#GMP #Cleanrooms & Cleanroom #HVAC: A Practical Approach


Identifying the most practical approach to achieve Good Manufacturing Practice (GMP) cleanrooms and cleanroom HVAC in the pharmaceutical industry does not require an “out of the box” or innovative approach. It rests, rather, on the comprehension of and adherence to a set of basic rules that have been penned by several GMP regulatory authorities. Rules are nevertheless open to subjective interpretation and herein lie some potential pitfalls. The International Society for Pharmaceutical Engineering (ISPE) provides invaluable and much needed guidance in this regard.

The most commonly used GMP regulations that govern the design of pharmaceutical cleanrooms and cleanroom HVAC systems include the EU GMP, PIC/S GMP, FDA cGMP, and WHO GMP.

What is clean and how particlebased cleanliness is specified depends on what standards are applied. The world community of cleanroom designers mostly follows the ISO 14644 standard family for this purpose. Cleanroom designers and builders should concentrate on the first five parts of the standard family depicted in Table 1.

ISPE has provided a set of tools that facilitates a clearer understanding of the application of di€fferent GMP regulations, standards and guidelines in everyday work. Without these baselines it would be difficult to find suitable solutions to GMP matters. ISPE creates a bridge between pharmaceutical engineers and regulative authorities by writing baselines, good engineering practices and monthly articles in its ”Pharmaceutical Engineering” magazine.

The ISPE publication series includes ISPE Baselines, ISPE GAMP Guidance documents, ISPE Guides and Good Practice Guides, ISPE Investigational Products Resources, and ISPE Regulatory. This excellent source of reference information combined with engineers’ practical experience allow most clients’ GMP related questions to be solved. Local laws and codes naturally have to be applied in every aspect in order for project goals to be fulfilled. Once a project has started the ISPE general V-model should be followed (see Figure 1)

According to the V-model all GMP projects should start with a properly executed risk assessment based approach which leads to the appropriate Validation Master Plan (VMP) and finally to project-specific User Requirement Specification (URS). All GMP cleanroom projects should follow this generally accepted route:



Every phase has to be approved and completed with documented confirmation before proceeding to the next phase. It all starts from the user requirements and ends with the very same user requirements. All requirements in the URS need to be fulfilled or approved with documented deviations.



User requirements:

When the URS for cleanrooms or cleanroom HVAC is written in the conceptual design phase by the plant operator (often with engineers’ assistance) and approved, one can proceed to the basic design phase (or the functional design). the URS is arguably the most important document in the whole GMP project as it defines all the users’ (GMP) critical demands for the process, clean utilities, cleanrooms, cleanroom HVAC and black utility. URS in the GMP context means a documented definition of the key requirements stated by the user.

The URS must state what GMP regulations should be followed and what cleanliness grades are required. EU GMP and FDA cGMP requirements are the two mostly referred to but also WHO and Japanese regulations can be used if applicable.

Cleanrooms can be constructed in various ways, but the first thing that has to be solved is the layout, which is governed by basic rules. Figure 2 displays the main principle of a shell-like barrier system where the cleanliness grades are always separated by personnel airlocks, material airlocks or pass-through cabinets.

Layout design should only be started once a clear understanding of the user requirements has been gained. The basic rules for airlocks and pressure cascading regimes should, however, be checked first. Here again ISPE has some useful models. See Figure 3 (overleaf ) for a layout with cleanliness grades and pressure cascading.

There are some diff€erences in cleanliness grades between the US and EU which need to be kept in mind when designing cleanrooms for diff€erent regulatory environments. US GMP covers three cleanliness grades: supporting clean areas in two grades and critical areas. EU GMP includes four cleanliness grades: A, B, C and D. This di€fference aff€ects design especially when sterile drugs are produced in aseptic processing.

The crucial role of HVAC:

Once layout work is completed the HVAC designer can begin working. HVAC is only a small part of a cleanroom – but a very important part. Without a well-functioning HVAC system the desired conditions for production might not be achievable. HVAC systems for cleanrooms are relatively expensive and take up much space, but are essential for the critical product parameters.


HVAC systems also represent large operating costs. It firstly needs to be decided what type of ventilation principle would best satisfy the URS requirements: recirculated air or outside air? Recirculated air means a ventilation system where e.g. 80% of air flow is in constant circulation and only 20% is replaced with fresh air from outside. A 100% fresh air system uses non-recirculated air from the outside.

It is preferable to use recirculated air in a cleanroom ventilation system if it is not prohibited for any reason as the use of recirculated air reduces energy consumption and emission levels. Direct and indirect impact systems

It is of critical importance to determine whether the HVAC system has a direct or indirect impact on the product. According to the ISPE guidelines a direct impact system “is expected to have a direct impact on product quality. These systems are designed and commissioned in line with Good Engineering Practice and in addition, are subject to qualification practices that incorporates the enhanced review, control, and testing against specifications or other requirements necessary for cGMP compliance.” ISPE indicates that an indirect impact system “is not expected to have a direct impact on product quality, but typically will support a direct Impact system. These systems are designed and commissioned following Good Engineering Practice only. Indirect impact systems can affect the performance or operation of a direct impact system.”

Once the impact type of the system has been determined, the appropriate commissioning and validation activities can be applied.

Critical parameters:

The so-called critical parameters for products manufactured in a cleanroom environment need to be specified in the URS. According to the ISPE guidelines there are several factors that need to be considered and the onus is on the designer to gather all the relevant information (see the info box for details):

Validation of cleanrooms and cleanroom HVAC:

The first validation activity in cleanroom and cleanroom HVAC projects is the DQ whereas the last is the approval of all the required design documents. When everything is complete a design qualification (DQ) report is drawn up and signed. It is a generally applied approach that construction work cannot be started before design qualification is done.


The next step is construction, which is followed by commissioning (C), installation qualification (IQ), operational qualification (OQ) and finally performance qualification (PQ). Each has to be approved before the next step can start and approval of all steps has to be documented.

ISPE defines commissioning as a “well planned, documented, and managed engineering approach to the start-up and turnover of facilities, systems, and equipment to the end-user that results in a safe and functional environment that meets established design requirements and stakeholder expectations”.

Validation on the other hand “ensures that the facility and system qualification (DQ, IQ, OQ and PQ) requirements are communicated and met.”

How to Remove Water Spots & Oil On Laboratory Glassware


Q. We see white water spots on cleaned and depyrogenated vials. Looks to be scale from the washer. Do you have a recommended cleaning agent for vial washing? I am told that use of detergents is not an industry norm. We also have baked on process fluids (oils) and would like to know if it is an industry norm to use detergents to remove such substances from glass vials used for injectable drugs. Can you provide me any literature on the subject?
A. Use of free-rinsing, interfering residue free aqueous detergents, such as those manufacturing by Alconox, Inc. are very much industry norms.  This will cover lab and process equipment across biotech, medical device, pharma, cosmetics and scores of other industries as well.
White spots on automated washer cleaned vials is likely some variation of water spots. If any glassware or equipment is loaded in to a washer that has a geometry that tends to hold water, that will be a candidate source.  For example, an upturned cup shape such as the bottom of a graduated cylinder will hold a dose of dirty wash water which then gets spread on the other glassware by the rinse cycle.
It is very important to pay attention to how glassware and equipment is loaded in to a washer so as to minimize trapping any water from cycle to cycle – often by appropriately tilting vials, glassware and equipment, it can be oriented to drain completely and not trap dirty wash solution. 
The above is with the assumption that deionized water for rinsing is being used in your automated washer.  If you are using tap water for rinsing (or washing in some cases), then the white residues can be simply evaporated tap water that deposits calcium, magnesium and iron residues (water hardness).     
The scale/water spots from washing are typically the result of calcium, magnesium and iron hydroxide/oxide deposits that form from the alkalinity in the detergent in the wash cycle.  If deionized water is used in the wash cycle there is less likelihood of scale formation.  
If there is scale formation, water spots, white spots, etc. the use of an acid rinse cycle using Citrajet® Low-Foam Liquid Acid Cleaner/Rinse detergent will eliminate the scale buildup.  Many automated lab washers have the capability of an acid rinse cycle.  If the automated washer does not have an acid rinse system, then a periodic maintenance cycle done in an empty washer by adding Citrajet detergent to the bottom of the washer will help control scale formation in a washer.  

Pharmaceutical Facilities And Equipment Risk Management


This two-part article focuses on risk management of facilities and equipment. It describes how a risk-based approach to facilities and equipment management fits into an integrated, effective quality systems structure. The principles discussed are equally applicable to all quality systems. The focus is on facilities and equipment because they represent a broad range of risk to product quality and are one of the key quality systems commonly identified in the pharmaceutical manufacturing industry.
Also, the discussion of risk is limited here to quality: safety, purity, strength, and identity of product. Risk to health, safety, the environment (HSE), and business continuity are not considered; however, the principles discussed are fully applicable to those factors, while the risk controls applied may be different.
This first installment provides background and introduction to quality systems and quality risk management and their relationship. The second will look at facilities and equipment risk more specifically and provide a framework for effective risk assessment and quality system application.
Background
In 2002 the U.S. FDA announced Pharmaceutical CGMPs for the 21st Century — A Risk-based Approach. The purpose of this initiative was nominally to modernize the FDA’s regulation of pharmaceutical manufacturing. Moreover, the initiative encouraged a new culture of continuous improvement within the manufacturing segment of the industry. In the years following the launch of the program, the FDA has adopted its core principles, as follows:
  • Risk-based orientation
  • Science-based policies
  • Integrated quality systems orientation
  • International cooperation
  • Strong public health protection
Throughout the initiation, implementation, and continuation of this modern approach, the primary focus has remained “to minimize the risks to public health associated with pharmaceutical product manufacturing.”
During this same time, these core elements have become increasingly accepted and practiced by the international regulatory community. This is most in evidence in the works of the International Conference on Harmonization (ICH), particularly its Q8, Q9, and Q10 working groups.
FDA publications, and presentations made by FDA representatives, emphasize that the approach remains aligned and fully compliant with cGMP regulations. In addition, exhaustive comparison of U.S. cGMPs with international regulations resulted in the conclusion that, although there are differences, the regulations are substantively the same and share the same focus of protecting the public health.
Perspective
First rule of quality risk management: “At the end of the day, the only thing that really matters is the patient.”
From the industry perspective, the response to this initiative has been somewhat mixed. Although many companies have established a quality systems structure and introduced some risk-based approaches, there seems to be considerable lag in developing a truly integrated and systematic approach to quality.
This is not entirely surprising considering the history of cGMPs in the U.S. and the rest of the world, which strongly emphasizes “compliance.” Of course, compliance with regulations is necessary to legally and ethically manufacture drug products. However, over time and through long and painful experience, the industry apparently lost sight of the real reason for compliance. That reason is, of course, to ensure quality.
The quest for compliance should not be misinterpreted as a failure to strive for quality. It should be recognized, however, that the emphasis on compliance, often in the pursuit of even the most insignificant and trivial internal procedural requirements, often detracted from the true goal of product quality. It is revealing that during an era when firms were rigorous in pursuing procedural perfection, there were still many warning letters and consent decrees, even among major manufacturers. Resources were frequently expended on issues that meant little with regard to quality. Quality remained important, but it seemed as if much that did not contribute to actual quality was equally as important. The result was that these “faux-critical” issues diverted resources from their proper focus.
Second rule of quality risk management: “If everything is critical, then nothing is critical.”
Until 2002, even the FDA did little to alter this trend. There was an industry perception that the FDA, the EU, and other regulators abhorred change, even change for the better. Consequently, the industry fell into the trap of compliance for the sake of compliance. Even 21CFR Part 11, which was intended to move technology forward by providing a framework for the use of computers in the industry, resulted in the development of such a dogmatic approach to computer validation that actually inhibited our use of automation.
Third rule of quality risk management: “Dogma breeds ritual; ritual breeds waste.”
As the industry adopted dogmatic and ritualistic approaches to deal with quality issues, we tended to crowd out science and common sense. This is not to say that rigorous procedures and documentation are not necessary. It is to say, however, that these procedures and documents are not sufficient to ensure quality. The core of quality is in the product and the process.
In its Final Report on GMPs for the 21st Century (2004), the FDA resolved to regulate the industry through sound science and to encourage the application of technology and the principles of quality systems to facilitate continuing quality improvement. The FDA and ICH set the framework for the future with guidance on pharmaceutical development, risk management, and quality systems in ICH Q8, Q9, and Q10, respectively. The emphasis has clearly become product quality and the protection of patient welfare. It remains for the industry to understand and follow suit.
Quality Systems
ICH Q10 describes the application of a quality systems approach across the life cycle of a product. This paper focuses primarily on the commercial manufacturing stage of the life cycle, as this is where cGMP regulations have the greatest impact. Some elements of technology transfer are included as being necessary to define product quality attributes and contributing parameters.
ICH Q10 stops short of delineating quality systems in detail, although it does stress four major areas:
  • Process performance and product quality monitoring system
  • Corrective action and preventive action (CAPA) system
  • Change management system
  • Management review of process performance and product quality
These core activities can be organized into many different structures, all of which may be effective. A firm’s choice depends on product mix, size and resource availability, process complexity, and many other factors. Some firms choose a major system/sub-system approach. Others organize into many separate systems. It is, however, convenient to organize along the lines that the U.S. FDA uses for its systematic approach to facility inspections. The following are major quality system categories defined by the FDA:
  • Quality management (usually includes change management, documentation, deviation management, training)
  • Laboratories (includes testing for materials and product)
  • Materials management (includes receipt, control and shipping of materials, components, and products)
  • Production and process controls
  • Packaging and labeling
  • Facilities and equipment
The quality systems provide the foundation for recognizing and controlling risk in manufacturing of product for human and animal use. Some of these controls are specific to the system within which they usually reside (e.g., maintenance and calibration within facilities and equipment), while others are foundational and applicable to all systems (e.g., documentation, change management, deviation management/CAPA, training).
The Product And The Process
Common interpretation of GMP regulations notwithstanding, all quality is embodied in the product. The most important factors in achieving quality product are the processes that are employed to manufacture it (including packaging and labeling). These principles are basic to establishment of a science- and risk-based approach to quality. Therefore, a thorough understanding of the product and its quality attributes is essential.
Quality attributes are criteria specified to meet the expectations of the user. In our industry, the user group is a diverse one, including not only patients, but physicians, nurses, pharmacists, other medical professionals, distributors, retailers, and, of course, regulators. Quality attributes may range from the sterility of an injectable to the position of a label on a shipping carton. The quality impact of this broad range of attributes is equally as broad. In order to apply a sense of risk, we often describe a select set of attributes as “critical to quality.” The designation of quality-critical attributes (CQAs) is a relatively recent movement (at least in my experience) aligned with the current emphasis on risk management. By defining CQAs as having a direct effect on product quality (according to U.S. regulation: safety, identity, strength, purity, quality), we enhance our focus on those elements actually affecting the patient.
For many, if not most of us, our influence on product quality is related not to our ability to test for CQAs, but more to the process that manufactures the product. For the sake of this discussion, “process” needs to be broadened to include all influences on quality, including materials, personnel, facilities, controls, and equipment. Those parameters or characteristics that have a direct effect on CQAs we will call “quality-critical requirements” (QCRs). (Note that this is a broader category within which “critical process parameters” – CPPs – fall.) In order to understand and manage the risks associated with facilities and equipment, we need be aware of the QCRs.
The most effective way I have found to identify QCRs is by developing a user requirements specification (URS) for each system, wherein each requirement is categorized as essential for quality (includes GMP compliance), HSE, or business. For example, the throughput or capacity of a system relates to business requirements, while the accuracy of temperature control might be necessary to limit byproduct formation, making it essential to quality and thus a QCR.

What Should You Know About Pharmaceutical Primary Packaging


Starting Early

In phase I, researchers are testing a new drug or treatment in a small group of volunteers for the first time to evaluate its safety, to determine a safe dosage range and to identify side effects. The amount of drug product manufactured is relatively small and easy to control at this point in the product’s development.

But when a clinical trial progresses, the formulation itself and the dosage form may change as factors like stability become better characterized. As a result, sponsors may also want to consider the desirable commercial format in later stage clinical studies

Patient and clinic needs will also change as the drug travels from small phase I trials to large multi-center phase III trials.

For flexibility around dosage it is often best to use a granule dosage form in capsules, and the dose can then be adjusted simply by changing the weight or number given.

Catalent conducts stability trials of the study drug alongside the clinical trial and will likely be testing different primary packaging configurations such as solid dose tablets or capsules in blister packs and bottles as part of that process.

Stability data will influence the pack design, and so the dosage form used at this stage may bear little resemblance to the final commercial form.

Packaging is also based on the volume of anticipated patient recruitment. Randomization of patients to treatment types can be stratified to test dosing of patients by gender, age, body weight and other factors. From a complexity point of view, the pack designs themselves don’t tend to be complex.

“What does get more complex after phase I are factors like randomizations and labeling because the customer and Catalent are trying to develop the best dosage form for patients. Stratified randomizations may be used when testing characteristics such as body weight, gender, age alongside dosage strength. As a result, the randomizations tend to be quite complex due to the number of variables involved ,” said Steve McMahon, process leader at Catalent.

Most often, supplies are sent to the clinical site rather than directly to patients, and labeling might be more complex because labels will include a unique number that links back to the randomization and how a patient is being dosed, McMahon said. Clinical sites usually manage inventory in-house due to shorter turn-around times, and so their ability to store items might also come into play.

More becomes known about the study drug as the trial progresses to phase II trials, where the drug is given to a larger group of patients to test for the right dosage and further evaluate its safety.

Based on stability data and other feedback, pack design and materials may change to a more appropriate dosage form, such as tablets, extended-release capsules, infusions or injections.

“If a long expiry date is not possible, then you might try and simplify the pack design,” he said, noting that in phase II, more stability data will be collected in a rolling stability trial, in which case expiry dates are extended as more data are captured.

“There are lots of different factors that we’ll start with, depending on the expiry date,” McMahon said.  “All the time you have to be mindful about what the customer wants to do in phase III and what is required when you commercialize the product.”

Some packaging protects stability better

One of the best forms of primary packaging to protect the product and improve the stability of a product is cold form blister packets. Materials used have a polyvinyl chloride (PVC) or polyvinylidene chloride (PVDC) seal, which is dense and resistant to temperature and humidity.

“The good thing about blistering is that only one tablet is popped at a time, so the stability of the rest of the drug product is maintained,” McMahon stressed. “Whereas, when using a bottle, once the bottle is opened, the entire pack is exposed to humidity.”

Storage conditions need to be considered in the dosage form and pack design he said, noting that a site could be storing a product at -80˚C while the trial is ongoing, but the product might be distributed at -20˚C.

These types of decisions surrounding the dosage form can also affect turn-around times for trial sites.

“For example, Catalent had a customer for a relatively large-scale phase II trial, and were doing packaging runs about every three months. Catalent did the initial primary packaging and then secondary packaging. Then every three months the customer received more stability data and Catalent was required to re-label packaged supplies with new expiry dates.”

“From a customer point of view this was costing them more money.”

He explains that the practice of putting expiry dates on clinical trial packaging is required in some jurisdictions. In the U.S., expiry dates are not compulsory on packaging for clinical trials because they can be controlled by interactive technology [IRT]. Expiry date information is also sent to the site.

The trial medication is managed through an automated pre-determined order system based on initial order and resupply parameters in the IRT system. Since the IRT manages expiry dates, it makes the whole supply chain a lot smoother. It also helps control costs due to less risk of over producing patient kits which may expire before they can be dispensed.

But in Europe, even if an IRT is used, the clinical trial packaging still needs to be labeled with expiry dates, he noted.

“It’s important to have strict standard operating procedures of how to do these things, and Catalent has extensive experience in doing multi-pack designs and labeling,” McMahon stressed.

“Even though it can be complex, we will produce batch documents that detail exactly what we need to do to for quality reviews internally and for the client, and after execution of that production run, it gets reviewed again. The controls in place for clinical trials are more restrictive than in commercial drug manufacturing, because the controls include management of blinded materials.”

By the time a drug gets to phase III trials, where it will be given to larger groups of patients often multi-country or global to confirm its effectiveness, a significant increase in production is quite common.

Companies will also start running multi-center studies in multiple countries, and production could shift from 250 units at phase I to two million units or more at phase III.

Ensuring patient compliance

Packaging and labeling in phase III are developed with regulatory and legal requirements, patient and sponsor needs as well as commercial requirements in mind, and the study may be randomized and blinded against a placebo or more likely a market leading comparator. Clinical trial packaging will often move to wallets or cards with blister packs or individual bottles that are sent to the clinical sites, which then distribute them to patients who generally self-administer at home.

 In phase I, the drug is administered at a specialized Phase I clinical site that has dispensing pharmacists with GMP training who can make-up dose capsules  or dispense tablets from a bottle. This translates into strict patient compliance. But when patients start taking the study drug at home, this is when compliance can start to slip. One solution is to use packaging designs that make it easier for the patient to comply with the protocol dosing schedule, such as sun and moon symbols on blister cards to indicate AM vs. PM doses.

“One of the big things that put patients off taking medication is non- patient centric packaging and labeling,” McMahon stressed. “If they don't understand the pack or the labeling or it's cumbersome, they may not take the drug.”

Why outsource clinical supplies?

McMahon said that most pharmaceutical companies are geared up for mass manufacturing, and they don't really have the scale and capability to do smaller scale clinical runs.

Some companies have separate units for clinical trials, but that often ends up costing more in the long run to keep such a unit up and running.

“It's beneficial for big pharmaceutical companies to use integrated providers like Catalent because we've got the experience and expertise of doing clinical trials, and we have the equipment to handle most packaging designs and clinical trial packaging. Some customers might have a high potency or controlled drug and their facility might not be set up for that.”

There are several factors that biopharmaceutical companies should take into account when vetting a drug delivery solution provider such as Catalent or contract manufacturer (CMO) for outsourcing primary packaging. Some of these vendor considerations include:

  1. The ability of a vendor to expand packaging capacity on short notice.  This may be due to constraints on storage, labor or equipment capacity at the customer’s own sites;
  2. Taking advantage of a vendors experience/specialty in a particular packaging field such as blister packaging of solid dose forms;
  3. Reduced risk to drug development timelines due to vendor’s’s experience in packaging and distribution;
  4. Major cost savings in not having to incur capital expenditure on specialized equipment, recruitment and training of staff;
  5. Vendors can help reduce drug development time with different packaging strategies;
  6. A vendor’s geographical presence of packaging and distribution centers can provide logistical savings;   
  7. The vendor will usually have more flexible production timelines with the capability to manage milestones on a project critical path; and
  8. Some vendors have the necessary experience and contacts to procure comparator products from multiple sources.

McMahon said the quality unit and the Qualified Person (In Europe) at a company will be able to advise if a planned packaging process or pack design is compliant with Regulatory Authorities such as the FDA, MHRA good manufacturing practices regulations or country specific requirements.

The customer quality unit would also typically audit and approve the CMO before placing work.

The CMO’s packaging design group can also confidentially leverage their collective experience across a variety of projects to help current customers devise their optimal packaging solution. .  This guidance can take the form of best practices or lessons learned that the customer may not have been aware of on their own.

The CMO can provide feedback on whether the pack design is suitable for packaging and distribution to clinical sites and patients across the world. It can also inform on whether the pack is the correct size to match the various phases of the protocol and the dispensing visits.

Costs can also be managed by consulting with the CMO, because the components used to make a pack design can increase or decrease in cost but can also add value to a patient through ease of use. This would give increased probability of patient compliance to the trial.

Investigator and clinical staff, as well as the patient can also provide valuable feedback on the pack design, particularly about whether the product is easy to dose and if labeling instructions are easily followed.

“We have a patient-centric initiative at Catalent with the aim of putting the patient first.  Part of this initiative resulted in some of our pre-production people visiting a clinical site to get feedback,” McMahon said, noting that staff are often surprised how small the clinical site can be and why storage might be a problem. Alternatively, a doctor or a nurse in a large hospital will want packaging that stands out and very clear labelling because they could be running multiple trials, and they need to be able to identify the right clinical supplies quickly.

Make primary packaging decisions early

To accelerate drug development, companies need to consider clinical trial packaging strategies as early as possible in the development process.

“There are several factors that need to be considered from the customer’s point of view, such as whether they have the capacity and resources for the packaging for a trial. If the answer is no, then the customer has to start thinking about outsourcing straightaway,” he said.

Some customers will have capacity for a phase I trial or a small phase II trial, but then a CMO will be needed to handle Phase III. For a smooth transition, companies need to plan for this early to leave room for auditing the CMO to make sure they have the necessary quality standards.  Formal agreements also need to be put in place and disclosure that outsourcing is going to occur should be included in any trial documentation.

“That is not a quick turnaround, and it could take up to six months to prepare, which is a long time during that critical stage,” McMahon advised.  

Companies should also consider the CMO’s packaging and distribution network. “For example, what if a trial needs to be conducted in Australia, what are you going to do? Does the customer require trial material to be shipped from the UK? If it's a temperature-sensitive product, I don't think that's necessarily a good idea. Working with a CMO with an extensive distribution network and the right expertise can give you other options that may be more advisable.”

He said that Catalent has global manufacturing capability across six continents, and can seamlessly switch gears to meet clinical trial needs, so shipping to remote regions in Australia wouldn’t be a problem.

Another challenge for drug companies can be sourcing a comparator product, because innovator companies may put up road blocks to sourcing their material until they are assured the product will be used within an appropriate setting and not subject to parallel trade as they are legally responsible for that drug. 

“This can either be a high mark up on the original cost of the drug, quantity or lot limits, an extremely long lead time on it or difficulty in obtaining the necessary paperwork,” McMahon said.  Catalent has a specialized team with well-established relationships with innovators (preferred route) and wholesalers to expedite the sourcing and purchasing of comparators.

The complexity of some study designs in clinical trial packaging can also create challenges, and the different types of clinical supply models that a customer may follow can have a major impact on the packaging strategy.

The traditional clinical supply model, called supply-led packaging, is a centralized stock-based approach that uses discrete primary and secondary packaging runs to bulk-ship finished patient kits to clinical sites and depots based on estimated demand.

Under this model, primary and secondary packaging is undertaken at centralized GMP packaging facilities where stock is built up well in advance of actual need. This model can cause an under- or over supply due to variations in patient recruitment rates, which can end up being quite wasteful or carry a greater risk of supply not meeting demand.

Just-in-time labeling

The just-in-time labeling model uses discrete primary and secondary packaging runs the same as in the traditional model to produce base-labeled patient kits that are held within a central physical inventory to await final labeling. 

Under this scenario, packaging is accomplished via large-batch runs. This model is efficient when the study involves materials that are not likely to require expiry update management. Or, it could a good option when the expiry date is very short and the product is on a concurrent stability program, and the most up-to-date expiry date can be added at the point of shipment.

“A potential issue is the customer still has all these bottles produced, so you could still be producing too much product. What does give you the flexibility is when recruitment is even across different countries.”

However, labeling at the time of shipment can result in longer associated lead times and result in potential bottle necks. In addition, more quality resources are needed.

Yet another option is multi-language booklets that have individual country-specific instructions inside, and then that carton can be sent anywhere around the world. 

Demand-led supply model

The demand-led supply (DLS) model is a dynamic, continuous GMP approach to secondary packaging, labeling, release and distribution. 

Under this model, made-to-order patient kits are shipped to clinical sites from regional facilities based on actual patient demand. Secondary packaging takes place at a regional, full-service packaging facility where a supply of unlabeled but uniquely coded ‘bright stock’ is placed in advance. Accordingly, the primary packaging plan should take into consideration the lead time required to ship bright stock from the central primary packaging facility into the regional packaging facilities. By using bright stock, not only can the exact kits needed be made-to-order, but drug product can potentially be pooled for use across multiple protocols which can potential reduce the amount of drug that goes unused..

Then, based on forecasted demand, this bright stock is distributed to regional GMP facilities where it awaits further processing. Secondary packaging is completed and the finished patient kit is shipped to the investigator site where it is needed within a matter of days in response to on-demand orders received via IRT.

The advantage of this model is that secondary orders are fulfilled based on what is actually needed by the sites, leading to more efficient use of stock, significantly reduced risk of stock-outs and virtually eliminating the need to update expiry labeling at the investigator site.

Since labeling is not completed until just before the material is sent to the site, expiry dates can reflect the most current stability data available. This translates into minimal drug waste and can be a major cost savings for some products.

One future enhancement for labeling is 2D bar codes. Some will even have hyperlinks inside the bar codes that enable the user to scan them with a smart phone to get country-specific instructions.

The demand-led model is also a good choice when a comparator drug is needed and is difficult to source, very expensive, or in short supply. This model is also well suited for orphan drug products and certain specialty products that are often subject to extreme limited availability and very tight distribution control.

Quality and due diligence

The approach to good manufacturing practices should be the same regardless of the phase of a clinical trial.  It is critical that quality personnel are involved in the development of a study from the start to ensure compliance to GMP at all stages.

The complexity of a study means greater involvement and understanding of quality personnel.  “Compliance to GMP, the Orange Guide, FDA regulations for labeling and packaging prevents issues from arising,” he said. 

“Spending more time upfront in planning, taking care to use established standard operating procedures and best practices produces greater efficiencies and reduced errors,” McMahon stressed.

And, if the complexity of a study requires a deviation from an SOP, then this needs to be documented pre-production appropriately.

Compliance ensures faster approvals for label artwork, batch documentation and ultimately faster availability to the patient. That translates into fewer delays in production and ultimately clinical trial data that is accepted by regulators.

For inspecting primary packaging, Catalent uses cameras to inspect the blister packs after they've been filled, or the camera can also be set up before the packs are sealed as well.

The primary packaging materials themselves can be challenging, because certain materials are more difficult to work with. For cold form blister packing, the actual pocket is formed by pressure, and the material basically bends into a pocket.

When it comes to high potency drug products, many CMOs have specialized packaging suites that are used exclusively for these types of drugs because they typically can’t be packaged in normal primary packaging rooms. Pharmaceutical companies that do not typically have potent and cytotoxic drugs in their portfolios may find that they do not have the necessary infrastructure in place to package these drugs on their own.

Laboratory Glassware Washer Validation Benefits

To appreciate how labwasher cleaning validation can help your organization, it helps to take a bird’s eye view of its objectives. They are to ensure consistency, quality and compliance with the FDA’s Current Good Manufacturing Practice (cGMP) regulations — regulations with a laser focus on a manufacturing process’ ability to create safe and effective products. Again and again.
The validation procedure starts with examining the big picture, the cleaning process, and then investigates the equipment that plays a pivotal role in executing each task. The purpose of the exercise is to furnish evidence to assure regulators and customers. This data you gather shows in black and white that your cleaning process will consistently deliver laboratory glassware that meets required specifications for cleanliness.
Many benefits flow from these objectives. They include:
1. Conformance with Regulatory Requirements
Validating all of the processes involved in bringing a drug to market is not an option. It is an FDA requirement. The most obvious reason for validation, therefore, is to ensure that your lab glassware cleaning process is in compliance with the FDA’s regulations.
2. Reduced Downtime
Since labwasher validation guarantees residue-free washing results every time, it enables you to avoid downtime due to dirty glassware.
3. Improved Quality
In pharmaceuticals, quality cannot be an afterthought. Because validation focuses the two essential ingredients of pharmaceutical quality, safety and efficacy, it is a natural step toward a goal of quality improvement. It make sure that medicinal products will perform according to expectations and the risk of contamination from impurities is brought as close to zero as possible.
4. Reduced Risk of Recalls
Contamination of drugs due to poor cleaning processes can compromise their safety and effectiveness, resulting in health hazards and even fatalities. Product recalls follow quickly on the heels of tainted drugs, shaving bottom lines, eating up time and sullying reputations. Clearly, all manufacturers strive to avoid such disasters.
5. Productivity
The validation process makes sure you have a systematic approach that produces good results without fail. In this way, it improves productivity. Because of the efficiency-boosting power of validation, even labs involved strictly with preliminary research and discovery that work outside of cGMP environments, are validating their cleaning processes. These labs include, for example:
  • Non-pharmaceutical labs that create products for clinical testing under cGMP
  • Contract labs that must use ironclad to avoid cross-contamination between multiple customers’ orders
  • Quality control labs
  • Labs involved in preliminary research and discovery
Obviously, there are many benefits to labwasher validation that go beyond compliance with cGMP regulations. These include increasing quality, productivity, and speed to market as well as reducing downtime and recalls. Given the many advantages of labwasher validation, you may be ready to get started with the process. How you do so will depend on the precise nature of your operations. However, there are usually three steps. First, define the cleaning process. Next, describe it in detail, usually including written standard operating procedures. Finally, qualify the labwasher and validate the cleaning process.

The Greatest Return From Your Poorly Soluble Molecule


Currently, about 70% of new molecular entities (NMEs) exhibit poor solubility in water and require some form of enhancement in order to achieve sufficient bioavailability, leading to tough questions about how to proceed for biopharmaceutical companies developing such compounds.

The sheer number of potential strategies for improving the solubility of a compound — which include various methods of particle size reduction, solid dispersions, salt formation, lipid formulations, inclusion complexes, nanocrystals, and a wide variety of other technologies — can overwhelm many developers, leaving them unsure how to choose a path forward.

At the same time, companies are under significant pressure to advance development programs in order to move to the next stage of funding as quickly as possible. In their rush to advance their molecules into the clinic, some biopharma companies ignore questions about solubility until later in development when the issue becomes unavoidable. Many other companies are willing to roll the dice and pick a solubility-enhancement technology based on a low initial cost or because they think that the process would be simple to scale up or a trusted consultant advises that the technology works for most molecules.

Of course, one of the reasons so many options exist is because there is no such thing as a one­-size-fits-all strategy for low solubility compounds; an enhancement approach that works for one molecule will not necessarily work for another with different physical and chemical properties.

Many developers are unaware that it can take just a few weeks to fundamentally evaluate a molecule’s physical and chemical properties, determine the ideal approach for that molecule and recommend a customized solubility solution. As a consequence, companies often choose a technology without undertaking a rigorous assessment. In some cases, that gamble may pay off, and they may luck into a formulation that provides the required exposure on the first attempt. In too many cases, however, biopharma companies find themselves going down a dead-end path that eats up tens of millions of dollars and years of time, while their competitors go straight to Proof of Concept with the first formulation strategy they try. Unfortunately, it is not uncommon for companies to formulate and reformulate compounds dozens of times over as many as 4 to 6 years without success, unnecessarily delaying or even killing the project.

If a formulation fails in Phase I due to insufficient bioavailability, experience has shown that redoing all of the necessary formulation and process development activities, including stability testing and validation of analytical methods, can easily require up to 12 months of time at a cost of $500,000 to $600,000.

Switching to a new approach is also not desirable, as it can require finding a new solubility vendor, a process that is likely to take an additional three to six months of dedicated effort to perform due diligence and negotiate a new master service agreement. Add in the cost of repeating a Phase I trial, which according to a 2014 U.S. Department of Health and Human Services report averages $4 million, and each subsequent attempt adds $5 million to R&D costs. In the meantime, the company is failing to collect milestone payments, and according to a 2012 Journal of Applied Clinical Trials article by Ken Getz of the Tufts Center for the Study of Drug Development, each day of delay in getting to market costs an average of $1.3 million in lost prescription sales.

Without a deep understanding of the science behind solubility enhancement, there is no guarantee that a second attempt will work any better than the first. When developers lack that understanding, they also lack the ability to discriminate the potential benefit of one technology over another, so they may wind up trying all of them. And when all of their attempts fail, it can be very difficult to draw a definitive conclusion as to whether or not the molecule can be developed.

In the end, if the company’s empirical search for a solubility solution fails to generate the necessary bioavailability, or if it produces a formulation that can’t be scaled up, or if the company has spent years on a compound that was never amenable to development, the entire company could be at stake. Fortunately, it is possible to get reliable, scientifically based guidance to select the approach with the best chance of success in a very short amount of time.

For example, in one case, a company that was developing a poorly soluble molecule for a pain indication had tried almost 50 different formulations, using five different drug delivery technologies including micronization, wet granulation and nanocrystals, while failing repeatedly to meet exposure targets in pre-clinical and Phase I studies. Following numerous failures, the company’s business partner decided not to advance the molecule any further and returned the rights to the developer.

Once the developer decided to consult an experienced solubility enhancement team at a large contract development and manufacturing organization (CDMO), it took only a few weeks for the team to evaluate the molecule and recommend a spray-dried formulation that succeeded immediately in pre-clinical studies. The new formulation demonstrated substantially higher bioavailability in a dog model, then achieved an 8 ½-fold improvement in Cmax and a five-fold increase in bioavailability when it advanced to a Phase I PK study.

For another company that had also tried more than 50 formulations of its poorly soluble compound over the course of several years without achieving any significant increase in bioavailability, a scientific evaluation of the molecule and potential delivery technologies determined definitively that further development was neither financially nor technically feasible at that time, allowing the company to end that program in favor of more promising candidates.

It generally takes just two to three weeks for a team with extensive experience in solubility solutions using a proprietary formulation design platform to evaluate a molecule, recommend a customized formulation strategy that is likely to provide sufficient bioavailability, and prepare additional formulation scenarios as a fallback. Or, the team may quickly determine conclusively that formulation of the compound is infeasible, allowing the developer to take a “fail fast” approach and end the program without wasting significant amounts of time and money.  

Understanding the compound’s properties is critical. While all BCS Class II molecules exhibit low solubility and high permeability, the specific molecular characteristics require unique approaches. For example, some low soluble compounds will dissolve fully in gastric fluids if the dissolution rate can be increased, so micronization may lead to success in those cases. Different compounds, on the other hand, will never dissolve no matter how small the particles. One low-solubility molecule may be suited to the creation of an amorphous dispersions by spray drying, while another may not dissolve in the necessary solvents.

Once the analyses have been completed, the development team has access to a wide variety of mathematical and computer modeling tools, ranging from high level quantum mechanical modeling of compounds and excipients to molecular dynamics simulations and quantitative structure activity relationships (QSAR)-based models that are applied to the development program as appropriate. In conjunction with the experimental data, the modeling tools provide deep insight and a mechanistic understanding of the compound’s structure and behavior.

In determining what formulation has the best chance of success, the team looks beyond the immediate needs of pre-clinical and early phase trials necessary to achieve proof of concept and keeps in mind the requirements of later phase clinical trials and potential commercialization. After all, a delivery technology that produces sufficient bioavailability to get through an ascending dose study, but which cannot be scaled up might be considered a success by a solubility vendor, but would likely be considered a failure by a biopharma company looking either to take the drug to market or to sell it to a larger company for commercialization.

Global CDMOs have the ability to gather a team and leverage an extremely broad skill base and a breadth of experience that would be unusual to find in a single consultant or small vendor. And these days, more and more biopharma companies are recognizing the benefits of working with a CDMO to minimize risk and help them get their low-solubility molecule all the way to their ultimate goal.

A CDMO that provides commercial manufacturing services for hundreds of small molecule products in a wide variety of dosage forms has a solid basis to understand the nuances of formulation interactions, such as how a spray-dried intermediate may impact solid-dose manufacturing or how a lipid formulation may affect the filling process into a softgel capsule, or how a formulation with a micronized API will blend with excipients.

A global CDMO also has the expertise to consider a multitude of factors beyond bioavailability that may play a role in formulation selection and the ultimate success of the product. Only a large CDMO that has helped clients earn numerous approvals is likely to have experienced personnel in every area of drug development, approval, and life cycle management, and is able to anticipate issues that can affect pre-clinical, clinical development, scale up, regulatory submissions, and lifecycle management.

Given the availability of this type of expertise and a proven method of predicting successful solubility enhancement strategies for individual molecules, there is simply no reason to take a risky trial-and-error approach to a development program. Today, biopharma companies can opt for a simple, time-saving method of choosing the best path for a poorly soluble molecule by asking a reliable CDMO to draw them a map. And by doing so they can achieve a greater overall financial return on their investment, whatever their commercial strategy may be.

Learn From FDA & MHRA GMP Inspection Observations

A comprehensive GMP intelligence program includes monitoring of enforcement actions, including FDA form 483s, warning letters, recalls, import alerts, consent decree agreements, EU reports of GMDP noncompliance, and inspection summaries published by selected European health authorities. This article presents the most recent GMP inspection data from CDER and MHRA (Medicines and Healthcare Products Regulatory Agency).  The CDER data and the MHRA data come from GMP inspections conducted in 2016.

The CDER drug inspection observations supplement the information we published in a previous article regarding CDER drug GMP warning letters from the same time interval.  The analysis herein includes data from the FY2016 form 483 observations and compares results with those from the three previous fiscal years. Raw data comes from the FDA website, though it is presented in a different manner. For example, I have combined the frequencies of all observations that cite 21 CFR 211.192 into a single value. In the FDA data, there are multiple line items for 211.192, each with a different frequency.  For example, in the FDA listing the most frequently cited item is 211.22(d), procedures not in writing, fully followed.  When you combine the full collection of times that 211.192 and 211.42(c) are cited, however, they become tied for No. 1, with 211.22(d) becoming the third most frequent citation.  FDA uses the term “frequency” which seems to be the number of times a given citation was identified in the form 483 collection supporting these data. 

Only form 483s that were issued through the Turbo EIR (Establishment Inspection Report) system are considered in this data, which provides a distinct limitation.  No form 483s issued to API manufacturers or issued outside of the Turbo EIR system are included. This becomes important to consider with FDA’s increased focus on API manufacturers, particularly outside the U.S.   MHRA data is similar and only includes deficiencies identified at dosage form manufacturers.  Note that the MHRA data includes only the 10 most frequently cited groups, whereas the data on the FDA website includes all observations. 

Here are some highlights of the analysis:

FDA:

The number of form 483s included in this analysis remains reasonably constant over the past four fiscal years even though it does not represent all drug inspections conducted by the FDA, particularly inspection of sites that manufacture APIs.

Deficiencies in investigations remains at the top of this list over the past four years.  We as an industry cannot seem to get this quite right.
In general, the regulations cited and their relative order has remained reasonably constant over the past four fiscal years.  Even though a few items have changed place, none of the numbers are striking.

MHRA:

MHRA issued 143 “critical” deficiencies, in a total of 324 inspections of which 82 inspections (25 percent) were overseas inspections and 242 inspections (75 percent) were conducted in the U.K.
The EU GMP Guide Chapters and Annexes that were cited in critical observations include, in order of their frequency: Chapter 1, Annex 1, Chapters 5, 8, Annex 15, Chapter 3, Annex 11, Chapter 2, Chapters 4 and 6.  
The MHRA cited a total of 4,588 deficiencies in the 10 areas that received the critical observations.  Critical deficiencies constituted 3 percent of the total.
The sections that follow include more detailed discussion of the observations.

FDA Form 483 Inspection Observations

The following data is based on inspections generated using the FDA Turbo-EIR system.  The number of form 483s remained quite similar over the four years in question, with FY2014 having the fewest.  Form 483s issued to API manufacturers or issued outside of the Turbo EIR system are not included.

Table 1 shows only the most frequent group of inspection observations; the tabulation on the FDA website shows all observations.  Table 1 is organized in the order of those observations with the highest to lowest frequency for 2016.  In several instances, though, the order of the observations did change in FY2016 from previous years; these are highlighted in gray. 

Table 1: Inspection Observations Issued Through Turbo-EIR System per Fiscal Year. (These are shown in the order of highest to lowest for FY2016.)


Figure 1 below shows the data from Table 1 graphed over four fiscal years, 2013–2016.  While there is some variation from year to year, the frequency with which specific regulations are identified remains generally constant.  Figure 2 shows additional detail of several of the areas where the frequency of the observation did show some variation between FY2015 and FY2016.


Figure 1: Frequency of observations


Figure 2: Selected observation frequency

In conclusion, there is little change in the overall frequency of inspection observations, as characterized by the regulation cited, between FY2013 and 2016. This may have been different if all inspected sites, including API sites, had been included in the metrics. The three most frequent observations in FY2016 cite 211.192 (investigations), 211.42(c) (design of facilities to prevent cross contamination), and 211.160(b) (scientifically sound specifications). While 211.192 was in first place for all four fiscal years, in 2016 it tied with 211.42(c), Requirement for adequate facilities to prevent contamination or mix-ups, moved up from third place, even though the actual number of those observations decreased from 2015. Citations against 211.160(b) Development of scientifically sound specifications went from second place to fourth place. Observations citing 211.113(b) Validation of aseptic processes including sterilization dropped from fifth place to sixth place in 2016, and the actual number decreased significantly, to FY2013 levels. Finally, observations identifying 211.25(a) Staff shall have training, education and experience to perform their jobs dropped from eighth place to 10th place in 2016. 

MHRA Inspection Deficiencies

I won’t reproduce the graphics from the MHRA slide deck, but I do recommend reading those because they contain a wealth of information at a granular level. The MHRA conducted a total of 324 inspections in 2016; 242 inspections were conducted in the U.K. and 82 inspections were conducted overseas. The MHRA inspections identified 143 total “critical” deficiencies in 2016, a dramatic increase from 2015 when 51 were identified. We cannot compare this with the U.S. FDA inspection observations because the FDA does not classify the criticality of observations. In the future, perhaps health authorities will adopt a common classification category for inspection observations.

MHRA identified critical deficiencies in only five areas in 2015, and increased this to 10 areas in 2016. Several categories saw significant increases, for example:

  • Sterility Assurance had no critical observations in 2015 and 34 in 2016
  • Personnel had no critical observations in 2015 and eight in 2016
  • Premises and Equipment had no critical observation in 2015 and nine in 2016
  • Computerized Systems had one critical observation in 2015 and nine in 2016.

Table 2 identifies the areas with critical deficiencies identified in 2016. The groups included seven Chapters and three Annexes. Figure 3 clearly shows that approximately two-thirds of the deficiencies are included within three groups: Quality Systems, Sterility Assurance, and Production.

Table 2: Chapters and Annexes Associated with MHRA Critical GMP Inspection Deficiencies in 2016



                          Figure 3: Distribution of these critical MHRA deficiencies


Conclusions:

It is difficult to directly compare areas identified by the MHRA with those identified by the FDA, as the FDA does not categorize the criticality of inspection observations as do the MHRA and other health authorities. We can, however, say that with FDA observations addressing “investigations” at the top of the list, “quality unit responsibilities” third on the list, and “staff training” at No. 10, quality systems is a high priority for the FDA. Similarly, Quality Systems is the area with the most critical deficiencies identified by the MHRA in 2016. Validation of aseptic processing (21 CFR 211.113(b)) was sixth on the FDA list but was second on the list for MHRA.

Computer system requirements are identified in Annex 11, Computerized Systems. Data integrity and data governance deficiencies are identified by MHRA by citing either Chapter 4 or Annex 11, both of which were associated with critical deficiencies in 2016. Similar FDA regulations are found in 21 CFR 11, Electronic Records; Electronic Signatures, and it is rarely, if ever, identified in either form 483s or warning letters. The FDA frequently associates these types of inspection observations with predicate rules including 21 CFR 211.68(b) and 21 CFR 194.


MHRA has always had a reputation as one of the most rigorous health authority inspectorates. It seemed to have upped its game in 2016, as demonstrated by an increase in the number of critical deficiencies, along with an increase in the total number of deficiencies identified for essentially the same number of inspections.

Both agencies will likely continue to focus on sterility assurance, investigations, quality systems, and data integrity/governance in 2017. It would be interesting to see if the number and types of observations identified during API inspections were similar for the two health authorities. And finally, the Mutual Recognition Agreement (MRA) between FDA and the European Medicines Agency will likely not impact the number of inspections for 2017, though it may be possible to see that happen in 2018. Time will tell how this impacts the number and locations of both EMA and FDA inspections.


References:



Barbara Unger