Emily Whitehead made the history books in 2012 when she become the first pediatric patient with B-cell acute lymphoblastic leukemia to be given a chimeric antigen receptor (CAR) T-cell therapy. After receiving the therapy at Children’s Hospital of Philadelphia, Emily nearly died from side effects. But after 23 days, she was declared cancer free. She remains cancer free today.

Cell and gene therapies (CGTs) have decidedly advanced since 2012. Dozens of CGTs have secured FDA approval. Six of them are CAR T-cell therapies. But significant cell therapy manufacturing challenges need to be addressed for both autologous and allogeneic therapies to get these lifesaving cures to a greater population of individuals in need.

Tool providers are working on closed-process systems and bioreactors to address contamination risks and to take advantage of improved raw and ancillary components. Also, innovative processes are being developed to speed up bioburden testing and quality control. These processes could help manufacturers avoid wasting a week or more—a significant delay for a living therapy that needs to be administered quickly.

Cell therapy developers are optimistic that the current challenges can be resolved so that these approaches can go through another impactful evolution, including decentralized point-of-care manufacturing.

Itineraries for optimization journeys

Both autologous and allogeneic cell therapies share common features, operational complexities, and manufacturing challenges, from closed processing and liquid handling under aseptic conditions to process time constraints, due to the perishability of living cell–based therapies. Manufacturing currently leverages similar methods for cell isolation, genetic modification, and formulation, although the nature of the specific therapy dictates the choice and the scale of cell expansion platform.

Closed systems, automation, standardization, process analytical technology, efficient cell expansion systems, and rapid testing methods will be the main success enablers of the next generation of advanced cell therapies. “As the sector matures, more templated processes will help ensure process optimization, robustness, and continuous production improvement by providing a structured framework for monitoring and controlling critical process parameters across scales and applications,” says Philip Schaefer Jaramillo, head of business franchises, process solutions, Life Science, Merck KGaA, Darmstadt, Germany.

Every day counts for manufacturers of these treatments. Products need to be released and administered to patients as fast as possible. Classical quality control micro-sterility testing takes 14 days to complete due to lengthy incubation requirements.

Manufacturers are increasingly turning to alternative rapid microbiological testing methods such as the Milliflex Rapid System 2.0 for testing cell-based therapeutic products for microbial contaminants, which reduces their time to results by up to 75%. The system combines Milliflex membrane filtration sample preparation, suitable cell lysis protocols, ATP bioluminescence technology, and advanced image analysis to deliver fast, reliable microbial detection in colony-forming units.

In addition, BioReliance Contract Testing Services offer risk-mitigating solutions. Client-based testing programs are developed that draw upon an array of available platform assays for final product release. “We also advance fully closed and sterile sampling solutions to ensure compliance and quality of cell therapy products,” Schaefer Jaramillo remarks. “The Mobius Breez Microbioreactor is a unique automated cell expansion platform for higher throughput, small-scale cell therapy process optimization.”

Mapping efficient routes

A common manufacturing platform for all cell therapies does not exist currently. However, according to Matthew Hewitt, PhD, vice president, technical officer, CGT and Biologics, Charles River Laboratories, manufacturing processes are beginning to be “bucketed.” He says that applying general know-how from prior processes is vital to decreasing development timelines and ensuring robust manufacturing processes. For example, program-to-program autologous CAR T-cell therapy manufacturing processes tend to be quite similar, and prior knowledge can be quickly applied to future programs to reduce development costs and timelines.

“Generally, when one considers how to develop or structure a manufacturing process, it is a good idea to assess the cell numbers needed for clinical dosing and quality control,” Hewitt advises. “Once that is established, you can work backward to design a process that meets those needs and balances being ready for clinical activities while having ‘line of sight’ as to whether the process is suitable for commercial manufacturing.”

Cell Therapy lab picture
This image from Charles River Laboratories shows a technician preparing for a cell processing run on the CliniMACS Prodigy, a cell manufacturing platform developed by Miltenyi Biotech.

Streamlined manufacturing and quality control testing is needed as it seems market penetration for cell therapies is dictated by the number of doses that can be manufactured rather than by patient adoption. As mentioned previously, sterility testing for product release tends to be a challenge because it takes seven days on average. “This timeframe needs to be shortened while maintaining quality and safety, perhaps with molecular methods,” Hewitt notes.

Charles River employs a “concept to cure” portfolio, which provides developers a one-stop shop for advanced therapy development. The portfolio connected teams across the company’s CDMO operations in plasmid DNA, viral vector, and cell therapy manufacturing to condense timelines, and the preclinical CRO portfolio is being leveraged to further streamline advanced therapy development.

“We will continue to see an acceleration of commercial approvals,” Hewitt predicts. “In addition, I envisage regulators from multiple jurisdictions will develop a pathway where if one regulator approves a therapy, then a network of others will also grant the therapy approval in their jurisdictions.”

There have been promising discussions with the FDA and other regulators about decentralized, point-of-care cell therapy manufacturing. “Once we innovate our way out of the manufacturing problem, we will then have to contend with challenges in quality control release testing and eventually quality assessment review of batch records for product release, as well as decentralizing our quality reviews,” Hewitt adds.

Switching gears smoothly

Some cell therapy models are based on stem cells; others, not. In either case, there is support for clinical trials and commercial applications in both autologous and allogeneic settings. Despite all these contingencies, manufacturing processes for cell and gene therapies rely on a common modular platform that can be adapted to different cell types and therapeutics.

Closed automated systems address many error-prone and labor-intensive open handling steps, leading to a significant reduction of contamination risks, costs, and timelines. Several effective automated systems specific for CGT application are available. Some represent a “one solution” automated platform that could perform all the manufacturing steps in a fully closed system. Other modular instruments are designed to perform defined steps and to work in combination.

“Some constraints still need to be overcome,” says Sara Morlacchi, PhD, cell process development manager, AGC Biologics. “In general, these devices lose accuracy when working with small volumes, and in the case of allogeneic production, the challenge is to guarantee production scalability.”

Manufacturing processes are highly variable in terms of cell type, length, volumes, reagents, equipment, test procedures, and processes for cell modification or cell expansion. To accommodate customer requirements, AGC Biologics offers a flexible service to meet preclinical through commercial needs. To support cell therapy products, the company has expanded operations at its site in Italy (the Milan Cell and Gene Center of Excellence) and at its site in Longmont, CO. The company has also announced it is building a new facility in Yokohama, Japan, that is scheduled to begin providing services in 2025.

Cell Therapy lab picture
AGC Biologics offers cell therapy services at two locations: the company’s Longmont, CO, site and, as shown here, the Milan Cell and Gene Center of Excellence. (The Milan facility was the first cell and gene therapy site approved in Europe for GMP manufacturing.) AGC Biologics’ capabilities encompass the creation of engineered cells at various scales, backed by analytical method development and qualification.

AGC Biologics has experience with both open and closed processes and proven expertise in lentiviral and retroviral transduction. “We also focus on implementing the latest technologies,” Morlacchi declares. “These include genome editing systems and mesenchymal stem cell–derived exosomes.”

AGC Biologics’ cell and gene platform includes flexible processes based on different cell types such as hematopoietic stem cells, T cells, natural killer cells, and mesenchymal stem cells, either small scale for autologous production or large scale for allogeneic batches. Process development is performed with a GMP-like approach to de-risk the transfer phase. “We offer more than 160 in-house analytical tests to characterize the product through its life cycle and to follow all development phases required for the establishment of a manufacturing platform,” Morlacchi adds.

Transitioning to higher-grade inputs

“Demonstrating comparability and reproducibility are essential for the advancement of novel cell and gene therapies,” says Julia Hatler, PhD, vice president of CGT solutions, Bio-Techne. “Developers have become more conscious of the supply of critical reagents and ancillary materials throughout their workflows.”

This includes transitioning to reliable, higher-grade materials such as animal-free and GMP-grade earlier in the development pipeline to reduce late-stage bridging efforts and to ensure future supply, performance, and regulatory compliance.

“Bio-Techne offers a wide range of reagents to support this transition. Our research-use-only, animal-free, and GMP cell culture reagents include cytokines and growth factors, small molecules, antibodies, media, and extracellular matrices for robust, reliable, and consistent cell cultures,” Hatler details. “Most animal-free research-use-only and GMP proteins are manufactured at the same facility, on the same equipment, and by the same team to ensure consistent specifications and a seamless transition from research to clinical manufacturing.”

To further decrease time and cost for cell engineering, nonviral genome engineering solutions are increasingly being implemented. “Nonviral cell engineering with our nucleic acid–based TcBuster transposon system can deliver larger gene payloads more quickly and cost effectively than traditional viral-based approaches,” Hatler asserts.

Clinical and commercial success also relies on the ability to scale manufacturing and analysis, including eliminating manual touchpoints by using integrated, aseptic, closed-cell manufacturing systems that have relatively small physical footprints. “We also provide scalable immune cell therapy manufacturing solutions,” Hatler notes. “For example, we provide the closed-system G-Rex bioreactor and Lovo cell processing system through our strategic partnership with ScaleReady.”

There is a strong movement toward flexible automated analytical assay platforms for both rapid and accurate multiparameter critical quality attribute measurements, which are required for regulatory approvals. “Bio-Techne’s solutions include Simple Western for complex cell lysates, Simple Plex automated ELISA for cytokine monitoring, and Maurice for imaged capillary isoelectric focusing assays and capillary electrophoresis sodium dodecyl sulfate assays for higher-purity samples,” Hatler says. “These analysis systems combine accuracy and consistency with the automated scalability required for cell and gene therapy manufacturing.”

Finally, there is a growing interest in tracking expression to evaluate a therapeutic’s performance. Spatial biology solutions include the RNAscope ISH platform, which evaluates therapeutic biodistribution and cellular tropism in preclinical model systems.

Driving toward consistency

The cell therapy field is witnessing significant advancements, especially with increased automation and the development of allogeneic “off the shelf” therapies. Automation is crucial for scaling up traditionally labor-intensive manufacturing processes while
allogeneic therapies offer the promise of more accessible treatments by eliminating the patient-specific requirement. The approval of gene editing therapeutics, such as the first CRISPR-based gene editing therapy for sickle cell disease, highlights the field’s potential.

ACROBiosystems offers a comprehensive range of high-quality, affordable, and validated premium and GMP-grade cytokines and growth factors crucial for cell culturing, particularly during the cell therapy manufacturing phase. These products play a pivotal role in ensuring consistent results, addressing the challenge of maintaining cell viability, potency, and functionality throughout the development process.

A diverse array of products to help ensure cell therapy manufacturing safety and efficacy is available, including GMP-grade nucleases and residue detection kits. “These products are instrumental in eliminating and detecting traces of DNA/RNA and other contaminants, effectively mitigating significant concerns related to CMC QC,” says Peter Hsueh, PhD, marketing product manager, ACROBiosystems. “We support the field in overcoming key challenges from R&D to manufacturing.”

The company’s focus on GMP-grade quality management and stringent controls ensures that ancillary materials meet the highest standards for safety and consistency. Adherence to regulatory guidelines such as USP <1043>, USP <92>, Ph. Eur. 5.2.12, and ICH guidances ensures batch-to-batch consistency, stability, and efficacy. An integrated approach to aseptic production prevents contamination and upholds product integrity.

“Our GMP growth factors and cytokines are engineered for exceptional consistency, tailored to specific cell culture applications, ensuring product potency and streamlining the transition from preclinical research to clinical manufacturing,” Hsueh asserts. Comprehensive material regulatory documentation and expertise support IND or BLA applications.

Although significant progress has been made in providing GMP-grade raw/ancillary materials for cell therapy applications, several gaps persist. Specifically, a need exists for a broader range of GMP-grade growth factors crucial for induced pluripotent stem cell culture and differentiation, which are essential for effective allogenic cell therapy development. ACROBiosystems is expanding its offerings of key GMP-grade growth factors and cytokines. It also provides custom GMP protein services to tailor cytokines to specific project needs.

Previous articleProtein Microparticles Turn Infusions into Injectables
Next articleLonza Launches AI Service for Small Molecule Development