Microbiology Roundtable

Bioavailability Enhancement By Attenuating Presystemic Metabolism

In general, what are some of the current critical issues/trends facing pharmaceutical manufacturers in regards to microbiology testing and remediation?

Michaël Reynier, VP Healthcare Business Industrial Microbiology Unit, Health & Personal Care Business, bioMérieux SA: Microbiology testing still relies heavily on culture media and manual interpretation. Therefore, it remains a slow and sometimes subjective process - especially when compared to analytical methods used in chemistry. Consequently, pharmaceutical manufacturers suff er from recurring data integrity weaknesses and sub-optimization of their workforce due to limited automation.

Additionally, time to result continues to be a growing concern as more complex and fragile drug products that are short-lived or non-sterile by design gain in popularity.

Jordi Iglesias, Technology and Market Development Manager II, Microbial Solutions, Charles River: Data integrity: Most compendial microbiology tests are old techniques based on visual growth. These techniques rely on operator interpretation of growth and this subjective reading of the test is hard to reconcile with data integrity requirements. In my opinion the standardization of the readings in microbiology and removal of operator subjectivity is the most critical issue that microbiology has to face in the coming years.

Parametric release: Parametric release will also be frequently discussed in the coming years. Parametric release requires an extensive, sustained effort to obtain representative data to support final product release without performing release testing.

Real-time tests: Real-time testing is very closely related to parametric release as it can provide a huge amount of consistent data for environmental monitoring and water systems. This data will ensure your process is continuously under control if all the parameters are in the defined range. To release the product without batch to batch testing, only a periodic re-qualification will be needed.

The main concerns for these technologies are how to define the limits as they are not defined by regulators. Also, if these techniques are destructive; you will know that there is a bug present, but you will not be able to identify it.

Tony Cundell, Ph.D., Principal Consultant, Microbiological Consulting, LLC: In terms of microbiological testing the limitation of microbial methods must be accepted, especially chronic sampling issues and poor analytical capabilities of our methods. This is critical when determining the best application of modern microbiological methods that will advance microbial contamination control.

Another trend that will improve our understanding of product safety is risk-based microbial contamination assessments. However, these assessments require a broad understanding of formulation development, manufacturing processes, drug product attributes and clinical microbiology. These assessments must be conducted with rigor and have credibility with our management and regulators alike.

Suzanne Williams, Manager, Eurofins Lancaster Laboratories Inc.: The evaluation of microbiological results and implication of those results on the product being manufactured is challenging, specifically in regard to out-of-trend and out-of-specification results. Smaller companies or virtual companies may not have immediate, readily available or on-site access to microbiologists that can assist them when questions or issues arise.

Frank Panofen, Ph.D., Business Development Director (EMEA), Particle Measuring Systems: In microbiology, the pharmaceutical world and regulators are moving more to a science-based analytical approach. Unfortunately, historical data and quality systems are mainly based on traditional technologies, which causes the reference system to be wrong by default. This inhibits progress and implementation of systems more suitable for modern manufacturing in the 21st century.

Subscribe to our e-Newsletters
Stay up to date with the latest news, articles, and events. Plus, get special offers
from American Pharmaceutical Review – all delivered right to your inbox! Sign up now!

Paula Peacos, Senior Consultant, ValSource, Inc.: Data integrity is probably the biggest issue. There has been intense scrutiny around issues such as plate count accuracy, impact of sample hold times, recovery of slow growing organisms or viable but not culturable organisms (VNBC) etc. Lab microbiologists need to have a detailed understanding of the product and process in order to implement a strong, well-designed, risk-based and scientifically justified testing program.Thisalsoappliestoremediationinthatthisdeeperknowledge is critical to successful root cause analyses and CAPA. Lab staff in many firms do not have this deeper knowledge, and so the regulatory bodies are beginning to set this as an expectation. Firms must also to be able to defend the accuracy of their data, which is not always easy to do given the inherent variability of our current methodologies. This requires the implementation of additional controls. For example, some firms are implementing second operator verification of plate counts.

A holistic facility contamination control program is also now an expectation. This program requires identification of all process critical control points, justification of all monitoring and testing procedures, and integration of all supporting procedures, such as cleaning and disinfection, investigations, training, etc.

Tim Sandle, Ph.D., Head of Microbiology and Sterility Assurance, Bio Products Laboratory Limited: One of the biggest issues is with time-to-result, which is affected by the (largely) continued dependency upon culture-based methods and which is symptomatic by the slow take-up of rapid microbiological methods. Being able to obtain data faster, enables better responses.

While rapid methods will undoubtedly help, it remains that contamination control and good design are the most important considerations. There is little value testing if a given process has not been correctly designed to minimize the ingress of contamination, Of the different routes in, the main one remains people and the way they behave. Be it sterile or non-sterile manufacturing, designing systems and putting in place barriers to reduce the opportunity for personnel to get close to the product are paramount.

Quinton C. Inglet, B.S., RM(NRCM), Senior Lab Operations Manager, Nelson Laboratories, LLC: The Parenteral Drug Association (PDA) recently released Technical Report No. 82 Low Endotoxin Recovery. This is the first guidance document that describes the method needed to perform low endotoxin recover (LER) studies. Fortunately, the scope of this document is limited to biological drug products rather than all injectable drug product. The scope of this document is limited because the mechanism associated with LER appears to involve two ingredients often used in the manufacturing of biological drug products a chelating agent and a surfactant. LER studies are required for FDA Biologics Licensing Applications (BLA).

Some key concepts that should be considered when designing a LER study are:

  • Undiluted drug products should be spiked with Control Standard Endotoxin (CSE)
  • The volume of CSE should be less than 10% of the total volume
  • The spiked value should account for any dilution of the sample necessary to obtain valid positive product control
  • The final endotoxin concentration should be near the midpoint of the standard curve
  • Representative samples should be taken from three batches
  • A minimum of four time-points should be included

Significantly more information is included in this document making it a useful resource for those interested in low endotoxin recover endotoxin testing.

Cameron Barnard, VP Global Marketing and Product, Rapid Micro Biosystems: Topics that are relevant in Pharmaceutical QC cover the spectrum of tests performed in the microbiology lab, interference in endotoxin tests, B. cepacia complex for non-sterile products, automation should we or do we need to do it given the growing scarcity of trained microbiologists,- EM incubation strategies and implementation of “Data Integrity” compliance. The last two of these are confounded by uncertainty of the regulatory requirements with differing interpretations globally.

Have new types of drugs/new manufacturing processes put additional pressure on companies to ensure their facilities are as clean as possible? What have you observed?

Reynier: Developers of new cell and gene therapies usually start in a research-oriented lab setting and must scale their manufacturing practices considerably when moving to Phase 2 or Phase 3 trials. The industrialization and subsequent microbial control of the production process of these advanced therapies is more demanding than ever. Rapid, automated technologies offer greater control of the microbial risk at each stage of the production - from needle to needle.

Similarly, biologic and biosimilar drugs leverage increased speed and automation of microbial control to deliver critical information in a way that can actually contribute to the improvement of the production processes.

Iglesias: As sterility is only a statistical test, regulatory bodies have asked companies to improve their environmental and water system monitoring plans. This provides companies with more data, allowing more timely decisions on issues such as whether more efficient cleaning or disinfection programs are needed, if there are needs in training, or trends for specific points need to be addressed.

In this case, reliable and high-throughput ID equipment is needed for thorough investigations. It is also important to notice that sometimes we are focused on A/B area identifications, but the most important IDs are class C because contaminations detected in class A and B can be related to bad practices in the surrounding class C areas.

In my opinion, regulatory bodies should place more weight on monitoring the process and drive to improve EM programs because the amount of data obtained through this procedure is more representative, and has many more points to be tested (daily, per batch, at rest) and is more likely to find an OOS in EM than in the sterility.

For example, you may have a passing sterility result but have an EM OOS in class A which will generate a deviation and risk assessment before product release.

Cundell: The trend towards continuous manufacturing requires the attention of pharmaceutical microbiologists in terms of process equipment cleaning, in-process intermediate hold times, and the maintenance of environmental control.

The emerging field of cellular therapies poses questions as diverse as what are the appropriate manufacturing facilities for these products, e.g., biological safety cabinets, restrictive access barrier systems or isolator systems, what environmental monitoring and control programs are needed, whether sterility testing will improve patient safety, and how good manufacturing practices can be applied to cellular therapy production.

Williams: With the increase in cell therapy and gene therapy products, the need for better cleaning and disinfection processes or systems to be in place has also increased. These products have a great therapeutic potential but due to limited shelf-life and availability of material, they are administered in some cases prior to microbiological results (i.e., sterility) being available. Any cleaning or sterilization process utilized must also be shown to not have an adverse impact on the product or cells.

Panofen: There is a trend to work with more hazardous ingredients (API) with drugs (biopharmaceuticals) becoming more valuable. This puts more pressure on manufacturing to eliminate all contamination risks to the drug and the environment. Removing operators from all steps of the production is key, robotization and automation taking more import roles.

Peacos: The turnaround times for cell and gene therapy products, particularly the autologous “personalized” products can be a little as three days. As most of these are sterile products, this is a huge challenge.

Many of these new processes still require a large amount of manual manipulation to execute. Rapid sterility testing methods are not always appropriate for cell-based products, and some common slow-growing organisms like Propionibacterium acnes still require at least seven days for detection. To compensate, firms are implementing much more robust contamination control (e.g., cleaning and disinfection, gowning) programs, aseptic processing, monitoring and testing programs. The use of closed systems and single-use technologies is also being implemented. All of this is being done to protect the product and the patient through exclusion or microbial and other contaminants.

These new technologies are also putting a great deal of pressure on the industry in general to develop faster and more accurate methods of microbial detection and sample processing. The challenge here is that the method must not destroy or kill any organism as identification of any recoveries is required for investigation and root cause analyses.

Sandle: As technology advances, pharmaceutical manufacturers should be making the most of the best available solutions to enhance aseptic practices. With sterile manufacturing, for instance, the wider availability of single-use, sterile disposable technologies should be designed into the process. This extends from bioreactors to biocontainer bags for taking samples for bioburden analysis. Perhaps the most important single invention in recent years is the aseptic connector. These provide secure, simple and economical connection for high fluid throughput in upstream and downstream, and eliminate the risk that is presented from the gloved hands of personnel attempting to make a connection. With aseptically filled products, ideally there are no connections downstream from the final sterilizing grade filter. If there are (and in some cases PUPSIT has hampered the ideal), then aseptic connectors are a must.

Barnard: The introduction of personalized medicines has increased the criticality of a clean environment around the critical manufacturing site as the product is released before the sterility test is complete. Any slow growing EM organism that enters the product would be given to the patient before its presence in the sterility test could be verified. In addition, each batch is specific for one person so not only is the testing volume at a facility much larger than at a Pharma facility producing non personalized products but also the timing of this material is critical. These patients can’t wait 30 days for an investigation to be closed out and the material released.

What advice can you give to pharmaceutical companies striving to keep up-to-date on microbiology regulations and technology?

Reynier: More and more pharmaceutical companies, especially those developing the most innovative therapies, realize that using automated rapid methods to perform critical tests such as sterility and in-process controls is not only possible but also well accepted by regulators. Rewards in terms of productivity, better process control and ultimately improved patient safety, especially in case of short I would advise the companies which have not yet started to consider implementing rapid microbial methods to explore doing so. The revolution has begun; and these methods are often a competitive advantage for those who have embedded them in their processes.

Iglesias: Challenge your processes, techniques, and tests with the new requirements within the industry (data integrity, sustainability, human error, robustness) and when a gap is detected, design your transition plan from the old technology to one that will fit your process and fill in all the gaps.

Be in control of the change. It’s better to define your own transition plan. Don’t wait until an auditor raises a concern before looking to improve your process.

Cundell: With the advance of information technologies that allows an organization to readily access specialized areas without maintaining specialists like pharmaceutical microbiology in-house; this is a new challenge for general management. What is the right balance between allowing generalists to keep up-to-date in a broad range of areas, having microbiologists on staff or using consultants to provide specialized expertise? Allocation of resources in R&D is a challenge when priorities constantly change, expertise becomes dated, and new classes of drug products emerge. These are critical decisions.

Williams:

  • Research guidance documents and compendial requirements.
  • Join informational groups such as microbiology networks where you can ask microbiological questions of peers and others in the industry
  • Reach out to industry experts or attend training

Panofen: The 21st century is the time for changing approaches and evaluating options to make your science right. This includes not only implementing the right technologies but making them available for audits in the right way. The only option I see is to use paperless systems that can manage big data in microbiology.

Peacos: Laboratory management and staff should assume personal responsibility for keeping themselves up to date on these issues, and this should be a job requirement. Failure to do so greatly restricts the ability of a firm to be proactive in addressing new issues, but also to be compliant in general as expectations are changing so quickly.

Firms can facilitate this by providing an internal website with all of the necessary links to sites such as USP, Eudralex, European Pharmacopeia, ISO, Parenteral Drug Association, etc. These sites not only contain the regulations and guidance but the reasons for and thoughts behind them. This knowledge is critical for microbiologists to develop and justify appropriate, robust and justified programs and procedures.

Regular attendance at industry meetings is also important, as it is one of the best ways to find out what other firms are doing to address problematic issues. Firms should also pay for critical industry memberships as employees are much less likely to do this out of pocket. Many firms are cutting back on these things due to the expense, but this is a mistake. For the reasons stated above, the old adage “being a penny wise and a pound foolish” applies here.

Sandle: In terms of regulations, the main ones in the past year or so have emanated from Europe. These are the draft Annex 1 (which looks set to remain in draft for a while and possibly be subject to a second draft and round of public consultation) and the EMA concept paper on sterilization (which includes some good advice on controlling times in relation to aseptic processing, since the longer the time then the greater the chance for microbial growth to occur). Other than that, there is the long-waited replacement for the ISO biocontamination control standard ISO 14698, but that’s spent years going backwards and forwards.

With technology, there are an array of rapid and alternative methods. I feel that the first technologies to gain wider acceptance will be the airborne spectrophotometric particle counters that can differentiate between biologic and inert particles. Simply, the conventional particle counter will be replaced with these devices.

Another promising technology is based around bioburden testing. In this area, technologies that use ATP bioluminescence are promising. With these methods, a luciferin/luciferase enzyme reagent catalyzes the conversion of microbial adenosine triphosphate into ADP and light, which can be used to assess microbial cell numbers rapidly. The next wave of developments should see improvements to both limit of detection and limit of quantification.

Jonathan Swenson, B.S. SM(NRCM), Sr. Laboratory Operations Manager, Nelson Laboratories, LLC: The FDA website is an important resource for information to understand the focus with the industry. Obviously this is where the previous and recently published FDA guidance documents are found however there are additional areas that are helpful to review. The FDA site offers courses and webinars for continual education around guidance and emerging technology. There is a list topics for upcoming meetings and conferences the FDA will be attending. The most useful tool on the FDA website is the information listed under Warning Letters and Notice of Violation Letters to Pharmaceutical Companies. Regularly checking the letters is important to review findings, evaluate trends of inspections and help you understand if you would pass the same inspection.

Barnard: Staying abreast of regulatory changes and keeping pace with quality best practices is essential for proactive risk management. The best way to do this is by attending PDA micro and the Joint Regulatory conferences to get advance warning of changes. In addition, monitoring and reading through 483 reports received by other manufacturers through one of the companies that offers these summaries allows an update on trending regulatory concerns. New technologies will in general be launched through a web site such as Rapid Microbiology.com and will be on display at the PDA exhibition or other local chapter meetings.

Are there differences in international regulations/ guidelines for microbiology testing? If so, how do global companies handle differences at facilities internationally?

Reynier: While the main pharmacopeias are largely harmonized, there are definitely differences between countries in terms of regulations and guidelines. Navigating these differences can lead the most conservative or risk-averse companies to stick with culture media- based microbial control instead of moving to rapid methods.

Despite this, the general principles of risk assessment and evidence- based method validation and deployment are universal, and since most large companies export drugs outside of the countries in which they are manufactured they have no choice but to follow multiple sets of regulations. These companies tend to take a global approach to validation, designing their protocols to satisfy the criteria of multiple pharmacopeias. The design and pilot of the protocol is often performed at a center of excellence before transferring the method worldwide and ultimately accepted by local regulatory bodies.

Iglesias: Yes, there are differences. When you export your products to different countries, some tests will have different limits or acceptance criteria that must be met. When this happens, two approaches can be followed:

  • Perform only one test using the most stringent acceptance criteria.
  • Perform as many tests per different acceptance criteria you have.

The first approach is more desirable but risky, as you may have a batch that is acceptable for some country’s limits, but exceeds the specifications that you have defined. The second approach is difficult to manage by quality and supply departments as batches are split for different countries. You need to set a strong internal procedure to ensure that a batch that fits for one country’s specification is not delivered to a country with a more stringent limit.

Cundell: There appears to be a convergence in aseptic process compliance expectations globally. This will be driven by the revision to Annex 1 of the EU Good Manufacturing Practices, the expanding membership in PIC/S and mutual recognition of facility inspections. In the past, FDA microbiologists played leadership roles in membrane filtration methods in sterility testing, the implementation of the bacterial endotoxin assay, and aseptic processing guidelines. Regulators should step up their game.

In terms of managing aseptic process facilities, companies typically adopt the most stringent requirement, often at the expense of innovation and efficiency.

Williams: In some cases, compendial requirements or guidelines are different. While some test methods are harmonized, others are not and may differ significantly. The methods should be researched and any questions posed to the regulatory bodies. If compliance is needed for multiple methods due to product distribution, put procedures in place that will meet all requirements. This may not be possible in all cases and more than one method may be necessary to generate results that will meet the requirements.

Panofen: When you investigate the regulations there are, at this moment, differences existing since writing of such documents is always years behind the state-of-the-art. This year we will have the EU GMP ANNEX 1, which should not just be considered a European document, but one with worldwide reach. FDA and WHO significantly contributed to the exercise. This will be the future and furthers harmonization of standards.

Peacos: This is an area of difficulty as only the USP, EP and JP are currently harmonized, but not all methods in these compendia are fully harmonized. Other countries frequently model their regulations and guidance after one or more of these three, but they often incorporate some additional testing requirements and/or variations. This can be particularly challenging for firms that market world-wide and sometimes results in additional or duplicate testing requirements for a single lot.

Some firms adhere to the most stringent of the regulations, therefore meeting and exceeding the requirements of countries with less stringent requirements. Some have testing specification documents outlining specific requirements for each country a lot is to be shipped to. The batch is sub-lotted for shipment and testing is performed as required for the individual countries by the sub-lot. Some firms produce country-specific batches, although this often not cost-effective.

Others are using a risk-based approach to develop testing programs customized and justified for their specific product or process. This is most often done when alternative testing methods are utilized. Acceptance of such a program however will vary from country to country. For that matter, acceptance of alternative testing methods will vary from country to country.

Sandle: The most obvious difference is the gap that has grown between the U.S. FDA’s aseptic processing guidance and EU GMP Annex 1. I think FDA are holding back to see what happens when (if ) Annex 1 is finalized. Then, I think we’ll see closer alignment. The FDA guidance certainly needs to gravitate away from its emphasis upon monitoring and towards greater acceptance of design and the building of a contamination control strategy.

The USP remains ahead of the game when it comes to compendia. The European Pharmacopeia has some useful monographs, but the USP shows far greater research and this is due to the hard work put in by the microbiology committee. A key example is with the recent chapter on Burkholderia cepacia complex. Industry has been desperate for guidance on testing for this group of water-associated organisms, in light of the particular risk they pose to aqueous inhalation products.

As well as differences, what the industry needs is some clear guidance around a biocontamination control standard. There is a growing regulator expectation for each company to have guidance, but a paucity of information around how such a strategy should be designed and implemented.

Barnard: Key chapters such as Sterility, bioburden and objectionable organism testing are harmonized between USA, Japan and EU. Other areas such as validation requirements for RMM’s have more recently diverged in their focus and content with the USP moving away from the PDA TR33 and EP 5.1.6 texts. In this situation either make use of the technology company expertise in the area or try to combine the more severe requirements of the two Pharmacopeia to make the validation robust.

Looking ahead what advice would you give pharmaceutical companies? What areas or technologies would you advise them to concentrate on?

Reynier: Today, pharmaceutical companies can have a bigger share of voice in promoting the development and deployment of rapid microbial testing methods. With groups such as the B-POG (BioPhorum Operation group) they are starting to get organized, produce and publish evidence that help new technologies mature and be more accepted.

Based on their need for improved automation and more consistent monitoring, I would advise them to continue to structure their requests in open working groups where, together with technology providers, they can share, experiment and propose new approaches, to the scientific community and regulatory agencies that are best suited to their needs.

Iglesias: Staying in the comfort zone is not a good approach for a pharmaceutical company. Regulations, expectations and guidelines are in a slow but constant state of change, and as changes are not always easy to implement within our organizations, it’s part of our job to be prepared and ready for changes before they become mandatory.

My advice is to search for technologies that deliver advantages in lab efficiency, operational aspects (ease of training, ease of use), reduced lead times and objective readings that will keep pace with new regulations and expectations, but also meet all internal quality standards to ensure patient safety.

Cundell: It is well known that specialists do the worst job and generalists the best job making predictions. This understanding was illustrated by Isaiah Berlin’s essay “The Hedgehog and the Fox” where hedgehogs viewed the world through the lens of a single defining idea and foxes draw on a wider range of experience to form a prediction. Perhaps pharmaceutical microbiologists can be best characterized as hedgehogs. Too centered on the microbiology laboratory.

That being said, the area of most promise to revolutionize healthcare is gene and cell therapy. Huge challenges remain with this technology. They include cell potency and viability, microbial contamination control, good manufacturing practices, and treatment costs.

A single cellular therapy treatment costs hundreds of thousands of dollars. For comparison purposes, a pint of blood costs between $300- 400 and 14 million units are transfused annually.

Williams: Rapid technologies have the ability to impact micro- biological testing in ways we haven’t seen before. With those changes, a greater focus on data integrity may also be expected.

Panofen: There is a two-step approach: Use traditional methods to their limits, e.g. long-term viable air sampling. In the background, become comfortable with real-time solutions so that you can transition them to critical areas with confidence based on long-term, scientifically generated data.

Peacos: Microbial data trending and analysis is an area firms in general need to improve on. Many trending reports state only what was recovered from one period to the next, with no analysis of what that data means or what the impact is. Identifications may only be performed when an action level excursion occurs, resulting in very few identifications performed if that facility is operating in a state of control.

Microbiologists should ideally trend by more than one means. Excursion rate alone only shows how many individual samples exceeded the established alert/action limits. Adding recovery rates to the program allows one to see rising and falling populations, allowing for proactive mitigation before an excursion occurs. Regular routine identification of facility floras allows the microbiologist to effectively monitor changes in the amounts and types which can have an impact on the efficacy of the contamination control program.

A good trending software will enable one to easily, effectively and rapidly analyze the data from multiple angles for truly useful and meaningful trending reports. Using an appropriate software also greatly improves the level of data integrity of the trending program. Firms really need to invest in this. The results are more than worth it.

Sandle:Theadvice Iwouldgivewouldinvolvefocusingontechnologies that can assess the process as it happens; real-time data can ensure that processes are halted to avoid contamination of the finished product. Assessing water – the key ingredient of pharmaceuticals – in real time is a case in point. Technologies are available to assess bioburden and endotoxin in relation to water distribution loops.

Identification technologies remain important. While most companies will outsource for their occasional genotypic methods, having a robust and reliable phenotypic method is important. The fastest and most accurate is the MALDI-TOF. With this technology, cells are ionized by a laser. The ionized particles are accelerated in an electric field and enter a time of flight (TOF) tube. Here the protein and peptide molecules are separated based on their mass to charge ratio and the resulting MALDI-TOF mass spectrum is compared with an internal database. A further time saving is that Gram-staining is not necessary.

Barnard: For the future I greater focus on the automation of processes in the laboratory to minimize any human errors. The use of robots is becoming more frequent for both the endotoxin test and for enumeration of colonies on media plates. Other technologies are also allowing tests to be performed online to further remove the human from the equation.

  • <<
  • >>

Join the Discussion