Whitepaper: CMC Guidelines for Breakthrough Therapy Drug Development, from Concept to Compliance


This whitepaper offers a critical roadmap for navigating the complexities of regulatory compliance in biologic therapy development. Highlighting innovative strategies for chemical manufacturing and controls (CMC), this guide emphasizes laboratory practices and analytical methods to mitigate impurities, contamination, and toxicity, whilst ensuring strict adherence to ICH and FDA standards. Aimed at drug development teams, this document synthesizes expert insights, sophisticated methodologies, and regulatory guidelines into actionable knowledge, simplifying the intricate journey from laboratory bench to market. Biologic therapies have revolutionized the biopharmaceutical landscape with targeted, precision medicine. These treatments, including antibodies, complex proteins, peptides, and oligonucleotides, have fundamentally reshaped healthcare by addressing diseases at their molecular level. But ensuring the safety and efficacy of these therapies requires navigating a complex and evolving regulatory framework. Chemical Manufacturing and Controls (CMC) meticulously scrutinize each biologic for risks such as instability, impurities, and degradation. This complexity affects stakeholders in the life sciences ecosystem, who must adhere to guidelines from bodies like the International Council for Harmonisation (ICH) and the U.S. FDA. Therefore, we have crafted a comprehensive whitepaper to help manufacturers understand key regulatory documents and stay ahead in the shifting regulatory environment. Navigating Regulatory Challenges Amidst Increased Regulatory Scrutiny Biologic drug therapies are rapidly evolving, prompting regulatory agencies like the FDA to continually update guidelines to ensure the safety, efficacy, and quality of these innovative treatments. Manufacturers are encouraged to rigorously adhere to Good Manufacturing Practices (GMP) that align with the three tiers of CMC testing: sterility, toxicity, and stability. However, it can be hard for manufacturers to know where to turn amidst a growing number of guidance documents. The complexity and frequent updates of these regulations, driven by the latest scientific advancements, emerging data, public health issues, and technological breakthroughs, make compliance a daunting process. Here, we provide a detailed list of key FDA guidance documents and regulatory sections essential for biologic development. This comprehensive guide helps developers navigate the regulatory landscape, ensuring compliance needs are met across the various vendors involved in the development of their biologic therapies amidst evolving requirements. The Three Tiers of CMC Testing The three tiers of CMC testing provide an essential framework for manufacturers to bring safe, effective, and high-quality biologic treatments to the market. This framework helps to ensure that biologic drugs are free from harmful microorganisms, safe for human use, and stable under specified conditions. Tier 1 – Sterility Testing: Typically conducted early in the manufacturing process to ensure that the product is free from microbial contamination before further processing or testing. Tier 2 – Toxicity Testing: Generally performed after initial product formulation or development to assess the safety of the product. Toxicity testing helps determine any adverse effects the product may have on biological systems and informs dosage levels and potential risks associated with use. Tier 3 – Stability Testing: Usually conducted later in the manufacturing process, after the product has been formulated and packaged, to assess its shelf life and how it retains its quality over time. Stability testing ensures that the product remains safe and effective throughout its intended shelf life under various storage conditions. Current Regulations Sterility Testing 21 CFR 610.12 (Sterility Test): Details the procedures and requirements for conducting sterility tests on biologic products to ensure they are free from harmful microorganisms. 21 CFR 211.94 (Drug Product Containers and Closures): Specifies the standards for the design, material, and construction of containers and closures to ensure they do not affect the safety, strength, quality, or purity of the drug. 21 CFR 211.167 (Special Testing Requirements): Outlines special testing requirements to guarantee the biologic product’s quality and efficacy, including additional tests not covered under general provisions. Toxicity Testing ICH Q6A (Specifications: Test Procedures and Acceptance Criteria for New Drug Substances and New Drug Products: Chemical Substances): Sets the standard for designing test procedures and establishing acceptance criteria for new chemical drug substances and products. ICH Q6B (Specifications: Test Procedures and Acceptance Criteria for Biotechnological/Biological Products): Offers guidance on the test procedures and acceptance criteria specifically for biotechnological and biological products. 87 FR 67037 (Current Good Manufacturing Practice Requirements for Combination Products): This Federal Register notice details the regulatory framework for combination products, underscoring the GMP standards that must be met when biological products are combined with either drug or device components. ICH Q5A(R2) (Viral Safety Evaluation of Biotechnology Products Derived from Cell Lines of Human or Animal Origin): Provides additional recommendations on the established and complementary approaches to control the potential viral contamination of biotechnology products and describes the evaluation of the viral safety of biotechnology products including viral clearance and testing. Stability Testing ICH Q5C ( Stability Testing of Biotechnological/Biological Products): Provides a framework for establishing the stability profile of biotechnological and biological products. ICH Q1A(R2) (Stability Testing of New Drug Substances and Products): Provides a comprehensive framework for the stability testing of new drug substances and products, which is critical for determining shelf life and storage conditions. Tier 1: GMP Sterility Testing Sterility is paramount in biologic development, as any microbial contamination can compromise the safety and efficacy of products. The major regulatory guidelines to reference when developing sterility testing protocols are 21 CFR 211.167(a), 21 CFR 610.12, and ICH Q1A(R2), Q6A, and Q6B. Adopting GMP as outlined in these regulations will enable manufacturers to ensure the integrity of biologic products and meet evolving regulatory requirements. Container Closure Systems Container and closure systems must comply with 21 CFR 211.94, which mandates that they be inert to prevent any reactive, additive, or absorptive interactions that could compromise the drug’s safety, identity, strength, quality, or purity. Additionally, depyrogenated containers are essential during high-risk phases, such as the final fill and packaging of injectable biologics, to mitigate risks of endotoxin or pyrogen contamination. However, maintaining these standards presents several challenges. Ensuring that containers are truly inert and free from contaminants requires rigorous quality assurance practices. Samples must accurately reflect the quality claims of the components and confirm the absence of contaminants such as animal proteins, DNA, endotoxins, other pyrogens, culture media constituents, viruses, and leachables. This process is complex and demands precise and reliable analytical methods. Advanced methods such as Gas Chromatography (GC) and Liquid Chromatography (LC) combined with with Mass Spectrometry (MS) techniques such as High Resolution Mass Spectrometry (HRMS) and Inductively Coupled Mass Spectrometry (ICP-MS) provide effective solutions to these challenges by providing high separation efficiency, high sensitivity and accuracy. Together, these methods provide a comprehensive solution for verifying the inertness of container and closure systems, detecting contaminants, and maintaining the highest standards of product integrity and regulatory compliance. Tier 2: Proactive Strategies for Toxicity Risk Mitigation 87 FR 67037 recommends employing specific methods to improve confidence in the ability of a bioanalytical method employed to detect and differentiate the analyte from other substances, including metabolites, isomers, impurities, degradation products formed during sample preparation. Biologic medicines are more targeted and efficacious than traditional medications, but the method-of-action (MOAs) that make biologics so valuable are also what raise the risks of toxicity and immunological responses. Taking a proactive stance to toxicity testing at clinical development stages helps to minimize the risks of toxicity by characterizing and mitigating potential toxicities before they threaten your product. Essentially allowing lab teams to function as smoke alarms rather than fire departments. Early-stage toxicological screenings can offer early indications of potential adverse effects, allowing for necessary adjustments in the formulation or manufacturing process. In particular, in-depth characterization of the drug products by HRMS & Ion Mobility Mass Spectrometry (IMS) early in development can help detect impurities, contaminants, and degradation products that may lead to toxicities. This proactive stance can also help to maximize resources and streamline regulatory review. Toxicity Testing Methods To select the optimal method for CMC (Chemistry, Manufacturing, and Controls) toxicity testing, manufacturers should thoroughly evaluate the factors of specificity, mixture complexity, and the necessity for accurate compound identification. The use of a stable isotope-labeled internal standard (IS) in Mass Spectrometry (MS) analysis is highly recommended to achieve the highest level of specificity. The integration of a stable isotope-labeled IS with MS can significantly improve the accuracy of quantification and is a commonly accepted practice in bioanalytical method development. UHPLC is the preferred technique for the robust separation of complex mixtures. When coupled with MS, UHPLC-MS can provide valuable information for quantitative analysis, peptide mapping, glycan analysis, and characterization of protein variants. This combination offers detailed insights into the composition, structure, and quality attributes of biological drugs thereby helping manufacturers to predict toxicity risks and take proactive strategies to maximize safety. HRMS offers the added advantage of high resolution and accurate mass measurements, which are particularly beneficial for analyzing complex biological entities like monoclonal antibodies (mABs), fusion proteins, or other large molecules. The accurate mass determination facilitated by these technologies aids in identifying and characterizing various post-translational modifications, impurities, and degradation products, which contributes to method optimization and confirms the molecular integrity of biologic products. Mesoscale Discovery (MSD) is another tool for biologics toxicity testing that provides highly sensitive and specific assays for biomarker detection and quantification. MSD’s electrochemiluminescence (ECL) technology allows for the simultaneous measurement of multiple biomarkers in a single sample, which is crucial for assessing the biological response and potential toxicity of mABs. This multiplexing capability enables a comprehensive analysis of cytokine release, immunogenicity, and other biomarkers associated with adverse effects, facilitating early detection of toxicity issues and improving the safety profile of biologic therapies. Viral Safety of Biotechnology Products ICH Q5A(R2) is a specific guideline that describes the evaluation of the viral safety of biotechnology products including viral clearance and testing. It is applicable for products derived from cell lines of human or animal origin, including cytokines, monoclonal antibodies (mAbs), and subunit vaccines produced from in vitro cell culture using recombinant DNA technologies. While mostly relevant for preclinical toxicity testing, section seven provides points to consider for continuous manufacturing. These include general and unique considerations such as the potential risk related to longer periods in production culture. A validated viral clearance study procedure is also described which uses chromatography to demonstrate the robustness of viral clearance. Tier 3: Optimizing Stability Testing and Compliance Stability testing is crucial in biologic drug development, ensuring safety, efficacy, and regulatory compliance over time. Tailored testing protocols are essential to address the unique characteristics of biologic drugs and meet global regulatory standards. Challenges arise from adapting standard guidelines to suit the nuanced demands of biologic drugs, requiring bespoke adjustments to ensure accurate analysis. The introduction of ICH Q5C represents a significant advancement in stability testing frameworks, specifically tailored to address the unique considerations of biologic drugs. Recognizing potential changes in molecular structure and biological activity over time, ICH Q5C emphasizes the importance of advanced analytical techniques in accurately assessing these variations. By providing guidance that considers the special characteristics of biologics, ICH Q5C enables the development of stable biologic drugs that meet regulatory requirements throughout their intended shelf life. Stability testing methods should therefore employ guidance from these regulations to measure the effects of environmental conditions such as pH, temperature, ionic strength, and the presence of excipients can influence solubility and conformational stability. Accelerated & Stress Studies It is strongly suggested that studies be conducted on the drug substance and drug product under accelerated and stress conditions. Studies under stress conditions may also be useful in determining whether accidental exposures to conditions other than those proposed (e.g., during transportation) are deleterious to the product, and also for evaluating which specific test parameters may be the best indicators of product stability. Utilizing solvents such as isopropyl alcohol or water at varying pH levels, are crucial to replicate real-world stress conditions. Though time-consuming, this testing is essential to ensure that packaging or manufacturing materials do not compromise the long-term integrity of the biologic. Shelf Life The shelf-lives of biotechnological/biological products may vary from days to several years. Thus, it is difficult to draft uniform guidelines regarding the stability study duration and testing frequency that would be applicable to all types of biotechnological/biological products. With only a few exceptions, however, the shelf lives for existing products and potential future products will be within the range of 0.5 to 5 years. When shelf-lives of 1 year or less are proposed, the real-time stability studies should be conducted monthly for the first 3 months and at 3 month intervals thereafter. For products with proposed shelf-lives of greater than 1 year, the studies should be conducted every 3 months during the first year of storage, every 6 months during the second year, and annually thereafter. Stability Testing Methods On the whole, there is no single stability-indicating assay or parameter that profiles the stability characteristics of a biotechnological/biological product. Therefore, manufacturers must provide assurance that changes in the identity, purity, and potency of the product will be detected using proposed methodologies. UHPLC is particularly effective in identifying and quantifying degradation products swiftly, a crucial aspect of stability testing. The technique’s high efficiency stems from its use of sub-2 µm particle size columns, which enhance separation capabilities, allowing for more nuanced assessments of a biologic’s stability profile. For analysis, HRMS stands out for its exceptional mass accuracy and the ability to analyze complex mixtures in detail. Its ability to determine subtle changes in molecular weights indicative of instability, including post-translational modifications in proteins make it especially useful for biologics stability studies. By offering precise differentiation between compounds of similar masses, HRMS is invaluable for elucidating the purity and integrity of biologics throughout their development lifecycle. Methods: The Importance of Flexibility In the realm of CMC, the role of precise and tailored analytical testing is paramount. It not only ensures compliance with stringent regulatory standards but also guarantees the safety and efficacy of pharmaceutical products. The FDA particularly underscores the value of flexibility in the selection and development of appropriate analytical methods, as well as testing conditions that are specifically aligned with the unique characteristics of each product. Partnering with a CRO that has deep expertise in method development offers significant advantages in this dynamic landscape. Such a CRO can enhance product development efficiency by rapidly adapting and optimizing testing methods to meet specific product profiles. This not only streamlines the regulatory approval process but also enhances the robustness of product quality assessments through customized stability and integrity testing. Ultimately, this partnership ensures rigorous quality control and maintains the highest standards of patient safety, providing a critical competitive edge in pharmaceutical development. Conclusion Biologics hold immense potential to treat disease, improve patient care and quality of life. However, their unique characteristics necessitate tailored CMC testing solutions. The major challenges include: Challenge 1: Checking for contamination and impurities, either derived from the manufacturing process or inherent characteristics of the biologic, Challenge 2: Sufficiently characterizing the biologic early enough in clinical phase to pre-empt degradation or other potentially toxic products. Challenge 3: Fully understanding the effects of environmental conditions on products to ensure stability under stress conditions. As we have seen in this whitepaper, advanced methods such as UHPLC and HRMS are invaluable to ensure the sterility, safety and stability required of biologic therapeutics by existing regulations. UHPLC allows us to separate complex solutions while HRMS lets us characterize these separated molecules in high resolution. Together, they empower us to understand our biologic products like never before and to make predictions that will maximize resources and streamline time-to-market.

Leave a Reply

Your email address will not be published. Required fields are marked *