Considering the Promises of Point-of-Care Manufacturing

Published on: 
Pharmaceutical Technology, Pharmaceutical Technology, November 2023, Volume 47, Issue 11
Pages: 12-14

Emergence of advanced manufacturing technology to ensure quality of biopharmaceutical drugs combined with efforts to identify a regulatory pathway indicate that a distributed manufacturing model is within reach.

Although biopharmaceutical production today generally takes place in centralized manufacturing facilities, industry and regulators are taking a close look at the benefits of decentralized or distributed manufacturing, which involves smaller and flexible-volume manufacturing operations in multiple locations closer to the site of use and even to point-of-care (POC) locations. POC manufacturing is seen as essential for efficiency in producing personalized medicines. In the near-term future, POC production is likely to be in a controlled environment such as a hospital, clinic, or pharmacy, while in the long term, POC production could extend to other locations. Such a model could enable quality drug production anywhere, from the battlefield to remote villages or even outer space, experts suggest.

The impetus for POC manufacturing comes in part from its potential to alleviate pressing problems, such as drug shortages, pandemic preparedness, and equitable availability of treatments. It is also driven by technological advances that promise to allow efficient and consistently high-quality production using new equipment, analytical tools, and quality control paradigms.

These technologies offer the benefit of making drugs much closer to where and when they are needed. “The advantages of making medicines on demand—to solve issues such as the difficulty of predicting demand and the complexity of the supply chain—are compelling,” states Govind Rao, professor at the University of Maryland, Baltimore County (UMBC) and director of UMBC’s Center for Advanced Sensor Technology.

FDA recognizes the need for flexible and agile manufacturing and sees the potential for portable, distributed manufacturing units to be used for POC manufacturing. In October 2022, the Center for Drug Evaluation and Research (CDER) published a discussion paper that highlighted areas to consider for drugs regulated by CDER as well as the Center for Biologics Evaluation and Research and called for public feedback (1). FDA and the Product Quality Research Institute (PQRI) also held a workshop in November 2022 to gather input from stakeholders. For advanced manufacturing technologies—particularly distributed manufacturing, POC manufacturing, artificial intelligence (AI), and end-to-end continuous manufacturing—seeking input is the first step in FDA’s new Framework for Regulatory Advanced Manufacturing Evaluation (FRAME) initiative, according to a presentation by Michael Kopcha, director of CDER’s Office of Pharmaceutical Quality (2).

The European Medicines Agency’s (EMA) Quality Innovation Group is focused on a similar list of advanced manufacturing technologies, which were discussed at a March 2023 focus group meeting (3). That decentralized manufacturing is being discussed is one reason for optimism regarding its uptake in Europe, suggests Celeste Lamm, director of Global Regulatory Affairs, CMC at Merck. In addition, she points to the European Commission’s proposed new directive (4) that includes a pathway for decentralized manufacturing within the European Union and provides an architecture for responsibility between a central site and decentralized sites. “However, the proposal limits decentralized manufacturing to applications where the central site is located within the EU, and it isn’t clear how the regulation would be applied if some of the decentralized sites were outside of the EU,” Lamm says.

Regulatory uncertainty is an ongoing challenge, with unanswered questions around how connected sites that are located in different regions will be regulated and a lack of global harmonization. Agencies in different regions are communicating with each other, and it is hoped that approaches will be similar. Because no final guidance has been released by any regulatory agency, any differences in requirements are still unknown, Lamm cautions.

Change will take time. Lamm sees similarities between the pace of adoption of distributed manufacturing and that of continuous manufacturing. “Both regulators and industry acknowledge the benefits, but adoption has been slow, because existing traditional manufacturing is frequently sufficient, and it can keep costs down to use existing facilities,” Lamm says. “Adoption of innovative manufacturing technology occurs gradually as there is opportunity to replace existing lines, where existing technology isn’t sufficient (for example, when local manufacturing is needed), and where there is enough clarity to calculate the long-term benefit to justify investment.”

Consistent quality

A significant hurdle for distributed and POC manufacturing is how to ensure consistent quality of the drug product, but there is a growing availability of technologies that can meet this challenge. For example, prefabricated, portable cleanrooms and automated processes that fit in these spaces meet the need for standardization of equipment, process, and systems that is crucial to consistency. In addition, digital technology and cloud-based systems in use today make it easier to connect the data and quality systems of different locations. With these tools, distributed manufacturing can even reduce risk and contribute to consistency.

“Using enterprise quality systems across distributed sites is the natural extension of how we currently work in a global environment,” says Lamm. She says that regulators accept aspects of digitally connected quality system solutions, but they note that it is important to ensure that all personnel are similarly trained and are following the same practices.

“There are many flavors of distributed manufacturing for consideration when training personnel,” Lamm adds. Variations include the number of sites and the complexity of the manufacturing process. “The conversation regarding appropriate quality approaches for distributed manufacturing is ongoing, particularly for high volumes of sites or complex manufacturing processes. It is critical that we continue the dialogue between industry and regulators to address concerns.”

Advertisement

ML in QA/QC

Quality assurance/quality control (QA/QC) methods for distributed manufacturing will need to be different from those currently used in centralized manufacturing, says Rao. His group at UMBC recently patented a method for using machine learning (ML) to ensure consistent product quality in UMBC’s Biological Medicines On-Demand (Bio-MOD) system, which uses a cell-free method for end-to-end continuous manufacturing of biologic drug substances (5).

“With cell-free manufacturing, one gene produces one protein in a process that is almost more like chemistry than biology in that it can be consistent,” says Rao. “Our ML algorithm can extract enough data from the process sensors and analytic measurements to get a high degree of confidence that everything about the process is consistent. If the process is the same, the product should be same. The algorithm can also identify any deviation from the expected, such as an air bubble or impurity. This automated function raises the safety level to a much higher level than the current method of testing a product after it is made.”

The Bio-MOD system is currently suitcase-sized, but could be scaled-out to larger volumes, says Rao. He adds that the ML approach can also be used in cell-based manufacturing.

POC in CGT

Decentralized, POC manufacturing is particularly helpful for autologous cell and gene therapies (CGTs), because it solves some of the problems of long-distance, cold-chain shipping of patient-specific raw materials and finished products. Various technology providers have created closed, automated CGT manufacturing systems that aim to provide scalable throughput to meet the growing demand for CGTs in centralized or decentralized locations (6).

Biotech company Orgenesis has targeted POC CGT manufacturing with its POCare Network and its Octomera Mobile Production Units and Labs (OMPULs). In June 2023, the company and one of its network partners, the University of California (UC), Davis, announced that OMPULs will be installed and operated by Orgenesis at multiple UC medical and academic institutions (7).

Decentralized and standardized manufacturing is crucial for getting CGTs to patients at a reasonable cost, says Vered Caplan, CEO of Orgenesis. She says that the OMPULs are standardized units that are operated with standard procedures.

“We have defined everything—raw materials, disposables, all the process parameters, cleaning parameters, the environment in and around the unit, all the equipment, and procedures,” Caplan explains. “The unit is not just a mobile cleanroom. It is a closed, automated mini-production facility with standardized, validated isolation units inside. These units can then be duplicated and located at different sites, either in a hospital or nearby. Because of their agnostic nature, we can augment them to many modalities and even applications such as apheresis, if needed, to complete the full value chain.”

Comparability testing of products from the different units will demonstrate that standardization produces comparability; the extent of testing is decided using risk analysis. Caplan adds that although product quality is built-in to the standard process, the decentralized sites will also have quality assurance oversight through centralized audits. “Our work is driven by compliance to the most current guidelines, and our flexible nature allows us to be responsive as we start to see [new regulatory guidance] come to fruition,” she says.

Caplan suggests that hospitals are driving demand for decentralized manufacturing so they can meet their patients’ needs. She says standardized manufacturing platforms like the OMPUL will provide consistent quality at lower cost than each hospital creating their own platform. Orgenesis is also working with solution and equipment providers to ensure that the team is aware of the latest technologies and that they can easily be incorporated into the platform, Caplan says.

References

  1. CDER. Distributed Manufacturing and Point-of-Care Manufacturing of Drugs, Discussion Paper. FDA.gov (October 2022).
  2. Kopcha, M. A Regulatory Perspective on Innovations in Pharmaceutical Manufacturing. Presentation at the FDA/PQRI Workshop of the Regulatory Framework for Distributed and Point of Care Pharmaceutical Manufacturing (Nov. 14, 2022).
  3. EMA Quality Innovation Group. Listen and Learn Focus Group Meeting Report, May 12, 2023.
  4. EC, Proposal for a Directive Of The European Parliament And Of The Council on the Union code Relating to Medicinal Products for Human Use, and Repealing Directive 2001/83/EC and Directive 2009/35/EC (April 26, 2023).
  5. Rao, G. et al. US Patent 11,685,892 B2. “Methods to Incorporate Machine Learning Analytics for Optimizing Protein Purity, Potency, and Quality in an On-Demand Production System for Point-of-Care Delivery” (June 27, 2023).
  6. Markarian, J. Automation Aids Cell and Gene Therapy Production. BioPharm Intern., 2023 36 (7) pp. 10-13
  7. Orgenesis. Orgenesis and University of California, Davis Sign Partnership Agreement for Rollout of Cell and Gene Therapy Mobile Processing Units and Labs Throughout California. Press Release, June 7, 2023.

About the Author

Jennifer Markarian is manufacturing reporter for Pharmaceutical Technology.

Article Details

Pharmaceutical Technology
Volume 47, No. 11
November 2023
Pages: 12–14

Citation

When referring to this article, please cite it as Markarian, J. Considering the Promises of Point-of-Care Manufacturing. Pharmaceutical Technology 2023, 47 (11), 12–14.