1
|
Fan Y, Feng R, Zhang X, Wang ZL, Xiong F, Zhang S, Zhong ZF, Yu H, Zhang QW, Zhang Z, Wang Y, Li G. Encoding and display technologies for combinatorial libraries in drug discovery: The coming of age from biology to therapy. Acta Pharm Sin B 2024; 14:3362-3384. [PMID: 39220863 PMCID: PMC11365444 DOI: 10.1016/j.apsb.2024.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/19/2024] [Accepted: 04/08/2024] [Indexed: 09/04/2024] Open
Abstract
Drug discovery is a sophisticated process that incorporates scientific innovations and cutting-edge technologies. Compared to traditional bioactivity-based screening methods, encoding and display technologies for combinatorial libraries have recently advanced from proof-of-principle experiments to promising tools for pharmaceutical hit discovery due to their high screening efficiency, throughput, and resource minimization. This review systematically summarizes the development history, typology, and prospective applications of encoding and displayed technologies, including phage display, ribosomal display, mRNA display, yeast cell display, one-bead one-compound, DNA-encoded, peptide nucleic acid-encoded, and new peptide-encoded technologies, and examples of preclinical and clinical translation. We discuss the progress of novel targeted therapeutic agents, covering a spectrum from small-molecule inhibitors and nonpeptidic macrocycles to linear, monocyclic, and bicyclic peptides, in addition to antibodies. We also address the pending challenges and future prospects of drug discovery, including the size of screening libraries, advantages and disadvantages of the technology, clinical translational potential, and market space. This review is intended to establish a comprehensive high-throughput drug discovery strategy for scientific researchers and clinical drug developers.
Collapse
Affiliation(s)
- Yu Fan
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- Zhuhai UM Science and Technology Research Institute, Zhuhai 519031, China
| | - Ruibing Feng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Xinya Zhang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- Zhuhai UM Science and Technology Research Institute, Zhuhai 519031, China
| | - Zhen-Liang Wang
- Geriatric Medicine, First People's Hospital of XinXiang and the Fifth Affiliated Hospital of Xinxiang Medical College, Xinxiang 453100, China
| | - Feng Xiong
- Shenzhen Innovation Center for Small Molecule Drug Discovery Co., Ltd., Shenzhen 518000, China
| | - Shuihua Zhang
- Shenzhen Innovation Center for Small Molecule Drug Discovery Co., Ltd., Shenzhen 518000, China
| | - Zhang-Feng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Hua Yu
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Qing-Wen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Zhang Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of People's Republic of China, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Department of Pharmacy, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou 510632, China
| | - Yitao Wang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Guodong Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- Zhuhai UM Science and Technology Research Institute, Zhuhai 519031, China
| |
Collapse
|
2
|
Dattola A, Tolone M, Amore E, Bennardo L, Trovato F, Amato S, Grieco T, Richetta AG, Pellacani G, Skroza N, Nisticò SP. Interleukin-13 Inhibitors in the Treatment of Atopic Dermatitis: The Role of Tralokinumab. Dermatol Pract Concept 2024; 14:dpc.1403a204. [PMID: 39122503 PMCID: PMC11314215 DOI: 10.5826/dpc.1403a204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 08/12/2024] Open
Abstract
INTRODUCTION The advent of biotechnological drugs has significantly changed the management of atopic dermatitis (AD) and the approach to the moderate-to-severe form of this chronic relapsing disease. OBJECTIVES The aim of our review is to summarize the current literature on anti-interleukin (IL)-13 in atopic dermatitis. METHODS A literature search was organized and a systematic review was performed to summarize the most recent evidence supporting the efficacy and safety of tralokinumab. RESULTS Tralokinumab (anti-IL-13) 300 mg every 2 weeks subcutaneously has proven effective in several clinical trials in adults and adolescents with moderate to severe atopic dermatitis inadequately controlled with other topical or systemic therapies. Tralokinumab was found to be significantly superior in terms of efficacy in reducing Investigator's Global Assessment (IGA), Eczema Area and Severity Index (EASI) -75, Numeric Pain Rating Scale (NRS) pruritus, and Dermatology Life Quality Index (DLQI) scale numbers. During follow-up, tralokinumab was well tolerated with limited severity of adverse events. CONCLUSIONS Tralokinumab leads to statistically significant improvements in disease severity and outcome scores. It represents an effective treatment option for adults with moderate to severe AD, but further large-scale studies are needed to verify long-term superiority over other treatments.
Collapse
Affiliation(s)
| | - Martina Tolone
- Department of Health Sciences- Unit of Dermatology, Magna Graecia University, Catanzaro, Italy
| | - Emanuele Amore
- Department of Dermatology University of Rome "La Sapienza", Rome, Italy
| | - Luigi Bennardo
- Department of Health Sciences- Unit of Dermatology, Magna Graecia University, Catanzaro, Italy
| | - Federica Trovato
- Department of Dermatology University of Rome "La Sapienza", Rome, Italy
| | - Simone Amato
- Department of Dermatology University of Rome "La Sapienza", Rome, Italy
| | - Teresa Grieco
- Department of Dermatology University of Rome "La Sapienza", Rome, Italy
| | | | | | - Nevena Skroza
- Dermatology Unit 'Daniele Innocenzi', Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome Polo Pontino, Latina, Italy
| | - Steven Paul Nisticò
- Department of Dermatology University of Rome "La Sapienza", Rome, Italy
- Department of Health Sciences- Unit of Dermatology, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
3
|
Guttman-Yassky E, Kabashima K, Staumont-Salle D, Nahm WK, Pauser S, Da Rosa JC, Martel BC, Madsen DE, Røpke M, Arlert P, Steffensen L, Blauvelt A, Reich K. Targeting IL-13 with tralokinumab normalizes type 2 inflammation in atopic dermatitis both early and at 2 years. Allergy 2024; 79:1560-1572. [PMID: 38563683 DOI: 10.1111/all.16108] [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: 09/06/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Tralokinumab is a monoclonal antibody that specifically neutralizes interleukin (IL)-13, a key driver of skin inflammation and barrier abnormalities in atopic dermatitis (AD). This study evaluated early and 2-year impacts of IL-13 neutralization on skin and serum biomarkers following tralokinumab treatment in adults with moderate-to-severe AD. METHODS Skin biopsies and blood samples were evaluated from a subset of patients enrolled in the Phase 3 ECZTRA 1 (NCT03131648) and the long-term extension ECZTEND (NCT03587805) trials. Gene expression was assessed by RNA sequencing; protein expression was assessed by immunohistochemistry and immunoassay. RESULTS Tralokinumab improved the transcriptomic profile of lesional skin by Week 4. Mean improvements in the expression of genes dysregulated in AD were 39% at Week 16 and 85% at 2 years with tralokinumab, with 15% worsening at Week 16 with placebo. At Week 16, tralokinumab significantly decreased type 2 serum biomarkers (CCL17/TARC, periostin, and IgE), reduced epidermal thickness versus placebo, and increased loricrin coverage versus baseline. Two years of tralokinumab treatment significantly reduced expression of genes in the Th2 (IL4R, IL31, CCL17, and CCL26), Th1 (IFNG), and Th17/Th22 (IL22, S100A7, S100A8, and S100A9) pathways as well as increased expression of epidermal differentiation and barrier genes (CLDN1 and LOR). Tralokinumab also shifted atherosclerosis signaling pathway genes (SELE, IL-37, and S100A8) toward non-lesional expression. CONCLUSION Tralokinumab treatment improved epidermal pathology, reduced systemic markers of type 2 inflammation, and shifted expression of key AD biomarkers in skin towards non-lesional levels, further highlighting the key role of IL-13 in the pathogenesis of AD. CLINICAL TRIAL REGISTRATION NCT03131648, NCT03587805.
Collapse
Affiliation(s)
- Emma Guttman-Yassky
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kenji Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Delphine Staumont-Salle
- Department of Dermatology, University Hospital of Lille, INFINITE (Institute for Translational Research) U1286 Inserm, University of Lille, Lille, France
| | - Walter K Nahm
- University of California, San Diego School of Medicine, San Diego, California, USA
| | | | - Joel Correa Da Rosa
- Mount Sinai Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | | | | | | | | | - Kristian Reich
- Translational Research in Inflammatory Skin Diseases, Institute for Health Services Research in Dermatology and Nursing, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
4
|
Bernstein ZJ, Shenoy A, Chen A, Heller NM, Spangler JB. Engineering the IL-4/IL-13 axis for targeted immune modulation. Immunol Rev 2023; 320:29-57. [PMID: 37283511 DOI: 10.1111/imr.13230] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/19/2023] [Indexed: 06/08/2023]
Abstract
The structurally and functionally related interleukin-4 (IL-4) and IL-13 cytokines play pivotal roles in shaping immune activity. The IL-4/IL-13 axis is best known for its critical role in T helper 2 (Th2) cell-mediated Type 2 inflammation, which protects the host from large multicellular pathogens, such as parasitic helminth worms, and regulates immune responses to allergens. In addition, IL-4 and IL-13 stimulate a wide range of innate and adaptive immune cells, as well as non-hematopoietic cells, to coordinate various functions, including immune regulation, antibody production, and fibrosis. Due to its importance for a broad spectrum of physiological activities, the IL-4/IL-13 network has been targeted through a variety of molecular engineering and synthetic biology approaches to modulate immune behavior and develop novel therapeutics. Here, we review ongoing efforts to manipulate the IL-4/IL-13 axis, including cytokine engineering strategies, formulation of fusion proteins, antagonist development, cell engineering approaches, and biosensor design. We discuss how these strategies have been employed to dissect IL-4 and IL-13 pathways, as well as to discover new immunotherapies targeting allergy, autoimmune diseases, and cancer. Looking ahead, emerging bioengineering tools promise to continue advancing fundamental understanding of IL-4/IL-13 biology and enabling researchers to exploit these insights to develop effective interventions.
Collapse
Affiliation(s)
- Zachary J Bernstein
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anjali Shenoy
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amy Chen
- Department of Molecular and Cellular Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
- Division of Allergy and Clinical Immunology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jamie B Spangler
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Cancer Center, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Barker KH, Higham JP, Pattison LA, Chessell IP, Welsh F, Smith ESJ, Bulmer DC. Sensitization of colonic nociceptors by IL-13 is dependent on JAK and p38 MAPK activity. Am J Physiol Gastrointest Liver Physiol 2023; 324:G250-G261. [PMID: 36749569 PMCID: PMC10010921 DOI: 10.1152/ajpgi.00280.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The effective management of visceral pain is a significant unmet clinical need for those affected by gastrointestinal diseases, such as inflammatory bowel disease (IBD). The rational design of novel analgesics requires a greater understanding of the mediators and mechanisms underpinning visceral pain. Interleukin-13 (IL-13) production by immune cells residing in the gut is elevated in IBD, and IL-13 appears to be important in the development of experimental colitis. Furthermore, receptors for IL-13 are expressed by neurons innervating the colon, though it is not known whether IL-13 plays any role in visceral nociception per se. To resolve this, we used Ca2+ imaging of cultured sensory neurons and ex vivo electrophysiological recording from the lumbar splanchnic nerve innervating the distal colon. Ca2+ imaging revealed the stimulation of small-diameter, capsaicin-sensitive sensory neurons by IL-13, indicating that IL-13 likely stimulates nociceptors. IL-13-evoked Ca2+ signals were attenuated by inhibition of Janus (JAK) and p38 kinases. In the lumbar splanchnic nerve, IL-13 did not elevate baseline firing, nor sensitize the response to capsaicin application, but did enhance the response to distention of the colon. In line with Ca2+ imaging experiments, IL-13-mediated sensitization of the afferent response to colon distention was blocked by inhibition of either JAK or p38 kinase signaling. Together, these data highlight a potential role for IL-13 in visceral nociception and implicate JAK and p38 kinases in pronociceptive signaling downstream of IL-13.
Collapse
Affiliation(s)
- Katie H Barker
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - James P Higham
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Luke A Pattison
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Iain P Chessell
- Department of Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Fraser Welsh
- Department of Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Ewan St J Smith
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Ramírez-Jiménez F, Pavón-Romero GF, Velásquez-Rodríguez JM, López-Garza MI, Lazarini-Ruiz JF, Gutiérrez-Quiroz KV, Teran LM. Biologic Therapies for Asthma and Allergic Disease: Past, Present, and Future. Pharmaceuticals (Basel) 2023; 16:270. [PMID: 37259416 PMCID: PMC9963709 DOI: 10.3390/ph16020270] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 09/18/2024] Open
Abstract
The discovery of the mechanism underlying allergic disease, mouse models of asthma, and bronchoscopy studies provided initial insights into the role of Th2-type cytokines, including interlukin (IL)-4, IL-5 and IL-13, which became the target of monoclonal antibody therapy. Omalizumab, Benralizumab, Mepolizumab, Reslizumab, and Tezepelumab have been approved. These biologicals have been shown to be good alternative therapies to corticosteroids, particularly in severe asthma management, where they can improve the quality of life of many patients. Given the success in asthma, these drugs have been used in other diseases with type 2 inflammation, including chronic rhinosinusitis with nasal polyps (CRSwNP), atopic dermatitis, and chronic urticaria. Like the Th2-type cytokines, chemokines have also been the target of novel monoclonal therapies. However, they have not proved successful to date. In this review, targeted therapy is addressed from its inception to future applications in allergic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Luis M. Teran
- Immunogenetics and Allergy Department, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, (INER), Mexico City 14080, Mexico
| |
Collapse
|
7
|
Soehoel A, Larsen MS, Timmermann S. Population Pharmacokinetics of Tralokinumab in Adult Subjects With Moderate to Severe Atopic Dermatitis. Clin Pharmacol Drug Dev 2022; 11:910-921. [PMID: 35671038 PMCID: PMC9796478 DOI: 10.1002/cpdd.1113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/02/2022] [Indexed: 01/01/2023]
Abstract
Tralokinumab is the first biologic therapy for moderate-to-severe atopic dermatitis (AD) that specifically neutralizes interleukin-13 activity, a key driver of AD signs and symptoms. Tralokinumab is a human immunoglobulin G4 monoclonal antibody administered subcutaneously every 2 weeks (with possibility of maintenance dosing every 4 weeks). This population pharmacokinetic (PK) analysis aimed to identify sources of PK variability and relevant predictors of tralokinumab exposure in adults with moderate to severe AD. Nonlinear mixed-effect modeling, including covariate analysis, was used on a data set including 2561 subjects (AD, asthma, healthy) from 10 clinical trials. A 2-compartment model with first-order absorption and elimination adequately described the tralokinumab PK. Body weight was identified as a relevant predictor of tralokinumab exposure; other covariates including age, sex, race, ethnicity, disease type, AD severity, and renal and hepatic impairment were not. For body weight, the difference in exposure between the upper- and lower-weight quartiles in patients with AD was <2-fold, supporting the appropriateness of flat dosing (300 mg). Given the reduced exposure associated with higher body weight, coupled with the reduced exposure provided by dosing every 4 weeks, it is uncertain whether higher-weight patients will achieve sufficient exposure to maintain efficacy if dosed every 4 weeks instead of the standard every 2 weeks.
Collapse
Affiliation(s)
| | - Malte Selch Larsen
- Clinical PharmacologyLEO Pharma A/SBallerupDenmark,Present address:
Novo NordiskSøborgDenmark
| | | |
Collapse
|
8
|
Wang B, Goodman J, Roskos LK. Mechanistic modeling of a human IgG
4
monoclonal antibody (tralokinumab) Fab‐arm exchange with endogenous IgG
4
in healthy volunteers. CPT Pharmacometrics Syst Pharmacol 2022; 11:438-446. [PMID: 35023315 PMCID: PMC9007600 DOI: 10.1002/psp4.12738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/16/2020] [Accepted: 03/29/2020] [Indexed: 11/23/2022] Open
Abstract
Therapeutic IgG4 antibodies engage in Fab‐arm exchange with endogenous human immunoglobulin G4 (IgG4) to form monovalent hybrid molecules. A mechanistic population model was developed to quantitatively characterize the dynamic Fab‐arm exchange of tralokinumab, a human IgG4 monoclonal antibody currently being developed for the treatment of atopic dermatitis, with endogenous IgG4 in healthy volunteers. The estimated pharmacokinetic parameters for IgG4 were similar to those of immunoglobulin G1 or immunoglobulin G2 in humans. However, the mechanistically modeled clearance of half molecules is 21‐fold higher, likely due to the loss of avidity for the neonatal Fc receptor. Half molecules of tralokinumab randomly associate with those of endogenous IgG4 to form monovalent hybrid molecules, which became the dominant form of tralokinumab within 1 day postdose in healthy volunteers. As the potency of monovalent tralokinumab is comparable with that of bivalent tralokinumab, the IgG4 Fab‐arm exchange with endogenous IgG4 is not expected to affect the potency of neutralization of interleukin‐13 in vivo.
Collapse
Affiliation(s)
- Bing Wang
- Amador Bioscience Pleasanton California USA
| | - Jo Goodman
- Clinical Pharmacology and Safety Sciences AstraZeneca BioPharmaceuticals, R&D Cambridge UK
| | - Lorin K. Roskos
- Clinical Pharmacology and Safety Sciences AstraZeneca BioPharmaceuticals, R&D Gaithersburg Maryland USA
| |
Collapse
|
9
|
Chiricozzi A, Gori N, Maurelli M, Gisondi P, Caldarola G, De Simone C, Peris K, Girolomoni G. Biological agents targeting interleukin-13 for atopic dermatitis. Expert Opin Biol Ther 2022; 22:651-659. [PMID: 35081849 DOI: 10.1080/14712598.2022.2035356] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Atopic dermatitis (AD) is a chronic inflammatory skin disease that is pathogenically driven by type-2 inflammation. Interleukin-13 (IL-13) plays a central role in AD pathogenesis, as confirmed by the clinical efficacy of agents that selectively block IL-13, although their therapeutic value and place-in-therapy are incompletely defined. AREAS COVERED The aim of this review article is to describe preclinical and clinical data regarding selective IL-13 inhibitors investigated in AD. In particular, we discuss the clinical outcomes obtained with lebrikizumab and tralokinumab, which are in a more advanced phase of development. EXPERT OPINION Biological agents that neutralize IL-13 have demonstrated clinical benefits in treating AD with excellent safety profiles. Robust clinical evidence exists in support of tralokinumab, which underwent phase III trials, met the predefined primary endpoints, and is approaching the market. In contrast, clinical trial testing for lebrikizumab needs to be completed to fully assess its therapeutic potential. PLAIN LANGUAGE SUMMARY Atopic dermatitis (AD) is a chronic pathological inflammatory skin disease that results from type-2 inflammation. Selective interleukin-13 (IL-13) inhibitors have shown clinical efficacy against AD, suggesting that IL-13 plays a central in AD pathogenesis. However, the therapeutic value and place-in-therapy of IL-13 inhibitors are incompletely defined. The aim of this review article is to describe preclinical and clinical data for selective IL-13 inhibitors against AD, including lebrikizumab and tralokinumab, which are in a more advanced phase of development. The up-to-date overview of the strengths and limitations of different agents used to treat AD discussed in this article might be useful in driving treatment decision.
Collapse
Affiliation(s)
- Andrea Chiricozzi
- Dermatologia, Dipartimento Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Dermatologia, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Niccolò Gori
- Dermatologia, Dipartimento Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Dermatologia, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Martina Maurelli
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - Paolo Gisondi
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - Giacomo Caldarola
- Dermatologia, Dipartimento Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Dermatologia, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Clara De Simone
- Dermatologia, Dipartimento Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Dermatologia, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ketty Peris
- Dermatologia, Dipartimento Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Dermatologia, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giampiero Girolomoni
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
10
|
Gevenois PJLY, De Pauw P, Schoonooghe S, Delporte C, Sebti T, Amighi K, Muyldermans S, Wauthoz N. Development of Neutralizing Multimeric Nanobody Constructs Directed against IL-13: From Immunization to Lead Optimization. THE JOURNAL OF IMMUNOLOGY 2021; 207:2608-2620. [PMID: 34645688 DOI: 10.4049/jimmunol.2100250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/16/2021] [Indexed: 11/19/2022]
Abstract
IL-13 is a pleiotropic cytokine mainly secreted by Th2 cells. It reacts with many different types of cells involved in allergy, inflammation, and fibrosis, e.g., mastocytes, B cells, and fibroblasts. The role of IL-13 in conditions involving one or several of these phenotypes has therefore been extensively investigated. The inhibition of this cytokine in animal models for various pathologies yielded highly promising results. However, most human trials relying on anti-IL-13 conventional mAbs have failed to achieve a significant improvement of the envisaged disorders. Where some studies might have suffered from several weaknesses, the strategies themselves, such as targeting only IL-13 using conventional mAbs or employing a systemic administration, could be questioned. Nanobodies are recombinant Ag-binding fragments derived from the variable part of H chain-only Abs occurring in Camelidae. Thanks to their single-domain structure, small size (≈15 kDa), good stability, and solubility, they can be engineered into multispecific constructs for combined therapies or for use in new strategies such as formulations for local administration, e.g., pulmonary administration. In this study, we describe the generation of 38 nanobodies that can be subdivided into five CDR3 families. Nine nanobodies were found to have a good affinity profile (KD = 1-200 nM), but none were able to strongly inhibit IL-13 biological activity in vitro (IC50 > 50 µM: HEK-Blue IL-13/IL-4 cells). Multimeric constructs were therefore designed from these inhibitors and resulted in an up to 36-fold improvement in affinity and up to 300-fold enhancement of the biological activity while conserving a high specificity toward IL-13.
Collapse
Affiliation(s)
- Philippe J-L Y Gevenois
- Unit of Pharmaceutics and Biopharmaceutics, Free University of Brussels, Faculty of Pharmacy, Brussels, Belgium;
| | - Pieter De Pauw
- Laboratory of Cellular and Molecular Immunology, Free University of Brussels, Ixelles, Belgium
| | - Steve Schoonooghe
- Flemish Institute for Biotechnology Nanobody Core, Free University of Brussels, Brussels, Belgium
| | - Cédric Delporte
- Unit of Pharmacognosy, Bioanalysis and Drug Discovery, RD3 and Analytical Platform of the Faculty of Pharmacy, Free University of Brussels, Brussels, Belgium; and
| | | | - Karim Amighi
- Unit of Pharmaceutics and Biopharmaceutics, Free University of Brussels, Faculty of Pharmacy, Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Free University of Brussels, Ixelles, Belgium
| | - Nathalie Wauthoz
- Unit of Pharmaceutics and Biopharmaceutics, Free University of Brussels, Faculty of Pharmacy, Brussels, Belgium
| |
Collapse
|
11
|
Zhou S, Qi F, Gong Y, Zhang J, Zhu B. Biological Therapies for Atopic Dermatitis: A Systematic Review. Dermatology 2021; 237:542-552. [PMID: 33735876 DOI: 10.1159/000514535] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/17/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a widely acquired, relapsing inflammatory skin disease. Biologics are now widely used in patients with moderate-to-severe AD. OBJECTIVE This work aims to summarize both label and off-label biologics on AD treatment in phase II and phase III stages, and compile evidence on the efficacy of the most-studied biologics. METHODS We conducted a comprehensive literature search through PubMed, EMBASE, and ClinicalTrials.gov to identify all documented biological therapies for AD. The criteria were further refined to focus on those treatments with the highest evidence level for AD with at least one randomized clinical trial supporting their use. Only studies or articles published in English were enrolled in this study. FINDINGS Primary searches identified 525 relevant articles and 27 trials. Duplicated articles and papers without a full text were excluded. Only completed trials were enrolled. We included 28 randomized controlled trials, 4 unpublished trials, 2 observational studies, and 1 meta-analysis. Eight kinds of biologics, including IL-4/IL-13 inhibitors, JAK inhibitors, anti-IL-13 antibodies, anti-IL-22 antibodies, anti-IL-33 antibodies, thymic stromal lymphopoietin inhibitor (TSLP), OX40 antibodies, and H4R-antagonists were included in this work. Dupliumab, as the most widely used and investigated biologic, was reported in 1 meta-analysis and 4 trials exploring its long-term use and application in both adults and pediatric patients. Besides dupilumab, four other IL-4/IL-13 inhibitors recruited were all randomized, clinical trials at phase 2-3 stage. Six different kinds of JAK inhibitors were summarized with strong evidence revealing their significant therapeutic effects on AD. There were 3 trials for nemolizumab, an anti-IL-13 antibody, all of which were in the phase 2 clinical trial stage. Results showed nemolizumab could be another alternative therapy for moderate-to-severe AD with long-term efficiency and safety. CONCLUSION The biological therapies with the most robust evidence on efficacy and long-term safety for AD treatment include dupilumab, barcitinib, abrocitinib, and delgocitinib. Most of the biologics mentioned in this review were still at the exploratory stage. This review will help practitioners advise patients seeking suitable biological therapies and offer experimental study directions for treatment.
Collapse
Affiliation(s)
- Shuying Zhou
- Department of Dermatology, The 305 Hospital of PLA, Beijing, China
| | - Fei Qi
- Capital Medical University Affiliated with Beijing Chaoyang Hospital, Beijing, China,
| | - Yue Gong
- Department of Dermatology, The 305 Hospital of PLA, Beijing, China
| | - Jinping Zhang
- Department of Dermatology, The 305 Hospital of PLA, Beijing, China
| | - Binghua Zhu
- Department of Dermatology, The 305 Hospital of PLA, Beijing, China
| |
Collapse
|
12
|
Silverberg JI, Toth D, Bieber T, Alexis AF, Elewski BE, Pink AE, Hijnen D, Jensen TN, Bang B, Olsen CK, Kurbasic A, Weidinger S. Tralokinumab plus topical corticosteroids for the treatment of moderate-to-severe atopic dermatitis: results from the double-blind, randomized, multicentre, placebo-controlled phase III ECZTRA 3 trial. Br J Dermatol 2021; 184:450-463. [PMID: 33000503 PMCID: PMC7986183 DOI: 10.1111/bjd.19573] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2020] [Indexed: 12/13/2022]
Abstract
Background Tralokinumab is a fully human monoclonal antibody that specifically neutralizes interleukin‐13, a key driver of atopic dermatitis (AD). Objectives To evaluate the efficacy and safety of tralokinumab in combination with topical corticosteroids (TCS) in patients with moderate‐to‐severe AD who were candidates for systemic therapy. Methods This was a double‐blind, placebo plus TCS controlled phase III trial. Patients were randomized 2 : 1 to subcutaneous tralokinumab 300 mg or placebo every 2 weeks (Q2W) with TCS as needed over 16 weeks. Patients who achieved an Investigator’s Global Assessment (IGA) score of 0/1 and/or 75% improvement in Eczema Area and Severity Index (EASI 75) at week 16 with tralokinumab were rerandomized 1 : 1 to tralokinumab Q2W or every 4 weeks (Q4W), with TCS as needed, for another 16 weeks. Results At week 16, more patients treated with tralokinumab than with placebo achieved IGA 0/1: 38·9% vs. 26·2% [difference (95% confidence interval): 12·4% (2·9–21·9); P = 0·015] and EASI 75: 56·0% vs. 35·7% [20·2% (9·8–30·6); P < 0·001]. Of the patients who were tralokinumab responders at week 16, 89·6% and 92·5% of those treated with tralokinumab Q2W and 77·6% and 90·8% treated with tralokinumab Q4W maintained an IGA 0/1 and EASI 75 response at week 32, respectively. Among patients who did not achieve IGA 0/1 and EASI 75 with tralokinumab Q2W at 16 weeks, 30·5% and 55·8% achieved these endpoints, respectively, at week 32. The overall incidence of adverse events was similar across treatment groups. Conclusions Tralokinumab 300 mg in combination with TCS as needed was effective and well tolerated in patients with moderate‐to‐severe AD. What is already known about this topic?Atopic dermatitis (AD) is a chronic interleukin (IL)‐13‐mediated disease. In clinical practice, biologics are commonly initiated as add‐on therapy to topical corticosteroids (TCS). Tralokinumab is a fully human monoclonal antibody that binds specifically to the IL‐13 cytokine with high affinity, thereby preventing receptor interaction and subsequent downstream signalling. Tralokinumab combined with TCS showed early and sustained efficacy and safety in a 12‐week, phase IIb trial in moderate‐to‐severe AD.
What does this study add?This is the first phase III trial evaluating a targeted anti‐IL‐13 biologic in combination with TCS. These data demonstrate that tralokinumab plus TCS can achieve significant improvements in AD signs and symptoms and quality of life, as well as exert a steroid‐sparing effect. Response with tralokinumab in combination with TCS was maintained over 32 weeks. Tralokinumab may be considered a targeted biological treatment option for patients with moderate‐to‐severe AD.
Linked Comment: Morra and Drucker. Br J Dermatol 2021; 184:386–387. Plain language summary available online
Collapse
Affiliation(s)
- J I Silverberg
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - D Toth
- XLR8 Medical Research and Probity Medical Research, Windsor, ON, Canada
| | - T Bieber
- Department of Dermatology and Allergy, University Medical Center, Bonn, Germany
| | - A F Alexis
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - B E Elewski
- Department of Dermatology, University of Alabama, Birmingham, AL, USA
| | - A E Pink
- St John's Institute of Dermatology, Guy's and St Thomas' Hospitals, London, UK
| | - D Hijnen
- Department of Dermatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - B Bang
- LEO Pharma A/S, Ballerup, Denmark
| | | | | | - S Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | |
Collapse
|
13
|
León B, Ballesteros-Tato A. Modulating Th2 Cell Immunity for the Treatment of Asthma. Front Immunol 2021; 12:637948. [PMID: 33643321 PMCID: PMC7902894 DOI: 10.3389/fimmu.2021.637948] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
It is estimated that more than 339 million people worldwide suffer from asthma. The leading cause of asthma development is the breakdown of immune tolerance to inhaled allergens, prompting the immune system's aberrant activation. During the early phase, also known as the sensitization phase, allergen-specific T cells are activated and become central players in orchestrating the subsequent development of allergic asthma following secondary exposure to the same allergens. It is well-established that allergen-specific T helper 2 (Th2) cells play central roles in developing allergic asthma. As such, 80% of children and 60% of adult asthma cases are linked to an unwarranted Th2 cell response against respiratory allergens. Thus, targeting essential components of Th2-type inflammation using neutralizing antibodies against key Th2 modulators has recently become an attractive option for asthmatic patients with moderate to severe symptoms. In addition to directly targeting Th2 mediators, allergen immunotherapy, also known as desensitization, is focused on redirecting the allergen-specific T cells response from a Th2-type profile to a tolerogenic one. This review highlights the current understanding of the heterogeneity of the Th2 cell compartment, their contribution to allergen-induced airway inflammation, and the therapies targeting the Th2 cell pathway in asthma. Further, we discuss available new leads for successful targeting pulmonary Th2 cell responses for future therapeutics.
Collapse
Affiliation(s)
- Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andre Ballesteros-Tato
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
14
|
Rolla G, Heffler E, Pizzimenti S, Michils A, Malinovschi A. An Emerging Role for Exhaled Nitric Oxide in Guiding Biological Treatment in Severe Asthma. Curr Med Chem 2021; 27:7159-7167. [PMID: 32660394 DOI: 10.2174/0929867327666200713184659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/15/2020] [Accepted: 03/16/2020] [Indexed: 11/22/2022]
Abstract
Asthma is a heterogeneous disease with regard to the inflammatory pathways activated. In recent years, biologic drugs (monoclonal antibodies) directed towards specific components of type 2 inflammation have been approved for the treatment of severe asthma. Phenotyping of patients with severe asthma and evaluation of biomarkers have been recommended to help identify patients who are candidates for treatment with biologics and to monitor treatment responses. Fractional exhaled Nitric Oxide (FeNO) is a biomarker of type 2 inflammation in asthma, signaling activation of Interleukin (IL)-4/IL-13 pathway. FeNO could be useful to assess treatment response or identify candidates for a specific drug that acts on type 2 inflammation mechanisms linked to Nitric Oxide (NO) production, such as the IL-4/IL-13 pathway or upstream processes. The value of FeNO as a biomarker predictive of responses to the biologics available for treating severe asthma is discussed based on the published studies at the moment of the review.
Collapse
Affiliation(s)
- Giovanni Rolla
- Department of Medical Sciences, Allergy and Clinical Immunology AO Mauriziano, University of Torino,
Torino, Italy
| | - Enrico Heffler
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy,Personalized Medicine, Asthma and Allergy, Humanitas Clinical and Research Center, IRCCS, Rozzano,
Italy
| | - Stefano Pizzimenti
- Respiratory Medicine Unit, National Health System, ASL Città di Torino, Torino, Italy
| | - Alain Michils
- Chest Department, Erasme University Hospital, Université libre de Bruxelles, Brussels, Belgium
| | - Andrei Malinovschi
- Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
Dotter H, Boll M, Eder M, Eder AC. Library and post-translational modifications of peptide-based display systems. Biotechnol Adv 2021; 47:107699. [PMID: 33513435 DOI: 10.1016/j.biotechadv.2021.107699] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 01/04/2021] [Accepted: 01/14/2021] [Indexed: 12/27/2022]
Abstract
Innovative biotechnological methods empower the successful identification of new drug candidates. Phage, ribosome and mRNA display represent high throughput screenings, allowing fast and efficient progress in the field of targeted drug discovery. The identification range comprises low molecular weight peptides up to whole antibodies. However, a major challenge poses the stability and affinity in particular of peptides. Chemical modifications e.g. the introduction of unnatural amino acids or cyclization, have been proven to be essential tools to overcome these limitations. This review article particularly focuses on available methods for the targeted chemical modification of peptides and peptide libraries in selected display approaches.
Collapse
Affiliation(s)
- Hanna Dotter
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; Division of Radiopharmaceutical Development, German Cancer Consortium, partner site Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany, and German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Melanie Boll
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; Division of Radiopharmaceutical Development, German Cancer Consortium, partner site Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany, and German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Matthias Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; Division of Radiopharmaceutical Development, German Cancer Consortium, partner site Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany, and German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Ann-Christin Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; Division of Radiopharmaceutical Development, German Cancer Consortium, partner site Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany, and German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
16
|
Nusbaum KB, Nguyen CM, Fleischer AB. Emerging systemic therapies for atopic dermatitis: biologics. J DERMATOL TREAT 2020; 33:1269-1273. [PMID: 33045848 DOI: 10.1080/09546634.2020.1836314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND The mainstay of atopic dermatitis treatment has been largely unchanged over the last few decades. With improved understanding of the immunologic pathways underlying atopic dermatitis in recent years, targeted biologic therapies are being developed. OBJECTIVE Discuss efficacy and safety profiles of emerging biologics in phase 2 and 3 clinical trials. METHODS A systemic literature review was conducted to identify results of randomized, placebo-controlled trials of monoclonal antibodies up to March 1, 2020 for the treatment of atopic dermatitis. RESULTS Targeted biologics appear to have acceptable safety profiles. Dupilumab, lebrikizumab, and nemolizumab demonstrate efficacy as agents producing improvement in clinical severity and pruritus. CONCLUSIONS The growing class of biologics shows promise in meeting the needs of treatment-resistant atopic dermatitis. The use of validated core measurements is necessary for future trials in order to adequately compare agents and progress evidence-based medicine.
Collapse
Affiliation(s)
- Kelsey B Nusbaum
- University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Catherine M Nguyen
- Department of Dermatology, University of Cincinnati, Cincinnati, OH, USA
| | - Alan B Fleischer
- Department of Dermatology, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
17
|
In vitro evolution of antibody affinity via insertional scanning mutagenesis of an entire antibody variable region. Proc Natl Acad Sci U S A 2020; 117:27307-27318. [PMID: 33067389 DOI: 10.1073/pnas.2002954117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We report a systematic combinatorial exploration of affinity enhancement of antibodies by insertions and deletions (InDels). Transposon-based introduction of InDels via the method TRIAD (transposition-based random insertion and deletion mutagenesis) was used to generate large libraries with random in-frame InDels across the entire single-chain variable fragment gene that were further recombined and screened by ribosome display. Knowledge of potential insertion points from TRIAD libraries formed the basis of exploration of length and sequence diversity of novel insertions by insertional-scanning mutagenesis (InScaM). An overall 256-fold affinity improvement of an anti-IL-13 antibody BAK1 as a result of InDel mutagenesis and combination with known point mutations validates this approach, and suggests that the results of this InDel mutagenesis and conventional exploration of point mutations can synergize to generate antibodies with higher affinity.
Collapse
|
18
|
Abstract
Chronic pruritus, defined as an unpleasant sensation resulting in a need to scratch that lasts more than 6 weeks, is a prevalent and bothersome symptom associated with both cutaneous and systemic conditions. Due to complex pathogenesis and profuse contributing factors, chronic pruritus therapy remains challenging. Regardless of the well-established antipruritic properties of classic pharmacotherapy (topical therapy, phototherapy and systemic therapy), these methods often provide insufficient relief for affected individuals. Owing to the growing interest in the field of pruritic research, further experimental and clinical data have emerged, continuously supporting the possibility of instigating novel therapeutic measures. This review covers the most relevant current modalities remaining under investigation that possess promising perspectives of approval in the near future, especially opioidergic drugs (mu-opioid antagonists and kappa-opioid agonists), neurokinin-1 receptor antagonists, biologic drugs, Janus kinase inhibitors, ileal bile acid transporter inhibitors, aryl hydrocarbon receptor agonists and histamine H4 receptor antagonists.
Collapse
Affiliation(s)
- Radomir Reszke
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, 1 Chalubinskiego Street, 50-368, Wrocław, Poland
| | - Piotr Krajewski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, 1 Chalubinskiego Street, 50-368, Wrocław, Poland
| | - Jacek C Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, 1 Chalubinskiego Street, 50-368, Wrocław, Poland.
| |
Collapse
|
19
|
Alfaleh MA, Alsaab HO, Mahmoud AB, Alkayyal AA, Jones ML, Mahler SM, Hashem AM. Phage Display Derived Monoclonal Antibodies: From Bench to Bedside. Front Immunol 2020; 11:1986. [PMID: 32983137 PMCID: PMC7485114 DOI: 10.3389/fimmu.2020.01986] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage–derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
20
|
Sharma V, Chouhan P, Pandey RK, Ojha R, Aathmanathan VS, Krishnan M, Prajapati VK. Immunoglobulin interface redesigning to enhance lebrikizumab mediated immunomodulation of IL-13 hyper-response. J Biomol Struct Dyn 2020; 39:4051-4065. [PMID: 32448082 DOI: 10.1080/07391102.2020.1773316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The overexpression of interleukin-13 (IL-13) leads to autoimmune and inflammatory diseases. These adverse responses can be neutralized by using lebrikizumab as a therapeutic monoclonal antibody (mAb). Herein, we have attempted to modulate the lebrikizumab mAb to enhance its binding affinity towards IL-13. The interface residues of the lebrikizumab-IL-13 complex were determined by the PyMOL and verified by the artificial neural network-based B-cell epitope prediction server (ABCpred server) and the Paratome web server. The Cologne University Protein Stability Analysis Tool (CUPSAT) web server based mutational approach was used to identify the stable and favorable interface mutations in the lebrikizumab. Only 40 mutations were selected to generate a single mutant library, and their binding affinity for IL-13 was analyzed by using the Z-Dock server. Based on high Z-score, mutants having a better affinity with IL-13 were selected to create a multi-mutant library. The multi-mutant library was again subjected to the Z-Dock server, and their binding affinity was determined. The highest-scoring ten mAb mutants were validated by using PatchDock and ClusPro servers. The best two potential mAb mutants were identified and subjected to molecular dynamics (MD) simulations to ensure its structural stability at the microscopic level. The changes in the different bonds as the effect of mutation were assessed by LigPlot + v2.1. The AllerTOP and ToxinPred web servers were used to analyze the non-allergic and nontoxic nature of the selected mutants. Therefore, these redesigned mAb could be used for potential treatment against IL-13 associated diseased conditions.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vinita Sharma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Priya Chouhan
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Rajan Kumar Pandey
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Rupal Ojha
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | | | - Muthukalingan Krishnan
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, India.,Madurai Kamraj University, Madurai, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
21
|
Synthesized atropine nanoparticles ameliorate airway hyperreactivity and remodeling in a murine model of chronic asthma. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
22
|
Evaluation of Antibody Properties and Clinically Relevant Immunogenicity, Anaphylaxis, and Hypersensitivity Reactions in Two Phase III Trials of Tralokinumab in Severe, Uncontrolled Asthma. Drug Saf 2020; 42:769-784. [PMID: 30649752 PMCID: PMC6520328 DOI: 10.1007/s40264-018-00788-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Introduction Tralokinumab is a monoclonal antibody (mAb) that neutralizes interleukin (IL)-13, a cytokine involved in the pathogenesis of asthma. Objective The objectives of this study were to characterize the potential immunogenic properties of tralokinumab and report data for anti-drug antibodies (ADAs) and hypersensitivity reactions from two phase III clinical trials. Methods The oligosaccharide structure of tralokinumab, Fab-arm exchange, and ADAs were characterized by standard techniques. Hypersensitivity adverse events (AEs) were evaluated in two pivotal clinical trials of tralokinumab in severe, uncontrolled asthma: STRATOS 1 and 2 (NCT02161757 and NCT02194699). Results No galactose-α-1,3-galactose (α-Gal) epitopes were found in the Fab region of tralokinumab and only 4.5% of glycoforms contained α-Gal in the Fc region. Under non-reducing conditions, Fab-arm exchange did not take place with another immunoglobulin (Ig) G4 mAb (mavrilimumab). However, following glutathione reduction, a hybrid antibody with monovalent bioactivity was detected. ADA incidences (titers) were as follows: STRATOS 1—every 2 weeks (Q2 W) 0.8% (26.0), every 4 weeks (Q4 W) 0.5% (26.0), placebo 0.8% (52.0); STRATOS 2—Q2 W 1.2% (39.0), placebo 0.8% (13.0). Participant-reported hypersensitivity AE rates were as follows: STRATOS 1—Q2 W 25.9%, Q4 W 25.0%, placebo 25.5%; STRATOS 2—Q2 W 13.2%, placebo 9.0%. External evaluation for anaphylaxis by Sampson criteria found no tralokinumab-related severe hypersensitivity or anaphylaxis reactions. Conclusion Preclinical assessments suggested a low likelihood of immunogenicity for tralokinumab. In STRATOS 1 and 2, ADA incidence was low, no differences were found between tralokinumab-treated and placebo groups in reporting of hypersensitivity reactions, and there were no Sampson criteria-evaluated anaphylaxis events with tralokinumab treatment. Together, the results suggest that tralokinumab treatment would not increase the risk for severe hypersensitivity or anaphylactic reactions. Electronic supplementary material The online version of this article (10.1007/s40264-018-00788-w) contains supplementary material, which is available to authorized users.
Collapse
|
23
|
Alfaleh MA, Alsaab HO, Mahmoud AB, Alkayyal AA, Jones ML, Mahler SM, Hashem AM. Phage Display Derived Monoclonal Antibodies: From Bench to Bedside. Front Immunol 2020. [PMID: 32983137 DOI: 10.3389/fimmu.2020.01986/bibtex] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage-derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
24
|
Varricchi G, Marone G, Spadaro G, Russo M, Granata F, Genovese A, Marone G. Novel Biological Therapies in Severe Asthma: Targeting the Right Trait. Curr Med Chem 2019; 26:2801-2822. [PMID: 29318959 DOI: 10.2174/0929867325666180110094542] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 11/09/2017] [Accepted: 11/09/2017] [Indexed: 12/23/2022]
Abstract
Asthma is a heterogeneous disease characterized by chronic airway inflammation that results in a wide spectrum of clinical manifestations. Patients with severe asthma represent a substantial share of consumption of healthcare resources and hospitalization. Moreover, these patients are at risk of increased morbidity and mortality. Recently, several phenotypes and endotypes of asthma have been identified. The identification of specific subtypes of asthma is fundamental for optimizing the clinical benefit of novel treatments. Although in most patients the disease can be controlled by some combination of pharmacologic agents, in some 5-10% of patients the disease remains uncontrolled. Several monoclonal antibodies (mAbs) targeting pathogenetic molecules (e.g., IgE, IL-5, IL- 5Rα, IL-4, IL-13, TSLP) are currently available or under development for the treatment of different forms of severe type 2 asthma. The identification of diagnostic and predictive biomarkers (e.g., IgE, blood eosinophil count, FeNO, periostin, etc.) has revolutioned the field of targeted therapy in severe asthma. Monoclonal antibodies targeting Th2-driven inflammation are generally safe in adult patients with moderate-to-severe asthma. The long-term safety of these biologics is a relevant issue that should be addressed. Unfortunately, little is known about non-type 2 asthma. Further studies are needed to identify biomarkers to guide targeted therapies of different forms of non-type 2 asthma.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy
| | - Giancarlo Marone
- Department of Public Health, University of Naples Federico II, Naples, Italy.,Monaldi Hospital, Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy
| | - Michele Russo
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, Naples, Italy
| | - Francescopaolo Granata
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy
| | - Arturo Genovese
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council (CNR), Naples, Italy
| |
Collapse
|
25
|
Abstract
Atopic dermatitis (AD) is a chronic, inflammatory skin disease characterized by pruritus, inflammatory erythematous skin lesions, and skin-barrier defect. Current mainstay treatments of emollients, steroids, calcineurin inhibitors, and immunosuppressants have limited efficacy and potentially serious side effects. Recent advances and understanding of the pathogenesis of AD have resulted in new therapies that target specific pathways with increased efficacy and the potential for less systemic side effects. New FDA-approved therapies for AD are crisaborole and dupilumab. The JAK-STAT inhibitors (baricitinib, upadacitinib, PF-04965842, ASN002, tofacitinib, ruxolitinib, and delgocitinib) have the most promising results of the emerging therapies. Other drugs with potential include the aryl hydrocarbon receptor modulating agent tapinarof, the IL-4/IL-13 antagonists lebrikizumab and tralokinumab, and the IL-31Rα antagonist nemolizumab. In this review, new and emerging AD therapies will be discussed along with their mechanisms of action and their potential based on clinical study data.
Collapse
Affiliation(s)
- Henry L Nguyen
- Department of Dermatology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55902, USA
| | - Katelyn R Anderson
- Department of Dermatology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55902, USA
| | - Megha M Tollefson
- Department of Dermatology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55902, USA.
| |
Collapse
|
26
|
Gottlow M, Svensson DJ, Lipkovich I, Huhn M, Bowen K, Wessman P, Colice G. Application of structured statistical analyses to identify a biomarker predictive of enhanced tralokinumab efficacy in phase III clinical trials for severe, uncontrolled asthma. BMC Pulm Med 2019; 19:129. [PMID: 31315668 PMCID: PMC6637533 DOI: 10.1186/s12890-019-0889-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 07/02/2019] [Indexed: 11/10/2022] Open
Abstract
Background Tralokinumab is an anti–interleukin (IL)-13 monoclonal antibody investigated for the treatment of severe, uncontrolled asthma in two Phase III clinical trials, STRATOS 1 and 2. The STRATOS 1 biomarker analysis plan was developed to identify biomarker(s) indicative of IL-13 activation likely to predict tralokinumab efficacy and define a population in which there was an enhanced treatment effect; this defined population was then tested in STRATOS 2. Methods The biomarkers considered were blood eosinophil counts, fractional exhaled nitric oxide (FeNO), serum dipeptidyl peptidase-4, serum periostin and total serum immunoglobulin E. Tralokinumab efficacy was measured as the reduction in annualised asthma exacerbation rate (AAER) compared with placebo (primary endpoint measure of STRATOS 1 and 2). The biomarker analysis plan included negative binomial and generalised additive models, and the Subgroup Identification based on Differential Effect Search (SIDES) algorithm, supported by robustness and sensitivity checks. Effects on the key secondary endpoints of STRATOS 1 and 2, which included changes from baseline in standard measures of asthma outcomes, were also investigated. Prior to the STRATOS 1 read-out, numerous simulations of the methodology were performed with hypothetical data. Results FeNO and periostin were identified as the only biomarkers potentially predictive of treatment effect, with cut-offs chosen by the SIDES algorithm of > 32.3 ppb and > 27.4 ng/ml, respectively. The FeNO > 32.3 ppb subgroup was associated with greater AAER reductions and improvements in key secondary endpoints compared with the periostin > 27.4 ng/ml subgroup. Upon further evaluation of AAER reductions at different FeNO cut-offs, ≥37 ppb was chosen as the best cut-off for predicting tralokinumab efficacy. Discussion A rigorous statistical approach incorporating multiple methods was used to investigate the predictive properties of five potential biomarkers and to identify a participant subgroup that demonstrated an enhanced tralokinumab treatment effect. Using STRATOS 1 data, our analyses identified FeNO at a cut-off of ≥37 ppb as the best assessed biomarker for predicting enhanced treatment effect to be tested in STRATOS 2. Our findings were inconclusive, which reflects the complexity of subgroup identification in the severe asthma population. Trial registration STRATOS 1 and 2 are registered on ClinicalTrials.gov (NCT02161757 registered on June 12, 2014, and NCT02194699 registered on July 18, 2014). Electronic supplementary material The online version of this article (10.1186/s12890-019-0889-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mattis Gottlow
- Biometrics and Information Sciences, AstraZeneca, Pepparedsleden 1, SE-431 83, Mölndal, Sweden
| | - David J Svensson
- Biometrics and Information Sciences, AstraZeneca, Pepparedsleden 1, SE-431 83, Mölndal, Sweden
| | - Ilya Lipkovich
- IQVIA, 4820 Emperor Blvd, Durham, NC, 27703, USA.,Present Address: Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, 46285, USA
| | - Monika Huhn
- Biometrics and Information Sciences, AstraZeneca, Pepparedsleden 1, SE-431 83, Mölndal, Sweden
| | - Karin Bowen
- Biometrics and Information Sciences, AstraZeneca, One MedImmune Way, Gaithersburg, MD, 20878, USA
| | - Peter Wessman
- Biometrics and Information Sciences, AstraZeneca, Pepparedsleden 1, SE-431 83, Mölndal, Sweden
| | - Gene Colice
- Global Medicines Development, AstraZeneca, One MedImmune Way, Gaithersburg, MD, 20878, USA.
| |
Collapse
|
27
|
Zhang Y, Cheng J, Li Y, He R, Pan P, Su X, Hu C. The Safety and Efficacy of Anti-IL-13 Treatment with Tralokinumab (CAT-354) in Moderate to Severe Asthma: A Systematic Review and Meta-Analysis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2019; 7:2661-2671.e3. [PMID: 31152798 DOI: 10.1016/j.jaip.2019.05.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/14/2019] [Accepted: 05/11/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Several clinical studies have evaluated the use of tralokinumab (CAT-354) administration in patients with moderate to severe asthma; no consensus on tralokinumab efficacy and safety was reached. Thus, further analysis is required on the efficacy and safety of tralokinumab as an asthma biologic. OBJECTIVE To assess the efficacy and safety of subcutaneous injection of tralokinumab in patients with moderate to severe asthma. METHODS Clinical trials were identified from MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov from their inception to November 4, 2018. Only randomized clinical trials (RCTs) with tralokinumab versus placebo treatment in patients with moderate to severe asthma were evaluated. Efficacy and safety outcomes were extracted, and a meta-analysis was performed using a random-effects model. The Cochrane Collaboration's risk-of-bias assessment tool was used to assess the risk of bias. RESULTS Five studies describing 6 RCTs (including 2928 adults with moderate to severe asthma) were pooled and analyzed in this study. Absolute FEV1 was statistically improved in patients receiving tralokinumab at 300 mg every 2 weeks (mean difference [MD], 0.14 L; 95% CI, 0.08-0.21) and 600 mg every 2 weeks (MD, 0.20 L; 95% CI, 0.01-0.39), as well as FEV1% changes (MD, 5.82%, 95% CI, 3.58-8.06, and MD, 11.8%, 95% CI, 0.79-22.81, respectively). Also, absolute forced vital capacity volume changes (MD, 0.11 L; 95% CI, 0.01-0.21) and percentage changes (MD, 4.44%; 95% CI, 0.84-8.04) improved in tralokinumab at 300 mg every 2 weeks. Asthma Quality of Life Questionnaire scores were not significantly different, and absolute Asthma Control Questionnaire 6 scores were statistically improved but did not reach the clinically meaningful difference. Tralokinumab treatment did not decrease annualized asthma exacerbation rate in unselected patients with moderate to severe asthma, but it was associated with improved annualized asthma exacerbation rate in patients with severe asthma with high fractional exhaled nitric oxide levels (rate ratio, 0.72; 95% CI, 0.53-0.97). Tralokinumab was not associated with an increased incidence of serious adverse events, but it did show an increase in mild injection-site reactions (odds ratio, 5.92; 95% CI, 1.61-21.76). CONCLUSION This pooled analysis of 6 RCTs suggested that tralokinumab was well tolerated and it modestly improved FEV1 and forced vital capacity in patients with moderate to severe asthma. It did not render clinically important improvements in asthma-related quality of life, and nor did it reduce asthma exacerbations.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Respiratory Medicine, Xiangya Hospital (Key Site of National Clinical Research Center for Respiratory Disease), Central South University, Changsha, Hunan, China
| | - Jun Cheng
- Department of Gastrointestinal Surgery and Breast and Thyroid Surgery, Changsha Hospital of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Yuanyuan Li
- Department of Respiratory Medicine, Xiangya Hospital (Key Site of National Clinical Research Center for Respiratory Disease), Central South University, Changsha, Hunan, China
| | - Ruoxi He
- Department of Respiratory Medicine, Xiangya Hospital (Key Site of National Clinical Research Center for Respiratory Disease), Central South University, Changsha, Hunan, China
| | - Pinhua Pan
- Department of Respiratory Medicine, Xiangya Hospital (Key Site of National Clinical Research Center for Respiratory Disease), Central South University, Changsha, Hunan, China
| | - Xiaoli Su
- Department of Respiratory Medicine, Xiangya Hospital (Key Site of National Clinical Research Center for Respiratory Disease), Central South University, Changsha, Hunan, China
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Hospital (Key Site of National Clinical Research Center for Respiratory Disease), Central South University, Changsha, Hunan, China.
| |
Collapse
|
28
|
Del Rosso JQ. MONOCLONAL ANTIBODY THERAPIES for Atopic Dermatitis: Where Are We Now in the Spectrum of Disease Management? THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2019; 12:39-41. [PMID: 30881583 PMCID: PMC6415707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Atopic dermatitis (AD) is a chronic disorder that requires thorough patient education and a therapeutic management strategy designed to control flares, decrease recurrences, and reduce pruritus. In cases that cannot be controlled by proper skin care and barrier repair, topical therapy, and avoidance of triggers, systemic therapy is often required to control flares and maintain remission. It is important for clinicians to avoid becoming overly dependent on the intermittent use of systemic corticosteroid therapy to control flares, without incorporating other treatment options that might more optimally control AD over time. This article provides an overview of systemic therapies, including conventional oral therapy options and injectable biologic agents, that modulate the immune dysregulation in AD. Major emphasis is placed on the monoclonal antibodies currently available (e.g., dupilumab) for the treatment of AD, as well as those in latter stages of development, with a focus on agents targeting IL-4and/or IL-13.
Collapse
Affiliation(s)
- James Q Del Rosso
- Dr. Del Rosso is Research Director of JDR Dermatology Research in Las Vegas, Nevada; is with Thomas Dermatology in Las Vegas, Nevada; and is Adjunct Clinical Professor (Dermatology) with Touro University Nevada in Henderson, Nevada
| |
Collapse
|
29
|
Busse WW, Brusselle GG, Korn S, Kuna P, Magnan A, Cohen D, Bowen K, Piechowiak T, Wang MM, Colice G. Tralokinumab did not demonstrate oral corticosteroid-sparing effects in severe asthma. Eur Respir J 2019; 53:13993003.00948-2018. [PMID: 30442714 DOI: 10.1183/13993003.00948-2018] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/30/2018] [Indexed: 12/15/2022]
Abstract
Long-term oral corticosteroid (OCS) use in patients with severe asthma is associated with significant adverse effects.This 40-week, randomised, double-blind trial evaluated the OCS-sparing potential of tralokinumab in patients with severe, uncontrolled asthma requiring maintenance OCS treatment plus inhaled corticosteroids/long-acting β2-agonists. Overall, 140 patients were randomised to tralokinumab 300 mg or placebo (n=70 in each group) administered subcutaneously every 2 weeks. The primary end-point was percentage change from baseline in average OCS dose at week 40, while maintaining asthma control. Secondary end-points included proportion of patients with a prescribed maintenance OCS dose of ≤5 mg, those with a ≥50% reduction in prescribed maintenance OCS dose and asthma exacerbation rate. Safety was also assessed.At week 40, the percentage reduction from baseline in the final daily average OCS dose was not significantly different between tralokinumab and placebo (37.62% versus 29.85%; p=0.271). There were no significant between-treatment differences for any secondary end-point. Overall, reporting of adverse events and serious adverse events were similar for the tralokinumab and placebo groups. Although a greater proportion of tralokinumab-treated patients reported upper respiratory tract infections (35.7% versus 14.3%), there were no reported cases of pneumonia.Overall, tralokinumab did not demonstrate an OCS-sparing effect in patients with severe asthma.
Collapse
Affiliation(s)
- William W Busse
- Dept of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Guy G Brusselle
- Dept of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Stephanie Korn
- Pulmonary Dept, Mainz University Hospital, Mainz, Germany
| | - Piotr Kuna
- Dept of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Lodz, Poland
| | - Antoine Magnan
- Institut du Thorax, INSERM CNRS, Université de Nantes, CHU de Nantes, Nantes, France
| | | | | | | | | | | |
Collapse
|
30
|
Sato S, Yanagihara T, Kolb MRJ. Therapeutic targets and early stage clinical trials for pulmonary fibrosis. Expert Opin Investig Drugs 2018; 28:19-28. [DOI: 10.1080/13543784.2019.1554054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Seidai Sato
- Firestone Institute for Respiratory Health, Departments of Medicine, McMaster University, Hamilton,
Ontario, Canada
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University,
Tokushima, Japan
| | - Toyoshi Yanagihara
- Firestone Institute for Respiratory Health, Departments of Medicine, McMaster University, Hamilton,
Ontario, Canada
| | - Martin R. J. Kolb
- Firestone Institute for Respiratory Health, Departments of Medicine, McMaster University, Hamilton,
Ontario, Canada
| |
Collapse
|
31
|
Potential Risks Related to Modulating Interleukin-13 and Interleukin-4 Signalling: A Systematic Review. Drug Saf 2018; 41:489-509. [PMID: 29411337 PMCID: PMC5938313 DOI: 10.1007/s40264-017-0636-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction Interleukin-13 and interleukin-4 are type-II cytokines signalling through the shared type II interleukin-4 receptor. As a result of their structural similarity, interleukin-13 and interleukin-4 have overlapping functions in the mediation of type-II-driven diseases and are, therefore, promising targets of biologic drugs currently in development for the treatment of such diseases, including asthma and atopic dermatitis. Objective This systematic review was conducted to assess preclinical evidence of potential safety concerns related to blockade of interleukin-13 alone or interleukin-13 and interleukin-4 in combination. Methods We specifically examined risks related to infection, malignancy and the cardiovascular system. We systematically searched the BIOSIS, MEDLINE and EMBASE databases to identify preclinical studies published between January 2006 and October 2016 that addressed the effects of interleukin-13/interleukin-4 blockade and modulation on the risk of infection, malignancy and cardiovascular events. To provide a clinical context, we also performed a search for clinical trials targeting the interleukin-13/interleukin-4 pathways. Relevant data from preclinical and clinical trials were abstracted and presented descriptively. Results Aside from expected evidence that inhibition of interleukin-13 and interleukin-4 impaired host responses to helminth infections, we did not identify other preclinical evidence suggesting safety risks relating to infection, malignancy or cardiovascular events. We found no evidence in clinical trials suggesting serious safety concerns, i.e. increased risk for infections, malignancy or cardiovascular events from therapeutic modulation of the interleukin-13 pathway alone or the combined interleukin-13/interleukin-4 pathways. Conclusions Although our findings are reassuring, long-term safety assessments of biologics that target the interleukin-13/interleukin-4 pathways currently in clinical development are needed. Electronic supplementary material The online version of this article (10.1007/s40264-017-0636-9) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Emson C, Pham TH, Manetz S, Newbold P. Periostin and Dipeptidyl Peptidase-4: Potential Biomarkers of Interleukin 13 Pathway Activation in Asthma and Allergy. Immunol Allergy Clin North Am 2018; 38:611-628. [PMID: 30342583 DOI: 10.1016/j.iac.2018.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Periostin and dipeptidyl peptidase-4 (DPP-4) are proteins induced by type 2 cytokines interleukin (IL)-4 and IL-13 and show increased expression in asthma and diseases with type 2 inflammation, including atopic dermatitis and chronic rhinosinusitis. Both proteins can also be induced by other stimuli, such as profibrotic factors, which may confound their specificity as biomarkers of IL-13 pathway activation and type 2-driven disease. DPP-4 is important in glucose metabolism; therefore, serum concentrations may be confounded by the presence of concomitant metabolic disease. This review evaluates the potential of these biomarkers for anti-IL-13-directed therapy in asthma and diseases with type 2 inflammation.
Collapse
Affiliation(s)
- Claire Emson
- MedImmune, One MedImmune Way, Gaithersburg, MD 20878, USA.
| | | | - Scott Manetz
- MedImmune, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Paul Newbold
- MedImmune, One MedImmune Way, Gaithersburg, MD 20878, USA
| |
Collapse
|
33
|
Wollenberg A, Howell MD, Guttman-Yassky E, Silverberg JI, Kell C, Ranade K, Moate R, van der Merwe R. Treatment of atopic dermatitis with tralokinumab, an anti-IL-13 mAb. J Allergy Clin Immunol 2018; 143:135-141. [PMID: 29906525 DOI: 10.1016/j.jaci.2018.05.029] [Citation(s) in RCA: 279] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/23/2018] [Accepted: 05/22/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND IL-13 has an important role in atopic dermatitis (AD) pathogenesis. Tralokinumab is a fully human mAb that potently and specifically neutralizes IL-13. OBJECTIVE We sought to evaluate the efficacy and safety of tralokinumab in adults with moderate-to-severe AD. METHODS In this phase 2b study (NCT02347176), 204 adults were randomized 1:1:1:1 to receive 45, 150, or 300 mg of subcutaneous tralokinumab, or placebo, every 2 weeks for 12 weeks with concomitant topical glucocorticoids. Coprimary end points were change from baseline in Eczema Area Severity Index score and percentage of participants with an Investigator's Global Assessment response (0/1 score and reduction of ≥2 grades from baseline) at week 12. RESULTS At week 12, 300 mg of tralokinumab significantly improved change from baseline in Eczema Area Severity Index score versus placebo (adjusted mean difference, -4.94; 95% CI, -8.76 to -1.13; P = .01), and a greater percentage of participants achieved an Investigator's Global Assessment response (26.7% vs 11.8%). Greater responses were found in participants with greater concentrations of biomarkers of increased IL-13 activity. Participants treated with 300 mg of tralokinumab demonstrated improvements in SCORAD, Dermatology Life Quality Index, and pruritus numeric rating scale (7-day mean) scores versus placebo. Upper respiratory tract infection was the most frequent treatment-emergent adverse event reported as related to study drug in the placebo (3.9%) and pooled tralokinumab (3.9%) groups. CONCLUSIONS Tralokinumab treatment was associated with early and sustained improvements in AD symptoms and an acceptable safety and tolerability profile, thereby providing evidence for targeting IL-13 in patients with AD.
Collapse
Affiliation(s)
- Andreas Wollenberg
- Department of Dermatology and Allergy, Ludwig Maximillian University, Munich, Germany.
| | | | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai Medical Center Medicine, New York, NY
| | - Jonathan I Silverberg
- Department of Dermatology, Preventive Medicine and Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | | | | | - Rachel Moate
- Clinical Statistics and Data, MedImmune, Cambridge, United Kingdom
| | | |
Collapse
|
34
|
Panettieri RA, Wang M, Braddock M, Bowen K, Colice G. Tralokinumab for the treatment of severe, uncontrolled asthma: the ATMOSPHERE clinical development program. Immunotherapy 2018. [PMID: 29536781 DOI: 10.2217/imt-2017-0191] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tralokinumab, a fully human IgG4 monoclonal antibody, specifically neutralizes IL-13. The ATMOSPHERE clinical development program comprised four randomized, placebo-controlled clinical trials and an open-label study that aimed to assess the efficacy and safety of tralokinumab for the treatment of severe, uncontrolled asthma. The two pivotal trials (STRATOS 1 and STRATOS 2; NCT02161757 and NCT02194699) evaluated the efficacy and safety of tralokinumab, with STRATOS 1 identifying a subgroup most likely to demonstrate enhanced response to treatment. Further trials have assessed the ability of tralokinumab to reduce oral corticosteroid use (TROPOS; NCT02281357) and determined its mechanistic effects (MESOS; NCT02449473). An open-label study in Japanese individuals (NCT02902809) assessed the long-term safety and tolerability of tralokinumab in this population.
Collapse
Affiliation(s)
- Reynold A Panettieri
- Rutgers Institute for Translational Medicine & Science, Rutgers, The State University of New Jersey, NJ 08901, USA
| | - Millie Wang
- Global Medicines Development, AstraZeneca, Cambridge, UK
| | | | - Karin Bowen
- Global Medicines Development, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Gene Colice
- Global Medicines Development, AstraZeneca, Gaithersburg, MD 20878, USA
| |
Collapse
|
35
|
Parker JM, Glaspole IN, Lancaster LH, Haddad TJ, She D, Roseti SL, Fiening JP, Grant EP, Kell CM, Flaherty KR. A Phase 2 Randomized Controlled Study of Tralokinumab in Subjects with Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2018; 197:94-103. [DOI: 10.1164/rccm.201704-0784oc] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
| | - Ian N. Glaspole
- Department of General Respiratory Medicine and Lung Transplantation, The Alfred Hospital, Melbourne, Australia
- Department of Medicine, Monash University, Melbourne, Australia
| | - Lisa H. Lancaster
- Allergy, Pulmonary, and Critical Care, Vanderbilt University, Nashville, Tennessee
| | - Tarik J. Haddad
- Pulmonary and Critical Care Medicine, Tampa General Hospital, Tampa, Florida
| | - Dewei She
- Clinical Biostatistics and Data Management
| | | | | | - Ethan P. Grant
- Translational Medicine, MedImmune, Gaithersburg, Maryland
| | - Chris M. Kell
- Product Development, MedImmune, Cambridge, United Kingdom; and
| | - Kevin R. Flaherty
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
36
|
Tralokinumab pharmacokinetics and tolerability when administered by different subcutaneous injection methods and rates
. Int J Clin Pharmacol Ther 2017; 55:606-620. [PMID: 28590244 PMCID: PMC5480250 DOI: 10.5414/cp203023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2017] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Tralokinumab, administered as two 1-mL subcutaneous injections every 2 weeks, at the target dose 300 mg, has been shown to improve lung function in patients with asthma. This study evaluated the pharmacokinetic (PK) and tolerability profile of tralokinumab 300 mg when administered by different rates of subcutaneous injection, as part of a pilot investigation of new injection regimens. METHODS This phase I study randomized 60 healthy adults to receive 300 mg tralokinumab, as two 1-mL subcutaneous injections, each delivered over 10 seconds, or one 2-mL injection delivered over 10 seconds (12 mL/min), 1 minute (2 mL/min), or 12 minutes (0.167 mL/min). RESULTS No differences in the PK profile of tralokinumab were observed between cohorts. Immediately following injection, injection-site pain intensity (mean (SD)) was lowest following 0.167 mL/min injection (5.1 mm (8.0) via visual analog scale (VAS)) and greatest following 12 mL/min injection (41 mm (27.7) via VAS); with mean injection-site pruritus intensity low for all participants. Two types of local injection-site reactions were observed: erythema (58.3%) and hematoma/bleeding (18.3%). All treatment-emergent adverse events were mild. CONCLUSIONS Tralokinumab 300 mg is well tolerated, with comparable PK, when administered by a single 2-mL injection at different rates of subcutaneous injection vs. two 1-mL injections.
.
Collapse
|
37
|
Baverel PG, White N, Vicini P, Karlsson MO, Agoram B. Dose-Exposure-Response Relationship of the Investigational Anti-Interleukin-13 Monoclonal Antibody Tralokinumab in Patients With Severe, Uncontrolled Asthma. Clin Pharmacol Ther 2017; 103:826-835. [PMID: 28758192 DOI: 10.1002/cpt.803] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 01/09/2023]
Abstract
Interleukin (IL)-13 is involved in the pathogenesis of some types of asthma. Tralokinumab is a human immunoglobulin G4 monoclonal antibody that specifically binds to IL-13. Two placebo-controlled phase II studies (phase IIa, NCT00873860 and phase IIb, NCT01402986) have been conducted in which tralokinumab was administered subcutaneously. This investigation aimed to characterize tralokinumab's dose-exposure-response (forced expiratory volume in 1 s (FEV1 )) relationship in patients with asthma and to predict the most appropriate dose for phase III. An integrated population pharmacokinetic-pharmacodynamic (PK/PD) modeling analysis was required for phase III dose selection, due to differing phase II patient populations, designs, and regimens. Analysis of combined datasets enabled the identification of tralokinumab's dose-exposure-FEV1 response relationship in patients with asthma. Near-maximal FEV1 increase was predicted at a dose of 300 mg SC once every 2 weeks (Q2W). This dose was chosen for tralokinumab in the phase III clinical development program for treatment of severe, uncontrolled asthma.
Collapse
Affiliation(s)
- Paul G Baverel
- Clinical Pharmacology, Drug Metabolism and Pharmacokinetics, MedImmune, Cambridge, UK
| | - Nicholas White
- Clinical Pharmacology, Drug Metabolism and Pharmacokinetics, MedImmune, Cambridge, UK
| | - Paolo Vicini
- Clinical Pharmacology, Drug Metabolism and Pharmacokinetics, MedImmune, Cambridge, UK
| | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Balaji Agoram
- Clinical Pharmacology, Drug Metabolism and Pharmacokinetics, MedImmune, Cambridge, UK.,Clinical Pharmacology and DMPK, FortySeven Inc, Menlo Park, California, USA
| |
Collapse
|
38
|
Karelina T, Voronova V, Demin O, Colice G, Agoram BM. A Mathematical Modeling Approach to Understanding the Effect of Anti-Interleukin Therapy on Eosinophils. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 5:608-616. [PMID: 27885827 PMCID: PMC5192997 DOI: 10.1002/psp4.12129] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/25/2016] [Accepted: 08/26/2016] [Indexed: 11/25/2022]
Abstract
Emerging T‐helper type 2 (Th2) cytokine‐based asthma therapies, such as tralokinumab, lebrikizumab (anti‐interleukin (IL)‐13), and mepolizumab (anti‐IL‐5), have shown differences in their blood eosinophil (EOS) response. To better understand these effects, we developed a mathematical model of EOS dynamics. For the anti‐IL‐13 therapies, lebrikizumab and tralokinumab, the model predicted an increase of 30% and 10% in total and activated EOS in the blood, respectively, and a decrease in the total and activated EOS in the airways. The model predicted a rapid decrease in total and activated EOS levels in blood and airways for the anti‐IL‐5 therapy mepolizumab. All model‐based predictions were consistent with published clinical observations. The modeling approach provided insights into EOS response after treatment with Th2‐targeted therapies, and supports the hypothesis that an increase in blood EOS after anti‐IL‐13 therapy is part of the pharmacological action of these therapies.
Collapse
Affiliation(s)
- T Karelina
- Institute for Systems Biology, Moscow, Russia
| | - V Voronova
- Institute for Systems Biology, Moscow, Russia
| | - O Demin
- Institute for Systems Biology, Moscow, Russia
| | - G Colice
- Astrazeneca, Global Medicines Development, Gaithersburg, Maryland, USA
| | - B M Agoram
- MedImmune LLC, Clinical Pharmacology, Drug Metabolism, and Pharmacokinetics, Mountain View, California, USA
| |
Collapse
|
39
|
Dahlmann F, Sewald K. Use of nonhuman primates in obstructive lung disease research - is it required? Primate Biol 2017; 4:131-142. [PMID: 32110701 PMCID: PMC7041527 DOI: 10.5194/pb-4-131-2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/05/2017] [Indexed: 12/20/2022] Open
Abstract
In times of increasing costs for health insurances, obstructive lung
diseases are a burden for both the patients and the economy. Pulmonary symptoms
of asthma and chronic obstructive pulmonary disease (COPD) are similar;
nevertheless, the diseases differ in pathophysiology and therapeutic
approaches. Novel therapeutics are continuously developed, and nonhuman
primates (NHPs) provide valuable models for investigating novel biologicals
regarding efficacy and safety. This review discusses the role of nonhuman primate models for drug
development in asthma and COPD and investigates whether alternative methods
are able to prevent animal experiments.
Collapse
Affiliation(s)
- Franziska Dahlmann
- German Primate Center GmbH, Infection Pathology Unit, Kellnerweg 4, 37077 Göttingen, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine, Preclinical Pharmacology and Immunology, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Nikolai-Fuchs-Straße 1, 30625 Hanover, Germany
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine, Preclinical Pharmacology and Immunology, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Nikolai-Fuchs-Straße 1, 30625 Hanover, Germany
| |
Collapse
|
40
|
Popovic B, Breed J, Rees DG, Gardener MJ, Vinall LMK, Kemp B, Spooner J, Keen J, Minter R, Uddin F, Colice G, Wilkinson T, Vaughan T, May RD. Structural Characterisation Reveals Mechanism of IL-13-Neutralising Monoclonal Antibody Tralokinumab as Inhibition of Binding to IL-13Rα1 and IL-13Rα2. J Mol Biol 2016; 429:208-219. [PMID: 27956146 DOI: 10.1016/j.jmb.2016.12.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/02/2016] [Accepted: 12/05/2016] [Indexed: 12/12/2022]
Abstract
Interleukin (IL)-13 is a pleiotropic T helper type 2 cytokine frequently associated with asthma and atopic dermatitis. IL-13-mediated signalling is initiated by binding to IL-13Rα1, which then recruits IL-4Rα to form a heterodimeric receptor complex. IL-13 also binds to IL-13Rα2, considered as either a decoy or a key mediator of fibrosis. IL-13-neutralising antibodies act by preventing IL-13 binding to IL-13Rα1, IL-4Rα and/or IL-13Rα2. Tralokinumab (CAT-354) is an IL-13-neutralising human IgG4 monoclonal antibody that has shown clinical benefit in patients with asthma. To decipher how tralokinumab inhibits the effects of IL-13, we determined the structure of tralokinumab Fab in complex with human IL-13 to 2 Å resolution. The structure analysis reveals that tralokinumab prevents IL-13 from binding to both IL-13Rα1 and IL-13Rα2. This is supported by biochemical ligand-receptor interaction assay data. The tralokinumab epitope is mainly composed of residues in helices D and A of IL-13. It is mostly light chain complementarity-determining regions that are driving paratope interactions; the variable light complementarity-determining region 2 plays a key role by providing residue contacts for a network of hydrogen bonds and a salt bridge in the core of binding. The key residues within the paratope contributing to binding were identified as Asp50, Asp51, Ser30 and Lys31. This study demonstrates that tralokinumab prevents the IL-13 pharmacodynamic effect by binding to IL-13 helices A and D, thus preventing IL-13 from interacting with IL-13Rα1 and IL-13Rα2.
Collapse
Affiliation(s)
- B Popovic
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK.
| | - J Breed
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge Science Park, Milton Road, Cambridge CB4 0WG, UK
| | - D G Rees
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - M J Gardener
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - L M K Vinall
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - B Kemp
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - J Spooner
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - J Keen
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - R Minter
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - F Uddin
- Department of Biopharmaceutical Development, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - G Colice
- Inflammation, Neuroscience, Respiratory, Global Medicines Development, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - T Wilkinson
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - T Vaughan
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - R D May
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| |
Collapse
|
41
|
Hagmann BR, Odermatt A, Kaufmann T, Dahinden CA, Fux M. Balance between IL-3 and type Iinterferons and their interrelationship with FasL dictates lifespan and effector functions of human basophils. Clin Exp Allergy 2016; 47:71-84. [PMID: 27910206 DOI: 10.1111/cea.12850] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 09/13/2016] [Accepted: 10/10/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND In contrast to eosinophils and neutrophils, the regulation of the lifespan of human basophils is poorly defined, with the exception of the potent anti-apoptotic effect of IL-3 that also promotes pro-inflammatory effector functions and phenotypic changes. Type I IFNs (IFN-α, IFN-β), which are well known for their anti-viral activities, have the capacity to inhibit allergic inflammation. OBJECTIVE To elucidate whether type I IFNs have the potential to abrogate the lifespan and/or effector functions of human basophils. METHODS We cultured human basophils, and for comparison, eosinophils and neutrophils, with IL-3, interferons, FasL and TRAIL, alone or in combination, and studied cell survival, effector functions and signalling pathways involved. RESULTS Despite an identical pattern of early signalling in basophils, eosinophils and neutrophils in response to different types of interferons, only basophils displayed enhanced apoptosis after type I IFN treatment. IFN-γ prolonged survival of eosinophils but did not affect the lifespan of basophils. IFN-α-mediated apoptosis required STAT1-STAT2 heterodimers and the contribution of constitutive p38 MAPK activity. Whereas the death ligands FasL and TRAIL-induced apoptosis in basophils per se, IFN-α-mediated apoptosis did neither involve autocrine TRAIL signalling nor did it sensitize basophils to FasL-induced apoptosis. However, IFN-α and FasL displayed an additive effect in killing basophils. Interestingly, IL-3, which protected basophils from IFN-α-, TRAIL- or FasL-mediated apoptosis, did not completely block the additive effect of combined IFN-α and FasL treatment. Moreover, we demonstrate that IFN-α suppressed IL-3-induced release of IL-8 and IL-13. In contrast to IFN-α-mediated apoptosis, these inhibitory effects of IFN-α were not dependent on p38 MAPK signalling. CONCLUSIONS AND CLINICAL RELEVANCE Our study defines the unique and granulocyte-type-specific inhibitory and pro-apoptotic function of type I IFNs and their cooperation with death ligands in human blood basophils, which may be relevant for the anti-allergic properties of type I IFNs.
Collapse
Affiliation(s)
- B R Hagmann
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,University Institute of Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - A Odermatt
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,University Institute of Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - T Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - C A Dahinden
- University Institute of Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - M Fux
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,University Institute of Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
42
|
Nixon J, Newbold P, Mustelin T, Anderson GP, Kolbeck R. Monoclonal antibody therapy for the treatment of asthma and chronic obstructive pulmonary disease with eosinophilic inflammation. Pharmacol Ther 2016; 169:57-77. [PMID: 27773786 DOI: 10.1016/j.pharmthera.2016.10.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Eosinophils have been linked with asthma for more than a century, but their role has been unclear. This review discusses the roles of eosinophils in asthma and chronic obstructive pulmonary disease (COPD) and describes therapeutic antibodies that affect eosinophilia. The aims of pharmacologic treatments for pulmonary conditions are to reduce symptoms, slow decline or improve lung function, and reduce the frequency and severity of exacerbations. Inhaled corticosteroids (ICS) are important in managing symptoms and exacerbations in asthma and COPD. However, control with these agents is often suboptimal, especially for patients with severe disease. Recently, new biologics that target eosinophilic inflammation, used as adjunctive therapy to corticosteroids, have proven beneficial and support a pivotal role for eosinophils in the pathology of asthma. Nucala® (mepolizumab; anti-interleukin [IL]-5) and Cinquair® (reslizumab; anti-IL-5), the second and third biologics approved, respectively, for the treatment of asthma, exemplifies these new treatment options. Emerging evidence suggests that eosinophils may contribute to exacerbations and possibly to lung function decline for a subset of patients with COPD. Here we describe the pharmacology of therapeutic antibodies inhibiting IL-5 or targeting the IL-5 receptor, as well as other cytokines contributing to eosinophilic inflammation. We discuss their roles as adjuncts to conventional therapeutic approaches, especially ICS therapy, when disease is suboptimally controlled. These agents have achieved a place in the therapeutic armamentarium for asthma and COPD and will deepen our understanding of the pathogenic role of eosinophils.
Collapse
Affiliation(s)
| | | | | | - Gary P Anderson
- Lung Health Research Centre, University of Melbourne, Melbourne, Victoria, Australia
| | | |
Collapse
|
43
|
Walsh GM. Biologics targeting IL-5, IL-4 or IL-13 for the treatment of asthma - an update. Expert Rev Clin Immunol 2016; 13:143-149. [PMID: 27459348 DOI: 10.1080/1744666x.2016.1216316] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION The development of monoclonal antibody-based biologics targeted at inhibition of the Th2 cytokines IL-4, IL-5 and IL-13 represent potentially effective treatments for patients with the glucocorticoid refractory eosinophilic asthma phenotype. Areas covered: Asthma exhibits marked heterogeneity both clinically and at the molecular phenotypic level, requiring specifically targeted treatments to block the key pathways of the disease. It is becoming apparent that significant clinical effects with anti-cytokine-based biologic therapies are more likely in carefully selected patient populations that take asthma phenotypes into account. The development of reproducible and straightforward discriminatory biomarkers may aid identification of those patients most likely to benefit from treatment with these expensive interventions. This narrative review is based on English-language original articles in PubMed or Med-Line that reported significant clinical findings published in the last two years on the evidence demonstrating the effectiveness or otherwise of the targeting of IL-4, IL-5, or IL-13 in carefully selected patients with severe refractory asthma. Expert commentary: The use of a baseline peripheral blood eosinophilia as a simple reproducible biomarker to identify patients with particular sub-phenotypes of asthma to guide the effective use of biologic therapy represents a significant step forward.
Collapse
Affiliation(s)
- Garry M Walsh
- a School of Medicine and Dentistry , University of Aberdeen , Scotland , UK
| |
Collapse
|
44
|
May RD, Fung M. Strategies targeting the IL-4/IL-13 axes in disease. Cytokine 2016; 75:89-116. [PMID: 26255210 DOI: 10.1016/j.cyto.2015.05.018] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/15/2015] [Indexed: 02/07/2023]
Abstract
IL-4 and IL-13 are pleiotropic Th2 cytokines produced by a wide variety of different cell types and responsible for a broad range of biology and functions. Physiologically, Th2 cytokines are known to mediate host defense against parasites but they can also trigger disease if their activities are dysregulated. In this review we discuss the rationale for targeting the IL-4/IL-13 axes in asthma, atopic dermatitis, allergic rhinitis, COPD, cancer, inflammatory bowel disease, autoimmune disease and fibrotic disease as well as evaluating the associated clinical data derived from blocking IL-4, IL-13 or IL-4 and IL-13 together.
Collapse
|
45
|
Nambiar J, Clarke AW, Shim D, Mabon D, Tian C, Windloch K, Buhmann C, Corazon B, Lindgren M, Pollard M, Domagala T, Poulton L, Doyle AG. Potent neutralizing anti-CD1d antibody reduces lung cytokine release in primate asthma model. MAbs 2016; 7:638-50. [PMID: 25751125 PMCID: PMC4623119 DOI: 10.1080/19420862.2015.1016693] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
CD1d is a receptor on antigen-presenting cells involved in triggering cell populations, particularly natural killer T (NKT) cells, to release high levels of cytokines. NKT cells are implicated in asthma pathology and blockade of the CD1d/NKT cell pathway may have therapeutic potential. We developed a potent anti-human CD1d antibody (NIB.2) that possesses high affinity for human and cynomolgus macaque CD1d (KD ∼100 pM) and strong neutralizing activity in human primary cell-based assays (IC50 typically <100 pM). By epitope mapping experiments, we showed that NIB.2 binds to CD1d in close proximity to the interface of CD1d and the Type 1 NKT cell receptor β-chain. Together with data showing that NIB.2 inhibited stimulation via CD1d loaded with different glycolipids, this supports a mechanism whereby NIB.2 inhibits NKT cell activation by inhibiting Type 1 NKT cell receptor β-chain interactions with CD1d, independent of the lipid antigen in the CD1d antigen-binding cleft. The strong in vitro potency of NIB.2 was reflected in vivo in an Ascaris suum cynomolgus macaque asthma model. Compared with vehicle control, NIB.2 treatment significantly reduced bronchoalveolar lavage (BAL) levels of Ascaris-induced cytokines IL-5, IL-8 and IL-1 receptor antagonist, and significantly reduced baseline levels of GM-CSF, IL-6, IL-15, IL-12/23p40, MIP-1α, MIP-1β, and VEGF. At a cellular population level NIB.2 also reduced numbers of BAL lymphocytes and macrophages, and blood eosinophils and basophils. We demonstrate that anti-CD1d antibody blockade of the CD1d/NKT pathway modulates inflammatory parameters in vivo in a primate inflammation model, with therapeutic potential for diseases where the local cytokine milieu is critical.
Collapse
Key Words
- AHR, airway hyper-reactivity; APC, antigen-presenting cell; AUC, area under the curve; BAL, broncho-alveolar lavage; BSA, bovine serum albumin; CHO, Chinese hamster ovary; ELISA, enzyme-linked immunosorbent assay; G-CSF, granulocyte colony stimulating fac
- CD1d, NKT cell, antibody, asthma, cytokine
Collapse
Affiliation(s)
- Jonathan Nambiar
- a Teva Pharmaceuticals Australia Pty. Ltd. ; North Ryde , NSW Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Landolina N, Levi-Schaffer F. Monoclonal antibodies: the new magic bullets for allergy: IUPHAR Review 17. Br J Pharmacol 2016; 173:793-803. [PMID: 26620589 DOI: 10.1111/bph.13396] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 11/08/2015] [Accepted: 11/22/2015] [Indexed: 12/18/2022] Open
Abstract
Allergic diseases and conditions are widespread and their incidence is on the increase. They are characterized by the activation of mast cells resident in tissues and the consequent infiltration and stimulation of several inflammatory cells, predominantly eosinophils. Cell-cell cross-talk and the release of mediators are responsible for the symptoms and for the modulation of the response. The gold standard of therapeutic intervention is still glucocorticosteroids, although they are not effective in all patients and may cause numerous side effects. Symptomatic medications are also widespread. As research has led to deeper insights into the mechanisms governing the diseases, new avenues have been opened resulting in recent years in the development of monoclonal antibodies (mAbs) such as anti-IgE mAbs (omalizumab) and others still undergoing clinical trials aimed to specifically target molecules involved in the migration and stimulation of inflammatory cells. In this review, we summarize new developments in the field of anti-allergic mAbs with special emphasis on the treatment of asthma, particularly severe forms of this condition, and atopic dermatitis, which are two unmet clinical needs.
Collapse
Affiliation(s)
- N Landolina
- Pharmacology and Experimental Therapeutics Unit, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - F Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
47
|
Abstract
Since the development of therapeutic antibodies the demand of recombinant human antibodies is steadily increasing. Traditionally, therapeutic antibodies were generated by immunization of rat or mice, the generation of hybridoma clones, cloning of the antibody genes and subsequent humanization and engineering of the lead candidates. In the last few years, techniques were developed that use transgenic animals with a human antibody gene repertoire. Here, modern recombinant DNA technologies can be combined with well established immunization and hybridoma technologies to generate already affinity maturated human antibodies. An alternative are in vitro technologies which enabled the generation of fully human antibodies from antibody gene libraries that even exceed the human antibody repertoire. Specific antibodies can be isolated from these libraries in a very short time and therefore reduce the development time of an antibody drug at a very early stage.In this review, we describe different technologies that are currently used for the in vitro and in vivo generation of human antibodies.
Collapse
|
48
|
Fallahi M, Keyhanmanesh R, Khamaneh AM, Ebrahimi Saadatlou MA, Saadat S, Ebrahimi H. Effect of Alpha-Hederin, the active constituent of Nigella sativa, on miRNA-126, IL-13 mRNA levels and inflammation of lungs in ovalbumin-sensitized male rats. AVICENNA JOURNAL OF PHYTOMEDICINE 2016; 6:77-85. [PMID: 27247924 PMCID: PMC4884220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE In previous studies the therapeutic effects of Nigella sativa have been demonstrated on asthmatic animals. In the present study, the preventive effect of single dose of alpha-hederin, its active constituent, has been evaluated on lung inflammation and some inflammatory mediators in lungs of ovalbumin sensitized rat in order to elicit its mechanism. MATERIALS AND METHODS Forty rats were randomly grouped in 4 groups; control (C), sensitized (S), sensitized pretreated groups with thymoquinone (3 mg/kg i.p., S+TQ) and alpha-hederin (0.02 mg/kg i.p., S+AH). Levels of IL-13 mRNA and miRNA-126 in lung tissue and its pathological changes in each group were assessed. RESULTS Elevated levels of miRNA-126, IL-13 mRNA and pathological changes were observed in the sensitized group compared to the control group (p<0.001 to p<0.05). All of these factors were significantly reduced in S+TQ and S+AH groups in comparison to S group (p<0.001 to p<0.05). Although alpha-hederin decreased the levels of miRNA-126, IL-13 mRNA and pathological changes in comparison with thymoquinone, the results were statistically not significant. CONCLUSION The results suggested that alpha-hederin had preventive effect on sensitized rats like thymoquinone. It may intervene in miRNA-126 expression, which consequently could interfere with IL-13 secretion pathway leading to a reduction in inflammatory responses.
Collapse
Affiliation(s)
- Maryam Fallahi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Medical Education Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mahdi Khamaneh
- Department of molecular medicine, School of advanced medical sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Saeideh Saadat
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Physiology, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Hadi Ebrahimi
- Tuberculosis and lung diseases research center, Tabriz University of Medical Sciences, Tabriz, Iran,Corresponding Author: Tel: 041-33364664, Fax: 041-33364664,
| |
Collapse
|
49
|
Urbach C, Gordon NC, Strickland I, Lowne D, Joberty-Candotti C, May R, Herath A, Hijnen D, Thijs JL, Bruijnzeel-Koomen CA, Minter RR, Hollfelder F, Jermutus L. Combinatorial Screening Identifies Novel Promiscuous Matrix Metalloproteinase Activities that Lead to Inhibition of the Therapeutic Target IL-13. ACTA ACUST UNITED AC 2015; 22:1442-1452. [PMID: 26548614 DOI: 10.1016/j.chembiol.2015.09.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/10/2015] [Accepted: 09/28/2015] [Indexed: 12/20/2022]
Abstract
The practical realization of disease modulation by catalytic degradation of a therapeutic target protein suffers from the difficulty to identify candidate proteases, or to engineer their specificity. We identified 23 measurable, specific, and new protease activities using combinatorial screening of 27 human proteases against 24 therapeutic protein targets. We investigate the cleavage of monocyte chemoattractant protein 1, interleukin-6 (IL-6), and IL-13 by matrix metalloproteinases (MMPs) and serine proteases, and demonstrate that cleavage of IL-13 leads to potent inhibition of its biological activity in vitro. MMP-8 degraded human IL-13 most efficiently in vitro and ex vivo in human IL-13 transgenic mouse bronchoalveolar lavage. Hence, MMP-8 is a therapeutic protease lead against IL-13 for inflammatory conditions whereby reported genetic and genomics data suggest an involvement of MMP-8. This work describes the first exploitation of human enzyme promiscuity for therapeutic applications, and reveals both starting points for protease-based therapies and potential new regulatory networks in inflammatory disease.
Collapse
Affiliation(s)
- Carole Urbach
- Department of Antibody Discovery and Protein Engineering, MedImmune, Granta Park, Cambridge CB21 6GH, UK.
| | - Nathaniel C Gordon
- Department of Antibody Discovery and Protein Engineering, MedImmune, Granta Park, Cambridge CB21 6GH, UK; Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK
| | - Ian Strickland
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune, Granta Park, Cambridge CB21 6GH, UK
| | - David Lowne
- Department of Antibody Discovery and Protein Engineering, MedImmune, Granta Park, Cambridge CB21 6GH, UK
| | | | - Richard May
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune, Granta Park, Cambridge CB21 6GH, UK
| | - Athula Herath
- Non Clinical Biostatistics, MedImmune, Granta Park, Cambridge CB21 6GH, UK
| | - DirkJan Hijnen
- Department of Dermatology, University Medical Center, 3508 GA Utrecht, the Netherlands
| | - Judith L Thijs
- Department of Dermatology, University Medical Center, 3508 GA Utrecht, the Netherlands
| | | | - Ralph R Minter
- Department of Antibody Discovery and Protein Engineering, MedImmune, Granta Park, Cambridge CB21 6GH, UK
| | - Florian Hollfelder
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK
| | - Lutz Jermutus
- Department of Antibody Discovery and Protein Engineering, MedImmune, Granta Park, Cambridge CB21 6GH, UK
| |
Collapse
|
50
|
Baverel PG, Jain M, Stelmach I, She D, Agoram B, Sandbach S, Piper E, Kuna P. Pharmacokinetics of tralokinumab in adolescents with asthma: implications for future dosing. Br J Clin Pharmacol 2015; 80:1337-49. [PMID: 26182954 DOI: 10.1111/bcp.12725] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/10/2015] [Accepted: 07/13/2015] [Indexed: 12/31/2022] Open
Abstract
AIMS Tralokinumab, an investigational human immunoglobulin G4 monoclonal antibody, potently and specifically neutralizes interleukin-13, a central mediator of asthma. Tralokinumab has shown improvements in clinical endpoints in adults with uncontrolled asthma. The present study explored the pharmacokinetics (PK) and safety of a single tralokinumab dose, and utilized a population PK modelling and simulation approach to evaluate the optimal dosing strategy for adolescents. METHODS Adolescent subjects with asthma, using daily controller medication, received a single subcutaneous dose of tralokinumab 300 mg. Safety, immunogenicity and PK data were collected during a 57-day follow-up. A population PK model was developed using data from the present study and prior studies in adults. Simulations were performed to evaluate dose adjustment requirements for adolescents. RESULTS Twenty adolescents (12-17 years) were enrolled; all completed the study. No clinically relevant safety findings or antidrug antibodies were detected. PK parameters were similar to those observed in adults. PK modelling showed that body weight was a minor predictor of tralokinumab PK; after incorporating body weight into the PK model, a 15% (nonparametric 95% confidence interval 5%, 26%) lower clearance was found in adolescents compared with adults [173 (151, 209) vs. 204 (191, 229) ml day(-1)]. Simulations showed no therapeutically relevant differences in exposures between adolescent and adult populations, and similar PK profiles for weight-based (4 mg kg(-1)) and fixed (300 mg) fortnightly subcutaneous doses of tralokinumab. CONCLUSION Single-dose administration of tralokinumab 300 mg in adolescents was well tolerated, with a PK profile similar to that in adults. Exposure predictions suggest that dose adjustment is not required for adolescents.
Collapse
Affiliation(s)
- Paul G Baverel
- Clinical Pharmacology, Drug Metabolism and Pharmacokinetics, MedImmune, Cambridge, UK
| | - Meena Jain
- Clinical Development, MedImmune, Cambridge, UK
| | - Iwona Stelmach
- Department of Paediatrics and Allergy, Medical University of Łódź, Poland
| | - Dewei She
- Biostatistics, MedImmune, Gaithersburg, MD, USA
| | - Balaji Agoram
- Clinical Pharmacology, Drug Metabolism and Pharmacokinetics, MedImmune, Cambridge, UK
| | | | | | - Piotr Kuna
- Department of Internal Medicine, Asthma and Allergy, Barlicki University Hospital, Medical University of Łódź, Poland
| |
Collapse
|