1
|
Prathapan V, Eipert P, Wigger N, Kipp M, Appali R, Schmitt O. Modeling and simulation for prediction of multiple sclerosis progression. Comput Biol Med 2024; 175:108416. [PMID: 38657465 DOI: 10.1016/j.compbiomed.2024.108416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
In light of extensive work that has created a wide range of techniques for predicting the course of multiple sclerosis (MS) disease, this paper attempts to provide an overview of these approaches and put forth an alternative way to predict the disease progression. For this purpose, the existing methods for estimating and predicting the course of the disease have been categorized into clinical, radiological, biological, and computational or artificial intelligence-based markers. Weighing the weaknesses and strengths of these prognostic groups is a profound method that is yet in need and works directly at the level of diseased connectivity. Therefore, we propose using the computational models in combination with established connectomes as a predictive tool for MS disease trajectories. The fundamental conduction-based Hodgkin-Huxley model emerged as promising from examining these studies. The advantage of the Hodgkin-Huxley model is that certain properties of connectomes, such as neuronal connection weights, spatial distances, and adjustments of signal transmission rates, can be taken into account. It is precisely these properties that are particularly altered in MS and that have strong implications for processing, transmission, and interactions of neuronal signaling patterns. The Hodgkin-Huxley (HH) equations as a point-neuron model are used for signal propagation inside a small network. The objective is to change the conduction parameter of the neuron model, replicate the changes in myelin properties in MS and observe the dynamics of the signal propagation across the network. The model is initially validated for different lengths, conduction values, and connection weights through three nodal connections. Later, these individual factors are incorporated into a small network and simulated to mimic the condition of MS. The signal propagation pattern is observed after inducing changes in conduction parameters at certain nodes in the network and compared against a control model pattern obtained before the changes are applied to the network. The signal propagation pattern varies as expected by adapting to the input conditions. Similarly, when the model is applied to a connectome, the pattern changes could give an insight into disease progression. This approach has opened up a new path to explore the progression of the disease in MS. The work is in its preliminary state, but with a future vision to apply this method in a connectome, providing a better clinical tool.
Collapse
Affiliation(s)
- Vishnu Prathapan
- Medical School Hamburg University of Applied Sciences and Medical University, Am Kaiserkai 1, 20457, Hamburg, Germany.
| | - Peter Eipert
- Medical School Hamburg University of Applied Sciences and Medical University, Am Kaiserkai 1, 20457, Hamburg, Germany.
| | - Nicole Wigger
- Department of Anatomy, University of Rostock Gertrudenstr 9, 18057, Rostock, Germany.
| | - Markus Kipp
- Department of Anatomy, University of Rostock Gertrudenstr 9, 18057, Rostock, Germany.
| | - Revathi Appali
- Institute of General Electrical Engineering, University of Rostock, Albert-Einstein-Straße 2, 18059, Rostock, Germany; Department of Aging of Individuals and Society, Interdisciplinary Faculty, University of Rostock, Universitätsplatz 1, 18055, Rostock, Germany.
| | - Oliver Schmitt
- Medical School Hamburg University of Applied Sciences and Medical University, Am Kaiserkai 1, 20457, Hamburg, Germany; Department of Anatomy, University of Rostock Gertrudenstr 9, 18057, Rostock, Germany.
| |
Collapse
|
2
|
Benito‐Villalvilla C, de la Rocha‐Muñoz A, López‐Abente J, Eggel A, Bottoli I, Severin T, Woisetschläger M, Palomares O. Ligelizumab impairs
IgE
‐binding to plasmacytoid dendritic cells more potently than omalizumab and restores
IFN
‐α production and
FOXP3
+
Treg generation. Allergy 2022; 78:1060-1072. [PMID: 36315052 DOI: 10.1111/all.15567] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Ligelizumab is an anti-IgE monoclonal antibody binding IgE with higher affinity than omalizumab that is under clinical investigation for several IgE-mediated diseases. We previously showed that omalizumab removes IgE bound to FcεRI on plasmacytoid dendritic cells (pDCs) and restores their ability to produce IFN-α and regulatory T cells (Tregs). The aim of this work is to investigate the capacity of ligelizumab to regulate functional properties of pDCs in comparison with omalizumab. METHODS pDCs were isolated from atopic donors and IgE was detached from FcεRI on pDCs with designed ankyrin repeat protein (DARPin) bi53-79. pDCs were resensitized with IgE alone or in the presence of ligelizumab or omalizumab prior to IgE-FcεRI crosslinking and Toll-like receptor 9 (TLR9) stimulation. Flow cytometry, ELISA, coculture experiments and intranuclear staining were performed to determine cytokine production and Treg generation. An antigen-specific model of resensitization and IgE-crosslinking was also performed. RESULTS The levels of serum total free IgE show a non-linear positive correlation with the frequency of IgE+ pDCs displaying IgE bound to FcεRI within the 43 individual donors included in the study. Ligelizumab displays stronger capacity than omalizumab to block the binding of free IgE to FcεRI on human pDCs, resulting in a greater restoration of TLR9-L-induced IFN-α production. Ligelizumab also restores the ability of pDCs to generate FOXP3+ Tregs as previously reported for omalizumab. CONCLUSIONS The uncovered novel molecular mechanisms of ligelizumab to regulate functional properties of pDCs from atopic donors might have important clinical implications for anti-IgE treatments in different IgE-mediated diseases.
Collapse
Affiliation(s)
| | - Andrés de la Rocha‐Muñoz
- Department of Biochemistry and Molecular Biology School of Chemistry Complutense University Madrid Spain
- Autonomous University of Madrid Madrid Spain
| | - Jacobo López‐Abente
- Department of Biochemistry and Molecular Biology School of Chemistry Complutense University Madrid Spain
| | - Alexander Eggel
- Department of BioMedical Research University of Bern Bern Switzerland
- Department of Rheumatology and Immunology University Hospital of Bern Bern Switzerland
| | | | | | | | - Oscar Palomares
- Department of Biochemistry and Molecular Biology School of Chemistry Complutense University Madrid Spain
| |
Collapse
|
3
|
Ménochet K, Yu H, Wang B, Tibbitts J, Hsu CP, Kamath AV, Richter WF, Baumann A. Non-human primates in the PKPD evaluation of biologics: Needs and options to reduce, refine, and replace. A BioSafe White Paper. MAbs 2022; 14:2145997. [PMID: 36418217 PMCID: PMC9704389 DOI: 10.1080/19420862.2022.2145997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
Monoclonal antibodies (mAbs) deliver great benefits to patients with chronic and/or severe diseases thanks to their strong specificity to the therapeutic target. As a result of this specificity, non-human primates (NHP) are often the only preclinical species in which therapeutic antibodies cross-react with the target. Here, we highlight the value and limitations that NHP studies bring to the design of safe and efficient early clinical trials. Indeed, data generated in NHPs are integrated with in vitro information to predict the concentration/effect relationship in human, and therefore the doses to be tested in first-in-human trials. The similarities and differences in the systems defining the pharmacokinetics and pharmacodynamics (PKPD) of mAbs in NHP and human define the nature and the potential of the preclinical investigations performed in NHPs. Examples have been collated where the use of NHP was either pivotal to the design of the first-in-human trial or, inversely, led to the termination of a project prior to clinical development. The potential impact of immunogenicity on the results generated in NHPs is discussed. Strategies to optimize the use of NHPs for PKPD purposes include the addition of PD endpoints in safety assessment studies and the potential re-use of NHPs after non-terminal studies or cassette dosing several therapeutic agents of interest. Efforts are also made to reduce the use of NHPs in the industry through the use of in vitro systems, alternative in vivo models, and in silico approaches. In the case of prediction of ocular PK, the body of evidence gathered over the last two decades renders the use of NHPs obsolete. Expert perspectives, advantages, and pitfalls with these alternative approaches are shared in this review.
Collapse
Affiliation(s)
| | - Hongbin Yu
- R&D Project Management and Development Strategies, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Bonnie Wang
- Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Inc, Princeton, NJ, USA
| | - Jay Tibbitts
- Nonclinical Development, South San Francisco, CA, USA
| | - Cheng-Pang Hsu
- Preclinical Development and Clinical Pharmacology, AskGene Pharma Inc, Camarillo, CA, USA
| | - Amrita V. Kamath
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, South San Francisco, CA, USA
| | - Wolfgang F. Richter
- Roche Pharma Research and Early Development, Roche Innovation, Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Andreas Baumann
- R&D, Bayer Pharma AG, Berlin, Germany & Non-clinical Biotech Consulting, Potsdam, Germany °(° present affiliation)
| |
Collapse
|
4
|
Wang Y, Pan D, Huang C, Chen B, Li M, Zhou S, Wang L, Wu M, Wang X, Bian Y, Yan J, Liu J, Yang M, Miao L. Dose escalation PET imaging for safety and effective therapy dose optimization of a bispecific antibody. MAbs 2021; 12:1748322. [PMID: 32275842 PMCID: PMC7153848 DOI: 10.1080/19420862.2020.1748322] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Selecting the dose for efficacy and first-in-human studies of bispecific antibodies (BsAbs) is a challenging process. Herein, positron emission tomography (PET) imaging with 89Zr-labeled IBI322, an anti-CD47/PD-L1 BsAb, was used to optimize the safety and effective therapy dose. By labeling with 89Zr, we aimed to assess the pharmacokinetics (PK), safety, and target engagement of IBI322 with dose escalation dynamic PET imaging in humanized transgenic animal models bearing MC38 tumors (knock-in of hCD47 and hPDL1). 89Zr-labeled IBI322 specifically accumulated in tumors with a tumor-to-muscle ratio of 12.37 ± 1.42 at 168 h (0.22 mg/kg) and the biodistribution of normal tissues from PET imaging could be used for preliminary safety prediction. According to the Pearson correlation analysis between the ELISA-quantified serum concentration and heart uptake (%ID/g) (r = 0.980), a modified Patlak model was proposed. The exploratory target-mediated 50% (0.38 mg/kg) and 90% (0.63 mg/kg) inhibitory mass doses were calculated with the current modified Patlak model. The preliminary pharmacodynamics (PD) study with 0.34 mg/kg revealed that the dose prediction was rational. In conclusion, dose escalation PET imaging with 89Zr-labeled antibodies is promising for PK/PD modeling and safety prediction, and helpful for determining rational dosing for preclinical and clinical trials of BsAbs.
Collapse
Affiliation(s)
- Yan Wang
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Donghui Pan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Chenrong Huang
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Bingliang Chen
- Drug Discovery Department, Innovent Biopharmaceutical (Suzhou) Co., Ltd, Suzhou, China
| | - Mingzhu Li
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Shuaixiang Zhou
- Drug Discovery Department, Innovent Biopharmaceutical (Suzhou) Co., Ltd, Suzhou, China
| | - Lizhen Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Min Wu
- Drug Discovery Department, Innovent Biopharmaceutical (Suzhou) Co., Ltd, Suzhou, China
| | - Xinyu Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yicong Bian
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Junjie Yan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Junjian Liu
- Drug Discovery Department, Innovent Biopharmaceutical (Suzhou) Co., Ltd, Suzhou, China
| | - Min Yang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Liyan Miao
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
5
|
Zhu J, Beechinor RJ, Thompson T, Schorzman AN, Zamboni W, Crona DJ, Weiner DL, Tarantino LM. Pharmacokinetic and pharmacodynamic analyses of cocaine and its metabolites in behaviorally divergent inbred mouse strains. GENES, BRAIN, AND BEHAVIOR 2021; 20:e12666. [PMID: 32383297 PMCID: PMC7941260 DOI: 10.1111/gbb.12666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 11/26/2022]
Abstract
Cocaine (COC) is a psychostimulant with a high potential for abuse and addiction. Risk for COC use disorder is driven, in part, by genetic factors. Animal models of addiction-relevant behaviors have proven useful for studying both genetic and nongenetic contributions to drug response. In a previous study, we examined initial locomotor sensitivity to COC in genetically diverse inbred mouse strains. That work highlighted the relevance of pharmacokinetics (PK) in initial locomotor response to COC but was limited by a single dose and two sampling points. The objective of the present study was to characterize the PK and pharmacodynamics of COC and its metabolites (norcocaine and benzoylecgonine) in six inbred mouse strains (I/LnJ, C57BL/6J, FVB/NJ, BTBR T+ tf/J, LG/J and LP/J) that exhibit extreme locomotor responses to cocaine. Mice were administered COC at one of four doses and concentrations of cocaine, norcocaine and benzoylecgonine were analyzed in both plasma and brain tissue at 5 different time points. Initial locomotor sensitivity to COC was used as a pharmacodynamic endpoint. We developed an empirical population PK model that simultaneously characterizes cocaine, norcocaine and benzoylecgonine in plasma and brain tissues. We observed interstrain variability occurring in the brain compartment that may contribute to pharmacodynamic differences among select strains. Our current work paves the way for future studies to explore strain-specific pharmacokinetic differences and identify factors other than PK that are responsible for the diverse behavioral response to COC across these inbred mouse strains.
Collapse
Affiliation(s)
- Jing Zhu
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ryan J Beechinor
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
- UC Davis Comprehensive Cancer Center, Davis, California, USA
| | - Trey Thompson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - William Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Daniel J Crona
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
- Department of Pharmacy, UNC Hospitals and Clinics, Chapel Hill, North Carolina, USA
| | - Daniel L Weiner
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lisa M Tarantino
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
6
|
Ahmed S, Ellis M, Li H, Pallucchini L, Stein AM. Guiding dose selection of monoclonal antibodies using a new parameter (AFTIR) for characterizing ligand binding systems. J Pharmacokinet Pharmacodyn 2019; 46:287-304. [PMID: 31037615 DOI: 10.1007/s10928-019-09638-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 04/16/2019] [Indexed: 01/07/2023]
Abstract
Guiding the dose selection for monoclonal antibody oncology drugs is often done using methods for predicting the receptor occupancy of the drug in the tumor. In this manuscript, previous work on characterizing target inhibition at steady state using the AFIR metric (Stein and Ramakrishna in CPT Pharmacomet Syst Pharmacol 6(4):258-266, 2017) is extended to include a "target-tissue" compartment and the shedding of membrane-bound targets. A new potency metric average free tissue target to initial target ratio (AFTIR) at steady state is derived, and it depends on only four key quantities: the equilibrium binding constant, the fold-change in target expression at steady state after binding to drug, the biodistribution of target from circulation to target tissue, and the average drug concentration in circulation. The AFTIR metric is useful for guiding dose selection, for efficiently performing sensitivity analyses, and for building intuition for more complex target mediated drug disposition models. In particular, reducing the complex, physiological model to four key parameters needed to predict target inhibition helps to highlight specific parameters that are the most important to estimate in future experiments to guide drug development.
Collapse
Affiliation(s)
- Sameed Ahmed
- Department of Applied Mathematics, University of Waterloo, Waterloo, Canada
| | - Miandra Ellis
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, USA
| | - Hongshan Li
- Department of Mathematics, Purdue University, Lafayette, USA
| | | | - Andrew M Stein
- Novartis Institute for BioMedical Research, 45 Sidney St., Cambridge, MA, 02140, USA.
| |
Collapse
|
7
|
An LC–MS/MS approach to assess total and free protein target in the serum of cynomolgus monkey. Bioanalysis 2019; 11:393-406. [DOI: 10.4155/bio-2018-0294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: Develop LC–MS/MS-based assays to measure total and free complement C5 in cynomolgus monkey serum as a target engagement biomarker for pharmacokinetic/pharmacodynamic correlation study. Materials & methods/results: The C5-specific signature peptide derived from pellet digestion of serum proteins with and without prior immunodepletion of the drug-bound C5 by protein A beads was quantified to assess free and total C5 levels, respectively. Conditions for immunodepletion by protein A were optimized to ensure complete depletion of IgGs (and drug-bound C5). The effect of sample dilution on drug-target dissociation and thus free C5 measurement was evaluated by applying a mathematical simulation. Conclusion: The procedure described here allows for the assessment of protein target engagement, aiding in pharmacokinetic/pharmacodynamic correlation analysis and human dose projection.
Collapse
|
8
|
Eckl-Dorna J, Villazala-Merino S, Linhart B, Karaulov AV, Zhernov Y, Khaitov M, Niederberger-Leppin V, Valenta R. Allergen-Specific Antibodies Regulate Secondary Allergen-Specific Immune Responses. Front Immunol 2019; 9:3131. [PMID: 30705676 PMCID: PMC6344431 DOI: 10.3389/fimmu.2018.03131] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/18/2018] [Indexed: 01/08/2023] Open
Abstract
Immunoglobulin E (IgE)-associated allergy is the most common immunologically-mediated hypersensensitivity disease. It is based on the production of IgE antibodies and T cell responses against per se innocuous antigens (i.e., allergens) and subsequent allergen-induced inflammation in genetically pre-disposed individuals. While allergen exposure in sensitized subjects mainly boosts IgE production and T cell activation, successful allergen-specific immunotherapy (AIT) induces the production of allergen-specific IgG antibodies and reduces T cell activity. Under both circumstances, the resulting allergen-antibody complexes play a major role in modulating secondary allergen-specific immune responses: Allergen-IgE complexes induce mast cell and basophil activation and perpetuate allergen-specific T cell responses via presentation of allergen by allergen presenting cells to T cells, a process called IgE-facilitated antigen presentation (FAP). In addition, they may induce activation of IgE memory B cells. Allergen-induced production of specific IgGs usually exerts ameliorating effects but under certain circumstances may also contribute to exacerbation. Allergen-specific IgG antibodies induced by AIT which compete with IgE for allergen binding (i.e., blocking IgG) inhibit formation of IgE-allergen complexes and reduce activation of effector cells, B cells and indirectly T cells as FAP is prevented. Experimental data provide evidence that by binding of allergen-specific IgG to epitopes different from those recognized by IgE, allergen-specific IgG may enhance IgE-mediated activation of mast cells, basophils and allergen-specific IgE+ B cells. In this review we provide an overview about the role of allergen-specific antibodies in regulating secondary allergen-specific immune responses.
Collapse
Affiliation(s)
- Julia Eckl-Dorna
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | | | - Birgit Linhart
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Alexander V Karaulov
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Yury Zhernov
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia
| | - Musa Khaitov
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia
| | | | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia.,NRC Institute of Immunology FMBA of Russia, Moscow, Russia
| |
Collapse
|
9
|
Explaining Interpatient Variability in Adalimumab Pharmacokinetics in Patients With Crohn's Disease. Ther Drug Monit 2018. [DOI: 10.1097/ftd.0000000000000494] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
10
|
Stein AM, Ramakrishna R. AFIR: A Dimensionless Potency Metric for Characterizing the Activity of Monoclonal Antibodies. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:258-266. [PMID: 28375563 PMCID: PMC5397564 DOI: 10.1002/psp4.12169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/06/2016] [Accepted: 01/03/2017] [Indexed: 12/14/2022]
Abstract
For monoclonal antibody (mAb) drugs, soluble targets may accumulate several thousand fold after binding to the drug. Time course data of mAb and total target is often collected and, although free target is more closely related to clinical effect, it is difficult to measure. Therefore, mathematical models of this data are used to predict target engagement. In this article, a “potency factor” is introduced as an approximation for the model‐predicted target inhibition. This potency factor is defined to be the time‐Averaged Free target concentration to Initial target concentration Ratio (AFIR), and it depends on three key quantities: the average drug concentration at steady state; the binding affinity; and the degree of target accumulation. AFIR provides the intuition for how changes in dosing regimen and binding affinity affect target capture and AFIR can be used to predict the druggability of new targets and the expected benefits of more potent, second‐generation mAbs.
Collapse
Affiliation(s)
- A M Stein
- Novartis Institute for BioMedical Research, Cambridge, Massachusetts, USA
| | - R Ramakrishna
- Novartis Institute for BioMedical Research, Cambridge, Massachusetts, USA
| |
Collapse
|
11
|
Gericke J, Metz M, Ohanyan T, Weller K, Altrichter S, Skov PS, Falkencrone S, Brand J, Kromminga A, Hawro T, Church MK, Maurer M. Serum autoreactivity predicts time to response to omalizumab therapy in chronic spontaneous urticaria. J Allergy Clin Immunol 2017; 139:1059-1061.e1. [DOI: 10.1016/j.jaci.2016.07.047] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/10/2016] [Accepted: 07/26/2016] [Indexed: 10/20/2022]
|
12
|
Samineni D, Girish S, Li C. Impact of Shed/Soluble targets on the PK/PD of approved therapeutic monoclonal antibodies. Expert Rev Clin Pharmacol 2016; 9:1557-1569. [DOI: 10.1080/17512433.2016.1243055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
13
|
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
|
14
|
Gauvreau GM, Arm JP, Boulet LP, Leigh R, Cockcroft DW, Davis BE, Mayers I, FitzGerald JM, Dahlen B, Killian KJ, Laviolette M, Carlsten C, Lazarinis N, Watson RM, Milot J, Swystun V, Bowen M, Hui L, Lantz AS, Meiser K, Maahs S, Lowe PJ, Skerjanec A, Drollmann A, O'Byrne PM. Efficacy and safety of multiple doses of QGE031 (ligelizumab) versus omalizumab and placebo in inhibiting allergen-induced early asthmatic responses. J Allergy Clin Immunol 2016; 138:1051-1059. [PMID: 27185571 DOI: 10.1016/j.jaci.2016.02.027] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/11/2016] [Accepted: 02/10/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Omalizumab is an established anti-IgE therapy for the treatment of allergic diseases that prevents IgE from binding to its receptor. QGE031 is an investigational anti-IgE antibody that binds IgE with higher affinity than omalizumab. OBJECTIVE This study compared the effects of QGE031 with those of omalizumab on clinical efficacy, IgE levels, and FcεRI expression in a clinical model of allergic asthma. METHODS Thirty-seven patients with mild allergic asthma were randomized to subcutaneous omalizumab, placebo, or QGE031 at 24, 72, or 240 mg every 2 weeks for 10 weeks in a double-blind, parallel-group multicenter study. Inhaled allergen challenges and skin tests were conducted before dosing and at weeks 6, 12, and 18, and blood was collected until 24 weeks after the first dose. RESULTS QGE031 elicited a concentration- and time-dependent change in the provocative concentration of allergen causing a 15% decrease in FEV1 (allergen PC15) that was maximal and approximately 3-fold greater than that of omalizumab (P = .10) and 16-fold greater than that of placebo (P = .0001) at week 12 in the 240-mg cohort. Skin responses reached 85% suppression at week 12 in the 240-mg cohort and were maximal at week 18. The top doses of QGE031 consistently suppressed skin test responses among subjects but had a variable effect on allergen PC15 (2-fold to 500-fold change). QGE031 was well tolerated. CONCLUSION QGE031 has greater efficacy than omalizumab on inhaled and skin allergen responses in patients with mild allergic asthma. These data support the clinical development of QGE031 as a treatment of asthma.
Collapse
Affiliation(s)
- Gail M Gauvreau
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan P Arm
- Novartis Pharmaceuticals, East Hanover, and Novartis Pharma AG, Basel, Switzerland
| | - Louis-Philippe Boulet
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec City, Quebec, Canada
| | - Richard Leigh
- Departments of Medicine and Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Donald W Cockcroft
- Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Beth E Davis
- Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Irvin Mayers
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - J Mark FitzGerald
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Barbro Dahlen
- Karolinska Institutet, Department of Medicine, Karolinska University, Stockholm, Sweden
| | - Kieran J Killian
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michel Laviolette
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec City, Quebec, Canada
| | - Christopher Carlsten
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nikolaos Lazarinis
- Karolinska Institutet, Department of Medicine, Karolinska University, Stockholm, Sweden
| | - Richard M Watson
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Joanne Milot
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec City, Quebec, Canada
| | - Veronica Swystun
- Departments of Medicine and Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Miranda Bowen
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Linda Hui
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ann-Sofie Lantz
- Karolinska Institutet, Department of Medicine, Karolinska University, Stockholm, Sweden
| | - Karin Meiser
- Novartis Pharmaceuticals, East Hanover, and Novartis Pharma AG, Basel, Switzerland
| | | | - Philip J Lowe
- Novartis Pharmaceuticals, East Hanover, and Novartis Pharma AG, Basel, Switzerland
| | - Andrej Skerjanec
- Novartis Pharmaceuticals, East Hanover, and Novartis Pharma AG, Basel, Switzerland
| | - Anton Drollmann
- Novartis Pharmaceuticals, East Hanover, and Novartis Pharma AG, Basel, Switzerland.
| | - Paul M O'Byrne
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
15
|
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
|
16
|
Honma W, Gautier A, Paule I, Yamaguchi M, Lowe PJ. Ethnic sensitivity assessment of pharmacokinetics and pharmacodynamics of omalizumab with dosing table expansion. Drug Metab Pharmacokinet 2015; 31:173-84. [PMID: 27238573 DOI: 10.1016/j.dmpk.2015.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/07/2015] [Accepted: 12/21/2015] [Indexed: 11/15/2022]
Abstract
A three-part license expansion for omalizumab (Xolair(®)), humanized anti-IgE antibody, was recently made in Japan for paediatric use, additional higher doses and revised dosing frequency in allergic asthma. The dosing level and frequency of omalizumab are guided by a dosing table based on the total serum IgE and bodyweight. Nonlinear mixed-effect pharmacokinetic (PK) and pharmacodynamic (PD) modeling and simulation techniques described the binding between omalizumab and its target IgE. The population PKPD analysis was conducted using data from the nine studies included originally in the European application of dosing table expansion together with three Japanese clinical studies to assess the influence of the ethnicity. Statistically significant differences between the ethnic groups were detected. These were small, within or close to bioequivalence criteria. The model described the primary pharmacology in Caucasian and Japanese patients, both adult and paediatric, with simulations showing that the interplay between the clearance, volume and binding affinity parameters was such that there was no clinical impact of the Japanese ethnic differences on either drug PK or free IgE suppression and hence the required posology.
Collapse
Affiliation(s)
- Wataru Honma
- Novartis Pharma K.K., 1-23-1 Toranomon, Minato-ku, Tokyo 105-6333, Japan.
| | - Aurélie Gautier
- Novartis Pharma AG, Lichtstrasse 35, CH-4056 Basel, Switzerland.
| | - Ines Paule
- Novartis Pharma AG, Lichtstrasse 35, CH-4056 Basel, Switzerland.
| | - Masayuki Yamaguchi
- Novartis Pharma K.K., 1-23-1 Toranomon, Minato-ku, Tokyo 105-6333, Japan.
| | - Philip J Lowe
- Novartis Pharma AG, Lichtstrasse 35, CH-4056 Basel, Switzerland.
| |
Collapse
|
17
|
Deciphering the In Vivo Performance of a Monoclonal Antibody to Neutralize Its Soluble Target at the Site of Action in a Mouse Collagen-Induced Arthritis Model. Pharm Res 2015; 33:1040-9. [PMID: 26718954 DOI: 10.1007/s11095-015-1850-8] [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] [Received: 10/14/2015] [Accepted: 12/22/2015] [Indexed: 12/25/2022]
Abstract
PURPOSE Study the disposition and target-neutralization capability of an anti-interleukin-6 (IL-6) monoclonal antibody (mAb) at the joint in a mouse collagen-induced arthritis (CIA) model. METHODS A mechanistic pharmacokinetic/pharmacodynamic study was conducted in a mouse CIA model using CNTO 345, a rat anti-mouse IL-6 mAb, as model compound. The drug, total/free IL-6 concentrations in both serum and joint lavage fluid were quantitatively assessed and compared to those in the normal control mice. RESULTS CNTO 345 exhibited higher clearance and significantly higher joint lavage/serum ratio in the CIA mice than in the normal control mice. The mAb concentrations in the joint lavage are approximately proportional to the serum concentrations at all the time points being examined. Dosing of CNTO 345 led to sustained free IL-6 suppression in both serum and joint lavage in a dose-dependent manner. A dose-dependent increase in total IL-6 was observed in serum, but not in the joint lavage fluid. Though no change in disease activity was observed following a single dose of anti-IL-6 mAb at peak of the disease, a dose-dependent decrease in serum amyloid A, a downstream biomarker of IL-6, was observed. CONCLUSIONS This study provided quantitative assessments of the distribution and target-neutralization capability of an anti-IL-6 mAb at the site of action in an animal disease model.
Collapse
|
18
|
Tibbitts J, Canter D, Graff R, Smith A, Khawli LA. Key factors influencing ADME properties of therapeutic proteins: A need for ADME characterization in drug discovery and development. MAbs 2015; 8:229-45. [PMID: 26636901 PMCID: PMC4966629 DOI: 10.1080/19420862.2015.1115937] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Protein therapeutics represent a diverse array of biologics including antibodies, fusion proteins, and therapeutic replacement enzymes. Since their inception, they have revolutionized the treatment of a wide range of diseases including respiratory, vascular, autoimmune, inflammatory, infectious, and neurodegenerative diseases, as well as cancer. While in vivo pharmacokinetic, pharmacodynamic, and efficacy studies are routinely carried out for protein therapeutics, studies that identify key factors governing their absorption, distribution, metabolism, and excretion (ADME) properties have not been fully investigated. Thorough characterization and in-depth study of their ADME properties are critical in order to support drug discovery and development processes for the production of safer and more effective biotherapeutics. In this review, we discuss the main factors affecting the ADME characteristics of these large macromolecular therapies. We also give an overview of the current tools, technologies, and approaches available to investigate key factors that influence the ADME of recombinant biotherapeutic drugs, and demonstrate how ADME studies will facilitate their future development.
Collapse
|
19
|
Zheng S, McIntosh T, Wang W. Utility of free and total target measurements as target engagement and efficacy biomarkers in biotherapeutic development--opportunities and challenges. J Clin Pharmacol 2015; 55 Suppl 3:S75-84. [PMID: 25707966 DOI: 10.1002/jcph.357] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/27/2014] [Indexed: 01/09/2023]
Abstract
For biotherapeutics directed against soluble targets, most often monoclonal antibodies (mAbs), their therapeutic efficacy theoretically is driven by the magnitude and duration of free target suppression. However, for soluble targets of rapid turnover and low abundance, it can be technically challenging to directly measure the lowering of free target following treatment with biologics. The opportunities, challenges, and practical approaches to assess free and bound soluble targets and the utility of free and bound target measurements as biomarkers for target engagement and efficacy are covered in this review. In particular, case examples are presented to illustrate the interplay between drug and free/bound target, and how an integrated bioanalytical and pharmacokinetic/target engagement/pharmacodynamic (PK/TE/PD) modeling approach can be used to assess the target engagement for biologics directed against soluble targets with rapid turnover. Important caveats of the modeling approach in the absence of free target measurements are also discussed.
Collapse
Affiliation(s)
- Songmao Zheng
- Biologics Clinical Pharmacology, Janssen R&D, 1400 McKean Road, Spring House, PA, 19438, USA
| | | | | |
Collapse
|
20
|
Dua P, Hawkins E, van der Graaf PH. A Tutorial on Target-Mediated Drug Disposition (TMDD) Models. CPT Pharmacometrics Syst Pharmacol 2015; 4:324-37. [PMID: 26225261 PMCID: PMC4505827 DOI: 10.1002/psp4.41] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/07/2015] [Indexed: 12/16/2022] Open
Abstract
Target-mediated drug disposition (TMDD) is the phenomenon in which a drug binds with high affinity to its pharmacological target site (such as a receptor) to such an extent that this affects its pharmacokinetic characteristics.1 The aim of this Tutorial is to provide an introductory guide to the mathematical aspects of TMDD models for pharmaceutical researchers. Examples of Berkeley Madonna2 code for some models discussed in this Tutorial are provided in the Supplementary Materials.
Collapse
Affiliation(s)
- P Dua
- Pharmatherapeutics Research Clinical Pharmacology, Pfizer NeusentisCambridge, UK
| | - E Hawkins
- Pharmatherapeutics Research Clinical Pharmacology, Pfizer NeusentisCambridge, UK
- Department of Mathematics, University of SurreyGuildford, UK
| | - PH van der Graaf
- Leiden Academic Centre for Drug Research (LACDR), Systems PharmacologyLeiden, The Netherlands
| |
Collapse
|
21
|
Sundqvist M, Lundahl A, Någård MB, Bredberg U, Gennemark P. Quantifying and Communicating Uncertainty in Preclinical Human Dose-Prediction. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015. [PMID: 26225248 PMCID: PMC4429578 DOI: 10.1002/psp4.32] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human dose-prediction is fundamental for ranking lead-optimization compounds in drug discovery and to inform design of early clinical trials. This tutorial describes how uncertainty in such predictions can be quantified and efficiently communicated to facilitate decision-making. Using three drug-discovery case studies, we show how several uncertain pieces of input information can be integrated into one single uncomplicated plot with key predictions, including their uncertainties, for many compounds or for many scenarios, or both.
Collapse
Affiliation(s)
- M Sundqvist
- CVMD iMed DMPK, AstraZeneca R&D SE-431, 83, Mölndal, Sweden
| | - A Lundahl
- CVMD iMed DMPK, AstraZeneca R&D SE-431, 83, Mölndal, Sweden
| | - M B Någård
- CVMD iMed DMPK, AstraZeneca R&D SE-431, 83, Mölndal, Sweden
| | - U Bredberg
- CVMD iMed DMPK, AstraZeneca R&D SE-431, 83, Mölndal, Sweden
| | - P Gennemark
- CVMD iMed DMPK, AstraZeneca R&D SE-431, 83, Mölndal, Sweden
| |
Collapse
|
22
|
Jensen RK, Plum M, Tjerrild L, Jakob T, Spillner E, Andersen GR. Structure of the omalizumab Fab. ACTA CRYSTALLOGRAPHICA SECTION F-STRUCTURAL BIOLOGY COMMUNICATIONS 2015; 71:419-26. [PMID: 25849503 DOI: 10.1107/s2053230x15004100] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 02/26/2015] [Indexed: 11/10/2022]
Abstract
Omalizumab is a humanized anti-IgE antibody that inhibits the binding of IgE to its receptors on mast cells and basophils, thus blocking the IgE-mediated release of inflammatory mediators from these cells. Omalizumab binds to the Fc domains of IgE in proximity to the binding site of the high-affinity IgE receptor FcℇRI, but the epitope and the mechanisms and conformations governing the recognition remain unknown. In order to elucidate the molecular mechanism of its anti-IgE activity, the aim was to analyse the interaction of omalizumab with human IgE. Therefore, IgE Fc Cℇ2-4 was recombinantly produced in mammalian HEK-293 cells. Functionality of the IgE Fc was proven by ELISA and mediator-release assays. Omalizumab IgG was cleaved with papain and the resulting Fab was purified by ion-exchange chromatography. The complex of IgE Fc with omalizumab was prepared by size-exclusion chromatography. However, crystals containing the complex were not obtained, suggesting that the process of crystallization favoured the dissociation of the two proteins. Instead, two structures of the omalizumab Fab with maximum resolutions of 1.9 and 3.0 Å were obtained. The structures reveal the arrangement of the CDRs and the position of omalizumab residues known from prior functional studies to be involved in IgE binding. Thus, the structure of omalizumab provides the structural basis for understanding the function of omalizumab, allows optimization of the procedure for complex crystallization and poses questions about the conformational requirements for anti-IgE activity.
Collapse
Affiliation(s)
- Rasmus K Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wiedsvej 10C, 8000 Aarhus, Denmark
| | - Melanie Plum
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wiedsvej 10C, 8000 Aarhus, Denmark
| | - Luna Tjerrild
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wiedsvej 10C, 8000 Aarhus, Denmark
| | - Thilo Jakob
- Allergy Research Group, Department of Dermatology, University Freiburg Medical Center, Hauptstrasse 7, 79104 Freiburg, Germany
| | - Edzard Spillner
- Department of Engineering - Biotechnology, Aarhus University, Gustav Wiedsvej 10C, 8000 Aarhus, Denmark
| | - Gregers Rom Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wiedsvej 10C, 8000 Aarhus, Denmark
| |
Collapse
|
23
|
Penney M, Agoram B. At the bench: the key role of PK-PD modelling in enabling the early discovery of biologic therapies. Br J Clin Pharmacol 2015; 77:740-5. [PMID: 23962236 DOI: 10.1111/bcp.12225] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 07/26/2013] [Indexed: 12/14/2022] Open
Abstract
Pharmacokinetic-pharmacodynamic (PK-PD) modelling is already used extensively in pre-clinical and clinical drug development to characterize drug candidates quantitatively, aid go/no-go decisions and to inform future trial design and optimal dosing regimens. Less well known, although arguably as powerful, is its application at the earliest stages of drug development, at target selection and lead selection, where these same techniques can be used to predict and so bring forward drug candidates with the necessary characteristics or, for unachievable requirements, allow the abandonment of the programme for the minimum spend of time and cost. We consider three examples that illustrate the power of the application of modelling at this early stage. We start with the simple case of determining the optimal characteristics for a monoclonal antibody against a soluble ligand with its application to the investment decision for the development of best-in-class compounds. This is extended to the more complex situation of the target protein having an endogenous, inhibitory binding protein. We then illustrate how using physiologically-based pharmacokinetic modelling enables the appropriate engineering and testing of biological therapeutics for optimal PK-PD characteristics. These examples illustrate how a minimal investment in modelling achieves orders of magnitude better returns in choosing the correct targets, mechanism of action and candidate characteristics to progress to clinical trials, streamlining drug development and delivering better medicines to patients.
Collapse
Affiliation(s)
- Mark Penney
- Clinical Pharmacology & DMPK, MedImmune plc, Granta Park, Cambridge, CB21 6GH, UK
| | | |
Collapse
|
24
|
Sepp A, Berges A, Sanderson A, Meno-Tetang G. Development of a physiologically based pharmacokinetic model for a domain antibody in mice using the two-pore theory. J Pharmacokinet Pharmacodyn 2015; 42:97-109. [PMID: 25577033 DOI: 10.1007/s10928-014-9402-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 12/20/2014] [Indexed: 10/24/2022]
Abstract
Domain antibodies (dAbs) are the smallest antigen-binding fragments of immunoglobulins. To date, there is limited insight into the pharmacokinetics of dAbs, especially their distribution into tissues and elimination. The objective of this work was to develop a physiologically-based pharmacokinetic model to investigate the biodisposition of a non-specific dAb construct in mice. Following a single IV administration of 10 mg/kg dummy dAb protein to twenty four female mice, frequent blood samples were collected and whole body lateral sections were analyzed by quantitative whole-body autoradiography. The model is based on the two-pore hypothesis of extravasation where organ-specific isogravimetric flow rates (Jorg,ISO) and permeability-surface area products (PSorg) are expressed as linear functions of the lymph flow rate (Jorg) and the kidney compartment is modified to account for glomerular filtration of dAb. As a result, only Jorg, glomerular filtration coefficient and the combined volume of Bowman's capsule, proximal and distal renal tubules and loop of Henle were optimized by fitting simultaneously all blood and organ data to the model. Our model captures the pharmacokinetic profiles of dAb in blood and all organs and shows that extravasation into interstitial space is a predominantly diffusion-driven process. The parameter values were estimated with good precision (%RMSE ≈ 30) and low cross-correlation (R(2) < 0.2). We developed a flexible model with a limited parameter number that may be applied to other biotherapeutics after adapting for size-related effects on extravasation and renal elimination processes.
Collapse
Affiliation(s)
- Armin Sepp
- BioPharm Innovation, Biopharm R&D, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | | | | | | |
Collapse
|
25
|
Lowe PJ, Georgiou P, Canvin J. Revision of omalizumab dosing table for dosing every 4 instead of 2 weeks for specific ranges of bodyweight and baseline IgE. Regul Toxicol Pharmacol 2014; 71:68-77. [PMID: 25497995 DOI: 10.1016/j.yrtph.2014.12.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 11/26/2022]
Abstract
The dosing level and frequency of omalizumab are guided by a dosing table based on total serum immunoglobulin E (IgE) and bodyweight. Using a validated, mathematical simulation model (based on concentration data from 8 studies), we evaluated the impact of a revised omalizumab dosing table (every 4 weeks dosing regimen) on the pharmacokinetic and pharmacodynamic profiles of free and total IgE. Safety analysis, in patients with high levels of exposure to omalizumab, was done using data from the clinical and post-marketing databases. The model accurately predicted observed omalizumab, free and total IgE concentrations. After reaching steady-state, the average increase in exposure was 10%, even for patients with the highest concentrations at the upper 97.5th percentile. Free IgE suppression slightly increased in the initial phase, and slightly reduced at the trough of the dosing cycle, but average suppression remained similar for both regimens. The safety profile of omalizumab was similar for patients receiving higher or lower doses. Thus, doubling the dose of omalizumab, in a subset of patients receiving 225-300 mg of omalizumab (every 2 weeks dosing regimen) can efficiently suppress free IgE without compromising safety or efficacy.
Collapse
Affiliation(s)
| | | | - Janice Canvin
- Novartis Pharmaceuticals UK Limited, Horsham, West Sussex, UK
| |
Collapse
|
26
|
Arm JP, Bottoli I, Skerjanec A, Floch D, Groenewegen A, Maahs S, Owen CE, Jones I, Lowe PJ. Pharmacokinetics, pharmacodynamics and safety of QGE031 (ligelizumab), a novel high-affinity anti-IgE antibody, in atopic subjects. Clin Exp Allergy 2014; 44:1371-85. [PMID: 25200415 PMCID: PMC4278557 DOI: 10.1111/cea.12400] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 08/05/2014] [Accepted: 08/19/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Using a monoclonal antibody with greater affinity for IgE than omalizumab, we examined whether more complete suppression of IgE provided greater pharmacodynamic effects, including suppression of skin prick responses to allergen. OBJECTIVE To explore the pharmacokinetics, pharmacodynamics and safety of QGE031 (ligelizumab), a novel high-affinity humanized monoclonal IgG1κ anti-IgE. METHODS Preclinical assessments and two randomized, placebo-controlled, double-blind clinical trials were conducted in atopic subjects. The first trial administered single doses of QGE031 (0.1-10 mg/kg) or placebo intravenously, while the second trial administered two to four doses of QGE031 (0.2- 4 mg/kg) or placebo subcutaneously at 2-week intervals. Both trials included an open-label omalizumab arm. RESULTS Sixty of 73 (82%) and 96 of 110 (87%) subjects completed the intravenous and subcutaneous studies, respectively. Exposure to QGE031 and its half-life depended on the QGE031 dose and serum IgE level. QGE031 had a biexponential pharmacokinetic profile after intravenous administration and a terminal half-life of approximately 20 days. QGE031 demonstrated dose- and time-dependent suppression of free IgE, basophil FcεRI and basophil surface IgE superior in extent (free IgE and surface IgE) and duration to omalizumab. At Day 85, 6 weeks after the last dose, skin prick wheal responses to allergen were suppressed by > 95% and 41% in subjects treated subcutaneously with QGE031 (2 mg/kg) or omalizumab, respectively (P < 0.001). Urticaria was observed in QGE031- and placebo-treated subjects and was accompanied by systemic symptoms in one subject treated with 10 mg/kg intravenous QGE031. There were no serious adverse events. CONCLUSION AND CLINICAL RELEVANCE These first clinical data for QGE031, a high-affinity IgG1κ anti-IgE, demonstrate that increased suppression of free IgE compared with omalizumab translated to superior pharmacodynamic effects in atopic subjects, including those with high IgE levels. QGE031 may therefore benefit patients unable to receive, or suboptimally treated with, omalizumab.
Collapse
Affiliation(s)
- J P Arm
- Translational Medicine, Novartis Institute for Biomedical ResearchCambridge, MA, USA
| | - I Bottoli
- Primary Care, Novartis Pharma AGBasel, Switzerland
| | - A Skerjanec
- Preclinical Safety, Novartis Institute for Biomedical ResearchBasel, Switzerland
| | - D Floch
- Preclinical Safety, Novartis Institute for Biomedical ResearchBasel, Switzerland
| | - A Groenewegen
- Biomarker Development, Novartis Pharma AGBasel, Switzerland
| | - S Maahs
- Clinical Sciences and Innovation, Novartis Institute for Biomedical ResearchEast Hanover, NJ, USA
| | - C E Owen
- Novartis Institute for Biomedical ResearchHorsham, West Sussex, UK
| | - I Jones
- NIBR Biometrics and Statistical Science, Novartis Pharma AGBasel, Switzerland
| | - P J Lowe
- Advanced Quantitative Sciences, Novartis Pharma AGBasel, Switzerland
| |
Collapse
|
27
|
Krause A, Lowe PJ. Visualization and communication of pharmacometric models with berkeley madonna. CPT Pharmacometrics Syst Pharmacol 2014; 3:e116. [PMID: 24872204 PMCID: PMC4055786 DOI: 10.1038/psp.2014.13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 03/27/2014] [Indexed: 11/08/2022] Open
Abstract
Population or other pharmacometric models are a useful means to describe, succinctly, the relationships between drug administration, exposure (concentration), and downstream changes in pharmacodynamic (PD) biomarkers and clinical endpoints, including the mixed effects of patient factors and random interpatient variation (fixed and random effects). However, showing a set of covariate equations to a drug development team is perhaps not the best way to get a message across. Visualization of the consequences of the knowledge encapsulated within the model is the key component. Yet in many instances, it can take hours, perhaps days, to collect ideas from teams, write scripts, and run simulations before presenting the results-by which time they have moved on. How much better, then, to seize the moment and work interactively to decide on a course of action, guided by the model. We exemplify here the visualization of pharmacometric models using the Berkeley Madonna software with a particular focus on interactive sessions. The examples are provided as Supplementary Material.
Collapse
Affiliation(s)
- A Krause
- Actelion Pharmaceuticals Ltd, Department of Clinical Pharmacology, Allschwil, Switzerland
| | - P J Lowe
- Novartis Pharma AG, Advanced Quantitative Sciences, Basel, Switzerland
| |
Collapse
|
28
|
Brennan FR, Cauvin A, Tibbitts J, Wolfreys A. Optimized nonclinical safety assessment strategies supporting clinical development of therapeutic monoclonal antibodies targeting inflammatory diseases. Drug Dev Res 2014; 75:115-61. [PMID: 24782266 DOI: 10.1002/ddr.21173] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 02/23/2014] [Indexed: 12/19/2022]
Abstract
An increasing number of immunomodulatory monoclonal antibodies (mAbs) and IgG Fc fusion proteins are either approved or in early-to-late stage clinical trials for the treatment of chronic inflammatory conditions, autoimmune diseases and organ transplant rejection. The exquisite specificity of mAbs, in combination with their multi-functional properties, high potency, long half-life (permitting intermittent dosing and prolonged pharamcological effects), and general lack of off-target toxicity makes them ideal therapeutics. Dosing with mAbs for these severe and debilitating but often non life-threatening diseases is usually prolonged, for several months or years, and not only affects adults, including sensitive populations such as woman of child-bearing potential (WoCBP) and the elderly, but also children. Immunosuppression is usually a therapeutic goal of these mAbs and when administered to patients whose treatment program often involves other immunosuppressive therapies, there is an inherent risk for frank immunosuppression and reduced host defence which when prolonged increases the risk of infection and cancer. In addition when mAbs interact with the immune system they can induce other adverse immune-mediated drug reactions such as infusion reactions, cytokine release syndrome, anaphylaxis, immune-complex-mediated pathology and autoimmunity. An overview of the nonclinical safety assessment and risk mitigation strategies utilized to characterize these immunomodulatory mAbs and Fc fusion proteins to support first-in human (FIH) studies and futher clinical development in inflammatory disease indications is provided. Specific emphasis is placed on the design of studies to qualify animal species for toxicology studies, early studies to investigate safety and define PK/PD relationships, FIH-enabling and chronic toxicology studies, immunotoxicity, developmental, reproductive and juvenile toxicity studies and studies to determine the potential for immunosuppression and reduced host defence against infection and cancer. Nonclinical strategies to facilitate clinical and market entry in the most efficient timeframe are presented.
Collapse
Affiliation(s)
- Frank R Brennan
- Preclinical Safety, New Medicines, UCB-Celltech, Slough, SL1 3WE, UK
| | | | | | | |
Collapse
|
29
|
Bashaw ED, DeSilva B, Rose MJ, Wang YMC, Shukla C. Bioanalytical method validation: concepts, expectations and challenges in small molecule and macromolecule--a report of PITTCON 2013 symposium. AAPS JOURNAL 2014; 16:586-91. [PMID: 24700273 DOI: 10.1208/s12248-014-9597-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 03/21/2014] [Indexed: 01/29/2023]
Abstract
The concepts, importance, and implications of bioanalytical method validation has been discussed and debated for a long time. The recent high profile issues related to bioanalytical method validation at both Cetero Houston and former MDS Canada has brought this topic back in the limelight. Hence, a symposium on bioanalytical method validation with the aim of revisiting the building blocks as well as discussing the challenges and implications on the bioanalysis of both small molecules and macromolecules was featured at the PITTCON 2013 Conference and Expo. This symposium was cosponsored by the American Chemical Society (ACS)-Division of Analytical Chemistry and Analysis and Pharmaceutical Quality (APQ) Section of the American Association of Pharmaceutical Scientists (AAPS) and featured leading speakers from the Food & Drug Administration (FDA), academia, and industry. In this symposium, the speakers shared several unique examples, and this session also provided a platform to discuss the need for continuous vigilance of the bioanalytical methods during drug discovery and development. The purpose of this article is to provide a concise report on the materials that were presented.
Collapse
Affiliation(s)
- Edward D Bashaw
- Office of Clinical Pharmacology, Center for Drug Evaluations and Research, United States Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | | | | | | | | |
Collapse
|
30
|
Mortensen DL, Prabhu S, Stefanich EG, Kadkhodayan-Fischer S, Gelzleichter TR, Baker D, Jiang J, Wallace K, Iyer S, Fielder PJ, Putnam WS. Effect of antigen binding affinity and effector function on the pharmacokinetics and pharmacodynamics of anti-IgE monoclonal antibodies. MAbs 2014; 4:724-31. [PMID: 23778267 DOI: 10.4161/mabs.22216] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Modulating the binding affinities to IgE or changing the FcγR binding properties of anti-IgE antibodies offers an opportunity to enhance the therapeutic potential of anti-IgE antibodies, but the influence of increased affinity to IgE or reduced Fc effector function on the pharmacological properties of anti-IgE therapies remains unclear. Our studies were designed to characterize the pharmacokinetics, pharmacodynamics and immune-complex distribution of two high-affinity anti-IgE monoclonal antibodies, high-affinity anti-IgE antibody (HAE) 1 and 2, in mice and monkeys. HAE1, also known as PRO98498, is structurally similar to omalizumab (Xolair®), a humanized anti-IgE IgG1 marketed for the treatment of asthma, but differs by 9 amino acid changes in the complementarity-determining region resulting in a 23-fold improvement in affinity. HAE2 is similar to HAE1, but its Fc region was altered to reduce binding to Fcγ receptors. As expected given the decreased binding to Fcγ receptors, systemic exposure to pre-formed HAE2:IgE complexes in mice was greater (six-fold) and distribution to the liver lower (four-fold) compared with HAE1:IgE complexes. In monkeys, systemic exposure to HAE1 was similar to that previously observed for omalizumab in this species, but required comparatively lower serum drug concentrations to suppress free IgE levels. HAE2 treatment resulted in greater exposure and greater increase of total IgE, relative to HAE1, because of decreased clearance of HAE2:IgE complexes. Overall, these data suggest that increased binding affinity to IgE may provide a more effective therapeutic for asthma patients, and that retaining FcγR binding of the anti-IgE antibody is important for elimination of anti-IgE:IgE complexes.
Collapse
Affiliation(s)
- Deborah L Mortensen
- Departments of Pharmacokinetic and Pharmacodynamic Sciences, Genentech, South San Francisco, CA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Wang W, Wang X, Doddareddy R, Fink D, McIntosh T, Davis HM, Zhou H. Mechanistic pharmacokinetic/target engagement/pharmacodynamic (PK/TE/PD) modeling in deciphering interplay between a monoclonal antibody and its soluble target in cynomolgus monkeys. AAPS JOURNAL 2013; 16:129-39. [PMID: 24287601 DOI: 10.1208/s12248-013-9545-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 11/04/2013] [Indexed: 11/30/2022]
Abstract
For therapeutic monoclonal antibodies (mAbs) against soluble ligands, the free ligand level can, theoretically, be used as a surrogate for efficacy. However, it can be extremely challenging technically to measure free ligand level in the presence of an excessive amount of antibody-ligand complex. The interplay among such mAbs, ligands, and the downstream pharmacodynamic (PD) effects has not been well defined. Using siltuximab and interleukin-6 (IL-6) as model compounds, a pharmacokinetic (PK)/target engagement (TE) model was established via simultaneous fitting of total siltuximab, total IL-6, and free IL-6 concentration profiles following a low dose of siltuximab in cynomolgus monkeys. The model adequately captured the observed data and provided estimation of model parameters with good precision. The PK/TE model was used to predict free IL-6 profiles at higher siltuximab doses, where the accurate determination of free IL-6 concentration became technically too difficult. The measured free IL-6 levels from the low-dose groups and PK/TE model-predicted free IL-6 levels from the high-dose groups were used to drive an indirect response TE/PD model to describe the concentration-effect relationship between free IL-6 and C-reactive protein (CRP). The TE/PD model adequately captured both CRP elevation and CRP suppression in response to free IL-6 concentration change from baseline with a linear stimulation function, providing direct evidence that the PK/TE model-predicted free IL-6 levels from the high-dose groups were accurate. Overall, the results provided an integrated PK/TE/PD modeling and bioanalytical framework for prediction of efficacious dose levels and duration of action for mAbs against soluble ligands with rapid turnover.
Collapse
Affiliation(s)
- Weirong Wang
- Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Pharmacokinetics, pharmacodynamics and physiologically-based pharmacokinetic modelling of monoclonal antibodies. Clin Pharmacokinet 2013; 52:83-124. [PMID: 23299465 DOI: 10.1007/s40262-012-0027-4] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Development of monoclonal antibodies (mAbs) and their functional derivatives represents a growing segment of the development pipeline in the pharmaceutical industry. More than 25 mAbs and derivatives have been approved for a variety of therapeutic applications. In addition, around 500 mAbs and derivatives are currently in different stages of development. mAbs are considered to be large molecule therapeutics (in general, they are 2-3 orders of magnitude larger than small chemical molecule therapeutics), but they are not just big chemicals. These compounds demonstrate much more complex pharmacokinetic and pharmacodynamic behaviour than small molecules. Because of their large size and relatively poor membrane permeability and instability in the conditions of the gastrointestinal tract, parenteral administration is the most usual route of administration. The rate and extent of mAb distribution is very slow and depends on extravasation in tissue, distribution within the particular tissue, and degradation. Elimination primarily happens via catabolism to peptides and amino acids. Although not definitive, work has been published to define the human tissues mainly involved in the elimination of mAbs, and it seems that many cells throughout the body are involved. mAbs can be targeted against many soluble or membrane-bound targets, thus these compounds may act by a variety of mechanisms to achieve their pharmacological effect. mAbs targeting soluble antigen generally exhibit linear elimination, whereas those targeting membrane-bound antigen often exhibit non-linear elimination, mainly due to target-mediated drug disposition (TMDD). The high-affinity interaction of mAbs and their derivatives with the pharmacological target can often result in non-linear pharmacokinetics. Because of species differences (particularly due to differences in target affinity and abundance) in the pharmacokinetics and pharmacodynamics of mAbs, pharmacokinetic/pharmacodynamic modelling of mAbs has been used routinely to expedite the development of mAbs and their derivatives and has been utilized to help in the selection of appropriate dose regimens. Although modelling approaches have helped to explain variability in both pharmacokinetic and pharmacodynamic properties of these drugs, there is a clear need for more complex models to improve understanding of pharmacokinetic processes and pharmacodynamic interactions of mAbs with the immune system. There are different approaches applied to physiologically based pharmacokinetic (PBPK) modelling of mAbs and important differences between the models developed. Some key additional features that need to be accounted for in PBPK models of mAbs are neonatal Fc receptor (FcRn; an important salvage mechanism for antibodies) binding, TMDD and lymph flow. Several models have been described incorporating some or all of these features and the use of PBPK models are expected to expand over the next few years.
Collapse
|
33
|
Eckl-Dorna J, Niederberger V. What is the source of serum allergen-specific IgE? Curr Allergy Asthma Rep 2013; 13:281-7. [PMID: 23585215 DOI: 10.1007/s11882-013-0348-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Immunoglobulin E (IgE), the key effector element in the induction and propagation of allergic diseases, is the least abundant antibody class. In allergic patients, class switch recombination to IgE in B cells is induced by allergen contact in conjunction with T cell interaction and a Th2 cytokine environment. With regard to future therapeutic approaches, the sites of IgE production in human subjects and the nature and characteristics of IgE-producing cells are of great interest. In this context, it has been shown that allergen-specific IgE levels can be boosted by contact with allergens via the respiratory mucosa of the nose. Also, it has been proposed that allergy effector organs (e.g., the nasal mucosa and the lung) may be important sites of IgE production in allergic patients. IgE-producing cells have also been found in the blood, but their numbers are extremely low. Transfer of specific sensitization during bone marrow transplantation indicates the presence of IgE-producing B memory cells or plasma cells also in the bone marrow. This review summarizes data on the induction of IgE production, IgE memory and the sites of IgE production in human allergic patients.
Collapse
Affiliation(s)
- Julia Eckl-Dorna
- Department of Otorhinolaryngology, Medical University of Vienna, General Hospital Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | | |
Collapse
|
34
|
Pharmacokinetic studies of protein drugs: past, present and future. Adv Drug Deliv Rev 2013; 65:1065-73. [PMID: 23541379 DOI: 10.1016/j.addr.2013.03.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/18/2013] [Accepted: 03/18/2013] [Indexed: 12/11/2022]
Abstract
Among the growing number of therapeutic proteins on the market, there is an emergence of biotherapeutics designed from our comprehension of the physiological mechanisms responsible for their peripheral and tissue pharmacokinetics. Most of them have been optimized to increase their half-life through glycosylation engineering, polyethylene glycol conjugation or Fc fusion. However, our understanding of biological drug behaviors is still its infancy compared to the huge amount of data regarding small molecular weight drugs accumulated over half a century. Unfortunately, therapeutic proteins share few resemblances with these drugs. For instance drug-targeted-mediated disposition, binding to glycoreceptors, lysosomal recycling, large hydrodynamic volume and electrostatic charge are typical critical characteristics that cannot be derived from our anterior knowledge of classical drugs. However, the numerous discoveries made in the two last decades have driven and will continue to drive new options in biochemical engineering and support the design of complex delivery systems. Most of these new developments will be supported by novel analytical methods for assessing in vitro or in vivo metabolism parameters.
Collapse
|
35
|
Aston PJ, Derks G, Agoram BM, van der Graaf PH. A mathematical analysis of rebound in a target-mediated drug disposition model: I.without feedback. J Math Biol 2013; 68:1453-78. [PMID: 23591581 DOI: 10.1007/s00285-013-0675-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 04/05/2013] [Indexed: 11/29/2022]
Abstract
We consider the possibility of free receptor (antigen/cytokine) levels rebounding to higher than the baseline level after one or more applications of an antibody drug using a target-mediated drug disposition model. Using geometry and dynamical systems analysis, we show that rebound will occur if and only if the elimination rate of the drug-receptor product is slower than the elimination rates of the drug and of the receptor. We also analyse the magnitude of rebound through approximations and simulations and demonstrate that it increases if the drug dose increases or if the difference between the elimination rate of the drug-receptor product and the minimum of the elimination rates of the drug and of the receptor increases.
Collapse
Affiliation(s)
- Philip J Aston
- Department of Mathematics, University of Surrey, Guildford, UK
| | | | | | | |
Collapse
|
36
|
Chimalakonda AP, Yadav R, Marathe P. Factors influencing magnitude and duration of target inhibition following antibody therapy: implications in drug discovery and development. AAPS JOURNAL 2013; 15:717-27. [PMID: 23588584 DOI: 10.1208/s12248-013-9477-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 03/20/2013] [Indexed: 01/01/2023]
Abstract
Antibodies or antibody-related fusion proteins binding to soluble antigens in plasma form an important subclass of approved therapeutics. Pharmaceutical companies are constantly trying to accelerate the pace of drug discovery and development of these antibodies and identify superior candidates in face of significant attrition rates. Understanding the interplay between drug- and target-related factors on magnitude and duration of target inhibition is imperative for successful advancement of these therapeutics. Simulations using a target-mediated drug disposition model were performed to evaluate the influence of antibody-target binding affinity, baseline target concentration, and target turnover on magnitude and duration of soluble target inhibition. These simulations assumed intravenous dosing of the antibody and evaluated multiple parameters over a wide range. These simulations reveal that improvement in affinity reaches a point of diminishing returns following which further improvement in affinity does not alter the magnitude and more importantly the duration of target inhibition. Evaluation of unbound antibody and target kinetics indicated that point of diminishing returns in duration of inhibition was due to target-mediated binding and subsequent elimination of antibody at later time points. Similarly, influence of baseline target concentration and target turnover on magnitude and duration of target inhibition in plasma is shown. Additionally, the fraction of dose eliminated via target mediated elimination (Fel(™)) can be a useful tool to enable selection of strategies to increase duration of target inhibition. The implications of these simulations in drug discovery and development with regard to target identification, antibody optimization, and backup candidate selection are discussed.
Collapse
Affiliation(s)
- Anjaneya P Chimalakonda
- Metabolism and Pharmacokinetics, Department of Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Co., Mail Stop: 17-2.04, Pennington, NJ 08534, USA.
| | | | | |
Collapse
|
37
|
Xu Z, Davis HM, Zhou H. Rational development and utilization of antibody-based therapeutic proteins in pediatrics. Pharmacol Ther 2013; 137:225-47. [DOI: 10.1016/j.pharmthera.2012.10.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 10/08/2012] [Indexed: 12/15/2022]
|
38
|
Disparate in vivo efficacy of FTY720 in xenograft models of Philadelphia positive and negative B-lineage acute lymphoblastic leukemia. PLoS One 2012; 7:e36429. [PMID: 22570713 PMCID: PMC3343039 DOI: 10.1371/journal.pone.0036429] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 04/01/2012] [Indexed: 12/22/2022] Open
Abstract
Most patients with acute lymphoblastic leukemia (ALL) respond well to standard chemotherapy-based treatments. However a significant proportion of patients, particularly adult patients, relapse with the majority dying of leukemia. FTY720 is an immunosuppressive drug that was recently approved for the treatment of multiple sclerosis and is currently under pre-clinical investigation as a therapy for a number of hematological malignancies. Using human ALL xenografts in NOD/SCIDγc−/− mice, we show for the first time that three Ph+ human ALL xenografts responded to FTY720 with an 80±12% (p = 0.048) reduction in overall disease when treatment was commenced early. In contrast, treatment of mice with FTY720 did not result in reduced leukemia compared to controls using four separate human Ph− ALL xenografts. Although FTY720 reactivated PP2A in vitro, this reactivation was not required for death of Ph− ALL cells. The plasma levels of FTY720 achieved in the mice were in the high nanomolar range. However, the response seen in the Ph+ ALL xenografts when treatment was initiated early implies that in vivo efficacy may be obtained with substantially lower drug concentrations than those required in vitro. Our data suggest that while FTY720 may have potential as a treatment for Ph+ ALL it will not be a useful agent for the treatment of Ph− B-ALL.
Collapse
|
39
|
Zeng X, Wang T, Zhu C, Ye Y, Song B, Lai X, Zeng Y. FTY720 mediates activation suppression and G0/G1 cell cycle arrest in a concanavalin A-induced mouse lymphocyte pan-activation model. Inflamm Res 2012; 61:623-34. [DOI: 10.1007/s00011-012-0454-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 12/31/2011] [Accepted: 02/15/2012] [Indexed: 12/18/2022] Open
|
40
|
Taylor S, Gray JRJ, Willis R, Deeks N, Haynes A, Campbell C, Gaskin P, Leavens K, Demont E, Dowell S, Cryan J, Morse M, Patel A, Garden H, Witherington J. The utility of pharmacokinetic–pharmacodynamic modeling in the discovery and optimization of selective S1P1agonists. Xenobiotica 2012; 42:671-86. [DOI: 10.3109/00498254.2011.645908] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
41
|
Lowe PJ, Renard D. Omalizumab decreases IgE production in patients with allergic (IgE-mediated) asthma; PKPD analysis of a biomarker, total IgE. Br J Clin Pharmacol 2012; 72:306-20. [PMID: 21392073 DOI: 10.1111/j.1365-2125.2011.03962.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT Omalizumab is a humanized anti-IgE monoclonal antibody that binds and captures circulating IgE, preventing interaction with receptors on mast cells and basophils, thereby interrupting the allergic cascade. It has a well-characterized efficacy and safety profile in patients with asthma. While omalizumab is known to reduce serum free IgE concentrations, effects on total IgE and IgE production are less well characterized. WHAT THIS STUDY ADDS (i) Confirmation of prior hypotheses that IgE production can decrease with time when patients are given anti-IgE therapy; (ii) guidance on a biomarker, total IgE, which can be used to ascertain whether individual patients experience a change in their IgE production; and (iii) a way to assess whether patients' IgE production has been sufficiently down-regulated such that they may consider stopping anti-IgE therapy. AIM To determine whether excessive IgE production by patients with atopic allergic asthma decreases with omalizumab therapy. METHODS Omalizumab, free and total IgE data were obtained from an epidemiological study and six randomized, double-blind, placebo-controlled trials in patients with allergic asthma. The binding between omalizumab and IgE together with the production and elimination of IgE were modelled as previously, except that, in order to explain why total IgE was decreasing over a period of 5 years, the expression of IgE was allowed to change. RESULTS The prior constant IgE production model failed to converge on the data once long-term observations were included, whereas models allowing IgE production to decrease fitted. A feedback model indicated that, on average, IgE production decreased by 54% per year. This model was further developed with covariate searches indicating clinically small but statistically significant effects of age, gender, body mass index and race on some parameters. Model predictions were checked internally and externally against 3-5 year data from paediatric and adult atopic asthmatic patients and externally against extensive total IgE data from a long-duration (>1 year) phase 1 study which was not used in the model building. CONCLUSIONS A pharmacokinetic-pharmacodynamic model incorporating omalizumab-IgE binding and feedback for control of IgE production indicates that omalizumab reduces production of IgE. This raises the possibility that indefinite treatment may not be required, only for perhaps a few years. After the initial accumulation, total IgE should provide a means to monitor IgE production and guide individual treatment decisions.
Collapse
|
42
|
Clienti S, Morjaria JB, Basile E, Polosa R. Monoclonal antibodies for the treatment of severe asthma. Curr Allergy Asthma Rep 2011; 11:253-60. [PMID: 21360131 DOI: 10.1007/s11882-011-0184-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Patients with severe asthma have a significant unmet need with persistent symptoms and/or frequent exacerbations despite treatment with high-dose steroid and other currently available therapies. These patients are also at risk of developing steroid-related side effects, and their severe, unrelenting symptoms have a huge impact on health care resources due to frequent hospital admissions and requirement for intensive medication use. Consequently, a compelling need exists for more effective and safer pharmacotherapies to help them achieve adequate disease control. Recent novel therapies for severe asthma are now emerging, some of the most promising of which are monoclonal antibodies. Monoclonal antibodies represent a form of immunotherapy used in a wide variety of therapeutic roles. The spectrum of disease states in which monoclonal antibodies have been approved for therapeutic use now includes respiratory and allergic diseases. At present, only one drug is licensed for allergic asthmatics with severe disease, omalizumab. We review some of the currently available biologics that are approved or under investigation for use in severe asthma. Some have shown to be useful in specifically targeted subpopulations of patients with severe asthma, whereas other have proven to be unsafe and/or unsuccessful. Despite these developments, more effort should be devoted to identifying new molecular targets, testing innovative approaches, and establishing the best use of what is available. Regarding this latter point, identifying individual characteristics that predict successful responses to these treatments is highly desirable.
Collapse
Affiliation(s)
- Salvatore Clienti
- Institute of Internal Medicine, S. Marta Hospital, Azienda Ospedaliero-Universitaria "Policlinico-Vittorio Emanuele", Università di Catania, Catania, Italy
| | | | | | | |
Collapse
|
43
|
Stratified medicine in selecting biologics for the treatment of severe asthma. Curr Opin Allergy Clin Immunol 2011; 11:58-63. [PMID: 21150434 DOI: 10.1097/aci.0b013e3283423245] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE OF REVIEW Despite optimal treatment, asthma symptoms in about 5-10% of patients remain poorly controlled. Apart from having an impact on their asthma-related quality of life and adverse effects of the medications used (especially corticosteroids), their severe symptoms also impact healthcare resources due to frequent admission and requirement for intensive medications use. The last decade has seen an improved understanding in the pathophysiology of the complex cellular and molecular networks involved in the inflammatory and immunological phenotype of severe asthma. This knowledge may help providing strategies by which these phenotypes operate and pave the way for drug development and individualized treatment. RECENT FINDINGS Here we review the current evidence of biological agents in patients with severe asthma recently assessed for safety and efficacy. Some of these agents have shown to be useful in specifically targeted subpopulations of patients with severe asthma, whereas others have proven to be unsafe and/or unsuccessful. In addition, we discuss recent data on clinical and pharmacokinetic-pharmacodynamic aspects of omalizumab, the only licensed anti-IgE therapy for severe atopic asthma. SUMMARY More basic science work is required to improve the current understanding of severe asthma pathophysiology and proof-of-concept clinical studies are required to explore relevant biomolecular targets in this small subset of patients. At present, only one drug is licensed for allergic asthmatic patients with severe disease, omalizumab. Novel therapies in the form of oligonucleotide therapies and other biological agents are also being investigated in the difficult-to-treat asthmatic patient group.
Collapse
|
44
|
Ohno T, Hasegawa C, Nakade S, Kitagawa J, Honda N, Ogawa M. The prediction of human response to ONO-4641, a sphingosine 1-phosphate receptor modulator, from preclinical data based on pharmacokinetic-pharmacodynamic modeling. Biopharm Drug Dispos 2011; 31:396-406. [PMID: 20623701 DOI: 10.1002/bdd.719] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The pharmacokinetic (PK) and pharmacodynamic (PD) parameters of ONO-4641 in humans were estimated using preclinical data in order to provide essential information to better design future clinical studies. The characterization of PK/PD was measured in terms of decreased lymphocyte counts in blood after administration of ONO-4641, a sphingosine 1-phosphate receptor modulator. Using a two-compartment model, human PK parameters were estimated from preclinical PK data of cynomolgus monkey and in vitro human metabolism data. To estimate human PD parameters, the relationship between lymphocyte counts and plasma concentrations of ONO-4641 in cynomolgus monkeys was determined. The relationship between lymphocyte counts and plasma concentrations of ONO-4641 was described by an indirect-response model. The indirect-response model had an I(max) value of 0.828 and an IC(50) value of 1.29 ng/ml based on the cynomolgus monkey data. These parameters were used to represent human PD parameters for the simulation of lymphocyte counts. Other human PD parameters such as input and output rate constants for lymphocytes were obtained from the literature. Based on these estimated human PK and PD parameters, human lymphocyte counts after administration of ONO-4641 were simulated. In conclusion, the simulation of human lymphocyte counts based on preclinical data led to the acquisition of useful information for designing future clinical studies.
Collapse
Affiliation(s)
- Tomoya Ohno
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd, Ibaraki, Japan.
| | | | | | | | | | | |
Collapse
|
45
|
Bioanalytical considerations in the comparability assessment of biotherapeutics. Bioanalysis 2011; 3:613-22. [PMID: 21417731 DOI: 10.4155/bio.11.25] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Comparison of biotherapeutic products before and after manufacturing changes is required to show that the products are highly similar. Besides in vitro assessment on the critical quality attributes and potency, biocomparability studies are sometimes required to demonstrate similarities in pharmacokinetic and pharmacodynamic characteristics. The complex and diverse nature of biotherapeutics requires multifaceted considerations in the biocomparability study design, bioanalytical measurements of drug concentrations and/or pharmacodynamic responses, immunogenicity analysis, data interpretation and decision making. A major perspective is to understand the structure and biological functions of the biotherapeutics in relation to the indication. Issues of a common standard and the importance of the use of ligand-binding assays that are sensitive to structural changes are discussed. It would not be possible to use the same process and one-size-fit-all criteria for biocomparability studies of all biologics. Previous examples from industry and our experience of the bioanalytical considerations for fit-for-purpose pharmacokinetic support and immunogenicity assessments are presented.
Collapse
|
46
|
Yu J, Karcher H, Feire AL, Lowe PJ. From target selection to the minimum acceptable biological effect level for human study: use of mechanism-based PK/PD modeling to design safe and efficacious biologics. AAPS JOURNAL 2011; 13:169-78. [PMID: 21336535 DOI: 10.1208/s12248-011-9256-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 01/31/2011] [Indexed: 11/30/2022]
Abstract
In this paper, two applications of mechanism-based modeling are presented with their utility from candidate selection to first-in-human dosage selection. The first example is for a monoclonal antibody against a cytomegalovirus glycoprotein complex, which involves an antibody binding model and a viral load model. The model was used as part of a feasibility analysis prior to antibody generation, setting the specifications for the affinity needed to achieve a desired level of clinical efficacy. The second example is a pharmacokinetic-pharmacodynamic model based on a single-dose pharmacology study in cynomolgus monkey using data on pharmacokinetics, receptor occupancy, and the dynamics of target cell depletion and recovery. The model was used to estimate the MABEL, here defined as the minimum acceptable biological effect level against which a dose is selected for a first-in-human study. From these applications, we demonstrate that mechanism-based PK/PD binding models are useful for predicting human response to biologics compounds. Especially, such models have the ability to integrate preclinical and clinical, in vitro and in vivo information and facilitate rational decision making during various stages of drug discovery and translational research.
Collapse
Affiliation(s)
- Jing Yu
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA.
| | | | | | | |
Collapse
|
47
|
Gibbs JP. Prediction of exposure-response relationships to support first-in-human study design. AAPS JOURNAL 2010; 12:750-8. [PMID: 20967521 DOI: 10.1208/s12248-010-9236-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 10/01/2010] [Indexed: 01/31/2023]
Abstract
In drug development, phase 1 first-in-human studies represent a major milestone as the drug moves from preclinical discovery to clinical development activities. The safety of human subjects is paramount to the conduct of these studies and regulatory considerations guide activities. Forces of evolution on the pharmaceutical industry are re-shaping the first-in-human dose selection strategy. Namely, high attrition rates in part due to lack of efficacy have led to the re-organization of research and development organizations around the umbrella of translational research. Translational research strives to bring basic research advances into the clinic and support the reverse transfer of information to enhance compound selection strategies. Pharmacokinetic/pharmacodynamic (PK/PD) modeling holds a unique position in translational research by attempting to integrate diverse sets of information. PK/PD modeling has demonstrated utility in dose selection and trial design for later stages of drug development and is now being employed with greater prevalence in the translational research setting to manage risk (i.e., oncology and inflammation/immunology). Moving from empirical E (max) models to more mechanistic representations of the biological system, a higher fidelity of human predictions is expected. Strategies that have proven useful for PK predictions are being applied to PK/PD predictions. This review article examines examples of the application of PK/PD modeling in establishing target concentrations for supporting first-in-human study design.
Collapse
|
48
|
Davda JP, Hansen RJ. Properties of a general PK/PD model of antibody-ligand interactions for therapeutic antibodies that bind to soluble endogenous targets. MAbs 2010; 2:576-88. [PMID: 20676036 DOI: 10.4161/mabs.2.5.12833] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Antibodies that target endogenous soluble ligands are an important class of biotherapeutic agents. While much focus has been placed on characterization of antibody pharmacokinetics, less emphasis has been given to characterization of antibody effects on their soluble targets. We describe here the properties of a generalized mechanism-based PK/PD model used to characterize the in vivo interaction of an antibody and an endogenous soluble ligand. The assumptions and properties of the model are explored, and situations are described when deviations from the basic assumptions may be necessary. This model is most useful for in vivo situations where both antibody and ligand levels are available following drug administration. For a given antibody exposure, the extent and duration of suppression of free ligand is impacted by the apparent affinity of the interaction, as well as by the rate of ligand turnover. The applicability of the general equilibrium model of in vivo antibody-ligand interaction is demonstrated with an anti-Aß antibody.
Collapse
Affiliation(s)
- Jasmine P Davda
- Eli Lilly and Company, Drug Disposition, Lilly Corporate Center, Indianapolis, IN, USA
| | | |
Collapse
|
49
|
Bugelski PJ, Achuthanandam R, Capocasale RJ, Treacy G, Bouman-Thio E. Monoclonal antibody-induced cytokine-release syndrome. Expert Rev Clin Immunol 2010; 5:499-521. [PMID: 20477639 DOI: 10.1586/eci.09.31] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Monoclonal antibodies (mAbs) are widely used in anti-inflammatory and tumor therapy. Although effective, mAbs can cause a variety of adverse effects. An important toxicity seen with a few mAbs is cytokine-release syndrome (CRS). These mAbs include: alemtuzumab, muromonab-CD3, rituximab, tosituzumab, CP-870,893, LO-CD2a/BTI-322 and TGN1412. By contrast, over 30 mAbs used clinically are not associated with CRS. In this review, the clinical aspects of CRS, the mAbs associated with CRS, the cytokines involved and putative mechanisms mediating cytokine release will be discussed. This will be followed by a discussion of the poor predictive value of studies in animals and the prospects for creating in vitro screens. Finally, approaches to decreasing the probability of CRS, decreasing the severity or treating CRS, should it occur, will be described.
Collapse
Affiliation(s)
- Peter J Bugelski
- Toxicology and Investigational Pharmacology, Centocor R&D, R-4-2, 145 King of Prussia Road, Radnor, PA 19087, USA.
| | | | | | | | | |
Collapse
|
50
|
Abstract
Most therapeutic monoclonal antibodies are designed to bind a specific antigen to elicit pharmacological effects. Accurate quantification of a therapeutic monoclonal antibody in biological matrices is essential for assessing its pharmacokinetics and selecting an effective dosing regimen. Therapeutic antibodies may exist in free, partially bound and fully bound forms in the bloodstream. The choice of which form(s) to measure and how to measure them is gaining much attention with the increase in the number of soluble therapeutic targets. This article will review the bioanalytical methods used in supporting the clinical development of the US FDA-approved therapeutic monoclonal antibodies and also discuss how different factors, such as assay format, target and antibody concentrations, and sample dilutions, can have an impact on the measurement of each form of antibody. Appreciation of which form of drug is being measured and what factors may impact measurement under different conditions are important for interpretation of the pharmacokinetics of therapeutic antibodies.
Collapse
|