Vol 79 – Strategies for Addressing Impurities in Biologic Drug Substance Manufacturing

August 17, 2023The Pathfinder 28 Min Read

In the intricate world of biologic drug substance manufacturing, impurities are not just a challenge—they’re a ticking time bomb. These unseen adversaries, lurking in the shadows of the development process, can explode into a full-blown crisis when least expected. From clinical trials to market shelves, biological impurities can derail a product’s entire lifecycle. How can industries arm themselves against these hidden threats, especially when they emerge later in development? This article delves into real-world scenarios, unraveling the complexities and unveiling strategies to defuse the impurity time bomb. 

 

Strategies for Addressing Impurities in Biologic Drug Substance Manufacturing 

 

The production of biologic drug substances presents unique challenges due to the inherent complexity of biological systems. One of the significant concerns in the manufacturing of biologic drugs is the presence of impurities, which can arise from various sources. This article will explore the key strategies for addressing impurities in the manufacturing of biologic drug substances, ensuring quality and safety. 

 

Understanding the Nature of Impurities  

Impurities in biologic drug substances can be classified into two main categories: 

 

  • Product-Related Impurities: These include variants, aggregates, and degradation products of the desired therapeutic protein.  
  • Process-Related Impurities: These impurities arise from the manufacturing process and include host cell proteins, DNA, endotoxins, and residual solvents. Understanding the nature of these impurities is critical for implementing effective control strategies. 

 

Robust Process Design and Optimization  

A well-designed manufacturing process is vital in minimizing impurities. Key strategies include: 

 

  • Selection of Appropriate Host Systems: Choosing the right host cell line and expression system can minimize certain impurities.  
  • Optimization of Upstream Processes: Control of media components, pH, temperature, and other culture conditions can reduce unwanted variations.  
  • Downstream Process Development: Employing various purification techniques, such as chromatography, can further reduce impurities. 

 

Analytical Development and Validation 

Implementing comprehensive analytical methods to detect and quantify impurities is essential. These include: 

 

  • Method Development: Employing techniques like mass spectrometry, liquid chromatography, and capillary electrophoresis to identify impurities.  
  • Validation: Ensuring that the methods are accurate, precise, and consistent across different lots and manufacturing sites. 

 

Regulatory Compliance and Quality Assurance  

Compliance with regulatory guidelines, such as those provided by the FDA, is crucial. Strategies include: 

 

  • Risk-Based Approach: Evaluating and understanding the potential risks associated with impurities.  
  • Stability Data Assessment: Assessing the impact of impurities on the stability of the product over its shelf life.  
  • Implementing Quality by Design (QbD): Integrating quality controls throughout the development and manufacturing process. 

 

Continuous Monitoring and Improvement  

Continuous monitoring and feedback allow for the ongoing assessment of the manufacturing process and the implementation of improvements as needed. Employing technologies like Process Analytical Technology (PAT) can provide real-time insights into the process and enable proactive adjustments. 

 

Know the Guidances: 

 

International Guidances  

  • ICH Q5A(R1): “Quality of Biotechnological Products: Viral Safety Evaluation of Biotechnology Products Derived from Cell Lines of Human or Animal Origin.”  
  • ICH Q5C: “Quality of Biotechnological Products: Stability Testing of Biotechnological/Biological Products.”  
  • ICH Q6B: “Specifications: Test Procedures and Acceptance Criteria for Biotechnological/Biological Products.” 

 

U.S. Food and Drug Administration (FDA)  

  • FDA Guidance for Industry: “Process Validation: General Principles and Practices.”  
  • FDA Guidance for Industry: “Q11 Development and Manufacture of Drug Substances (Chemical Entities and Biotechnological/Biological Entities).” 

 

European Medicines Agency (EMA)  

  • EMA Guideline on Similar Biological Medicinal Products: This guideline covers the quality, non-clinical, and clinical aspects of biosimilars.  
  • EMA Guideline on Development, Production, Characterization, and Specifications for Monoclonal Antibodies and Related Products. 

 

World Health Organization (WHO) 

WHO Guidelines on Evaluation of Similar Biotherapeutic Products (SBPs). 

Other Relevant Guidelines  

PDA Technical Report No. 42: “Process Validation of Protein Manufacturing.”  

USP Chapters on Biological Assay Validation: Includes information on validating assays for biological activity, specific methods for quantifying impurities, etc. 

 

These guidance’s provide a comprehensive framework for the control of impurities in biologic drug substance manufacturing. They cover various aspects such as the characterization of impurities, analytical methods, process design, validation, and quality controls. Compliance with these guidance’s ensures that the manufacturing process meets the required standards for quality, safety, and efficacy. 

 

Deal with Impurities in Biologic Impurities Early   

 

Scenario 1: Selection of Host Cell Line and Expression System 

Problem: 

A biopharmaceutical company is developing a monoclonal antibody and discovers host cell protein (HCP) impurities in the early stages of development. 

 

Solution: 

  • Evaluate Different Host Cell Lines: Assess various cell lines for their propensity to generate HCP impurities. 
  • Optimize Expression Systems: Modify the expression system to reduce the synthesis of HCPs. 
  • Implement Early Screening: Use specific assays to detect HCP impurities and monitor their levels throughout development. 

 

Scenario 2: Impurities in Upstream Processing 

Problem: 

Unwanted product-related impurities like aggregates and degradation products are detected during the fermentation process. 

 

Solution: 

  • Control Culture Conditions: Modify parameters like pH, temperature, and agitation to minimize aggregation. 
  • Use Appropriate Media Components: Select media components that minimize degradation. 
  • Implement In-Process Controls: Employ real-time monitoring to detect and control impurity formation early in the process. 

 

Scenario 3: Residual Solvents in Downstream Processing 

Problem: 

A company identifies residual solvents in the purification stage, which can affect the final product’s quality. 

Solution: 

  • Optimize Purification Steps: Adjust chromatography conditions to enhance the removal of solvents. 
  • Implement Rigorous Testing: Use validated analytical methods to detect residual solvents early in the process. 
  • Review Supplier Quality: Ensure that raw materials, including solvents, meet quality standards. 

 

Scenario 4: Risk of Contamination with Endotoxins 

Problem: 

A potential risk of bacterial endotoxin contamination is identified in a biologic drug substance derived from microbial fermentation. 

 

Solution: 

  • Implement Rigorous Sterilization: Use validated sterilization methods for all equipment and materials. 
  • Monitor for Contamination: Utilize sensitive assays to detect endotoxins at an early stage. 
  • Incorporate Endotoxin Removal Steps: Implement specific purification steps to remove or inactivate endotoxins. 

 

These scenarios highlight the forward-thinking approach required to address impurities in Biologic Drug Substance manufacturing. Early identification, thorough understanding of the source, and robust strategies for mitigation are vital. Collaboration between process development, analytical development, quality assurance, and regulatory teams is essential to ensure that the product meets the required purity, safety, and efficacy standards.  

By taking a proactive approach and implementing a robust regulatory development framework, sponsors working with their CMOs can build quality into the product from the outset, ensuring the successful development of biologic drug substances. 

 

Some Case Studies that illustrate how impurities can become an issue Later in Development 

 

Case Study 1: Formation of Protein Aggregates During Stability Testing 

Problem: 

During stability testing of a therapeutic protein, the formation of protein aggregates was detected. These aggregates could potentially lead to immunogenic responses in patients. 

 

Solution: Investigate the Cause: Analytical methods were employed to understand the cause of aggregation. 

  • Modify Formulation: The formulation was optimized by adjusting pH, buffer, and excipient levels to minimize aggregation. 
  • Implement Additional Monitoring: Enhanced testing protocols were put in place to monitor aggregation throughout shelf-life. 
  • Consult Regulatory Guidelines: Ensured compliance with relevant regulatory guidelines for addressing this impurity. 

 

Case Study 2: Detection of Host Cell DNA in Late-Stage Development 

Problem: 

Host cell DNA impurities were detected in late-stage development of a monoclonal antibody, exceeding acceptable levels. 

 

Solution: 

  • Enhance Purification Steps: Downstream processes were optimized to enhance the removal of DNA impurities. 
  • Implement In-Line Monitoring: Introduced real-time monitoring to detect and control DNA levels. 
  • Conduct Risk Assessment: Performed a comprehensive risk assessment in line with ICH guidelines. 
  • Engage with Regulatory Authorities: Collaborated with regulatory bodies to ensure the final product met all requirements. 

 

Case Study 3: Endotoxin Contamination During Scale-Up 

Problem: 

During the scale-up phase of a recombinant protein, endotoxin contamination was detected, posing a serious risk to patient safety. 

 

Solution: 

  • Identify the Source: Conducted a thorough investigation to identify the contamination source. 
  • Implement Endotoxin Removal Techniques: Utilized specialized chromatography and filtration techniques to remove endotoxins. 
  • Revise Sterilization Protocols: Improved sterilization protocols to prevent future contamination. 
  • Update Quality Assurance Measures: Integrated rigorous QA measures to ensure compliance with regulatory standards. 

 

Case Study 4: Residual Protein A in a Monoclonal Antibody Product 

Problem: 

Residual Protein A, used in purification, was detected in a monoclonal antibody product during phase III clinical trials, exceeding permitted levels. 

 

Solution: 

  • Optimize Protein A Removal: Adjusted chromatography conditions to enhance Protein A removal. 
  • Implement Rigorous Testing: Employed validated methods to monitor residual Protein A levels. 
  • Engage with Clinical Teams: Worked with clinical development teams to assess potential impacts on ongoing trials. 
  • Consult Regulatory Authorities: Collaborated with regulatory agencies to align with industry standards and ensure patient safety. 

 

These examples highlight that addressing biological impurities later in development requires a multifaceted approach. It involves rigorous investigation, optimization of manufacturing processes, implementation of robust analytical methods, and close collaboration with regulatory authorities.  

It’s essential to maintain a flexible and responsive development strategy that can adapt to unexpected challenges. Leveraging expertise in quality assurance, regulatory compliance, analytical development, and manufacturing can mitigate these challenges and ensure the successful development and commercialization of biologic drug substances. 

 

Conclusion 

 

The fight against biological impurities is an unending battle, demanding vigilance, innovation, and adaptability. The case studies explored demonstrate that these microscopic invaders can breach even the most fortified defenses. But surrender is not an option. The relentless pursuit of perfection in biologic drug substance manufacturing requires not just early intervention but a robust arsenal of strategies even in later stages of development. It’s a high-stakes game where the prize is nothing less than the safety and efficacy of life-saving medicines. The question is not if the impurity time bomb will strike, but when—and how well-prepared will we be when it does? The answer lies in our collective commitment to quality, precision, and unwavering regulatory compliance. The clock is ticking—are we ready? 

Upcoming Event

Oct 22

AAPS 2023 PHARMSCI 360

October 22-25, 2023 

Orlando, FL, USA

Orange County Convention Center

The Pathfinder
WILA
WILA

The latest in biotech for your ears

The VirtualPharm Podcast is an audio series that covers topics related to compliance and regulatory affairs in the Lifesciences industry. Each episode, hosted by an expert in the topic with decades of experience, offers listeners detailed insight into the changing regulatory environment.

Subscribe to the Pathfinder


    Lets discuss your product