GMPs for Sterile Manufacturing of Biologics

Published on: 
BioPharm International, BioPharm International, June 2022 Issue, Volume 35, Issue 6
Pages: 38-41

The complex nature of biologics creates challenges for GMPs in sterile manufacturing, and the EU’s Annex 1 is making an impact.

Biologics, by nature, are complex and come with certain challenges when producing these products under current good manufacturing practices (CGMPs). Biologics are, typically, parenterals that must be sterile and manufactured under sterile aseptic conditions, according to Hanns-Christian Mahler, CEO of ten23 health. Sterile manufacturing of these “sensitive molecules”, therefore, adds an additional layer of complexity, notes Mahler.

“[Biologics] are typically manufactured under aseptic conditions, using sterile filtration and would generally not withstand sterilizing conditions such as heat (terminal sterilization or autoclaving) and are expected to undergo significant degradation when using chemical sterilization or radiation. Even ‘normal’ unit operations during sterile production, such as pumping, can significantly damage and degrade biologics. Hence, sterile manufacturing of biologics requires intimate knowledge and experience in the field of formulation and process development and understanding of critical quality attributes [CQAs],” says Mahler.

Injected products require strong controls on manufacturing to prevent microbial and other types of contamination of the product, according to Julie Bonjean, head of Quality and Validation at Univercells Technologies. Control over the manufacturing environment, equipment, and consumables is crucial, specifies Bonjean, and validation must be performed to ensure processes run as expected and are reproducible.

“Any human intervention on the process must be performed by highly trained professionals or reduced whenever possible. In both cases, meeting GMP requirements will add quality costs because more controls and more oversight are needed. For biologics, however, there is a need to develop scalable processes much earlier to ensure scalability from discovery into commercial production. All of this has driven the industry towards automated equipment portfolios that simplify tech transfer activities and are operated with single use systems to reduce the burden,” Bonjean stresses.

Aspects of following CGMPs in manufacturing of biologics that may be challenging are the evaluation of the materials’ qualities, and the application of emerging technologies, according to Jeffrey Lacouchie, regulatory affair specialist at Polyplus. “In general, complying with the FDA’s GMP requirements is not difficult, but there are several intricacies that might provide a barrier. For instance, it is simple to do gap analysis and confirm that processes are in accordance with the many regulations, guidelines, and technical publications that comprise contemporary CGMPs. But the quantity of complicated formulation work required in biological products is quite challenging,” says Lacouchie.

“The core GMP principles are the same for solid-dosage products and biologics. Given the nature of the manufacturing process for biologics, however, GMP guidelines include specific requirements regarding microbiological contamination which can represent a significant risk when producing biologics,” agrees Giuseppe Mauro, Vice President, Incyte.

To gain perspective on sterile manufacturing of biologics under GMPs, BioPharm International asked industry experts about best practices for manufacturing these products and to provide an update on the latest regulations for sterile manufacturing. Joining Bonjean, Lacouchie, Mahler, and Mauro in the discussion are Kurt Brorson, vice president, technical and Siegfried Schmitt, vice president, technical, both at Parexel; Richard Johnson, president of the Parenteral Drug Association (PDA); Lindsay Fraser, head of technical services at Symbiosis Pharmaceutical Services; and Paul Gustafson, chairperson of the Pharmaceutical Inspection Co-operation Scheme (PIC/S).

Best practices for biologics

BioPharm: What are some best practices for manufacturing biologics under GMPs?

Mahler (ten23 health): As biologics are manufactured aseptically, environmental control [and] robust and qualified aseptic processing are mandatory to ensure sterility. To reduce intervention and thereby the risk for the sterility, the process must be well characterized and developed to maintain product quality. Small process changes may harm the functionality of the product, so degradation pathways shall be known. Good understanding of the interaction of process parameters to CQA are key for stable and reliable manufacturing.

Mauro (Incyte): Companies need to stay well informed of the latest guidance and trends, as the CGMPs are continuously changing, and it is up to the manufacturer of biologics to stay up to date. Applying quality risk management (QRM) principles is an effective best practice. It is important to periodically step back and assess the internal manufacturing practices versus the industry standard, and to maintain a scientific and pragmatic approach in managing the product lifecycle. Another good practice is to exchange our practices with other manufacturers on a regular basis and look for all the market innovations that could be applicable to our processes.

Brorson and Schmitt (Parexel): Best practices are when companies achieve more than mere compliance, for example by applying the principles of quality by design (QbD) from the early stages of development right throughout the lifecycle. In addition, best practice is establishing a supply chain that is robust, flexible, and responsive to changing patient needs (e.g., for vaccines). Maintaining low or no bioburden during drug substance manufacture as well as viral safety is another. For biologics, drug substances manufacture is complex with microbially vulnerable steps and care is required to produce a safe and high-quality product.

Johnson (PDA): Drug substance manufacture often includes living cells, and control of process conditions for maintenance of cell banks, protection from contamination (chemical, microbial, and viral contamination) is critical. Each process is highly specific and involves many steps. Single-use, closed systems have aided in this protection, and permitted scale out and scale up much easier than traditional stainless steel.

Gustafson (PIC/S): One best practice that goes beyond GMP requirements for manufacturing of biologics (or any medically necessary drug) is the security of the drug supply chain. In the case of biologics, consideration should be paid to building redundancies into the stages of global supply chain whether it be cell banks, critical material suppliers, manufacturing capacity, or analytical testing.

Other legally binding or best practices for manufacture of biologics under GMP can also be found in the wealth of additional materials from national competent regulatory authorities or leading international industry associations.

The Pharmaceutical Inspection Co-operation Scheme (PIC/S) guides provide an excellent source of legally binding requirements (PIC/S GMP Guide) and best practices (see other relevant voluntary guidance documents) for the manufacture of biologics under GMPs (1–5).

Fraser (Symbiosis Pharm-aceutical Services): Best practices would include building quality, safety, and efficacy into the product (QbD). Specific procedures include the selection of GMP/parenteral compliant raw materials going into the process, in-process controls, and validated procedures/assays required to manufacture and characterize the product. Quality compliance must be performed throughout the entire product lifecycle and must be maintained for any outsourced third-party organizations such as test houses.

Annex 1 revision and other regulations

BioPharm: What can you tell us about the European Union’s revision to Annex 1? What changes should manufacturers be aware of, specifically for biologics?

Gustafson (PIC/S): The new version of Annex 1: Manufacture of sterile products highlights a tremendous success for international co-operation and harmonization with PIC/S, European Medicines Agency (EMA), and World Health Organization (WHO) coming together to jointly develop this guidance for the manufacture of sterile products. We look forward to the upcoming joint publication of this guide by PIC/S, the European Commission (EC), and WHO in the coming months. This document is a much more comprehensive guidance to support industry, growing from 16 pages in the latest version to 57 pages with a completely revised chapter layout.

A key consideration in the new Annex is how pharmaceutical quality system requirements are incorporated into the guide to increase the use of quality risk management and to implement contamination control strategy. This will help enable good quality culture at manufacturing sites to help facilitate an increase in product quality.

Another, very important point about this guidance and other more recently published PIC/S annexes is the importance for regulatory frameworks to enable adoption of new technologies and not hinder scientific advancements.

Advertisement

To help better understand the changes, PIC/S encourages manufacturers to engage in one of the many conferences that leading international pharmaceutical associations have been hosting and additional information sessions that will be planned in the year following publication. Once published, this guide will have a one-year implementation period (with a two-year period for one section related to certain lyophilizer requirements).

Johnson (PDA): Annex 1 is going through its first major update in several years. The timing for publication is not certain, but it is expected around mid-2022. The new version embraces ICH [International Council for Harmonisation] Q9 risk management approaches, but still includes prescriptive requirements in many areas. For the most part, it is consistent with current GMP approaches, but there are still some areas of
concern, [such as:]

  • The use of prescriptive requirements and examples (perceived as prescriptive requirements), that may restrict or limit current and future innovative approaches
  • Mixed messaging on the allowance of alternative approaches based on risk, by alternating a language supporting a risk-based approach with very prescriptive requirements
  • A focus on reactive process monitoring and product testing as a primary means of process control, that results in less emphasis on process design, training, and failure prevention
  • The need for recognition of the impact and feasibility of certain Annex requirements and changes to existing manufacturing processes, facility, and operations, as compared the product quality benefit of those requirements and changes
  • The need to clarify the intent of and harmonize language in Annex sections, to prevent misunderstandings due to the wide geographical scope of this guidance document
  • The lack of clear distinction between and the perceived grouping of technologies that require different contamination control strategies, including RABS [restricted access barriers] and isolators, terminal sterilization and aseptic processing, and ATMPs [advanced therapy medicinal products] and conventional therapy manufacturing.

The requirement for a Contamination Control Strategy is an example of positive improvement.

Lacouchie (Polyplus): The EU’s revision to Annex 1 provides basic recommendations to produce all sterile products utilizing QRM concepts to guarantee that microbiological, particle, and pyrogen contamination is avoided in the final product. Much is dependent on the personnel’s ability, training, and attitudes. Quality assurance is very critical in this form of manufacturing, which must strictly adhere to well-established and validated methods of preparation and procedure. Any terminal procedure or finished product test should not rely only on sterility or other quality factors.

This revision is designed to improve clarity, integrate QRM concepts to enable the incorporation of new technology and innovative processes, and change the structure to a more logical flow. [Significant changes include] introduction of new sections, introduction of QRM principles, restructured to give more logical flow, [and] added detail to a number of the previous sections to provide further clarity.

Mahler (ten23 health): The Annex 1 revision, in its second draft as of Feb. 20, 2020, includes a variety of considerations in quite logical arrangement. One element is that the Annex 1 revision clearly stated barrier technologies as state-of-the-art manufacturing. Frequent contamination by bacterizidal gassing will be mandatory to keep the sterile environment for manufacturing. Potential update and residues of sterilizing agent (e.g., vaporized hydrogen peroxide [VHP]) has to be considered and evaluated for product quality impact. There are also important chapters related to Visual Inspection and Container-Closure-Integrity.

Mauro (Incyte): The EU’s revision to Annex 1 is a real improvement in our continuous effort for the protection of patients. It reinforces the importance for each manufacturer to ensure that an effective QRM is integrated into all areas of the product life cycle. One of the key objectives of this update is to improve the manufacturer’s controls over the potential microbiological contamination of products. Key changes and challenges will impact each manufacturer’s formal documentation for this risk-based contamination control strategy, including consideration of the testing filters in cleanrooms and barrier technologies. Added contents on aseptic processing, alignment with European Pharmacopoeia on WFI (Water for Injections) and barrier technologies will help the industry to clarify expectations.

Brorson and Schmitt (Parexel): Industry has not yet seen the final version of the revised Annex 1, which is likely to be published later this year. However, it is clear that state-of-the-art technology will be an expectation. This means that in order to achieve compliance with Annex 1, companies may need to update with different and new equipment or facilities. A new key concept is the Contamination Control Strategy, which requires companies to look at contamination control holistically.

BioPharm: Has the revised Annex 1 addressed some of industry’s concerns?

Mauro (Incyte): With more than 2000 comments generated by
contributors on the first draft of the Annex 1, we can see that the industry is deeply engaged and is looking for some clarification and guidance on the defined requirements.

The new version is bringing some clarity around the aseptic control measure concept and expectations. However, final alignments are still in progress and a final version regarding the testing filters in classrooms is still expected.

Johnson (PDA): During the lengthy revision process, there have been many concerns from the earliest drafts that have been addressed. The last public version still had some language that PDA and other associations were requesting changed. We will see in the final version whether these have been resolved. There is also concern about the dates for implementation; due to the long lead times for some equipment, it is hoped that the final version will take this into consideration.

Gustafson (PIC/S): The joint international team representing the working group for Annex 1: Manufacture of sterile products has been active in listening to industry feedback. PIC/S, EMA, and WHO hosted two separate rounds of consultation. The first in 2017 provided for approximately 6000 comments from industry stakeholders. After significant revision, a second consultation was held in 2020 with about 2000 comments.

The feedback provided by industry was carefully considered by the joint working group in advancing the version that will soon be published. PIC/S will be working to help ensure there is mutual understanding of the revised version that helps in protection of patient health.

Lacouchie (Polyplus): The specific changes in the newly revised guidance document that will significantly impact the industry will be technical and organizational. This revised document is an excellent illustration of what a coordinated effort can achieve, providing a connection with the ongoing mutual recognition process between the US and the EU. Industry experts anticipate changes with the proposed Annex 1, particularly in the areas of QRM, contamination control strategies, and environmental and process monitoring.

Mahler (ten23 health): From our perspective at ten23 health, the Annex 1 revision does not bear any surprising new requirements and matches our expectations for CGMP. Interestingly enough, a survey made during the PDA Visual Inspection conference suggested that roughly a third of attendees were not ready to address the new Annex 1 draft requirements related to ‘container closure integrity’ (CCI).

BioPharm: Are there any upcoming changes to FDA or ICH guidelines for sterile/aseptic manufacturing that manufacturers of biologics should be aware of?

Brorson and Schmitt (Parexel): The revisions underway to ICH Q9 on QRM will prompt all companies, including the biotech industry, to assess their approach to QRM, particularly with regards to the level of formality and appropriateness of the assessment outcomes. ICH Q5A is also under revision. While Q5A applies to adventitious agents (viruses) that are tested and controlled during drug substance manufacture, not fill/finish, viral safety is a critical aspect for the overall safety of the final dosage form.

Gustafson (PIC/S): The upcoming publication of PIC/S Annex 1: Manufacture of Sterile Products is perhaps one of the most significant GMP publications that will be advanced in the near future. This said, PIC/S seeks to co-operate with other international partners including ICH and International Coalition of Medicines Regulatory Authorities (ICMRA).

PIC/S closely co-operates with ICH. Since 2020, there has been an operating pilot for More Routine Engagement of ICH with PIC/S on ICH Q Guidelines that are related to inspection activities. This covers Q9 (Quality Risk Management) and Q12 (Pharmaceutical Product Lifecycle Management) for which PIC/S has been represented in ICH working groups. The current version of ICH Q9 was adopted by PIC/S in 2009 as Annex 20 (Quality Risk Management) of the PIC/S GMP Guide. This will be updated once a new version of Q9 is ready. For Q12, PIC/S is supporting the effective implementation of ICH Q12, with regard to inspectors’ needs, through training and various tools in development, with the support of ICH.

Another important area that PIC/S has observed for co-operation is with ICMRA in establishment of a Pharmaceutical Quality Knowledge Management System (PQ KMS) for which there is a recent publication.

References

  1. PIC/S, Guide to Good Manufacturing Practice for Medicinal Products, Part I (PIC/S, Feb. 1, 2022).
  2. PIC/S, Guide to Good Manufacturing Practice for Medicinal Products, Part II (PIC/S, Feb. 1, 2022).
  3. PIC/S, Annex 1: Manufacture of sterile products (revision currently unpublished).
  4. PIC/S, Annex 2A: Manufacture of advanced therapy medicinal products (ATMP) for human use, (in force since May 1, 2021).
  5. PIC/S, Annex 2B: Manufacture of biological medicinal substances and products for human use. (in force since May 1, 2021).

About the author

Susan Haigney is managing editor of BioPharm International.

Article details

BioPharm International
Vol. 35, No. 6
June 2022
Pages: 38-41

Citation

When referring to this article, please cite it as. S. Haigney, “GMPs for Sterile Manufacturing of Biologics,” BioPharm International 35 (6) (2022).