1
|
Eggel A, Pennington LF, Jardetzky TS. Therapeutic monoclonal antibodies in allergy: Targeting IgE, cytokine, and alarmin pathways. Immunol Rev 2024; 328:387-411. [PMID: 39158477 PMCID: PMC11659931 DOI: 10.1111/imr.13380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
The etiology of allergy is closely linked to type 2 inflammatory responses ultimately leading to the production of allergen-specific immunoglobulin E (IgE), a key driver of many allergic conditions. At a high level, initial allergen exposure disrupts epithelial integrity, triggering local inflammation via alarmins including IL-25, IL-33, and TSLP, which activate type 2 innate lymphoid cells as well as other immune cells to secrete type 2 cytokines IL-4, IL-5 and IL-13, promoting Th2 cell development and eosinophil recruitment. Th2 cell dependent B cell activation promotes the production of allergen-specific IgE, which stably binds to basophils and mast cells. Rapid degranulation of these cells upon allergen re-exposure leads to allergic symptoms. Recent advances in our understanding of the molecular and cellular mechanisms underlying allergic pathophysiology have significantly shaped the development of therapeutic intervention strategies. In this review, we highlight key therapeutic targets within the allergic cascade with a particular focus on past, current and future treatment approaches using monoclonal antibodies. Specific targeting of alarmins, type 2 cytokines and IgE has shown varying degrees of clinical benefit in different allergic indications including asthma, chronic spontaneous urticaria, atopic dermatitis, chronic rhinosinusitis with nasal polyps, food allergies and eosinophilic esophagitis. While multiple therapeutic antibodies have been approved for clinical use, scientists are still working on ways to improve on current treatment approaches. Here, we provide context to understand therapeutic targeting strategies and their limitations, discussing both knowledge gaps and promising future directions to enhancing clinical efficacy in allergic disease management.
Collapse
Affiliation(s)
- Alexander Eggel
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
- Department of Rheumatology and ImmunologyUniversity Hospital BernBernSwitzerland
| | | | - Theodore S. Jardetzky
- Department of Structural BiologyStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
2
|
Combined IgE neutralization and Bifidobacterium longum supplementation reduces the allergic response in models of food allergy. Nat Commun 2022; 13:5669. [PMID: 36167830 PMCID: PMC9515155 DOI: 10.1038/s41467-022-33176-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/06/2022] [Indexed: 11/09/2022] Open
Abstract
IgE is central to the development of allergic diseases, and its neutralization alleviates allergic symptoms. However, most of these antibodies are based on IgG1, which is associated with an increased risk of fragment crystallizable-mediated side effects. Moreover, omalizumab, an anti-IgE antibody approved for therapeutic use, has limited benefits for patients with high IgE levels. Here, we assess a fusion protein with extracellular domain of high affinity IgE receptor, FcεRIα, linked to a IgD/IgG4 hybrid Fc domain we term IgETRAP, to reduce the risk of IgG1 Fc-mediated side effects. IgETRAP shows enhanced IgE binding affinity compared to omalizumab. We also see an enhanced therapeutic effect of IgETRAP in food allergy models when combined with Bifidobacterium longum, which results in mast cell number and free IgE levels. The combination of IgETRAP and B. longum may therefore represent a potent treatment for allergic patients with high IgE levels. IgE is a critical component of the allergic response and therapeutic targeting can alleviate symptomology. Here the authors propose the combined use of Bifidobacterium longum and a FcεRIα extracellular domain linked to a IgD/IgG4 hybrid Fc domain fusion protein called IgETRAP and show reduction of mast cell and IgE levels in models of food allergy.
Collapse
|
3
|
Wang Y, Zheng C, Zhuang C, Fu Q, Zhang B, Bian Y, Qi N, Zhu J. Characterization and pre-clinical assessment of a proposed biosimilar to its originator Omalizumab. Eur J Pharm Sci 2022; 178:106292. [PMID: 36089232 DOI: 10.1016/j.ejps.2022.106292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/28/2022] [Accepted: 09/04/2022] [Indexed: 11/03/2022]
Abstract
Omalizumab is an anti-IgE monoclonal antibody (mAb) approved for the treatment of moderate-to-severe asthma. Herein, we report physicochemical, biological, pharmacological, and toxicological characteristics of an Omalizumab biosimilar mAb named KA. We show that KA and its originator present only minimum differences. Their charge heterogeneity and primary, secondary structures are similar. The two molecules are comparable regarding in vitro activity, including molecular binding and cell-based inhibition. Pharmacological and toxicological properties were assessed using a mouse model of allergy and cynomolgus monkeys, and we determined that the efficacy, safety, and pharmacokinetic characteristics of KA are comparable to its originator. Our data, which demonstrated that KA has similar activity to the Omalizumab reference product in relevant preclinical models, calls for a clinical evaluation of its bio-similarity.
Collapse
Affiliation(s)
- Yanchao Wang
- Engineering Research Center of Cell & Therapeutic Antibody, MOE, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Chen Zheng
- Shanghai Taiyin Biotechnology Co., Ltd., 2066 Wangyuan Road, Shanghai 201403, China
| | - Chao Zhuang
- Shanghai Taiyin Biotechnology Co., Ltd., 2066 Wangyuan Road, Shanghai 201403, China
| | - Qiang Fu
- Shanghai Taiyin Biotechnology Co., Ltd., 2066 Wangyuan Road, Shanghai 201403, China
| | - Baohong Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, MOE, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yanling Bian
- Engineering Research Center of Cell & Therapeutic Antibody, MOE, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Nianmin Qi
- Shanghai Taiyin Biotechnology Co., Ltd., 2066 Wangyuan Road, Shanghai 201403, China.
| | - Jianwei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody, MOE, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
4
|
Liu S, Shah DK. Mathematical Models to Characterize the Absorption, Distribution, Metabolism, and Excretion of Protein Therapeutics. Drug Metab Dispos 2022; 50:867-878. [PMID: 35197311 PMCID: PMC11022906 DOI: 10.1124/dmd.121.000460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/31/2022] [Indexed: 11/22/2022] Open
Abstract
Therapeutic proteins (TPs) have ranked among the most important and fastest-growing classes of drugs in the clinic, yet the development of successful TPs is often limited by unsatisfactory efficacy. Understanding pharmacokinetic (PK) characteristics of TPs is key to achieving sufficient and prolonged exposure at the site of action, which is a prerequisite for eliciting desired pharmacological effects. PK modeling represents a powerful tool to investigate factors governing in vivo disposition of TPs. In this mini-review, we discuss many state-of-the-art models that recapitulate critical processes in each of the absorption, distribution, metabolism/catabolism, and excretion pathways of TPs, which can be integrated into the physiologically-based pharmacokinetic framework. Additionally, we provide our perspectives on current opportunities and challenges for evolving the PK models to accelerate the discovery and development of safe and efficacious TPs. SIGNIFICANCE STATEMENT: This minireview provides an overview of mechanistic pharmacokinetic (PK) models developed to characterize absorption, distribution, metabolism, and elimination (ADME) properties of therapeutic proteins (TPs), which can support model-informed discovery and development of TPs. As the next-generation of TPs with diverse physicochemical properties and mechanism-of-action are being developed rapidly, there is an urgent need to better understand the determinants for the ADME of TPs and evolve existing platform PK models to facilitate successful bench-to-bedside translation of these promising drug molecules.
Collapse
Affiliation(s)
- Shufang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
5
|
Impact of anti-PEG antibody affinity on accelerated blood clearance of pegylated epoetin beta in mice. Biomed Pharmacother 2021; 146:112502. [PMID: 34891120 DOI: 10.1016/j.biopha.2021.112502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/22/2021] [Accepted: 12/02/2021] [Indexed: 11/24/2022] Open
Abstract
Antibodies that bind polyethylene glycol (PEG) can be induced by pegylated biomolecules and also exist in a significant fraction of healthy individuals who have never received pegylated medicines. The binding affinity of antibodies against PEG (anti-PEG antibodies) likely varies depending on if they are induced or naturally occurring. Anti-PEG antibodies can accelerate the clearance of pegylated medicines from the circulation, resulting in loss of drug efficacy, but it is unknown how accelerated blood clearance is affected by anti-PEG antibody affinity. We identified a panel of anti-PEG IgG and IgM antibodies with binding avidities ranging over several orders of magnitude to methoxy polyethylene glycol-epoetin beta (PEG-EPO), which is used to treat patients suffering from anemia. Formation of in vitro immune complexes between PEG-EPO and anti-PEG IgG or IgM antibodies was more obvious as antibody affinity increased. Likewise, high affinity anti-PEG antibodies produced greater accelerated blood clearance of PEG-EPO as compared to low affinity antibodies. The molar ratio of anti-PEG antibody to PEG-EPO that accelerates drug clearance in mice correlates with antibody binding avidity. Our study indicates that the bioactivity of PEG-EPO may be reduced due to rapid clearance in patients with either high concentrations of low affinity or low concentrations of high affinity anti-PEG IgG and IgM antibodies.
Collapse
|
6
|
Pennington LF, Gasser P, Kleinboelting S, Zhang C, Skiniotis G, Eggel A, Jardetzky TS. Directed evolution of and structural insights into antibody-mediated disruption of a stable receptor-ligand complex. Nat Commun 2021; 12:7069. [PMID: 34862384 PMCID: PMC8642555 DOI: 10.1038/s41467-021-27397-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 11/11/2021] [Indexed: 11/15/2022] Open
Abstract
Antibody drugs exert therapeutic effects via a range of mechanisms, including competitive inhibition, allosteric modulation, and immune effector mechanisms. Facilitated dissociation is an additional mechanism where antibody-mediated “disruption” of stable high-affinity macromolecular complexes can potentially enhance therapeutic efficacy. However, this mechanism is not well understood or utilized therapeutically. Here, we investigate and engineer the weak disruptive activity of an existing therapeutic antibody, omalizumab, which targets IgE antibodies to block the allergic response. We develop a yeast display approach to select for and engineer antibody disruptive efficiency and generate potent omalizumab variants that dissociate receptor-bound IgE. We determine a low resolution cryo-EM structure of a transient disruption intermediate containing the IgE-Fc, its partially dissociated receptor and an antibody inhibitor. Our results provide a conceptual framework for engineering disruptive inhibitors for other targets, insights into the failure in clinical trials of the previous high affinity omalizumab HAE variant and anti-IgE antibodies that safely and rapidly disarm allergic effector cells. Facilitated dissociation is a mechanism where antibody-mediated disruption of high-affinity complexes can enhance the therapeutic effects of a drug. Here the authors present a yeast display approach to select and engineer omalizumab variants that dissociate receptor-bound IgE to accelerate its inhibition of the allergic response.
Collapse
Affiliation(s)
- Luke F Pennington
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Progam in Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Sean N. Parker Center for Allergy Research at Stanford University, Stanford, CA, 94305, USA
| | - Pascal Gasser
- Department of Rheumatology and Immunology, University Hospital Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Silke Kleinboelting
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chensong Zhang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Georgios Skiniotis
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Alexander Eggel
- Department of Rheumatology and Immunology, University Hospital Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Theodore S Jardetzky
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA. .,Progam in Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA. .,Sean N. Parker Center for Allergy Research at Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
7
|
Xiang H, Chan AG, Ahene A, Bellovin DI, Deng R, Hsu AW, Jeffry U, Palencia S, Powers J, Zanghi J, Collins H. Preclinical characterization of bemarituzumab, an anti-FGFR2b antibody for the treatment of cancer. MAbs 2021; 13:1981202. [PMID: 34719330 PMCID: PMC8565817 DOI: 10.1080/19420862.2021.1981202] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Bemarituzumab (FPA144) is a first-in-class, humanized, afucosylated immunoglobulin G1 monoclonal antibody (mAb) directed against fibroblast growth factor receptor 2b (FGFR2b) with two mechanisms of action against FGFR2b-overexpressing tumors: inhibition of FGFR2b signaling and enhanced antibody-dependent cell-mediated cytotoxicity (ADCC). Bemarituzumab is being developed as a cancer therapeutic, and we summarize here the key nonclinical data that supported moving it into clinical trials. Bemarituzumab displayed sub-nanomolar cross-species affinity for FGFR2b receptors, with >20-fold enhanced binding affinity to human Fc gamma receptor IIIa compared with the fucosylated version. In vitro, bemarituzumab induced potent ADCC against FGFR2b-expressing tumor cells, and inhibited FGFR2 phosphorylation and proliferation of SNU-16 gastric cancer cells in a concentration-dependent manner. In vivo, bemarituzumab inhibited tumor growth through inhibition of the FGFR2b pathway and/or ADCC in mouse models. Bemarituzumab demonstrated enhanced anti-tumor activity in combination with chemotherapy, and due to bemarituzumab-induced natural killer cell-dependent increase in programmed death-ligand 1, also resulted in enhanced anti-tumor activity when combined with an anti-programmed death-1 antibody. Repeat-dose toxicity studies established the highest non-severely-toxic dose at 1 and 100 mg/kg in rats and cynomolgus monkeys, respectively. In pharmacokinetic (PK) studies, bemarituzumab exposure increase was greater than dose-proportional, with the linear clearance in the expected dose range for a mAb. The PK data in cynomolgus monkeys were used to project bemarituzumab linear PK in humans, which were consistent with the observed human Phase 1 data. These key nonclinical studies facilitated the successful advancement of bemarituzumab into the clinic.
Collapse
Affiliation(s)
- Hong Xiang
- Five Prime Therapeutics, Inc, South San Francisco, California.,Clinical Pharmacology, Modeling and Simulation, Amgen Inc, Thousand Oaks, California
| | - Abigael G Chan
- Five Prime Therapeutics, Inc, South San Francisco, California.,Global Project Management, Zai Lab (US) LLC, Menlo Park, California
| | - Ago Ahene
- Five Prime Therapeutics, Inc, South San Francisco, California.,Bioanalytic Sciences, Amgen Inc, South San Francisco, California
| | - David I Bellovin
- Five Prime Therapeutics, Inc, South San Francisco, California.,Bioanalytic Sciences, Amgen Inc, South San Francisco, California
| | - Rong Deng
- R&D Q-Pharm Consulting LLC, Pleasanton
| | - Amy W Hsu
- Five Prime Therapeutics, Inc, South San Francisco, California.,Research, Merck & Co., Inc, South San Francisco, California
| | - Ursula Jeffry
- Five Prime Therapeutics, Inc, South San Francisco, California.,Toxicology Department, NGM Biopharmaceuticals, Inc, San Francisco, California
| | - Servando Palencia
- Five Prime Therapeutics, Inc, South San Francisco, California.,Research, Teva Pharmaceuticals, Redwood city, California
| | - Janine Powers
- Five Prime Therapeutics, Inc, South San Francisco, California.,Translational Medicine, Nurix Therapeutics, San Francisco, California
| | - James Zanghi
- Five Prime Therapeutics, Inc, South San Francisco, California.,Bioanalytic Sciences, Genentech Inc., South San Francisco, California
| | - Helen Collins
- Five Prime Therapeutics, Inc, South San Francisco, California.,Clinic, Amgen Inc., South San Francisco, California
| |
Collapse
|
8
|
Chen X, Peng F, Ji Y, Xiang H, Wang X, Liu T, Wang H, Han Y, Wang C, Zhang Y, Kong X, Lang JY. Brca2 deficiency drives gastrointestinal tumor formation and is selectively inhibited by mitomycin C. Cell Death Dis 2020; 11:812. [PMID: 32980867 PMCID: PMC7519908 DOI: 10.1038/s41419-020-03013-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022]
Abstract
BRCA2 is crucial for repairing DNA double-strand breaks with high fidelity, and loss of BRCA2 increases the risks of developing breast and ovarian cancers. Herein, we show that BRCA2 is inactively mutated in 10% of gastric and 7% of colorectal adenocarcinomas, and that this inactivation is significantly correlated with microsatellite instability. Villin-driven Brca2 depletion promotes mouse gastrointestinal tumor formation when genome instability is increased. Whole-genome screening data showed that these BRCA2 monoallelic and biallelic mutant tumors were selectively inhibited by mitomycin C. Mechanistically, mitomycin C provoked double-strand breaks in cancer cells that often recruit wild-type BRCA2 for repair; the failure to repair double-strand breaks caused cell-cycle arrest at the S phase and p53-mediated cell apoptosis of BRCA2 monoallelic and biallelic mutant tumor cells. Our study unveils the role of BRCA2 loss in the development of gastrointestinal tumors and provides a potential therapeutic strategy to eliminate BRCA2 monoallelic and biallelic mutant tumors through mitomycin C.
Collapse
Affiliation(s)
- Xiaomin Chen
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Fangfei Peng
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yan Ji
- Bioinformatics Core, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Honggang Xiang
- Department of General Surgery, Pudong New Area People's Hospital affiliated to Shanghai University of Medicine & Health Science, 201299, Shanghai, China
| | - Xiang Wang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Tingting Liu
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Heng Wang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yumin Han
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Changxu Wang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yongfeng Zhang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiangyin Kong
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Jing-Yu Lang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
9
|
Meyer DW, Bou LB, Shum S, Jonas M, Anderson ME, Hamilton JZ, Hunter JH, Wo SW, Wong AO, Okeley NM, Lyon RP. An in Vitro Assay Using Cultured Kupffer Cells Can Predict the Impact of Drug Conjugation on in Vivo Antibody Pharmacokinetics. Mol Pharm 2020; 17:802-809. [DOI: 10.1021/acs.molpharmaceut.9b00991] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- David W Meyer
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| | - Lauren B Bou
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| | - Sara Shum
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| | - Mechthild Jonas
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| | - Martha E Anderson
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| | - Joe Z Hamilton
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| | - Joshua H Hunter
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| | - Serena W Wo
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| | - Abbie O Wong
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| | - Nicole M Okeley
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| | - Robert P Lyon
- Seattle Genetics, Inc., 21823 30th Dr SE Bothell, Washington 98021, United States
| |
Collapse
|
10
|
Hasni S, Gupta S, Davis M, Poncio E, Temesgen-Oyelakin Y, Joyal E, Fike A, Manna Z, Auh S, Shi Y, Chan D, Carlucci P, Biehl A, Dema B, Charles N, Balow JE, Waldman M, Siegel RM, Kaplan MJ, Rivera J. Safety and Tolerability of Omalizumab: A Randomized Clinical Trial of Humanized Anti-IgE Monoclonal Antibody in Systemic Lupus Erythematosus. Arthritis Rheumatol 2019; 71:1135-1140. [PMID: 30597768 DOI: 10.1002/art.40828] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/27/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Autoreactive IgE antibodies have been implicated in the pathogenesis of systemic lupus erythematosus (SLE). We hypothesize that omalizumab, a monoclonal antibody binding IgE, may improve SLE activity by reducing type I interferon (IFN) production by hampering plasmacytoid dendritic cells and basophil activation. This study was undertaken to assess the safety, tolerability, and clinical efficacy of omalizumab in mild to moderate SLE. METHODS Sixteen subjects with SLE and a Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) score of ≥4 and elevated autoreactive IgE antibody levels were randomized to receive omalizumab or placebo (2:1) for 16 weeks, followed by 16 weeks of open-label treatment and a 4-week washout period. The SLEDAI-2K score, British Isles Lupus Assessment Group index (BILAG 2004) score, and physician's global assessment of disease activity were recorded at each visit. The type I IFN-induced gene signature was determined using quantitative polymerase chain reaction. RESULTS Omalizumab was well tolerated with no allergic reactions, and mostly mild adverse events comparable to those experienced with placebo treatment. SLEDAI-2K scores improved in the omalizumab group compared to the placebo group at week 16 (P = 0.038), as well as during the open-label phase in subjects initially receiving placebo (P = 0.02). No worsening in BILAG scores or the physician's global assessment was detected. There was a trend toward a reduction in IFN gene signature in subjects treated with omalizumab (P = 0.11), especially in subjects with a high baseline IFN signature (P = 0.052). CONCLUSION Our findings indicate that omalizumab is well tolerated in SLE and is associated with improvement in disease activity. Larger randomized clinical trials will be needed to assess the efficacy of omalizumab in patients with SLE.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Sungyoung Auh
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland
| | | | | | | | - Ann Biehl
- Clinical Center, NIH, Bethesda, Maryland
| | | | | | - James E Balow
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland
| | - Meryl Waldman
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland
| | | | - Mariana J Kaplan
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Juan Rivera
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| |
Collapse
|
11
|
Chung S, Nguyen V, Lin YL, Lafrance-Vanasse J, Scales SJ, Lin K, Deng R, Williams K, Sperinde G, Li JJ, Zheng K, Sukumaran S, Tesar D, Ernst JA, Fischer S, Lazar GA, Prabhu S, Song A. An in vitro FcRn- dependent transcytosis assay as a screening tool for predictive assessment of nonspecific clearance of antibody therapeutics in humans. MAbs 2019; 11:942-955. [PMID: 30982394 PMCID: PMC6601550 DOI: 10.1080/19420862.2019.1605270] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
A cell-based assay employing Madin–Darby canine kidney cells stably expressing human neonatal Fc receptor (FcRn) heavy chain and β2-microglobulin genes was developed to measure transcytosis of monoclonal antibodies (mAbs) under conditions relevant to the FcRn-mediated immunoglobulin G (IgG) salvage pathway. The FcRn-dependent transcytosis assay is modeled to reflect combined effects of nonspecific interactions between mAbs and cells, cellular uptake via pinocytosis, pH-dependent interactions with FcRn, and dynamics of intracellular trafficking and sorting mechanisms. Evaluation of 53 mAbs, including 30 marketed mAb drugs, revealed a notable correlation between the transcytosis readouts and clearance in humans. FcRn was required to promote efficient transcytosis of mAbs and contributed directly to the observed correlation. Furthermore, the transcytosis assay correctly predicted rank order of clearance of glycosylation and Fv charge variants of Fc-containing proteins. These results strongly support the utility of this assay as a cost-effective and animal-sparing screening tool for evaluation of mAb-based drug candidates during lead selection, optimization, and process development for desired pharmacokinetic properties.
Collapse
Affiliation(s)
- Shan Chung
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Van Nguyen
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Yuwen Linda Lin
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | | | - Suzie J Scales
- c Department of Molecular Biology , Genentech Inc ., South San Francisco , CA , USA
| | - Kevin Lin
- d Department of Analytical Operations , Genentech Inc ., South San Francisco , CA , USA
| | - Rong Deng
- e Department of Clinical Pharmacology , Genentech Inc ., South San Francisco , CA , USA
| | - Kathi Williams
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Gizette Sperinde
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Juan Jenny Li
- f Department of Biochemistry and Cellular Pharmacology , Genentech Inc ., South San Francisco , CA , USA
| | - Kai Zheng
- g Department of Late Stage Pharmaceutical Development , Genentech Inc ., South San Francisco , CA , USA
| | - Siddharth Sukumaran
- h Department of Pharmacokinetics & Pharmacodynamics , Genentech Inc ., South San Francisco , CA , USA
| | - Devin Tesar
- i Department of Drug Delivery , Genentech Inc ., South San Francisco , CA , USA
| | - James A Ernst
- b Department of Protein Chemistry , Genentech Inc ., South San Francisco , CA , USA
| | - Saloumeh Fischer
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Greg A Lazar
- j Department of Antibody Engineering , Genentech Inc ., South San Francisco , CA , USA
| | - Saileta Prabhu
- h Department of Pharmacokinetics & Pharmacodynamics , Genentech Inc ., South San Francisco , CA , USA
| | - An Song
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| |
Collapse
|
12
|
Yang Z, Du M, Wang W, Xin X, Ma P, Zhang H, Lerner RA. Affinity maturation of an TpoR targeting antibody in full-length IgG form for enhanced agonist activity. Protein Eng Des Sel 2019; 31:233-241. [PMID: 29474709 DOI: 10.1093/protein/gzy002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/31/2018] [Indexed: 01/08/2023] Open
Abstract
It has been observed that converting scFv formatted antibodies to full-length IgG often associates with loss of affinity. We aim to address this issue in this paper by establishing an integrated affinity maturation method applying yeast display technology platform. To demonstrate that, we employed a human thrombopoietin receptor targeting antibody named 3D9 which was identified previously from a combinational antibody library in scFv-Fc fusion protein form. We have observed that significant potency loss happened when 3D9 was transformed to full-length IgG form. In this study, we tested whether the potency of the full-length IgG can be improved by affinity maturation of 3D9 using a modified Fab yeast display platform. An efficient CDR3 targeted mutagenesis strategy was designed for Fab library with pre-designed CDR diversity. Next generation sequencing was also used for evaluation of the enrichment process and investigation of sequence-function relationship of the antibody. A variant with improved affinity and higher potency was identified. The study demonstrates that the strategy we used here are efficient for optimizing affinity and activity of full-length IgGs.
Collapse
Affiliation(s)
- Zhuo Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mingjuan Du
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Xiu Xin
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Peixiang Ma
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Hongkai Zhang
- State key laboratory of medicinal chemical biology, Nankai University, Tianjin.,College of Life Science, Nankai University, Tianjin, China
| | - Richard A Lerner
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China.,Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
13
|
Leipold D, Prabhu S. Pharmacokinetic and Pharmacodynamic Considerations in the Design of Therapeutic Antibodies. Clin Transl Sci 2018; 12:130-139. [PMID: 30414357 PMCID: PMC6440574 DOI: 10.1111/cts.12597] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022] Open
Abstract
The design and development of therapeutic monoclonal antibodies (mAbs) through optimizing their pharmacokinetic (PK) and pharmacodynamic (PD) properties is crucial to improve efficacy while minimizing adverse events. Many of these properties are interdependent, which highlights the inherent challenges in therapeutic antibody design, where improving one antibody property can sometimes lead to changes in others. Here, we discuss optimization approaches for PK/PD properties of therapeutic mAbs.
Collapse
Affiliation(s)
- Douglas Leipold
- Preclinical and Translational Pharmacokinetics/Pharmacodynamics, Genentech, South San Francisco, California, USA
| | - Saileta Prabhu
- Preclinical and Translational Pharmacokinetics/Pharmacodynamics, Genentech, South San Francisco, California, USA
| |
Collapse
|
14
|
Improvement of pharmacokinetic properties of therapeutic antibodies by antibody engineering. Drug Metab Pharmacokinet 2018; 34:25-41. [PMID: 30472066 DOI: 10.1016/j.dmpk.2018.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/13/2018] [Accepted: 10/23/2018] [Indexed: 01/17/2023]
Abstract
Monoclonal antibodies (mAbs) have become an important therapeutic option for several diseases. Since several mAbs have shown promising efficacy in clinic, the competition to develop mAbs has become severe. In efforts to gain a competitive advantage over other mAbs and provide significant benefits to patients, innovations in antibody engineering have aimed at improving the pharmacokinetic properties of mAbs. Because engineering can provide therapeutics that are more convenient, safer, and more efficacious for patients in several disease areas, it is an attractive approach to provide significant benefits to patients. Further advances in engineering mAbs to modulate their pharmacokinetics were driven by the increase of total soluble target antigen concentration that is often observed after injecting a mAb, which then requires a high dosage to antagonize. To decrease the required dosage, several antibody engineering techniques have been invented that reduce the total concentration of soluble target antigen. Here, we review the various ways that antibody engineering can improve the pharmacokinetic properties of mAbs.
Collapse
|
15
|
Translational pharmacokinetics and pharmacodynamics of monoclonal antibodies. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 21-22:75-83. [PMID: 27978991 DOI: 10.1016/j.ddtec.2016.09.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 01/10/2023]
Abstract
Monoclonal antibodies (mAbs) are an important therapeutic class with complex pharmacology and interdependent pharmacokinetic (PK) and pharmacodynamics (PD) properties. Understanding the PK and PD of mAbs and their biological and mechanistic underpinnings are crucial in enabling their design and selection, designing appropriate efficacy and toxicity studies, translating PK/PD parameters to humans, and optimizing dose and regimen to maximize success in the clinic. Significant progress has been made in this field however many critical questions still remain. This article gives a brief overview of the PK and PD of mAbs, factors that influence them, and areas of ongoing inquiry. Current tools and translational approaches to predict the PK/PD of mAbs in humans are also discussed.
Collapse
|
16
|
Liour SS, Tom A, Chan YH, Chang TW. Treating IgE-mediated diseases via targeting IgE-expressing B cells using an anti-CεmX antibody. Pediatr Allergy Immunol 2016; 27:446-51. [PMID: 27090058 DOI: 10.1111/pai.12584] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2016] [Indexed: 01/17/2023]
Abstract
Targeting the IgE pathway is a clinically validated strategy for treating IgE-mediated diseases. Omalizumab, an anti-IgE antibody, which binds to free IgE and prevents the binding of IgE to FcεRI on mast cells and basophils has been approved for severe persistent allergic asthma and chronic spontaneous (idiopathic) urticaria. The therapeutic efficacy of anti-IgE has also been reported in allergic rhinitis, allergic bronchopulmonary aspergillosis, latex allergy, atopic dermatitis, allergic urticaria, anaphylaxis, and others. Anti-CεmX, which binds to membrane-bound IgE (mIgE) on IgE-switched B cells, lyses mIgE-expressing B lymphoblasts and prevents the allergen-induced generation of IgE-producing plasma cells, offers an alternative mechanism of intervening with the IgE inflammatory pathway. Because anti-CεmX does not bind to free IgE, it can modulate the IgE pathway regardless of the serum IgE levels in treated patients. These unique pharmacologic mechanisms potentially enable anti-CεmX to provide different clinical utilities from anti-IgE and serve as a therapeutic and a prophylactic in some IgE-mediated diseases, which are not adequately treated with current medicine.
Collapse
Affiliation(s)
| | - Andrew Tom
- Genomics Research Center, Taipei, Taiwan
| | | | | |
Collapse
|
17
|
Weeratna RD, Chikh G, Zhang L, Fraser JD, Thorn JM, Merson JR, McCluskie MJ, Champion BR, Davis HL. Immunogenicity of a peptide-based anti-IgE conjugate vaccine in non-human primates. Immun Inflamm Dis 2016; 4:135-147. [PMID: 27957325 PMCID: PMC4879460 DOI: 10.1002/iid3.98] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 01/06/2016] [Accepted: 01/12/2016] [Indexed: 01/02/2023] Open
Abstract
The anti-human immunoglobulin E (IgE) monoclonal antibody, omalizumab (Xolair®, Genentech, South San Fransisco, CA), is effective in the treatment of poorly controlled moderate to severe allergic asthma and chronic idiopathic urticaria. It acts by specifically binding to the constant domain (Cϵ3) of free human IgE in the blood and interstitial fluid. Although efficacious, use of omalizumab is limited due to restrictions on patient weight and pre-existing IgE levels, and frequent dosing (q2-4 weeks). A vaccine inducing anti-IgE antibodies has the potential for similar clinical benefits with less frequent dosing and relatively lower cost of goods. We developed a vaccine containing two IgE peptide-conjugates targeting the Cϵ3 domain of human IgE. As part of preclinical evaluation of the vaccine to optimize formulation and dose prior to initiating clinical studies, we evaluated the vaccine in non-human primates, and demonstrate the induction of anti-peptide antibodies that can bind to conformationally intact human IgE and are capable, at least in some animals, of substantial lowering circulating IgE levels.
Collapse
Affiliation(s)
| | - Ghania Chikh
- Pfizer Vaccine ImmunotherapeuticsOttawa LaboratoriesOttawaOntarioCanada
| | - Lu Zhang
- Pfizer Vaccine ImmunotherapeuticsOttawa LaboratoriesOttawaOntarioCanada
| | | | | | - James R. Merson
- Pfizer Biotherapeutics Pharmaceutical SciencesSt. LouisMissouriUSA
| | | | | | - Heather L. Davis
- Pfizer Vaccine ImmunotherapeuticsOttawa LaboratoriesOttawaOntarioCanada
| |
Collapse
|
18
|
Haraya K, Tachibana T, Nezu J. Predicting pharmacokinetic profile of therapeutic antibodies after iv injection from only the data after sc injection in cynomolgus monkey. Xenobiotica 2016; 47:194-201. [PMID: 27151820 DOI: 10.1080/00498254.2016.1174792] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
1. The number of developed therapeutic monoclonal antibodies (mAbs) has increased in this decade. This study aims to predict their pharmacokinetic profiles after intravenous (iv) injection using only the data taken after subcutaneous (sc) injection in cynomolgus monkey. 2. Two-compartment model parameters, Q, Vc and Vp, were collected from the published data after iv injection in cynomolgus monkey for 21 mAbs (Group A). Bioavailability after sc injection (F), CL and serum/plasma concentration after iv injection of other published 19 mAbs (Group B) were predicted using the estimated geometric means of Q, Vc and Vp in Group A and the serum/plasma concentration after sc injection in Group B. 3. F and CL of 18 out of 19 mAbs in Group B were successfully predicted within 30% difference of observed value. Moreover, most of the observed serum/plasma concentrations after iv injection of mAbs in Group B were successfully predicted within 2-fold difference. Our approach suggests that iv injection might not be required to evaluate absorption of mAbs after sc injection in cynomolgus monkey. Therefore, our approach might reduce the time and cost of drug development, reduce the burden on resources, and also contribute to animal welfare.
Collapse
Affiliation(s)
- Kenta Haraya
- a Chugai Pharmabody Research Pte. Ltd , Synapse , Singapore
| | | | - Junichi Nezu
- a Chugai Pharmabody Research Pte. Ltd , Synapse , Singapore
| |
Collapse
|
19
|
Mould DR, D'Haens G, Upton RN. Clinical Decision Support Tools: The Evolution of a Revolution. Clin Pharmacol Ther 2016; 99:405-18. [PMID: 26785109 DOI: 10.1002/cpt.334] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/06/2016] [Accepted: 01/07/2016] [Indexed: 12/23/2022]
Abstract
Dashboard systems for clinical decision support integrate data from multiple sources. These systems, the newest in a long line of dose calculators and other decision support tools, utilize Bayesian approaches to fully individualize dosing using information gathered through therapeutic drug monitoring. In the treatment of inflammatory bowel disease patients with infliximab, dashboards may reduce therapeutic failures and treatment costs. The history and future development of modern Bayesian dashboard systems is described.
Collapse
Affiliation(s)
- D R Mould
- Projections Research Inc., Phoenixville, Pennsylvania, USA
| | - G D'Haens
- Inflammatory Bowel Disease Centre Academic Medical Centre 1105 AZ, Amsterdam, The Netherlands
| | - R N Upton
- Projections Research Inc., Phoenixville, Pennsylvania, USA.,Australian Centre for Pharmacometrics and Sansom Institute, School of Pharmacy and Medical Sciences, University of South Australia, South Australia, Australia
| |
Collapse
|
20
|
Igawa T, Haraya K, Hattori K. Sweeping antibody as a novel therapeutic antibody modality capable of eliminating soluble antigens from circulation. Immunol Rev 2016; 270:132-51. [DOI: 10.1111/imr.12392] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tomoyuki Igawa
- Research Division; Chugai Pharmaceutical Co. Ltd.; Shizuoka Japan
| | - Kenta Haraya
- Chugai Pharmabody Research Pte. Ltd.; Synapse Singapore
| | - Kunihiro Hattori
- Research Division; Chugai Pharmaceutical Co. Ltd.; Shizuoka Japan
| |
Collapse
|
21
|
Sheldon E, Schwickart M, Li J, Kim K, Crouch S, Parveen S, Kell C, Birrell C. Pharmacokinetics, Pharmacodynamics, and Safety of MEDI4212, an Anti-IgE Monoclonal Antibody, in Subjects with Atopy: A Phase I Study. Adv Ther 2016; 33:225-51. [PMID: 26843086 DOI: 10.1007/s12325-016-0287-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Indexed: 01/15/2023]
Abstract
INTRODUCTION The anti-IgE therapy omalizumab is currently licensed for the treatment of moderate to severe allergic asthma and chronic idiopathic urticaria. Owing to limitations in the use of omalizumab, a need exists for optimized anti-IgE therapies to broaden clinical indications and patient populations, and to improve dosing schedules. The objective of this phase I, randomized, placebo/omalizumab-controlled, first-in-human, dose-escalation study was to evaluate the pharmacokinetics, pharmacodynamics, and safety of the high-affinity, anti-IgE therapy MEDI4212 in non-Japanese and Japanese subjects with atopy and/or diagnostic IgE ≥ 30 IU/mL. METHODS Subjects with atopy and/or baseline IgE ≥ 30 IU/mL were randomized to a single dose of subcutaneous (5, 15, 60, 150, or 300 mg) or intravenous (300 mg) MEDI4212, subcutaneous omalizumab, or placebo. Following administration, pharmacokinetic, pharmacodynamic [IgE (free and total), and cellular FcεRI expression], and safety assessments were made. RESULTS MEDI4212 rapidly suppressed free serum IgE to a greater extent than omalizumab; however, recovery of free IgE to baseline in MEDI4212-dosed subjects was rapid when compared with the slow and gradual recovery seen in omalizumab-dosed individuals. The loss of IgE suppression corresponded with a rapid decrease of serum MEDI4212. FcεRI expression on dendritic cells and basophils was reduced following MEDI4212 dosing. MEDI4212 was well tolerated by subjects; adverse events were generally of low severity and no subjects discontinued due to adverse events. CONCLUSIONS The increased potency of MEDI4212 may be of clinical interest for individuals with high-diagnostic IgE levels where more extensive IgE suppression is required for clinical response. However, the modest duration of free IgE suppression below the target concentration noted with MEDI4212 in this study suggests limited potential for dosing schedule advantages over omalizumab. FUNDING MedImmune. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT01544348.
Collapse
Affiliation(s)
| | | | - Jing Li
- MedImmune, Mountain View, CA, USA
| | | | | | | | | | | |
Collapse
|
22
|
Haraya K, Tachibana T, Iwayanagi Y, Maeda A, Ozeki K, Nezu J, Ishigai M, Igawa T. PK/PD analysis of a novel pH-dependent antigen-binding antibody using a dynamic antibody-antigen binding model. Drug Metab Pharmacokinet 2016; 31:123-32. [PMID: 26944099 DOI: 10.1016/j.dmpk.2015.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/01/2015] [Accepted: 12/21/2015] [Indexed: 01/26/2023]
Abstract
Previously, we have reported novel engineered antibody with pH-dependent antigen-binding (recycling antibody), and with both pH-dependent antigen-binding and increased FcRn-binding at neutral pH (sweeping antibody). The purpose of this study is to perform PK/PD predictions to better understand the potential applications of the antibodies as therapeutics. To demonstrate the applicability of recycling and sweeping antibodies over conventional antibodies, PK/PD analyses were performed. PK/PD parameters for antibody and antigen dynamics were estimated from the results of a pharmacokinetic study in human FcRn transgenic mice. A simulation study was performed using the estimated PK/PD parameters with various target antigen profiles. In comparison to conventional antibody, recycling antibody enhanced antibody-antigen complex clearance by 3 folds, while sweeping antibody accelerated antigen clearance by 10 folds in a pharmacokinetic study. Simulation results showed that recycling and sweeping antibodies can improve dosage frequency and reduce the required dose for target antigens with various clearances, plasma concentrations or binding kinetics. Moreover, importance of the association rate constant to enhance the beneficial effect of antibodies was shown. These results support the conclusion that recycling and sweeping antibodies can be applied to various target antigens with different profiles, and expand the number of antigens that antibodies can target.
Collapse
Affiliation(s)
- Kenta Haraya
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #04-11 to 17 Synapse, 138623, Singapore.
| | - Tatsuhiko Tachibana
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #04-11 to 17 Synapse, 138623, Singapore
| | - Yuki Iwayanagi
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Atsuhiko Maeda
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Kazuhisa Ozeki
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Junichi Nezu
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #04-11 to 17 Synapse, 138623, Singapore
| | - Masaki Ishigai
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Tomoyuki Igawa
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| |
Collapse
|
23
|
Mould DR, Dubinsky MC. Dashboard systems: Pharmacokinetic/pharmacodynamic mediated dose optimization for monoclonal antibodies. J Clin Pharmacol 2015; 55 Suppl 3:S51-9. [PMID: 25707964 DOI: 10.1002/jcph.370] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/18/2014] [Accepted: 07/18/2014] [Indexed: 12/15/2022]
Abstract
Many marketed drugs exhibit high variability in exposure and response. While these drugs are efficacious in their approved indications, finding appropriate dose regimens for individual patients is not straightforward. Similar dose adjustment problems are also seen with drugs that have a complex relationship between exposure and response and/or a narrow therapeutic window. This is particularly true for monoclonal antibodies, where prolonged dosing at a sub-therapeutic dose can also elicit anti-drug antibodies which will further compromise safety and efficacy. Thus, finding appropriate doses quickly would represent a substantial improvement in healthcare. Dashboard systems, which are decision-support tools, offer an improved, convenient means of tailoring treatment for individual patients. This article reviews the clinical need for this approach, particularly with monoclonal antibodies, the design, development, and testing of such systems, and the likely benefits of dashboard systems in clinical practice. We focus on infliximab for reference.
Collapse
|
24
|
Chirumbolo S. Immunotherapy in allergy and cellular tests: state of art. Hum Vaccin Immunother 2014; 10:1595-610. [PMID: 24717453 PMCID: PMC5396242 DOI: 10.4161/hv.28592] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/10/2014] [Accepted: 03/18/2014] [Indexed: 12/13/2022] Open
Abstract
The basophil activation test (BAT) is an in vitro assay where the activation of basophils upon exposure to various IgE-challenging molecules is measured by flow cytometry. It is a cellular test able to investigate basophil behavior during allergy and allergy immunotherapy. A panoply of critical issues and suggestive advances have rendered this assay a promising yet puzzling tool to endeavor a full comprehension of innate immunity of allergy desensitization and manage allergen or monoclonal anti-IgE therapy. In this review a brief state of art of BAT in immunotherapy is described focusing onto the analytical issue pertaining BAT performance in allergy specific therapy.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Medicine; University of Verona; Verona, Italy
- Laboratory of Physiopathology of Obesity; Depertment of Medicine-University of Verona; LURM Est Policlinico GB Rossi; Verona, Italy
| |
Collapse
|
25
|
Chirumbolo S, Olivieri M. Increase in human basophils IgE-mediated stimulation by omalizumab: A role for membrane FcγRs? J Allergy Clin Immunol 2014; 133:1493-4. [DOI: 10.1016/j.jaci.2013.12.1094] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/05/2013] [Indexed: 12/18/2022]
|