how-to-extend-biologic-drug-shelf-life

Beyond the Two-Year Mark: A Practical Look at Extending Biologic Drug Shelf Life

Beyond the Two-Year Mark: A Practical Look at Extending Biologic Drug Shelf Life

Beyond the Two-Year Mark: A Practical Look at Extending Biologic Drug Shelf Life

11.09.2025

6

Minutes

Leukocare Editorial Team

11.09.2025

6

Minutes

Leukocare Editorial Team

As a CMC leader, balancing complex biologic development with urgent market needs makes achieving a long shelf life challenging. A longer shelf life offers a strategic advantage, impacting supply chains and patient access. Discover practical strategies to extend biologic drug shelf life effectively.

Menu

Beyond the Two-Year Mark: A Practical Look at Extending Biologic Drug Shelf Life

FAQ

1. Current Situation

2. Typical Market Trends

4. How Leukocare Can Support These Challenges

5. Value Provided to Customers

Beyond the Two-Year Mark: A Practical Look at Extending Biologic Drug Shelf Life

As a leader in CMC or Drug Product Development, you're balancing a lot. You're pushing complex molecules through the pipeline at an accelerated pace while ensuring the final product is stable, safe, and effective. Shelf life is a key part of that puzzle. A longer shelf life isn't just a technical achievement; it's a strategic advantage that impacts supply chains, market access, and patient care. Getting it right is rarely straightforward.

1. Current Situation

There's a lot of pressure. Your team develops formulations for increasingly complex biologics: monoclonal antibodies, viral vectors, RNA therapies, and antibody-drug conjugates. These molecules are sensitive to their environment by nature [1]. Temperature shifts, pH changes, and even physical stress can lead to aggregation, degradation, and loss of potency [2].

The traditional approach to stability testing, while essential for regulatory approval, is a waiting game [3]. Real-time, long-term studies are the gold standard, but they take years; a timeline that often conflicts with the urgent need to get new medicines to patients [1]. This forces teams to make critical formulation decisions early, often with incomplete data, introducing risk that can surface late in development [5].

2. Typical Market Trends

Market trends are making shelf life more critical.

  • The Rise of High-Concentration Formulations: The industry is moving toward subcutaneous (SC) self-administration to improve patient convenience. This requires developing high-concentration formulations, often exceeding 100 mg/mL, which can increase challenges like high viscosity and a greater tendency for proteins to aggregate [6, 8].

  • Global Supply Chains: A longer shelf life, ideally 24 months or more, is necessary for managing global logistics [6, 8]. It provides a buffer for shipping, distribution, and storage in different climate zones, reducing the risk of product loss [9].

  • Complex New Modalities: The pipeline is no longer dominated by standard mAbs. Newer modalities like viral vectors and cell therapies have unique instability issues that require specialized formulation strategies to ensure they remain viable until administration.

3. Current Challenges and How They Are Solved

Achieving a long shelf life means tackling several basic stability challenges directly. Biologics can degrade through physical pathways like aggregation or chemical pathways like oxidation.

One of the first big decisions is choosing between a liquid formulation or a lyophilized (freeze-dried) powder [11]. Lyophilization removes water, which generally leads to a more stable product with a longer shelf life, but it adds complexity and cost to manufacturing and requires reconstitution before use. Liquid formulations are convenient but often demand strict cold-chain storage [12, 13].

To stabilize the molecule, formulation scientists use excipients: inactive ingredients that protect the active protein [9].

  • Buffers are used to maintain the optimal pH [14].

  • Sugars like sucrose and trehalose can help protect the protein's structure, especially during freeze-drying.

  • Surfactants such as polysorbates prevent proteins from sticking to surfaces or aggregating at interfaces [15].

  • Amino acids can be used to reduce aggregation or viscosity [14].

The traditional way to find the right mix is through extensive screening studies, which take a lot of time and expensive drug substance. A more modern approach involves using Design of Experiments (DoE) to more efficiently map the formulation landscape. This statistical method helps identify the most important factors and their interactions, but it still requires a considerable amount of lab work. For a deeper dive into modern approaches, check out our article on how AI is accelerating preformulation studies.

4. How Leukocare Can Support These Challenges

The core challenge isn't just finding a stable formulation; it's finding it quickly and with great confidence. A data-centric approach changes everything here. Waiting years for real-time stability data to confirm your formulation choices is no longer a viable strategy in a competitive market.

At Leukocare, we pair our formulation expertise with advanced data modeling. Our platform uses AI to analyze how a molecule behaves and to predict its long-term stability from short-term data. This is about more than just fitting data to a curve; it's about building a kinetic model that reflects the specific degradation pathways of your molecule [1].

This method allows us to [17]:

  • Screen a wider design space: We can explore more formulation possibilities computationally, focusing lab work on the most promising candidates.

  • Reduce material needs: By minimizing extensive physical screening, we preserve your valuable drug substance.

  • Provide early insights: You get a reliable forecast of your product's shelf life months, not years, into development. This helps reduce risks and build a stronger CMC package for regulatory filings. For more on this, our post on the future of shelf life prediction models offers further context.

This combination of lab science and data science provides a clearer, faster path to a stable, commercially viable formulation.

5. Value Provided to Customers

Working with a strategic partner for formulation development provides more than just a data package. For a Director in CMC, it offers clear benefits that ease your biggest pressures:

  • Accelerated Timelines: Getting reliable stability predictions early helps you meet aggressive project deadlines and move toward IND and BLA filings faster.

  • Reduced Risk: Making formulation decisions based on robust predictive data lowers the risk of late-stage failures, which can be incredibly costly [1]. A stable formulation is also a key component of a successful biologic drug life cycle management strategy [17].

  • A Stronger Case for Regulators: A stability package supported by both real-world data and a scientifically sound predictive model gives regulatory agencies like the FDA and EMA greater confidence in your proposed shelf life.

  • Making Informed Decisions: With a clear understanding of your molecule’s stability profile, you can make better strategic decisions, from selecting the final container closure to planning your supply chain [18, 19].

A well-designed formulation strategy turns stability from a roadblock into a strategic asset.

FAQ

1. How early in development should we focus on optimizing for a long shelf life?
As early as possible. Initial formulation work during preclinical stages should aim to find conditions that provide basic stability. By Phase 1, you should be actively working on a formulation that can support your target shelf life for commercial launch. Early investment prevents costly reformulation work later on.

2. Is lyophilization always the best option for achieving a long shelf life?
Not necessarily. While lyophilization often provides superior stability, especially for highly sensitive molecules, it adds manufacturing complexity and cost. A stable liquid formulation is often preferred for convenience if it can meet the target shelf life of 24 months or more with refrigerated storage [20]. The choice depends on the molecule's inherent stability, the target product profile, and the commercial strategy [9].

3. How do predictive models improve upon standard accelerated stability studies?
Standard accelerated studies show if a product passes or fails at specific high-stress conditions. A kinetic model uses the rate of degradation from those studies to build a predictive tool. This lets you forecast stability at intended storage conditions and even predict the impact of temperature excursions during shipping, providing a much deeper understanding of your product's behavior [1].

4. Can these predictive approaches handle novel modalities with unique degradation pathways [17]?
Yes, but it requires a tailored approach. A one-size-fits-all model won't work for a viral vector or an mRNA vaccine. Advanced modeling's strength, including AI solutions for bioprocess and formulation, is the ability to build custom models that account for the specific ways these complex molecules degrade. This ensures the prediction is scientifically relevant and defensible.

Literature

  1. leukocare.com

  2. bioprocessonline.com

  3. mdpi.com

  4. ascendiacdmo.com

  5. nih.gov

  6. westpharma.com

  7. pharmaceutical-technology.com

  8. pharmasalmanac.com

  9. stabilitystudies.in

  10. bioprocessonline.com

  11. nih.gov

  12. pharmasalmanac.com

  13. lyophilizationcore.com

  14. nanoscience.com

  15. jmchemsci.com

  16. pharmasalmanac.com

  17. mdpi.com

  18. bdo.com

  19. stabilitystudies.in

  20. idt-biologika.com

Further Articles

Further Articles

Further Articles