Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes

Total Page:16

File Type:pdf, Size:1020Kb

Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes Paula Denney Eason, Renée C. Benson, Jenny T. Ly, Rachel A. Saxer, James L. Wilbur, George B. Sigal, Eli N. Glezer, Hans A. Biebuyck, Jacob N. Wohlstadter, and Robert M. Umek TM TM A division of Meso Scale Diagnostics,TM LLC. Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes Abstract This poster presents a robust, receptor-ligand binding assay based upon a novel assay platform developed by Meso Scale DiscoveryTM (MSDTM). MSD’s platform combines array technologies and electrochemiluminescence detection to achieve ultra-fast, highly sensitive assays in a homogeneous format. Cellular membranes containing the EGF receptor were passively adsorbed to MSD proprietary coated electrodes embedded in multi-well plates. Binding of EGF to the EGF receptor was detected by inducing and measuring electrochemi- luminescence from a labeled EGF ligand. Approximately 1000 cell equivalents per well yielded a signal to background ratio of 20. The observed KD agrees with that reported in the literature and demonstrates that immobilization of the membranes and modification of the ligand do not alter the binding affinity. Binding specificity was confirmed with two inhibitors. The assay can be readily adapted to facilitate analysis of a broad array of receptor-ligand interactions. TM TM A division of Meso Scale Diagnostics,TM LLC. Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes TM Multi-Array TechnologyTM Multi-Array Technology Unified technology platform with instruments, plates and reagents for drug discovery for drug discovery. Combines the power of microarrays with the sensitivity of electrochemiluminescence Combines the power of microarrays with the sensitivity of electrochemiluminescence.96-, 384- and 1536 microplate formats 96-,Multi-Spot 384- TMand plates 1536 with microplate high density formats. arrays for multiplexing Multi-SpotSector HTSTMTM platesInstrument: with highHigh densityresolution arrays imaging for multiplexing. detection and robotic integration for HTS and large-scale proteomics TM Sector HTS Instrument: High resolution imaging detection and Sector HTS roboticSector PRintegrationTM Instrument: for HTSMedium and throughputlarge-scale proteomics.benchtop reader for assay development, cellular and molecular biology, research in therapeutic areas, secondary screening, QC. Assays developed on Sector PRTM portInstrument: to Sector Medium HTS. throughput benchtop reader for assay development, cellular and molecular biology, research in therapeutic areas, secondary screening, QC. Assays developed on Sector PR port to Sector HTS. Sector PR TM TM A division of Meso Scale Diagnostics,TM LLC. Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes EGF-EGFR Assay Format Extra-Cellular Domain Trans-Membrane Domain EGFR A-431 Cells Cytoplasmic Domain 1) Lyse cells 2) Purify membrane fragments 3) Passively adsorb membrane fragments on electrode, 30min MSD Proprietary Coated Carbon Electrodes Bind labeled EGF (STAG-EGF), 1h STAG-EGF 1)Wash away unbound STAG-EGF 1)Add TPA 2)Add TPA 2)Induce and Measure 3)Induce and Measure electrochemiluminescence (ECL) electrochemiluminescence(ECL) ECL ECL ECL ECL Washed Non-washed Format Format TM TM A division of Meso Scale Diagnostics,TM LLC. Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes Direct Immobilization of Biologically Active Membranes on Carbon Electrodes 10µg 10µg Total Total Supernatant Supernatant 1 2 1 2 A-431 K-562 Western Silver Quantification of Membrane Immobilization Blot Stain on the Electrode Surface Ten micrograms of total membrane protein was loaded directly onto the gel. For comparison, the supernatant containing the unbound fraction recovered after 10µg total membrane protein was incubated on the electrode band used to quantify for 30 min is shown. α-EGFR antibody was used for the western blot. 4000 Optimal Mass Immobilized on the Electrode Titration of total membrane protein to determine optimal plating 3000 A431 mass. In this 96-well washed format, membranes were immobilized K562 onto MSD proprietary coated carbon electrodes in 5µL/well for 1 hour. 2000 Next, labeled ligand was added in 3% BSA for 1 hour. Unbound Average Signal ligand was washed, followed by the addition of assay buffer just prior 1000 to electrochemiluminescence. 0 01234 Total Membrane Protein (µg/well) TM TM A division of Meso Scale Diagnostics,TM LLC. Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes Binding Characteristics of Membranes are Conserved After Immobilization on Carbon Electrodes Saturation-Binding Curve Saturation-Binding Curve 96 - Well Washed Format 384 - Well Washed Format 12500 1000 10000 A431 750 A431 K562 K562 7500 500 5000 Average Signal Average Signal KD = 4.7 KD = 9.9 250 2500 0 0 0 50 100 150 200 250 300 0 100 200 300 STAG-EGF (nM) STAG-EGF (nM) EGF was purchased (Sigma) and labeled with a sulfonated derivative of Ruthenium (II) tris-bipyridine (the electrochemiluminescent label, here abbreviated as STAG). STAG-EGF was purified by FPLC and the molecular weight of the labeled ligand was confirmed by mass-spectral analysis. Purified STAG-EGF was quantified by both a BCA protein assay and ruthenium absorbance at 455nm. As little as 1000 or 300 cell equivalents/well in the 96- and 384-well formats, respectively, produced maximum signal to background. The assay was performed using the standard protocol. The observed KD values in both the 96- and 384-well assays were within 3-5 fold of that reported in scientific literature, demonstrating a very modest change in binding affinity of STAG-EGF relative to native EGF. TM TM A division of Meso Scale Diagnostics,TM LLC. Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes Receptors in Immobilized Membranes Retain Sensitivity to Known Inhibitors Suramin Competition Cold EGF Competition 96-Well Washed Format 96-Well Washed Format 10000 4000 A431 A431 K562 K562 7500 3000 5000 IC50 = 20nM 2000 IC50 = 1.8mM Average Signal STAG-EGF = 120nM Average Signal STAG-EGF = 15nM 2500 1000 0 0 1e -6 1e -5 1e -4 1e -3 .01 0.1 1 10 0.001 0.01 0.1 1 10 100 EGF (nM) Suramin (mM) Receptor binding to STAG-EGF was assayed as a function of varying concentrations of unlabeled EGF. The observed IC50 value of the unmodified ligand relative to the STAG-EGF challenge is in agreement with the slightly greater KD observed for the STAG-EGF. Suramin, another compound known to inhibit binding of several growth factor receptors, also inhibited STAG-EGF binding in the assay. The assay was conducted according to the standard protocol except that the labeled ligand and the inhibitor were incubated simultaneously in the plate. TM TM A division of Meso Scale Diagnostics,TM LLC. Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes Membranes Immobilized on Carbon Electrodes are Compatible with HTS 384-Well Washed Format 60nM STAG-EGF A431 K562 Z -factors for all 5 plates >0.5 100 Signal Signal to Background = 6.3 A-431 CV = 10.2% K-562 CV = 7.3% CV for all 5 plates = 8.7% plate 1 plate 2 plate 3 plate 4 plate 5 0.57 0.56 0.63 0.53 0.68 Z -factor 10 Well Number In this 384-well, washed format, membranes were deposited with a PixSysTM instrument (Cartesian Technologies) and R allowed to incubate on carbon electrodes for 30min. Labeled ligand, 60nM STAG-EGF, in 3% BSA was added by a RapidPlate instrument (Zymark) and incubated for 1 hour at ambient temperature. Unbound ligand was washed away, followed by the addition of assay buffer on the Rapid-Plate just prior to inducing electrochemiluminescence. The assay is adaptable to a fully integrated work system for high-throughput screens. TM TM A division of Meso Scale Diagnostics,TM LLC. Simple, Rapid Ligand-Binding Assay Using Immobilized Cellular Membranes Conclusion 1. We have developed a simple, rapid assay to detect the binding of the EGF ligand to EGFR via passive adsorption of biologically active membranes onto disposable, MSD proprietary coated carbon electrodes. 2. Quantitative membrane immobilization demonstrates biologically active A-431 and K-562 membranes. As little as 0.5µg (1000 cell equivalents) and 0.15µg (300 cell equivalents) total membrane protein/well, produced maximum signal to background in the 96- and 384-well assays, respectively. 3. Immobilization of the membranes onto MSD proprietary coated carbon electrodes and modification of the ligand result in a very modest alteration in binding affinity. Observed KDs in both the washed and non-washed formats agree with that reported in literature, while characteristic competition curves with cold ligand and a known inhibitor demonstrate receptor-ligand binding specificity. 4. The assay is adaptable to completely automated 96- and 384-well formats for HTS. A 5-plate screen produced Z-factors > 0.5/plate and interplate CV’s = 8.7% in a 384-well washed format. TM TM A division of Meso Scale Diagnostics,TM LLC..
Recommended publications
  • Clinical Pharmacology 1: Phase 1 Studies and Early Drug Development
    Clinical Pharmacology 1: Phase 1 Studies and Early Drug Development Gerlie Gieser, Ph.D. Office of Clinical Pharmacology, Div. IV Objectives • Outline the Phase 1 studies conducted to characterize the Clinical Pharmacology of a drug; describe important design elements of and the information gained from these studies. • List the Clinical Pharmacology characteristics of an Ideal Drug • Describe how the Clinical Pharmacology information from Phase 1 can help design Phase 2/3 trials • Discuss the timing of Clinical Pharmacology studies during drug development, and provide examples of how the information generated could impact the overall clinical development plan and product labeling. Phase 1 of Drug Development CLINICAL DEVELOPMENT RESEARCH PRE POST AND CLINICAL APPROVAL 1 DISCOVERY DEVELOPMENT 2 3 PHASE e e e s s s a a a h h h P P P Clinical Pharmacology Studies Initial IND (first in human) NDA/BLA SUBMISSION Phase 1 – studies designed mainly to investigate the safety/tolerability (if possible, identify MTD), pharmacokinetics and pharmacodynamics of an investigational drug in humans Clinical Pharmacology • Study of the Pharmacokinetics (PK) and Pharmacodynamics (PD) of the drug in humans – PK: what the body does to the drug (Absorption, Distribution, Metabolism, Excretion) – PD: what the drug does to the body • PK and PD profiles of the drug are influenced by physicochemical properties of the drug, product/formulation, administration route, patient’s intrinsic and extrinsic factors (e.g., organ dysfunction, diseases, concomitant medications,
    [Show full text]
  • Affinity-Based, Biophysical Methods to Detect and Analyze Ligand Binding
    Journal of Structural Biology 172 (2010) 142–157 Contents lists available at ScienceDirect Journal of Structural Biology journal homepage: www.elsevier.com/locate/yjsbi Affinity-based, biophysical methods to detect and analyze ligand binding to recombinant proteins: Matching high information content with high throughput Geoff A. Holdgate a, Malcolm Anderson a, Fredrik Edfeldt b, Stefan Geschwindner b,* a Lead Generation Sciences, AstraZeneca R&D Alderley Park, 50F49 Mereside, Alderley Park, United Kingdom b Lead Generation Sciences, AstraZeneca R&D Mölndal, S-43183 Mölndal, Sweden article info abstract Article history: Affinity-based technologies have become impactful tools to detect, monitor and characterize molecular Available online 4 July 2010 interactions using recombinant target proteins. This can aid the understanding of biological function by revealing mechanistic details, and even more importantly, enables the identification of new improved Keywords: ligands that can modulate the biological activity of those targets in a desired fashion. The selection of the Affinity appropriate technology is a key step in that process, as each one of the currently available technologies Thermodynamic offers a characteristic type of biophysical information about the ligand-binding event. Alongside the Interaction indisputable advantages of each of those technologies they naturally display diverse restrictions that Fragment are quite frequently related to the target system to be studied but also to the affinity, solubility and Ligand Screening molecular size of the ligands. This paper discusses some of the theoretical and experimental aspects of the most common affinity-based methods, what type of information can be gained from each one of those approaches, and what requirements as well as limitations are expected from working with recombinant proteins on those platforms and how those can be optimally addressed.
    [Show full text]
  • Recommendations for the Bioanalytical Method Validation Of
    Pharmaceutical Research, Vol. 20, No. 11, November 2003 (© 2003) Research Paper Recommendations for the studies was held more than a decade ago in Crystal City, VA (1,2). The proceedings and recommendations of that meeting Bioanalytical Method Validation of essentially became a de facto guideline for bioanalytical meth- Ligand-binding Assays to Support ods validation within the pharmaceutical industry. The con- ference addressed validation of bioanalytical methods in gen- Pharmacokinetic Assessments eral, but acknowledged differences between chromatographic of Macromolecules and nonchromatographic assays, including immunoassays and microbiological-based methods. After the original Crystal City conference, bioanalytical methods validation was ad- dressed subsequently several times at meetings and in publi- Binodh DeSilva,1 Wendell Smith,2 Russell Weiner,3 cations (3–6). Most emphasis to date has been on validation of Marian Kelley,4,11 JoMarie Smolec,5 Ben Lee,6 bioanalytical methods for conventional small molecular Masood Khan,7 Richard Tacey,8 Howard Hill,9 and weight drugs, principally due to the rapid growth during the Abbie Celniker10 1990s in the use of hyphenated mass-spectrometry as a rou- tine analytical tool. Because of the evolution in divergent analytical tech- Received July 2, 2003; accepted July 30, 2003 nologies for conventional small molecules and macromol- Purpose. With this publication a subcommittee of the AAPS Ligand ecules and the growth in the interest of macromolecular Binding Assay Bioanalytical Focus Group (LBABFG) makes recom- therapeutics, the topic of bioanalytical methods validation mendations for the development, validation, and implementation of was revisited in 2000 in 2 meetings, one focused on small ligand binding assays (LBAs) that are intended to support pharma- molecule analytes (7) and the other focused on macromol- cokinetic and toxicokinetic assessments of macromolecules.
    [Show full text]
  • Challenges Associated with the Development and Validation of Flow Cytometry Assays
    Challenges associated with the development and validation of flow cytometry assays Carolyne Dumont, Eliane Moisan, Martin Poirier, Marie-Hélène Côté, and Marie-Soleil Piché 1 INTRODUCTION 2 Case Study 1 3 Case Study 2 Development of a PD marker by flow cytometry to assess the efficacy of Development of a flow cytometry assay for the measurement of basophil Flow cytometry is a technology allowing multi-parametric analysis of thousands of particles per second and helps to a chemokine neutralizing antibody activation in the context of a Phase III clinical study adequately identify or functionally characterize complex cell populations of interest. It is often used in basic research, Assay Design: The assay was required for the evaluation of pharmacodynamics (inhibition an agonist’s granulocytes activation activity) in Assay Design: Human whole blood samples were spiked with the different controls (or compounds in the clinical study) and further discovery, preclinical and clinical trials. With the increasing proportion of biologics in the pipeline, flow cytometry has proven preclinical studies. Non-Human Primate (NHP) or rat blood from treated animals was incubated at 3 conditions and granulocyte activation stained with an anti-CCR3 and anti-CD63 antibody. The validations stimulation conditions tested included a negative control (PBS) and itself to be an indispensable tool to assess safety, receptor occupancy (RO) or pharmacodynamics (PD). was measured as CD11b expression by flow cytometry (MFI). The conditions tested included a negative control (PBS), a positive control two positive controls (anti-FcεRI and fMLP). Basophils were identified as CCR3+ and upon activation, CD63 became externalized and (fMLP) and the test condition (agonist).
    [Show full text]
  • Pharmacokinetic-Pharmacodynamic Modelling of Systemic IL13 Blockade by Monoclonal Antibody Therapy: a Free Assay Disguised As Total
    pharmaceutics Article Pharmacokinetic-Pharmacodynamic Modelling of Systemic IL13 Blockade by Monoclonal Antibody Therapy: A Free Assay Disguised as Total John Hood 1,*, Ignacio González-García 1 , Nicholas White 1, Leeron Marshall 1,2, Vincent F. S. Dubois 1 , Paolo Vicini 1,3 and Paul G. Baverel 1,4 1 Clinical Pharmacology and Quantitative Pharmacology, AstraZeneca, Cambridge CB21 6GH, UK; [email protected] (I.G.-G.); [email protected] (N.W.); [email protected] (L.M.); [email protected] (V.F.S.D.); [email protected] (P.V.); [email protected] (P.G.B.) 2 Salford Royal Foundation Trust, Salford M6 8HD, UK 3 Confo Therapeutics, 9052 Ghent, Zwijnaarde, Belgium 4 Roche Pharma Research and Early Development, Clinical Pharmacology, Pharmaceutical Sciences, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland * Correspondence: [email protected]; Tel.: +44-1223-749-6288 Abstract: A sequential pharmacokinetic (PK) and pharmacodynamic (PD) model was built with Nonlinear Mixed Effects Modelling based on data from a first-in-human trial of a novel biologic, MEDI7836. MEDI7836 is a human immunoglobulin G1 lambda (IgG1λ-YTE) monoclonal antibody, Citation: Hood, J.; González-García, with an Fc modification to reduce metabolic clearance. MEDI7836 specifically binds to, and function- I.; White, N.; Marshall, L.; Dubois, ally neutralizes interleukin-13. Thirty-two healthy male adults were enrolled into a dose-escalation V.F.S.; Vicini, P.; Baverel, P.G. clinical trial. Four active doses were tested (30, 105, 300, and 600 mg) with 6 volunteers enrolled Pharmacokinetic-Pharmacodynamic per cohort. Eight volunteers received placebo as control.
    [Show full text]
  • UBI® SARS-Cov-2 ELISA INSTRUCTIONS for USE
    UBI® SARS-CoV-2 ELISA INSTRUCTIONS FOR USE FOR IN VITRO DIAGNOSTIC USE ONLY FOR EMERGENCY USE AUTHORIZATION ONLY FOR PRESCRIPTION USE ONLY INTENDED USE The UBI® SARS-CoV-2 ELISA is an Enzyme-Linked Immunosorbent Assay (ELISA) intended for qualitative detection of IgG antibodies to SARS-CoV-2 in human serum and plasma (sodium heparin or dipotassium (K2) EDTA). The UBI® SARS-CoV-2 ELISA is intended for use as an aid in identifying individuals with an adaptive immune response to SARS-CoV-2, indicating recent or prior infection. At this time, it is unknown for how long antibodies persist following infection and if the presence of antibodies confers protective immunity. The UBI® SARS-CoV-2 ELISA should not be used to diagnose or exclude acute SARS-CoV-2 infection. Testing is limited to laboratories certified under the Clinical Laboratory Improvement Amendments of 1988 (CLIA), 42 U.S.C 263a, that meet requirements to perform high complexity testing. Results are for the detection of IgG SARS CoV-2 antibodies. IgG antibodies to SARS-CoV-2 are generally detectable in blood several days after initial infection, although the duration of time antibodies are present post-infection is not well characterized. Individuals may have detectable virus present for several weeks following seroconversion. Laboratories within the United States and its territories are required to report all results to the appropriate public health authorities. The sensitivity of the UBI® SARS-CoV-2 ELISA early after infection is unknown. Negative results do not preclude acute SARS-CoV-2 infection. If acute infection is suspected, direct testing for SARS-CoV-2 is necessary.
    [Show full text]
  • Molecular Diagnostic Assay Validation Update to the 2009 AMP Molecular Diagnostic Assay Validation White Paper
    Molecular Diagnostic Assay Validation Update to the 2009 AMP Molecular Diagnostic Assay Validation White Paper Members of the 2013 and 2014 Clinical Practice Committees of the Association for Molecular Pathology The 2013 AMP Clinical Practice Committee consisted of Matthew J. Bankowski, Milena Cankovic, Jennifer Dunlap, Larissa V. Furtado*, Jerald Gong, Thomas Huard, Linda Jeng*, Loren Joseph (Chair), Annette Kim, Bryan Krock, Marilyn M. Li, Mary Lowery-Nordberg, Melissa Miller, Caroline Sue Richards, and Paul Rothberg. *Project leads Standard of practice is not defined by this article, and there may be alternatives. See Disclaimer for further details. INTRODUCTION The goal of method validation in the molecular diagnostics laboratory is to ensure that a given test is ready for implementation in the clinical laboratory. To reach that goal, each step of the testing process must be carefully evaluated and documented. Such validation is relatively standardized for high volume automated assays in fields such as clinical chemistry. It is challenging to apply those standardized practices to molecular diagnostics laboratories, which rely heavily on low-volume labor-intensive tests, both FDA approved and laboratory- developed. Excellent and comprehensive documents have recently been published on this topic 1-10. This overview is intended to provide a brief practical reference that can be used to guide the validation process in molecular diagnostics laboratories. In addition, a sample summary checklist is provided in the appendix as a template to facilitate documentation of laboratory director review and sign off on the validation process when new assays are developed or adopted. SUMMARY CLIA (42 CFR 493.1253) and CAP (MOL 30785-31705) require that laboratories validate the performance of tests before reporting patient results.
    [Show full text]
  • Real-Time Quantitative Measurement of Autocrine Ligand Binding Indicates That Autocrine Loops Are Spatially Localized
    Proc. Natl. Acad. Sci. USA Vol. 95, pp. 15368–15373, December 1998 Cell Biology Real-time quantitative measurement of autocrine ligand binding indicates that autocrine loops are spatially localized DOUGLAS A. LAUFFENBURGER*†‡,GREGORY T. OEHRTMAN†,LAURA WALKER†, AND H. STEVEN WILEY§ *Division of Bioengineering & Environmental Health and Center for Biomedical Engineering and †Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139; and §Department of Pathology, University of Utah Medical Center, Salt Lake City, UT 84132 Communicated by Edwin N. Lightfoot, Jr., University of Wisconsin, Madison, WI, October 15, 1998 (received for review July 21, 1998) ABSTRACT Autocrine ligands are important regulators rapid (,30 sec) and small (,0.1 unit) changes in solution pH of many normal tissues and have been implicated in a number in the cellular microenvironment in an '1 ml chamber above of disease states, including cancer. However, because by the sensor. These pH changes [termed ‘‘extracellular acidifi- definition autocrine ligands are synthesized, secreted, and cation rate’’ (ECAR)] can arise from both metabolic and bound to cell receptors within an intrinsically self-contained regulatory events and have been shown to be quantitatively ‘‘loop,’’ standard pharmacological approaches cannot be used related to specific activation of many types of cell receptors, to investigate relationships between ligandyreceptor binding including tyrosine kinase receptors, G protein receptors, and and consequent cellular responses. We demonstrate here a ion channel receptors (16) with EC50 values similar to those new approach for measurement of autocrine ligand binding to derived from direct-labeled ligand binding (17). Hence, for cells, using a microphysiometer assay originally developed for exogenous ligands the microphysiometer can be used to obtain investigating cell responses to exogenous ligands.
    [Show full text]
  • A Live Cell Nanobret Binding Assay Allows the Study of Ligand-Binding Kinetics to the Adenosine A3 Receptor
    Purinergic Signalling https://doi.org/10.1007/s11302-019-09650-9 ORIGINAL ARTICLE A live cell NanoBRET binding assay allows the study of ligand-binding kinetics to the adenosine A3 receptor Monica Bouzo-Lorenzo1,2 & Leigh A. Stoddart 1,2 & Lizi Xia3 & Adriaan P. IJzerman3 & Laura H. Heitman 3 & Stephen J. Briddon1,2 & Stephen J. Hill1,2 Received: 25 November 2018 /Accepted: 14 February 2019 # The Author(s) 2019 Abstract There is a growing interest in understanding the binding kinetics of compounds that bind to G protein-coupled receptors prior to progressing a lead compound into clinical trials. The widely expressed adenosine A3 receptor (A3AR) has been implicated in a range of diseases including immune conditions, and compounds that aim to selectively target this receptor are currently under development for arthritis. Kinetic studies at the A3AR have been performed using a radiolabelled antagonist, but due to the kinetics of this probe, they have been carried out at 10 °C in membrane preparations. In this study, we have developed a live cell NanoBRET ligand binding assay using fluorescent A3AR antagonists to measure kinetic parameters of labelled and unlabelled compounds at the A3AR at physiological temperatures. The kinetic profiles of four fluorescent antagonists were determined in kinetic association assays, and it was found that XAC-ser-tyr-X-BY630 had the longest residence time (RT = 288 ± 62 min) at the A3AR. The association and dissociation rate constants of three antagonists PSB-11, compound 5, and LUF7565 were also determined using two fluorescent ligands (XAC-ser- tyr-X-BY630 or AV039, RT = 6.8 ± 0.8 min) as the labelled probe and compared to those obtained using a radiolabelled antagonist ([3H]PSB-11, RT = 44.6 ± 3.9 min).
    [Show full text]
  • White Paper Recommendations for the Development and Validation Of
    The AAPS Journal ( # 2017) DOI: 10.1208/s12248-017-0181-6 White Paper Recommendations for the Development and Validation of Neutralizing Antibody Assays in Support of Biosimilar Assessment D. Gouty,1,16 C. C. Cai,2 X. Y. Cai,3 A. Kasinath,4 V. Kumar,5 S. Alvandkouhi,6 J. Yang,7 S. Pederson,8 B. Babbitt,2 D. Peritt,9 A. Rudy,10 V. Koppenburg,11 A. Dasilva,12 M. Ullmann,13 S. Liu,14 and C. Satterwhite15 Received 23 May 2017; accepted 21 November 2017 Abstract. The American Association of Pharmaceutical Scientists (AAPS) biosimilar focus group on nonclinical and clinical assays has developed this manuscript to guide the industry on best practices and testing strategies when developing neutralizing antibody (NAb) assays for biosimilar programs. The immunogenicity assessment to biosimilar and originator drug products is one of the key aspects of clinical programs for biosimilars to demonstrate biosimilarity. Establishing that there are no clinically meaningful differences in immune response between a proposed product and the originator product is a key element in the demonstration of biosimilarity. It is critical to collect, evaluate, and compare the safety and immunogenicity data from the clinical pharmacology, safety, and/or efficacy studies especially when the originator drug product is known to have potential for immune-mediated toxicity. This manuscript aims to provide a comprehensive review and recommendations on assay formats, critical reagents, approaches to method development, and validation of the neutralizing antibody assays in extrapolation within the scope of biosimilar drug development programs. Even if there are multiple options on the development and validation of NAb assays for biosimilar programs, the type of drug and its MoA will help determine the assay format and technical platform for NAb assessment (e.g., cell-based or non-cell-based assay).
    [Show full text]
  • Assay Qualification Recall…
    Assay Qualification Recall… Assay Qualification: • ACCURACY: Orthogonal method • PRECISION: Reproducibility: same day replicates, day to day, different operators • ROBUSTNESS: sensitivity to assay parameters • SPECIFICITY: sensitivity to matrix effects • DYNAMIC RANGE AND RESPONSE FUNCTION: Instrument benchmarking. +/- controls. Calibration curve. Limit of detection Goal Establish confidence in genomic assays used to identify on-target and off- target genome editing genomic locations and sequence variants. Simplified Genome Editing Process Design & Execute Source sample Screen for genome disruption Target Editing molecule Targeted Confirm sequence change Editing molecule sequence (e.g. T7 Endonuclease, Surveyor, HRMA, TIDE) Targeted (sequencing, qPCR) Editing molecule formulation Genome Wide Whole exome sequencing Delivery (e.g. GUIDE-Seq, Digenome-Seq, Whole genome sequencing CIRCLE-Seq, SITE-Seq, BLESS/BLISS) Assay Reference qualification Materials Activities and outputs: 1. Qualify genomic assays used to identify on-target and off-target genome editing genomic locations and sequence variants. 2. Develop & qualify reference materials – designed to mimic genome edited DNA complexity for use in benchmarking sequence verification assay limits of detection and sensitivity. Assay Qualification Process Assess assay precision (initially within NIST laboratories) Assess sources of variability and improve assay protocols and robustness (DoE) (NIST) Identify assay(s) of primary importance (community consensus) Refined assay protocols will be evaluated
    [Show full text]
  • Directed Evolution of a G Protein-Coupled Receptor for Expression, Stability, and Binding Selectivity
    Directed evolution of a G protein-coupled receptor for expression, stability, and binding selectivity Casim A. Sarkar*†‡, Igor Dodevski*†, Manca Kenig*§, Stefan Dudli*, Anja Mohr*, Emmanuel Hermans¶, and Andreas Plu¨ckthun*ʈ *Biochemisches Institut, Universita¨t Zu¨ rich, Winterthurerstrasse 190, CH-8057 Zu¨rich, Switzerland; and ¶Laboratoire de Pharmacologie Expe´rimentale, Universite´catholique de Louvain, Avenue Hippocrate 54.10, B-1200 Bruxelles, Belgium. Edited by William F. DeGrado, University of Pennsylvania, Philadelphia, PA, and approved July 18, 2008 (received for review April 1, 2008) We outline a powerful method for the directed evolution of sequence of a GPCR, keeping all other variables constant, to integral membrane proteins in the inner membrane of Escherichia yield more functionally expressed protein in a convenient het- coli. For a mammalian G protein-coupled receptor, we arrived at a erologous host, Escherichia coli. We used as a model system the sequence with an order-of-magnitude increase in functional ex- rat neurotensin receptor-1 (NTR1), which has been shown to pression that still retains the biochemical properties of wild type. give a detectable yield in E. coli (10, 11) but which still needs to This mutant also shows enhanced heterologous expression in be improved to allow more convenient preparation of milligram eukaryotes (12-fold in Pichia pastoris and 3-fold in HEK293T cells) quantities of receptor. and greater stability when solubilized and purified, indicating that Detailed characterization of the best variant from the selec- the biophysical properties of the protein had been under the tion reported here reveals that it exhibits an order-of-magnitude pressure of selection.
    [Show full text]