long-term-stability-testing-for-bispecific-antibodies

Beyond the Ticking Clock: Advancing Long-Term Stability Testing for Bispecific Antibodies

Beyond the Ticking Clock: Advancing Long-Term Stability Testing for Bispecific Antibodies

Beyond the Ticking Clock: Advancing Long-Term Stability Testing for Bispecific Antibodies

06.08.2025

6

Minutes

Leukocare Editorial Team

06.08.2025

6

Minutes

Leukocare Editorial Team

Directors of CMC and Drug Product Development know bispecific antibody stability is tough. These complex molecules are prone to issues, but ensuring long-term stability is crucial for getting them safely to patients. Discover how a robust stability program provides the data you need for confident decisions.

Menu

Beyond the ticking clock: A new look at long-term stability for bispecific antibodies

1. Current Situation

2. Typical Market Trends

3. Current Challenges and How They Are Solved

4. How Leukocare Can Support These Challenges

5. Value Provided to Customers

6. FAQ

Beyond the ticking clock: A new look at long-term stability for bispecific antibodies

If you're a Director of CMC or Drug Product Development, you know 'stability' is everything. For bispecific antibodies (BsAbs), that's always tough. These complex molecules are opening new frontiers in medicine, but their intricate structures make them prone to issues that can derail a development program. Making sure a bispecific stays stable throughout its shelf life isn't just a regulatory checkbox; it's key to getting it safely from the lab to patients [1, 19].

This isn't just about ticking boxes. It's about really understanding the molecule so it's safe, effective, and easy to make. A good stability program gives you the data you need to make confident decisions at every step.

1. Current Situation

Bispecific antibodies are a rapidly growing class of therapeutics, with nearly 160 in clinical trials. Their ability to engage two different targets simultaneously offers new ways to fight complex diseases, particularly cancer [3]. But this complexity also creates their biggest development challenge [4, 5, 13]. Compared to traditional monoclonal antibodies (mAbs), BsAbs are often harder to manufacture and more susceptible to aggregation, fragmentation, and loss of function [6].

These molecules aren't just two mAbs stuck together [7, 8, 11]. Their unique architectures, whether IgG-like or smaller fragments, create novel stability problems. Incorrect chain association, for example, can lead to a mixture of inactive or even harmful variants [5]. This makes chemistry, manufacturing, and controls (CMC) tricky. You need really strong analytical methods to keep product quality and consistency solid [10, 21].

2. Typical Market Trends

The bispecific antibody market is expanding quickly, with projections suggesting it could reach over $220 billion by 2032 [11, 7]. This growth is fueled by the demand for more effective cancer therapies and advancements in antibody engineering [12].

Here are some main trends shaping things for CMC leaders [13, 4]:

  • Accelerated Timelines: Many bispecifics are on fast-track development paths, putting immense pressure on teams to deliver a robust data package for IND and BLA filings in record time. Some programs aim for DNA to IND in as little as 13 months.

  • Outsourcing Models: Virtual and small biotech companies are major innovators in this space. Lacking internal labs, they rely on a network of external partners for everything from cell line development to formulation. This makes clear communication and shared strategic vision between partners essential [14].

  • Increasing Complexity: The field is moving beyond simple bispecifics to even more complex multi-specific formats, engaging natural killer (NK) cells, macrophages, and other immune effectors. This introduces new stability and manufacturing hurdles that require specialized knowledge [15].

  • Focus on Subcutaneous Delivery: To improve patient convenience, many companies are aiming for high-concentration liquid formulations suitable for subcutaneous injection. This presents significant challenges related to viscosity and aggregation [16].

3. Current Challenges and How They Are Solved

For a CMC director, a bispecific's theoretical promise doesn't mean much without a clear way to make it a stable, manufacturable drug [17, 18]. The usual stability challenges get even bigger with bispecifics:

  • Aggregation: BsAbs often have more exposed hydrophobic regions, making them prone to clumping together. This can reduce efficacy and, more critically, increase the risk of an immunogenic reaction.

  • Fragmentation: The molecule can break apart, leading to a loss of binding and function [1, 19].

  • Chemical Degradation: Oxidation and deamidation can alter the molecule's structure and compromise its activity [8].

  • Loss of Potency: A stable formulation must preserve the binding affinity and function of both arms of the antibody [20].

Traditionally, addressing these issues involves a long and expensive process of trial-and-error. Teams screen dozens of buffer conditions and excipients in long-term, real-time studies that can take years. While necessary, this approach is slow and consumes large amounts of expensive drug substance, a scarce resource in early development [22, 31, 32].

4. How Leukocare Can Support These Challenges

The conventional approach to formulation is often reactive. A more strategic, data-driven method can identify and solve stability issues much earlier. This is where a specialized partner can shift the dynamic from simply executing tasks to co-piloting development strategy.

For a fast-track virtual biotech, the goal is a clean path to BLA. They have a promising molecule but no internal lab. They face extreme time pressure and are skeptical of vendors who lack strategic depth. A partner that offers a smart formulation platform using predictive, AI-based modeling can provide data-driven insights quickly. This allows for parallel optimization of the cell line, process, and formulation, saving precious time and material.

A mid-size biotech may have an internal drug product team but hits bandwidth limits with tricky projects or new modalities. They have existing service partners but find them too rigid. Onboarding a new vendor through procurement is a slow process. For them, the right partner can enter via a specific challenge, like lyostability or a new modality. By delivering results on a single, complex pilot project, the partner can prove its value and build the trust needed to scale the relationship. It's about augmenting the internal team, not replacing it.

For a large pharma company tackling a new modality like viral vectors or RNA, the challenge is different. They have internal resources but may lack specific experience, leading to uncertainty. They need specific insights, case studies, and a true sparring partner to help navigate internal discussions and make sound regulatory decisions. They are not looking for a templated solution but for deep technical understanding that de-risks their entry into a new area.

Finally, for a CDMO that wants to offer a full-service package without building an internal formulation team, a neutral, external partner is key. The CDMO needs a collaborator that can work silently in the background, delivering reliable formulation data without creating coordination burdens or attempting to poach the client relationship.

5. Value Provided to Customers

In the end, formulation development is all about making things better by cutting down risk and speeding up timelines. The right partner gives you more than just data; they give you confidence.

  • Speed and Efficiency: By using predictive models based on data from accelerated stability studies, it's possible to get reliable long-term stability insights in weeks, not years. This provides a faster, more direct path to a stable formulation ready for Phase I [22, 31, 32].

  • Data-Driven Decisions: A robust data package strengthens investor conversations and regulatory filings. It demonstrates a deep understanding of the molecule and its critical quality attributes, building confidence with stakeholders and health authorities.

  • De-risking Development: Identifying and solving stability problems early prevents costly delays and failures down the line [24]. A formulation designed by science and guided by data is built for regulatory success [25, 26, 27].

  • Strategic Partnership: The value lies not just in executing work but in thinking proactively. A true partner acts as a co-strategist, bringing an independent opinion and scientific excellence to the table to help navigate the complex path of drug development.

6. FAQ

How early should we start thinking about formulation for a bispecific?
Early. Manufacturability and stability should be considered during the discovery stage. Initial assessments can help select the best candidates and de-risk the program before significant resources are committed [19, 28].

What are the key differences in stability testing for a BsAb versus a mAb?
The main difference is complexity. You need to confirm that both binding domains remain active and that the molecule doesn’t mispair or aggregate in unique ways. This often requires more sophisticated analytical methods and a deeper understanding of the specific failure modes of your molecular format [10, 21].

Can predictive modeling truly reduce the need for long-term stability studies [29, 30]?
Predictive modeling, especially when based on robust accelerated data, can provide highly accurate forecasts of long-term stability. While not a complete replacement for real-time data required by regulators, it allows you to select the most promising formulation candidates much faster, focusing your long-term studies on the formulations most likely to succeed [22, 31, 32].

How much material is needed for an early, data-driven formulation approach?
Modern, high-throughput analytics allow for comprehensive screening with much less material than traditional methods. This is a significant advantage in early development when the drug substance is limited and expensive.

How do you ensure the formulation is suitable for high-concentration applications?
Developing a high-concentration formulation requires a specific focus on viscosity and colloidal stability. Predictive tools and targeted excipient screening can identify formulation conditions that keep the molecule soluble and deliverable at concentrations required for subcutaneous injection [18].

Literature

  1. nih.gov

  2. tandfonline.com

  3. oup.com

  4. stellarmr.com

  5. nih.gov

  6. leadventgrp.com

  7. bioprocessonline.com

  8. nih.gov

  9. tandfonline.com

  10. nih.gov

  11. filab.fr

  12. globenewswire.com

  13. straitsresearch.com

  14. lonza.com

  15. drugtargetreview.com

  16. pharmasalmanac.com

  17. youtube.com

  18. nih.gov

  19. oup.com

  20. hilarispublisher.com

  21. parexel.com

  22. nih.gov

  23. nih.gov

  24. casss.org

  25. pharmtech.com

  26. biopharminternational.com

  27. nih.gov

  28. nih.gov

  29. revvity.com

  30. researchgate.net

  31. casss.org

  32. stabilitystudies.in

Further Articles

Further Articles

Further Articles