19 november 2024
Continuous Manufacturing (CM) is an innovative approach to drug manufacturing that applies to drug substances and drug products containing chemical entities and therapeutic proteins. It is applicable to new products (e.g., new drugs, new pharmaceutical forms or dosages, generic drugs, biosimilars) as well as to the conversion of batch manufacturing to CM for existing products (European Medicines Agency, 2023).
CM is regulated by ICH Guideline Q13, which became effective in July 2023.
CM involves the seamless supply of input materials, the transformation of materials in-process, and the simultaneous removal of output materials within a manufacturing process, without any interruption.
CM can be applied to some or to all unit operations in a process. Examples of CM modes based on the ICH Guideline Q13 (2021) include:
The ICH Q7 definition of a batch applies to all modes of CM. Regarding batch size, CM has flexible options as explained below:
Real-Time Release Testing: Release testing of the drug product produced with CM is usually performed using a combination of testing on the core tablets during manufacturing (RTRT) and testing on the film-coated tablets in the quality control lab (end product testing).
Continuous manufacturing is an innovative manufacturing approach that involves the continuous production of a product without interruption. It is a highly efficient process that can help businesses to reduce costs, increase productivity, and improve product quality. However, companies need to be aware of the challenges associated with continuous manufacturing
(Karkhana.io., n.d.).
Figure 1
Comparison of Continuous Manufacturing and Batch Manufacturing process
(Sia, C. H., Teh, K. S., & Chan, L. W. 2021, Figure 1)
According to ICH Q13 (2021), CM is defined as follows:
CM involves the continuous feeding of input materials into, the transformation of in-process materials within, and the concomitant removal of output materials from a manufacturing process.
Within Q13, there is very little guidance on process transfer and equivalency for CM processes. The guideline does address scale-up, but comes primarily from the standpoint of utilization within the same process train, or a “like-for-like.” It doesn’t really address process or equipment changes other than to say, “It may be possible.”
This creates a situation where technology users will either:
No, we did not face any significant regulatory hurdles during our Continuous Manufacturing (CM) production process. We ensured that all regulatory requirements were met throughout the development and implementation phases, including comprehensive data submissions and adherence to quality guidelines.
However, it is important to note that according to an FDA statement published in February 2019 by then FDA Commissioner Scott Gottlieb, MD, and Janet Woodcock, MD, Director of the FDA’s Center for Drug Evaluation and Research (CDER), it was clear what direction the FDA is taking toward CM. The statement read:“We’re encouraged to see a growing number of companies embracing CM. It’s a key step towards promoting drug quality and improving the efficiency of pharmaceutical manufacturing. We’ve worked hard to help the industry develop the tools to start advancing these goals. The FDA is committed to helping more companies advance these CM platforms, owing to the public health benefits of these more modern approaches. We support the early adopters that are embracing this innovative technology, and we look forward to working with other interested companies.”
This reflects the FDA’s proactive stance on modernizing manufacturing processes, which aligns with our experience in CM.
CM can enhance the pharmaceutical manufacturing process with a magnified development approach of Quality by Design (QbD) and the utilization of Process Analytical Technology (PAT). As depicted in the figure below, a comprehensive QbD approach allows for continuous enhancement through product and process understanding to ensure better product quality (Sia, C. H., Teh, K. S., & Chan, L. W., 2021).
Figure 2
Quality by Design (QbD) Framework: Integrating Science and Risk-Based Principles
(Sia, C. H., Teh, K. S., & Chan, L. W. 2021, Figure 2)
PAT is necessary for highly automated continuous processing, as it fulfills quality requirements such as residence time distribution (RTD), and CQAs such as the percentage of active pharmaceutical ingredient (API), particle-size distribution, granule size, and many others, which can be monitored with PAT. RTD refers to the distribution of time that materials remain in a unit operation; thus, it is critical for material characterization. PAT’s primary goal is to measure critical process parameters (CPPs) that directly impact the essential quality attributes of active pharmaceutical ingredients (APIs) (Mettler Toledo, n.d.; Sia, C. H., Teh, K. S., & Chan, L. W., 2021).
PAT is an important element in a Quality by Design (QbD) approach, which supports the idea that quality cannot simply be tested in the final product but rather must be inherent by design. This approach also facilitates a smoother transition from BM to CM (Mettler Toledo, n.d.).
This article and interview were written by:
Author:
Jimmy Gandhi
CMC Specialist
ProductLife Group
Co-Authors:
Mélissa Bou Jaoudeh
Innovative Product Development Officer
Research & Innovation
ProductLife Group
Alaa Abdellatif, PhD
Innovation Product Development Officer
Research & Innovation
ProductLife Group
Supported by:
Fabio Pedna
Team Lead
ProductLife Group
The interview was conducted with and answered by Jimmy Gandhi.
Go to our Events to register
Go to our News to get insights