Call for Abstract

17th European Biosimilars Congress, will be organized around the theme “Biopharmaceutical Innovations: Ensuring Quality in Biosimilar Production”

Eurobiosimilars 2024 is comprised of keynote and speakers sessions on latest cutting edge research designed to offer comprehensive global discussions that address current issues in Eurobiosimilars 2024

Submit your abstract to any of the mentioned tracks.

Register now for the conference by choosing an appropriate package suitable to you.

The development of biosimilars presents a myriad of current challenges. One major hurdle is the complex nature of biologic drugs, which are derived from living organisms and exhibit intricate structures and functions. Replicating these characteristics with high accuracy requires extensive scientific knowledge and advanced technology. Additionally, regulatory requirements for biosimilars demand comprehensive data demonstrating similarity to the reference biologic, including rigorous comparability studies. This necessitates significant investment in research and development, as well as clinical trials to ensure safety and efficacy. Moreover, intellectual property considerations and legal frameworks pose additional obstacles for biosimilar manufacturers. Overcoming these challenges requires collaboration between scientific experts, regulatory authorities, and industry stakeholders to foster a robust biosimilar development ecosystem.

Biosimilars have emerged as a promising development in the field of oncology, offering new avenues for the treatment of cancer. These specialized biologic drugs are designed to be highly similar to existing, FDA-approved reference biologics, and they have the potential to provide cost-effective alternatives for cancer patients. Biosimilars can target various aspects of cancer management, including tumor cell growth inhibition, immune system modulation, and supportive care. By providing more affordable options for cancer therapies, biosimilars can help reduce the financial burden on patients and healthcare systems while expanding access to high-quality cancer treatments.


Chemical and analytical strategies play a vital role in the development and characterization of biosimilars. These strategies aim to establish comparability between the biosimilar and the reference biologic by assessing their physicochemical properties, purity, and potency. Various analytical techniques, such as mass spectrometry, chromatography, and spectroscopy, are employed to evaluate critical quality attributes (CQAs) of the biosimilar. Additionally, advanced structural analysis techniques, including X-ray crystallography and nuclear magnetic resonance (NMR) spectroscopy, are used to examine the higher-order structure of the biosimilar. These strategies enable a comprehensive understanding of the biosimilar's molecular characteristics, facilitating the identification of potential differences and ensuring the safety and efficacy of the product.

Intellectual property rights (IPR) refer to legal rights granted to individuals or organizations to protect their creations or inventions. These rights provide exclusive ownership and control over intangible assets, including patents, trademarks, copyrights, and trade secrets. Intellectual property serves as an incentive for innovation by granting creators and inventors the ability to monetize their work and prevent unauthorized use or reproduction by others. It encourages research and development, fosters economic growth, and promotes creativity in various industries. However, managing intellectual property can be complex, as it involves navigating legal frameworks, enforcing rights, and addressing infringement issues. Striking a balance between protecting intellectual property and promoting access to knowledge remains a challenge, particularly in areas such as technology, pharmaceuticals, and entertainment, where rapid advancements and evolving business models constantly reshape the IPR landscape.

Biological medicine, also known as biologics, refers to therapeutic products derived from living organisms or their components. Unlike traditional chemical-based drugs, biological medicines are typically large and complex molecules, including proteins, antibodies, and nucleic acids. These medicines are designed to target specific disease mechanisms, offering innovative treatment options for various diseases such as cancer, autoimmune disorders, and chronic illnesses. Biological medicines are produced using advanced biotechnological processes, such as genetic engineering and cell culture techniques. Due to their complexity and manufacturing intricacies, the development, production, and regulation of biological medicines require specialized expertise and stringent quality control measures. Despite the challenges, biological medicine continues to revolutionize healthcare by providing targeted and personalized therapies that have the potential to improve patient outcomes and quality of life.

Emerging biosimilars in therapeutics are poised to revolutionize the healthcare landscape by expanding access to effective and more affordable treatment options. As patents for reference biologics expire, biosimilar versions are being developed and introduced into the market. These emerging biosimilars aim to demonstrate comparable safety, efficacy, and quality to their reference biologics through rigorous analytical and clinical evaluations. By offering cost-effective alternatives, biosimilars have the potential to improve patient access to life-saving treatments for diseases such as cancer, autoimmune disorders, and chronic conditions. Additionally, biosimilars stimulate competition in the biopharmaceutical industry, driving innovation and reducing healthcare costs. With continued research, development, and regulatory support, emerging biosimilars have the potential to transform patient care and enhance the sustainability of healthcare systems worldwide.

Innovative clinical approaches in biosimilars aim to streamline the development and evaluation process while ensuring patient safety and efficacy. Adaptive clinical trial designs, such as seamless phase transitions or Bayesian methods, allow for more efficient and flexible study protocols. These approaches optimize sample size, enhance statistical power, and enable real-time adjustments based on emerging data. Additionally, the use of biomarkers and predictive modeling helps identify patient populations likely to respond favorably to biosimilars, facilitating targeted treatment approaches. Furthermore, real-world evidence studies and post-marketing surveillance play a crucial role in monitoring the long-term safety and effectiveness of biosimilars. By embracing innovative clinical approaches, the development and adoption of biosimilars can be accelerated, benefiting patients and healthcare systems worldwide.

Regulatory updates on biosimilars play a vital role in ensuring patient safety, fostering competition, and promoting access to affordable biological therapies. Regulatory authorities, such as the FDA and EMA, continuously refine and update guidelines to provide clear pathways for the development, approval, and post-marketing surveillance of biosimilars. These updates often include guidance on demonstrating similarity, interchangeability, and extrapolation of indications. Additionally, regulatory agencies strive to enhance transparency and education for healthcare professionals and patients regarding biosimilar products. As the field of biosimilars evolves, regulatory updates address emerging challenges, such as interchangeability and naming conventions, to provide a robust framework for market entry and to inspire confidence in these cost-effective alternatives. Regular updates ensure that regulatory frameworks keep pace with scientific advancements and contribute to the continued growth and acceptance of biosimilars in clinical practice.

Biosimilars development in markets has gained significant traction, offering potential benefits in terms of cost savings, increased access to treatments, and fostering competition. As patents for reference biologics expire, biosimilar manufacturers seize the opportunity to enter the market with their comparable versions. However, successful market entry for biosimilars is dependent on several factors, including regulatory approval, pricing strategies, physician and patient acceptance, and reimbursement policies. Education and awareness campaigns are crucial to address misconceptions and build confidence in biosimilars among healthcare professionals and patients. Market dynamics vary across regions, with some markets being more receptive to biosimilars than others. As biosimilars continue to gain momentum, strategic partnerships, effective marketing, and robust supply chains will play a pivotal role in driving their adoption and market success.

The approval and integration of biosimilars, also known as biogenerics, into clinical practice have the potential to transform patient care by expanding treatment options and improving affordability. Regulatory agencies, such as the FDA and EMA, rigorously assess the comparability of biosimilars to their reference biologics through comprehensive scientific evaluations and clinical trials. Once approved, biosimilars are prescribed by healthcare professionals who consider factors such as safety, efficacy, and cost-effectiveness. Clinical practice guidelines and educational initiatives aid in the appropriate and informed use of biosimilars, ensuring patient safety and optimizing outcomes. As healthcare systems embrace biosimilars, their integration into clinical practice holds promise for reducing treatment costs, increasing patient access, and improving overall healthcare sustainability.

Biopharmaceutical informatics, also known as bioinformatics in the context of pharmaceuticals, involves the application of computational and information science methods to collect, analyze, and interpret biological and pharmaceutical data. It encompasses a wide range of techniques and tools that aid in the discovery, development, and optimization of biopharmaceutical products. Biopharmaceutical informatics enables the efficient handling and integration of complex data sets, such as genomics, proteomics, and clinical trial data, to gain insights into disease mechanisms, target identification, and drug design. It also plays a crucial role in pharmacovigilance, drug safety assessment, and personalized medicine approaches. By leveraging computational models, machine learning algorithms, and data mining techniques, biopharmaceutical informatics accelerates research and development processes, facilitates decision-making, and supports evidence-based healthcare practices.

Brexit, the United Kingdom's withdrawal from the European Union (EU), has had several consequences on biosimilars. Prior to Brexit, the EU provided a unified regulatory framework for the approval and market access of biosimilars across member states. However, with the UK's departure, the Medicines and Healthcare products Regulatory Agency (MHRA) now oversees the regulation and approval of biosimilars specifically for the UK market. This has led to additional regulatory complexities and increased costs for biosimilar manufacturers who need to navigate separate approval processes for the UK and EU markets. Moreover, the divergence in regulations between the UK and the EU has resulted in uncertainties regarding mutual recognition and potential delays in launching biosimilars in both regions. These consequences have introduced challenges for biosimilar developers and impacted patient access to more affordable treatment options in both the UK and EU markets.

Biopharmaceutics Classification System (BCS) and In Vitro-In Vivo Correlation (IVIVC) are important concepts in the pharmaceutical industry that can facilitate the approval process for generic drugs, including biowaivers for certain dosage forms. BCS categorizes drugs based on their solubility and permeability characteristics, allowing for predictions about their bioavailability. Drugs falling into BCS Class I and III, which are highly soluble and highly permeable, respectively, are often eligible for biowaivers. IVIVC is a scientific approach that establishes a relationship between in vitro dissolution profiles and in vivo drug absorption, helping to bridge the gap between lab testing and clinical outcomes. The utilization of BCS and IVIVC based biowaivers in drug development can save time and costs, enabling the faster introduction of generic and biosimilar products to the market, ultimately benefiting patients by increasing accessibility to affordable medications.

The biosimilars market and cost analysis are crucial aspects in evaluating the economic impact of biosimilar products. Biosimilars offer the potential for cost savings compared to their reference biologics, providing opportunities to reduce healthcare expenditure. Market analysis involves assessing factors such as market size, competition, and pricing strategies of biosimilar manufacturers. Cost analysis evaluates the pricing and cost-effectiveness of biosimilars in comparison to reference biologics, taking into account factors such as production costs, research and development investments, and market dynamics. By conducting comprehensive market and cost analyses, stakeholders, including healthcare providers, payers, and policymakers, can make informed decisions regarding the adoption and utilization of biosimilars, leading to improved affordability and accessibility of these important therapies.

Biosimilars pharmacovigilance presents unique challenges due to the complex nature of these products and the need for comprehensive monitoring of their safety profiles. One challenge is the potential for immunogenicity, as biosimilars can elicit immune responses in patients, leading to adverse reactions. Robust pharmacovigilance systems must be in place to detect and evaluate such events. Additionally, post-marketing surveillance for biosimilars requires effective traceability systems to distinguish between different products, ensuring accurate reporting and monitoring of adverse events. Furthermore, the global nature of biosimilar markets poses challenges in harmonizing pharmacovigilance practices and exchanging safety information across different regulatory authorities. Addressing these challenges necessitates close collaboration among stakeholders, standardized reporting frameworks, and ongoing vigilance to ensure the continued safety and confidence in biosimilars.

Legal issues surrounding biosimilars revolve around intellectual property rights, patent disputes, and regulatory frameworks. The Biologics Price Competition and Innovation (BPCI) Act, enacted in the United States, created a pathway for the approval and market entry of biosimilars. However, legal challenges arise from patent litigation initiated by reference biologic manufacturers to protect their exclusivity. These disputes can delay the launch of biosimilars and extend market exclusivity for reference products. The BPCI Act also addresses the interchangeability of biosimilars, providing a framework for automatic substitution at the pharmacy level. However, state-specific laws and regulations can introduce further legal complexities. Navigating these legal issues requires a thorough understanding of intellectual property laws, regulatory guidelines, and litigation strategies to ensure fair market competition and patient access to affordable biosimilars.

Formulation strategies for follow-on biologics, also known as biosimilars, are essential for ensuring product stability, efficacy, and safety. These strategies focus on developing formulations that maintain the critical quality attributes (CQAs) of the reference biologic while optimizing manufacturing processes and minimizing variability. Formulation considerations include excipient selection, pH adjustment, buffer systems, and stabilization techniques to prevent protein degradation and maintain structural integrity. Additionally, advanced technologies such as protein engineering, controlled release systems, and drug delivery platforms can be employed to enhance the performance and patient experience of biosimilars. Formulation strategies play a crucial role in achieving bioequivalence between biosimilars and their reference products, ensuring consistent therapeutic outcomes and facilitating their successful development and commercialization.

Current agency expectations for the approval of biosimilars focus on demonstrating a high level of similarity to the reference biologic in terms of quality, safety, and efficacy. Regulatory authorities, such as the FDA and EMA, require comprehensive analytical and preclinical data to establish a robust scientific foundation for biosimilarity. This includes extensive physicochemical and biological characterization, comparative pharmacokinetic and pharmacodynamic studies, and non-clinical toxicity assessments. Clinical trials are also conducted to demonstrate similar clinical efficacy and safety profiles. In addition to comparative data, agencies emphasize the importance of a thorough risk management plan and post-marketing surveillance to ensure ongoing safety monitoring. By meeting these agency expectations, biosimilar manufacturers can obtain regulatory approval and provide patients with cost-effective alternatives to reference biologics while maintaining high standards of quality and safety.

The biosimilars research pipeline is an active area of investigation, with numerous molecules undergoing development and evaluation. Researchers are focusing on expanding the range of therapeutic indications for biosimilars, targeting diseases such as cancer, autoimmune disorders, and inflammatory conditions. Additionally, efforts are underway to enhance the manufacturing processes, improve characterization techniques, and explore novel drug delivery systems for biosimilars. The research pipeline also encompasses the development of biosimilars for complex biologics, such as monoclonal antibodies and fusion proteins, requiring innovative approaches and technologies. As research progresses, the biosimilars research pipeline holds promise for introducing a wider array of cost-effective treatment options, increasing patient access to essential therapies, and promoting competition in the biopharmaceutical market.

The globalization of biosimilars has gained momentum as these products continue to expand their presence in international markets. Increased harmonization of regulatory frameworks, such as the EMA in Europe and the FDA in the United States, has facilitated the development, approval, and cross-border acceptance of biosimilars. Moreover, collaborations between biosimilar manufacturers, contract research organizations, and global supply chains have enabled efficient production and distribution of these products worldwide. The globalization of biosimilars has significant implications for patient access to affordable treatments, fostering competition, and reducing healthcare costs on a global scale. However, challenges remain in terms of navigating varying regulatory landscapes, addressing local market dynamics, and ensuring robust pharmacovigilance practices. By overcoming these challenges, the globalization of biosimilars has the potential to transform healthcare by improving access to life-saving therapies and promoting sustainable healthcare systems globally.

Drug delivery and development play a crucial role in optimizing the efficacy, safety, and patient experience of pharmaceutical products. Advancements in drug delivery technologies, such as nanoparticle-based systems, targeted drug delivery, and sustained-release formulations, enable precise and controlled release of drugs at specific sites in the body. These technologies enhance therapeutic outcomes, minimize side effects, and improve patient adherence to treatment regimens. In drug development, formulation scientists employ various strategies to improve drug solubility, stability, and bioavailability. They also explore novel delivery routes, such as transdermal, inhalation, and implantable systems, to enhance drug absorption and patient convenience. By integrating innovative drug delivery approaches into the development process, pharmaceutical researchers strive to maximize the therapeutic potential of drugs and enhance patient care.

Monoclonal antibody (mAb) biosimilars are an important segment of the biosimilars market, offering more affordable alternatives to reference mAb therapies. Developing biosimilars for mAbs presents unique challenges due to their complex structures and intricate manufacturing processes. Extensive analytical characterization, including physicochemical and functional assessments, is crucial to establish similarity to the reference mAb. Comparative clinical studies are conducted to demonstrate similar efficacy, safety, and immunogenicity profiles. Regulatory agencies have specific guidelines for the development and approval of mAb biosimilars, ensuring a comprehensive evaluation of quality attributes. The availability of mAb biosimilars expands treatment options for patients, increases competition, and potentially reduces healthcare costs, while maintaining high standards of safety and effectiveness.

The USFDA has been actively approving biosimilars to increase patient access to more affordable treatment options. Some notable USFDA-approved biosimilars include those for reference biologics such as adalimumab, infliximab, and trastuzumab. These biosimilars have undergone rigorous evaluations to demonstrate similarity in terms of safety, efficacy, and quality to their respective reference products. The USFDA's approval of these biosimilars provides healthcare providers and patients with alternatives that offer similar clinical benefits while potentially reducing healthcare costs. The expanding portfolio of USFDA-approved biosimilars signifies the growing acceptance and adoption of these products in the United States, contributing to increased competition and improved accessibility to critical biologic therapies.