated and by which receptors, and which neighbouring cells they interact with and by which mechanisms. Considerable progress has also been made regarding the potential contribution of mast cells to disease, which, in turn, has led to the development of novel approaches for preventing key pathogenic effects of mast cells, heralding the era of mast cell-targeted therapeutics. In this review, we present and discuss a selection of some of the most significant advancements and remaining gaps in our understanding of human mast cells during the last 25 years, with a focus on clinical relevance.
Collapse
Affiliation(s)
- Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Bernhard F Gibbs
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Jenny Hallgren
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Carlo Pucillo
- Laboratory of Immunology, Department of Medicine, University of Udine, Udine, Italy
| | - Frank Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Frank Siebenhaar
- Institute for Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology, ITMP Allergology and Immunology, Berlin, Germany
| | - Joana Vitte
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France; IDESP, INSERM UA 11, Montpellier, France
| | | | - Moïse Michel
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France; Immunology Laboratory, CHU Nîmes, Nîmes, France
| | - Pier Giorgio Puzzovio
- Pharmacology and Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Marcus Maurer
- Institute for Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology, ITMP Allergology and Immunology, Berlin, Germany.
| |
Collapse
|
16
|
Abstract
INTRODUCTION Across the globe, chronic urticaria (CU), i.e. chronic spontaneous urticaria (CSU) and chronic inducible urticaria (CINDU), is common, long-persisting and difficult to manage. Still, at least one-fifth is not sufficiently controlled by guideline-recommended treatment with H1-antihistamines and add-on therapy with the anti-IgE monoclonal antibody omalizumab. AREAS COVERED Using PubMed, ClinicalTrials.gov, Congress websites, and websites of the manufacturers, this review explored the pipeline, namely anti-IgE-, anti-cytokine-, anti-receptor biologics, and small molecules, in clinical development for CU. EXPERT OPINION The CU pipeline is promising. While three omalizumab biosimilars are investigated, the assumed early approval of ligelizumab will expand the effective and safe anti-IgE approach observed with omalizumab. For other anti-IgEs like UB-221, the development is behind. Data are too limited so far to clearly define the role of anti-cytokine and anti-cytokine receptor biologics such as dupilumab, tezepelumab, mepolizumab, benralizumab, and CDX-0159, of which only dupilumab is actually investigated in phase 3. Among three selective oral BTK inhibitors, remibrutinib, rilzabrutinib, and fenebrutinib, the development of remibrutinib is most advanced (phase 3). As the pipeline addresses different targets, study results will give deeper insights into the pathomechanisms of CU. Hopefully, in the next future additional approved and also more targeted approaches will be available.
Collapse
Affiliation(s)
- Bettina Wedi
- Department of Dermatology and Allergy, Comprehensive Allergy Center, Hannover Medical School, Hannover, Germany
| |
Collapse
|
17
|
Mendes‐Bastos P, Brasileiro A, Kolkhir P, Frischbutter S, Scheffel J, Moñino‐Romero S, Maurer M. Bruton's tyrosine kinase inhibition-An emerging therapeutic strategy in immune-mediated dermatological conditions. Allergy 2022; 77:2355-2366. [PMID: 35175630 PMCID: PMC9545595 DOI: 10.1111/all.15261] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023]
Abstract
Bruton's tyrosine kinase (BTK), a member of the Tec kinase family, is critically involved in a range of immunological pathways. The clinical application of BTK inhibitors for B‐cell malignancies has proven successful, and there is strong rationale for the potential benefits of BTK inhibitors in some autoimmune and allergic conditions, including immune‐mediated dermatological diseases. However, the established risk‐to‐benefit profile of “first‐generation” BTK inhibitors cannot be extrapolated to these emerging, non‐oncological, indications. “Next‐generation” BTK inhibitors such as remibrutinib and fenebrutinib entered clinical development for chronic spontaneous urticaria (CSU); rilzabrutinib and tirabrutinib are being studied as potential treatments for pemphigus. Promising data from early‐phase clinical trials in CSU suggest potential for these agents to achieve strong pathway inhibition, which may translate into measurable clinical benefits, as well as other effects such as the disruption of autoantibody production. BTK inhibitors may help to overcome some of the shortcomings of monoclonal antibody treatments for immune‐mediated dermatological conditions such as CSU, pemphigus, and systemic lupus erythematosus. In addition, the use of BTK inhibitors may improve understanding of the pathophysiological roles of mast cells, basophils, and B cells in such conditions.
Collapse
Affiliation(s)
| | - Ana Brasileiro
- Department of Dermatology Hospital Santo António dos Capuchos Centro Hospitalar Universitário Lisboa Central Lisbon Portugal
- NOVA Medical School Universidade NOVA de Lisboa Lisbon Portugal
| | - Pavel Kolkhir
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology and Allergy, Charité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
- Division of Immune‐Mediated Skin Diseases I.M. Sechenov First Moscow State Medical University (Sechenov University) Moscow Russia
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology Berlin Germany
| | - Stefan Frischbutter
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology and Allergy, Charité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology Berlin Germany
| | - Jörg Scheffel
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology and Allergy, Charité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology Berlin Germany
| | - Sherezade Moñino‐Romero
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology and Allergy, Charité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology Berlin Germany
| | - Marcus Maurer
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology and Allergy, Charité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology Berlin Germany
| |
Collapse
|
18
|
Optimizing drug inhibition of IgE-mediated anaphylaxis in mice. J Allergy Clin Immunol 2022; 149:671-684.e9. [PMID: 34186142 PMCID: PMC9187951 DOI: 10.1016/j.jaci.2021.06.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 05/19/2021] [Accepted: 06/17/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Administering allergens in increasing doses can temporarily suppress IgE-mediated allergy and anaphylaxis by desensitizing mast cells and basophils; however, allergen administration during desensitization therapy can itself induce allergic responses. Several small molecule drugs and nutraceuticals have been used clinically and experimentally to suppress these allergic responses. OBJECTIVES This study sought to optimize drug inhibition of IgE-mediated anaphylaxis. METHODS Several agents were tested individually and in combination for ability to suppress IgE-mediated anaphylaxis in conventional mice, FcεRIα-humanized mice, and reconstituted immunodeficient mice that have human mast cells and basophils. Hypothermia was the readout for anaphylaxis; therapeutic efficacy was measured by degree of inhibition of hypothermia. Serum mouse mast cell protease 1 level was used to measure extent of mast cell degranulation. RESULTS Histamine receptor 1 (HR1) antagonists, β-adrenergic agonists, and a spleen tyrosine kinase (Syk) inhibitor were best at individually inhibiting IgE-mediated anaphylaxis. A Bruton's tyrosine kinase (BTK) inhibitor, administered alone, only inhibited hypothermia when FcεRI signaling was suboptimal. Combinations of these agents could completely or nearly completely inhibit IgE-mediated hypothermia in these models. Both Syk and BTK inhibition decreased mast cell degranulation, but only Syk inhibition also blocked desensitization. Many other agents that are used clinically and experimentally had little or no beneficial effect. CONCLUSIONS Combinations of an HR1 antagonist, a β-adrenergic agonist, and a Syk or a BTK inhibitor protect best against IgE-mediated anaphylaxis, while an HR1 antagonist plus a β-adrenergic agonist ± a BTK antagonist is optimal for inhibiting IgE-mediated anaphylaxis without suppressing desensitization.
Collapse
|
19
|
Zhu S, Jung J, Victor E, Arceo J, Gokhale S, Xie P. Clinical Trials of the BTK Inhibitors Ibrutinib and Acalabrutinib in Human Diseases Beyond B Cell Malignancies. Front Oncol 2021; 11:737943. [PMID: 34778053 PMCID: PMC8585514 DOI: 10.3389/fonc.2021.737943] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/11/2021] [Indexed: 12/13/2022] Open
Abstract
The BTK inhibitors ibrutinib and acalabrutinib are FDA-approved drugs for the treatment of B cell malignances. Both drugs have demonstrated clinical efficacy and safety profiles superior to chemoimmunotherapy regimens in patients with chronic lymphocytic leukemia. Mounting preclinical and clinical evidence indicates that both ibrutinib and acalabrutinib are versatile and have direct effects on many immune cell subsets as well as other cell types beyond B cells. The versatility and immunomodulatory effects of both drugs have been exploited to expand their therapeutic potential in a wide variety of human diseases. Over 470 clinical trials are currently registered at ClinicalTrials.gov to test the efficacy of ibrutinib or acalabrutinib not only in almost every type of B cell malignancies, but also in hematological malignancies of myeloid cells and T cells, solid tumors, chronic graft versus host disease (cGHVD), autoimmune diseases, allergy and COVID-19 (http:www.clinicaltrials.gov). In this review, we present brief discussions of the clinical trials and relevant key preclinical evidence of ibrutinib and acalabrutinib as monotherapies or as part of combination therapies for the treatment of human diseases beyond B cell malignancies. Adding to the proven efficacy of ibrutinib for cGVHD, preliminary results of clinical trials have shown promising efficacy of ibrutinib or acalabrutinib for certain T cell malignancies, allergies and severe COVID-19. However, both BTK inhibitors have no or limited efficacy for refractory or recurrent solid tumors. These clinical data together with additional pending results from ongoing trials will provide valuable information to guide the design and improvement of future trials, including optimization of combination regimens and dosing sequences as well as better patient stratification and more efficient delivery strategies. Such information will further advance the precise implementation of BTK inhibitors into the clinical toolbox for the treatment of different human diseases.
Collapse
Affiliation(s)
- Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Jaeyong Jung
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Eton Victor
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Johann Arceo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
20
|
Zhu S, Gokhale S, Jung J, Spirollari E, Tsai J, Arceo J, Wu BW, Victor E, Xie P. Multifaceted Immunomodulatory Effects of the BTK Inhibitors Ibrutinib and Acalabrutinib on Different Immune Cell Subsets - Beyond B Lymphocytes. Front Cell Dev Biol 2021; 9:727531. [PMID: 34485307 PMCID: PMC8414982 DOI: 10.3389/fcell.2021.727531] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022] Open
Abstract
The clinical success of the two BTK inhibitors, ibrutinib and acalabrutinib, represents a major breakthrough in the treatment of chronic lymphocytic leukemia (CLL) and has also revolutionized the treatment options for other B cell malignancies. Increasing evidence indicates that in addition to their direct effects on B lymphocytes, both BTK inhibitors also directly impact the homeostasis, phenotype and function of many other cell subsets of the immune system, which contribute to their high efficacy as well as adverse effects observed in CLL patients. In this review, we attempt to provide an overview on the overlapping and differential effects of ibrutinib and acalabrutinib on specific receptor signaling pathways in different immune cell subsets other than B cells, including T cells, NK cells, monocytes, macrophages, granulocytes, myeloid-derived suppressor cells, dendritic cells, osteoclasts, mast cells and platelets. The shared and distinct effects of ibrutinib versus acalabrutinib are mediated through BTK-dependent and BTK-independent mechanisms, respectively. Such immunomodulatory effects of the two drugs have fueled myriad explorations of their repurposing opportunities for the treatment of a wide variety of other human diseases involving immune dysregulation.
Collapse
Affiliation(s)
- Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Jaeyong Jung
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Eris Spirollari
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Jemmie Tsai
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Johann Arceo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Ben Wang Wu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Eton Victor
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
21
|
Kaul M, End P, Cabanski M, Schuhler C, Jakab A, Kistowska M, Kinhikar A, Maiolica A, Sinn A, Fuhr R, Cenni B. Remibrutinib (LOU064): A selective potent oral BTK inhibitor with promising clinical safety and pharmacodynamics in a randomized phase I trial. Clin Transl Sci 2021; 14:1756-1768. [PMID: 33834628 PMCID: PMC8504815 DOI: 10.1111/cts.13005] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
Safe and effective new oral therapies for autoimmune, allergic, and inflammatory conditions remain a significant therapeutic need. Here, we investigate the human pharmacokinetics, pharmacodynamics (PDs), and safety of the selective, covalent Bruton's tyrosine kinase (BTK) inhibitor, remibrutinib. Study objectives were explored in randomized single and multiple ascending dose (SAD and MAD, respectively) cohorts with daily doses up to 600 mg, and a crossover food effect (FE) cohort, in adult healthy subjects without (SAD [n =80]/FE [n =12]) or with asymptomatic atopic diathesis (MAD [n =64]). A single oral dose of remibrutinib (0.5-600 mg) was rapidly absorbed (time to maximum concentration = 0.5 h-1.25 h) with an apparent blood clearance of 280-560 L/h and apparent volume of distribution of 400-15,000 L. With multiple doses (q.d. and b.i.d.), no pronounced accumulation of remibrutinib was detected (mean residence time was <3 h). Food intake showed no clinically relevant effect on remibrutinib exposure suggesting no need for dose adaptation. With remibrutinib doses greater than or equal to 30 mg, blood BTK occupancy was greater than 95% for at least 24 h (SAD). With MAD, remibrutinib reached near complete blood BTK occupancy at day 12 predose with greater than or equal to 10 mg q.d. Near complete basophil or skin prick test (SPT) inhibition at day 12 predose was achieved at greater than or equal to 50 mg q.d. for CD63 and at greater than or equal to 100 mg q.d. for SPT. Remibrutinib was well-tolerated at all doses without any dose-limiting toxicity. Remibrutinib showed encouraging blood and skin PDs with a favorable safety profile, supporting further development for diseases driven by mast cells, basophils, and B-cells, such as chronic spontaneous urticaria, allergic asthma, or Sjögren's syndrome.
Collapse
Affiliation(s)
- Martin Kaul
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| | - Peter End
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| | | | | | | | | | - Arvind Kinhikar
- Novartis Institutes for Biomedical ResearchCambridgeMassachusettsUSA
| | | | - Angela Sinn
- Early Phase Clinical UnitParexel InternationalBerlinGermany
| | - Rainard Fuhr
- Early Phase Clinical UnitParexel InternationalBerlinGermany
| | - Bruno Cenni
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| |
Collapse
|
22
|
Recent Advances in BTK Inhibitors for the Treatment of Inflammatory and Autoimmune Diseases. Molecules 2021; 26:molecules26164907. [PMID: 34443496 PMCID: PMC8399599 DOI: 10.3390/molecules26164907] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 11/17/2022] Open
Abstract
Bruton’s tyrosine kinase (BTK) plays a crucial role in B-cell receptor and Fc receptor signaling pathways. BTK is also involved in the regulation of Toll-like receptors and chemokine receptors. Given the central role of BTK in immunity, BTK inhibition represents a promising therapeutic approach for the treatment of inflammatory and autoimmune diseases. Great efforts have been made in developing BTK inhibitors for potential clinical applications in inflammatory and autoimmune diseases. This review covers the recent development of BTK inhibitors at preclinical and clinical stages in treating these diseases. Individual examples of three types of inhibitors, namely covalent irreversible inhibitors, covalent reversible inhibitors, and non-covalent reversible inhibitors, are discussed with a focus on their structure, bioactivity and selectivity. Contrary to expectations, reversible BTK inhibitors have not yielded a significant breakthrough so far. The development of covalent, irreversible BTK inhibitors has progressed more rapidly. Many candidates entered different stages of clinical trials; tolebrutinib and evobrutinib are undergoing phase 3 clinical evaluation. Rilzabrutinib, a covalent reversible BTK inhibitor, is now in phase 3 clinical trials and also offers a promising future. An analysis of the protein–inhibitor interactions based on published co-crystal structures provides useful clues for the rational design of safe and effective small-molecule BTK inhibitors.
Collapse
|
23
|
Abstract
Chronic spontaneous urticaria (CSU) is characterized by the presence of wheals, angioedema, or both for at least 6 weeks. It may persist for a long time-up to 50% of the patients have been reported to be symptomatic 5 years after the onset. Some patients can suffer more than one episode of CSU during their lifetime. Considering the recurrences, disabling symptoms, and significant impact on quality of life, proper and effective treatment of CSU is critical. The use of antihistamines (AHs) is still the mainstay of treatment. However, given the low rates of response to AHs (38.6% and 63.2% to standard doses and higher doses, respectively), the complete control of symptoms seems difficult to attain. The use of omalizumab for CSU has been a major breakthrough in the care of patients with CSU. However, the partial response and lack of response to omalizumab in a subgroup of patients, as high as 70% in some studies, make the development of alternative treatments desirable. Ever-increasing knowledge on the pathogenesis is making new target molecules available and enabling drug development for CSU. In addition to drug repurposing as in anti-IL-4/13, IL-5, and IL-17 antibodies, novel targeted therapy options such as ligelizumab and Bruton's tyrosine kinase inhibitors are currently undergoing clinical trials and will be available in the near future. This article reviews the current challenges in the treatment of CSU, the pathogenesis and potential target molecules, and the rationale for novel treatments and their rapidly developing status.
Collapse
|
24
|
Neys SFH, Hendriks RW, Corneth OBJ. Targeting Bruton's Tyrosine Kinase in Inflammatory and Autoimmune Pathologies. Front Cell Dev Biol 2021; 9:668131. [PMID: 34150760 PMCID: PMC8213343 DOI: 10.3389/fcell.2021.668131] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
Bruton's tyrosine kinase (BTK) was discovered due to its importance in B cell development, and it has a critical role in signal transduction downstream of the B cell receptor (BCR). Targeting of BTK with small molecule inhibitors has proven to be efficacious in several B cell malignancies. Interestingly, recent studies reveal increased BTK protein expression in circulating resting B cells of patients with systemic autoimmune disease (AID) compared with healthy controls. Moreover, BTK phosphorylation following BCR stimulation in vitro was enhanced. In addition to its role in BCR signaling, BTK is involved in many other pathways, including pattern recognition, Fc, and chemokine receptor signaling in B cells and myeloid cells. This broad involvement in several immunological pathways provides a rationale for the targeting of BTK in the context of inflammatory and systemic AID. Accordingly, numerous in vitro and in vivo preclinical studies support the potential of BTK targeting in these conditions. Efficacy of BTK inhibitors in various inflammatory and AID has been demonstrated or is currently evaluated in clinical trials. In addition, very recent reports suggest that BTK inhibition may be effective as immunosuppressive therapy to diminish pulmonary hyperinflammation in coronavirus disease 2019 (COVID-19). Here, we review BTK's function in key signaling pathways in B cells and myeloid cells. Further, we discuss recent advances in targeting BTK in inflammatory and autoimmune pathologies.
Collapse
|
25
|
Miyake K, Shibata S, Yoshikawa S, Karasuyama H. Basophils and their effector molecules in allergic disorders. Allergy 2021; 76:1693-1706. [PMID: 33205439 DOI: 10.1111/all.14662] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/26/2020] [Accepted: 11/12/2020] [Indexed: 12/20/2022]
Abstract
Basophils are the rarest granulocytes which represent <1% of peripheral blood leukocytes. Basophils bear several phenotypic similarities to tissue-resident mast cells and therefore had been erroneously considered as blood-circulating mast cells. However, recent researches have revealed that basophils play nonredundant roles in allergic inflammation, protective immunity against parasitic infections and regulation of innate and acquired immunity. Basophils are recruited to inflamed tissues and activated in an IgE-dependent or IgE-independent manner to release a variety of effector molecules. Such molecules, including IL-4, act on various types of cells and play versatile roles, including the induction and termination of allergic inflammation and the regulation of immune responses. Recent development of novel therapeutic agents has enabled us to gain further insights into basophil biology in human disorders. In this review, we highlight the recent advances in the field of basophil biology with a particular focus on the role of basophils in allergic inflammation. Further studies on basophils and their effector molecules will help us identify novel therapeutic targets for treating allergic disorders.
Collapse
Affiliation(s)
- Kensuke Miyake
- Inflammation, Infection and Immunity Laboratory TMDU Advanced Research Institute Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Sho Shibata
- Department of Respiratory Medicine Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Soichiro Yoshikawa
- Department of Cell Physiology Graduate School of Medicine, Dentistry and Pharmaceutical Sciences Okayama University Okayama Japan
| | - Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory TMDU Advanced Research Institute Tokyo Medical and Dental University (TMDU) Tokyo Japan
| |
Collapse
|
26
|
Rajani HF, Shahidi S, Gomari MM. Protein and Antibody Engineering: Suppressing Degranulation of the Mast Cells and Type I Hypersensitivity Reaction. Curr Protein Pept Sci 2021; 21:831-841. [PMID: 32392111 DOI: 10.2174/1389203721666200511094717] [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: 01/10/2020] [Revised: 03/09/2020] [Accepted: 04/23/2020] [Indexed: 11/22/2022]
Abstract
With an increase in atopic cases and owing to a significant role of mast cells in type I hypersensitivity, a therapeutic need to inhibit degranulation of mast cells has risen. Mast cells are notorious for IgE-mediated allergic response. Advancements have allowed researchers to improve clinical outcomes of already available therapies. Engineered peptides and antibodies can be easily manipulated to attain desired characteristics as per the biological environment. A number of these molecules are designed to target mast cells in order to regulate the release of histamine and other mediators, thereby controlling type I hypersensitivity response. The aim of this review paper is to highlight some of the significant molecules designed for the purpose.
Collapse
Affiliation(s)
- Huda Fatima Rajani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical
Sciences, Tehran, Iran
| | - Solmaz Shahidi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahmoudi Gomari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical
Sciences, Tehran, Iran
| |
Collapse
|
27
|
Dispenza MC. The Use of Bruton's Tyrosine Kinase Inhibitors to Treat Allergic Disorders. CURRENT TREATMENT OPTIONS IN ALLERGY 2021; 8:261-273. [PMID: 33880321 PMCID: PMC8050815 DOI: 10.1007/s40521-021-00286-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/20/2021] [Indexed: 12/19/2022]
Abstract
Purpose of review Studies show that inhibitors of Bruton’s tyrosine kinase (BTKis), currently FDA-approved for the treatment of B cell malignancies, can prevent IgE-mediated reactions through broad inhibition of the FcεRI signaling pathway in human mast cells and basophils. This review will summarize recent data supporting the use of these drugs as novel therapies in various allergic disorders. Recent findings Recent studies have shown that BTKis can prevent IgE-mediated degranulation and cytokine production in primary human mast cells and basophils. Two oral doses of the second-generation BTKi acalabrutinib can completely prevent moderate passive systemic anaphylaxis in humanized mice and even protect against death during severe anaphylaxis. Furthermore, two doses of ibrutinib can reduce or eliminate skin prick test responses to foods and aeroallergens in allergic subjects. BTKis in development also show efficacy in clinical trials for chronic urticaria. Unlike other therapies targeting IgE, such as omalizumab, BTKis appear to have rapid onset and transient effects, making them ideal candidates for intermittent use to prevent acute reactions such as IgE-mediated anaphylaxis. Summary These studies suggest that BTKis may be capable of preventing IgE-mediated anaphylaxis, paving the way for future trials in food allergy and urticaria.
Collapse
Affiliation(s)
- Melanie C Dispenza
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
28
|
Albuhairi S, Rachid R. Biologics and Novel Therapies for Food Allergy. Immunol Allergy Clin North Am 2021; 41:271-283. [PMID: 33863483 DOI: 10.1016/j.iac.2021.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Food allergy is a significant public health burden affecting around 10% of adults and 8% of children. Although the first peanut oral immunotherapy product received Food and Drug Administration approval in 2020, there is still an unmet need for more effective therapeutic options that minimize the risk of anaphylaxis, nutritional deficiencies, and patient's quality of life. Biologics are promising modalities, as they may improve compliance, target multiple food allergies, and treat other concomitant atopic diseases. Although omalizumab has been evaluated extensively, most biologics are more novel and have broader immunologic impact. Careful evaluation of their safety profile should therefore be conducted.
Collapse
Affiliation(s)
- Sultan Albuhairi
- Department of Pediatrics, Allergy and Immunology Section, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Rima Rachid
- Division of Immunology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Dispenza MC, Krier-Burris RA, Chhiba KD, Undem BJ, Robida PA, Bochner BS. Bruton's tyrosine kinase inhibition effectively protects against human IgE-mediated anaphylaxis. J Clin Invest 2021; 130:4759-4770. [PMID: 32484802 DOI: 10.1172/jci138448] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/28/2020] [Indexed: 01/30/2023] Open
Abstract
No known therapies can prevent anaphylaxis. Bruton's tyrosine kinase (BTK) is an enzyme thought to be essential for high-affinity IgE receptor (FcεRI) signaling in human cells. We tested the hypothesis that FDA-approved BTK inhibitors (BTKis) would prevent IgE-mediated responses including anaphylaxis. We showed that irreversible BTKis broadly prevented IgE-mediated degranulation and cytokine production in primary human mast cells and blocked allergen-induced contraction of isolated human bronchi. To address their efficacy in vivo, we created and used what we believe to be a novel humanized mouse model of anaphylaxis that does not require marrow ablation or human tissue implantation. After a single intravenous injection of human CD34+ cells, NSG-SGM3 mice supported the population of mature human tissue-resident mast cells and basophils. These mice showed excellent responses during passive systemic anaphylaxis using human IgE to selectively evoke human mast cell and basophil activation, and response severity was controllable by alteration of the amount of allergen used for challenge. Remarkably, pretreatment with just 2 oral doses of the BTKi acalabrutinib completely prevented moderate IgE-mediated anaphylaxis in these mice and also significantly protected against death during severe anaphylaxis. Our data suggest that BTKis may be able to prevent anaphylaxis in humans by inhibiting FcεRI-mediated signaling.
Collapse
Affiliation(s)
- Melanie C Dispenza
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rebecca A Krier-Burris
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Krishan D Chhiba
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bradley J Undem
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Piper A Robida
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bruce S Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
30
|
Dispenza MC, Bochner BS, MacGlashan DW. Targeting the FcεRI Pathway as a Potential Strategy to Prevent Food-Induced Anaphylaxis. Front Immunol 2021; 11:614402. [PMID: 33391286 PMCID: PMC7773654 DOI: 10.3389/fimmu.2020.614402] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/16/2020] [Indexed: 12/25/2022] Open
Abstract
Despite attempts to halt it, the prevalence of food allergy is increasing, and there is an unmet need for strategies to prevent morbidity and mortality from food-induced allergic reactions. There are no known medications that can prevent anaphylaxis, but several novel therapies show promise for the prevention of food-induced anaphylaxis through targeting of the high-affinity IgE receptor (FcϵRI) pathway. This pathway includes multiple candidate targets, including tyrosine kinases and the receptor itself. Small molecule inhibitors of essential kinases have rapid onset of action and transient efficacy, which may be beneficial for short-term use for immunotherapy buildup or desensitizations. Short courses of FDA-approved inhibitors of Bruton’s tyrosine kinase can eliminate IgE-mediated basophil activation and reduce food skin test size in allergic adults, and prevent IgE-mediated anaphylaxis in humanized mice. In contrast, biologics may provide longer-lasting protection, albeit with slower onset. Omalizumab is an anti-IgE antibody that sequesters IgE, thereby reducing FcϵRI expression on mast cells and basophils. As a monotherapy, it can increase the clinical threshold dose of food allergen, and when used as an adjunct for food immunotherapy, it decreases severe reactions during buildup phase. Finally, lirentelimab, an anti-Siglec-8 antibody currently in clinical trials, can prevent IgE-mediated anaphylaxis in mice through mast cell inhibition. This review discusses these and other emerging therapies as potential strategies for preventing food-induced anaphylaxis. In contrast to other food allergy treatments which largely focus on individual allergens, blockade of the FcϵRI pathway has the advantage of preventing clinical reactivity from any food.
Collapse
Affiliation(s)
- Melanie C Dispenza
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Bruce S Bochner
- Department of Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Donald W MacGlashan
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
31
|
Abstract
Despite recent advances in the treatment of autoimmune and inflammatory diseases, unmet medical needs in some areas still exist. One of the main therapeutic approaches to alleviate dysregulated inflammation has been to target the activity of kinases that regulate production of inflammatory mediators. Small-molecule kinase inhibitors have the potential for broad efficacy, convenience and tissue penetrance, and thus often offer important advantages over biologics. However, designing kinase inhibitors with target selectivity and minimal off-target effects can be challenging. Nevertheless, immense progress has been made in advancing kinase inhibitors with desirable drug-like properties into the clinic, including inhibitors of JAKs, IRAK4, RIPKs, BTK, SYK and TPL2. This Review will address the latest discoveries around kinase inhibitors with an emphasis on clinically validated autoimmunity and inflammatory pathways.
Collapse
Affiliation(s)
- Ali A Zarrin
- Discovery Department, TRex Bio, South San Francisco, CA, USA.
| | - Katherine Bao
- Early Discovery Biochemistry Department, Genentech, South San Francisco, CA, USA
| | | | - Domagoj Vucic
- Early Discovery Biochemistry Department, Genentech, South San Francisco, CA, USA
| |
Collapse
|
32
|
Litzenburger T, Steffgen J, Benediktus E, Müller F, Schultz A, Klein E, Ramanujam M, Harcken C, Gupta A, Wu J, Wiebe S, Li X, Flack M, Padula SJ, Visvanathan S, Hünnemeyer A, Hui J. Safety, pharmacokinetics and pharmacodynamics of BI 705564, a highly selective, covalent inhibitor of Bruton's tyrosine kinase, in Phase I clinical trials in healthy volunteers. Br J Clin Pharmacol 2020; 87:1824-1838. [PMID: 32986868 PMCID: PMC9290462 DOI: 10.1111/bcp.14571] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022] Open
Abstract
Aims To evaluate the safety, pharmacokinetics and pharmacodynamics of single‐ and multiple‐rising doses (MRDs) of BI 705564 and establish proof of mechanism. Methods BI 705564 was studied in 2 placebo‐controlled, Phase I clinical trials testing single‐rising doses (1–160 mg) and MRDs (1–80 mg) of BI 705564 over 14 days in healthy male volunteers. Blood samples were analysed for BI 705564 plasma concentration, Bruton's tyrosine kinase (BTK) target occupancy (TO) and CD69 expression in B cells stimulated ex vivo. A substudy was conducted in allergic, otherwise healthy, MRD participants. Safety was assessed in both studies. Results All doses of BI 705564 were well tolerated. Geometric mean BI 705564 plasma terminal half‐life ranged from 10.1 to 16.9 hours across tested doses, with no relevant accumulation after multiple dosing. Doses ≥20 mg resulted in ≥85% average TO that was maintained for ≥48 hours after single‐dose administration. Functional effects of BTK signalling were demonstrated by dose‐dependent inhibition of CD69 expression. In allergic participants, BI 705564 treatment showed a trend in wheal size reduction in a skin prick test and complete inhibition of basophil activation. Mild bleeding‐related adverse events were observed with BI 705564; bleeding time increased in 1/12 participants (8.3%) who received placebo vs 26/48 (54.2%) treated with BI 705564. Conclusion BI 705564 showed efficient target engagement through durable TO and inhibition of ex vivo B‐cell activation, and proof of mechanism through effects on allergic skin responses. Mild bleeding‐related adverse events were probably related to inhibition of platelet aggregation by BTK inhibition.
Collapse
Affiliation(s)
| | | | | | - Fabian Müller
- Boehringer Ingelheim Pharma GmbH & Co. KG Biberach Germany
| | - Armin Schultz
- CRS Clinical Research Services Mannheim GmbH Mannheim Germany
| | - Elliott Klein
- Boehringer Ingelheim Pharmaceuticals Ridgefield CT USA
| | | | | | - Alpana Gupta
- Boehringer Ingelheim Pharmaceuticals Ridgefield CT USA
| | - Jing Wu
- Boehringer Ingelheim Pharmaceuticals Ridgefield CT USA
| | - Sabrina Wiebe
- Boehringer Ingelheim Pharma GmbH & Co. KG Biberach Germany
| | - Xiujiang Li
- Boehringer Ingelheim Pharmaceuticals Ridgefield CT USA
| | - Mary Flack
- Boehringer Ingelheim Pharmaceuticals Ridgefield CT USA
| | | | | | | | - Jianan Hui
- Boehringer Ingelheim Pharmaceuticals Ridgefield CT USA
| |
Collapse
|
33
|
Valent P, Akin C, Hartmann K, Nilsson G, Reiter A, Hermine O, Sotlar K, Sperr WR, Escribano L, George TI, Kluin-Nelemans HC, Ustun C, Triggiani M, Brockow K, Gotlib J, Orfao A, Kovanen PT, Hadzijusufovic E, Sadovnik I, Horny HP, Arock M, Schwartz LB, Austen KF, Metcalfe DD, Galli SJ. Mast cells as a unique hematopoietic lineage and cell system: From Paul Ehrlich's visions to precision medicine concepts. Am J Cancer Res 2020; 10:10743-10768. [PMID: 32929378 PMCID: PMC7482799 DOI: 10.7150/thno.46719] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
The origin and functions of mast cells (MCs) have been debated since their description by Paul Ehrlich in 1879. MCs have long been considered 'reactive bystanders' and 'amplifiers' in inflammatory processes, allergic reactions, and host responses to infectious diseases. However, knowledge about the origin, phenotypes and functions of MCs has increased substantially over the past 50 years. MCs are now known to be derived from multipotent hematopoietic progenitors, which, through a process of differentiation and maturation, form a unique hematopoietic lineage residing in multiple organs. In particular, MCs are distinguishable from basophils and other hematopoietic cells by their unique phenotype, origin(s), and spectrum of functions, both in innate and adaptive immune responses and in other settings. The concept of a unique MC lineage is further supported by the development of a distinct group of neoplasms, collectively referred to as mastocytosis, in which MC precursors expand as clonal cells. The clinical consequences of the expansion and/or activation of MCs are best established in mastocytosis and in allergic inflammation. However, MCs have also been implicated as important participants in a number of additional pathologic conditions and physiological processes. In this article, we review concepts regarding MC development, factors controlling MC expansion and activation, and some of the fundamental roles MCs may play in both health and disease. We also discuss new concepts for suppressing MC expansion and/or activation using molecularly-targeted drugs.
Collapse
|
34
|
Ma B, Bohnert T, Otipoby KL, Tien E, Arefayene M, Bai J, Bajrami B, Bame E, Chan TR, Humora M, MacPhee JM, Marcotte D, Mehta D, Metrick CM, Moniz G, Polack E, Poreci U, Prefontaine A, Sheikh S, Schroeder P, Smirnakis K, Zhang L, Zheng F, Hopkins BT. Discovery of BIIB068: A Selective, Potent, Reversible Bruton's Tyrosine Kinase Inhibitor as an Orally Efficacious Agent for Autoimmune Diseases. J Med Chem 2020; 63:12526-12541. [PMID: 32696648 DOI: 10.1021/acs.jmedchem.0c00702] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Autoreactive B cell-derived antibodies form immune complexes that likely play a pathogenic role in autoimmune diseases. In systemic lupus erythematosus (SLE), these antibodies bind Fc receptors on myeloid cells and induce proinflammatory cytokine production by monocytes and NETosis by neutrophils. Bruton's tyrosine kinase (BTK) is a non-receptor tyrosine kinase that signals downstream of Fc receptors and plays a transduction role in antibody expression following B cell activation. Given the roles of BTK in both the production and sensing of autoreactive antibodies, inhibitors of BTK kinase activity may provide therapeutic value to patients suffering from autoantibody-driven immune disorders. Starting from an in-house proprietary screening hit followed by structure-based rational design, we have identified a potent, reversible BTK inhibitor, BIIB068 (1), which demonstrated good kinome selectivity with good overall drug-like properties for oral dosing, was well tolerated across preclinical species at pharmacologically relevant doses with good ADME properties, and achieved >90% inhibition of BTK phosphorylation (pBTK) in humans.
Collapse
Affiliation(s)
- Bin Ma
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Tonika Bohnert
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Kevin L Otipoby
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Eric Tien
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Million Arefayene
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Judy Bai
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Bekim Bajrami
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Eris Bame
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Timothy R Chan
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Michael Humora
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - J Michael MacPhee
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Douglas Marcotte
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Devangi Mehta
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Claire M Metrick
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - George Moniz
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Evelyne Polack
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Urjana Poreci
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Annick Prefontaine
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Sarah Sheikh
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Patricia Schroeder
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Karen Smirnakis
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Lei Zhang
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Fengmei Zheng
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Brian T Hopkins
- Research & Development, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
35
|
Jörg L, Mueller-Wirth N, Pecaric-Petkovic T, Diaz C, Pichler W, Hausmann O. The Fcε receptor I pathway is crucial but not exclusive for basophil activation in patients with autoimmune forms of chronic spontaneous urticaria. J Eur Acad Dermatol Venereol 2020; 34:e825-e827. [PMID: 32491222 DOI: 10.1111/jdv.16703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- L Jörg
- Department of Rheumatology, Immunology and Allergology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - N Mueller-Wirth
- ADR-AC GmbH, Adverse Drug Reactions, Analysis and Consulting, Bern, Switzerland
| | - T Pecaric-Petkovic
- ADR-AC GmbH, Adverse Drug Reactions, Analysis and Consulting, Bern, Switzerland
| | - C Diaz
- ADR-AC GmbH, Adverse Drug Reactions, Analysis and Consulting, Bern, Switzerland
| | - W Pichler
- ADR-AC GmbH, Adverse Drug Reactions, Analysis and Consulting, Bern, Switzerland
| | - O Hausmann
- ADR-AC GmbH, Adverse Drug Reactions, Analysis and Consulting, Bern, Switzerland.,Löwenpraxis Luzern, Lucerne, Switzerland
| |
Collapse
|
36
|
Corzo CA, Varfolomeev E, Setiadi AF, Francis R, Klabunde S, Senger K, Sujatha-Bhaskar S, Drobnick J, Do S, Suto E, Huang Z, Eastham-Anderson J, Katewa A, Pang J, Domeyer M, Dela Cruz C, Paler-Martinez A, Lau VWC, Hadadianpour A, Ramirez-Carrozi V, Sun Y, Bao K, Xu D, Hunley E, Brightbill HD, Warming S, Roose-Girma M, Wong A, Tam L, Emson CL, Crawford JJ, Young WB, Pappu R, McKenzie BS, Asghari V, Vucic D, Hackney JA, Austin CD, Lee WP, Lekkerkerker A, Ghilardi N, Bryan MC, Kiefer JR, Townsend MJ, Zarrin AA. The kinase IRAK4 promotes endosomal TLR and immune complex signaling in B cells and plasmacytoid dendritic cells. Sci Signal 2020; 13:13/634/eaaz1053. [PMID: 32487715 DOI: 10.1126/scisignal.aaz1053] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The dysregulation of multiple signaling pathways, including those through endosomal Toll-like receptors (TLRs), Fc gamma receptors (FcγR), and antigen receptors in B cells (BCR), promote an autoinflammatory loop in systemic lupus erythematosus (SLE). Here, we used selective small-molecule inhibitors to assess the regulatory roles of interleukin-1 receptor (IL-1R)-associated kinase 4 (IRAK4) and Bruton's tyrosine kinase (BTK) in these pathways. The inhibition of IRAK4 repressed SLE immune complex- and TLR7-mediated activation of human plasmacytoid dendritic cells (pDCs). Correspondingly, the expression of interferon (IFN)-responsive genes (IRGs) in cells and in mice was positively regulated by the kinase activity of IRAK4. Both IRAK4 and BTK inhibition reduced the TLR7-mediated differentiation of human memory B cells into plasmablasts. TLR7-dependent inflammatory responses were differentially regulated by IRAK4 and BTK by cell type: In pDCs, IRAK4 positively regulated NF-κB and MAPK signaling, whereas in B cells, NF-κB and MAPK pathways were regulated by both BTK and IRAK4. In the pristane-induced lupus mouse model, inhibition of IRAK4 reduced the expression of IRGs during disease onset. Mice engineered to express kinase-deficient IRAK4 were protected from both chemical (pristane-induced) and genetic (NZB/W_F1 hybrid) models of lupus development. Our findings suggest that kinase inhibitors of IRAK4 might be a therapeutic in patients with SLE.
Collapse
Affiliation(s)
- Cesar A Corzo
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | - Ross Francis
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Sha Klabunde
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kate Senger
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Joy Drobnick
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Steven Do
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Eric Suto
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zhiyu Huang
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Arna Katewa
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jodie Pang
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Melanie Domeyer
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | - Vivian W C Lau
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | - Yonglian Sun
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Katherine Bao
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Daqi Xu
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Emily Hunley
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Soren Warming
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Alfred Wong
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Lucinda Tam
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Claire L Emson
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - James J Crawford
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Wendy B Young
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Rajita Pappu
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Brent S McKenzie
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Vida Asghari
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Domagoj Vucic
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jason A Hackney
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Cary D Austin
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Wyne P Lee
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Nico Ghilardi
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Marian C Bryan
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - James R Kiefer
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Ali A Zarrin
- Research, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
37
|
Munir MA, Badri KH. The Importance of Derivatizing Reagent in Chromatography Applications for Biogenic Amine Detection in Food and Beverages. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2020; 2020:5814389. [PMID: 32377440 PMCID: PMC7199571 DOI: 10.1155/2020/5814389] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 05/19/2023]
Abstract
Biogenic amines (BA) are chemical compounds formed in foods that contain protein, allowing the foods to undergo a bacterial degradation process. Biogenic amines are labeled as toxic food because its consumption exceeding the FDA regulation (50 mg/kg) can be harmful to humans. Some countries also have regulations that prohibit the consumption of biogenic amines in high concentrations, especially histamine. The chromatography methods generally applied by researchers are liquid chromatography (LC) and gas chromatography (GC), where the use of a derivatization reagent is necessary to increase their sensitivity. This review is based on past and present studies about biogenic amine detection related to food samples. The rationale of this study is also to provide data on the comparison of the analytical approaches between LC and GC methods. Furthermore, the various approaches of biogenic amine determination and the most applied analytical methods have been reviewed.
Collapse
Affiliation(s)
| | - Khairiahi Haji Badri
- Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| |
Collapse
|
38
|
Ye B, Zhou C, Guo H, Zheng M. Effects of BTK signalling in pathogenic microorganism infections. J Cell Mol Med 2019; 23:6522-6529. [PMID: 31397086 PMCID: PMC6787465 DOI: 10.1111/jcmm.14548] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/22/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023] Open
Abstract
As a cytoplasmic protein tyrosine kinase, Bruton's tyrosine kinase (Btk) is widely considered as a vital kinase in many aspects of different physiologic processes. It is engaged in many important signalling pathways related to the immune response, such as the B cell receptor pathway, pattern-recognition receptor pathway, and triggering receptor expressed on myeloid cell pathway. Recent studies have increasingly focused on the important role of Btk in various inflammatory diseases, which are related to Btk expression in myeloid innate immune cells, such as macrophages, dendritic cells and neutrophils. Although some investigations have explored the role of Btk in microbial infections, many aspects remain elusive, and some of the results are opposite and controversial. Considering the complicated and multiple roles of Btk in the immune system, we summarized the engagement of Btk signalling in various pathogenic microorganism infections, the possible mechanisms involved and its therapeutic potential in the control of infectious diseases.
Collapse
Affiliation(s)
- Bingjue Ye
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of School of MedicineZhejiang UniversityHangzhouChina
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesHangzhouChina
| | - Cheng Zhou
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of School of MedicineZhejiang UniversityHangzhouChina
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesHangzhouChina
| | - Huiting Guo
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of School of MedicineZhejiang UniversityHangzhouChina
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesHangzhouChina
| | - Min Zheng
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of School of MedicineZhejiang UniversityHangzhouChina
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesHangzhouChina
| |
Collapse
|
39
|
Gamperl S, Stefanzl G, Peter B, Smiljkovic D, Bauer K, Willmann M, Valent P, Hadzijusufovic E. Effects of ibrutinib on proliferation and histamine release in canine neoplastic mast cells. Vet Comp Oncol 2019; 17:553-561. [PMID: 31286638 PMCID: PMC6900099 DOI: 10.1111/vco.12520] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 01/08/2023]
Abstract
The Bruton's tyrosine kinase (BTK) inhibitor ibrutinib is effective in the treatment of human chronic lymphocytic leukaemia and mantle cell lymphoma. Recent data have shown that ibrutinib also blocks IgE-dependent activation and histamine release in human basophils (BAs) and mast cells (MCs). The aim of this study was to investigate whether BTK serves as a novel therapeutic target in canine mast cell tumours (MCTs). We evaluated the effects of ibrutinib on two canine MC lines, C2 and NI-1 and on primary MCs obtained from canine MCTs (n = 3). Using flow cytometry, we found that ibrutinib suppresses phosphorylation of BTK and of downstream STAT5 in both MC lines. In addition, ibrutinib decreased proliferation of neoplastic MCs, with IC50 values ranging between 0.1 and 1 μM in primary MCT cells and between 1 and 3 μM in C2 and NI-1 cells. In C2 cells, the combination "ibrutinib + midostaurin" produced synergistic growth-inhibitory effects. At higher concentrations, ibrutinib also induced apoptosis in both MC lines. Finally, ibrutinib was found to suppress IgE-dependent histamine release in primary MCT cells, with IC50 values ranging from 0.05 to 0.1 μM in NI-1 cells, and from 0.05 to 1 μM in primary MCT cells. In summary, ibrutinib exerts anti-proliferative effects in canine neoplastic MCs and counteracts IgE-dependent histamine release in these cells. Based on our data, ibrutinib may be considered as a novel therapeutic agent for the treatment of canine MCT. The value of BTK inhibition in canine MCT patients remains to be elucidated in clinical trials.
Collapse
Affiliation(s)
- Susanne Gamperl
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Gabriele Stefanzl
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Barbara Peter
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Dubravka Smiljkovic
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Karin Bauer
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Michael Willmann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria.,Department/Hospital for Companion Animals and Horses, University Clinic for Small Animals, Internal Medicine Small Animals, University of Veterinary Medicine, Vienna, Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Emir Hadzijusufovic
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria.,Department/Hospital for Companion Animals and Horses, University Clinic for Small Animals, Internal Medicine Small Animals, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
40
|
Abstract
Higher organisms are all born with general immunity as well as with, increasingly, more specific immune systems. All immune mechanisms function with the intent of aiding the body in defense against infection. Internal and external factors alike have varying effects on the immune system, and the immune response is tailored specifically to each one. Accompanying the components of the human innate and adaptive immune systems are the other intermingling systems of the human body. Increasing understanding of the body's immune interactions with other systems has opened new avenues of study, including that of the microbiome. The microbiome has become a highly active area of research over the last 10 to 20 years since the NIH began funding the Human Microbiome Project (HMP), which was established in 2007. Several publications have focused on the characterization, functions, and complex interplay of the microbiome as it relates to the rest of the body. A dysfunction between the microbiome and the host has been linked to various diseases including cancers, metabolic deficiencies, autoimmune disorders, and infectious diseases. Further understanding of the microbiome and its interaction with the host in relation to diseases is needed in order to understand the implications of microbiome dysfunction and the possible use of microbiota in the prevention of disease. In this review, we have summarized information on the immune system, the microbiome, the microbiome's interplay with other systems, and the association of the immune system and the microbiome in diseases such as diabetes and colorectal cancer.
Collapse
Affiliation(s)
| | - Sohail Siraj
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX
| | - Krishna Patel
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX
| | - Umesh T. Sankpal
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX
| | - Stephen Mathew
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX
| | - Riyaz Basha
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX
| |
Collapse
|
41
|
L'Italien L, Orozco O, Abrams T, Cantagallo L, Connor A, Desai J, Ebersbach H, Gelderblom H, Hoffmaster K, Lees E, Maacke H, Schleyer S, Skegro D, Lee-Hoeflich ST. Mechanistic Insights of an Immunological Adverse Event Induced by an Anti-KIT Antibody Drug Conjugate and Mitigation Strategies. Clin Cancer Res 2018; 24:3465-3474. [PMID: 29615457 DOI: 10.1158/1078-0432.ccr-17-3786] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/06/2018] [Accepted: 03/27/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Hypersensitivity reactions (HSRs) were observed in three patients dosed in a phase I clinical trial treated with LOP628, a KIT targeted antibody drug conjugate. Mast cell degranulation was implicated as the root cause for the HSR. Underlying mechanism of this reported HSR was investigated with an aim to identifying potential mitigation strategies.Experimental Design: Biomarkers for mast cell degranulation were evaluated in patient samples and in human peripheral blood cell-derived mast cell (PBC-MC) cultures treated with LOP628. Mitigation strategies interrogated include pretreatment of mast cells with small molecule inhibitors that target KIT or signaling pathways downstream of FcεR1, FcγR, and treatment with Fc silencing antibody formats.Results: Transient elevation of serum tryptase was observed in patients 1-hour posttreatment of LOP628. In agreement with the clinical observation, LOP628 and its parental antibody LMJ729 induced degranulation of human PBC-MCs. Unexpectedly, KIT small molecule inhibitors did not abrogate mast cell degranulation. By contrast, small molecule inhibitors that targeted pathways downstream of Fc receptors blunted degranulation. Furthermore, interference of the KIT antibody to engage Fc receptors by pre-incubation with IgG or using engineered Fc silencing mutations reduced or prevented degranulation. Characterization of Fcγ receptors revealed human PBC-MCs expressed both FcγRII and low levels of FcγRI. Interestingly, increasing the level of FcγRI upon addition of IFNγ, significantly enhanced LOP628-mediated mast cell degranulation.Conclusions: Our data suggest LOP628-mediated mast cell degranulation is the likely cause of HSR observed in the clinic due to co-engagement of the FcγR and KIT, resulting in mast cell activation. Clin Cancer Res; 24(14); 3465-74. ©2018 AACR.
Collapse
Affiliation(s)
| | - Olivia Orozco
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Tinya Abrams
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Lisa Cantagallo
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Anu Connor
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Jayesh Desai
- Royal Melbourne Hospital, Parkville VIC, Australia
| | - Hilmar Ebersbach
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | | | - Keith Hoffmaster
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Emma Lees
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Heiko Maacke
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Siew Schleyer
- Novartis Institutes for Biomedical Research, Shanghai, China
| | - Darko Skegro
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | | |
Collapse
|
42
|
Chirumbolo S, Bjørklund G, Sboarina A, Vella A. The role of basophils as innate immune regulatory cells in allergy and immunotherapy. Hum Vaccin Immunother 2018; 14:815-831. [PMID: 29257936 DOI: 10.1080/21645515.2017.1417711] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Basophils are circulating cells that are associated quite exclusively with allergy response and hypersensitivity reactions but their role in the immune network might be much more intriguing and complex than previously expected. The feasibility of testing their biology in vitro for allergy research and diagnosis, due fundamentally to their quite easy availability in the peripheral blood, made them the major source for assessing allergy in the laboratory assay, when yet many further cells such as mast cells and eosinophils are much more involved as effector cells in allergy than circulating basophils. Interestingly, basophil numbers change rarely in peripheral blood during an atopic response, while we might yet observe an increase in eosinophils and modification in the biology of mast cells in the tissue during an hypersensitivity response. Furthermore, the fact that basophils are very scanty in numbers suggests that they should mainly serve as regulatory cells in immunity, rather than effector leukocytes, as still believed by the majority of physicians. In this review we will try to describe and elucidate the possible role of these cells, known as "innate IL4-producing cells" in the immune regulation of allergy and their function in allergen immunotherapy.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- a Department of Neurological and Movement Sciences , University of Verona , Verona , Italy
| | - Geir Bjørklund
- b Council for Nutritional and Environmental Medicine (CONEM) , Mo i Rana , Norway
| | - Andrea Sboarina
- c Department of Surgery , Dentistry, Paediatrics and Gynaecology-University of Verona , Verona , Italy
| | - Antonio Vella
- d Unit of Immunology-Azienda Ospedaliera Universitaria Integrata (AOUI) , Verona , Italy
| |
Collapse
|
43
|
Smiljkovic D, Blatt K, Stefanzl G, Dorofeeva Y, Skrabs C, Focke‐Tejkl M, Sperr WR, Jaeger U, Valenta R, Valent P. BTK inhibition is a potent approach to block IgE-mediated histamine release in human basophils. Allergy 2017; 72:1666-1676. [PMID: 28328081 PMCID: PMC5655929 DOI: 10.1111/all.13166] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2017] [Indexed: 12/29/2022]
Abstract
Background Recent data suggest that Bruton's tyrosine kinase (BTK) is an emerging therapeutic target in IgE receptor (IgER)‐cross‐linked basophils. Methods We examined the effects of four BTK inhibitors (ibrutinib, dasatinib, AVL‐292, and CNX‐774) on IgE‐dependent activation and histamine release in blood basophils obtained from allergic patients (n=11) and nonallergic donors (n=5). In addition, we examined the effects of these drugs on the growth of the human basophil cell line KU812 and the human mast cell line HMC‐1. Results All four BTK blockers were found to inhibit anti‐IgE‐induced histamine release from basophils in nonallergic subjects and allergen‐induced histamine liberation from basophils in allergic donors. Drug effects on allergen‐induced histamine release were dose dependent, with IC50 values ranging between 0.001 and 0.5 μmol/L, and the following rank order of potency: ibrutinib>AVL‐292>dasatinib>CNX‐774. The basophil‐targeting effect of ibrutinib was confirmed by demonstrating that IgE‐dependent histamine release in ex vivo blood basophils is largely suppressed in a leukemia patient treated with ibrutinib. Dasatinib and ibrutinib were also found to counteract anti‐IgE‐induced and allergen‐induced upregulation of CD13, CD63, CD164, and CD203c on basophils, whereas AVL‐292 and CNX‐774 showed no significant effects. Whereas dasatinib and CNX‐774 were found to inhibit the growth of HMC‐1 cells and KU812 cells, no substantial effects were seen with ibrutinib or AVL‐292. Conclusions BTK‐targeting drugs are potent inhibitors of IgE‐dependent histamine release in human basophils. The clinical value of BTK inhibition in the context of allergic diseases remains to be determined.
Collapse
Affiliation(s)
- D. Smiljkovic
- Department of Internal Medicine I Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
| | - K. Blatt
- Department of Internal Medicine I Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
- Ludwig Boltzmann Cluster Oncology Medical University of Vienna Vienna Austria
| | - G. Stefanzl
- Department of Internal Medicine I Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
- Ludwig Boltzmann Cluster Oncology Medical University of Vienna Vienna Austria
| | - Y. Dorofeeva
- Division of Immunopathology Department of Pathophysiology and Allergy Research Center for Pathophysiology, Immunology and Infectiology Medical University of Vienna Vienna Austria
| | - C. Skrabs
- Department of Internal Medicine I Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
| | - M. Focke‐Tejkl
- Division of Immunopathology Department of Pathophysiology and Allergy Research Center for Pathophysiology, Immunology and Infectiology Medical University of Vienna Vienna Austria
| | - W. R. Sperr
- Department of Internal Medicine I Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
- Ludwig Boltzmann Cluster Oncology Medical University of Vienna Vienna Austria
| | - U. Jaeger
- Department of Internal Medicine I Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
- Ludwig Boltzmann Cluster Oncology Medical University of Vienna Vienna Austria
| | - R. Valenta
- Division of Immunopathology Department of Pathophysiology and Allergy Research Center for Pathophysiology, Immunology and Infectiology Medical University of Vienna Vienna Austria
| | - P. Valent
- Department of Internal Medicine I Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
- Ludwig Boltzmann Cluster Oncology Medical University of Vienna Vienna Austria
| |
Collapse
|
44
|
Akin C. Mast cell activation syndromes. J Allergy Clin Immunol 2017; 140:349-355. [PMID: 28780942 DOI: 10.1016/j.jaci.2017.06.007] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 06/20/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022]
Abstract
Mast cell activation is common and possibly necessary for maintenance of survival. Disordered mast cell activation occurs when mast cells are pathologically overproduced or if their activation is out of proportion to the perceived threat to homeostasis. Mast cell activation syndrome refers to a group of disorders with diverse causes presenting with episodic multisystem symptoms as the result of mast cell mediator release. Despite introduction of diagnostic criteria and some advances in treatment in the last decade, many areas of mast cell activation syndrome are in need of research. This article reviews our current knowledge about the various types of mast cell activation disorders, their treatment, and areas of uncertainty in need of future investigation.
Collapse
Affiliation(s)
- Cem Akin
- Division of Allergy and Clinical Immunology, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
45
|
Yu Y, Wang P, Bian L, Hong S. Rare Death Via Histamine Poisoning Following Crab Consumption: A Case Report. J Forensic Sci 2017; 63:980-982. [DOI: 10.1111/1556-4029.13611] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/19/2017] [Accepted: 07/05/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Yang Yu
- Department of Anatomy; Kunming Medical University; Kunming China
| | - Ping Wang
- Department of Anatomy; Kunming Medical University; Kunming China
| | - Ligong Bian
- Department of Anatomy; Kunming Medical University; Kunming China
| | - Shijun Hong
- School of Forensic Medicine; Kunming Medical University; Kunming China
| |
Collapse
|