PDA Letter Article

Bridging the Gap to Patients Overcoming the Fill Finish Complexities of Tomorrow’s Therapies

Tommy Schornak, Kindeva

Sponsored by Kindeva

Kindeva company logoFoundational shifts in healthcare, including the increasing prevalence of chronic diseases, are fueling a period of unprecedented expansion in the sterile fill finish market.

Valued at approximately $15.4 billion in 2023, the sector is projected to grow to $33.5 billion by 2032 (1). The dramatic rise in demand for patient-centric treatments such as glucagon-like peptide-1 (GLP-1) receptor agonists is a clear indicator of this new reality. This specific surge is part of a much broader trend: a pharmaceutical pipeline where nearly two-thirds of all therapies now require injectable delivery.

However, this growth is not a simple matter of scaling existing capacity. It is creating a convergence of technical, regulatory and strategic challenges that demand a new and more integrated approach to manufacturing. The therapies themselves are more complex. Regulatory standards for sterility are more stringent. The nature of the relationship between drug innovators and their manufacturing partners is fundamentally changing. This new landscape creates a clear opportunity for partners who can integrate adaptability, deep technical expertise and true partnership into a forward-thinking strategy that delivers value far beyond the production line.

The New Reality of Product and Process Complexity

One of the most defining features of today’s market is the sheer diversity of products requiring sterile manufacturing. The industry is essentially operating at two speeds simultaneously:

High-volume commercial therapies: At one end of the spectrum are high-production-volume commercial products like GLP-1 therapies, which require high-speed, high-throughput capacity and dedicated supply chains to meet global demand.

High-value targeted therapies: At the other end are highly targeted therapies for oncology, rare diseases or personalized medicine. These applications depend on smaller batch sizes, highly specialized handling protocols and accelerated production schedules to serve specific patient populations.

This diversification is most apparent in the shift toward patient-centric delivery formats. The move away from traditional vials and toward devices like prefilled syringes, cartridges for autoinjectors and microneedle array patches adds a significant layer of operational complexity. Each configuration demands a unique manufacturing approach, impacting everything from the initial filling and inspection to the final packaging. These new formats also raise the bar for critical quality attributes, placing a greater emphasis on ensuring robust container closure integrity, guaranteeing precise dose accuracy and protecting user safety.

Many legacy fill finish lines, optimized for the simplicity of vial production, are not equipped to handle this variety. Meeting this challenge requires investment in modular or combinatory line configurations that can accommodate a broader range of container types, batch sizes and fill volumes while maintaining both throughput and sterility assurance. Designing for this adaptability from the outset is critical to future-proofing infrastructure and ensuring a manufacturer can respond to the next modality shift with minimal disruption.

Beyond delivery formats, the pharmaceutical pipeline features a growing number of sophisticated therapeutic products, including advanced immunotherapies, cell and gene therapies and mRNA vaccines. These biologics present a distinct set of manufacturing challenges that require specialized solutions:

Significant financial risk: A primary concern is the high cost of the drug substance itself. This makes minimizing line losses and preventing batch failures a critical financial imperative, as even minor deviations can result in the loss of millions of dollars of valuable product.

Sensitive product handling: Many advanced therapies are inherently sensitive. Temperature sensitivity demands rigorous cold chain management throughout the entire manufacturing and distribution process. High viscosity or shear sensitivity requires specialized filling equipment and gentle handling techniques to prevent aggregation or degradation. For example, highly viscous formulations can be challenging to pump accurately, risking dose variation (2), while shear-sensitive proteins can be damaged during standard filtration or filling processes, potentially leading to aggregation that impacts both product efficacy and patient safety.

Meeting these complex demands effectively requires specialized facilities and tailored formulation suites designed for handling delicate, high-value products.

A Blueprint for the Modern Facility: Integrating Compliance, Automation and Sustainability

As manufacturers adapt to a broader range of products and formats, they must also navigate increasingly detailed regulatory expectations for sterile facility design and contamination control. The 2022 revision of EU GMP Annex 1 represents a significant evolution in regulatory thinking, placing a clear emphasis on modern technical solutions like isolators and automation over traditional procedural controls. The guideline’s core philosophy is to physically separate operators from sterile products to the greatest extent possible, thereby reducing reliance on manual procedures and minimizing the primary source of contamination risk (3).

This renewed focus on engineering controls is making isolator-based systems foundational to modern aseptic facility design. While these technologies have been available for years, many pharmaceutical companies have now adopted a policy requiring isolators as a minimum qualification for their contract development and manufacturing organization (CDMO) partners. For many organizations, adapting to these expectations means reevaluating both physical layouts and process strategies. Retrofitting legacy lines is sometimes possible but is often financially or operationally impractical. Consequently, a growing share of industry investment is shifting toward greenfield builds designed to meet Annex 1 compliance from the outset. These new installations pair isolator containment with high-speed performance and manufacturing flexibility, reinforcing both regulatory readiness and commercial scalability.

Automation is a critical part of this modern approach, and its benefits extend far beyond efficiency. Automated systems minimize human error, increase throughput, generate comprehensive data for quality control and ensure consistent dosing accuracy. These gains are being realized through the adoption of key technologies such as automated filling and stoppering lines, robotic handling for sterile transfers and automated visual inspection systems. The significant capital investment required to implement these technologies delivers clear returns in quality, compliance and reliability.

Beyond efficiency, automation provides an unparalleled level of data integrity. Every critical process parameter, from temperature and pressure to fill volume and stopper placement, is monitored in real-time and recorded in a comprehensive, unalterable digital batch record. This not only streamlines regulatory audits but also enables a move away from reactive, end-product testing and toward proactive quality assurance through the adoption of Process Analytical Technology (PAT). This data-rich approach allows for a deeper understanding of the manufacturing process, ensuring consistent quality and enabling continuous improvement.

This technological shift also requires a corresponding evolution in the workforce. The demand is moving away from operators skilled in manual aseptic techniques and toward technicians and engineers with expertise in robotics, data analysis and system maintenance. This necessitates a significant investment in training and development to cultivate the advanced skills needed to run the manufacturing facilities of the future.

Forward-thinking facility design, particularly for new greenfield builds, now extends beyond regulatory compliance to include another critical priority: sustainability. Drug developers face increasing pressure to reduce the environmental footprint of their supply chains and these expectations now apply to their CDMO partners. The focus has broadened beyond materials and waste to include energy efficiency, water usage and overall facility emissions. Integrating these considerations from the ground up during the design phase is essential for aligning environmental priorities with regulatory and operational goals. CDMOs that design with this level of foresight and transparency are better positioned to meet the long-term expectations of their partners and the planet.

Redefining the Partnership: A Framework of Expectation and Response

As fill finish operations become more intricate, the traditional client-vendor dynamic is evolving. The need for simple production capacity has developed into a demand for a true strategic partner, creating a new dynamic between sponsor expectations and the CDMO response.

The expectation: Early-stage expertise and shared accountability

Sponsors need technical insight at the earliest stages of development to support critical decisions on formulation, container selection and scalable process design. This is essential for navigating the complexities of advanced therapies and new delivery formats. Furthermore, evolving regulatory standards mean that sponsors and CDMOs are now held jointly accountable for product knowledge, compliance and risk mitigation.

→The CDMO response: An integrated partnership model

In response, successful partnerships are now being built on a foundation of transparency, shared data systems and collaborative problem-solving across the entire development lifecycle. The goal is to design for manufacturability from the outset, preventing costly and time-consuming changes later on. This proactive approach is critical for protecting product quality, satisfying global compliance requirements and supporting accelerated timelines.

The expectation: Future-proofing and proactive compliance

A growing trend among biopharma companies is to seek partners who demonstrably exceed current regulatory guidance. This future-proofing mindset often translates into a preference for CDMOs utilizing advanced isolator technology as a minimum qualification for partnership. By selecting a partner already operating at the highest standard, sponsors can build a more resilient and reliable supply chain capable of weathering future regulatory shifts or unexpected market surges.

→The CDMO response: Building trust through investment and expertise

Ultimately, trust is built through a combination of demonstrated technical expertise, investment in state-of-the-art infrastructure and a culture of open communication and collaborative engagement.

Kindeva's Bridge to the Future of Fill Finish

This convergence of challenges was the driving force behind Kindeva’s investment in our new state-of-the-art sterile fill finish facility in Bridgeton, Missouri. The site was designed with the sole purpose of addressing the industry's key pain points, combining isolator-based sterility assurance, format flexibility and end-to-end integration to optimize even the most complex programs. It serves as a clear example of the forward-thinking investment required to succeed in today's environment.

As a purpose-built greenfield facility, Bridgeton embodies the principles of a modern manufacturing facility. It was engineered from the ground up for complete compliance with the most stringent global regulatory standards, including EU GMP Annex 1. The facility’s high-speed filling lines are housed within advanced isolator systems, providing a closed and controlled environment that physically separates the product from human operators, ensuring the highest levels of sterility assurance.

This state-of-the-art infrastructure was designed for flexibility. The facility can readily handle a wide range of injectable formats, including vials, prefilled syringes and cartridges, and is equipped to manage everything from small clinical trial batches to large-scale commercial manufacturing. This integrated design is the practical application of the strategic partnership model, bringing together fill finish, product assembly and final packaging to eliminate the risks of a fragmented supply chain and accelerate timelines.

Building Better Tomorrows, Today

The sterile fill finish landscape is no longer defined by a single process model. It reflects a dynamic mix of therapeutic innovation, evolving compliance standards and increasing operational complexity. The facilities and partnerships that succeed in this environment will be those designed with adaptability and expertise at their core. The choices being made today in how lines are configured, how automation is applied and how partnerships are structured will determine how effectively the industry delivers the next generation of therapies. With the right infrastructure, technical insight and strategic alignment, manufacturing partners can help ensure that growing complexity becomes a driver of progress rather than a barrier to getting life-changing medicines to patients.

References

  1. https://www.globenewswire.com/news-release/2025/01/29/3017150/0/en/Fill-Finish-Manufacturing-Market-Size-to-Hit-USD-33-5-Billion-by-2032-Growing-at-a-CAGR-of-9-1-SNS-Insider.html
  2. https://pubmed.ncbi.nlm.nih.gov/15124199/
  3. EU GMP Annex 1: Manufacture of Sterile Medicinal Products (2022).