101
|
Groell F, Jordan O, Borchard G. In vitro models for immunogenicity prediction of therapeutic proteins. Eur J Pharm Biopharm 2018; 130:128-142. [DOI: 10.1016/j.ejpb.2018.06.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 05/09/2018] [Accepted: 06/08/2018] [Indexed: 12/21/2022]
|
102
|
Háda V, Bagdi A, Bihari Z, Timári SB, Fizil Á, Szántay C. Recent advancements, challenges, and practical considerations in the mass spectrometry-based analytics of protein biotherapeutics: A viewpoint from the biosimilar industry. J Pharm Biomed Anal 2018; 161:214-238. [PMID: 30205300 DOI: 10.1016/j.jpba.2018.08.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 01/22/2023]
Abstract
The extensive analytical characterization of protein biotherapeutics, especially of biosimilars, is a critical part of the product development and registration. High-resolution mass spectrometry became the primary analytical tool used for the structural characterization of biotherapeutics. Its high instrumental sensitivity and methodological versatility made it possible to use this technique to characterize both the primary and higher-order structure of these proteins. However, even by using high-end instrumentation, analysts face several challenges with regard to how to cope with industrial and regulatory requirements, that is, how to obtain accurate and reliable analytical data in a time- and cost-efficient way. New sample preparation approaches, measurement techniques and data evaluation strategies are available to meet those requirements. The practical considerations of these methods are discussed in the present review article focusing on hot topics, such as reliable and efficient sequencing strategies, minimization of artefact formation during sample preparation, quantitative peptide mapping, the potential of multi-attribute methodology, the increasing role of mass spectrometry in higher-order structure characterization and the challenges of MS-based identification of host cell proteins. On the basis of the opportunities in new instrumental techniques, methodological advancements and software-driven data evaluation approaches, for the future one can envision an even wider application area for mass spectrometry in the biopharmaceutical industry.
Collapse
Affiliation(s)
- Viktor Háda
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary.
| | - Attila Bagdi
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary
| | - Zsolt Bihari
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary
| | | | - Ádám Fizil
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary
| | - Csaba Szántay
- Spectroscopic Research Department, Gedeon Richter Plc, Hungary.
| |
Collapse
|
103
|
Ovacik M, Lin K. Tutorial on Monoclonal Antibody Pharmacokinetics and Its Considerations in Early Development. Clin Transl Sci 2018; 11:540-552. [PMID: 29877608 PMCID: PMC6226118 DOI: 10.1111/cts.12567] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/11/2018] [Indexed: 12/21/2022] Open
Abstract
The tutorial introduces the readers to the fundamentals of antibody pharmacokinetics (PK) in the context of drug development. Topics covered include an overview of antibody development, PK characteristics, and the application of antibody PK/pharmacodynamics (PD) in research and development decision-making. We also discuss the general considerations for planning a nonclinical PK program and describe the types of PK studies that should be performed during early development of monoclonal antibodies.
Collapse
Affiliation(s)
- Meric Ovacik
- Department of Preclinical and Translational Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Kedan Lin
- Clinical Pharmacology, NGM Biopharmaceuticals, Inc., South San Francisco, California, USA
| |
Collapse
|
104
|
Sensitivity and drug tolerance of antidrug antibody assays in relation to positive control characteristics. Bioanalysis 2018; 10:1289-1306. [DOI: 10.4155/bio-2018-0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: Detection and characterization of antidrug antibodies (ADA) play a key role in understanding the relation of ADA with safety and efficacy. Positive controls (PCs) are used to estimate the sensitivity and assay sensitivity in the presence of drug (drug tolerance) of assays to detect ADA. We investigated a number of factors that may drive sensitivity and drug tolerance. Results: We found no correlation between affinity and sensitivity and sensitivity in the presence of drug in multiple assays with two antibody–drug conjugates. Multiple factors that influenced sensitivity and drug tolerance were observed, yet these had limited overall predictive value for all investigated assay formats, PCs and compounds.Conclusion: Assay sensitivity and drug tolerance as studied here, are likely driven by multiple factors such as cut point confidence level, stoichiometry of PC–drug complexes and presentation of epitopes.
Collapse
|
105
|
Polumuri SK, Haile LA, Ireland DDC, Verthelyi D. Aggregates of IVIG or Avastin, but not HSA, modify the response to model innate immune response modulating impurities. Sci Rep 2018; 8:11477. [PMID: 30065306 PMCID: PMC6068171 DOI: 10.1038/s41598-018-29850-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/06/2018] [Indexed: 12/11/2022] Open
Abstract
Therapeutic proteins can induce immune responses that affect their safety and efficacy. Product aggregates and innate immune response modulating impurities (IIRMI) are risk factors of product immunogenicity. In this study, we use Intravenous Immunoglobulin (IVIG), Avastin, and Human Serum Albumin (HSA) to explore whether increased aggregates activate innate immune cells or modify the response to IIRMI. We show that increased aggregates (shaken or stirred) in IVIG and Avastin, but not HSA, induced activation of MAPKs (pp38, pERK and pJNK) and transcription of immune-related genes including IL8, IL6, IL1β, CSF1, CCL2, CCL7, CCL3, CCL24, CXCL2, IRAK1, EGR2, CEBPβ, PPARg and TNFSF15 in human PBMC. The immunomodulatory effect was primarily mediated by FcγR, but not by TLR. Interestingly, increased aggregates in IVIG or Avastin magnified innate immune responses to TLR2/4 agonists, but diminished responses to TLR3/9 agonists. This study shows that IIRMI and aggregates can modify the activity of immune cells potentially modifying the milieu where the products are delivered highlighting the complex interplay of different impurities on product immunogenicity risk. Further, we show that aggregates could modify the sensitivity of PBMC-based assays designed to detect IIRMI. Understanding and managing immunogenicity risk is a critical component of product development and regulation.
Collapse
Affiliation(s)
- Swamy Kumar Polumuri
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Lydia A Haile
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Derek D C Ireland
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Daniela Verthelyi
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA.
| |
Collapse
|
106
|
Expanding Bedside Filtration-A Powerful Tool to Protect Patients From Protein Aggregates. J Pharm Sci 2018; 107:2775-2788. [PMID: 30059660 DOI: 10.1016/j.xphs.2018.07.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 12/29/2022]
Abstract
Protein immunogenicity is intensively researched by academics, biopharmaceutical companies, and authorities as it can compromise the safety and efficacy of a biopharmaceutical drug. So far, the exact protein aggregate properties inducing immune responses are not known. Possible protein-related factors could be size, chemical modifications, or higher order structures. It is impossible to achieve an absolute absence of protein aggregates even for very stable formulations. The application of "bedside filtration," meaning filtration during the preparation or administration of the drug product immediately before injection, has the potential to increase the safety of every drug container and could prevent the undesired injection of particulate matter into the patient. In this study, the high efficiency of filtration for reducing the amount of protein particles was demonstrated with more than 19 stressed and nonstressed biopharmaceutical products which covered a broad concentration and molecular weight range. Furthermore, critical aspects regarding the usage of filters such as particle shedding from filters, protein loss as a result of protein adsorption, or the hold-up volume of the filters were assessed. Although differences between the filters were observed, no negative impact by the investigated filters could be found. A broader application of bedside filtration is therefore proposed.
Collapse
|
107
|
Schack MM, Møller EH, Carpenter JF, Rades T, Groenning M. A Platform for Preparing Homogeneous Proteinaceous Subvisible Particles With Distinct Morphologies. J Pharm Sci 2018; 107:1842-1851. [DOI: 10.1016/j.xphs.2018.03.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 01/23/2018] [Accepted: 03/08/2018] [Indexed: 11/30/2022]
|
108
|
Faraji F, Karjoo Z, Moghaddam MV, Heidari S, Emameh RZ, Falak R. Challenges related to the immunogenicity of parenteral recombinant proteins: Underlying mechanisms and new approaches to overcome it. Int Rev Immunol 2018; 37:301-315. [PMID: 29851534 DOI: 10.1080/08830185.2018.1471139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immune response elicited by therapeutic proteins is an important safety and efficacy issue for regulatory agencies, drug manufacturers, clinicians, and patients. Administration of therapeutic proteins can potentially induce the production of anti-drug antibodies or cell-mediated immune responses. At first, it was speculated that the immunogenicity is related to the non-human origin of these proteins. Later on, it was confirmed that the human proteins may also show immunogenicity. In this review article, we will focus on a number of factors, which play crucial roles in the human protein immunogenicity. These factors are related to the patient's status (or intrinsic properties) and molecular characteristics of the therapeutic protein's (or extrinsic properties). Furthermore, we will discuss available in silico, in vitro, and in vivo methods for the prediction of sequences, which may generate an immune response following parenteral administration of these proteins. In summary, nowadays, it is possible for drug manufacturers to evaluate the risk of immunogenicity of therapeutic proteins and implement a management plan to overcome the problems prior to proceeding to human clinical trials.
Collapse
Affiliation(s)
- Fatemeh Faraji
- a Immunology Research Center , Iran University of Medical Sciences (IUMS) , Tehran , Iran.,b Department of Immunology, School of Medicine , Iran University of Medical Sciences (IUMS) , Tehran , Iran
| | - Zahra Karjoo
- a Immunology Research Center , Iran University of Medical Sciences (IUMS) , Tehran , Iran
| | | | - Sahel Heidari
- a Immunology Research Center , Iran University of Medical Sciences (IUMS) , Tehran , Iran.,b Department of Immunology, School of Medicine , Iran University of Medical Sciences (IUMS) , Tehran , Iran
| | - Reza Zolfaghari Emameh
- c Department of Energy and Environmental Biotechnology, Division of Industrial & Environmental Biotechnology , National Institute of Genetic Engineering and Biotechnology (NIGEB) , Tehran , Iran
| | - Reza Falak
- a Immunology Research Center , Iran University of Medical Sciences (IUMS) , Tehran , Iran.,b Department of Immunology, School of Medicine , Iran University of Medical Sciences (IUMS) , Tehran , Iran
| |
Collapse
|
109
|
Choy E, Caporali R, Xavier R, Fautrel B, Sanmarti R, Bao M, Bernasconi C, Pethö-Schramm A. Subcutaneous tocilizumab in rheumatoid arthritis: findings from the common-framework phase 4 study programme TOZURA conducted in 22 countries. Rheumatology (Oxford) 2018; 57:499-507. [PMID: 29244149 PMCID: PMC5850727 DOI: 10.1093/rheumatology/kex443] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Indexed: 12/20/2022] Open
Abstract
Objectives The aim of this pooled analysis of the TOZURA study programme was to evaluate the efficacy and safety of subcutaneous tocilizumab (TCZ-SC) as monotherapy or in combination with conventional synthetic DMARDs (csDMARDs) in patients with moderate to severe RA who had an inadequate response to csDMARD or anti-TNF agent therapy or who were MTX naïve. Methods TOZURA is a multinational, open-label, single-arm, common-framework, phase 4 study programme (11 protocols, 22 countries). Patients received TCZ-SC 162 mg each week for ⩾24 weeks, administered at the investigator’s discretion, as monotherapy or in combination with a csDMARD. Efficacy, safety and immunogenicity were evaluated; propensity score–based matching was used for between-group comparisons. Results Of 1804 patients, 353 (19.6%) received monotherapy and 1451 (80.4%) received combination therapy. The 28-joint DAS using ESR (DAS28-ESR) in both groups decreased significantly from baseline to week 24 (mean change: monotherapy −3.40, combination therapy −3.46), with no significant difference between groups (P = 0.46). The proportion of patients who achieved DAS28-ESR or Clinical Disease Activity Index remission or ACR 20/50/70/90 responses was similar between groups. Overall, 13.9% of patients withdrew—6.2% for safety reasons and 1.6% for insufficient therapeutic response; 5.8% of patients experienced one or more serious adverse events [14.6/100 patient-years (PY)]; six deaths occurred (0.64/100 PY). Conclusion In a common framework of 11 studies in 22 countries, this phase 4 study programme confirmed TCZ-SC’s known efficacy and safety profile with comparable effects as monotherapy and in combination with csDMARDs. Trial registration ClinicalTrials.gov (http://www.clinicaltrials.gov) NCT01941940, NCT01941095, NCT01951170, NCT01987479, NCT01988012, NCT01995201, NCT02001987, NCT02011334, NCT02031471, NCT02046603 and NCT02046616.
Collapse
Affiliation(s)
- Ernest Choy
- CREATE Centre, Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Roberto Caporali
- Division of Rheumatology, University of Pavia and IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Ricardo Xavier
- Department of Medicine, Universidade Federal do Rio Grande do Sul Porto Alegre, Rio Grande do Sul, Brazil
| | - Bruno Fautrel
- Department of Rheumatology, APHP, Pitie Salpetriere Hospital, Pierre and Marie Curie University, Paris, France
| | - Raimón Sanmarti
- Department of Rheumatology, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Min Bao
- Genentech, South San Francisco, CA, USA
| | | | | |
Collapse
|
110
|
Turner MR, Balu-Iyer SV. Challenges and Opportunities for the Subcutaneous Delivery of Therapeutic Proteins. J Pharm Sci 2018; 107:1247-1260. [PMID: 29336981 PMCID: PMC5915922 DOI: 10.1016/j.xphs.2018.01.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/21/2017] [Accepted: 01/03/2018] [Indexed: 12/22/2022]
Abstract
Biotherapeutics is a rapidly growing drug class, and over 200 biotherapeutics have already obtained approval, with about 50 of these being approved in 2015 and 2016 alone. Several hundred protein therapeutic products are still in the pipeline, including interesting new approaches to treatment. Owing to patients' convenience of at home administration and reduced number of hospital visits as well as the reduction in treatment costs, subcutaneous (SC) administration of biologics is of increasing interest. Although several avenues for treatment using biotherapeutics are being explored, there is still a sufficient gap in knowledge regarding the interplay of formulation conditions, immunogenicity, and pharmacokinetics (PK) of the absorption of these compounds when they are given SC. This review seeks to highlight the major concerns and important factors governing this route of administration and suggest a holistic approach for effective SC delivery.
Collapse
Affiliation(s)
- Michael R Turner
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214.
| |
Collapse
|
111
|
Passey C, Suryawanshi S, Sanghavi K, Gupta M. Reporting, Visualization, and Modeling of Immunogenicity Data to Assess Its Impact on Pharmacokinetics, Efficacy, and Safety of Monoclonal Antibodies. AAPS JOURNAL 2018; 20:35. [PMID: 29484520 DOI: 10.1208/s12248-018-0194-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/17/2018] [Indexed: 12/21/2022]
Abstract
The rapidly increasing number of therapeutic biologics in development has led to a growing recognition of the need for improvements in immunogenicity assessment. Published data are often inadequate to assess the impact of an antidrug antibody (ADA) on pharmacokinetics, safety, and efficacy, and enable a fully informed decision about patient management in the event of ADA development. The recent introduction of detailed regulatory guidance for industry should help address many past inadequacies in immunogenicity assessment. Nonetheless, careful analysis of gathered data and clear reporting of results are critical to a full understanding of the clinical relevance of ADAs, but have not been widely considered in published literature to date. Here, we review visualization and modeling of immunogenicity data. We present several relatively simple visualization techniques that can provide preliminary information about the kinetics and magnitude of ADA responses, and their impact on pharmacokinetics and clinical endpoints for a given therapeutic protein. We focus on individual sample- and patient-level data, which can be used to build a picture of any trends, thereby guiding analysis of the overall study population. We also discuss methods for modeling ADA data to investigate the impact of immunogenicity on pharmacokinetics, efficacy, and safety.
Collapse
Affiliation(s)
- Chaitali Passey
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Satyendra Suryawanshi
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Kinjal Sanghavi
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Manish Gupta
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA.
| |
Collapse
|
112
|
Kim J, Krebs MRH, Trout BL. Retracted: Molecular characterization of excipients' preferential interactions with therapeutic monoclonal antibodies. J Pharm Pharmacol 2018; 70:289-304. [PMID: 28776673 DOI: 10.1111/jphp.12787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 06/18/2017] [Indexed: 10/19/2022]
Abstract
Retraction: Molecular characterization of excipients' preferential interactions with therapeutic monoclonal antibodies by Jehoon Kim, Mark R. H. Krebs and Bernhardt L. Trout The above article from the Journal of Pharmacy and Pharmacology, first published online on 4 August 2017 in Wiley Online Library (wileyonlinelibrary.com), has been retracted by agreement between the authors, the journal Editor-in-Chief, Professor David Jones, and John Wiley & Sons Ltd. The authors discovered that the analysis of simulations was faulty making the data incorrect. Reference Kim J et al. Molecular characterization of excipients' preferential interactions with therapeutic monoclonal antibodies. J Pharm Pharmacol 2017. https://doi.org/10.1111/jphp.12787.
Collapse
Affiliation(s)
- Jehoon Kim
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mark R H Krebs
- Protein Pharmaceutical Development, Biogen, Cambridge, MA, USA
| | - Bernhardt L Trout
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
113
|
Daniels AL, Randolph TW. Flow Microscopy Imaging Is Sensitive to Characteristics of Subvisible Particles in Peginesatide Formulations Associated With Severe Adverse Reactions. J Pharm Sci 2018; 107:1313-1321. [PMID: 29409840 DOI: 10.1016/j.xphs.2018.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/26/2017] [Accepted: 01/17/2018] [Indexed: 01/01/2023]
Abstract
The presence of subvisible particles in formulations of therapeutic proteins is a risk factor for adverse immune responses. Although the immunogenic potential of particulate contaminants likely depends on particle structural characteristics (e.g., composition, size, and shape), exact structure-immunogenicity relationships are unknown. Images recorded by flow imaging microscopy reflect information about particle morphology, but flow microscopy is typically used to determine only particle size distributions, neglecting information on particle morphological features that may be immunologically relevant. We recently developed computational techniques that utilize the Kullback-Leibler divergence and multidimensional scaling to compare the morphological properties of particles in sets of flow microscopy images. In the current work, we combined these techniques with expectation maximization cluster analyses and used them to compare flow imaging microscopy data sets that had been collected by the U.S. Food and Drug Administration after severe adverse drug reactions (including 7 fatalities) were observed in patients who had been administered some lots of peginesatide formulations. Flow microscopy images of particle populations found in the peginesatide lots associated with severe adverse reactions in patients were readily distinguishable from images of particles in lots where severe adverse reactions did not occur.
Collapse
Affiliation(s)
- Austin L Daniels
- Center for Pharmaceutical Biotechnology, Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80309-0596
| | - Theodore W Randolph
- Center for Pharmaceutical Biotechnology, Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80309-0596.
| |
Collapse
|
114
|
Song Y, Yu D, Mayani M, Mussa N, Li ZJ. Monoclonal antibody higher order structure analysis by high throughput protein conformational array. MAbs 2018; 10:397-405. [PMID: 29313446 DOI: 10.1080/19420862.2017.1421880] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
The elucidation of antibody higher order structure (HOS) is critical in therapeutic antibody development. Since HOS determines the protein bioactivity and chemo-physical properties, this knowledge can help to ensure that the safety and efficacy attributes are not compromised. Protein conformational array (PCA) is a novel method for determining the HOS of monoclonal antibodies. Previously, we successfully utilized an enzyme-linked immunosorbent assay (ELISA)-based PCA along with other bioanalytical tools to elucidate the structures of antibody aggregates. In this study, applying a new multiplex-based PCA with 48-fold higher throughput than the ELISA-based one we revealed structural differences between different antibody molecules and antibody structure changes affected by various processing conditions. The PCA analysis of antibody molecules clearly demonstrated significant differences between IgG1 and IgG4 subclasses in epitope exposure and folding status. Furthermore, we applied small angle X-ray scattering to decipher mechanistic insights of PCA technology and validate structural information obtained using PCA. These findings enhance our fundamental understanding of mAbs' HOS in general. The PCA analysis of antibody samples from various processing conditions also revealed that antibody aggregation caused significantly higher exposure of antibody epitopes, which potentially led to a "foreign" molecule that could cause immunogenicity. The PCA data correlated well with protein stability results from traditional methods such as size-exclusion chromatography and protein thermal shift assay. Our study demonstrated that high throughput PCA is a suitable method for HOS analysis in the discovery and development of therapeutic antibodies.
Collapse
Affiliation(s)
- Yuanli Song
- a Biologics Process Development, Bristol-Myers Squibb , 38 Jackson Road, Devens , MA , USA
| | - Deqiang Yu
- a Biologics Process Development, Bristol-Myers Squibb , 38 Jackson Road, Devens , MA , USA
| | - Mukesh Mayani
- a Biologics Process Development, Bristol-Myers Squibb , 38 Jackson Road, Devens , MA , USA
| | - Nesredin Mussa
- a Biologics Process Development, Bristol-Myers Squibb , 38 Jackson Road, Devens , MA , USA
| | - Zheng Jian Li
- a Biologics Process Development, Bristol-Myers Squibb , 38 Jackson Road, Devens , MA , USA
| |
Collapse
|
115
|
Kannan A, Shieh IC, Leiske DL, Fuller GG. Monoclonal Antibody Interfaces: Dilatation Mechanics and Bubble Coalescence. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:630-638. [PMID: 29251942 DOI: 10.1021/acs.langmuir.7b03790] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Monoclonal antibodies (mAbs) are proteins that uniquely identify targets within the body, making them well-suited for therapeutic applications. However, these amphiphilic molecules readily adsorb onto air-solution interfaces where they tend to aggregate. We investigated two mAbs with different propensities to aggregate at air-solution interfaces. The understanding of the interfacial rheological behavior of the two mAbs is crucial in determining their aggregation tendency. In this work, we performed interfacial stress relaxation studies under compressive step strain using a custom-built dilatational rheometer. The dilatational relaxation modulus was determined for these viscoelastic interfaces. The initial value and the equilibrated value of relaxation modulus were larger in magnitude for the mAb with a higher tendency to aggregate in response to interfacial stress. We also performed single-bubble coalescence experiments using a custom-built dynamic fluid-film interferometer (DFI). The bubble coalescence times also correlated to the mAbs aggregation propensity and interfacial viscoelasticity. To study the influence of surfactants in mAb formulations, polyethylene glycol (PEG) was chosen as a model surfactant. In the mixed mAb/PEG system, we observed that the higher aggregating mAb coadsorbed with PEG and formed domains at the interface. In contrast, for the other mAb, PEG entirely covered the interface at the concentrations studied. We studied the mobility of the interfaces, which was manifested by the presence or the lack of Marangoni stresses. These dynamics were strongly correlated with the interfacial viscoelasticity of the mAbs. The influence of competitive destabilization in affecting the bubble coalescence times for the mixed mAb/PEG systems was also studied.
Collapse
Affiliation(s)
- Aadithya Kannan
- Department of Chemical Engineering, Stanford University , Stanford, California 94305, United States
- Late Stage Pharmaceutical Development and §Early Stage Pharmaceutical Development, Genentech , South San Francisco, California 94080, United States
| | - Ian C Shieh
- Department of Chemical Engineering, Stanford University , Stanford, California 94305, United States
- Late Stage Pharmaceutical Development and §Early Stage Pharmaceutical Development, Genentech , South San Francisco, California 94080, United States
| | - Danielle L Leiske
- Department of Chemical Engineering, Stanford University , Stanford, California 94305, United States
- Late Stage Pharmaceutical Development and §Early Stage Pharmaceutical Development, Genentech , South San Francisco, California 94080, United States
| | - Gerald G Fuller
- Department of Chemical Engineering, Stanford University , Stanford, California 94305, United States
- Late Stage Pharmaceutical Development and §Early Stage Pharmaceutical Development, Genentech , South San Francisco, California 94080, United States
| |
Collapse
|
116
|
Totality of Evidence and the Role of Clinical Studies in Establishing Biosimilarity. BIOSIMILARS 2018. [DOI: 10.1007/978-3-319-99680-6_22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
117
|
Dacoba TG, Olivera A, Torres D, Crecente-Campo J, Alonso MJ. Modulating the immune system through nanotechnology. Semin Immunol 2017; 34:78-102. [PMID: 29032891 PMCID: PMC5774666 DOI: 10.1016/j.smim.2017.09.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022]
Abstract
Nowadays, nanotechnology-based modulation of the immune system is presented as a cutting-edge strategy, which may lead to significant improvements in the treatment of severe diseases. In particular, efforts have been focused on the development of nanotechnology-based vaccines, which could be used for immunization or generation of tolerance. In this review, we highlight how different immune responses can be elicited by tuning nanosystems properties. In addition, we discuss specific formulation approaches designed for the development of anti-infectious and anti-autoimmune vaccines, as well as those intended to prevent the formation of antibodies against biologicals.
Collapse
Affiliation(s)
- Tamara G Dacoba
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Ana Olivera
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Dolores Torres
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - José Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain.
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain.
| |
Collapse
|
118
|
Bellinvia S, Ashraf M, Polosa R, Edwards C. A review article on biosimilar infliximab SB2 in the treatment of rheumatoid arthritis. Immunotherapy 2017; 9:1133-1142. [DOI: 10.2217/imt-2017-0068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
TNF inhibition has had a major impact as an approach for treating rheumatoid arthritis and a series of biologic agents directed against TNF have been developed for clinical use. Infliximab, a chimeric monoclonal antibody against soluble and membrane-bound TNF-α, was the biopharmaceutical to lead this ‘biologics revolution’. However, with expiration of patent protection of the originator medicinal product, biosimilar versions of infliximab have been developed through biosimilarity studies and randomized controlled trials aiming to assess pharmacokinetic, pharmacodynamic and clinical equivalence to their originator (reference product) in patients with moderate-to-severe disease activity. This review summarizes the clinical development of SB2, a biosimilar of infliximab, in rheumatoid arthritis.
Collapse
Affiliation(s)
- Salvatore Bellinvia
- Department of Rheumatology & NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton, Tremona Road, SO16 6YD, UK
- Institute of Internal & Emergency Medicine, Rheumatology Outpatient Clinic, Teaching Hospital “Policlinico - V. Emanuele”, University of Catania, Via Santa Sofia 78, 95123, Italy
| | - Madiha Ashraf
- Department of Rheumatology & NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton, Tremona Road, SO16 6YD, UK
| | - Riccardo Polosa
- Institute of Internal & Emergency Medicine, Rheumatology Outpatient Clinic, Teaching Hospital “Policlinico - V. Emanuele”, University of Catania, Via Santa Sofia 78, 95123, Italy
| | - Christopher Edwards
- Department of Rheumatology & NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton, Tremona Road, SO16 6YD, UK
| |
Collapse
|
119
|
Wong HE, Huang CJ, Zhang Z. Amino acid misincorporation in recombinant proteins. Biotechnol Adv 2017; 36:168-181. [PMID: 29107148 DOI: 10.1016/j.biotechadv.2017.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/12/2017] [Accepted: 10/24/2017] [Indexed: 11/26/2022]
Abstract
Proteins provide the molecular basis for cellular structure, catalytic activity, signal transduction, and molecular transport in biological systems. Recombinant protein expression is widely used to prepare and manufacture novel proteins that serve as the foundation of many biopharmaceutical products. However, protein translation bioprocesses are inherently prone to low-level errors. These sequence variants caused by amino acid misincorporation have been observed in both native and recombinant proteins. Protein sequence variants impact product quality, and their presence can be exacerbated through cellular stress, overexpression, and nutrient starvation. Therefore, the cell line selection process, which is used in the biopharmaceutical industry, is not only directed towards maximizing productivity, but also focuses on selecting clones which yield low sequence variant levels, thereby proactively avoiding potentially inauspicious patient safety and efficacy outcomes. Here, we summarize a number of hallmark studies aimed at understanding the mechanisms of amino acid misincorporation, as well as exacerbating factors, and mitigation strategies. We also describe key advances in analytical technologies in the identification and quantification of sequence variants, and some practical considerations when using LC-MS/MS for detecting sequence variants.
Collapse
Affiliation(s)
- H Edward Wong
- Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, United States
| | - Chung-Jr Huang
- Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, United States
| | - Zhongqi Zhang
- Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, United States.
| |
Collapse
|
120
|
Cell based assay identifies TLR2 and TLR4 stimulating impurities in Interferon beta. Sci Rep 2017; 7:10490. [PMID: 28874687 PMCID: PMC5585229 DOI: 10.1038/s41598-017-09981-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 08/01/2017] [Indexed: 01/05/2023] Open
Abstract
Immunogenicity can have devastating consequences on the safety and efficacy of therapeutic proteins. Therefore, evaluating and mitigating the risk of product immunogenicity is critical for the development these products. This study, showed that Betaseron and Extavia, which are reported to be more immunogenic among IFNβ products in clinical usage, contain residual innate immune response modulating impurities (IIRMIs) capable of activating NF-κB and induced expression of inflammatory mediators. These IIRMIs were undetectable in Rebif or Avonex. The stimulatory effect was attributed solely to IIRMIs because it was evident in murine cells lacking the interferon receptor (IFNAR). The IIRMIs in Betaseron and Extavia triggered NF-κB activation in HEK-293 cells bearing TLR2 and TLR4 in MyD88 dependent manner. Importantly, the IIRMIs in Betaseron induced up-regulation of IL-6, IL-1β, and ccl5 in the skin of IFNAR knock out mice following subcutaneous administration. This indicates that trace level IIRMIs in Betaseron could contribute to the higher immunogenicity rates seen in clinics. Together these data suggest that cell based assays can reveal subtle but clinically relevant differences in IIRMIs following manufacturing changes or between products with the same active ingredients but different manufacturing processes. Appreciating these differences may inform immunogenicity risk assessments.
Collapse
|
121
|
Wakshull E, Quarmby V, Mahler HC, Rivers H, Jere D, Ramos M, Szczesny P, Bechtold-Peters K, Masli S, Gupta S. Advancements in Understanding Immunogenicity of Biotherapeutics in the Intraocular Space. AAPS JOURNAL 2017; 19:1656-1668. [PMID: 28795351 DOI: 10.1208/s12248-017-0128-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/21/2017] [Indexed: 01/08/2023]
Abstract
Therapeutic breakthroughs in a number of retinal degenerative diseases have come about through the development of biotherapeutics administered directly into the eye. As a consequence of their use, we have gained more insight into the immune privileged status of the eye and the various considerations that development, manufacturing, and use of these drugs require. It has been observed that therapeutic proteins injected into the vitreous can elicit an immune response resulting in the production of anti-drug antibodies (ADAs) which can have clinical consequences. This review includes discussion of the anatomy, physiology, and specific area of the eye that are targeted for drug administration. The various immunologic mechanisms involved in the immune responses to intraocularly administered protein are discussed. This review entails discussion on chemistry, manufacturing, and control (CMC) and formulation-related issues that may influence the risk of immunogenicity. Based on the available immunogenicity profile of the marketed intraocular drugs and their reported adverse events, the animal models and the translational gap from animals to human are discussed. Thus, the objective of this review article is to assess the factors that influence immunogenicity in relation to intraocular administration and the steps taken for mitigating immunogenicity risks.
Collapse
Affiliation(s)
- Eric Wakshull
- BioAnalytical Sciences Genentech, South San Francisco, California, USA
| | - Valerie Quarmby
- BioAnalytical Sciences Genentech, South San Francisco, California, USA
| | | | | | | | - Meg Ramos
- AbbVie, Preclinical Safety, North Chicago, Illinois, USA
| | | | | | | | - Swati Gupta
- Nonclinical and Translational Sciences, Allergan, Irvine, California, USA.
| |
Collapse
|
122
|
Challenges in Predicting Protein-Protein Interactions from Measurements of Molecular Diffusivity. Biophys J 2017; 111:1831-1842. [PMID: 27806265 DOI: 10.1016/j.bpj.2016.09.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/26/2016] [Accepted: 09/14/2016] [Indexed: 01/11/2023] Open
Abstract
Dynamic light scattering can be used to measure the diffusivity of a protein within a formulation. The dependence of molecular diffusivity on protein concentration (traditionally expressed in terms of the interaction parameter kD) is often used to infer whether protein-protein interactions are repulsive or attractive, resulting in solutions that are colloidally stable or unstable, respectively. However, a number of factors unrelated to intermolecular forces can also impact protein diffusion, complicating this interpretation. Here, we investigate the influence of multicomponent diffusion in a ternary protein-salt-water system on protein diffusion and kD in the context of Nernst-Planck theory. This analysis demonstrates that large changes in protein diffusivity with protein concentration can result even for hard-sphere systems in the absence of protein-protein interactions. In addition, we show that dynamic light scattering measurements of diffusivity made at low ionic strength cannot be reliably used to detect protein conformational changes. We recommend comparing experimentally determined kD values to theoretically predicted excluded-volume contributions, which will allow a more accurate assessment of protein-protein interactions.
Collapse
|
123
|
Uchino T, Miyazaki Y, Yamazaki T, Kagawa Y. Immunogenicity of protein aggregates of a monoclonal antibody generated by forced shaking stress with siliconized and nonsiliconized syringes in BALB/c mice. ACTA ACUST UNITED AC 2017. [PMID: 28639328 DOI: 10.1111/jphp.12765] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE In this study, we aimed to investigate the immunogenicity of protein aggregates of monoclonal antibodies (mAbs), generated by forced shaking stress with siliconized and nonsiliconized syringes in a mouse model. METHODS Samples were filled in siliconized and nonsiliconized syringes with shaking and headspace air. Characterization studies were performed using high-performance size-exclusion chromatography, nanoparticle tracking analysis, flow cytometry, micro-flow imaging and resonant mass measurement. The samples (10 or 100 μg) were subcutaneously injected into BALB/c mice for 21 days, and the anti-drug antibody (ADA) concentrations were monitored. KEY FINDINGS In samples shaken with siliconized syringes [SO (+)], large amounts of submicron and subvisible protein aggregates were formed by interactions with silicone oil droplets. The characteristics of protein aggregates differed between the mAb solution and shaken samples, which strongly indicates that silicone oil accelerates protein aggregation. When administered at low doses, the ADA concentration in all samples increased with repeated injections, and SO (+) induced the highest immunogenicity. However, when administered at high doses, ADA concentration decreased following prolonged repeated administration for tolerance. CONCLUSIONS These results indicated that mAb protein aggregation induced immunogenicity in mice, and SO (+) induced higher immunogenicity than samples shaken with nonsiliconized syringe.
Collapse
Affiliation(s)
- Tomonobu Uchino
- Department of Clinical Pharmaceutics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yasunori Miyazaki
- Department of Clinical Pharmaceutics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takuto Yamazaki
- Department of Clinical Pharmaceutics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yoshiyuki Kagawa
- Department of Clinical Pharmaceutics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
124
|
Kayser V, Françon A, Pinton H, Saluzzo JF, Trout BL. Rational design of rabies vaccine formulation for enhanced stability. Turk J Med Sci 2017; 47:987-995. [PMID: 28618756 DOI: 10.3906/sag-1610-82] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/12/2017] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND/AIM Vaccines are often lyophilized in order to retain their stability and efficacy for a longer period of time. However, the same lyophilization process may also cause a major degradation of the vaccine, especially during early phases of manufacturing, leading to a loss of potency of the product. Many viral diseases, such as rabies, are acute and fatal unless the vaccine is administered prior to exposure or the onset of symptoms in the case of postexposure treatment. MATERIALS AND METHODS We investigated the effect of lyophilization on the stability of the virus structure during rabies vaccine manufacturing using dynamic light scattering and transmission electron microscopy. RESULTS Our results indicate that some viruses lose their stability and efficacy in the course of lyophilization if the pH of the cell culture medium is controlled by solvated CO2 because the structure of the rabies virus is very sensitive to the solution pH: the virus either aggregates or its shape is deformed at low solution pH, whereas at high pH empty capsid shells are formed. CONCLUSION Based on our findings, we developed a new formulation for the rabies vaccine that is stable in different buffers owing to the prevention of pH upshift upon lyophilization.
Collapse
Affiliation(s)
- Veysel Kayser
- Department of Chemical Engineering, Faculty of Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Faculty of Pharmacy, The University of Sydney, Sydney, Australia
| | | | | | | | - Bernhardt L Trout
- Department of Chemical Engineering, Faculty of Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
125
|
Targeting ligand–receptor interactions for development of cancer therapeutics. Curr Opin Chem Biol 2017; 38:62-69. [DOI: 10.1016/j.cbpa.2017.03.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/14/2022]
|
126
|
Schultz HS, Reedtz-Runge SL, Bäckström BT, Lamberth K, Pedersen CR, Kvarnhammar AM. Quantitative analysis of the CD4+ T cell response to therapeutic antibodies in healthy donors using a novel T cell:PBMC assay. PLoS One 2017; 12:e0178544. [PMID: 28562666 PMCID: PMC5451071 DOI: 10.1371/journal.pone.0178544] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 05/15/2017] [Indexed: 12/22/2022] Open
Abstract
Many biopharmaceuticals (BPs) are known to be immunogenic in the clinic, which can result in modified pharmacokinetics, reduced efficacy, allergic reactions and anaphylaxis. During recent years, several technologies to predict immunogenicity have been introduced, but the predictive value is still considered low. Thus, there is an unmet medical need for optimization of such technologies. The generation of T cell dependent high affinity anti-drug antibodies plays a key role in clinical immunogenicity. This study aimed at developing and evaluating a novel in vitro T cell:PBMC assay for prediction of the immunogenicity potential of BPs. To this end, we assessed the ability of infliximab (anti-TNF-α), rituximab (anti-CD20), adalimumab (anti-TNF-α) and natalizumab (anti-α4-integrin), all showing immunogenicity in the clinic, to induce a CD4+ T cells response. Keyhole limpet hemocyanin (KLH) and cytomegalovirus pp65 protein (CMV) were included as neo-antigen and recall antigen positive controls, respectively. By analyzing 26 healthy donors having HLA-DRB1 alleles matching the European population, we calculated the frequency of responding donors, the magnitude of the response, and the frequency of BP-specific T cells, as measured by 3[H]-thymidine incorporation and ELISpot IL-2 secretion. KLH and CMV demonstrated a strong T cell response in all the donors analyzed. The frequency of responding donors to the BPs was 4% for infliximab, 8% for adalimumab, 19% for rituximab and 27% for natalizumab, which is compared to and discussed with their respective observed clinical immunogenicity. This study further complements predictive immunogenicity testing by quantifying the in vitro CD4+ T cell responses to different BPs. Even though the data generated using this modified method does not directly translate to the clinical situation, a high sensitivity and immunogenic potential of most BPs is demonstrated.
Collapse
Affiliation(s)
- Heidi S. Schultz
- Immunogenicity Prediction and Tolerance, Global Research, Novo Nordisk A/S, Måløv, Denmark
- * E-mail:
| | | | - B. Thomas Bäckström
- Immunogenicity Prediction and Tolerance, Global Research, Novo Nordisk A/S, Måløv, Denmark
| | - Kasper Lamberth
- Immunogenicity Prediction and Tolerance, Global Research, Novo Nordisk A/S, Måløv, Denmark
| | | | - Anne M. Kvarnhammar
- Immunogenicity Prediction and Tolerance, Global Research, Novo Nordisk A/S, Måløv, Denmark
| | | |
Collapse
|
127
|
Pang L, Macauley MS, Arlian BM, Nycholat CM, Paulson JC. Encapsulating an Immunosuppressant Enhances Tolerance Induction by Siglec-Engaging Tolerogenic Liposomes. Chembiochem 2017; 18:1226-1233. [PMID: 28231415 DOI: 10.1002/cbic.201600702] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Indexed: 12/29/2022]
Abstract
Unwanted antibody responses significantly impact human health, and current options for treating deleterious antibody responses largely rely on broad immunosuppressants that can compromise overall immunity. A desirable alternative is to induce antigen-specific immune tolerance. We have shown that co-presentation of antigen and ligands of B cell sialic acid-binding immunoglobulin-like lectins (Siglecs) on a liposomal nanoparticle induces antigen-specific tolerance. Although Siglec-engaging tolerance-inducing antigenic liposomes (STALs) induce robust B cell tolerance in naïve mice, the full potential of STALs requires long-term tolerance induction and suppression of an ongoing immune response. We hypothesized that STALs encapsulated with rapamycin (RAPA), an immunomodulator, could improve the efficacy of STALs and potentially enable their use in the context of immunological memory. Here, we showed that formulation of STALs with RAPA produced enhanced tolerance induction in naïve mice compared to STALs without RAPA but had minimal impact on inducing tolerance in previously sensitized mice. These findings indicate that the addition of immunomodulators to STALs could be beneficial in tolerance induction and support future development of STALs for the treatment of allergy and autoimmune diseases.
Collapse
Affiliation(s)
- Lijuan Pang
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA.,Department of Immunology and Microbiology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Matthew S Macauley
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Britni M Arlian
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA.,Department of Immunology and Microbiology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Corwin M Nycholat
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA.,Department of Immunology and Microbiology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA.,Department of Immunology and Microbiology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|
128
|
Prabakaran R, Goel D, Kumar S, Gromiha MM. Aggregation prone regions in human proteome: Insights from large-scale data analyses. Proteins 2017; 85:1099-1118. [DOI: 10.1002/prot.25276] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/10/2017] [Accepted: 02/24/2017] [Indexed: 12/25/2022]
Affiliation(s)
- R. Prabakaran
- Department of Biotechnology, Bhupat Jyoti Mehta School of Biosciences; Indian Institute of Technology Madras; Chennai 600036 India
| | - Dhruv Goel
- Department of Computer Science and Engineering; Motilal Nehru National Institute of Technology; Allahabad 211004 India
| | - Sandeep Kumar
- Biotherapeutics Pharmaceutical Sciences, Pfizer Inc; 700 Chesterfield Parkway West Chesterfield Missouri 63017, USA
| | - M. Michael Gromiha
- Department of Biotechnology, Bhupat Jyoti Mehta School of Biosciences; Indian Institute of Technology Madras; Chennai 600036 India
| |
Collapse
|
129
|
Thomson AS, Mai S, Byrne MP. A novel approach to characterize host cell proteins associated with therapeutic monoclonal antibodies. Biotechnol Bioeng 2017; 114:1208-1214. [DOI: 10.1002/bit.26256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/05/2017] [Accepted: 01/16/2017] [Indexed: 01/20/2023]
Affiliation(s)
- Andrew S. Thomson
- R&D Platform Technology & Science; GlaxoSmithKline; King of Prussia Pennsylvania 19406
| | - Shing Mai
- R&D Platform Technology & Science; GlaxoSmithKline; King of Prussia Pennsylvania 19406
| | - Michael P. Byrne
- R&D Platform Technology & Science; GlaxoSmithKline; King of Prussia Pennsylvania 19406
| |
Collapse
|
130
|
Smith C, Li Z, Holman R, Pan F, Campbell RA, Campana M, Li P, Webster JRP, Bishop S, Narwal R, Uddin S, van der Walle CF, Lu JR. Antibody adsorption on the surface of water studied by neutron reflection. MAbs 2017; 9:466-475. [PMID: 28353420 DOI: 10.1080/19420862.2016.1276141] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Surface and interfacial adsorption of antibody molecules could cause structural unfolding and desorbed molecules could trigger solution aggregation, resulting in the compromise of physical stability. Although antibody adsorption is important and its relevance to many mechanistic processes has been proposed, few techniques can offer direct structural information about antibody adsorption under different conditions. The main aim of this study was to demonstrate the power of neutron reflection to unravel the amount and structural conformation of the adsorbed antibody layers at the air/water interface with and without surfactant, using a monoclonal antibody 'COE-3' as the model. By selecting isotopic contrasts from different ratios of H2O and D2O, the adsorbed amount, thickness and extent of the immersion of the antibody layer could be determined unambiguously. Upon mixing with the commonly-used non-ionic surfactant Polysorbate 80 (Tween 80), the surfactant in the mixed layer could be distinguished from antibody by using both hydrogenated and deuterated surfactants. Neutron reflection measurements from the co-adsorbed layers in null reflecting water revealed that, although the surfactant started to remove antibody from the surface at 1/100 critical micelle concentration (CMC) of the surfactant, complete removal was not achieved until above 1/10 CMC. The neutron study also revealed that antibody molecules retained their globular structure when either adsorbed by themselves or co-adsorbed with the surfactant under the conditions studied.
Collapse
Affiliation(s)
- Charles Smith
- a Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester , Manchester , UK
| | - Zongyi Li
- a Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester , Manchester , UK
| | - Robert Holman
- a Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester , Manchester , UK
| | - Fang Pan
- a Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester , Manchester , UK
| | | | - Mario Campana
- c ISIS Neutron Facility, STFC , Chilton, Didcot , UK
| | - Peixun Li
- c ISIS Neutron Facility, STFC , Chilton, Didcot , UK
| | | | - Steven Bishop
- d Formulation Sciences, MedImmune LLC , Gaithersburg , MD , USA
| | | | - Shahid Uddin
- e Formulation Sciences , MedImmune Ltd , Cambridge , UK
| | | | - Jian R Lu
- a Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester , Manchester , UK
| |
Collapse
|
131
|
Cheung CSF, Anderson KW, Patel PM, Cade KL, Phinney KW, Turko IV. A new approach to quantification of mAb aggregates using peptide affinity probes. Sci Rep 2017; 7:42497. [PMID: 28186164 PMCID: PMC5301252 DOI: 10.1038/srep42497] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/11/2017] [Indexed: 12/19/2022] Open
Abstract
Using mAbs as therapeutic molecules is complicated by the propensity of mAbs to aggregate at elevated concentrations, which can lead to a variety of adverse events in treatment. Here, we describe a proof-of-concept for new methodology to detect and quantify mAb aggregation. Assay development included using an aggregated mAb as bait for screening of phage display peptide library and identifying those peptides with random sequence which can recognize mAb aggregates. Once identified, the selected peptides can be used for developing quantitative methods to assess mAb aggregation. Results indicate that a peptide binding method coupled with mass spectrometric detection of bound peptide can quantify mAb aggregation and potentially be useful for monitoring aggregation propensity of therapeutic protein candidates.
Collapse
Affiliation(s)
- Crystal S F Cheung
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.,Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - Kyle W Anderson
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.,Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - Pooja M Patel
- Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - Keale L Cade
- Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - Karen W Phinney
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Illarion V Turko
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.,Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| |
Collapse
|
132
|
Salazar-Fontana LI, Desai DD, Khan TA, Pillutla RC, Prior S, Ramakrishnan R, Schneider J, Joseph A. Approaches to Mitigate the Unwanted Immunogenicity of Therapeutic Proteins during Drug Development. AAPS JOURNAL 2017; 19:377-385. [DOI: 10.1208/s12248-016-0030-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/15/2016] [Indexed: 12/17/2022]
|
133
|
Emery P, Vencovský J, Sylwestrzak A, Leszczyński P, Porawska W, Baranauskaite A, Tseluyko V, Zhdan VM, Stasiuk B, Milasiene R, Barrera Rodriguez AA, Cheong SY, Ghil J. A phase III randomised, double-blind, parallel-group study comparing SB4 with etanercept reference product in patients with active rheumatoid arthritis despite methotrexate therapy. Ann Rheum Dis 2017; 76:51-57. [PMID: 26150601 PMCID: PMC5264222 DOI: 10.1136/annrheumdis-2015-207588] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/11/2015] [Accepted: 06/06/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To compare the efficacy and safety of SB4 (an etanercept biosimilar) with reference product etanercept (ETN) in patients with moderate to severe rheumatoid arthritis (RA) despite methotrexate (MTX) therapy. METHODS This is a phase III, randomised, double-blind, parallel-group, multicentre study with a 24-week primary endpoint. Patients with moderate to severe RA despite MTX treatment were randomised to receive weekly dose of 50 mg of subcutaneous SB4 or ETN. The primary endpoint was the American College of Rheumatology 20% (ACR20) response at week 24. Other efficacy endpoints as well as safety, immunogenicity and pharmacokinetic parameters were also measured. RESULTS 596 patients were randomised to either SB4 (N=299) or ETN (N=297). The ACR20 response rate at week 24 in the per-protocol set was 78.1% for SB4 and 80.3% for ETN. The 95% CI of the adjusted treatment difference was -9.41% to 4.98%, which is completely contained within the predefined equivalence margin of -15% to 15%, indicating therapeutic equivalence between SB4 and ETN. Other efficacy endpoints and pharmacokinetic endpoints were comparable. The incidence of treatment-emergent adverse events was comparable (55.2% vs 58.2%), and the incidence of antidrug antibody development up to week 24 was lower in SB4 compared with ETN (0.7% vs 13.1%). CONCLUSIONS SB4 was shown to be equivalent with ETN in terms of efficacy at week 24. SB4 was well tolerated with a lower immunogenicity profile. The safety profile of SB4 was comparable with that of ETN. TRIAL REGISTRATION NUMBERS NCT01895309, EudraCT 2012-005026-30.
Collapse
Affiliation(s)
- Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | | | | | | | | | - Vira Tseluyko
- Kharkiv Medical Academy of Postgraduate Education, Kharkiv, Ukraine
| | | | | | | | | | | | - Jeehoon Ghil
- Samsung Bioepis Co., Ltd., Incheon, Republic of Korea
| |
Collapse
|
134
|
Hong J, Lee Y, Lee C, Eo S, Kim S, Lee N, Park J, Park S, Seo D, Jeong M, Lee Y, Yeon S, Bou-Assaf G, Sosic Z, Zhang W, Jaquez O. Physicochemical and biological characterization of SB2, a biosimilar of Remicade® (infliximab). MAbs 2016; 9:364-382. [PMID: 28005456 PMCID: PMC5297515 DOI: 10.1080/19420862.2016.1264550] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A biosimilar is a biological medicinal product that contains a version of the active substance of an already authorized original biological medicinal product. Biosimilarity to the reference product (RP) in terms of quality characteristics, such as physicochemical and biological properties, safety, and efficacy, based on a comprehensive comparability exercise needs to be established. SB2 (Flixabi® and Renflexis®) is a biosimilar to Remicade® (infliximab). The development of SB2 was performed in accordance with relevant guidelines of the International Conference on Harmonisation, the European Medicines Agency, and the United States Food and Drug Administration. To determine whether critical quality attributes meet quality standards, an extensive characterization test was performed with more than 80 lots of EU- and US-sourced RP. The physicochemical characterization study results revealed that SB2 was similar to the RP. Although a few differences in physicochemical attributes were observed, the evidence from the related literature, structure-activity relationship studies, and comparative biological assays showed that these differences were unlikely to be clinically meaningful. The biological characterization results showed that SB2 was similar to the RP in terms of tumor necrosis factor–α (TNF-α) binding and TNF-α neutralization activities as a main mode of action. SB2 was also similar in Fc-related biological activities including antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity, neonatal Fc receptor binding, C1q binding, and Fc gamma receptor binding activities. These analytical findings support that SB2 is similar to the RP and also provide confidence of biosimilarity in terms of clinical safety and efficacy.
Collapse
Affiliation(s)
- Juyong Hong
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Yuhwa Lee
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Changsoo Lee
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Suhyeon Eo
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Soyeon Kim
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Nayoung Lee
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Jongmin Park
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Seungkyu Park
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Donghyuck Seo
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Min Jeong
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Youngji Lee
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - Soojeong Yeon
- a Quality Evaluation Team, Samsung Bioepis Co., Ltd , Incheon , South Korea
| | - George Bou-Assaf
- b Department of Analytical Development , Biogen, Inc. , Cambridge , MA , USA
| | - Zoran Sosic
- b Department of Analytical Development , Biogen, Inc. , Cambridge , MA , USA
| | - Wei Zhang
- b Department of Analytical Development , Biogen, Inc. , Cambridge , MA , USA
| | - Orlando Jaquez
- c Department of Medical Affairs , Biosimilars , Biogen , Zug , Switzerland
| |
Collapse
|
135
|
Burmester GR, Choy E, Kivitz A, Ogata A, Bao M, Nomura A, Lacey S, Pei J, Reiss W, Pethoe-Schramm A, Mallalieu NL, Wallace T, Michalska M, Birnboeck H, Stubenrauch K, Genovese MC. Low immunogenicity of tocilizumab in patients with rheumatoid arthritis. Ann Rheum Dis 2016; 76:1078-1085. [PMID: 28007755 DOI: 10.1136/annrheumdis-2016-210297] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/22/2016] [Accepted: 11/26/2016] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Subcutaneous (SC) and intravenous formulations of tocilizumab (TCZ) are available for the treatment of patients with rheumatoid arthritis (RA), based on the efficacy and safety observed in clinical trials. Anti-TCZ antibody development and its impact on safety and efficacy were evaluated in adult patients with RA treated with intravenous TCZ (TCZ-IV) or TCZ-SC as monotherapy or in combination with conventional synthetic disease-modifying antirheumatic drugs (csDMARDs). METHODS Data from 5 TCZ-SC and 8 TCZ-IV phase III clinical trials and 1 TCZ-IV clinical pharmacology safety study (>50 000 samples) were pooled to assess the immunogenicity profile of TCZ-SC and TCZ-IV (8974 total patients). The analysis included antidrug antibody (ADA) measurement following TCZ-SC or TCZ-IV treatment as monotherapy or in combination with csDMARDs, after dosing interruptions or in TCZ-washout samples, and the correlation of ADAs with clinical response, adverse events or pharmacokinetics (PK). RESULTS The proportion of patients who developed ADAs following TCZ-SC or TCZ-IV treatment was 1.5% and 1.2%, respectively. ADA development was also comparable between patients who received TCZ monotherapy and those who received concomitant csDMARDs (0.7-2.0%). ADA development did not correlate with PK or safety events, including anaphylaxis, hypersensitivity or injection-site reactions, and no patients who developed ADAs had loss of efficacy. CONCLUSIONS The immunogenicity risk of TCZ-SC and TCZ-IV treatment was low, either as monotherapy or in combination with csDMARDs. Anti-TCZ antibodies developed among the small proportion of patients had no evident impact on PK, efficacy or safety.
Collapse
Affiliation(s)
- Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Free University and Humboldt University of Berlin, Berlin, Germany
| | | | - Alan Kivitz
- Altoona Center for Clinical Research, Duncansville, Pennsylvania, USA
| | - Atsushi Ogata
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Osaka, Japan.,Division of Allergy, Rheumatology and Connective Tissue Diseases, Department of Internal Medicine, NTT West Osaka Hospital, Osaka, Japan
| | - Min Bao
- Genentech, Inc., South San Francisco, California, USA
| | | | | | - Jinglan Pei
- Genentech, Inc., South San Francisco, California, USA
| | - William Reiss
- Genentech, Inc., South San Francisco, California, USA
| | | | | | | | | | - Herbert Birnboeck
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Kay Stubenrauch
- Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Mark C Genovese
- Division of Immunology and Rheumatology, Stanford University Medical Center, Palo Alto, California, USA
| |
Collapse
|
136
|
Kintzing JR, Filsinger Interrante MV, Cochran JR. Emerging Strategies for Developing Next-Generation Protein Therapeutics for Cancer Treatment. Trends Pharmacol Sci 2016; 37:993-1008. [PMID: 27836202 PMCID: PMC6238641 DOI: 10.1016/j.tips.2016.10.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/11/2016] [Accepted: 10/11/2016] [Indexed: 12/12/2022]
Abstract
Protein-based therapeutics have been revolutionizing the oncology space since they first appeared in the clinic two decades ago. Unlike traditional small-molecule chemotherapeutics, protein biologics promote active targeting of cancer cells by binding to cell-surface receptors and other markers specifically associated with or overexpressed on tumors versus healthy tissue. While the first approved cancer biologics were monoclonal antibodies, the burgeoning field of protein engineering is spawning research on an expanded range of protein formats and modifications that allow tuning of properties such as target-binding affinity, serum half-life, stability, and immunogenicity. In this review we highlight some of these strategies and provide examples of modified and engineered proteins under development as preclinical and clinical-stage drug candidates for the treatment of cancer.
Collapse
Affiliation(s)
- James R Kintzing
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Stanford Cancer Institute, Stanford, CA, USA
| | - Maria V Filsinger Interrante
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Stanford Cancer Institute, Stanford, CA, USA
| | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Stanford Cancer Institute, Stanford, CA, USA; Department of Chemical Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
137
|
Identification of an IgG CDR sequence contributing to co-purification of the host cell protease cathepsin D. Biotechnol Prog 2016; 33:140-145. [DOI: 10.1002/btpr.2397] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/30/2016] [Indexed: 11/07/2022]
|
138
|
Jacobs JF, Wevers RA, Lefeber DJ, van Scherpenzeel M. Fast, robust and high-resolution glycosylation profiling of intact monoclonal IgG antibodies using nanoLC-chip-QTOF. Clin Chim Acta 2016; 461:90-7. [DOI: 10.1016/j.cca.2016.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 12/31/2022]
|
139
|
Li W, Prabakaran P, Chen W, Zhu Z, Feng Y, Dimitrov DS. Antibody Aggregation: Insights from Sequence and Structure. Antibodies (Basel) 2016; 5:antib5030019. [PMID: 31558000 PMCID: PMC6698864 DOI: 10.3390/antib5030019] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/03/2016] [Accepted: 08/04/2016] [Indexed: 12/12/2022] Open
Abstract
Monoclonal antibodies (mAbs) are the fastest-growing biological therapeutics with important applications ranging from cancers, autoimmunity diseases and metabolic disorders to emerging infectious diseases. Aggregation of mAbs continues to be a major problem in their developability. Antibody aggregation could be triggered by partial unfolding of its domains, leading to monomer-monomer association followed by nucleation and growth. Although the aggregation propensities of antibodies and antibody-based proteins can be affected by the external experimental conditions, they are strongly dependent on the intrinsic antibody properties as determined by their sequences and structures. In this review, we describe how the unfolding and aggregation susceptibilities of IgG could be related to their cognate sequences and structures. The impact of antibody domain structures on thermostability and aggregation propensities, and effective strategies to reduce aggregation are discussed. Finally, the aggregation of antibody-drug conjugates (ADCs) as related to their sequence/structure, linker payload, conjugation chemistry and drug-antibody ratio (DAR) is reviewed.
Collapse
Affiliation(s)
- Wei Li
- Protein Interactions Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| | | | - Weizao Chen
- Protein Interactions Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| | - Zhongyu Zhu
- Protein Interactions Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| | - Yang Feng
- Protein Interactions Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| | - Dimiter S Dimitrov
- Protein Interactions Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| |
Collapse
|
140
|
Unraveling the Effect of Immunogenicity on the PK/PD, Efficacy, and Safety of Therapeutic Proteins. J Immunol Res 2016; 2016:2342187. [PMID: 27579329 PMCID: PMC4992793 DOI: 10.1155/2016/2342187] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/12/2016] [Indexed: 12/14/2022] Open
Abstract
Biologics have emerged as a powerful and diverse class of molecular and cell-based therapies that are capable of replacing enzymes, editing genomes, targeting tumors, and more. As this complex array of tools arises a distinct set of challenges is rarely encountered in the development of small molecule therapies. Biotherapeutics tend to be big, bulky, polar molecules comprised of protein and/or nucleic acids. Compared to their small molecule counterparts, they are fragile, labile, and heterogeneous. Their biodistribution is often limited by hydrophobic barriers which often restrict their administration to either intravenous or subcutaneous entry routes. Additionally, their potential for immunogenicity has proven to be a challenge to developing safe and reliably efficacious drugs. Our discussion will emphasize immunogenicity in the context of therapeutic proteins, a well-known class of biologics. We set out to describe what is known and unknown about the mechanisms underlying the interplay between antigenicity and immune response and their effect on the safety, efficacy, pharmacokinetics, and pharmacodynamics of these therapeutic agents.
Collapse
|
141
|
Joubert MK, Deshpande M, Yang J, Reynolds H, Bryson C, Fogg M, Baker MP, Herskovitz J, Goletz TJ, Zhou L, Moxness M, Flynn GC, Narhi LO, Jawa V. Use of In Vitro Assays to Assess Immunogenicity Risk of Antibody-Based Biotherapeutics. PLoS One 2016; 11:e0159328. [PMID: 27494246 PMCID: PMC4975389 DOI: 10.1371/journal.pone.0159328] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 06/30/2016] [Indexed: 12/29/2022] Open
Abstract
An In Vitro Comparative Immunogenicity Assessment (IVCIA) assay was evaluated as a tool for predicting the potential relative immunogenicity of biotherapeutic attributes. Peripheral blood mononuclear cells from up to 50 healthy naïve human donors were monitored up to 8 days for T-cell proliferation, the number of IL-2 or IFN-γ secreting cells, and the concentration of a panel of secreted cytokines. The response in the assay to 10 monoclonal antibodies was found to be in agreement with the clinical immunogenicity, suggesting that the assay might be applied to immunogenicity risk assessment of antibody biotherapeutic attributes. However, the response in the assay is a measure of T-cell functional activity and the alignment with clinical immunogenicity depends on several other factors. The assay was sensitive to sequence variants and could differentiate single point mutations of the same biotherapeutic. Nine mAbs that were highly aggregated by stirring induced a higher response in the assay than the original mAbs before stirring stress, in a manner that did not match the relative T-cell response of the original mAbs. In contrast, mAbs that were glycated by different sugars (galactose, glucose, and mannose) showed little to no increase in response in the assay above the response to the original mAbs before glycation treatment. The assay was also used successfully to assess similarity between multiple lots of the same mAb, both from the same manufacturer and from different manufacturers (biosimilars). A strategy for using the IVCIA assay for immunogenicity risk assessment during the entire lifespan development of biopharmaceuticals is proposed.
Collapse
Affiliation(s)
- Marisa K. Joubert
- Department of Attribute Sciences, Amgen Inc., Thousand Oaks, California, United States of America
- * E-mail: (MJ); (VJ)
| | - Meghana Deshpande
- Department of Clinical Immunology, Amgen Inc., Thousand Oaks, California, United States of America
| | - Jane Yang
- Department of Attribute Sciences, Amgen Inc., Thousand Oaks, California, United States of America
| | - Helen Reynolds
- Antitope Limited, Babraham Research Campus, Cambridge, United Kingdom
| | - Christine Bryson
- Antitope Limited, Babraham Research Campus, Cambridge, United Kingdom
| | - Mark Fogg
- Antitope Limited, Babraham Research Campus, Cambridge, United Kingdom
| | - Matthew P. Baker
- Antitope Limited, Babraham Research Campus, Cambridge, United Kingdom
| | - Jonathan Herskovitz
- Department of Clinical Immunology, Amgen Inc., Thousand Oaks, California, United States of America
| | - Theresa J. Goletz
- Department of Clinical Immunology, Amgen Inc., Seattle, Washington, United States of America
| | - Lei Zhou
- Department of Medical Sciences, Amgen Inc., Thousand Oaks, California, United States of America
| | - Michael Moxness
- Department of Clinical Immunology, Amgen Inc., Thousand Oaks, California, United States of America
| | - Gregory C. Flynn
- Department of Attribute Sciences, Amgen Inc., Thousand Oaks, California, United States of America
| | - Linda O. Narhi
- Department of Attribute Sciences, Amgen Inc., Thousand Oaks, California, United States of America
| | - Vibha Jawa
- Department of Clinical Immunology, Amgen Inc., Thousand Oaks, California, United States of America
- * E-mail: (MJ); (VJ)
| |
Collapse
|
142
|
Shah CA. ‘Lower anti-drug antibodies with etanercept biosimilar: can Ctrough explain the differences?’. Ann Rheum Dis 2016; 75:e60. [DOI: 10.1136/annrheumdis-2016-210089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 11/04/2022]
|
143
|
Broomfield A, Jones SA, Hughes SM, Bigger BW. The impact of the immune system on the safety and efficiency of enzyme replacement therapy in lysosomal storage disorders. J Inherit Metab Dis 2016; 39:499-512. [PMID: 26883220 DOI: 10.1007/s10545-016-9917-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/27/2016] [Accepted: 01/29/2016] [Indexed: 12/31/2022]
Abstract
In the light of clinical experience in infantile onset Pompe patients, the immunological impact on the tolerability and long-term efficacy of enzyme replacement therapy (ERT) for lysosomal storage disorders has come under renewed scrutiny. This article details the currently proposed immunological mechanisms involved in the development of anti-drug antibodies and the current therapies used in their treatment. Given the current understanding of the adaptive immune response, it focuses particularly on T cell dependent mechanisms and the paradigm of using lymphocytic negative selection as a predictor of antibody formation. This concept originally postulated in the 1970s, stipulated that the genotypically determined lack of production or production of a variant protein determines an individual's lymphocytic repertoire. This in turn is the key factor in determining the potential severity of an individual's immunological response to ERT. It also highlights the need for immunological assay standardization particularly those looking at describing the degree of functional impact, robust biochemical or clinical endpoints and detailed patient subgroup identification if the true evaluations of impact are to be realised.
Collapse
Affiliation(s)
- A Broomfield
- Willink Biochemical genetics unit, Manchester center for genomic medicine, St Mary's Hospital, Central Manchester Foundation Trust, Manchester, M13 9WL, UK.
| | - S A Jones
- Willink Biochemical genetics unit, Manchester center for genomic medicine, St Mary's Hospital, Central Manchester Foundation Trust, Manchester, M13 9WL, UK
| | - S M Hughes
- Department of Immunology, Royal Manchester children's Hospital, Central Manchester Foundation Trust, Manchester, M13 9WL, UK
| | - B W Bigger
- Stem Cell & Neurotherapies Laboratory, Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
144
|
Ratanji KD, Dearman RJ, Kimber I, Thorpe R, Wadhwa M, Derrick JP. Editor's Highlight: Subvisible Aggregates of Immunogenic Proteins Promote a Th1-Type Response. Toxicol Sci 2016; 153:258-70. [PMID: 27370416 PMCID: PMC5036615 DOI: 10.1093/toxsci/kfw121] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Protein aggregation is associated with enhanced immunogenicity of biotherapeutics. As a result, regulatory guidelines recommend screening for aggregation during bioprocessing. However, the mechanisms underlying the enhanced immunogenicity of aggregates are poorly understood. In the investigations described herein, the immunogenicity in mice of a humanized single chain variable antibody fragment (scFv) purified after expression in Escherichia coli has been examined. Reproducible scFv aggregates were obtained within the subvisible particle size range (mean diameter 2 µm) using thermal and mechanical stresses. Intraperitoneal immunization of BALB/c strain mice with 1 mg/ml of aggregated or monomeric scFv induced similar IgG and IgG1 antibody responses. In contrast, aggregate preparations stimulated significantly higher levels of anti-scFv IgG2a antibody than did the monomer. In comparative studies, aggregates of ovalbumin (OVA) within the subvisible particle size range were prepared by stir stress, and their immunogenicity compared with that of monomeric OVA in mice. Aggregated and monomeric OVA induced similar anti-OVA IgG and IgG1 antibody responses, whereas IgG2a antibody levels were significantly higher in aggregate-immunized mice. Furthermore, cytokine profiles in supernatants taken from splenocyte-dendritic cell co-cultures were consistent with aggregated preparations inducing a T helper (Th) 1-type response. Aggregated proteins within the subvisible range were therefore shown to induce a preferential Th1 type response, whereas monomeric proteins elicited a selective Th2 response. These data indicate that protein aggregation can impact on both the vigor and quality of immune responses.
Collapse
Affiliation(s)
- Kirsty D Ratanji
- *Faculty of Life Sciences, The University of Manchester, C1256, Michael Smith Building, Dover St, Manchester M13 9PT, UK
| | - Rebecca J Dearman
- *Faculty of Life Sciences, The University of Manchester, C1256, Michael Smith Building, Dover St, Manchester M13 9PT, UK
| | - Ian Kimber
- *Faculty of Life Sciences, The University of Manchester, C1256, Michael Smith Building, Dover St, Manchester M13 9PT, UK
| | - Robin Thorpe
- National Institute for Biological Standards and Control, Potters Bar, Hertfordshire EN6 3QG
| | - Meenu Wadhwa
- National Institute for Biological Standards and Control, Potters Bar, Hertfordshire EN6 3QG
| | - Jeremy P Derrick
- *Faculty of Life Sciences, The University of Manchester, C1256, Michael Smith Building, Dover St, Manchester M13 9PT, UK;
| |
Collapse
|
145
|
Immunogenicity of Biotherapeutics: Causes and Association with Posttranslational Modifications. J Immunol Res 2016; 2016:1298473. [PMID: 27437405 PMCID: PMC4942633 DOI: 10.1155/2016/1298473] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/09/2016] [Accepted: 06/12/2016] [Indexed: 12/21/2022] Open
Abstract
Today, potential immunogenicity can be better evaluated during the drug development process, and we have rational approaches to manage the clinical consequences of immunogenicity. The focus of the scientific community should be on developing sensitive diagnostics that can predict immunogenicity-mediated adverse events in the small fraction of subjects that develop clinically relevant anti-drug antibodies. Here, we discuss the causes of immunogenicity which could be product-related (inherent property of the product or might be picked up during the manufacturing process), patient-related (genetic profile or eating habits), or linked to the route of administration. We describe various posttranslational modifications (PTMs) and how they may influence immunogenicity. Over the last three decades, we have significantly improved our understanding about the types of PTMs of biotherapeutic proteins and their association with immunogenicity. It is also now clear that all PTMs do not lead to clinical immunogenicity. We also discuss the mechanisms of immunogenicity (which include T cell-dependent and T cell-independent responses) and immunological tolerance. We further elaborate on the management of immunogenicity in preclinical and clinical setting and the unique challenges raised by biosimilars, which may have different immunogenic potential from their parent biotherapeutics.
Collapse
|
146
|
Lauer TM, Wood GPF, Farkas D, Sathish HA, Samra HS, Trout BL. Molecular Investigation of the Mechanism of Non-Enzymatic Hydrolysis of Proteins and the Predictive Algorithm for Susceptibility. Biochemistry 2016; 55:3315-28. [PMID: 27194363 DOI: 10.1021/acs.biochem.5b01376] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Timothy M. Lauer
- Department
of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Geoffrey P. F. Wood
- Department
of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - David Farkas
- Department
of Formulation Sciences, MedImmune LLC, Gaithersburg, Maryland 20878, United States
| | - Hasige A. Sathish
- Department
of Formulation Sciences, MedImmune LLC, Gaithersburg, Maryland 20878, United States
| | - Hardeep S. Samra
- Department
of Formulation Sciences, MedImmune LLC, Gaithersburg, Maryland 20878, United States
| | - Bernhardt L. Trout
- Department
of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
147
|
Gould J, Callis CM, Dolan DG, Stanard B, Weideman PA. Special endpoint and product specific considerations in pharmaceutical acceptable daily exposure derivation. Regul Toxicol Pharmacol 2016; 79 Suppl 1:S79-93. [PMID: 27233924 DOI: 10.1016/j.yrtph.2016.05.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/19/2016] [Indexed: 12/12/2022]
Abstract
Recently, a guideline has been published by the European Medicines Agency (EMA) on setting safe limits, permitted daily exposures (PDE) [also called acceptable daily exposures (ADE)], for medicines manufactured in multi-product facilities. The ADE provides a safe exposure limit for inadvertent exposure of a drug due to cross-contamination in manufacturing. The ADE determination encompasses a standard risk assessment, requiring an understanding of the toxicological and pharmacological effects, the mechanism of action, drug compound class, and the dose-response as well as the pharmacokinetic properties of the compound. While the ADE concept has broad application in pharmaceutical safety there are also nuances and specific challenges associated with some toxicological endpoints or drug product categories. In this manuscript we discuss considerations for setting ADEs when the following specific adverse health endpoints may constitute the critical effect: genotoxicity, developmental and reproductive toxicity (DART), and immune system modulation (immunostimulation or immunosuppression), and for specific drug classes, including antibody drug conjugates (ADCs), emerging medicinal therapeutic compounds, and compounds with limited datasets. These are challenging toxicological scenarios that require a careful evaluation of all of the available information in order to establish a health-based safe level.
Collapse
|
148
|
Lee YJ, Shin D, Kim Y, Kang J, Gauliard A, Fuhr R. A randomized phase l pharmacokinetic study comparing SB4 and etanercept reference product (Enbrel®) in healthy subjects. Br J Clin Pharmacol 2016; 82:64-73. [PMID: 26972584 PMCID: PMC4917797 DOI: 10.1111/bcp.12929] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 02/24/2016] [Accepted: 03/07/2016] [Indexed: 01/13/2023] Open
Abstract
Aims SB4 has been developed as a biosimilar of etanercept. The primary objective of the present study was to demonstrate the pharmacokinetic (PK) equivalence between SB4 and European Union ‐sourced etanercept (EU‐ETN), SB4 and United States‐sourced etanercept (US‐ETN), and EU‐ETN and US‐ETN. The safety and immunogenicity were also compared between the treatments. Methods This was a single‐blind, three‐part, crossover study in 138 healthy male subjects. In each part, 46 subjects were randomized at a 1:1 ratio to receive a single 50 mg subcutaneous dose of the treatments (part A: SB4 or EU‐ETN; part B: SB4 or US‐ETN; and part C: EU‐ETN or US‐ETN) in period 1, followed by the crossover treatment in period 2 according to their assigned sequences. PK equivalence between the treatments was determined using the standard equivalence margin of 80–125%. Results The geometric least squares means ratios of AUCinf, AUClast and Cmax were 99.04%, 98.62% and 103.71% (part A: SB4 vs. EU‐ETN); 101.09%, 100.96% and 104.36% (part B: SB4 vs. US‐ETN); and 100.51%, 101.27% and 103.29% (part C: EU‐ETN vs. US‐ETN), respectively, and the corresponding 90% confidence intervals were completely contained within the limits of 80–125 %. The incidence of treatment‐emergent adverse events was comparable, and the incidence of the antidrug antibodies was lower in SB4 compared with the reference products. Conclusions The present study demonstrated PK equivalence between SB4 and EU‐ETN, SB4 and US‐ETN, and EU‐ETN and US‐ETN in healthy male subjects. SB4 was well tolerated, with a lower immunogenicity profile and similar safety profile compared with those of the reference products.
Collapse
Affiliation(s)
- Yoon Jung Lee
- Samsung Bioepis Co., Ltd., Incheon, Republic of Korea
| | - Donghoon Shin
- Samsung Bioepis Co., Ltd., Incheon, Republic of Korea
| | - Youngdoe Kim
- Samsung Bioepis Co., Ltd., Incheon, Republic of Korea
| | - Jungwon Kang
- Samsung Bioepis Co., Ltd., Incheon, Republic of Korea
| | - Anke Gauliard
- PAREXEL International Early Phase Clinical Unit, Berlin, Germany
| | - Rainard Fuhr
- PAREXEL International Early Phase Clinical Unit, Berlin, Germany
| |
Collapse
|
149
|
Jiskoot W, Kijanka G, Randolph TW, Carpenter JF, Koulov AV, Mahler HC, Joubert MK, Jawa V, Narhi LO. Mouse Models for Assessing Protein Immunogenicity: Lessons and Challenges. J Pharm Sci 2016; 105:1567-1575. [PMID: 27044944 PMCID: PMC4846475 DOI: 10.1016/j.xphs.2016.02.031] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/06/2016] [Accepted: 02/18/2016] [Indexed: 12/11/2022]
Abstract
The success of clinical and commercial therapeutic proteins is rapidly increasing, but their potential immunogenicity is an ongoing concern. Most of the studies that have been conducted over the past few years to examine the importance of various product-related attributes (in particular several types of aggregates and particles) and treatment regimen (such as dose, dosing schedule, and route of administration) in the development of unwanted immune responses have utilized one of a variety of mouse models. In this review, we discuss the utility and drawbacks of different mouse models that have been used for this purpose. Moreover, we summarize the lessons these models have taught us and some of the challenges they present. Finally, we provide recommendations for future research utilizing mouse models to improve our understanding of critical factors that may contribute to protein immunogenicity.
Collapse
Affiliation(s)
- Wim Jiskoot
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, RA Leiden 2300, The Netherlands
| | - Grzegorz Kijanka
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, RA Leiden 2300, The Netherlands
| | - Theodore W Randolph
- Center for Pharmaceutical Biotechnology, Department of Chemical and Biological Engineering, University of Colorado - Boulder, Boulder, Colorado 80309
| | - John F Carpenter
- Center for Pharmaceutical Biotechnology, Department of Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Atanas V Koulov
- Pharma Technical Development (Europe) Biologics, Basel 4070, Switzerland
| | | | - Marisa K Joubert
- Amgen Inc., Process Development, Thousand Oaks, California 91320
| | - Vibha Jawa
- Amgen Inc., Medical Sciences, Thousand Oaks, California 91320
| | - Linda O Narhi
- Amgen Inc., Process Development, Thousand Oaks, California 91320.
| |
Collapse
|
150
|
Hjorth CF, Norrman M, Wahlund PO, Benie AJ, Petersen BO, Jessen CM, Pedersen TÅ, Vestergaard K, Steensgaard DB, Pedersen JS, Naver H, Hubálek F, Poulsen C, Otzen D. Structure, Aggregation, and Activity of a Covalent Insulin Dimer Formed During Storage of Neutral Formulation of Human Insulin. J Pharm Sci 2016; 105:1376-86. [DOI: 10.1016/j.xphs.2016.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/11/2015] [Accepted: 01/06/2016] [Indexed: 10/22/2022]
|