Vol 56: Developing a Drug from an Academic Discovery: Best Practices for Early CMC Development

June 05, 2023The Pathfinder 46 Min Read

Navigating Biotech Product Development: Balancing Priorities, Risks, and Regulatory Compliance for Success

 

Transforming a promising academic discovery into a marketable drug is a complex and challenging journey. Chemistry, Manufacturing, and Controls (CMC) development is a critical aspect of this process. CMC encompasses the formulation, manufacturing process, and quality control procedures necessary to ensure a drug’s safety, efficacy, and reproducibility. This article will explore the best practices for CMC development, shedding light on the vital steps and considerations required to bring an academic discovery closer to the clinic.

Collaboration between Academia and Industry: Successfully translating an academic discovery into a drug often requires seamless collaboration between academic researchers and industry experts. Academia provides the initial discovery, while industry brings the necessary expertise in drug development, regulatory compliance, and commercialization. Open communication, clear expectations, and well-defined roles and responsibilities are crucial for a productive partnership.

Early Engagement with Regulatory Agencies: Engaging regulatory agencies early in drug development can significantly streamline CMC development. Seek guidance from regulatory experts to ensure your CMC strategy aligns with regulatory requirements. Early interaction allows you to understand the agency’s expectations, anticipate challenges, and proactively address potential roadblocks, saving time and resources.

Robust Formulation Development: Developing a stable and bioavailable formulation is vital for the success of a drug. Formulation development should consider factors like drug solubility, bioavailability, dosing route, and stability. Employ a systematic approach, including pre-formulation studies, excipient selection, and optimization techniques, to develop a formulation that ensures consistent drug delivery and therapeutic effect.

Scalable Manufacturing Process: A robust and scalable manufacturing process is critical to meet the demand for large-scale production. During process development, identify critical process parameters and their impact on product quality. Employ Quality by Design (QbD) principles to design a manufacturing process that minimizes variability and ensures batch-to-batch consistency. Additionally, consider implementing advanced manufacturing technologies, such as continuous manufacturing, to enhance efficiency and reduce costs.

Risk Assessment and Management: Conducting a thorough risk assessment throughout the CMC development process is crucial to identify and mitigate potential risks. Implement a Quality Risk Management (QRM) approach, analyzing process and product-related risks. Proactively address identified risks through appropriate control strategies and mitigation plans. Regularly reassess risks as new data becomes available or process changes occur.

Robust Analytical Methods: Developing and validating robust analytical methods is vital for ensuring the drug’s quality, safety, and efficacy. Analytical methods should accurately quantify the drug substance, assess impurities, and evaluate product quality attributes. Employ advanced techniques such as high-performance liquid chromatography (HPLC), mass spectrometry (MS), and nuclear magnetic resonance (NMR) spectroscopy to ensure accurate and reliable results.

Regulatory Compliance: Throughout CMC development, maintain compliance with relevant regulatory guidelines, including good manufacturing practices (GMP) and International Council for Harmonisation (ICH) guidelines. Document all development activities and establish a comprehensive CMC regulatory package. Implement a robust quality management system to ensure adherence to regulatory standards, minimize deviations, and facilitate a smooth regulatory submission process.

 

Advancing Academic Discoveries Towards IND: Considerations for Preparing a Product for Clinical Trials

 

When an academic discovery shows potential for translation into a drug, bridging the gap between academic research and clinical development is crucial. As academia typically operates outside the realm of Good Manufacturing Practices (GMPs), specific considerations must be considered to advance the product more effectively into the Investigational New Drug (IND) phase. In this article, we will explore critical factors that academic researchers should consider enhancing the readiness of their product as it enters clinical trials.

Preparing for IND Requirements: While academic research may not be conducted under GMPs, understanding the IND requirements is crucial for a smooth transition. Familiarize yourself with the regulatory guidelines the relevant health authorities set, such as the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA). Ensure that your preclinical data meets the necessary safety, efficacy, and toxicology requirements to support the IND application.

Robust Preclinical Studies: Prior to initiating clinical trials, extensive preclinical studies should be conducted to assess the safety and efficacy of the product. Considerations include animal model selection, dose range-finding studies, and evaluation of pharmacokinetics and pharmacodynamics. Robust preclinical data, focusing on the mechanism of action and potential side effects, will provide a solid foundation for the IND application.

CMC Development in Line with IND: Even though academia may not adhere to GMPs during research, it is essential to align the Chemistry, Manufacturing, and Controls (CMC) development with IND requirements. Work closely with contract research organizations (CROs) or manufacturing partners experienced in GMP compliance. Collaborate to develop a scalable and GMP-compliant manufacturing process and establish protocols for release testing, stability studies, and quality control measures.

Robust Analytical Methods and Specifications: Develop and validate robust analytical methods to characterize the drug substance and product. Ensure that the methods are transferable to GMP-compliant laboratories. Establish appropriate specifications for the product’s identity, purity, strength, and quality attributes. This will enable comparability studies throughout development and facilitate a seamless transition to GMP manufacturing.

Stability and Formulation Optimization: Academic research often focuses on the proof-of-concept stage and may not extensively evaluate stability or formulation optimization. However, it is essential to understand the drug’s stability profile under various storage conditions. Conduct stability studies to assess degradation pathways, identify critical stability-indicating parameters, and optimize the formulation to ensure stability over the desired shelf life.

Early Collaboration with Regulatory Experts: Engage regulatory experts or consultants with experience in IND applications and clinical development. They can provide guidance on regulatory requirements, assist in preparing the IND submission package, and help address any regulatory concerns. Early collaboration can ensure that the necessary documentation, data, and supporting information meet the IND submission standards.

Quality Assurance and Control: Implement a quality management system within academia to instill a culture of quality and control. Establish appropriate documentation practices, develop standard operating procedures (SOPs), and ensure data integrity. Although not GMP-compliant, adhering to quality principles during academic research will facilitate a smoother transition to GMP manufacturing during clinical development.

Core Regulatory CMC Compliance Areas to focus on to generate data for the IND

When academia generates data for an Investigational New Drug (IND) application, focusing on specific core regulatory Chemistry, Manufacturing, and Controls (CMC), compliance areas are crucial. While academic institutions may not operate under Good Manufacturing Practices (GMPs), addressing the following areas will help ensure the generated data aligns with regulatory requirements.

Drug Substance Characterization: Thoroughly characterize the drug substance, including its identity, purity, potency, and impurity profile. Provide comprehensive analytical data, such as structure elucidation, spectroscopic analysis, chromatographic purity, and elemental analysis. Address potential genotoxic impurities and confirm compliance with limits set by regulatory agencies.

Manufacturing Process Description: Provide a detailed description of the drug substance manufacturing process, highlighting critical steps, reaction conditions, and process controls. Include information on starting materials, reagents, and intermediate products. Although GMP compliance may not be mandatory at the academic stage, clearly define the process and anticipate any challenges in scaling up for later stages.

Formulation and Drug Product Development: Describe the formulation development process, including the selection and justification of excipients and the composition and manufacturing methods for the drug product. Provide data on the dosage form, stability studies, and compatibility testing. Focus on factors such as stability under different storage conditions, bioavailability, and control of critical quality attributes.

Analytical Method Validation: Validate analytical methods used to assess drug substance and product characteristics. Ensure that the methods are specific, accurate, precise, and robust. Validate methods for quantification, impurity analysis, and characterization. Provide validation data, including system suitability, specificity, linearity, accuracy, precision, and robustness.

Stability Studies: Conduct stability studies to assess the drug substance and product’s stability over the desired shelf life and under various storage conditions. Implement ICH guidelines for stability testing and establish appropriate stability-indicating parameters. Provide comprehensive data on degradation pathways, impurity profiles, and any observed changes in critical quality attributes over time.

Control of Excipients and Raw Materials: Demonstrate control over excipients and raw materials used in the drug substance and drug product manufacturing. Provide specifications, certificates of analysis, and proof of testing for critical materials. Address the identification, purity, potency, and control of potential impurities or contaminants in these materials.

Container Closure System: Evaluate the compatibility between the drug product and the proposed container closure system. Demonstrate that the chosen packaging materials maintain the product’s quality, stability, and sterility throughout its shelf life. Provide information on the container closure integrity and the packaging process.

Manufacturing Site Information: While academic research may not be conducted in GMP-compliant facilities, provide detailed information about the manufacturing site, including equipment, facility design, and process controls. Address any potential issues when scaling up the process, such as cross-contamination, process reproducibility, and batch-to-batch consistency.

GMP Compliance Planning: Develop a comprehensive plan for transitioning from academic research to GMP-compliant manufacturing during the later stages of drug development. Anticipate the necessary infrastructure, equipment, and personnel to meet GMP standards. Consider engaging external manufacturing partners or contract manufacturing organizations (CMOs) experienced in GMP compliance.

Documentation and Record Keeping: Maintain meticulous documentation throughout the CMC development process. Establish appropriate documentation practices, including laboratory notebooks, batch records, protocols, reports, and data integrity controls. Adhere to good documentation practices (GDP) to ensure the generated data’s traceability, accuracy, and reliability.

By focusing on these core regulatory CMC compliance areas, academia can generate data for the IND application that aligns with regulatory requirements. While GMP compliance may not be mandatory at the academic stage, it never hurts to think about it.

 

General Guidance that Should Be Considered

 

While academic researchers may not be directly subject to the same regulatory guidelines as pharmaceutical companies, being aware of certain general guidance documents can help inform their CMC development efforts and align them with regulatory expectations. Here are some key guidance documents to consider:

1.    ICH Guidelines: The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) has developed numerous guidelines that provide recommendations for CMC development. Key guidelines include ICH Q8 (R2) Pharmaceutical Development, ICH Q9 Quality Risk Management, and ICH Q11 Development and Manufacture of Drug Substances.

2.    FDA Guidance Documents: The U.S. Food and Drug Administration (FDA) publishes various guidance documents offering regulatory recommendations and expectations. Relevant guidance documents for CMC development include “CMC Guidance for INDs” and “Chemistry, Manufacturing, and Controls (CMC) Information— Human Gene Therapy Investigational New Drug Applications (INDs).”

3.    EMA Guidelines: The European Medicines Agency (EMA) provides guidelines that offer scientific and technical advice on developing and evaluating medicinal products. Guidelines such as “Guideline on the Requirements for Quality Documentation Concerning Biological Investigational Medicinal Products in Clinical Trials” and “Guideline on Pharmaceutical Quality of Inhalation and Nasal Products” can be helpful for CMC development.

4.    Pharmacopeial Standards: Reference standards established by pharmacopeial organizations, such as the United States Pharmacopeia (USP) and the European Pharmacopoeia (Ph. Eur.), provide important guidelines for drug substances and products. Understanding the requirements and specifications outlined in these compendia can aid in CMC development.

5.    Regulatory Agency Websites: Regularly check the websites of regulatory agencies such as the FDA, EMA, Health Canada, or the Pharmaceuticals and Medical Devices Agency (PMDA) in Japan. These agencies often provide updates, guidelines, and regulatory expectations related to CMC development and IND applications.

6.    Industry Best Practices: Staying informed about industry best practices can provide valuable insights. Industry organizations like the International Society for Pharmaceutical Engineering (ISPE) and the Parenteral Drug Association (PDA) publish guidance documents and organize conferences and webinars that cover various aspects of drug development, including CMC.

7.    Collaborative Networks: Engaging in collaborative networks and partnerships with industry professionals, CROs, or academic-industry consortia can provide access to valuable expertise and resources. These networks often share knowledge and best practices related to CMC development and regulatory compliance.

It is important to note that while regulatory guidelines may not directly bind academic researchers during the academic discovery phase, being aware of these guidance documents can help align their efforts with future regulatory requirements as the product progresses toward clinical trials and commercialization.

Consulting with regulatory experts or seeking advice from CROs experienced in regulatory compliance can further assist academic researchers in navigating the regulatory landscape and ensuring their CMC development aligns with industry standards.

 

Navigating Biotech Product Development: Balancing Priorities, Risks, and Regulatory Compliance

 

Congratulations! Your emerging biotech company has just secured funding and embarked on the exciting journey of developing a compound or in-licensed product. As you delve into the development process, you realize the complexity of managing various contractors, establishing clinical trials, and devising a concise CMC development and regulatory strategy. Additionally, the challenge of aligning your goals with budgetary constraints and the need for clinical milestones creates a delicate balance. This article explored how to navigate these hurdles, make informed decisions, mitigate risks, and approach regulatory agencies strategically to achieve success.

Assessing Development Progress: As you review the existing development work, evaluating the stage at which the project stands is crucial. Determine the completeness and quality of the data generated so far. Identify any gaps in knowledge or potential risks associated with the compound or product. This assessment will guide your decision-making and prioritize the areas that require immediate attention.

Optimizing Resource Allocation: Balancing the desire to achieve specific milestones and the need to manage costs can be challenging. Prioritize activities and allocate resources strategically based on the critical path. Identify the key activities directly impacting achieving clinical milestones and focus resources on those. Consider outsourcing non-core activities or establishing partnerships to optimize cost-efficiency while maintaining quality.

Risk Management and Mitigation: Product development inherently involves risks. Identify and evaluate risks associated with manufacturing, formulation, regulatory compliance, and clinical outcomes. Establish a comprehensive risk management plan to address and mitigate these risks proactively. Communicate potential risks to the Board of Directors, highlighting the importance of investing in risk mitigation strategies to ensure long-term success.

Regulatory Strategy and Planning: Developing a well-defined regulatory strategy early in the process is essential. Engage regulatory experts to help design a roadmap tailored to your product and target markets. Determine the optimal regulatory pathway, considering the product’s novelty, therapeutic area, and potential expedited development programs. Strategically plan interactions with regulatory agencies to seek guidance and ensure a favorable review process.

Building a Robust CMC Package: A concise and well-prepared CMC package is vital for regulatory submissions. Establish a CMC development plan that addresses key CMC aspects such as drug substance characterization, manufacturing process, formulation development, and analytical methods. Focus on generating high-quality data, adhering to regulatory guidelines, and considering long-term scalability and GMP compliance. Develop a comprehensive understanding of the critical quality attributes that impact product performance and safety.

Engaging Regulatory Agencies: Timing is crucial when engaging regulatory agencies. Initiate early interactions to seek guidance, clarify regulatory requirements, and confirm the adequacy of your development plans. Leverage scientific advice meetings and pre-submission interactions to address questions, refine your strategy, and ensure alignment with regulatory expectations. Approach these interactions with a well-prepared package, emphasizing your product’s potential public health impact and patient benefit.

 

Conclusion

Embarking on developing a compound or in-licensed product requires careful planning, strategic decision-making, and effective regulatory management. Your biotech company can navigate the complexities of product development by assessing development progress, optimizing resource allocation, managing risks, and strategically engaging regulatory agencies. Balancing funding limitations with the need to achieve clinical milestones and regulatory compliance is a delicate dance. Still, with a clear focus, comprehensive planning, and collaboration with experts, you can pave the way for success in bringing your innovative product to market.

For academic discoveries to progress successfully into clinical trials, it is essential to consider the IND requirements and bridge the gap between academic research and clinical development. Robust preclinical studies, alignment of CMC development with IND requirements, development of analytical methods, stability evaluation, early collaboration with regulatory experts, and establishing quality control practices within academia are critical steps. By addressing these considerations, academic researchers can significantly enhance the readiness of their product as it enters the IND phase.

 

By embracing these guidelines, companies can enhance their understanding of critical quality attributes, implement effective risk management strategies, establish robust quality systems, and manage post-approval changes in a harmonized manner. Staying informed about regulatory changes, engaging in early interactions with regulatory authorities, and leveraging regulatory expertise and partnerships are also crucial elements of a successful CMC regulatory strategy. Ultimately, by adopting a proactive and globally-minded approach, pharmaceutical companies can navigate the complexities of CMC regulations, ensure compliance, and achieve market access in various regions worldwide.

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