1
|
Dejager L, Banton S, Marques P, Rinikova G, Lory S, Hickford ES, Martin-Hamka C, Penney M, Grootjans S. BiSim Tool: a binding simulation tool to aid and simplify ligand-binding assay design and development. Bioanalysis 2024; 16:519-533. [PMID: 38629337 PMCID: PMC11299794 DOI: 10.4155/bio-2023-0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/29/2024] [Indexed: 08/02/2024] Open
Abstract
Ligand-binding assays (LBAs) rely on the reversible, noncovalent binding between the analyte of interest and the assay reagents, and understanding their dynamic equilibrium is key to building robust LBA methods. Although the dynamic interplay of free and bound fractions can be calculated using mathematical models, these are not routinely applied. This approach is costly in terms of both assay development time and reagents, and can result in an under-exploration of the possible parameter combinations. Therefore, we have created a user-friendly simulation tool to facilitate LBA development (the BiSim Tool). We describe the models driving the mathematical simulations and the main features of our software solution by means of case studies, illustrating the tool's value in drug development. To support drug development for all patients worldwide, the BiSim Tool is now available as an open-source code project and as a free web-based tool at https://proteinbindingsimulation.shinyapps.io/BiSim-ProteinBindingSimulation [1].
Collapse
Affiliation(s)
- Lien Dejager
- UCB Biopharma, Allée de la Recherche 60, 1070 Brussels, Belgium
| | - Sophia Banton
- UCB Biopharma, Allée de la Recherche 60, 1070 Brussels, Belgium
| | - Patricia Marques
- UCB Biopharma, Allée de la Recherche 60, 1070 Brussels, Belgium
- Employed by UCB at the time the work was undertaken
| | - Gabriela Rinikova
- UCB Biopharma, Allée de la Recherche 60, 1070 Brussels, Belgium
- Employed by UCB at the time the work was undertaken
| | - Sabrina Lory
- UCB Biopharma, Allée de la Recherche 60, 1070 Brussels, Belgium
| | | | | | - Mark Penney
- UCB Biopharma, Bath Road, Slough, SL1 3WE, UK
- Employed by UCB at the time the work was undertaken
| | | |
Collapse
|
2
|
Wen K, Dai W, Meng X, Lin Q, Wei J, Tong L, Taylor SK, Rudchenko SA, Stojanovic MN, Kalantarov G, Trakht I. Rapid isolation of anti-idiotype aptamers for quantification of human monoclonal antibodies against SARS-CoV-2 spike protein. Biosens Bioelectron 2024; 246:115842. [PMID: 38042051 PMCID: PMC10935567 DOI: 10.1016/j.bios.2023.115842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 12/04/2023]
Abstract
Therapeutic antibodies that block viral entry have already proven to be important, first line drugs for treatments of viral infections. In the case of SARS-CoV-2, combinations of multiple therapeutic antibodies may need to be rapidly identified and formulated in a way that blocks each new, predominant variant of the virus. For efficient introduction of any new antibody combination into patients, it is important to be able to monitor patient-specific pharmacokinetics of individual antibodies, which would include the time course of their specific capacity to block the viral spike proteins. Here, we present three examples of microfluidic-based rapid isolation of companion reagents useful for establishing combination antibody therapies. These reagents are specific three-dimensional imprints of variable regions of individual human monoclonal antibodies against the -spike protein of SARS-CoV-2 virus in the form of oligonucleotide-based ligands (aptamers). We implement these anti-idiotypic aptamers as bioreceptors in graphene-based field-effect transistor sensors to accomplish label free, rapid, and sensitive detection of matching antibodies within minutes. Through this work we have demonstrated the general applicability of anti-idiotype aptamers as capture reagents in quantification of active forms of monoclonal antibodies in complex biological mixtures.
Collapse
Affiliation(s)
- Kechun Wen
- Department of Mechanical Engineering, Columbia University, New York, NY, 10027, USA
| | - Wenting Dai
- Department of Mechanical Engineering, Columbia University, New York, NY, 10027, USA
| | - Xin Meng
- Department of Mechanical Engineering, Columbia University, New York, NY, 10027, USA
| | - Qiao Lin
- Department of Mechanical Engineering, Columbia University, New York, NY, 10027, USA.
| | - Jia Wei
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Liang Tong
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Steven K Taylor
- Division of Experimental Therapeutics, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Sergei A Rudchenko
- Division of Experimental Therapeutics, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Milan N Stojanovic
- Division of Experimental Therapeutics, Department of Medicine, Columbia University, New York, NY, 10032, USA; Departments of Biomedical Engineering and Systems Biology, Columbia University, New York, NY, 10032, USA.
| | - Gary Kalantarov
- Division of Experimental Therapeutics, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Ilya Trakht
- Division of Experimental Therapeutics, Department of Medicine, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
3
|
Li S, Guo H, Gao Y, Tian W, Wang S, Shen C, Xu L, Liu H, Zhang J, Wang Y. Development of a free cytokine immunoassay to maintain binding and dissociation equilibrium in vitro. J Pharm Biomed Anal 2024; 238:115813. [PMID: 37956554 DOI: 10.1016/j.jpba.2023.115813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/15/2023]
Abstract
Using competitive ELISA to detect free cytokines is limited as it can only reflect relative trends rather than accurately determine the real state and quantity of cytokines due to the dynamic equilibrium between dissociation and binding. This imprecise quantification adversely affects the usage of clinical medication and the validity assessment. In this study, we have developed a novel cytokine immunoassay that utilizes Rosetta molecular docking prediction technique, we screened two specific antibody pairs binding IL-1β and Durg respectively and then established the Total IL-1β and Total Drug ELISA assay. Protein A column could separate bound IL-1β and free IL-1β, and the bound IL-1β occupied for about 90% of the total. This innovative approach ensures the maintenance of equilibrium between the free cytokines and complex. We have developed a free cytokine content detection method that combines ELISA and solid phase extraction, which can detect the true concentration of free cytokines without destroying the free-binding dynamic equilibrium. It can be used to verify the accuracy of clinical PK/PD and other data, evaluate the applicability of detection methods, and guide clinical drug use and drug efficacy evaluation.
Collapse
Affiliation(s)
- Siqi Li
- GeneScience Pharmaceuticals Co, Ltd, Changchun 130012, China
| | - Hao Guo
- GeneScience Pharmaceuticals Co, Ltd, Changchun 130012, China
| | - Yan Gao
- GeneScience Pharmaceuticals Co, Ltd, Changchun 130012, China
| | - Wen Tian
- GeneScience Pharmaceuticals Co, Ltd, Changchun 130012, China
| | - Shan Wang
- GeneScience Pharmaceuticals Co, Ltd, Changchun 130012, China
| | - Chen Shen
- GeneScience Pharmaceuticals Co, Ltd, Changchun 130012, China
| | - Lili Xu
- GeneScience Pharmaceuticals Co, Ltd, Changchun 130012, China
| | - Hailong Liu
- GeneScience Pharmaceuticals Co, Ltd, Changchun 130012, China
| | - Jinliang Zhang
- School of Life Science, Jilin University, Changchun 130012, China; GeneScience Pharmaceuticals Co, Ltd, Changchun 130012, China
| | - Yingwu Wang
- School of Life Science, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun 130012, China.
| |
Collapse
|
4
|
Bouden S, Laadhar L, Soua J, Ben Messaoud M, Rouached L, Ayadi I, Saidane O, Ben Tekaya A, Mahmoud I, Rekik S, Srairi HS, Tekaya R, Bellakhal S, Fekih M, Abdelmoula L, Kallel M. No Correlation between Anti-drug Antibodies and Therapeutic Response in Tunisian Patients with Chronic Inflammatory Diseases Treated by TNF Blockers. Curr Rheumatol Rev 2024; 20:435-443. [PMID: 38314598 DOI: 10.2174/0115733971257151230919095431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 02/06/2024]
Abstract
INTRODUCTION Tumor necrosis factor alpha (TNF alpha) blockers such as infliximab (IFX) and adalimumab (ADA) had significantly changed the course of inflammatory diseases such as rheumatoid arthritis (RA), spondyloarthritis (SpA) and Crohn's disease (CD). However, about 30% of patients do not respond to these treatments. This lack of response may be due to the formation of antibodies against these drugs (anti-drug antibodies: ADAbs). The aim of this study was to determine the prevalence of ADAbs against IFX and ADA, and the trough serum concentration of IFX and ADA in RA, SpA or CD patients and to assess their impact on the therapeutic response. METHODS A cross sectional, multi-centric study was conducted, including patients with RA, SpA or CD treated with IFX or ADA as a first biotherapy for at least 6 months. ADAbs and trough levels were measured by an Enzyme Linked Immunosorbent assay (ELISA). RESULTS 197 patients were included (57 RA, 73 SpA and 67 CD). ADAbs were positive in 40% of cases for IFX and 25% for ADA. They were positive in 40% of SpA, 35% of RA, and 21% of CD. The presence of ADAbs was inversely correlated to the trough levels of IFX and ADA during RA (p = 0.01 and p < 0.0001), SpA (p < 0.01 and p < 0.0001) and CD (p = 0.001 and p = 0.04). For all pathologies, the presence of ADAbs was not correlated with disease activity. Concomitant methotrexate significantly reduced immunogenicity. CONCLUSION In our study, the presence of ADAb and low trough levels seem to not affect the therapeutic response in patients on TNF alpha antagonists. Other tracks more than immunogenicity should be investigated to explain the loss of response to these biotherapies.
Collapse
Affiliation(s)
- Selma Bouden
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
| | - Lilia Laadhar
- Department of Immunology, Rabta Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Jihene Soua
- Department of Rheumatology, Charles Nicoles Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Meriam Ben Messaoud
- Department of Rheumatology, Charles Nicoles Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Leila Rouached
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
| | - Imene Ayadi
- Department of Immunology, Rabta Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Olfa Saidane
- Department of Rheumatology, Charles Nicoles Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Aicha Ben Tekaya
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
| | - Ines Mahmoud
- Department of Rheumatology, Charles Nicoles Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Sonia Rekik
- Department of Immunology, Rabta Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Héla Sahli Srairi
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
| | - Rawdha Tekaya
- Department of Rheumatology, Charles Nicoles Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Syrine Bellakhal
- Department of Internal Medecine, FSI Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Monia Fekih
- Department of Gastro-enterology, Rabta Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Leila Abdelmoula
- Department of Rheumatology, Charles Nicoles Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Maryem Kallel
- Department of Immunology, Rabta Hospital, Tunis El Manar University, Tunis, Tunisia
| |
Collapse
|
5
|
Shim J, Chen J, Carrasco-Triguero M, Fischer SK. Overcoming Soluble Target Interference in Measurement of Total Bispecific Therapeutic Antibody Concentrations. AAPS J 2023; 25:82. [PMID: 37594571 DOI: 10.1208/s12248-023-00848-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/04/2023] [Indexed: 08/19/2023] Open
Abstract
The measurement of therapeutic drug concentrations is used to assess drug exposure and the relationship between therapeutic pharmacokinetics (PK) and pharmacodynamics (PD), which help determine the optimal dose for patients. Ligand binding assays (LBAs) are often the method of choice for evaluation of drug concentration and use either the therapeutic target protein or antibodies to the therapeutic as capture and/or detection reagents. Due to the bivalency of antibody therapeutics, heterogeneous states of the drug/target complex can exist in the presence of soluble targets which can complicate measurement of unbound drug. In the case of bispecific antibodies, measurement of drug can be even more complicated and depend upon the levels of both targets to each arm. Measuring the total drug allows for PKPD modeling prediction of human dose projections in addition to overcoming challenges associated with measuring free drug for bispecific antibodies. Here, we present a study in which a sandwich ELISA format was used to measure total anti-KLK5/KLK7 antibody concentrations. This assay utilized a non-blocking anti-idiotype (ID) antibody to one arm of the antibody for capture and an antibody to target bound to the other arm of the antibody for detection. Our qualified assay showed acceptable precision, accuracy, dilutional linearity, and reproducibility and enabled detection of a total bispecific antibody at high levels of two targets. To confirm that our assay was detecting total drug, a subset of samples was evaluated in a generic total LC-MS/MS assay.
Collapse
Affiliation(s)
- Jeongsup Shim
- BioAnalytical Sciences, Development Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080, USA.
| | - Jessica Chen
- BioAnalytical Sciences, Development Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Montserrat Carrasco-Triguero
- BioAnalytical Sciences, Development Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Saloumeh K Fischer
- BioAnalytical Sciences, Development Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| |
Collapse
|
6
|
Liu D, Hu L, Shao H. Therapeutic drug monitoring of immune checkpoint inhibitors: based on their pharmacokinetic properties and biomarkers. Cancer Chemother Pharmacol 2023:10.1007/s00280-023-04541-8. [PMID: 37410155 DOI: 10.1007/s00280-023-04541-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/03/2023] [Indexed: 07/07/2023]
Abstract
As a new means of oncology treatment, immune checkpoint inhibitors (ICIs) can improve survival rates in patients with resistant or refractory tumors. However, there are obvious inter-individual differences in the unsatisfactory response rate, drug resistance rate and the occurrence of immune-related adverse events (irAE). These questions have sparked interest in researchers looking for a way to screen sensitive populations and predict efficacy and safety. Therapeutic drug monitoring (TDM) is a way to ensure the safety and effectiveness of medication by measuring the concentration of drugs in body fluids and adjusting the medication regimen. It has the potential to be an adjunctive means of predicting the safety and efficacy of ICIs treatment. In this review, the author outlined the pharmacokinetic (PK) characteristics of ICIs in patients. The feasibility and limitations of TDM of ICIs were discussed by summarizing the relationships between the pharmacokinetic parameters and the efficacy, toxicity and biomarkers.
Collapse
Affiliation(s)
- Dongxue Liu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Linlin Hu
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Office of Medication Clinical Institution, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hua Shao
- Office of Medication Clinical Institution, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
7
|
Direct bioanalysis or indirect calculation of target engagement and free drug exposure: do we apply double standards? Bioanalysis 2023; 15:5-16. [PMID: 36762451 DOI: 10.4155/bio-2022-0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
Analysis of "free" drug/target concentrations is important to set up appropriate pharmacokinetic-pharmacodynamic models, to evaluate active-drug exposure and target engagement. Such "free-analyte" determination could be done by direct bioanalysis using an appropriate "free-analyte" assay. Development of "free" assays is often considered challenging from a technological and regulatory perspective. The application of a "total-total" approach, where the "free-analyte" concentration is determined mathematically, is considered a more convenient option. In this perspective, we examine and discuss the challenges of this "total-total" approach, from the affinity data, the importance of applying an appropriate "total" assay, the impact of additional binding partners and the variability of the total drug/target assays and their impact on the quality and variability of the final "free-analyte" dataset.
Collapse
|
8
|
Fairman D, Tang H. Best Practices in mAb and Soluble Target Assay Selection for Quantitative Modelling and Qualitative Interpretation. AAPS J 2023; 25:23. [PMID: 36759378 DOI: 10.1208/s12248-023-00788-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/22/2023] [Indexed: 02/11/2023] Open
Abstract
Biologics, especially monoclonal antibodies (mAbs), are an increasingly important part of the drug discovery and development portfolio across the pharmaceutical industry. To enable robust demonstration of pillars 1 and 2 [1] for mAbs, specialised assays are required to measure the complex interactions between mAb and target. This is especially important for the interpretation of soluble target interactions. In some instances, multiple assays with overlapping purposes (e.g., developing both complex and total assays) have been developed. In retrospect, these efforts may have led to excessive time and resources spent in assay development and the generation of data that is contradictory or misleading. Our recommendation is to invest resources early into the development of total assays for both mAb and target. Free target assay data may be inaccurate and report higher levels of free target than are present in the sample at collection due to re-equilibrium during measurement. Total assay formats are inherently less sensitive to the effects of sample preparation, assay conditions, and re-equilibration than free or complex assays. It is acknowledged that pathology/pharmacology is ultimately driven by the free target and knowledge of its dynamics are critical. However, generation of appropriate total target data and using model-based estimation of free target concentrations is a more robust approach than utilisation of direct assay derived estimates. Where free data are utilised, the potential biases should be prospectively considered when developing the assay and utilising the data for quantitative analyses.
Collapse
Affiliation(s)
- David Fairman
- Clinical Pharmacology Modelling and Simulation, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Huaping Tang
- Bioanalysis Immunogenicity and Biomarkers, GSK Research, 1250 South Collegeville Road, Pennsylvania, 19426, Collegeville, USA.
| |
Collapse
|
9
|
Millet A, Pescarmona R, Belot A, Machon C, Jamilloux Y, Guitton J. Quantification of canakinumab in human plasma by liquid chromatography-high resolution mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1211:123475. [PMID: 36179539 DOI: 10.1016/j.jchromb.2022.123475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/09/2022] [Accepted: 09/17/2022] [Indexed: 11/27/2022]
Abstract
Canakinumab is a fully-human monoclonal immunoglobulin gamma 1 kappa. This interleukin-1β blocker is used for the treatment of autoinflammatory diseases. Various studies have demonstrated the value of therapeutic drug monitoring of monoclonal antibodies in the management of inflammatory diseases. The purpose of this study was to develop a method to quantify canakinumab plasmatic concentration using liquid chromatography-high-resolution (Orbitrap®) mass spectrometry. The quantification was based on a bottom-up approach with the analysis of one surrogate peptide after an immunopurification of IgG followed by tryptic proteolysis. Rituximab and cetuximab, both IgG1, were tested as internal standards. Chromatographic separation was performed on a bioZenTM Peptide PS-C18 column. Mass detection was conducted in positive ionization mode with Parallel Reaction Monitoring at a resolution of 70,000. The method was fully validated in terms of linearity, sensitivity, selectivity, accuracy and matrix effect. Standards ranged from 2.5 to 75 µg/mL. Intra- and inter-day coefficients of variation ranged from 3.7 to 14.7 %, and accuracy from 97.4 to 104.1 %. This method allowed the determination of canakinumab plasmatic concentrations from eight treated patients. This method is efficient and suitable for routine use in therapeutic drug monitoring or pharmacokinetic studies.
Collapse
Affiliation(s)
- Aurélien Millet
- Biochemistry and Pharmacology-Toxicology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, F-69495 Pierre Bénite, France
| | - Rémi Pescarmona
- Immunology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, F-69495 Pierre Bénite, France
| | - Alexandre Belot
- Pediatric Nephrology, Rheumatology, Dermatology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France; Lyon Immunopathology FEderation (LIFE), Lyon, France
| | - Christelle Machon
- Biochemistry and Pharmacology-Toxicology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, F-69495 Pierre Bénite, France; Analytical Chemistry Laboratory, Faculty of Pharmacy ISPBL, University Lyon 1, F-69373 Lyon, France
| | - Yvan Jamilloux
- Lyon Immunopathology FEderation (LIFE), Lyon, France; Internal Medicine, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Guitton
- Biochemistry and Pharmacology-Toxicology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, F-69495 Pierre Bénite, France; Toxicology Laboratory, Faculty of Pharmacy ISPBL, University of Lyon 1, F-69373 Lyon, France.
| |
Collapse
|
10
|
Liu S, Shah DK. Mathematical Models to Characterize the Absorption, Distribution, Metabolism, and Excretion of Protein Therapeutics. Drug Metab Dispos 2022; 50:867-878. [PMID: 35197311 PMCID: PMC11022906 DOI: 10.1124/dmd.121.000460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/31/2022] [Indexed: 11/22/2022] Open
Abstract
Therapeutic proteins (TPs) have ranked among the most important and fastest-growing classes of drugs in the clinic, yet the development of successful TPs is often limited by unsatisfactory efficacy. Understanding pharmacokinetic (PK) characteristics of TPs is key to achieving sufficient and prolonged exposure at the site of action, which is a prerequisite for eliciting desired pharmacological effects. PK modeling represents a powerful tool to investigate factors governing in vivo disposition of TPs. In this mini-review, we discuss many state-of-the-art models that recapitulate critical processes in each of the absorption, distribution, metabolism/catabolism, and excretion pathways of TPs, which can be integrated into the physiologically-based pharmacokinetic framework. Additionally, we provide our perspectives on current opportunities and challenges for evolving the PK models to accelerate the discovery and development of safe and efficacious TPs. SIGNIFICANCE STATEMENT: This minireview provides an overview of mechanistic pharmacokinetic (PK) models developed to characterize absorption, distribution, metabolism, and elimination (ADME) properties of therapeutic proteins (TPs), which can support model-informed discovery and development of TPs. As the next-generation of TPs with diverse physicochemical properties and mechanism-of-action are being developed rapidly, there is an urgent need to better understand the determinants for the ADME of TPs and evolve existing platform PK models to facilitate successful bench-to-bedside translation of these promising drug molecules.
Collapse
Affiliation(s)
- Shufang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
11
|
Assessment of immunogenicity and drug activity in patient sera by flow-induced dispersion analysis. Sci Rep 2022; 12:4670. [PMID: 35304547 PMCID: PMC8933425 DOI: 10.1038/s41598-022-08682-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/09/2022] [Indexed: 01/20/2023] Open
Abstract
Biopharmaceuticals have revolutionized the treatment of many diseases such as diabetes, cancer, and autoimmune disorders. These complex entities provide unique advantages like high specificity towards their target. Unfortunately, biopharmaceuticals are also prone to elicit undesired immunogenic responses (immunogenicity), compromising treatment efficacy as well as patient safety due to severe adverse effects including life threatening conditions. Current immunogenicity assays are hampered by immobilization procedures, complicated sample pre-treatment, or rely on cell-based methods which all prevent reliable and continuous monitoring of patients. In this work, we present Flow Induced Dispersion Analysis (FIDA) for assessment of immunogenicity and drug activity in serum samples from arthritis patients receiving adalimumab. FIDA is a first principle technique for size-based characterization of biomolecules and their complexes under biologically relevant conditions. The FIDA methodology rely on an absolute and quantitative readout (hydrodynamic radius) thus reducing the need for positive and negative controls. Here, FIDA is applied for evaluating active adalimumab in serum by studying the interaction with its target tumor necrosis factor alpha (TNF-α). We report proof of principle for a quantitative approach for stratifying patients exhibiting presence of neutralizing and non-neutralizing antibodies based on their individual drug activity pattern. Further, it can be applied to any biopharmaceutical having soluble drug targets and it holds potential in a companion diagnostics setting.
Collapse
|
12
|
Furihata K, Hamada I, Niwa T, Watanabe T, Ezoe S. Pharmacokinetics, Safety, and Tolerability of NPC-21, an Anti-Cytomegalovirus Monoclonal Antibody, in Healthy Japanese and White Adult Men: A Randomized, Placebo-Controlled, First-in-Human Phase 1 Study. Clin Pharmacol Drug Dev 2022; 11:707-716. [PMID: 34989174 PMCID: PMC9303920 DOI: 10.1002/cpdd.1065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/07/2021] [Indexed: 11/09/2022]
Abstract
NPC-21 (EV2038) is a fully human monoclonal antibody that targets the antigenic domain 1 of glycoprotein B on the human cytomegalovirus (hCMV) envelope. NPC-21 has been shown to have broadly neutralizing activity and to inhibit cell-to-cell transmission of hCMV in preclinical studies. It is currently in development for the prophylactic or preemptive treatment of hCMV in patients receiving a solid-organ transplant or hematopoietic stem cell transplant. A first-in-human phase 1 study was conducted to assess the pharmacokinetics, safety, and tolerability of NPC-21 in healthy adult men. Forty participants (Japanese, n = 32; White, n = 8) were randomly assigned to receive a single intravenous dose of NPC-21 1, 3, 10, or 20 mg/kg or placebo. Six Japanese participants were included in each dose group and six White participants received a 10-mg/kg dose. The placebo group included 8 Japanese participants and 2 White participants. All 40 participants completed the study. Serum concentration, maximum serum concentration, area under the plasma concentration-time curve from time 0 to the last measurable concentration, and area under the plasma concentration-time curve from time 0 to infinity increased dose dependently; dose proportionality was linear. NPC-21 demonstrated a biphasic elimination pattern, with an estimated half-life between 612 and 790 hours. NPC-21 was safe and well tolerated up to 20 mg/kg. All adverse events were mild, and none led to treatment discontinuation or were considered related to the study drug. There were no differences in pharmacokinetics or safety between Japanese and White participants. These results support further investigation of NPC-21.
Collapse
Affiliation(s)
| | - Izumi Hamada
- Research & Development Division, Nobelpharma Co. Ltd., Tokyo, Japan
| | - Takuro Niwa
- Research & Development Division, Nobelpharma Co. Ltd., Tokyo, Japan
| | - Tatsuya Watanabe
- Research & Development Division, Nobelpharma Co. Ltd., Tokyo, Japan
| | - Sachiko Ezoe
- Department of Space Infection Control, Graduate School of Medicine, Division of Health Science, Osaka University, Osaka, Japan.,Medical Center for Translational Research, Osaka University Hospital, Osaka, Japan
| |
Collapse
|
13
|
Peña-Cabia S, Royuela Vicente A, Ramos Díaz R, Gutiérrez Nicolás F, Peñalver Vera Á, Siso García I, Hitt Sabag R, García Lacalle C, Peña-Cabia A, Iglesias-Peinado I, García Díaz B, López-Martín A. Assessment of exposure-response relationship for bevacizumab in patients with metastatic colorectal cancer. Biomed Pharmacother 2021; 141:111827. [PMID: 34153845 DOI: 10.1016/j.biopha.2021.111827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 12/27/2022] Open
Abstract
Limited literature is available for bevacizumab exposure-response relationship and there is not a concentration threshold associated with an optimal disease control. This prospective observational study in patients with metastatic colorectal cancer (mCRC) aims to evaluate, in a real-life setting, the relationship between bevacizumab through concentrations at steady state (Ctrough, SS) and disease control. Ctrough, SS were drawn, coinciding with the radiological evaluation of the response (progression or clinical benefit). Generalized estimating equations (GEE) analysis was performed. To test the association between Ctrough, SS in each patient with overall survival (OS) or progression-free survival (PFS), Cox proportional hazard models were developed. Data included 50 bevacizumab Ctrough, SS from 27 patients. The GEE model did not suggest any positive association between bevacizumab Ctrough, SS and clinical benefit (OR 0.99, 95% CI: 0.98-1.02, p = 0.863). The Cox regression showed association between higher median Ctrough, SS with better OS (HR 0.86, 95% CI: 0.73-1.01, p = 0.060), but not with PFS. We cannot confirm a relationship between bevacizumab Ctrough, SS and clinical benefit but this is the first real-world study trying to show a relationship between bevacizumab Ctrough, SS and disease control in mCRC. It was conducted in a small sample size which reduces the level of evidence. Further controlled randomized studies with a sufficient number of patients are required.
Collapse
Affiliation(s)
| | - Ana Royuela Vicente
- Biostatistics Unit, Puerta de Hierro Biomedical Research Institute (IDIPHISA), CIBERESP, Madrid, Spain
| | - Ruth Ramos Díaz
- Foundation Health Research Institute of Canary (FIISC), University Hospital Complex of Canary (CHUC), Tenerife, Spain
| | | | | | | | | | | | - Ana Peña-Cabia
- Medical Laboratory Unit, Virgen de la Luz Hospital, Cuenca, Spain
| | - Irene Iglesias-Peinado
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | | | - Ana López-Martín
- Medical Oncology Unit, Severo Ochoa University Hospital, Madrid, Spain
| |
Collapse
|
14
|
Walters RR, Boucher JF, De Toni F. Pharmacokinetics and Immunogenicity of Frunevetmab in Osteoarthritic Cats Following Intravenous and Subcutaneous Administration. Front Vet Sci 2021; 8:687448. [PMID: 34179175 PMCID: PMC8222533 DOI: 10.3389/fvets.2021.687448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/27/2021] [Indexed: 11/21/2022] Open
Abstract
Osteoarthritis and other degenerative joint diseases are common causes of chronic pain in cats. Frunevetmab is a felinized monoclonal antibody that binds to nerve growth factor (NGF) and provides relief from pain by blocking the receptor-mediated signaling cascade induced by NGF. Results from three studies were combined to provide an overview of frunevetmab pharmacokinetics (PK) and immunogenicity. The objective of the first study was to establish the pharmacokinetic parameters resulting from intravenous (IV) and subcutaneous (SC) administration of frunevetmab to the feline patient population at 3 mg/kg. Ten adult cats with naturally-occurring osteoarthritis were administered frunevetmab in a crossover design at 28 day intervals. Non-compartmental pharmacokinetic analysis of the plasma concentration-time data showed that the half-life was 10.1 ± 1.9 days after IV dosing and the SC bioavailability was 60.3 ± 15.8% with maximum drug levels observed at 3-7 days after dosing. Plasma samples were collected at ~28 days after dosing during two field safety and effectiveness studies of cats with degenerative joint disease. The doses ranged from 1.0 to 2.8 mg/kg; 2 or 3 doses were administered either SC/IV, SC/SC, or SC/SC/SC. The data from these studies along with the data from the laboratory pharmacokinetic study were analyzed using non-linear mixed-effects (NLME) modeling. The model closely predicted the trough concentrations from the two field studies, including the IV treatment in the pilot field study. The trough concentrations were predicted to be close to steady-state after 2 doses. A second objective was to determine the incidence and clinical relevance of frunevetmab immunogenicity. A three-tier anti-drug antibody assay (screen, confirm, titer) was developed and validated. Immunogenicity was assessed in 259 frunevetmab-treated animals enrolled in the two field studies. Only 4 of these animals (1.5%) appeared to develop immunogenicity to frunevetmab. None of the four exhibited adverse events attributed to immunogenicity and no impact on drug levels or efficacy was observed in three of the animals. In the placebo animals, 2.3% (3/131) appeared to develop treatment-emergent immunogenicity. Overall, frunevetmab administration resulted in a very low incidence of treatment-emergent immunogenicity with no safety findings and minimal effect on drug exposure and efficacy.
Collapse
Affiliation(s)
| | | | - Flavia De Toni
- Veterinary Medicine Research and Development, Zoetis, Kalamazoo, MI, United States
| |
Collapse
|
15
|
Increasing robustness, reliability and storage stability of critical reagents by freeze-drying. Bioanalysis 2021; 13:829-840. [PMID: 33890493 DOI: 10.4155/bio-2020-0299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim: Stabilization of critical reagents by freeze-drying would facilitate storage and transportation at ambient temperatures, and simultaneously enable constant reagent performance for long-term bioanalytical support throughout drug development. Freeze-drying as a generic process for stable performance and storage of critical reagents was investigated by establishing an universal formulation buffer and lyophilization process. Results: Using a storage-labile model protein, formulation buffers were evaluated to preserve reagent integrity during the freeze-drying process, and to retain functional performance after temperature stress. Application to critical reagents used in pharmacokinetics and anti-drug antibodies assays demonstrated stable functional performance of the reagents after 11 month at +40°C. Conclusion: Stabilization and storage of critical assay reagents by freeze-drying is an attractive alternative to traditional deep freezing.
Collapse
|
16
|
Harth S, Frisch C. Recombinant Anti-idiotypic Antibodies in Ligand Binding Assays for Antibody Drug Development. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2261:291-306. [PMID: 33420997 DOI: 10.1007/978-1-0716-1186-9_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Sensitive and reproducible pharmacokinetic (PK) assays and immunogenicity assessment are required as part of the complex and lengthy development process for biotherapeutic proteins. Ligand binding assays (LBAs) are included in a range of approaches applied to understand the nature and properties of the drug as well as the induction of anti-drug antibodies (ADA) against the therapeutic, which can cause adverse events and loss of efficacy. Currently, most biotherapeutics are monoclonal human or humanized antibodies. Anti-idiotypic antibodies, targeting the idiotopic determinants of individual antibody drugs are recognized as perfect reagents for such LBAs. Here we describe the typical setups for these assays and how different types of anti-biotherapeutic antibodies can be used to establish selective and sensitive assays.
Collapse
|
17
|
Klaus T, Deshmukh S. pH-responsive antibodies for therapeutic applications. J Biomed Sci 2021; 28:11. [PMID: 33482842 PMCID: PMC7821552 DOI: 10.1186/s12929-021-00709-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/15/2021] [Indexed: 11/29/2022] Open
Abstract
Therapeutic antibodies are instrumental in improving the treatment outcome for certain disease conditions. However, to enhance their efficacy and specificity, many efforts are continuously made. One of the approaches that are increasingly explored in this field are pH-responsive antibodies capable of binding target antigens in a pH-dependent manner. We reviewed suitability and examples of these antibodies that are functionally modulated by the tumor microenvironment. Provided in this review is an update about antigens targeted by pH-responsive, sweeping, and recycling antibodies. Applicability of the pH-responsive antibodies in the engineering of chimeric antigen receptor T-cells (CAR-T) and in improving drug delivery to the brain by the enhanced crossing of the blood-brain barrier is also discussed. The pH-responsive antibodies possess strong treatment potential. They emerge as next-generation programmable engineered biologic drugs that are active only within the targeted biological space. Thus, they are valuable in targeting acidified tumor microenvironment because of improved spatial persistence and reduced on-target off-tumor toxicities. We predict that the programmable pH-dependent antibodies become powerful tools in therapies of cancer.
Collapse
Affiliation(s)
- Tomasz Klaus
- Research and Development Department, Pure Biologics, Inc., Dunska 11, 54427, Wrocław, Poland
| | - Sameer Deshmukh
- Research and Development Department, Pure Biologics, Inc., Dunska 11, 54427, Wrocław, Poland.
| |
Collapse
|
18
|
Watanabe H, Shibuya M, Shibahara N, Ruike Y, Sampei Z, Haraya K, Tachibana T, Wakabayashi T, Sakamoto A, Tsunoda H, Murao N. A Novel Total Drug Assay for Quantification of Anti-C5 Therapeutic Monoclonal Antibody in the Presence of Abundant Target. AAPS JOURNAL 2021; 23:21. [PMID: 33415498 DOI: 10.1208/s12248-020-00539-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/24/2020] [Indexed: 11/30/2022]
Abstract
SKY59 or RO7112689 is a humanized monoclonal antibody against complement protein C5 with pH-dependent C5-binding and neonatal Fc receptor-mediated recycling capabilities, which result in long-lasting neutralization of C5. We developed and validated a novel total drug assay for quantification of target-binding competent SKY59 in the presence of endogenous C5 in cynomolgus monkey plasma. The target-binding competent SKY59 was determined after complex formation by the addition of recombinant monkey C5 using goat anti-human IgG-heavy chain monkey-adsorbed polyclonal antibody as a capture antibody and rabbit anti-C5 monoclonal antibody (mAb) non-competing with SKY59 for detection. The total SKY59 assay was shown to be accurate and precise over the range of 0.05-3.2 μg/mL as well as be tolerant to more than 400 μg/mL of C5 (~ 3000-fold molar excess of target). We also developed and validated a total C5 assay, confirmed selectivity and parallelism, and verified the utility of recombinant monkey C5 for the total C5 assay as well as the total SKY59 assay. Furthermore, we used these validated methods to measure SKY59 and C5 concentrations in cynomolgus monkey plasma samples in a toxicology study. This total drug assay can be applied not only to other antibody therapeutics against shed/soluble targets when a non-competing reagent mAb is available but also for clinical studies when a reagent mAb specific for engineered Fc region on a therapeutic mAb is available.
Collapse
Affiliation(s)
- Hiroo Watanabe
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan.
| | - Mitsuko Shibuya
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| | - Norihito Shibahara
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| | - Yoshinao Ruike
- Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Zenjiro Sampei
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| | - Kenta Haraya
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| | - Tatsuhiko Tachibana
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| | - Tetsuya Wakabayashi
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| | - Akihisa Sakamoto
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| | - Hiroyuki Tsunoda
- Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Naoaki Murao
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| |
Collapse
|
19
|
Mocenigo M, Porchetta A, Rossetti M, Brass E, Tonini L, Puzzi L, Tagliabue E, Triulzi T, Marini B, Ricci F, Ippodrino R. Rapid, Cost-Effective Peptide/Nucleic Acid-Based Platform for Therapeutic Antibody Monitoring in Clinical Samples. ACS Sens 2020; 5:3109-3115. [PMID: 32909731 DOI: 10.1021/acssensors.0c01046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We demonstrate here a homogeneous assay, named NanoHybrid, for monoclonal antibody quantification directly in serum samples in a single-step format. NanoHybrid is composed of both synthetic peptide nucleic acids (PNAs) and nucleic acid strands conjugated to recognition elements and optical labels and is designed to allow fast fluorescence quantification of a therapeutic antibody. More specifically, we have characterized our analytical assay for the detection of trastuzumab (Herceptin), a monoclonal antibody (mAb) drug used for breast cancer treatment and for tumors overexpressing the HER2/neu protein. We show here that NanoHybrid is capable of performing fast drug quantification directly in blood serum. The results obtained with a pool of samples from breast cancer patients under trastuzumab treatment are compared with CE-IVD ELISA (enzyme-linked immunosorbent assay) showing a good agreement (Cohen's K = 0.729). Due to the modular nature of the NanoHybrid platform, this technology can be programmed to potentially detect and quantify any antibody for which a high-affinity recognition element has been characterized. We envision the application of NanoHybrid in a point-of-care (POC) drug monitoring system based on disposable kits for therapeutic drug management.
Collapse
Affiliation(s)
- Marco Mocenigo
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
- Molecular Genetics and Biotechnology PhD Study Programme, University of Nova Gorica, Vipavska 13, 5000 Nova Gorica, Slovenia
| | - Alessandro Porchetta
- Department of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Marianna Rossetti
- Department of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Erik Brass
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Lucia Tonini
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Luca Puzzi
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Elda Tagliabue
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Tiziana Triulzi
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Bruna Marini
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Francesco Ricci
- Department of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Rudy Ippodrino
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| |
Collapse
|
20
|
Kielbasa W, Helton DL. A new era for migraine: Pharmacokinetic and pharmacodynamic insights into monoclonal antibodies with a focus on galcanezumab, an anti-CGRP antibody. Cephalalgia 2019; 39:1284-1297. [PMID: 30917684 PMCID: PMC6710614 DOI: 10.1177/0333102419840780] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/17/2018] [Accepted: 03/04/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE To review pharmacokinetic and pharmacodynamic characteristics of antibodies that bind to soluble ligands within the framework of calcitonin gene-related peptide antibodies. OVERVIEW Calcitonin gene-related peptide has been implicated in the pathophysiology of migraine. Galcanezumab is an antibody that binds to the ligand calcitonin gene-related peptide. Other antibodies that target calcitonin gene-related peptide include eptinezumab and fremanezumab. To understand how antibodies can affect the extent and duration of free ligand concentrations, it is important to consider the dose and pharmacokinetics of an antibody, and the kinetics of the ligand and antibody-ligand complex. Insights regarding the pharmacokinetic/pharmacodynamic properties of galcanezumab as a probe antibody drug and calcitonin gene-related peptide as its binding ligand regarding its clinical outcomes are provided. DISCUSSION Antibodies are administered parenterally because oral absorption is limited by gastrointestinal degradation and inefficient diffusion through the epithelium. The systemic absorption of antibodies following intramuscular or subcutaneous administration most likely occurs via convective transport through lymphatic vessels into blood. The majority of antibody elimination occurs via intracellular catabolism into peptides and amino acids following endocytosis. Binding of ligand to an antibody reduces the free ligand that is available to interact with the receptor and efficacy is driven by the magnitude and duration of the reduction in free ligand concentration. A galcanezumab pharmacokinetic/pharmacodynamic model shows that galcanezumab decreases free calcitonin gene-related peptide concentrations in a dose- and time-dependent manner and continues to suppress free calcitonin gene-related peptide with repeated dosing. The model provides evidence for a mechanistic linkage to galcanezumab therapeutic effects for the preventive treatment of migraine.
Collapse
|
21
|
Bogen JP, Hinz SC, Grzeschik J, Ebenig A, Krah S, Zielonka S, Kolmar H. Dual Function pH Responsive Bispecific Antibodies for Tumor Targeting and Antigen Depletion in Plasma. Front Immunol 2019; 10:1892. [PMID: 31447859 PMCID: PMC6697062 DOI: 10.3389/fimmu.2019.01892] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 07/26/2019] [Indexed: 01/08/2023] Open
Abstract
Shedding of membrane-bound cell surface proteins, where the extracellular domain is released and found in the circulation is a common phenomenon. A prominent example is CEACAM5 (CEA, CD66e), where the shed domain plays a pivotal role in tumor progression and metastasis. For treatment of solid tumors, the presence of the tumor-specific antigen in the plasma can be problematic since tumor-specific antibodies might be intercepted by the soluble antigen before invading their desired tumor target area. To overcome this problem, we developed a generic procedure to generate bispecific antibodies, where one arm binds the antigen in a pH-dependent manner thereby enhancing antigen clearance upon endosomal uptake, while the other arm is able to target tumor cells pH-independently. This was achieved by incorporating pH-sensitive binding modalities in the common light chain IGKV3-15*01 of a CEACAM5 binding heavy chain only antibody. Screening of a histidine-doped light chain library using yeast surface display enabled the isolation of pH-dependent binders. When such a light chain was utilized as a common light chain in a bispecific antibody format, only the respective heavy/light chain combination, identified during selections, displayed pH-responsive binding. In addition, we found that the altered common light chain does not negatively impact the affinity of other heavy chain only binders toward their respective antigen. Our strategy may open new avenues for the generation of bispecifics, where one arm efficiently removes a shed antigen from the circulation while the other arm targets a tumor marker in a pH-independent manner.
Collapse
Affiliation(s)
- Jan P Bogen
- Department of Applied Biochemistry, Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Steffen C Hinz
- Department of Applied Biochemistry, Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Julius Grzeschik
- Department of Applied Biochemistry, Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Aileen Ebenig
- Department of Applied Biochemistry, Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Simon Krah
- Protein Engineering and Antibody Technologies, Merck KGaA, Darmstadt, Germany
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Merck KGaA, Darmstadt, Germany
| | - Harald Kolmar
- Department of Applied Biochemistry, Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| |
Collapse
|
22
|
Winters A, McFadden K, Bergen J, Landas J, Berry KA, Gonzalez A, Salimi-Moosavi H, Murawsky CM, Tagari P, King CT. Rapid single B cell antibody discovery using nanopens and structured light. MAbs 2019; 11:1025-1035. [PMID: 31185801 PMCID: PMC6748590 DOI: 10.1080/19420862.2019.1624126] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Accelerated development of monoclonal antibody (mAb) tool reagents is an essential requirement for the successful advancement of therapeutic antibodies in today’s fast-paced and competitive drug development marketplace. Here, we describe a direct, flexible, and rapid nanofluidic optoelectronic single B lymphocyte antibody screening technique (NanOBlast) applied to the generation of anti-idiotypic reagent antibodies. Selectively enriched, antigen-experienced murine antibody secreting cells (ASCs) were harvested from spleen and lymph nodes. Subsequently, secreted mAbs from individually isolated, single ASCs were screened directly using a novel, integrated, high-content culture, and assay platform capable of manipulating living cells within microfluidic chip nanopens using structured light. Single-cell polymerase chain reaction–based molecular recovery on select anti-idiotypic ASCs followed by recombinant IgG expression and enzyme-linked immunosorbent assay (ELISA) characterization resulted in the recovery and identification of a diverse and high-affinity panel of anti-idiotypic reagent mAbs. Combinatorial ELISA screening identified both capture and detection mAbs, and enabled the development of a sensitive and highly specific ligand binding assay capable of quantifying free therapeutic IgG molecules directly from human patient serum, thereby facilitating important drug development decision-making. The ASC import, screening, and export discovery workflow on the chip was completed within 5 h, while the overall discovery workflow from immunization to recombinantly expressed IgG was completed in under 60 days.
Collapse
Affiliation(s)
- Aaron Winters
- a Department of Therapeutic Discovery, Amgen Research , Thousand Oaks , CA , USA
| | - Karyn McFadden
- a Department of Therapeutic Discovery, Amgen Research , Thousand Oaks , CA , USA
| | - John Bergen
- b Department of Therapeutic Discovery, Amgen Research , Burnaby , Canada
| | - Julius Landas
- b Department of Therapeutic Discovery, Amgen Research , Burnaby , Canada.,c Department of Pharmacokinetics & Drug Metabolism, University of British Columbia , Vancouver , Canada
| | - Kelly A Berry
- b Department of Therapeutic Discovery, Amgen Research , Burnaby , Canada
| | - Anthony Gonzalez
- a Department of Therapeutic Discovery, Amgen Research , Thousand Oaks , CA , USA
| | - Hossein Salimi-Moosavi
- a Department of Therapeutic Discovery, Amgen Research , Thousand Oaks , CA , USA.,c Department of Pharmacokinetics & Drug Metabolism, University of British Columbia , Vancouver , Canada
| | | | - Philip Tagari
- a Department of Therapeutic Discovery, Amgen Research , Thousand Oaks , CA , USA
| | - Chadwick T King
- b Department of Therapeutic Discovery, Amgen Research , Burnaby , Canada
| |
Collapse
|
23
|
Ren C, Bobst CE, Kaltashov IA. Exploiting His-Tags for Absolute Quantitation of Exogenous Recombinant Proteins in Biological Matrices: Ruthenium as a Protein Tracer. Anal Chem 2019; 91:7189-7198. [PMID: 31083917 DOI: 10.1021/acs.analchem.9b00504] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Metal labeling and ICP MS detection offer an alternative to commonly accepted techniques that are currently used to quantitate exogenous proteins in vivo, but modifying the protein surface with metal-containing groups inevitably changes its biophysical properties and is likely to affect trafficking and biodistribution. The approach explored in this work takes advantage of the presence of hexa-histidine tags in many recombinant proteins, which have high affinity toward a range of metals. While many divalent metals bind to poly histidine sequences reversibly, oxidation of imidazole-bound CoII or RuII is known to result in a dramatic increase of the binding strength. In order to evaluate the feasibility of using imidazole-bound metal oxidation as a means of attaching permanent tags to polyhistidine segments, a synthetic peptide YPDFEDYWMKHHHHHH was used as a model. RuII can be oxidized under ambient (aerobic) conditions, allowing any oxidation damage to the peptide beyond the metal-binding site to be avoided. The resulting peptide-RuIII complex is very stable, with the single hexa-histidine segment capable of accommodating up to three metal ions. Localization of RuIII within the hexa-histidine segment of the peptide was confirmed by tandem mass spectrometry. The RuIII/peptide binding appears to be irreversible, with both low- and high-molecular weight biologically relevant scavengers failing to strip the metal from the peptide. Application of this protocol to labeling a recombinant form of an 80 kDa protein transferrin allowed RuIII to be selectively placed within the His-tag segment. The metal label remained stable in the presence of ubiquitous scavengers and did not interfere with the receptor binding, while allowing the protein to be readily detected in serum at sub-nM concentrations. The results of this work suggest that ruthenium lends itself as an ideal metal tag for selective labeling of His-tag containing recombinant proteins to enable their sensitive detection and quantitation with ICP MS.
Collapse
Affiliation(s)
- Chengfeng Ren
- Department of Chemistry , University of Massachusetts-Amherst , Amherst , Massachusetts 01003 , United States
| | - Cedric E Bobst
- Department of Chemistry , University of Massachusetts-Amherst , Amherst , Massachusetts 01003 , United States
| | - Igor A Kaltashov
- Department of Chemistry , University of Massachusetts-Amherst , Amherst , Massachusetts 01003 , United States
| |
Collapse
|
24
|
Development and validation of indirect and generic immunoassays to quantify free and total evolocumab in rat serum. Bioanalysis 2019; 11:679-687. [DOI: 10.4155/bio-2019-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Evolocumab is a human monoclonal antibody used in the treatment of cardiovascular diseases, which targeted proprotein convertase subtilisin kexin type 9. To accurately quantify free (including partially bound) and total evolocumab concentrations in serum, indirect and generic ELISA methods were developed and validated in rat serum. Results: Indirect ELISA was accurate and precise over the concentration range of 23.4–1500 ng/ml, and the method was validated for selectivity, specificity, accuracy and precision, dilution linearity, parallelism and stability. Similarly, generic antihuman IgG ELISA method was validated for selectivity, accuracy and precision, and dilution linearity. Moreover, incurred sample reanalysis were carried out for the above two methods, and the percent difference met the acceptable criteria. Conclusion: The validated methods can be effectively used to evaluate pharmacokinetics of free and total evolocumab after subcutaneous administration of evolocumab to rats.
Collapse
|
25
|
Generation by phage display and characterization of drug-target complex-specific antibodies for pharmacokinetic analysis of biotherapeutics. MAbs 2018; 11:178-190. [PMID: 30516449 PMCID: PMC6343800 DOI: 10.1080/19420862.2018.1538723] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Anti-idiotypic antibodies play an important role in pre-clinical and clinical development of therapeutic antibodies, where they are used for pharmacokinetic studies and for the development of immunogenicity assays. By using an antibody phage display library in combination with guided in vitro selection against various marketed drugs, we generated antibodies that recognize the drug only when bound to its target. We have named such specificities Type 3, to distinguish them from the anti-idiotypic antibodies that specifically detect free antibody drug or total drug. We describe the generation and characterization of such reagents for the development of ligand binding assays for drug quantification. We also show how these Type 3 antibodies can be used to develop very specific and sensitive assays that avoid the bridging format. Abbreviations: BAP: bacterial alkaline phosphatase; CDR: complementarity-determining regions in VH or VL; Fab: antigen-binding fragment of an antibody; HRP: horseradish peroxidase; HuCAL®: Human Combinatorial Antibody Libraries; IgG: immunoglobulin G; LBA: ligand binding assay; LOQ: limit of quantitation; NHS: normal human serum; PK: pharmacokinetics; VH: variable region of the heavy chain of an antibody; VL: variable region of the light chain of an antibody.
Collapse
|
26
|
Critical considerations for immunocapture enrichment LC–MS bioanalysis of protein therapeutics and biomarkers. Bioanalysis 2018; 10:987-995. [DOI: 10.4155/bio-2018-0062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In recent years, immunocapture enrichment coupled with LC–MS technology has seen more applications for the measurement of low abundant protein therapeutics and biomarkers in biological matrices. In this article, several critical considerations for the application of immunocapture enrichment to LC–MS bioanalysis of protein therapeutics and biomarkers, including reagent selection, reagent characterization, designing of capture format, etc. are discussed. All these considerations are critical in developing reliable and robust bioanalytical assays with high assay specificity and sensitivity. Successful examples using the immunocapture LC–MS approach in the quantification of biotherapeutic and low abundant protein biomarkers will also be discussed.
Collapse
|
27
|
Cornec D, Kabat BF, Mills JR, Cheu M, Hummel AM, Schroeder DR, Cascino MD, Brunetta P, Murray DL, Snyder MR, Fervenza F, Hoffman GS, Kallenberg CGM, Langford CA, Merkel PA, Monach PA, Seo P, Spiera RF, St Clair EW, Stone JH, Barnidge DR, Specks U. Pharmacokinetics of rituximab and clinical outcomes in patients with anti-neutrophil cytoplasmic antibody associated vasculitis. Rheumatology (Oxford) 2018; 57:639-650. [PMID: 29340623 PMCID: PMC5888934 DOI: 10.1093/rheumatology/kex484] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 11/10/2017] [Indexed: 12/16/2022] Open
Abstract
Objectives To study the determinants of the pharmacokinetics (PK) of rituximab (RTX) in patients with ANCA-associated vasculitis (AAV) and its association with clinical outcomes. Methods This study included data from 89 patients from the RTX in AAV trial who received the full dose of RTX (four weekly infusions of 375 mg/m2). RTX was quantified at weeks 2, 4, 8, 16 and 24, and summarized by computing the trapezoidal area under the curve. We explored potential determinants of the PK-RTX, and analysed its association with clinical outcomes: achievement of remission at 6 months, duration of B-cell depletion and time to relapse in patients who achieved complete remission. Results RTX serum levels were significantly lower in males and in newly diagnosed patients, and negatively correlated with body surface area, baseline B-cell count and degree of disease activity. In multivariate analyses, the main determinants of PK-RTX were sex and new diagnosis. Patients reaching complete remission at month 6 had similar RTX levels compared with patients who did not reach complete remission. Patients with higher RTX levels generally experienced longer B-cell depletion than patients with lower levels, but RTX levels at the different time points and area under the curve were not associated with time to relapse. Conclusion Despite the body-surface-area-based dosing protocol, PK-RTX is highly variable among patients with AAV, its main determinants being sex and newly diagnosed disease. We did not observe any relevant association between PK-RTX and clinical outcomes. The monitoring of serum RTX levels does not seem clinically useful in AAV.
Collapse
Affiliation(s)
- Divi Cornec
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
- Rheumatology Department, Brest University Hospital, and INSERM U1227, Brest, France
| | - Brian F Kabat
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - John R Mills
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Amber M Hummel
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Melissa R Snyder
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Gary S Hoffman
- Division of Rheumatology, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | - Carol A Langford
- Division of Rheumatology, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Peter A Merkel
- Division of Rheumatology and Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA
| | - Paul A Monach
- Rheumatology, Boston University Medical Center, Boston, MA
| | - Philip Seo
- Rheumatology, Johns Hopkins University, Baltimore, MD
| | | | | | - John H Stone
- Rheumatology, Massachusetts General Hospital, Boston, MA, USA
| | - David R Barnidge
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
28
|
Legeron R, Xuereb F, Chaignepain S, Gadeau AP, Claverol S, Dupuy JW, Djabarouti S, Couffinhal T, Schmitter JM, Breilh D. A new reliable, transposable and cost-effective assay for absolute quantification of total plasmatic bevacizumab by LC–MS/MS in human plasma comparing two internal standard calibration approaches. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1070:43-53. [DOI: 10.1016/j.jchromb.2017.10.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 09/18/2017] [Accepted: 10/20/2017] [Indexed: 11/17/2022]
|
29
|
Immunoaffinity capture coupled with capillary electrophoresis - mass spectrometry to study therapeutic protein stability in vivo. Anal Biochem 2017; 539:118-126. [DOI: 10.1016/j.ab.2017.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 01/09/2023]
|
30
|
Chen X, Farrokhi V, Singh P, Ocana MF, Patel J, Lin LL, Neubert H, Brodfuehrer J. Biomeasures and mechanistic modeling highlight PK/PD risks for a monoclonal antibody targeting Fn14 in kidney disease. MAbs 2017; 10:62-70. [PMID: 29190188 DOI: 10.1080/19420862.2017.1398873] [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] [Indexed: 12/13/2022] Open
Abstract
Discovery of the upregulation of fibroblast growth factor-inducible-14 (Fn14) receptor following tissue injury has prompted investigation into biotherapeutic targeting of the Fn14 receptor for the treatment of conditions such as chronic kidney diseases. In the development of monoclonal antibody (mAb) therapeutics, there is an increasing trend to use biomeasures combined with mechanistic pharmacokinetic/pharmacodynamic (PK/PD) modeling to enable decision making in early discovery. With the aim of guiding preclinical efforts on designing an antibody with optimized properties, we developed a mechanistic site-of-action (SoA) PK/PD model for human application. This model incorporates experimental biomeasures, including concentration of soluble Fn14 (sFn14) in human plasma and membrane Fn14 (mFn14) in human kidney tissue, and turnover rate of human sFn14. Pulse-chase studies using stable isotope-labeled amino acids and mass spectrometry indicated the sFn14 half-life to be approximately 5 hours in healthy volunteers. The biomeasures (concentration, turnover) of sFn14 in plasma reveals a significant hurdle in designing an antibody against Fn14 with desired characteristics. The projected dose (>1 mg/kg/wk for 90% target coverage) derived from the human PK/PD model revealed potential high and frequent dosing requirements under certain conditions. The PK/PD model suggested a unique bell-shaped relationship between target coverage and antibody affinity for anti-Fn14 mAb, which could be applied to direct the antibody engineering towards an optimized affinity. This investigation highlighted potential applications, including assessment of PK/PD risks during early target validation, human dose prediction and drug candidate optimization.
Collapse
Affiliation(s)
- Xiaoying Chen
- a Department of Biomedicine Design , Pfizer Inc , Cambridge , MA , United States of America
| | - Vahid Farrokhi
- b Department of Biomedicine Design , Pfizer Inc , Andover , MA , United States of America
| | - Pratap Singh
- b Department of Biomedicine Design , Pfizer Inc , Andover , MA , United States of America
| | - Mireia Fernandez Ocana
- b Department of Biomedicine Design , Pfizer Inc , Andover , MA , United States of America
| | - Jenil Patel
- b Department of Biomedicine Design , Pfizer Inc , Andover , MA , United States of America
| | - Lih-Ling Lin
- c Inflammation and Immunology Research Unit , Pfizer Inc. , Cambridge , MA , United States of America
| | - Hendrik Neubert
- b Department of Biomedicine Design , Pfizer Inc , Andover , MA , United States of America
| | - Joanne Brodfuehrer
- a Department of Biomedicine Design , Pfizer Inc , Cambridge , MA , United States of America
| |
Collapse
|
31
|
Thoren KL, Pasi B, Delgado JC, Wu AH, Lynch KL. Quantitation of Infliximab and Detection of Antidrug Antibodies in Serum by Use of Surface Plasmon Resonance. ACTA ACUST UNITED AC 2017; 2:725-736. [DOI: 10.1373/jalm.2017.024380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 08/29/2017] [Indexed: 11/06/2022]
|
32
|
Evaluation of the potential use of hybrid LC–MS/MS for active drug quantification applying the ‘free analyte QC concept’. Bioanalysis 2017; 9:1705-1717. [DOI: 10.4155/bio-2017-0158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: Assessment of active drug exposure of biologics may be crucial for drug development. Typically, ligand-binding assay methods are used to provide free/active drug concentrations. To what extent hybrid LC–MS/MS procedures enable correct ‘active’ drug quantification is currently under consideration. Experimental & results: The relevance of appropriate extraction condition was evaluated by a hybrid target capture immuno-affinity LC–MS/MS method using total and free/active quality controls (QCs). The rapid extraction (10 min) provided correct results, whereas overnight incubation resulted in significant overestimation of the free/active drug (monclonal antibody) concentration. Conventional total QCs were inappropriate to determine optimal method conditions in contrast to free/active QCs. Conclusion: The ‘free/active analyte QC concept’ enables development of appropriate extraction conditions for correct active drug quantification by hybrid LC–MS/MS.
Collapse
|
33
|
Factors that impact pharmacokinetic measurements of antibody therapeutics: what is your PK assay telling you? Bioanalysis 2017; 9:1531-1533. [DOI: 10.4155/bio-2017-0173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
34
|
Gill KL, Machavaram KK, Rose RH, Chetty M. Potential Sources of Inter-Subject Variability in Monoclonal Antibody Pharmacokinetics. Clin Pharmacokinet 2017; 55:789-805. [PMID: 26818483 DOI: 10.1007/s40262-015-0361-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Understanding inter-subject variability in drug pharmacokinetics and pharmacodynamics is important to ensure that all patients attain suitable drug exposure to achieve efficacy and avoid toxicity. Inter-subject variability in the pharmacokinetics of therapeutic monoclonal antibodies (mAbs) is generally moderate to high; however, the factors responsible for the high inter-subject variability have not been comprehensively reviewed. In this review, the extent of inter-subject variability for mAb pharmacokinetics is presented and potential factors contributing to this variability are explored and summarised. Disease status, age, sex, ethnicity, body size, genetic polymorphisms, concomitant medication, co-morbidities, immune status and multiple other patient-specific details have been considered. The inter-subject variability for mAb pharmacokinetics most likely depends on the complex interplay of multiple factors. However, studies aimed at investigating the reasons for the inter-subject variability are sparse. Population pharmacokinetic models and physiologically based pharmacokinetic models are useful tools to identify important covariates, aiding in the understanding of factors contributing to inter-subject variability. Further understanding of inter-subject variability in pharmacokinetics should aid in development of dosing regimens that are more appropriate.
Collapse
Affiliation(s)
- Katherine L Gill
- Simcyp (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Krishna K Machavaram
- Simcyp (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Rachel H Rose
- Simcyp (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Manoranjenni Chetty
- Simcyp (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK.
| |
Collapse
|
35
|
Validation of a ligand-binding assay for active protein drug quantification following the ‘free analyte QC concept’. Bioanalysis 2016; 8:2537-2549. [DOI: 10.4155/bio-2016-0172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: Active drug assays are becoming increasingly important in protein drug development. We describe the validation of a ligand-binding assay for active protein drug quantification and address practical challenges as well as regulatory implications. Results: A bioanalytical method for active protein drug quantification was successfully validated. Validation data prove that this method can be routinely used applying the commonly accepted acceptance criteria for ligand-binding assays. Conclusion: Active drug assays are a powerful tool to elucidate the pharmacokinetic/pharmacodynamic relationship as they take into consideration the influence of various matrix components, such as soluble ligand and anti-drug antibodies. However, not all aspects of the validation concept described in the guidelines for pharmacokinetic assays can be applied to active drug assays and thus regulatory guidelines should be adapted in consequence.
Collapse
|
36
|
Bypassing nonparallelism of a monoclonal antibody ligand-binding assay by employment of alternative assay formats. Bioanalysis 2016; 8:2581-2593. [PMID: 27884079 DOI: 10.4155/bio-2016-0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Determination of concentration-time profiles in cynomolgus monkeys of a therapeutic monoclonal antibody against a soluble target revealed a substantial discrepancy between a generic anti-human IgG capture/detection and target bridging assay with the target bridging assay leading to dose- and time-dependent underquantification of drug concentrations, lack of parallelism and subsequently different pharmacokinetic parameters. In contrast, plasma levels derived from a target capture and an anti-idiotypic antibody bridging assay were in close concordance with the generic assay and demonstrated parallelism with high precision across several dilutions. The results provide a practical attempt to overcome nonparallelism by employing alternative assay formats utilizing tailored assay reagent combinations in order to obtain unbiased pharmacokinetic data.
Collapse
|
37
|
Samineni D, Girish S, Li C. Impact of Shed/Soluble targets on the PK/PD of approved therapeutic monoclonal antibodies. Expert Rev Clin Pharmacol 2016; 9:1557-1569. [DOI: 10.1080/17512433.2016.1243055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
38
|
Abstract
This article provides an overview of the information and factors relevant to designing bioanalytical strategies in support of in vivo nonclinical and clinical studies of protein therapeutics. The summarized information includes representative types of the therapeutic proteins, their key structural characteristics, the relationship between post-translational modifications and function, issues during purification and formulation, PK of therapeutic proteins and immunogenicity. The effect of each of those on bioanalysis strategy has been pointed out. The impacts of structural variant and ‘free’/‘bound’ forms on PK assessment have been discussed.
Collapse
|
39
|
Mayer AP, Hottenstein CS. Ligand-Binding Assay Development: What Do You Want to Measure Versus What You Are Measuring? AAPS JOURNAL 2015; 18:287-9. [PMID: 26669789 DOI: 10.1208/s12248-015-9855-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/04/2015] [Indexed: 01/11/2023]
Affiliation(s)
- Andrew P Mayer
- GlaxoSmithKline Pharmaceuticals Research and Development, 709 Swedeland Road, King of Prussia, Pennsylvania, 19406, USA. .,Platform Technology and Science, DMPK-Bioanalytical Science and Toxicokinetics, King of Prussia, Pennsylvania, USA.
| | - Charles S Hottenstein
- GlaxoSmithKline Pharmaceuticals Research and Development, 709 Swedeland Road, King of Prussia, Pennsylvania, 19406, USA.,Platform Technology and Science, DMPK-Bioanalytical Science and Toxicokinetics, King of Prussia, Pennsylvania, USA
| |
Collapse
|
40
|
Proposal for a harmonized descriptive analyte nomenclature for quantitative large-molecule bioanalysis. Bioanalysis 2015; 7:3057-62. [DOI: 10.4155/bio.15.218] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
41
|
Quantification of a bifunctional drug in the presence of an immune response: a ligand-binding assay specific for ‘active’ drug. Bioanalysis 2015; 7:3097-106. [DOI: 10.4155/bio.15.213] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Aim: During development of biologics, safety and efficacy assessments are often hampered by immune responses to the treatment. The raised antidrug antibodies (ADA) might interfere with the bioanalytical method and complicate result interpretation if non-fully characterized bioanalytical methods were applied. Methods: Here, we report an approach to characterize a ligand-binding assay (LBA) for the quantification of active drug exposure of a bifunctional therapeutic protein in the presence of antidrug antibodies, by correlating LBA results with those of a cell-based PK assay. Results: A clear correlation between both assays could be observed when monoclonal and polyclonal antibodies against the toxin moiety of the drug were used as ADA surrogates, and results were confirmed with human ADA-positive sera. Conclusion: The observed correlation between the LBA-based and cell-based PK assay indicated the suitability of the developed LBA for the determination of active drug exposure in the presence of an immune response.
Collapse
|
42
|
Willrich MAV, Murray DL, Barnidge DR, Ladwig PM, Snyder MR. Quantitation of infliximab using clonotypic peptides and selective reaction monitoring by LC–MS/MS. Int Immunopharmacol 2015. [DOI: 10.1016/j.intimp.2015.07.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
43
|
Peng K, Xu K, Liu L, Hendricks R, Delarosa R, Erickson R, Budha N, Leabman M, Song A, Kaur S, Fischer SK. Critical role of bioanalytical strategies in investigation of clinical PK observations, a Phase I case study. MAbs 2015; 6:1500-8. [PMID: 25484037 DOI: 10.4161/mabs.36208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RG7652 is a human immunoglobulin 1 (IgG1) monoclonal antibody (mAb) targeting proprotein convertase subtilisin/kexin type 9 (PCSK9) and is designed for the treatment of hypercholesterolemia. A target-binding enzyme-linked immunosorbent assay (ELISA) was developed to measure RG7652 levels in human serum in a Phase I study. Although target-binding assay formats are generally used to quantify free therapeutic, the actual therapeutic species being measured are affected by assay conditions, such as sample dilution and incubation time, and levels of soluble target in the samples. Therefore, in the presence of high concentrations of circulating target, the choice of reagents and assay conditions can have a significant effect on the observed pharmacokinetic (PK) profiles. Phase I RG7652 PK analysis using the ELISA data resulted in a nonlinear dose normalized exposure. An investigation was conducted to characterize the ELISA to determine whether the assay format and reagents may have contributed to the PK observation. In addition, to confirm the ELISA results, a second orthogonal method, liquid chromatography tandem mass spectrometry (LC-MS/MS) using a signature peptide as surrogate, was developed and implemented. A subset of PK samples, randomly selected from half of the subjects in the 6 single ascending dose (SAD) cohorts in the Phase I clinical study, was analyzed with the LC-MS/MS assay, and the data were found to be comparable to the ELISA data. This paper illustrates the importance of reagent characterization, as well as the benefits of using an orthogonal approach to eliminate bioanalytical contributions when encountering unexpected observations.
Collapse
Key Words
- 5, 5′-tetramethylbenzidine;
- BSA, bovine serum albumin
- CDR, complementarity-determining region
- ELISA, enzyme-linked immunosorbent assay
- HRP, horseradish peroxidase
- IS, internal standard
- IgG1, immunoglobulin G1
- LC-MS/MS
- LC-MS/MS, liquid chromatography tandem mass spectrometry
- LDL-c, low density lipoprotein cholesterol
- LDLR, low density lipoprotein receptor
- LLOQ, lower limit of quantification
- MAD, multiple-ascending dose
- MQC, minimum quantifiable concentration
- MRM, multiple reaction monitoring
- NHS, normal human sera
- PBS, phosphate buffered saline
- PCSK9, proprotein convertase subtilisin/kexin type 9;
- PD, pharmacodynamics
- PK, pharmacokinetics
- RG7652
- RT, room temperature
- S/N, signal-to-noise
- SA, streptavidin
- SAD, single-ascending dose
- SIL, stable isotope-labeled
- TMB, 3, 3′
- clinical pharmacokinetic assay
- enzyme-linked immunosorbent assay
- mAbs, monoclonal antibodies
- proprotein convertase subtilisin/kexin type 9
- rhuPCSK9, recombinant human PCSK9
- signature peptide
Collapse
Affiliation(s)
- Kun Peng
- a Department of BioAnalytical Sciences; Genentech Inc ; South San Francisco , CA USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Talbot JJ, Calamba D, Pai M, Ma M, Thway TM. Measurement of Free Versus Total Therapeutic Monoclonal Antibody in Pharmacokinetic Assessment is Modulated by Affinity, Incubation Time, and Bioanalytical Platform. AAPS JOURNAL 2015; 17:1446-54. [PMID: 26265093 DOI: 10.1208/s12248-015-9807-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 07/15/2015] [Indexed: 01/13/2023]
Abstract
Decisions about efficacy and safety of therapeutic proteins (TP) designed to target soluble ligands are made in part by their ex vivo quantification. Ligand binding assays (LBAs) are critical tools in measuring serum TP levels in pharmacokinetic, toxicokinetic, and pharmacodynamic studies. This study evaluated the impact of reagent antibody affinities, assay incubation times, and analytical platform on free or total TP quantitation. An ELISA-based LBA that measures monoclonal anti-sclerostin antibody (TPx) was used as the model system. To determine whether the method measures free or total TPx, the effects of K on, K off, and K D were determined. An 8:1 molar ratio of sclerostin (Scl) to TPx compared to a 1:1 molar ratio produced by rabbit polyclonal antibodies to TPx was required to achieve IC50, a measure of TPx interference effectiveness, making it unclear whether the ELISA truly measured free TPx. Kinetic analysis revealed that Scl had a rapid dissociation rate (K off) from TPx and that capture and detection antibodies had significantly higher binding affinities (K D) to TPx. These kinetic limitations along with long ELISA incubation times lead to the higher molar ratios (8:1) required for achieving 50% inhibition of TPx. However, a microfluidic platform with the same reagent pairs required shorter incubations to achieve a lower Scl IC50 molar ratio (1:1). The findings from this study provide the bioanalytical community with a deeper understanding of how reagent and platform selection for LBAs can affect what a particular method measures, either free or total TP concentrations.
Collapse
Affiliation(s)
- Jeffrey J Talbot
- Department of Pharmacokinetic and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, 91320, USA.,Regeneron Pharmaceuticals, 81 Columbia Turnpike, Rensselaer, New York, 12144, USA
| | - Dominador Calamba
- Department of Pharmacokinetic and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, 91320, USA
| | - Melody Pai
- Department of Pharmacokinetic and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, 91320, USA.,Department of Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA
| | - Mark Ma
- Department of Pharmacokinetic and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, 91320, USA
| | - Theingi M Thway
- Department of Pharmacokinetic and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, 91320, USA.
| |
Collapse
|
45
|
Choosing the right bioanalytical assay platform(s) to support the PK assessment of protein biotherapeutic programs. Bioanalysis 2015; 7:1197-9. [DOI: 10.4155/bio.15.62] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
46
|
Williams L, Sank M, Chimalakonda A, Ni Y, Saewert M, DeSilva B, Pillutla R. Development and characterization of a free therapeutic ligand binding assay with assistance from kinetics modeling. J Immunol Methods 2015; 419:18-24. [DOI: 10.1016/j.jim.2015.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 11/30/2022]
|
47
|
Salimi-Moosavi H, Winters A, Abbott C, Patel J, Hager T, Patel V, Shih J, Zhuang Y, Ma M. A multifactorial screening strategy to identify anti-idiotypic reagents for bioanalytical support of antibody therapeutics. Anal Biochem 2014; 470:52-60. [PMID: 25447458 DOI: 10.1016/j.ab.2014.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 09/24/2014] [Accepted: 10/13/2014] [Indexed: 01/28/2023]
Abstract
Antibodies are critical tools for protein bioanalysis; their quality and performance dictate the caliber and robustness of ligand binding assays. After immunization, polyclonal B cells generate a diverse antibody repertoire against constant and variable regions of the therapeutic antibody immunogen. Herein we describe a comprehensive and multifactorial screening strategy to eliminate undesirable constant region-specific antibodies and select for anti-idiotypic antibodies with specificity for the unique variable region. Application of this strategy is described for the therapeutic antibody Mab-A case study. Five different factors were evaluated to select a final antibody pair for the quantification of therapeutics in biological matrices: (i) matrix effect in preclinical and clinical matrices, (ii) assay sensitivity with lower limit of quantification goal of single-digit ng/ml (low pM) at a signal-to-background ratio greater than 5, (iii) epitope distinction or nonbridging antibody pair, (iv) competition with target and inhibitory capacity enabling measurement of free drug, and (v) neutralizing bioactivity using bioassay. The selected antibody pair demonstrated superior assay sensitivity with no or minimal matrix effect in common biological samples, recognized two distinct binding epitopes on the therapeutic antibody variable region, and featured inhibitory and neutralizing effects with respect to quantification of free drug levels.
Collapse
Affiliation(s)
- Hossein Salimi-Moosavi
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA 91320, USA.
| | - Aaron Winters
- Department of Discovery Science, Amgen, Thousand Oaks, CA 91320, USA
| | - Christina Abbott
- Department of Protein Technologies, Amgen, Thousand Oaks, CA 91320, USA
| | - Jennifer Patel
- Department of Clinical Immunology, Amgen, Thousand Oaks, CA 91320, USA
| | - Todd Hager
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA 91320, USA
| | - Vimal Patel
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA 91320, USA
| | - Judy Shih
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA 91320, USA
| | - Yao Zhuang
- Department of Clinical Immunology, Amgen, Thousand Oaks, CA 91320, USA
| | - Mark Ma
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA 91320, USA
| |
Collapse
|
48
|
Colbert A, Umble-Romero A, Prokop S, Xu R, Gibbs JP, Pederson S. Characterization of a quantitative method to measure free proprotein convertase subtilisin/kexin type 9 in human serum. MAbs 2014; 6:1103-13. [PMID: 24859266 PMCID: PMC4171013 DOI: 10.4161/mabs.28719] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 03/28/2014] [Accepted: 03/31/2014] [Indexed: 01/17/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a serine protease that plays an important role in the regulation of serum low-density lipoprotein (LDL) cholesterol by downregulation of LDL receptor, and as such is considered a novel target in cholesterol lowering therapy. In support of the drug development program for Evolocumab, a fully human IgG₂ antibody that targets PCSK9, a quantitative ELISA to measure free PCSK9 in human serum was developed. PCSK9 serves as a biomarker of pharmacological response during treatment, and measuring levels of the free ligand post-dosing was of interest as an aid to establishing the pharmacokinetic and pharmacodynamic properties of the therapeutic. Given the complexities associated with the measurement of free ligand in the presence of high concentrations of circulating drug, it was important to challenge the method with experiments designed to assess ex vivo conditions that have the potential to affect the binding equilibrium of drug and ligand within test samples during routine sampling handling and assay conditions. Herein, we report results of experiments that were conducted to characterize the assay in alignment with regulatory guidance and industry standards, and to establish evidence that the method is measuring the free ligand in circulation at the time serum was collected. A robust supporting data package was generated that demonstrates the method specifically and reproducibly measures the free ligand, and is suitable for its intended use.
Collapse
Affiliation(s)
- Alexander Colbert
- Department of Pharmacokinetics and Drug Metabolism; Amgen Inc.; Seattle, WA USA
| | - Amber Umble-Romero
- Department of Pharmacokinetics and Drug Metabolism; Amgen Inc.; Seattle, WA USA
| | - Samantha Prokop
- Department of Pharmacokinetics and Drug Metabolism; Amgen Inc.; Seattle, WA USA
| | - Ren Xu
- Department of Pharmacokinetics and Drug Metabolism; Amgen Inc.; Seattle, WA USA
| | - John P Gibbs
- Department of Pharmacokinetics and Drug Metabolism; Amgen Inc.; Seattle, WA USA
| | - Susan Pederson
- Department of Pharmacokinetics and Drug Metabolism; Amgen Inc.; Seattle, WA USA
| |
Collapse
|
49
|
The integration of ligand binding and LC-MS-based assays into bioanalytical strategies for protein analysis. Bioanalysis 2014; 6:1827-41. [DOI: 10.4155/bio.14.128] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Both LBAs and LC–MS-based assays are reviewed and summarized for applications in quantitative protein analysis. A strategy for platform selection is proposed based on several factors that contribute to the complexities of bioanalysis of biologics. Protein types, multiple co-existing forms, post-translational modifications, and affinities to ADA, targets, and endogenous proteins need to be considered when selecting the most appropriate platform. Other factors, such as intended use of data, assay sensitivity, available reagents, and multiple analytes also impact on the choice of bioanalytical platform. At BMS, strategies for the seamless integration of both platforms are being implemented to provide not only PK/PD data of the molecules but also useful information of the amino acid structure and functional relationship of the proteins.
Collapse
|
50
|
Hall MP. Biotransformation and in vivo stability of protein biotherapeutics: impact on candidate selection and pharmacokinetic profiling. Drug Metab Dispos 2014; 42:1873-80. [PMID: 24947971 DOI: 10.1124/dmd.114.058347] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Historically, since the metabolism of administered peptide/protein drugs ("biotherapeutics") has been expected to undergo predictable pathways similar to endogenous proteins, comprehensive biotherapeutic metabolism studies have not been widely reported in the literature. However, since biotherapeutics have rapidly evolved into an impressive array of eclectic modalities, there has been a shift toward understanding the impact of metabolism on biotherapeutic development. For biotherapeutics containing non-native chemical linkers and other moieties besides natural amino acids, metabolism studies are critical as these moieties may impart undesired toxicology. For biotherapeutics that are composed solely of natural amino acids, where end-stage peptide and amino acid catabolites do not generally pose toxicity concerns, the understanding of biotherapeutic biotransformation, defined as in vivo modifications such as peripherally generated intermediate circulating catabolites prior to end-stage degradation or elimination, may impact in vivo stability and potency/clearance. As of yet, there are no harmonized methodologies for understanding biotherapeutic biotransformation and its impact on drug development, nor is there clear guidance from regulatory agencies on how and when these studies should be conducted. This review provides an update on biotherapeutic biotransformation studies and an overview of lessons learned, tools that have been developed, and suggestions of approaches to address issues. Biotherapeutic biotransformation studies, especially for certain modalities, should be implemented at an early stage of development to 1) understand the impact on potency/clearance, 2) select the most stable candidates or direct protein re-engineering efforts, and 3) select the best bioanalytical technique(s) for proper drug quantification and subsequent pharmacokinetic profiling and exposure/response assessment.
Collapse
Affiliation(s)
- Michael P Hall
- Department of Pharmacokinetics & Drug Metabolism, Amgen Inc., Thousand Oaks, California
| |
Collapse
|