Harmonizing Biologics: Navigating Immunogenicity in Drug Development

The introduction of gene therapies and therapeutic proteins has marked a revolutionary step in combating disease. However, these advanced treatments must navigate the challenging landscape of immunogenicity, where the body’s defense system reacts to the introduced medication, potentially negating its therapeutic purpose or worse, reacting against the body’s own proteins. Early in the biologic development process, it’s critical to mitigate such immune reactions to ensure the treatment’s integrity and safety1.

The emergence of anti-drug antibodies (ADAs) in response to biologic medications poses clinical obstacles, from reduced drug effectiveness to serious safety complications. These ADAs have the potential to neutralize drug effects, alter pharmacokinetics (PK), and provoke autoimmune-like responses2. Drug developers are therefore tasked with predicting and curbing the formation of ADAs, crafting therapies that are both efficacious and safe3.

Global regulatory authorities, including the Food and Drug Administration (FDA) and the European Medicines Agency (EMA), mandate a thorough immunogenicity risk assessment before biologics can proceed to clinical trials and market introduction4. This rigorous evaluation process is essential for patient safety, aiding developers to make informed choices that bolster the success and acceptance of new biologic treatments. Comprehensive immunogenicity testing is thus a cornerstone in the journey towards safely integrating pioneering therapies into healthcare.

Advancements in Computational and Laboratory Techniques in Immunogenicity

For effective drug development, computational methods known as in silico tools are indispensable for forecasting the immunogenic risk. These enable the detection of peptide sequences that might bind to critical immune system proteins, thus predicting potential immune triggers5. The Immune Epitope Database is one such resource that assists in screening numerous variants efficiently6.

However, in silico platforms can be overly predictive, leading to superfluous modifications. Therefore, in vitro assays become crucial, offering validation through methods like peptide mapping. These laboratory techniques are particularly beneficial in later stages of drug candidate selection, providing experimental data to support computational findings7.

The synergy of in silico and in vitro approaches forms a comprehensive strategy for assessing immunogenic risk, vital for crafting safe and efficacious therapeutic proteins. Following up with in vitro assays then provides experimental data with human immune cells that helps refine interpretation of the in silico data8. This dual strategy not only increases confidence in the selection process but also plays a significant role in the regulatory approval for new drugs and an important component of the regulatory approval pathway9.

Leveraging Computational Strategies for Antibody Development

The landscape of drug development has been significantly altered by innovative strategies to manage immunogenicity. A key development was the transition from murine-derived antibodies to those that are fully human, particularly through the process of humanization first developed in the 1980s, which reduced the immune system’s response to these therapeutic agents10. Advances in epitope prediction algorithms have further enabled the tailoring of antibodies to diminish immunogenic potential, a step validated by reductions in T-cell epitope scores and in vitro testing. This journey has underscored the importance of proactive design in antibody development, where computational tools are used early to identify and modify potentially immunogenic regions, thus streamlining the development process and minimizing redesign of biologics at later-stages of the development process 11,12.

Pioneering Developmental Strategies for Drug Design

The concept of ‘design for success’ encapsulates the philosophy of early design and developability in drug creation. This comprehensive approach involves evaluating a drug’s therapeutic potential, safety, specificity, and manufacturability. Such assessments, fundamental in the nascent stages of development, guide the selection of candidates that not only meet therapeutic goals but are also feasible for producing at scale13. Abzena’s EpiScreen® platform is a key component of such an approach which assesses immunogenicity using a bioinformatics platform and suit of in vitro assays, in order to deliver better, more granular data that aids in the candidate selection process. This strategic analysis is key to optimizing resource allocation, enhancing the likelihood of clinical and market success, and fostering a streamlined, targeted approach to drug development.

Innovations in Assessing Immunogenicity

Recent years have witnessed a burgeoning of novel techniques aimed at tackling the intricacies of immunogenicity in protein therapy research. Notably, the advent of 3D organoid models replicating lymph nodes, along with synthetic skin and subcutaneous tissue constructs, signifies a transformative step in decoding immune reactions13,14. These emerging tools are integrating into holistic strategies to enhance predictions of bodily reactions to novel treatments, thereby refining the accuracy of immunogenicity evaluations15.

Continual research is reshaping our approach to immunogenicity in biologic treatments. By probing the depths of molecular and cellular foundations of immune reactions, we’re enhancing our predictive capabilities to prevent undesirable immunogenicity. The focus is on crafting refined in silico instruments that assess immunogenic regions with heightened precision and developing in vitro assays that replicate the human immune system’s complexity with remarkable exactness. The goal is to perfect these models to provide clear guidance on immunogenic risks, promoting the development of safer and more efficacious therapeutic proteins16.

Emerging technologies hold remarkable promise for revolutionizing the development of therapeutic proteins. They equip researchers with the means to detect and address immunogenicity risks early on, paving the way for safer and more effective biologic treatments. These advancements not only refine the safety profiles of upcoming therapies but also streamline the design and optimization process15. By facilitating the early pinpointing and modification of immunogenic elements, they promise to curtail both the time and expense involved in introducing new treatments to the market17.

Harmonizing Biologic Development and Regulation

The landscape of drug development is increasingly shaped by a cooperative approach between the biopharmaceutical industry and regulatory bodies, particularly in the crucial area of immunogenicity management18. This strategic alliance goes beyond shared objectives to forge practices that boost the effectiveness and safety of biologic therapies. Regulatory standards evolve in tandem with industry advancements in predictive technologies, ensuring a rigorous evaluation of drugs and fostering ongoing innovation. This joint endeavour reflects a deepening insight into biologic-immune interactions, prioritizing patient welfare and therapeutic efficacy19.

The fusion of industrial innovation with regulatory oversight has created a cohesive strategy for the future of biologic therapy development. This partnership is dedicated to formulating strong, scientifically based practices that surpass individual organizational objectives, focusing instead on the collective goal of ensuring the safety and efficacy of biologic medicines. Contract research organizations (CROs), like Abzena, can help support biopharma companies by employing these types of assessments earlier in the drug development process to ensure better downstream clinical outcomes. Such unified efforts equip the biopharmaceutical industry to better understand and address immunogenicity, thereby enhancing patient outcomes and fostering greater confidence in biologic treatments20.

In essence, this joint trajectory goes beyond merely aligning the objectives of the biopharmaceutical industry with regulatory standards; it is an embodiment of a unified commitment to enhance healthcare. This collaborative venture is crucial to ensuring that innovative treatments are not only secure for patient use but also effective in tackling diseases.

References

  1. Schellekens H. The immunogenicity of therapeutic proteins. Discov Med. 2010 Jun;9(49):560-4.
  2. Pizano-Martinez O, Mendieta-Condado E, Vázquez-Del Mercado M, Martínez-García EA, Chavarria-Avila E, Ortuño-Sahagún D, Márquez-Aguirre AL. Anti-Drug Antibodies in the Biological Therapy of Autoimmune Rheumatic Diseases. J Clin Med. 2023 May 4;12(9):3271.
  3. Sathish JG, Sethu S, Bielsky MC, de Haan L, French NS, Govindappa K, Green J, Griffiths CE, Holgate S, Jones D, Kimber I, Moggs J, Naisbitt DJ, Pirmohamed M, Reichmann G, Sims J, Subramanyam M, Todd MD, Van Der Laan JW, Weaver RJ, Park BK. Challenges and approaches for the development of safer immunomodulatory biologics. Nat Rev Drug Discov. 2013 Apr;12(4):306-24
  4. Vandivort TC, Horton DB, Johnson SB. Regulatory and strategic considerations for addressing immunogenicity and related responses in biopharmaceutical development programs. J Clin Transl Sci. 2020 Jun 15;4(6):547-555.
  5. Garcia KC, Adams EJ. How the T cell receptor sees antigen–a structural view. Cell. 2005 Aug 12;122(3):333-6.
  6. Vita R, Overton JA, Greenbaum JA, Ponomarenko J, Clark JD, Cantrell JR, Wheeler DK, Gabbard JL, Hix D, Sette A, Peters B. The immune epitope database (IEDB) 3.0. Nucleic Acids Res. 2015 Jan;43.
  7. Joubert MK, Deshpande M, Yang J, Reynolds H, Bryson C, Fogg M, Baker MP, Herskovitz J, Goletz TJ, Zhou L, Moxness M, Flynn GC, Narhi LO, Jawa V. Use of In Vitro Assays to Assess Immunogenicity Risk of Antibody-Based Biotherapeutics. PLoS One. 2016 Aug 5;11(8):e0159328.
  8. Cohen S, Chung S, Spiess C, Lundin V, Stefanich E, Laing ST, Clark V, Brumm J, Zhou Y, Huang C, Guerrero J, Myneni S, Yadav R, Siradze K, Peng K. An integrated approach for characterizing immunogenic responses toward a bispecific antibody. MAbs. 2021 Jan-Dec;13(1):1944017.
  9. U.S. Food and Drug Administration. Investigational New Drug (IND) Application. https://www.fda.gov/drugs/types-applications/investigational-new-drug-ind-application
  10. Gorman SD, Clark MR. Humanisation of monoclonal antibodies for therapy. Semin Immunol. 1990 Nov;2(6):457-66. Zinsli LV, Stierlin N, Loessner MJ, Schmelcher M. Deimmunization of protein therapeutics – Recent advances in experimental and computational epitope prediction and deletion. Comput Struct Biotechnol J. 2020 Dec 29;19:315-329.
  11. Zhang W, Wang H, Feng N, Li Y, Gu J, Wang Z. Developability assessment at early-stage discovery to enable development of antibody-derived therapeutics. Antib Ther. 2022 Nov 11;6(1):13-29.
  12. Jawa V, Terry F, Gokemeijer J, Mitra-Kaushik S, Roberts BJ, Tourdot S, De Groot AS. T-Cell Dependent Immunogenicity of Protein Therapeutics Pre-clinical Assessment and Mitigation-Updated Consensus and Review 2020. Front Immunol. 2020 Jun 30;11:1301.
  13. Yufeng Shou, Sarah C. Johnson, Ying Jie Quek, Xianlei Li, Andy Tay. Integrative lymph node-mimicking models created with biomaterials and computational tools to study the immune system. Materials Today Bio. Volume 14, 2022.
  14. Bédard P, Gauvin S, Ferland K, Caneparo C, Pellerin È, Chabaud S, Bolduc S. Innovative Human Three-Dimensional Tissue-Engineered Models as an Alternative to Animal Testing. Bioengineering (Basel). 2020 Sep 17;7(3):115.
  15. Sauna ZE, Jawa V, Balu-Iyer S, Chirmule N. Understanding preclinical and clinical immunogenicity risks in novel biotherapeutics development. Front Immunol. 2023 May 12;14:1151888.
  16. Gokemeijer J, Jawa V, Mitra-Kaushik S. How Close Are We to Profiling Immunogenicity Risk Using In Silico Algorithms and In Vitro Methods?: an Industry Perspective. AAPS J. 2017 Nov;19(6):1587-1592.
  17. Bauer J, Rajagopal N, Gupta P, Gupta P, Nixon AE, Kumar S. How can we discover developable antibody-based biotherapeutics? Front Mol Biosci. 2023 Aug 7;10:1221626.
  18. Ducret A, Ackaert C, Bessa J, Bunce C, Hickling T, Jawa V, Kroenke MA, Lamberth K, Manin A, Penny HL, Smith N, Terszowski G, Tourdot S, Spindeldreher S. Assay format diversity in pre-clinical immunogenicity risk assessment: Toward a possible harmonization of antigenicity assays. MAbs. 2022 Jan-Dec;14(1):1993522.
  19. Chang RB, Beatty GL. The interplay between innate and adaptive immunity in cancer shapes the productivity of cancer immunosurveillance. J Leukoc Biol. 2020 Jul;108(1):363-376.
  20. Wadhwa M, Thorpe R. Harmonization and standardization of immunogenicity assessment of biotherapeutic products. Bioanalysis. 2019 Sep;11(17):1593-1604.

Author
Author Image
Rob Holgate
Abzena
VP of Research & Innovation.
Abzena
Company Logo