News & Insights

The Chemistry, Manufacturing, and Controls (CMC) section of a cell therapy Investigational New Drug (IND) application is crucial in demonstrating to the FDA and other regulatory bodies that an investigational product can be consistently manufactured while meeting safety, identity, quality, purity, and potency standards. This section provides detailed information on composition, manufacturing processes, quality control measures, and stability, ensuring that the drug product is suitable for clinical investigation.
While the CMC section of a cell therapy’s IND application will obviously contain information specific to cell therapies and the therapeutic in question, the spirit and trajectory of the requirements are similar to those of any other biotherapeutic filing. However, understanding these established requirements and the most important steps developers can take when approaching the IND application process for a novel therapeutic, such as a cell therapy, is crucial.
New Therapeutic, Familiar CMC Requirements
Any scientist who is knowledgeable about the biotherapeutics regulatory filing and review process will be familiar with the cell therapy CMC requirements for IND filing.
Product Description
Reviewers first want to know exactly what you propose to administer to patients. Clearly define the cell therapy product, including:
- Cell Source and Type: Specify whether the cells are autologous or allogeneic, and detail the tissue origin and cell lineage.
- Genetic Modifications: Describe any genetic alterations, such as transduction with viral vectors or gene edits via endonuclease, and provide information on the vector construct and/or gene editing reagents (e.g., mRNA, lipid nanoparticles, etc).
- Mechanism of Action: Explain how the product is expected to work therapeutically, linking this narrative to measurable potency assays.
Manufacturing Process and Controls
A high-level flow diagram is not enough. Instead, the FDA expects a step-by-step process development narrative that tracks the cells from collection through cryopreservation.
- Process Steps: Outline each step in cell therapy manufacturing, from cell collection to final product formulation.
- Critical Process Parameters (CPPs): Identify parameters that must be controlled to ensure product quality, such as culture conditions and transduction efficiency.
- In-Process Controls: Describe tests and control technologies that ensure drug product consistency.
- Process Validation: Even in early-phase trials, provide a rationale for process parameters and controls, and outline plans for future validation studies.
Materials and Reagents
Detail all materials used in manufacturing, including:
- Raw Materials: Reagents, media, and supplements, specifying their grade, source, and vendor qualifications.
- Animal-Derived Components: If used, information on sourcing and testing for transmissible spongiform encephalopathies (TSEs), bovine spongiform encephalopathy (BSE), and other adventitious agents.
- Clearance of Residuals: Describe methods used to remove or reduce residual reagents to acceptable levels in the final product.
Analytical Methods and Release Specifications
An IND must tell a coherent, data-driven story that assures regulators that every lot released will be safe, pure, potent, and consistent. FDA reviewers expect a phase-appropriate but fully referenced description of each analytical method, its qualification status, and the numerical release specifications that trigger lot acceptance or rejection.
- Identity Testing: Confirm the product’s identity using appropriate markers or assays.
- Purity and Impurity Testing: Assess the presence of unwanted cell types, residual reagents, or contaminants.
- Potency Assays: Develop assays that measure the biological activity relevant to the product’s intended effect.
- Stability-Indicating Assays: Summarize real-time and accelerated studies measuring potency, viability, and container-closure integrity.
- Viability and Cell Count: Determine the percentage of viable cells and total cell number in the final product.
- Safety Testing: Include sterility, endotoxin levels, mycoplasma testing, and adventitious agent testing (if applicable).
Ensure that analytical methods are qualified for their intended use, with plans for full validation in later stages of development.
Cell Banking
If your therapy uses a master cell bank (MCB) and a working cell bank (WCB), provide complete production records and the full panel of sterility, mycoplasma, adventitious virus, identity, and stability data. For autologous therapies, provide a traceability package that links each donor lot to its corresponding vial of finished product. Any qualification testing performed outside a GMP suite should be justified because the FDA and other regulatory agencies now scrutinize the bank’s production environment as closely as the test results themselves.
Facilities and Equipment
The CMC narrative must include cleanroom classifications, personnel and material flows, HVAC zoning, and segregated areas for viral manipulations. Support the description with environmental monitoring trends and calibration schedules for critical equipment, particularly bioreactors, centrifuges, automated washers, and liquid nitrogen freezers. The aim is to demonstrate that your quality system controls the physical space and the instruments that come into contact with the product.
Stability and Storage
Provide real-time and accelerated stability data on the cryopreserved product and, where applicable, on the post-thaw “hold” interval at room or refrigerated temperature. Additionally, demonstrate that the packaging maintains sterility and product quality throughout storage and handling.
Chain of Identity and Chain of Custody
For autologous therapies, the vein-to-vein trail must be unbroken. The IND should describe the validated system that tracks donor, collection bag, in-process pool, cryovial, shipper, and infusion bag IDs, as well as the procedures for handling barcode mismatches or temperature excursions.
Comparability and Change Management
Process evolution is inevitable and necessary, but sponsors must present a prospective comparability protocol—analytical and functional bridges to support future changes to raw materials, equipment, or sites. If you have already modified the process, include side-by-side comparability data so reviewers can assess the impact on safety and potency without delaying the IND due to a significant change hold.
Quality Systems
Summarize the quality management systems in place, including confirmation of GMP compliance as outlined in 21 CFR Parts 210 and 211, as well as quality assurance and control measures throughout the processing steps. Finally, ensure that all processes, tests, and deviations are thoroughly documented.
Pre-IND Meetings are the Most Effective Step for Successful CMC Strategies
Early-stage sponsors often treat pre-IND meetings as a forum to confirm that their toxicology package and first-in-human trial design are on track; however, CMC needs to be meaningfully included in these initial discussions. Unlike more mature modalities, cell therapy approaches and technologies are evolving quickly. The regulatory framework is still catching up, and given the rapid pace of advancements, it will likely not be considered mature anytime soon.
Pre-IND dialogue is the first—and sometimes the only—opportunity to secure FDA buy-in on a phase-appropriate CMC strategy, which can help avoid development delays.
CMC and the Pre-IND Agenda
FDA’s Office of Therapeutic Products (OTP) expressly invites CMC questions during Type B Pre-IND meetings when the product or process is complex. Cell therapies routinely involve viral vectors, genome-editing components, or other cell manipulations that sit well outside conventional paradigms, and certainly fit this bill.
Regulatory authorities now expect a much greater understanding of products and processes across the board – the “Wild West” days are over. However, because cell therapy products, processes, and technologies are advancing so quickly, regulators often need to be educated and agree on approaches to avoid delayed first-in-human dosing.
4 Ways to Make the Most of Pre-IND CMC Discussions
- Prioritize High-Risk Elements: Raise pointed questions on assay qualification, raw material sourcing, and comparability protocols—the very topics the FDA flags most often in cell therapy holds.
- Present Data, Not Concepts: Summaries of pilot runs, draft specifications, and preliminary stability curves help reviewers gauge whether your controls are commensurate with patient risk.
- Map Change-Control Pathways: Explain how inevitable upgrades (vector swap, closed-system adoption) will be bridged so that regulators are comfortable approving the process and approach evolution, not just an initial filing.
- Confirm Phase-Appropriate Expectations: Use FDA feedback to gain consensus on what is “good enough” for Phase 1 and when full validation will be required, preventing moving targets in later stages.
Kincell Supports Successful INDs
For cell and gene therapy developers, postponing CMC dialogue until after IND submission is a decided gamble, but often, cell therapy startup innovators are less experienced with the regulatory approval process and lack relationships with regulators.
Kincell can help. As a cell therapy leader with deep scientific expertise, Kincell is an extension of your team, offering proactive collaboration, transparent communication, and tailored support that helps you file successfully and keep your program on track. Contact us to discuss your program.