Today we're diving into something that could literally save your company from becoming another biotech statistic.
Over 40% of biotherapeutic failures during clinical development stem from stability problems—and most trace back to protein aggregation that could have been prevented. We'll expose the hidden manufacturing crisis that derails promising biologics programs and delivers the systematic Quality by Design framework that elite biotech companies use to build quality into every process step.
This concept is discussed in greater detail in the Smart Biotech Scientist Podcast, hosted by David Brühlmann, founder of Brühlmann Consulting.
The Foundation Crisis - Why Most Biotech Companies Fail at Manufacturing
Let me tell you about a real case that shook the oncology world. Techno Pharma Sphere highlighted a recent monoclonal antibody drug development case study.
Clinical trials were going well - patients were responding, safety looked good, investors were excited. Everything was on track for a successful Phase III launch.
Then came the scale-up. What started as minor batch inconsistencies quickly revealed a catastrophic problem: protein aggregation. Despite extensive upstream optimization, their carefully engineered antibody was forming aggregates during shipping and storage at the very temperatures they'd specified for distribution.
The company faced an impossible choice. They could push forward and risk regulatory rejection, or halt everything. They chose to halt production, completely redesign their formulation, and submit entirely new data packages to regulators.
The cost? Nearly a year of delays. Production costs increased by over twenty-five percent. Their lead over competitors evaporated overnight. All because they'd never properly defined what "quality" meant at commercial scale.
This isn't an isolated incident. Industry analysis shows that over forty percent of biotherapeutic drug failures during clinical development are linked to stability issues, with protein aggregation being the primary culprit.
Sound familiar? If you're sitting there thinking "that could be us", then this episode could change everything for your company.
The Solution: Quality by Design Foundation
What if I told you there's a way to build quality in from the start? That's exactly what Quality by Design - or QbD - delivers.
QbD is a systematic approach to pharmaceutical development that begins with predefined objectives and emphasizes product and process understanding based on sound science and quality risk management. As the ICH Q8(R2) guidance states, it's fundamentally different from traditional approaches because it focuses on understanding your product and process deeply rather than just following established recipes.
The FDA puts it simply: "Quality by Design builds quality into the product instead of testing it." But here's what makes it powerful for biologics specifically - the NIH guidance reminds us that "for biologics, the process is the product." This means any changes in manufacturing can fundamentally change your biological molecule. QbD helps you understand and control these relationships from day one.
QbD rests on three fundamental pillars.
- First, product understanding - knowing exactly what you're building.
- Second, quality requirements - identifying what matters most for patient safety and efficacy.
- And third, process control - understanding how to make your product consistently.
The business case is compelling. QbD delivers predictable manufacturing, which reduces your risk. It builds regulatory confidence, which accelerates approval timelines. Bottom line? It's a faster, cheaper path to market.
Pillar One: Your Product Blueprint
Every successful product starts with knowing exactly what you're building. That's where the Quality Target Product Profile - or QTPP - comes in.
Think of your QTPP as your product's blueprint for success. It links patient needs directly to manufacturing specifications. The key insight? Start with the patient and work backward.
Your QTPP should define four critical components.
- First, route of administration - how will patients actually receive this therapy?
- Second, dosing regimen - how often and at what strength?
- Third, stability requirements - what are your storage and shipping constraints?
- Fourth, quality expectations - what purity and potency targets must you hit?
Let me give you a concrete example from the A-Mab case study - the industry's gold standard example developed jointly by regulators and industry. This hypothetical monoclonal antibody program demonstrates how a clear QTPP drives every manufacturing decision. Their QTPP specified subcutaneous administration for patient convenience, which immediately drove formulation requirements for high concentration and low viscosity. The stability requirement of 24 months at 2-8°C shaped their packaging and distribution strategy. Most importantly, their quality expectations - including aggregation below 2% and purity above 95% - became the foundation for all subsequent CQA definition.
Here's how to define your own QTPP.
- Start with the patient perspective - what do they actually need from your therapy?
- Consider your clinical requirements - what safety and efficacy targets must you achieve?
- Factor in commercial reality - how will market access and competition affect your positioning?
- And validate manufacturing feasibility - can you actually produce what you're promising?
Pillar Two: What Really Matters
Critical Quality Attributes (CQAs) are quality attributes that directly impact patient safety or efficacy. The ICH Q8 guideline defines them as properties that must be controlled to ensure quality. The focus principle is simple: identify the critical few, not the many.
Not everything that can be measured should be measured. That's the fundamental principle behind CQAs.
CQAs typically fall into three categories.
- Safety-critical attributes include things like aggregates, which create immunogenicity risk, and endotoxins or bioburden that could harm patients.
- Efficacy-critical attributes cover potency, binding affinity, and biological activity - the properties that make your drug work.
- Stability-critical attributes include fragmentation, oxidation, and charge variants that affect shelf life and performance.
How do you identify your CQAs?
- Start with a risk assessment - what could actually hurt patients?
- Consider your mechanism of action - how does your drug deliver its therapeutic effect?
- Leverage prior knowledge - what do similar approved products control?
- And validate analytical capability - can you measure these attributes reliably and consistently?
Let me give you a concrete example from the industry's most celebrated QbD success story. Roche/Genentech's obinutuzumab (Gazyva) became the first monoclonal antibody approved by the FDA using comprehensive QbD principles in 2013, including an approved design space. This breakthrough came after learning from their earlier Perjeta submission, which was rejected as a full QbD filing.
Here's how they systematically identified their CQAs. During the evaluation process, Roche discovered that ADCC (antibody-dependent cellular cytotoxicity) had initially not been identified as a CQA based on characterization studies. However, the regulatory review process highlighted ADCC's potential impact on safety, leading to the inclusion of fucosylation patterns as a critical quality attribute, since fucose levels strongly correlate with ADCC activity. The predominant afucosylated form, G0-F, showed strong correlation with ADCC and was added to their CQA list.
Their final CQA profile included several key attributes that directly impacted patient safety and efficacy.
- Aggregation levels were controlled below specific thresholds due to immunogenicity concerns.
- The fucosylation pattern (specifically G0-F levels) became critical for controlling ADCC activity, which is particularly important for safety in oncology applications.
- They also monitored potency, purity, and specific charge variants that could affect therapeutic performance.
This systematic approach delivered transformative business results. According to Genentech, the comprehensive QbD approach provided unprecedented regulatory flexibility, though it required extensive justification and significantly increased the CMC section of their biologics license application. The enhanced process understanding allowed them to make manufacturing changes within their approved design space without additional regulatory approval, providing both operational flexibility and reduced time to market for process improvements.
Roche/Genentech has since applied these refined QbD tools to their entire biologics pipeline, creating significant efficiencies and resource savings during late-stage development while enabling post-licensure flexibility for production. Most importantly, their rigorous CQA identification process ensures that every quality attribute they control has a direct scientific link to patient safety or therapeutic efficacy.
This systematic approach aligns perfectly with ICH Q8(R2) guidance on pharmaceutical development, which emphasizes that quality should be built into products by design. The guidance specifically states that "the product should be designed to meet patients' needs" - exactly what the QTPP accomplishes.
Now, here's something critical that many founders miss: the FDA and EMA actually want to see QbD approaches in your submissions. They're not just checking boxes - they want evidence that you understand your product and process. Start engaging with agencies early through pre-IND meetings. Present your QTPP and initial CQA thinking. They'll guide you on what matters most for your specific molecule and indication.
For small companies, this early dialogue is gold. It prevents you from over-investing in the wrong areas and shows regulators you're thinking systematically about quality.
Your Next Step
Before your next team meeting, I want you to do one simple exercise. It's called the CQA Reality Check.
List every quality attribute you currently test for your product. Now ask yourself this binary question for each one: "If this attribute failed, would it hurt patients or impact efficacy?" If the answer is yes, it's a CQA candidate. If the answer is no, it's just nice to know.
Your goal is to narrow your list to five to eight true CQAs. Focus on what actually matters for patients, not what's easy to measure.
This single exercise will give you the foundation everything else builds on. You'll stop over-testing irrelevant parameters and start focusing your resources on what truly drives product quality.
The Process Breakthrough - From Chaos to Control
Previously, we covered the foundation crisis that kills forty percent of biotherapeutic programs during clinical development - companies that never properly defined what "quality" meant at commercial scale. Now, I'm going to show you exactly how to build the process side of Quality by Design to prevent those failures. By the end of this episode, you'll have the roadmap to go from manufacturing chaos to regulatory approval in record time.
When Good Processes Go Bad
Here's a scenario that keeps biotech CEOs awake at night. Your process works perfectly at two-liter bench scale. You're confident, investors are excited, everything's on track.
Then you hit your first fifty-liter pilot run. Suddenly, pH profiles look different, titer drops by twenty percent. You troubleshoot, make adjustments, try again. The two-hundred-liter production run brings new surprises - aggregation appears out of nowhere, charge variant patterns shift.
Your management team asks the question you dread: "How long to fix this?"
Your honest answer? "We don't really know. Could be three months, could be twelve."
Why does this keep happening to smart people? The physics change. Mass transfer, mixing efficiency, heat dissipation - everything behaves differently at scale. The biology changes too. Cell stress, shear sensitivity, oxygen limitations create new challenges. Even the chemistry changes with buffer gradients and temperature variations you never saw coming.
Most companies make a critical mistake. They confuse specifications with control. They focus only on final product testing - like trying to control your car by only looking at the destination. By the time you see a problem in your final product, it's too late to fix it.
What's missing is real-time process understanding, predictive capability, and risk mitigation that catches problems before they happen.
The Solution: Process Intelligence
Let's quickly recap the Quality by Design framework we established in our last episode. We start with your Quality Target Product Profile - your QTPP - which defines exactly what you're building based on patient needs. From there, we identify your Critical Quality Attributes - your CQAs - the specific product characteristics that directly impact patient safety and efficacy. Today we're focusing on the third pillar: how to control your manufacturing process to consistently deliver those CQAs.
Not all process variables are created equal. The Pareto Principle applies perfectly to bioprocessing - eighty percent of your quality issues come from twenty percent of your process variables. Quality by Design helps you find that critical twenty percent.
These are your Critical Process Parameters - or CPPs. The ICH Q8 guideline defines CPPs as process parameters whose variability has an impact on a Critical Quality Attribute and therefore should be monitored or controlled to ensure the process produces the desired quality.
Think of CPPs as the vital signs of your manufacturing process. Just like monitoring heart rate and blood pressure tells you about human health, monitoring CPPs tells you about process health.
Let me share a well-documented industry success story. Genentech's development of their monoclonal antibody platform faced exactly this challenge - mysterious batch-to-batch variability that threatened their entire program. Through systematic QbD implementation, they discovered that small variations in feeding strategies during cell culture had dramatic impacts on product quality consistency.
Their solution? They moved from fixed feeding schedules to metabolic state-based feeding control, monitoring glucose consumption rates and lactate production in real-time. The result was a significant improvement in process consistency and became the foundation for their platform approach that's now used across their entire portfolio.
CPPs typically fall into two categories.
- Upstream parameters include pH setpoint and control ranges, dissolved oxygen levels, feed rates and timing, and temperature profiles.
- Downstream parameters cover column loading density, buffer pH and conductivity, flow rates and residence times, plus pool criteria and hold times.
Your Manufacturing GPS
Now, let's explore the control strategy - your manufacturing GPS that guides your entire production process from raw materials to final product.
Think of control strategy as your GPS system for manufacturing. Your destination is your CQAs - where you need to be quality-wise. Your route is your process steps - how to get there. Real-time guidance comes from CPP monitoring - staying on track. And when things go off-course, you have process adjustments to recalculate your path.
Control strategy has three integrated layers:
- Input controls cover raw material specifications, cell bank characterization, and media and buffer quality standards.
- Process controls include real-time CPP monitoring, in-process testing and release criteria, plus environmental monitoring for bioburden and endotoxin.
- Output controls encompass product release specifications, stability monitoring programs, and innovative approaches like real-time release testing.
Real-time release testing represents a major innovation. Instead of waiting fourteen days for traditional release testing, companies can release product based on validated process data. Amgen has publicly shared how their implementation of real-time release testing achieved faster lot release and a cost reduction across multiple products in their biosimilar portfolio.
The key insight is moving from reactive to predictive control. Traditional approaches detect problems after they occur. Smart control strategies prevent problems before they happen.
Working Smarter, Not Harder
While Quality by Design provides the overarching framework for systematic pharmaceutical development, one of its most powerful implementation tools is the Design of Experiments (DoE) methodology, which enables scientists to efficiently explore and optimize complex manufacturing processes.
Traditional Design of Experiments approaches have serious limitations. One-factor-at-a-time testing is slow and misses critical interactions. Full factorial designs test everything but require hundreds of experiments. The problem? Complex optimization takes two-plus years using traditional methods.
But there's a revolution happening in bioprocess development. It's called hybrid modeling, and it's transforming how smart companies approach process optimization.
Hybrid modeling combines your existing biological knowledge with machine learning. Instead of treating your process like a black box, it uses your understanding of cell biology, biochemistry, and engineering as a foundation. Then machine learning fills in the gaps and finds hidden patterns you'd never discover through traditional experiments.
The results are remarkable. DataHow, a leading process analytics company, has demonstrated sixty to eighty percent reduction in required experiments across multiple pharmaceutical clients. Instead of two years and two hundred experiments, companies are achieving better optimization in four months with twenty-five strategic experiments.
Here's how it works in practice. Take CHO cell culture optimization. Traditional approaches test pH, temperature, dissolved oxygen, and feed rates one at a time, then try combinations. Hybrid modeling starts with your knowledge of cell metabolism, then uses machine learning to optimize the interactions iteratively. Each experiment makes the model smarter, guiding you toward optimal conditions faster than ever before.
For detailed case studies and published validation of these results, check out our previous episodes with DataHow's founders Fabian Feidl and Michael Sokolov, where we dive deep into the science behind these impressive numbers.
Episode 99: From Raw Data to Actionable Insights: Unlocking the Power of Process Models with Fabian Feidl - Part 1
Episode 100: From Raw Data to Actionable Insights: Unlocking the Power of Process Models with Fabian Feidl - Part 2
Episode 05: Hybrid Modeling: The Key to Smarter Bioprocessing with Michael Sokolov - Part 1
Episode 06: Hybrid Modeling: The Key to Smarter Bioprocessing with Michael Sokolov - Part 2
The business impact is transformative, especially for smaller companies where every experiment counts. Months instead of years for development. Fewer experiments mean lower costs and faster decisions. Most importantly, you discover optimal conditions you'd never test using traditional approaches because the model identifies non-obvious parameter combinations.
Building Your Control Strategy
Your control strategy transforms process understanding into operational reality. Think of it as translating scientific insights into manufacturing instructions that deliver consistent quality every time.
Effective control strategies specify exactly which parameters to monitor, what ranges are acceptable, how to respond when parameters drift, and when to investigate versus when to take immediate action. They include decision trees that eliminate guesswork from manufacturing operations.
The most sophisticated companies are implementing model-based control. Real-time data feeds into process models that predict quality outcomes before final testing. This enables proactive adjustments rather than reactive responses.
Consider feed control in cell culture. Traditional approaches use fixed schedules. Model-based approaches monitor glucose consumption, lactate production, and cell density to calculate optimal feed timing and composition in real time. The difference? Predictable performance instead of batch-to-batch variability.
Training is crucial for success. Operators need to understand not just what to do, but why they're doing it. When everyone understands the underlying science, they make better decisions when unexpected situations arise.
Your Process Assessment
Here's what you can do before your next meeting. Take one sheet of paper and map your current process from cell culture to final product. At each step, ask two critical questions.
First: "What could go wrong here that impacts product quality?"
Second: "Will this step work the same way at two hundred liters as it does at two liters?"
Circle the three most critical steps - these become your focus areas for control.
This exercise reveals your biggest risks immediately and identifies scale-up vulnerabilities before they bite you. The steps you circle become your foundation for CPP identification and control strategy development.
Takes five minutes, could save you millions.
The Bottom Line
Every major biotech company that's succeeded at scale has mastered these fundamentals. Quality by Design isn't just regulatory compliance - it's competitive advantage.
Companies using these approaches achieve predictable manufacturing, reduced development timelines, lower costs of goods sold, and regulatory confidence that accelerates approvals. Most importantly, they deliver medicines to patients faster and more reliably.
The question isn't whether you need Quality by Design. The question is whether you'll implement it before your competitors do.
Remember what we covered today.
- Critical Process Parameters help you focus on what really drives quality.
- Smart control strategies prevent problems rather than just detecting them.
- And hybrid modeling can reduce your experimental burden by up to eighty percent while delivering better results.
Your homework? Complete that five-minute process assessment. Map your process, identify risks, circle your critical steps. That simple exercise gives you the foundation for everything else.
Turn CMC Chaos Into Predictable Execution
Before we wrap up, let me address something I hear constantly from biotech leaders: "David, this all sounds great, but how do I make sure my team doesn't miss any critical steps on our path to IND filing?"
Here's the problem. Most biotech founders spend two to four weeks just mapping their CMC roadmap, trying to figure out all the interdependencies. Meanwhile, every successful monoclonal antibody program follows the same proven path to IND submission.
That's exactly why I developed the Notion CMC Dashboard. This comprehensive digital workspace eliminates the guesswork from your CMC journey. It's more flexible than Excel, easier to use than MS Project, and more collaborative than SharePoint.
This isn't just another planning template. It's your CMC Command Center that includes a complete proven monoclonal antibody CMC roadmap to IND submission, multiple dashboard views for tasks, key activities, timeline, and risk and delay alerts. It's ready to go with no setup required, yet fully customizable to your specific program needs.
The result? You go from CMC chaos to predictable execution. You hit your IND timeline with certainty instead of hoping you didn't miss a critical dependency.
Picture this: Kanban boards that show your task flow, timeline views that reveal critical path bottlenecks, and automated risk alerts that flag potential delays before they derail your submission. Your investors get transparency they've never seen before, your team stays aligned across functions, and you actually sleep at night knowing nothing's falling through the cracks.
This isn't just project management - it's your CMC command center that adapts as your program evolves. Think of it as the GPS for your entire development journey.
David Brühlmann is a strategic advisor who helps C-level biotech leaders reduce development and manufacturing costs to make life-saving therapies accessible to more patients worldwide.
He is also a biotech technology innovation coach, technology transfer leader, and host of the Smart Biotech Scientist podcast—the go-to podcast for biotech scientists who want to master biopharma CMC development and biomanufacturing.
Hear It From The Horse’s Mouth
Want to listen to the full interview? Go to Smart Biotech Scientist Podcast.
Want to hear more? Do visit the podcast page and check out other episodes.
Do you wish to simplify your biologics drug development project? Contact Us
🧬 Stop second-guessing your CMC strategy. Our fast-track CMC roadmap assessment identifies critical gaps that could derail your timelines and gives you the clarity to build a submission package that regulators approve. Secure your assessment at https://stan.store/SmartBiotech/p/get-cmc-clarity-in-1-week--investor-ready