![Figure illustrating the context and nature of biosimilar research and development in the healthcare industry.](https://static.wixstatic.com/media/6d4460_6fe3804a546b407eb414414b94a6580d~mv2.jpg/v1/fill/w_980,h_560,al_c,q_85,usm_0.66_1.00_0.01,enc_auto/6d4460_6fe3804a546b407eb414414b94a6580d~mv2.jpg)
INTRODUCTION:
In the constantly evolving world of medicine and pharmacology, the emergence of biosimilars represents a significant revolution, offering new avenues for access to treatments and therapeutic innovation. These drugs, similar but not identical to their biological reference products, are at the heart of a rapidly expanding field, marked by unique scientific, regulatory, and commercial challenges. The importance of research and development (R&D) in this sector cannot be understated, as it shapes the future of treating many diseases, including the rarest and most difficult to treat.
Biosimilars promise to make innovative biological therapies more accessible and affordable for patients worldwide.
However, their development, regulatory approval, and commercialization are fraught with complexities. These drugs must demonstrate their biosimilarity to the reference products through rigorous processes, including analytical, preclinical, and clinical studies. Moreover, each region - Europe, the United Kingdom, the United States, Canada, and India - follows its own regulatory guidelines, creating a complex landscape for companies engaged in the development of biosimilars.
Through this article, we aim to provide a comprehensive and informed overview of biosimilars, demonstrating their transformative potential for the global health system and for improving the quality of life of patients around the world.
1.OVERVIEW OF BIOSIMILAR DEVELOPMENT
1.1 DEFINITION AND CHARACTERISTICS
Biomedicine
A biological drug, also known as "biomedicine" or "biological product," is a type of medication obtained from biological sources or by their synthesis. Unlike traditional drugs, generally manufactured through chemical synthesis, biological drugs are produced by living organisms such as animal cells, bacteria, or yeasts. Here are some key characteristics of biological drugs:
Biological source:
Biological drugs are derived from biological sources, including a wide range of products such as proteins (including antibodies), nucleotides (DNA, RNA), substances produced by genetic engineering, and vaccines.
Complexity:
They are generally more complex and larger than traditional chemical drugs, due to their larger molecular structure and complex manufacturing method.
Manufacturing:
The manufacturing process of biological drugs involves living organisms, requiring sterile culture conditions and advanced biotechnological processes.
Variability:
There can be some variability in the production batches of biological drugs due to their biological origin. Manufacturers must therefore follow strict procedures to ensure product consistency and quality.
Use:
They are used to treat a wide range of diseases and conditions, including cancers, autoimmune diseases like psoriatic arthritis and multiple sclerosis, endocrine disorders like diabetes, and many others.
Regulation:
Biological drugs are subject to stricter regulation and approval requirements by health authorities due to their complexity and specific manufacturing method[1], [2].
Biosimilars
Biosimilars are biological drugs highly similar to an already approved reference biological drug, with no clinically significant differences in terms of purity, potency, and safety. Unlike generic drugs, which are exact copies of chemical drugs, biosimilars are developed from living cells and may present slight variations from their reference product, due to the inherent complexity of their biological manufacturing. These drugs can cover a wide range of treatments, including monoclonal antibodies, growth factors, and hormones, used in the treatment of serious diseases like cancer, autoimmune diseases, and hematological disorders[3], [4], [5], [6].
![figure showing the general principles of biosimilar r&d](https://static.wixstatic.com/media/6d4460_29872a9ee5e14fb68bf2a47c6c222ce7~mv2.jpg/v1/fill/w_980,h_980,al_c,q_85,usm_0.66_1.00_0.01,enc_auto/6d4460_29872a9ee5e14fb68bf2a47c6c222ce7~mv2.jpg)
1.2. BIOSIMILARS RESEARCH AND INNOVATION ASPECT
Intellectual Property (IP) and Development Strategies for Biosimilars
Patent Expirations:
The opportunity to develop biosimilars generally arises when the patents for reference biological products expire. The IP landscape for biologics is complex, potentially including multiple patents covering the product, its manufacturing process, and therapeutic uses[7], [8].
Patent Litigations:
Manufacturers of reference biological products may engage in litigation to protect their patents and market exclusivities, which can delay the market entry of biosimilars. Resolving these litigations is often a crucial aspect of the development strategy for biosimilars[7], [8].
Dedicated Regulatory Pathway:
Many countries have established dedicated regulatory pathways for the approval of biosimilars, requiring the demonstration of similarity to the reference product without infringing on patents, involving a deep understanding of the IP surrounding the reference product[9], [10].
Development Strategy Aspect
Demonstration of Similarity:
The development of biosimilars involves analytical, preclinical, and clinical studies to demonstrate that the biosimilar is highly similar to the reference product in terms of efficacy, safety, and quality, despite the natural variations inherent in biological production[6], [10], [11], [12].
Development Costs:
Although less expensive than the development of original biologics, it requires significant investments due to regulatory requirements to prove similarity. Efficient development strategies are adopted to minimize costs while meeting regulatory standards[13], [14], [15].
Market Positioning:
Marketing and market positioning strategies are crucial for the success of biosimilars, including competitive pricing, education programs for healthcare professionals and patients, and negotiations with payers for reimbursement [13], [14], [15].
Continuous Innovation:
Although similar by definition to original products, investments in innovation are made, for example, by improving formulations or delivery devices, to differentiate in the market [13], [14], [15].
1.3 IMPORTANCE OF BIOSIMILARS R&D
Biosimilars R&D plays a crucial role in optimizing access to biological treatments, offering less expensive alternatives to innovative biologics. This allows for significant cost reductions for healthcare systems and better accessibility for patients. Biosimilars R&D requires substantial investment in time and resources but is essential to ensure therapeutic equivalence with the reference product and to obtain regulatory approval[11], [12], [13], [14], [15].
![Figure showing the strategic aspects and impact of biosimilars R&D](https://static.wixstatic.com/media/6d4460_d69d5f17161a4c8eb4b5d850b80d708a~mv2.jpg/v1/fill/w_980,h_980,al_c,q_85,usm_0.66_1.00_0.01,enc_auto/6d4460_d69d5f17161a4c8eb4b5d850b80d708a~mv2.jpg)
2. GENERAL PHASES OF BIOSIMILAR DEVELOPMENT
The development of a biosimilar is a complex and layered process, breaking down into several key phases:
Research and Positioning:
Identification of a reference biological product and market needs assessment, involving a thorough analysis of patents, a rigorous candidate selection, and strategic positioning.
Analytical Development:
Comprehensive characterization of the biosimilar and the reference product to establish biosimilarity, including an exhaustive comparison in terms of structure, function, and purity.
Preclinical Trials:
In vitro and in vivo studies to assess toxicity, pharmacokinetics, and pharmacodynamics, aiming to prove safety and biological activity.
Clinical Trials:
Conducting clinical trials to demonstrate the efficacy, safety, and immunogenicity of the biosimilar compared to the reference product, including planning clinical trials with relevant patient populations and clinical endpoints.
Regulatory Approach:
Submission of a complete dossier to regulatory authorities, including analytical, preclinical, and clinical data, to demonstrate biosimilarity and develop the regulatory dossier.
Manufacturing:
Establishment of manufacturing processes in compliance with Good Manufacturing Practices (GMP), ensuring the quality, safety, and efficacy of the biosimilar.
The proof of similarity relies on a set of comparative data, including analytical, preclinical, and clinical studies, demonstrating that minor variations do not affect the efficacy or safety of the biosimilar. Post-marketing surveillance is also essential for monitoring long-term safety and efficacy. [3], [5], [9], [10], [11], [12], [14], [15]
3. REGULATORY FRAMEWORK FOR BIOSIMILARS – EUROPE
The commercialization and use of biosimilars go through a labyrinth of regulations that vary significantly from one region to another. Each jurisdiction has its own guidelines and regulatory requirements, designed to ensure the safety, efficacy, and quality of biosimilars. These regulatory frameworks are essential not only for protecting public health but also for fostering innovation and accessibility of biological treatments. Understanding these different regulatory systems is crucial for biosimilar developers to navigate the approval process and optimize their market entry strategy.
Europe is often considered a pioneer in establishing a regulatory framework for biosimilars, thanks to the European Medicines Agency (EMA) with the implementation of detailed guidelines that have since evolved.
The approval of biosimilars is based on a robust structural and functional comparability assessment of the proposed biosimilar compared to the reference biological product. The gathered data, known as the totality of evidence, demonstrates the biosimilarity between the proposed biosimilar and its reference biological product in terms of quality, safety, and efficacy. Therefore, physicians and patients can expect the same clinical outcome.
![Figure showing the general R&I processes for biosimilars](https://static.wixstatic.com/media/6d4460_61a9636dc8ea427eacc689dad6badce7~mv2.jpg/v1/fill/w_980,h_980,al_c,q_85,usm_0.66_1.00_0.01,enc_auto/6d4460_61a9636dc8ea427eacc689dad6badce7~mv2.jpg)
Here's a detailed exploration of the regulatory framework's various aspects, adhering to the standardized structure requested:
3.1. QUALITY STUDIES - MOLECULAR SIMILARITY AND STRUCTURAL ANALYSES:
In-depth structural and functional analyses are required to demonstrate the molecular similarity between the biosimilar and its reference product. This includes characterizing purity, primary structure, secondary and tertiary structure conformation, post-translational modifications, and isoelectric focusing, among others[10], [16], [17].
Primary Structure and Conformation:
The biosimilar's nearly identical amino acid sequence must be characterized by sequencing (mass spectrometry or capillary electrophoresis). Any minor differences from the reference product must be justified.
Secondary and Tertiary Structure:
Techniques such as nuclear magnetic resonance (NMR), fluorescence spectroscopy, and X-ray diffraction are used. Tolerance windows for these structures are generally not quantified publicly, but profiles must be highly similar.
Post-Translational Modifications:
Glycosylation, in particular, must be similar, although minor acceptable variations are recognized. Similarity is assessed by comparative profiles using X-ray crystallography for three-dimensional structure, and chromatography for purity and product variants. There's no specific tolerance window, but a very similar profile is expected.
Purity and Impurities:
Analyzing purity and impurities, including process variants like deamidated or oxidized forms, is crucial for ensuring the biosimilar's quality and safety. High-performance liquid chromatography (HPLC) and mass spectrometry are commonly used techniques for these analyses. Profiles should be similar, with process-specific impurities well characterized and controlled.
Tolerance windows and exact criteria for acceptance are evaluated in the context of submitted data, the biological product's characteristics, and the specific requirements of the therapeutic indication on a case-by-case basis, based on the clinical relevance of observed differences. Structural analyses must demonstrate molecular similarity with a tolerance window defined by direct comparison with the reference product. This includes >90% similarity for primary characteristics, like amino acid sequence, and stricter criteria for secondary and tertiary characteristics. Variations must be justified by functional and clinical data showing no impact on efficacy and safety [5], [10]
3.2. QUALITY - MANUFACTURING CERTIFICATE:
The Good Manufacturing Practice (GMP) certificate defined by the European Commission is issued after inspection and verification that production facilities meet European quality standards. Criteria include compliance with standardized manufacturing procedures, material traceability, and the assurance of finished product quality[3], [5], [10], [18], [19].
3.3. PRECLINICAL STUDIES – SIMILARITY IN EFFICACY, SAFETY
Preclinical functional evaluation must demonstrate the biosimilar's biological activity and its similarity to the reference product. Evaluated criteria include studies of biological activity, pharmacodynamics, and toxicology, which must show comparable results for clinical transferability[2], [3], [10].
Efficacy and Biological Activity:
Results must demonstrate no clinically significant differences, with specific criteria defined by the study. The tolerance window for biological activity may vary depending on the product's mechanism of action, but results must support biosimilarity without significant deviations.
Toxicology:
Studies must generally show a comparable toxicological profile without new or unexpected safety signals.
Criteria:
Preclinical studies must show comparable bioactivity, generally within an 80-125% range compared to the reference product for pharmacodynamic parameters.
Results for Clinical Transferability:
Results must support clinical similarity without requiring an exact numerical match but rather functional equivalence, based on a solid demonstration of similarity in efficacy and absence of new or unexpected toxicity in animal models.
3.4. CLINICAL STUDIES – SIMILARITY IN EFFICACY, SAFETY
Clinical Trial Authorization: Clinical trial authorization in the EU is managed by competent national authorities in collaboration with the EMA. Criteria for admissibility include demonstrating a robust study design, justifying the patient population, and safety protocols[2], [3], [10], [20].
Primary Outcome Criteria:
Primary outcome criteria for biosimilar clinical trials focus on efficacy, safety, and immunogenicity, with results that must be statistically comparable to those of the reference product[2], [3], [10].
Efficacy and Safety:
Clinical studies must confirm biosimilarity with comparable safety and efficacy profiles to the reference product, and that differences observed in molecular and functional analyses do not have clinically significant impacts. Any equivalence margin must be scientifically justified to be approved by the EMA.
Immunogenic Data:
Studies must demonstrate that the biosimilar's risk of immunogenicity does not exceed that of the reference product, with 95% confidence intervals for antibody proportions.
3.5. MARKETING AUTHORIZATION AND BIOSIMILAR APPROVAL:
The EMA is the regulatory body concerned with the marketing authorization of biosimilars in the EU. The evaluation process is centralized and results in an authorization valid in all EU member states.
For approval,regulatory decisions are made based on a comprehensive assessment of each biosimilar's dossier, including quality, preclinical, and clinical data, considering the totality of evidence provided rather than isolated criteria.
Results must indicate similarity in terms of efficacy and safety, with a narrowly defined tolerance margin. Immunogenicity data are particularly scrutinized for their potential impact on efficacy and safety [2], [3], [10].
Efficacy and Safety:
Clinical outcomes that fall within the established tolerance windows of the original biological drug for one or more approved indications, using one or more primary clinical criteria to demonstrate non-inferiority or equivalence to the reference product.
Immunogenicity:
The rates of immunogenic antibody reactions (ADA) of biosimilars must be comparable to those of the reference product, without a significant increase in immune reactions in the studied populations.
No significant increase in ADA rates or negative safety profiles compared to the reference product is accepted.
Manufacturing Certificate attesting to GMP:
in accordance with European guidelines.
Molecular Similarity:
Results demonstrating molecular similarity between the biosimilar and its reference product. Including purity, primary structure, secondary and tertiary structure conformation, and post-translational modifications.
It's crucial to consult the EMA for product-specific guidelines, as requirements can vary significantly depending on the complexity of the biosimilar and its clinical application. Biosimilar developers should engage in early dialogue with the EMA to ensure their development program meets Europe-specific regulatory requirements [2], [3], [10]..
3.5.POST-MARKETING SURVEILLANCE:
Post-marketing surveillance is managed through the EudraVigilance system, which allows for the collection and analysis of drug safety information. Criteria include monitoring adverse effects and long-term efficacy[21].
HOPING THIS BRIEF SUMMARY HELPS YOU WITH YOUR FUTURE BIOSIMILARS R&D PROJECTS
If you need help with your scientific and regulatory R&D in Biosimilars, feel free to contact me at contact@orphanquantum.com
See you soon,
Tristan.
*Please let me know if you spot any errors or would like to suggest details I might have missed that could be relevant to include in this analysis.
SOURCES
[1] “FDA - Biological Product Definitions”.
[2] “EMA - biological medicinal products”, Accessed: Mar. 01, 2024. [Online]. Available: https://www.ema.europa.eu/en/documents/leaflet/biological-medicinal-products_en.pdf
[3] FDA, “FDA. Overview of Biosimilar Products.”, Accessed: Mar. 01, 2024. [Online]. Available: https://www.fda.gov/media/151058/download
[4] M. Weise et al., “Biosimilars: what clinicians should know,” Blood, vol. 120, no. 26, pp. 5111–5117, Dec. 2012, doi: 10.1182/blood-2012-04-425744.
[5] “EMA - Biosimilar medicines: Overview | European Medicines Agency.” Accessed: Mar. 01, 2024. [Online]. Available: https://www.ema.europa.eu/en/human-regulatory-overview/biosimilar-medicines-overview
[6] C. for D. E. and Research, “Scientific Considerations in Demonstrating Biosimilarity to a Reference Product.” Accessed: Mar. 01, 2024. [Online]. Available: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/scientific-considerations-demonstrating-biosimilarity-reference-product
[7] V. L. Van de Wiele, R. F. Beall, A. S. Kesselheim, and A. Sarpatwari, “The characteristics of patents impacting availability of biosimilars,” Nat Biotechnol, vol. 40, no. 1, pp. 22–25, Jan. 2022, doi: 10.1038/s41587-021-01170-5.
[8] “Biosimilars-economics-and-intellectual-property.pdf”, Accessed: Mar. 01, 2024. [Online]. Available: https://media.crai.com/sites/default/files/publications/Biosimilars-economics-and-intellectual-property.pdf
[9] L. Barbier, A. Mbuaki, S. Simoens, P. Declerck, A. G. Vulto, and I. Huys, “Regulatory Information and Guidance on Biosimilars and Their Use Across Europe: A Call for Strengthened One Voice Messaging,” Front Med (Lausanne), vol. 9, p. 820755, 2022, doi: 10.3389/fmed.2022.820755.
[10] “EMA - Biosimilars in the EU - Information guide for healthcare professionals”.
[11] P. Kurki, L. van Aerts, E. Wolff-Holz, T. Giezen, V. Skibeli, and M. Weise, “Interchangeability of Biosimilars: A European Perspective,” BioDrugs, vol. 31, no. 2, pp. 83–91, Apr. 2017, doi: 10.1007/s40259-017-0210-0.
[12] P. Kurki, S. Barry, I. Bourges, P. Tsantili, and E. Wolff-Holz, “Safety, Immunogenicity and Interchangeability of Biosimilar Monoclonal Antibodies and Fusion Proteins: A Regulatory Perspective,” Drugs, vol. 81, no. 16, pp. 1881–1896, Nov. 2021, doi: 10.1007/s40265-021-01601-2.
[13] M. Schiestl, M. Zabransky, and F. Sörgel, “Ten years of biosimilars in Europe: development and evolution of the regulatory pathways,” Drug Des Devel Ther, vol. 11, pp. 1509–1515, May 2017, doi: 10.2147/DDDT.S130318.
[14] H. P. Cohen, M. Turner, D. McCabe, and G. R. Woollett, “Future Evolution of Biosimilar Development by Application of Current Science and Available Evidence: The Developer’s Perspective,” BioDrugs, vol. 37, no. 5, pp. 583–593, Sep. 2023, doi: 10.1007/s40259-023-00619-0.
[15] I. Gherghescu and M. B. Delgado-Charro, “The Biosimilar Landscape: An Overview of Regulatory Approvals by the EMA and FDA,” Pharmaceutics, vol. 13, no. 1, p. 48, Dec. 2020, doi: 10.3390/pharmaceutics13010048.
[16] S. A. Berkowitz, J. R. Engen, J. R. Mazzeo, and G. B. Jones, “Analytical tools for characterizing biopharmaceuticals and the implications for biosimilars,” Nat Rev Drug Discov, vol. 11, no. 7, pp. 527–540, Jul. 2012, doi: 10.1038/nrd3746.
[17] N. Nupur, S. Joshi, D. Gulliarme, and A. S. Rathore, “Analytical Similarity Assessment of Biosimilars: Global Regulatory Landscape, Recent Studies and Major Advancements in Orthogonal Platforms,” Front Bioeng Biotechnol, vol. 10, p. 832059, Feb. 2022, doi: 10.3389/fbioe.2022.832059.
[18] C. for D. E. and Research, “FDA - Current Good Manufacturing Practice (CGMP) Regulations,” FDA, Dec. 2023, Accessed: Mar. 01, 2024. [Online]. Available: https://www.fda.gov/drugs/pharmaceutical-quality-resources/current-good-manufacturing-practice-cgmp-regulations
[19] “EMA - Good manufacturing practice | European Medicines Agency.” Accessed: Mar. 01, 2024. [Online]. Available: https://www.ema.europa.eu/en/human-regulatory-overview/research-and-development/compliance-research-and-development/good-manufacturing-practice
[20] “Clinical Trials Regulation | European Medicines Agency.” Accessed: Mar. 01, 2024. [Online]. Available: https://www.ema.europa.eu/en/human-regulatory-overview/research-and-development/clinical-trials-human-medicines/clinical-trials-regulation
[21] “EudraVigilance | European Medicines Agency.” Accessed: Mar. 01, 2024. [Online]. Available: https://www.ema.europa.eu/en/human-regulatory-overview/research-and-development/pharmacovigilance-research-and-development/eudravigilance
Комментарии