Immunogenicity is critical for measuring the potential for immune responses to ensure safe and effective therapies. As the field evolves and new approaches emerge, sponsors have a range of immunogenic considerations for therapeutic proteins and associated analytical methodologies for detecting and characterising anti-drug antibodies (ADAs). Here we share some industry best practices for achieving high-quality ADA assays and explore the emerging trends including the advantages and limitations of singlicate ADA analysis compared with alternative techniques.
How immunogenicity impacts the development and efficacy of therapeutic proteins
Understanding the propensity of a therapeutic protein to generate an immune response is important for several reasons. Dosing with a biotherapeutic can stimulate an immune response to the product itself or to proteins with a related structure. The immune response can potentially impact the efficacy of the therapeutic and when endogenous proteins with a related structure are targeted by ADAs, that presents a significant safety risk for the patient.
Therapeutic efficacy can be impacted via reductions in the amount of the drug in circulation or reduction of the ability of a therapeutic to bind to its target, which is referred to as a neutralising antibody or a NAb. A variety of factors – such as formulation, protein modifications and structure, target, route of administration and dose – all have the potential to impact the immunogenicity of a therapeutic and this should be carefully considered during development.
Over the years, regulations have been implemented to monitor immunogenicity as its importance is better understood. The field is still rapidly evolving and discussions on new approaches continue with the goal of monitoring effectively while also being pragmatic and efficient.
Selecting the right approach to immunogenicity
The field of immunogenicity is very broad in terms of drug modalities and analytical techniques, presenting unique challenges and opportunities in selecting the best approach. Different therapeutics each present their own limitations when choosing an assay format, determining assessments to include in validation or determining assay tiers to include. There are also a variety of analytical approaches to choose from to address a sponsor’s needs. For example, there are different platforms, assay formats, types of automation and different techniques at the bench that may better suit each study. The qualitative nature of immunogenicity assays also means that there are different opinions across the industry regarding the analysis and interpretation of data and the ideal approach for meeting regulatory requirements.
At ICON, we frequently see slight variations in the preferred statistical analysis approach for the determination of assay cut points among our sponsors. Experiments in criteria are not as clear as for pharmacokinetic (PK) assays regulated by ICH M10 harmonised guidance. These different aspects can often be challenging, but they also offer us the opportunity for the continued development of our own scientific knowledge and expertise, and for collaborative engagement with our sponsors.
Anti-drug antibodies and current analytical methodologies
There are multiple approaches to detecting and characterizing anti-drug antibodies (ADAs). Most often we use electrochemiluminescence assays (ECLA), typically with the MSD platform. We also use enzyme-linked immunosorbent assays (ELISAs), and occasionally enzyme activity assays. We also have experience using flow cytometry for readout of ADAs and mass spectrometry for IgG subclass typing of immune responses.
There is a variety of different method formats, including simple bridging assay to more drug-tolerant formats. These include:
- affinity capture elution (ACE)
- solid phase extraction with acid dissociation (SPEAD)
- bead extraction and acid dissociation (BEAD)
- precipitation and acid dissociation (PandA)
- high ionic strength dissociation assays (HISDA)
The most appropriate format is selected based on considerations such as the therapeutic, its target and the level of drug tolerance that’s needed.
Best practices for ensuring high-quality ADA detection and characterisation
The industry standard for the analysis of clinical samples is a three-tiered approach: screening, confirmatory and titration. Additional tiers should be considered where domain characterisation is necessary for things like multi-domain therapeutics such as bispecific antibodies or antibody-drug conjugates (ADCs) or the addition of antibody isotyping assays where there are relevant clinical concerns. It is also important to bring awareness and consideration of the required assay parameters to the fore at earlier stages, ensuring the assay can reliably tolerate ADAs in the presence of the therapeutic, its target, and/or other co-dosed medications in the presence of matrix components.
Careful selection of controls and cut-points
A good surrogate positive control is important for assessing and monitoring assay parameters, as is having either sufficient volume or reproducibility for long-term support of an assay. Monoclonal and polyclonal controls both have their own advantages and limitations. It can be challenging to generate a positive control, especially against some therapeutics such as peptides. The negative control pool should be representative of the samples used to establish cut-points. In selecting these cut-points, it is important to apply the appropriate statistical technique to establish meaningful and appropriate cut-points to obtain meaningful assay results.
This is inclusive of appropriate removal of outliers. Typically, data are assessed for normality followed by selection of a parametric or nonparametric cut-point calculation. Although, there is new interest in the industry around alternative approaches such as the application of random effects modeling. Consideration of the need for disease state population cut-points is also of importance, as is monitoring in-study false positive rates as an indication that in-study cut-points are needed. And finally, determining if a cell-based or a plate-based assay is most appropriate to monitor for the neutralising effects of ADAs should consider the therapeutic’s mechanism of action and assay parameters that can be achieved.
Evolving industry standards
ICON scientists and managers stay connected to current opinions in industry via attendance at conferences and meetings, participation in publications and conversations with our sponsors. Our three bioanalytical labs in Lenexa, Kansas; Assen, Netherlands; and Whitesboro, New York afford us insight in local environments and industry landscapes globally. Additionally, with approximately 500 bioanalytical staff members, ICON has the added benefit of a large team of experts and the exchange of information among our sites, ensuring that we are always abreast of changes as they evolve.
Advantages and limitations of singlicate ADA analysis
Singlicate ADA analysis is a topic of interest in industry. There are a few obvious advantages to single-well analysis:
- the ability to analyse twice as many samples per
- higher sample throughput
- reduction in utilisation of potentially costly reagents, materials and labor
From a regulatory perspective, there’s not an explicit requirement for duplicate-well analysis, provided an assay has very good duplicate precision, as many modern ligand binding assays do, particularly on platforms such as MSD. Therefore, moving to single-well analysis should theoretically have minimal impact.
Furthermore, ADA samples typically undergo multiple analyses within the three-tier approach, so a switch to a single-well analysis for each tier would likely have minimal impact. However, due in part to the qualitative nature of immunogenicity assays, there’s still ongoing discussion in the industry around how best to employ single-well analysis in assay validation and its impact on the statistical determination of parameters such as assay cut-point and 1% failure rate LPC (low positive control) setting.
Questions driving industry discussion on the topic of singlicate ADA analysis include: Should the validation be performed in duplicate wells with the results from duplicate- and single-wells compared to support the use of single-well analysis for samples? Or can the validation be performed in single wells without such a comparison? What are the impacts to variability in cut-point assessments? There’s certainly evidence in support of the use of single-well analysis for ADA. But at this point, it’s still a relatively new approach that I’m sure will become better characterised as it’s more widely used. Currently, we see more uptake with single-well analysis for PK.
Emerging trends in bioanalysis of immunogenicity
As biotherapeutics evolve, so must the methods we use to assess immunogenicity. ICON has invested in examining the best analytical approach for therapeutics such as gene therapies, which are increasingly common. In addition to classic ligand binding assays, techniques such as cell-based assays with flow cytometry readout for immunogenicity assessments for cell-based therapeutics, or hybrid LBA/LC-MS assays for IgG subclass-typing to further characterise immune responses can be advantageous.
As our understanding of immunogenicity continues to evolve, we can expect an industry-wide movement toward a more risk-based approach informed by upfront assessment of therapeutics and an improved understanding of the safety implications of an immunogenic response. This will allow for a more pragmatic and focused approach to assay design and validation.
Partner with ICON for the highest levels of scientific expertise and insight into the latest innovations and therapeutic advances for your clinical trial. Find out more about our bioanalytical capabilities.
In this section
-
Digital Disruption
-
Clinical strategies to optimise SaMD for treating mental health
-
Digital Disruption: Surveying the industry's evolving landscape
- AI and clinical trials
-
Clinical trial data anonymisation and data sharing
-
Clinical Trial Tokenisation
-
Closing the evidence gap: The value of digital health technologies in supporting drug reimbursement decisions
-
Digital disruption in biopharma
-
Disruptive Innovation
- Remote Patient Monitoring
-
Personalising Digital Health
- Real World Data
-
The triad of trust: Navigating real-world healthcare data integration
-
Clinical strategies to optimise SaMD for treating mental health
-
Patient Centricity
-
Agile Clinical Monitoring
-
Capturing the voice of the patient in clinical trials
-
Charting the Managed Access Program Landscape
-
Developing Nurse-Centric Medical Communications
- Diversity and inclusion in clinical trials
-
Exploring the patient perspective from different angles
-
Patient safety and pharmacovigilance
-
A guide to safety data migrations
-
Taking safety reporting to the next level with automation
-
Outsourced Pharmacovigilance Affiliate Solution
-
The evolution of the Pharmacovigilance System Master File: Benefits, challenges, and opportunities
-
Sponsor and CRO pharmacovigilance and safety alliances
-
Understanding the Periodic Benefit-Risk Evaluation Report
-
A guide to safety data migrations
-
Patient voice survey
-
Patient Voice Survey - Decentralised and Hybrid Trials
-
Reimagining Patient-Centricity with the Internet of Medical Things (IoMT)
-
Using longitudinal qualitative research to capture the patient voice
-
Agile Clinical Monitoring
-
Regulatory Intelligence
-
An innovative approach to rare disease clinical development
- EU Clinical Trials Regulation
-
Using innovative tools and lean writing processes to accelerate regulatory document writing
-
Current overview of data sharing within clinical trial transparency
-
Global Agency Meetings: A collaborative approach to drug development
-
Keeping the end in mind: key considerations for creating plain language summaries
-
Navigating orphan drug development from early phase to marketing authorisation
-
Procedural and regulatory know-how for China biotechs in the EU
-
RACE for Children Act
-
Early engagement and regulatory considerations for biotech
-
Regulatory Intelligence Newsletter
-
Requirements & strategy considerations within clinical trial transparency
-
Spotlight on regulatory reforms in China
-
Demystifying EU CTR, MDR and IVDR
-
Transfer of marketing authorisation
-
An innovative approach to rare disease clinical development
-
Therapeutics insights
- Endocrine and Metabolic Disorders
- Cardiovascular
- Cell and Gene Therapies
-
Central Nervous System
-
A mind for digital therapeutics
-
Challenges and opportunities in traumatic brain injury clinical trials
-
Challenges and opportunities in Parkinson’s Disease clinical trials
-
Early, precise and efficient; the methods and technologies advancing Alzheimer’s and Parkinson’s R&D
-
Key Considerations in Chronic Pain Clinical Trials
-
A mind for digital therapeutics
-
Glycomics
- Infectious Diseases
- NASH
- Oncology
- Paediatrics
-
Respiratory
-
Rare and orphan diseases
-
Advanced therapies for rare diseases
-
Cross-border enrollment of rare disease patients
-
Crossing the finish line: Why effective participation support strategy is critical to trial efficiency and success in rare diseases
-
Diversity, equity and inclusion in rare disease clinical trials
-
Identify and mitigate risks to rare disease clinical programmes
-
Leveraging historical data for use in rare disease trials
-
Natural history studies to improve drug development in rare diseases
-
Patient Centricity in Orphan Drug Development
-
The key to remarkable rare disease registries
-
Therapeutic spotlight: Precision medicine considerations in rare diseases
-
Advanced therapies for rare diseases
-
Transforming Trials
-
Accelerating biotech innovation from discovery to commercialisation
-
Ensuring the validity of clinical outcomes assessment (COA) data: The value of rater training
-
Linguistic validation of Clinical Outcomes Assessments
-
Optimising biotech funding
- Adaptive clinical trials
-
Best practices to increase engagement with medical and scientific poster content
-
Decentralised clinical trials
-
Biopharma perspective: the promise of decentralised models and diversity in clinical trials
-
Decentralised and Hybrid clinical trials
-
Practical considerations in transitioning to hybrid or decentralised clinical trials
-
Navigating the regulatory labyrinth of technology in decentralised clinical trials
-
Biopharma perspective: the promise of decentralised models and diversity in clinical trials
-
eCOA implementation
- Blended solutions insights
-
Implications of COVID-19 on statistical design and analyses of clinical studies
-
Improving pharma R&D efficiency
-
Increasing Complexity and Declining ROI in Drug Development
-
Innovation in Clinical Trial Methodologies
- Partnership insights
-
Risk Based Quality Management
-
Transforming the R&D Model to Sustain Growth
-
Accelerating biotech innovation from discovery to commercialisation
-
Value Based Healthcare
-
Strategies for commercialising oncology treatments for young adults
-
US payers and PROs
-
Accelerated early clinical manufacturing
-
Cardiovascular Medical Devices
-
CMS Part D Price Negotiations: Is your drug on the list?
-
COVID-19 navigating global market access
-
Ensuring scientific rigor in external control arms
-
Evidence Synthesis: A solution to sparse evidence, heterogeneous studies, and disconnected networks
-
Global Outcomes Benchmarking
-
Health technology assessment
-
Perspectives from US payers
-
ICER’s impact on payer decision making
-
Making Sense of the Biosimilars Market
-
Medical communications in early phase product development
-
Navigating the Challenges and Opportunities of Value Based Healthcare
-
Payer Reliance on ICER and Perceptions on Value Based Pricing
-
Payers Perspectives on Digital Therapeutics
-
Precision Medicine
-
RWE Generation Cross Sectional Studies and Medical Chart Review
-
Survey results: How to engage healthcare decision-makers
-
The affordability hurdle for gene therapies
-
The Role of ICER as an HTA Organisation
-
Strategies for commercialising oncology treatments for young adults
-
Blog
-
Videos
-
Webinar Channel