2.68 Aseptic Operations D Pollard, Merck Research Laboratories, Rahway, NJ, USA © 2011 Elsevier B.V. All rights reserved. 2.68.1 2.68.2 2.68.2.1 2.68.2.2 2.68.3 2.68.3.1 2.68.3.2 2.68.3.3 2.68.3.4 2.68.3.5 2.68.3.6 2.68.4 2.68.4.1 2.68.4.2 2.68.4.3 2.68.4.4 2.68.4.5 2.68.5 2.68.5.1 2.68.5.2 2.68.5.3 2.68.6 2.68.6.1 2.68.6.2 2.68.6.3 2.68.6.4 2.68.7 References Introduction Design and Procedural Approaches to Minimizing Contamination Room Classification Considerations Utility Design Considerations Fermentation/Cell Culture Considerations Equipment Design Use of Disposables Equipment Setup Considerations for Raw Materials and Media Preparation Seed Expansion Considerations Vessel-to-Vessel Transfers Considerations for Purification and Formulation/Fill Liquid Filtration Considerations for Applying Disposable Technology to Purification Formulation and Fill Considerations Maintenance Considerations Training of Personnel Validation and Verification Sterilization Considerations Sanitization Considerations Sanitary Cleaning Issues Sterility Analysis and Culture Purity Procedures Used for Process Sampling Procedures for Testing of Contaminants Criteria for Contaminated Samples Strategy for Contamination Investigation Summary Glossary contamination Accumulation of undesired biological particles from either a microbial, a viral, or a fungal source, which reach levels above the acceptance criteria set for the specific process step. sanitization A set of conditions typically chemical, such as disinfectant or caustic washes, for the reduction of bioburden within equipment or upon surfaces within the processing facility http://en.wikipedia.org/wiki/File: Disinfection.jpg. sterility analysis The application of test methods to quantify the level of contamination within a process. This 933 936 936 936 936 936 937 938 938 938 939 939 939 939 940 940 940 940 940 941 941 942 942 943 943 944 944 944 can be from online process attributes or direct measurement from sampling. validation Actions or activities that establish by objective evidence that a process consistently produces a result or product meeting predetermined requirements. viral clearance A measure of the capacity of a purification process to remove virus particles from the final drug bulk product. Typically, this includes multiple steps for multiple log reductions such as pH inactivation hold step and through specific filtration steps. 2.68.1 Introduction The aseptic operations during any bioprocess are vital for controlling the desired level of microbial load (bioburden) of a given biological product. Manufacturers follow the standards and procedures set by the regulatory agencies [1, 2] to ensure product quality. This minimizes the risk to patients of exposure to unacceptable levels of contamination such as failure to maintain product sterility. Practical and experience-based practices, not captured in regulatory documents, have been defined by organizations [3–5] and harmonization conferences [6, 7]. These regulatory compliant procedures are based upon rational, evidence-based science, and engineering with incorporation of risk assessment analysis. 933 934 Other Considerations The manufacturing of sterile products is acknowledged to be the most difficult of all pharmaceutical production activities [3]. For bioprocess production, such as aqueous protein or monoclonal antibody solutions (Figure 1), some form of aseptic operations usually encompass every production step from fermentation, purification, formulation, and fill. It is a regulatory requirement to assure that culture purity (single organism) is maintained from the master cell bank and throughout the upstream step (Figure 1). During purification, maintaining the low bioburden specifications of the drug substance is completed using a combination of chemical sanitization of equipment and filtration of all buffers, including those used for formulation. After formulation the final drug product is filter sterilized and aseptically filled into the final container (vial, syringe, or IV bag) and, in some cases, lyophilized. Managing contamination to minimal levels covers a wide range of activities, including facility design, equipment setup, process operations, process validation, process monitoring, and personnel training (Figure 2). Despite all these efforts including attention to detail for process operations, contaminations will always occur as no microbial fermentation or cell culture facility is contam­ ination free. So procedures need to be in place for vigorous investigations of contamination to build experience and a knowledge database. Literature contamination rates of 5–30% have been described for microbial fermentation with a contamination prob­ ability of 1 out of 100 acceptable [8]. Examples of rates <1% are considered commendable and indicated as good performance [8–10], while rates of 2% have been recorded for animal cell culture [8, 11, 12]. Facility improvements have shown to lower contamination rates. For example, the contamination rate for monosodium glutamate production was reduced from 4.5% to 0.6% by a combination of sparger air system upgrades, installation of a laminar flow hood in the inoculum room, and repair of holes in the heat exchangers of the continuous sterilization system [11]. For the microbial fermentation or cell culture process, microbial contamination is the most common cause of process failure over mechanical, electrical, or instrumentation problems that occur [10]. Microbial contamination can impact the process by changing the chemical conditions such as the conversion of nutrients to unwanted impurities, changing the pH, and triggering the formation of enzymes leading to product degradation [8]. Historically, there are only a few examples of facilities that will continue to process contaminated batches beyond fermentation. These were usually for small-molecule natural products/anti-infective products where the subsequent chemical steps achieved sufficient purity and removed contaminants. This is certainly not an option for the injectable products for biologics and vaccines. Upon contamination detection, the entire B D Raw materials D Nutrient addition D Media prep S S D Buffer D S D D S 0.22-µm filtration Microfiltration Seed train Disc stack centrifuge B Ultrafiltration D B D B D Production fermentor (1000–5000 l) D D Buffer Buffer D Buffer D Buffer Sterile fill for drug product S Sterile fill (vials, syringe, IV bags)l B D D Filtration (0.22 µm) Intermediate product reservoir (sterile disposable bag or stainless steel tank) D Polishing 2 chrom B Polishing 1 chrom Capture chrom Ultrafiltration Drug substance Bioreduction Final formulation Filtration 0.22 µm D Disposable option is available S Sterility testing B Bioburden testing B D Product intermediate filtration B D B D Figure 1 Bioprocess map for production of a therapeutic protein or monoclonal antibody using microbial fermentation using a Pichia expression host. Stages for sterility and bioburden testing are indicated. Steps that have disposable options are indicated. Aseptic Operations Raw materials Facility and Equipment design Process design and operational procedures 935 Process monitoring Preventative maintenance Minimizing contamination Sterility sampling and testing (low contamination rate and bioburden control) Personnel training Contamination investigation Regulatory framework (agency interaction) Microbiological process controls and validation Knowledge database Figure 2 Integrated approach for control of contamination in bioprocessing. batch is discarded, equipment shutdown, and a failure investigation initiated. This causes substantial losses of time, materials, and revenue, with disruption to the facility schedule. Contaminants vary by product type but the most frequent microbial contaminants are from two forms: (1) fast-growing sporeforming Gram-positive bacteria such as Bacillus subtilis, associated with incomplete sterilization such as from large-medium particles or residual dried batch in vessel crevices [10, 13]; and (2) Gram-negative rods, which are indicative of cooling water leak [14], water in the inlet air, or incomplete filter sterilization. Gram-positive bacteria often enter from non-sterile air [8], owing to improper air filter installation, sterilization, or integrity [15]. Multiple contaminants are usually indicative of general sterilization failure [16]. Mycoplasma is an important contamination to monitor for cell culture processes [17–19]. Mycoplasmas lack a cell wall, have filterability at 0.22 μm, and are easily killed at 60 °C. Cell culture media components are heat-labile sensitive, so sterilization by filtration is the only option. Mycoplasma infections can overwhelm production cell cultures achieving high densities (106–107 colony-forming unit (CFU) ml–1) but visually no turbidity is observed [19]. Twenty species of mycoplasmas are known to cause cell culture issues and five have shown to give >95% of contaminations (Mycoplasma arginini, M. fermentas, M. hyorhinis, M. orale, and Acholeplasma laidlawii) [19]. Contamination sources are commonly from human operators or from the cell lines. Viral contamina­ tion, via endogenous viruses or adventitious viral agents, is an important concern for cell culture [17, 20]. Cell lines contain retrovirus-coding sequences in their genomes and therefore inherently express retrovirus particles during production. Adventitious viral agents may be introduced through the use of cell lines derived from infected animals or virus-contaminated reagents or serum components. Safety assurance is accomplished by the combination of raw material control/testing, master cell and working bank testing, in-process control testing, and virus clearance studies. Typical viral testing includes a panel of viruses, ranging in size from 17 to 400 nm, such as bovine viruses (viral diarrhea virus, adenovirus, polyoma virus), reovirus, cache virus, and murine minute virus. Reported viral infections of recombinant CHO cell lines include murine minute virus, a parvovirus [21], and epizootic hemorrhagic disease virus [22]. The risk of contamination has to be evaluated for each particular bioprocess. Subtle changes in operating conditions between processes can have a large impact on the susceptibility to contamination. Certain factors have been identified that lower the risk of contamination such as pH range (<5 and >8), low initial bioburden of the media before sterilization, high osmotic pressure, high or low carbon concentrations, switching from complex media with insoluble solids content to soluble defined media [16, 23], or applying temperatures above 60 °C. Minimizing the risk of microbial contamination is a combination of prevention activities and contamination monitoring (Figure 2). Contamination risks occur in aging facilities that are susceptible to mechanical failures, and also new facilities that have operational unknowns [16]. A balance needs to be addressed between increasing time for preventative maintenance (PM) to reduce failures versus fast turnaround times to maximize the productivity of the facility [11]. Process design, testing, and training are all important. Protocols must be in place for each new bioprocess to minimize and investigate microbial contaminations [10, 16]. This is influenced by the nature of the fermentation/contaminant, equipment design, process operation procedures, and the microbiological process controls implemented (Figure 2). 936 Other Considerations 2.68.2 Design and Procedural Approaches to Minimizing Contamination 2.68.2.1 Room Classification Considerations For biologics and vaccine production, the use of validated classified areas with room engineering controls is a regulatory require­ ment to minimize the potential for contamination of the process. Details of classified area design have been previously described [3, 13, 24]. The flow of airborne particles is controlled by the facility design using high containment areas with HEPA. Prefiltered air with climate control (18–20 °C, <50% humidity), personnel gowning areas, and equipment air locks. The HEPA filters have a retention of >99.97% of particles larger than 0.3 µm [3]. Floors are typically monolithic with integral drains to prevent standing water, while the ceilings are impervious, windows are flush mounted, and all surfaces can withstand chemical sanitization. A grade C or class 100 000 room is controlled environment for processing with unidirectional air and people flow segregation with airflow locks. A grade B or class 10 000 area has segregation and airlocks used to control flow with inlet only and outlet flows for personnel [3]. Typically, this uses 100–600 air changes per hour airflow with strict standards of free from microbial contamination and particle specifications such as <3500 particles at greater than 0.5 µm m–3 [3]. For sterile operations such as making of master cell banks or formulation and filling of final product typically requires a grade A, class 100 environment. Environmental monitoring plans are used to quantify the room classifications, which will be discussed in the monitoring and sampling sections. Access to utilities for repair or maintenance must also be considered. For example, separating the utility header in a separate service corridor adjacent to the classified area, process area allows greater flexibility for ease of repair and maintenance. 2.68.2.2 Utility Design Considerations The design and quality of the utilities that are in the product (steam and sparger air) and non-product contact (instrument air and chilled water) can affect contamination. For example, it is recommended for fermentor sparger air to locate the air compressor intakes at elevation above and upwind from fermentor exhausts and cooling tower mist [11, 16]. There is estimated to be a 1-log reduction in live organism concentration with every 3 m of increased elevation [11]. As air flows through the compressor, it is heated to >100 °C and can be cooled directly downstream of the compressor outlet. It is recommended to maintain the air at elevated temperature for as long as possible by relocating the coolers between the retention chamber and the drier [16]. This utilizes the heat of compression to sterilize the air. Retention chambers (typically 5400 l for pilot plant facility) have been designed to include exterior insulation and interior baffles to extend the travel path of hot air in plug flow. This minimizes the inlet/exit temperature drop to ≤2 °C [16]. In a facility at 11 000 l min–1 and 2.8 kg cm−2, the compressor discharge and retention chamber temperatures typically are around 93 °C with residence times in the retention chamber of 20 s (>16 000 l min–1) to 1 min at 5000 l min–1 [16]. Condensate in the sparger air is removed using heat exchangers with driers and then filtered to remove desiccant before reaching fermentors. Moisture in fermentor inlet filters can be caused by poor system design or maintenance, or excessive pressure/ temperature drop in the air supply header. Typically, the dew point is monitored when sparger air leaves the utility building and as it enters the process building [16]. Target dew points after the dryer are below –20 °C and can change up to ±15 °C after flowing 150 ft depending on ambient temperature and demand [16]. High-quality dried and filtered instrument air ensures automatic valve reliability. Replacing plastic instrument airlines with copper minimizes leaks (particularly at fittings) and enables reliable instru­ ment air pressure to control valves. Installing backup compressors and receiver tanks can prevent fermentor backpressure loss caused by automatic valves reverting to their failure states during instrument air supply interruption. Installation of filter housings with lines that slope away from the filter enables moisture drainage, preventing condensate collection and blinding of the filters. The appropriate design and operation of the water systems that contact the process are critical to minimizing contamination. For biologics and vaccines manufacture, water for injection (WFI) made from thermal distillation is commonly used for buffers and final product formulation. It is common for supply loops to recirculate water at 70 °C to prevent microbial growth [11]. Then cold water is established at point of use drops using an individual heat exchanger. Proper utility system operation requires monitoring using a combination of automated data acquisition system and manual review via utilities checklists. Periodic reviews are necessary to confirm consistent execution of the operation. Measures undertaken due to utility failures should be documented to build a knowledge database. Experience of these procedures has shown to reduce the impact of product and non-product contact utility outages and promote uniformity of response [16]. After a utility failure, affected fermentors should be evaluated to determine whether to abort the batches. Evaluations are conducted using online trends and visual monitoring of field gauges during utility loss and subsequent restoration. 2.68.3 Fermentation/Cell Culture Considerations 2.68.3.1 Equipment Design To achieve desired sterility and culture purity goals, bioreactors are to be of high-quality design and well maintained [10, 16]. Many design features that are typically incorporated for sterility are also useful for containment (i.e., minimization of microorganism release) [25]. The steam seal heat barrier plays an important part against the entry of contaminants. Diaphragm valves are commonly used for sterility although constant steam service has shown to deform Teflon-backed ethylene polypropylene diene monomer (EPDM) diaphragms [10]. Alternatives include the use of three-piece ball valves that allow for easy ball replacement (75% less time) without the cutting and welding necessary for existing two-piece valves [16]. The most efficient facility allows for Aseptic Operations 937 piping arrangements that allow localized shutdown of equipment without impacting the steam seals of adjacent equipment. Establishment of piping installation preferences contributes to maintaining sterility control such as the minimization of dead legs, installation of steam entries on the top of piping, installation of condensate removal legs on the bottom of piping, smooth aligned manual welds, line sloping for drainage, and use of 316-l material of construction [16]. The agitator seal on a fermentor is a main area of contamination concern. For steam-lubricated, top-driven seals, steam should remain applied to the seal, even when not in use. For condensate-lubricated, bottom-driven seals, residual condensate should remain between batches. After long periods (2–3 months) of inactivity, fermentors with bottom seals should undergo sterility testing [16]. The use of high-temperature fluorinated elastomers (Kalrez, DuPont, Wilmington, DE, USA) on both double mechanical seal faces is recommended. Seal failure, due to external or internal leakage, can be determined visually and by measuring air pressure decay rates. A 2- to 3-year typical seal service life has been established based on tracking seal failures [16]. Ideally, stainless steel grade of 316 l is recommended for bioprocessing along with smooth welds and joints to give high cleanability. Often facilities use electropolished systems with an ultrasmooth finish, such as a 0.4 ra where no point is higher than 0.4 μm above the surface of the metal [26]. 2.68.3.2 Use of Disposables For cell culture, the industry is moving away from stainless steel bioreactors for inoculum expansion and production to single-use technologies such as Wave rocking bioreactor (500 l) or disposable stirred tank reactors (up to 2000 l) [20, 27]. This has mostly been driven by the industry’s desire to speed up timelines and reduce costs by the removal of clean-in-place (CIP) and steam-in-place (SIP) infrastructure and the associated validation/monitoring of such systems. The disposable reactors are based upon gas impermeable multilayered bags of polyethylene, polystyrene, or polypropylene (polyethersulfone, polyvinylidene fluoride) of 300 µm thickness. Sterilization is by steam or gamma irradiation using cobalt 60 isotope (25–40 kGy) to provide the electro­ magnetic energy that penetrates materials of construction, destroying any microorganisms and viruses. Reactor bags are held in support vessels that enable temperature control and process monitoring such as pH or dissolved oxygen (DO) through fluorescence patch technology [27]. The disposable bag technology is also used for preparation and storage of cell culture media, purification buffers, and product intermediates at 200-, 500-, and 1000-l scale [20, 28]. This removes the need for additional stainless steel vessels and the associated CIP infrastructure and validation. Analysis shows that total production costs of a 2000-l process are 30% cheaper using disposables rather than using stainless steel vessels [20]. However, at 8000-l scale bioreactor, there is little difference due to lower facility dependence and high material costs from the larger number of disposable bags required [20]. Issues still remain to be resolved such as the control of leachables, limitations of scalability, and process monitoring with the lack of standardization for materials of construction [27]. Despite these issues, an often overlooked advantage of single-use technology is the reduction in cross contam­ ination and improvement in sterility assurance. Single integrated disposable systems can now be purchased that combine the disposable bag and capsule filters using the highest purity silicone tubing (platinum cured) [29] (Figure 3). The single system includes the necessary clamps, adaptors, and connection devices into one single system as specified by the customer. This has greatly reduced the number of sterile manipulations that would have been required for sterile connectivity of each piece and to the process. The need for open connections has been eliminated by the ability to connect tubing-by-tubing weld connections. Tubing welder systems (Terumo, Terumo Medical Corp., MD, USA) provide sterile welds to dry or liquid-containing tubing but can be limited to the size of the tubing. Another approach is the use of disposable aseptic connectors (such as ReadyMate™, GE Healthcare) that provide high flexibility by the genderless design (Figure 3). This allows cross-size connectivity between a different size tubing and a tamper proof connection that can withstand process pressures up to 15 psi. Figure 3 Integrated design of sterile bag integrated with filter assembly and genderless connector. 938 Other Considerations 2.68.3.3 Equipment Setup The accurate setup of the equipment will assist in the minimization of contamination (Table 1). For contained systems, pre-batch integrity testing of hydrostatic and air pressurization hold testing is a routine operation for fermentors. A pressurization hold test of <0.07 kg cm−2 loss over 12 h is routinely deemed acceptable. This allows leaks to be identified and corrected that otherwise may cause grow back, especially if a channel (fluid layer) is involved [16]. This is usually followed by a 7–10-day sterility test. Before the production batch, it is usual to perform routine gasket and O-ring replacements, such as for probes and blind plugs, and clean up of all ports and threads. The next step is steam sterilization of the empty vessel at 1.3 kg cm−2 and 124.5 ± 1.0 °C for 1 h without installation of the sparger air filter or probes and is usually completed within 48 h before media batching [16]. The fermentor then remains under air pressure until ready for media batching. Personnel visually examine inside the vessel during setup and before batching. Often an agitator seal or head plate O-ring leak can be detectable simply by checking for unusual amounts of accumulated condensate. During and after sterilization, the temperature and pressure profiles should be evaluated to identify any unusual occurrences that should be documented to aid contamination investigation. Examples include difficulty in obtaining airflow through the sparger filter after sterilization, foaming during heat up and/or sterilization hold periods, and Templstiking failures [16]. Steam traps should be regularly checked, at least daily, during the fermentor operation as well as directly before sterilization. Inexpensive bleed valves can be installed for easy draining/clearing of plugged traps [16]. 2.68.3.4 Considerations for Raw Materials and Media Preparation Understanding the bioburden of raw materials and how it changes during raw material preparation can be important for minimizing contamination. For example, the media components for fermentation use complex components such as yeast extract and soy peptone [30] that supply an initial bioburden to the media, which can increase during media batching preparation. Minimizing the time between media batching and sterilization to <6 h is routinely employed. Other possible methods include lowering the media to a low pH (<4.0) and cooling the media (6–8 °C) before sterilization. For media with insoluble components or clumps, an in-line mixer (homogenizer) can be employed to ensure that solids are sufficiently wetted. Media are recirculated through the mixer for up to 1 h (3–10 turnovers), then passed once through the mixer before transfer to fermentors. For vessel steam sterilization, it is important to maintain a positive pressure thoughout the sterilization and cool down cycle. After sterilization, vessel pressurization with sterile sparger air before cooling is recommended to avoid vacuum upon cooling. Raising the backpressure to 1.5 kg cm−2 before introducing sparger air and start of cooling avoids foam, and large pressure drops [16]. Other facilities shut off steam and start cooling while permitting backpressure to decrease as low as 0.2 kg cm−2 before applying sterile air. This approach potentially increases the risk of pulling in nonsterile air [16]. 2.68.3.5 Seed Expansion Considerations Inoculum expansion using disposable shake flasks with vented caps is used routinely and replaces the use of glass Erlenmeyer flasks with cotton bungs that are prone to wetting and provide a potential contamination risk. Similarly, the use of tubing welders and sterile disposable connectors allows closed connections of inoculum to the process vessel. Presterilized, single-system disposable Table 1 Fermentor pre-batch setup procedure for lab, pilot, or manufacturing scale facility using steam in place for sterilization Procedure steps Operating conditions Acceptance criteria 1 Pre-batch integrity testing <0.7 kg cm–2 loss over 12 h 2 3 Sterility test Gasket and O-ring replacement 4 Empty vessel steam sterilization 5 Visual fermentor inspection prior to media batching Media batching: minimize hold time before sterilization Fermentor + media sterilization Reactor leak testing by hydrostatic and air pressurization hold testing Sterilization of media for 7–10 days Gaskets/O-rings change out for fermentor, probes, blind plugs steam sterilization at 1.3 kg cm–2, 124.5 °C for 1 h without sparger air filter or DO, pH probes. Reactor remains under pressure until media batching. Agitator seal or head plate O-ring leak can be detectable by condensate accumulation Media makeup and transfer to fermentor 6 7 Maintain temperature and pressure conditions during entire sterilization. Frequently templstick key areas – filter housing, harvest point, side ports, steam traps checked Free from microbial contamination All new gaskets/O-rings installed Acceptable sterilization maintaining desired temperature and pressure for duration No visible condensate accumulation in the fermentor Sterilize media < 6 h after media make up Sterilization maintained at temperature and pressure with no templstik failures and no steam trap failures. Adapted from Junker B (2006) Sustainable reduction of bioreactor contamination in an industrial fermentation pilot plant. Journal of Bioscience and Bioengineering 102(4): 251–268. Aseptic Operations 939 bags with capsule filters are used as nutrient feeding containers to avoid repeatedly assembling and autoclaving plastic containers with numerous fittings. 2.68.3.6 Vessel-to-Vessel Transfers Transfers between reactors such as inocula or media can be made by hard pipe or presterilized tubing connections using pressurized sterile air via dip tubes [16]. Transfers between harvest valve of the seed vessel side port of the receiving vessel maybe preferred as dip tubes can be difficult to clean additional validation difficulty. Hard transfer piping can be steamed constantly or steamed for 30 min at 130 °C (in excess of the required Fo). During tank-to-tank pressure transfers, backflow of broth is prevented by using a higher pressure in the source tank. Pressurized transfers are conducted with appropriate transfer line cooling such as emptying the initial hot broth to waste or using sterile airflow to vent. 2.68.4 Considerations for Purification and Formulation/Fill For purification, the first step is typically to separate out the cells via disc stack centrifugation (Figure 1) or high-pressure homogenization for cell breakage if the product is intracellular. Adding sterilization capability to these intricate machines adds significant complexity and cost. An additional 20–30% is required for a manufacturing capacity centrifuge, which is already a high capital expense item [31]. So the majority of the industry uses CIP and sanitization procedures to minimize bioburden for most purification unit operations. This includes chromatography and especially filtration (ultrafiltration (UF) and microfilteration (MF)) where the membranes are not capable of withstanding SIP [28, 31]. Chemical sanitization is usually completed with 0.5–1 N NaOH for a hold time of >1 h that provides adequate bioburden control. Regulatory authorities will expect maximum hold times to be used and justification for specifications needs to be defined. Water rinses are needed to remove the chemical sanitizer residues. Sufficient rinses to return the conductivity to <5 mS cm–1 can be used or specific unit operation criteria such as 150-l rinse of WFI m−2 membrane surface area for membrane filtration. 2.68.4.1 Liquid Filtration All buffers used for purification are filtered using presterilized 0.22-μm filter capsules and then typically stored in disposable bags. The disposable filter capsules are extensively used throughout bioprocessing for sterile filtration of gases (sparger air) or liquids such as nutrient feeding, bioburden reduction of purification buffers, product intermediates, and final product sterile filtration [31–33]. A sterilizing grade filter rated as 0.22 μm has been validated for bacterial retention to defined specifications. Bacterial retention occurs by the sieving action on the membrane surface but also by size exclusion entrapment in the membrane structure. For example, 10 layers of 0.8-µm cellulose is required for a reduction of 2.4 × 108 of a challenge organism such as Brevundimonas diminuta [31]. Membrane filters are porous materials so retain a certain amount of open area. This allows for fluid flow through the membrane as defined by the porosity of the membrane. The membranes are usually pleated and formed into a cylinder, then cast into a capsule of polypropylene. Layers of membrane are corrugated along with upstream and downstream layers of coarser material, allowing support and drainage for the membrane. Hydrophobic membrane filters (materials such as polytetrafluoroethy­ lene (PTFE) can be in situ steam sterilized such as 30 min exposure at 125 °C for repetitive cycles and are often used for gases, such as sparger air, as the hydrophobic nature allows them to dry quickly. Sterile liquids are often filtered using polymeric microporous hydrophilic materials such as polyvinylidene fluoride [31]. 2.68.4.2 Considerations for Applying Disposable Technology to Purification Presterilized disposable technology has also been applied to the filtration unit operations of depth filtration, UF, and MF (Figure 1) [20, 31, 34]. Depth filters are primarily cellulose fiber based combined with filters aids such as activated carbon or diatomaceous earth to give two modes of clarification: size exclusion of large particles entrapped on the surface and retained within the interior walls of the membrane, while amine chemistry enables particle adsorption [32]. Depth filters are used to remove cellular debris after centrifugation, but the charge surfaces also reduce contaminants such as residual DNA and host cell proteins. Modular flat sheet disposable stacks are available presterilized by gamma irradiation. A system with 1 m2 capacity enables the processing of between 250 and 500 l and disposable systems are available up to 12 000-l scale (43 m2). UF is used for product concentration and buffer exchanges that occur at multiple times during bioprocessing (Figure 1). Single-use tangential flow filtration (TFF) cartridge systems are available for UF. For example, single-use and single pass flat sheet membrane systems can be manifolded together to create a serpentine flow path of progressively smaller membrane areas. This configuration allows the cross flow across the membranes to be maintained, while the feed stream volume and volumetric flow rate is reduced [31]. For a cell culture process, the demonstration of viral clearance is required. The reduction of viral load (4–6 log) is typically achieved from a combination of a chromatography step, pH inactivation, and addition of a viral filtration step such as single-use nanofilter system, run in tangential flow UF mode [28]. Prebatch setup tests for purification equipment can include pressurization tests for centrifugation and filtration skids. The proper installation of filters assemblies can be confirmed from pressurization of the wetted and rinsed filters and the generation of stable volumetric flow rates. 940 2.68.4.3 Other Considerations Formulation and Fill Considerations For final drug formulation, UF (TFF) is used for concentration and buffer exchange into the final formulation. The presterilized UF disposable capsules can be used such as the Pellicon filters (Millipore) with disposable capsule availability of up to 2.5 m2 per capsule. Final drug product sterilization after formulation is completed by filter sterilization (0.22 μm) typically into a disposable presterilized bottle. This improves sterility assurance by removing the need to CIP or SIP of a suitable container and also reduces the validation issues. The sterile material is then filled into the desired container often using in-line sterilization. The vials, prefilled syringes or IV bags, and associated parts, such as vial caps, vial crimps, and syringe barrels, are washed before depyrogenation and fill. The depyrogenation of glass surfaces is typically at 250–260 °C. Any sterile gas requirement (air or nitrogen) such as for the automated filling stations or for lyophilization for freeze drying applications is filtered using disposable capsule filtration. 2.68.4.4 Maintenance Considerations Implementing a PM program is vital to maintaining control of contamination and minimizing downtime [16]. Examples based upon facility-specific experience include (1) quarterly infrared steam condensate trap surveys to identify traps that were plugged, blowing through, or leaking; (2) annual testing of transfer line valves for internal and external leaks; (3) annual replacement of diaphragms, especially those exposed to constant steam service; (4) annual testing of diaphragm valves, both before and after repair, using pressurized air to detect leaks from valves submerged in water; (5) annual internal vessel inspections to ensure bolts are present and tightened; and (6) annual inspection of vessel ports to remove burrs to prevent sticking [16]. Routine maintenance of key instrumentation is important such as backpressure and temperature control for fermentation sterilization reliability. Pressure gauge accuracy should be verified against pressure transmitters at 1.1–1.3 kg cm−2 and resistance temperature detectors (RTDs) characterized at three temperatures (0, 65, and 130 °C) using an oil bath. For fermentation the most common reasons for probe failures are compromised DO membranes and broken pH glass sensors [16]. These failures can be significant sterility risks owing to potential release of non-sterile electrolyte. DO probe failures are about threefold higher than pH probe failures. A pre-batch pH/DO probe response checkout can be conducted to ensure reliability. pH probe sensors should be replaced every six batches while DO probe membranes replaced every other batch, and DO probe sensors replaced annually. Both pH and DO can be tested using offsets between (1) pre- and poststerilization probe readings and (2) poststerilization probe readings and laboratory-analyzed grab samples. When a poststerilization failure of a DO or pH probe is detected, the vessel is usually discarded. The replacement of gaskets and external O-rings after each batch, such as on probes or vessel plugs, is common practice. This avoids the inconsistencies associated with visual inspection and reuse. It can be made cost effective by finding a suitable disposable, single-use gasket that can withstand the process conditions such as contact with steam. Examples include high-temperature peroxide-cured EPDM that can withstand >275 °F for >400 h and up to 600–800 h without splitting or substantially sticking to the vessel manway [16]. Examples of detailed maintenance to process filters include the uniform torquing of larger scale sparger filter housings to assure a uniform seal, implementing a air pre-filter and steam filter before the main air sparger filter [8] and testing the steam supply valve to the sparger air filter for leaks. This maintenance work must be documented using a suitable database that allows the tracking of work orders for initiation, completion, and repair testing. The confirmation of repair testing ensures that the repair meet expectations before returning equipment to service [16]. Regularly weekly review of the database identifies if additional investigation or more extensive repair might be required for repeat issues. Change control procedures for equipment and computer systems ensure that the changes are documented, communicated, and appropriately evaluated for potential effects on contamination as well as validation [16]. 2.68.4.5 Training of Personnel Consistent bioprocess operations are needed to minimize contamination and aseptic processing is highly dependent upon the proficiency of the personnel assigned. This proficiency must be firmly established before allowing personnel to conduct critical aseptic operations. Training and documented communication of procedures and best practices are vital to maintaining the operational effectiveness of a facility and minimize contamination [16]. This training should include both classroom and ‘hands-on’ dummy runs on the process floor to gain familiarity. Building the skill set of new staff should include monitoring their ability to execute procedures. Operational procedures should be explained with the reasons behind operational procedures to generate a logical framework to assist personnel to recall the order of execution. Benefits were acknowledged by Junker [16] from effective training of the maintenance personnel. This included training to identify problems, understanding of project status, and regular review of maintenance job prioritization for timely completion of the most critical repairs. 2.68.5 Validation and Verification 2.68.5.1 Sterilization Considerations To define the sterilization conditions for a particular piece of equipment and media, it is highly recommended to complete actual SIP studies, rather than relying on theoretical analysis of kill calculations [35]. For example, at the 120 000-l scale, a cycle of 122 °C for 12 min was reported based upon on computer kill calculations [11], whereas SIP studies with spore strips confirmed that batch Aseptic Operations 941 sterilization times for growth media were 45 min at ≥122 °C for 800–19 000 l were required [16]. Thermocouples with spore strips using Geobacillus stearothermophilus can be placed inside the equipment to be heat sterilized such as the vessel headspace. Spore solutions can be placed in the liquid-containing part of the equipment. Relative D-values of spores in the solution to be sterilized can be compared to those in water to adjust sterilization conditions (pressure and temperature for a particular piece of equipment) [16, 35]. Similarly the validation of autoclaves and load patterns can be conducted using thermocouples and spore strips/solutions. The SIP effectiveness can be confirmed by conducting three successful, successive, sterility tests, or inoculated batches. A typical sterility medium consists of 6 g l–1 yeast extract (autolyzed code 106; BioSpringer USA, Minneapolis, MN, USA), 6 g l–1 cerelose, and 1 ml l–1 polypropylene 2000 (P2000; Dow, Freeport, TX, USA), adjusted to a presterilization pH of 7.0 [16]. A low-end sterilization temperature range of 122–123 °C for 40 or 45 min depending on tank size is used, followed by a 7–10-day hold period at 35–37 °C. At least one sterility batch is conducted after each contaminated batch. 2.68.5.2 Sanitization Considerations To define the sanitization conditions for purification equipment requires experimentally confirming the hold times and chemical sanitizer concentration to give sufficient bioburden reduction to meet specifications for the particular bioprocess. The validation of filtration processes can be completed using bacterial challenge testing to demonstrate consistent removal of the standard bacteria. A sterilizing grade filter rated as 0.22 μm has been validated for bacterial retention which is defined as one capable of retaining 107 B. diminuta cells per cm2 of membrane surface under specific conditions as accepted by regulatory agencies [2]. Filter validation also includes integrity testing using a combination of bubble point, forward flow, and pressure hold tests. The bubble test point is to determine the pressure at which a continuous stream of gas bubbles (air or nitrogen) is initially seen downstream of a wetted filter under gas pressure. This can be accomplished using an automated filter integrity testing device (Integritest4, Millipore). The testing is applied to any sterilizing filter and associated vent filter of a process. The membranes are tested wet by water for hydrophilic membranes and using 60% IPA for hydrophobic filters. The demonstration of sufficient viral clearance (4–6 log reduction) can be validated by viral challenge studies of the chromato­ graphy step, pH inactivation, and viral filtration. The industry typically uses two species including enveloped and nonenveloped as a demonstration of two different viral particles with different sizes, examples of which are shown in Table 2. 2.68.5.3 Sanitary Cleaning Issues Controlling microbial contamination of bioprocesses requires effective sanitary cleaning procedures for all areas of operations. This even includes janitorial training for maintaining the sanitary conditions of the raw material warehouse and routine inspection of sanitation and pest control programs. The accumulation of solids within equipment can compromise the sterilization effectiveness, so good cleaning procedures are important to minimize contamination. Automated CIP systems with spray balls can be implemented. Alternatively, a manual approach can be used with high-pressure nozzles of high-velocity water streams to remove adhering residues located on areas such as the upper sidewalls or agitator mounting flange. This is typically followed by filling the tank with cleaning agent up to contact with the tank dome, which ensures coverage of all internal crevices. The cleaning agent such as CIP 200 or sulfamic acid (1.2 wt.%) is maintained at its optimal temperature of 80 °C (±10 °C), under agitation at 75% of maximum speed, for a 1–3-day soaking duration. Cleaning and rinse solutions should also be passed through the fermentor internals by air-pressurization or recirculation using a pump and include the flushing the vent line if a contamination occurred during the batch [16]. For fermentors with repeated contaminations, boiling of the vessel with Na2CO3 or Na3PO4 (high pH, metal-chelating agents) and a germicide can then be applied. For each new process, a cleanalibity assessment is completed using media and samples of the process to verify the cleaning process. Clean ability is assessed by a combination of methods: visual inspection for surface cleanliness, direct swab testing of surface, and direct testing of rinsate. Typically, rinsates are tested for pH, conductivity, total organic carbon (TOC), bioburden, and endotoxin, and acceptance criteria are defined by the agencies [1, 2]. Cleaning validation criteria will be process specific as cleaning residues may be removed by subsequent purification steps so the earlier cleaning processes may not require cleaning validation. TOC analysis is considered to be a good measure of overall cleanliness as it will incorporate residues from all possible sources, such as the product, cell culture or fermentation media, and buffers [4]. The removal of cleaning agents Table 2 Typical viruses used for viral clearance challenges studies for validation Virus cell Murine leukemia Human herpes simplex virus Parainfluenza virus SF4 Murine virus of mouse Reovirus type 3 Simian vacuolating virus 40 (SV40) Virus classification Envelope containing Genome Retroviridae Herpesviridae Paramyxoviridae Parvoviridae Reoviridae Polyomaviridae ✓ ✓ ✓ ✗ ✗ ✗ RNA DNA RNA DNA RNA DNA Size (nm) 80–130 150–200 150–300 18–26 60–80 45–55 Shape Indicator cells Spherical Spherical Spherical Icosahedral Icosahedral Icosahedral PG4 Vero BT cells 324 K LLC-MK cells Monkey kidney cells 942 Other Considerations such as CIP 200 is indicated by phosphorus or rinsate conductivity. Examples have been shown that rinsate analysis can be more effective as swab sampling routinely exhibits lower recovery [4]. The cleaning process should have documentation that the procedure does not allow microbial proliferation. The bioburden is monitored during cleaning validation and clean hold time studies. Bioburden acceptance criteria are based upon the equipment process step, the final rinse water quality, and the capability of the cleaning and sampling processes. 2.68.6 Sterility Analysis and Culture Purity Sampling for contamination is required from multiple steps during bioprocess manufacturing (Table 3), can prevent the use of contaminated inoculum, and assist the investigation of the source of contamination. Testing includes confirmation of culture purity throughout the fermentation or cell culture process, from working cell bank vial to production vessel as well as sterility of uninoculated media and nutrients. Additional areas include monitoring of sterile raw materials, possible product intermediate steps during purification, and the final product. A cell culture process will also need to confirm virus reduction after the first chromatography step. The excursions during fermentation online profiles such as pH or oxygen uptake rate from vent gas analysis or excessive foaming are indicators that can quickly identify to contamination issues. Occasionally, low-level contaminations cannot be detected by online parameters resorting in reliance on off-line culture purity testing. 2.68.6.1 Procedures Used for Process Sampling For each process, the inoculum seed train should be sampled from the vial, first-stage flask, and pooled inoculum. Sterility and culture purity analysis for fermentors should be conducted daily, as well as after sterilization, after any additions such as inoculum or nutrients and at the final harvest point. Fermentor samples can be taken from the vessel’s sample nipple into presterilized test tubes (open transfer) with push on caps. A constant steam bleed is used for steam- sterilization of this nipple (maintaining the surrounding area hot between samples) and is cleaned regularly. Alternatively, a contained (closed transfer) sample system can be used, as for biologics processing, with a 0.2-µm filter-vented glass sample bottle and an integrity-tested valve array to provide a steamable connection. For purification, samples for bioburden may be routinely analyzed from buffer makeup, after sanitization rinses, process intermediates, drug substance, and from the final formulation. During sterile fill samples, the routine procedure is to take samples for sterility at the start, mid, and end of the fill. For the classified grade areas, environmental monitoring sample collection is a regulatory requirement. Sampling includes surface wipe tests where swabs are moistened with water, wiped on the surface, and then placed into a tube with sterile water, sonicated, and mixed [13]. The absorbency of the swab can have significant impact on the quantification of the organisms. For example, cotton swabs can produce lower cell counts than calcium alginate swabs, as cotton contained fatty acids that may inhibit microbe growth [37]. Calcium alginate swabs were deemed superior for quantitative analysis as they can dissolve in 1% sodium hexametaphosphate [37]. An alternative to swabbing surfaces is direct contact of the test surface with an agar plate to determine the number of viable CFUs of bacteria and mold on a test surface. For this RODAC (replicate organism direct agar contact) procedure, the tryptic soy agar (TSA) surface is ‘rolled’ on the test surface such as table, wall, or personnel gown or finger tip and then incubated for set times and temperature. Other techniques for surface sampling include the vacuum suction sampler that has a filter sample wand that contacts the surface and the vacuum suction sends exposed water through a filter that is then incubated on agar plates [38]. The contamination of the air can be sampled by passive air monitoring using settling agar plates placed throughout the facility. Air filtration systems can be implemented that can collect more than 98% of particles >0.3 µm [24, 39]. The slit to agar sampler (New Brunswick Scientific or Table 3 A sterility and bioburden testing matrix for bioprocesses Microbial and cell culture processes Cell culture processes only Step Type Bacteria Mold Endotoxin Virus Mycoplasma Raw materials Master and working cell banks Growth medium (sterile) Fermentation/cell culture End of fermentation (pre-harvest) Product intermediates during purification Drug substance Drug product Bioburden Culture purity Sterility Sterility/culture purity Sterility/culture purity Bioburden Bioburden Sterility ✓ ✓ ✓ ✓ ✓ ✗ ✗ ✓ ✓ ✓ ✓ ✓ ✓ ✗ ✗ ✓ ✓ ✓ ✗ ✗ ✓ ✓ ✓ ✓ ✓ ✓ ✗ ✗ ✓ ✗ ✗ ✓ ✓ ✓ ✗ ✗ ✓ ✗ ✗ ✓ Adapted from Adamson SR (2000) Process validation and characterization: Animal cell culture process. In: Sofer G and Zabriskie D (eds.) Biopharmaceutical Process Validation, 1st edn., pp. 101–128. New York, NY: Marcel Dekker, Inc. Aseptic Operations 943 Barramundi Corp.) directs air onto an agar plate, which is subsequently incubated. Caution is required such that sufficient disinfection is completed to minimize contamination of the sample air and the generation of false positives [40]. Agars such as TSA are used for non-fastidious microbes and Sabouraud dextrose agar (SDA) for the cultivation of fungi. After incubation, the plates can be read on a colony counter for total viable cell count. An alternative system is a compact centrifugal air sampler (RCS plus, Biotest AG, Dreieich, Germany), which can allow 1 m3 of air to be sampled onto soybean casein digest medium strips that are then removed and incubated. 2.68.6.2 Procedures for Testing of Contaminants A common monitoring approach of samples is direct microscopic examination of liquid broth samples using Gram staining. The preparation of multiple slides can be aided by a commercially available automated Gram stainer (Midas III; EMD Chemicals, Gibbstown, NJ, USA/Merck KGaA, Darmstadt, Germany) [16]. A second technique is direct streak of the process sample onto agar media plates such as SDA, blood agar, or potato dextrose agar and incubated at 25–37 °C for 7–10 days. Plates are examined for unusual colony formation on an interim and final basis, respectively. A third technique is the subculturing enrichment technique that selectively enriches for contaminants. Tryptic soy broth (TSB) is commonly used as it detects most bacteria. Test procedures for fermentation broth samples usually involve inoculating 1 ml of sample into 4 ml TSB in inoculation tubes shaken at 220 rpm for 15–30 h at 34–38 °C. After shaking, tubes are used to streak onto TSA plates, incubated statically for an additional 6 days, then examined visually. Viability has been defined as the ability to form colonies, thus turbidity in a TSB tube without subsequent colony formation was not considered contamination. An alternative is the phenol red (PR) dextrose broth that detects bacteria by turning yellow with acidic contaminants as well as acidic production cultures. The bioburden analysis for raw materials, process purification, and final product sterility samples is usually tested by concen­ trating cells by membrane filtration using a 50-ml sample filtered through a membrane filtration disc (47 mm diameter), then incubating the filters on two types of agar at two different temperatures between 3 and 7 days. Typical limits of detection are down to <5 CFU per 50 ml. Alternatively, serial dilutions can be completed and spread plate technique used on agar to quantify a yield count between 30 and 300 CFU. Although these standard microbiological methods for contaminant detection are reliable, they are time consuming as the typical duration of sterility testing for injectables is 14 days. Alternate rapid methods include total and viable microbial population measured using total ATP and intracellular ATP [38]. Commercial luminescence systems are available (Celsis RapiScreen from Celsis, Milliflex from Millipore, and PallChek from Pall) for ATP assays that give similar total viable count to conventional aerobic plate counts. The ATP assay methods can be sensitive to the sample matrix particularly from cleaning solution residues, which can denature the luciferase enzyme [41]. Alternative methods such as quantitative polymerase chain reaction (Q-PCR) methods are becoming mainstream tools for rapid microbial detection. Methods have been demonstrated for the estimation of total bacterial population by determination of 16S RNA copy number using a fluorescent label probe. The process takes <1 day as an autolyzer is used to automate the nucleic acid extraction and has been shown to perform equally to manual phenol/chloroform extraction [38]. Favorable detection limits (1 CFU ml–1) of such PCR methods have shown to be demonstrated. The detection of a contaminant, based upon isolation of the contaminant cultivation on nutrient media, is followed by morphological and biochemical tests. This commonly includes transmission electron microscopy to assist virus typing. The most likely match for a genus and species can come from database profiling from fatty acid methyl ester (FAME) analysis and DNA sequencing. A number of commercial identification systems are available (Microcompass (Lonza) and MicroSeq system (AB biosystems)) which use PCR techniques by automating the sequencing of the universal 16S ribosomal RNA gene and then comparing it to known libraries to find the closest match. Q-PCR methodology has been developed for endogenous retrovirus load qualification and adventitious viral agent testing in production cell culture [42, 43]. Cell lines contain retrovirus-coding sequences in their genomes and therefore inherently express retrovirus particles during production. The viral load in the final product needs to be quantified and has traditionally used in vitro cellbased infectivity plaque assays [43], taking 28 days for cell culture passaging as opposed to 1–2 days for PCR assays. The Q-PCR methods allow virus identification by comparison of rRNA sequences to known libraries [42]. New technology such as Virochip virus detection has been demonstrated to identify new virus families via hybridization to similar sequences of established virus strains [44]. The chip system has an automated workflow of random PCR, hybridization, and fluorescent detection. A typical current chip array has >30 000 oligos from >3000 viruses [44, 45]. Virus detection by rapid sequencing from mRNA nucleic acid pools is also feasible using massively parallel sequencing that allows entire genomes (25 million bases) within 4 h then screened against the viral database. The mycoplasma testing of cell culture is usually completed using standard agar plating (7–14-day incubation) and culturebased testing by exposing Vero indicator cells (28-day incubation). Specific PCR methods have also been implemented [19, 38] which can be completed in <1 day with limits of detection to 10 CFU ml–1. The Q-PCR method amplifies a 280-bp DNA fragment of the gene encoding for the 16S rDNA and has been validated using reference strains M. orale and M. pneumoniae with sufficient detection ranges for all mycoplasma species found in cell culture. 2.68.6.3 Criteria for Contaminated Samples Early contamination detection avoids wasting resources for continued processing of contaminated batches. Wherever possible, it is routine operation to confirm seed inoculum sterility before transfer to the next growth stage. This includes microscopic evaluation 944 Other Considerations of the broth samples and enrichment culture tubes. The detection of two consecutive samples (taken >2 h apart) is usually used as a confirmation of contamination. The frequency of false positives, when a contaminated sample is followed by a sample that is contamination free, is around 0.18–0.42% per year (from 5000 samples) [16] and is more common with direct broth samples and TSB tubes than in plated agar medium. 2.68.6.4 Strategy for Contamination Investigation Thorough investigation of contaminations can significantly help to reduce the contamination rate of a process or facility. Performing the investigation by promoting information gathering in a non-judgmental manner promotes openness [10, 16] and can lead to a more useful and faster narrowing down of the key issues. It is often experienced that these investigations find a matrix of interrelated issues carrying through from different operational steps. So it is recommended to investigate a number of key points of the process rather than a narrow focus on a single issue. Visual examination of the equipment setup and operation is vital, including evaluation of alarms action items of planned and unplanned should be reviewed. Where ever possible the equipment should be disassembled and evaluated including any probes and valves assessed for leakage. Integrity testing of all associated filters is recommended. Steam traps should be checked for function, condensate traps for blockage, and internal vessel surfaces for cleanliness. An agitator seal pressure test is conducted for internal and external leaks. For a repeat contamination, particularly of Gram-negative rods, the jacket should be leak tested using 90 psig air and a vessel full of water. Common issues for fermentation contamination include raw material issues, inadequate sterilization of equipment/air/media, instrumentation errors, inadequate procedures, operator errors, insufficient training, procedures not followed, and lack of routine PM-contaminated transfers [10, 16]. These issues need to be tracked to improve awareness of key issues. For example, Junker [16] showed that the highest cause of contamination in a microbial pilot plant facility during the period 1Q1990–3Q1997 was to unknown causes (55.3%), followed by contaminated tank-to-tank transfers (25.1%). Substantial efforts were then implemented to improve thoroughness of postcontamination mechanical checkouts (to reduce unknowns) and pretransfer clearing of seed tanks (to improve detection methods). These actions not only reduced the overall number of contaminated batches during the following period of 4Q1997–3Q2004, but shifted the highest percentage to equipment (54.6%) followed by unknown causes (25.5%). Further reduction in the percentage of equipment-classified contaminations might be realized by enhanced setup or PM procedures. Tracking the timing of the contamination detection and the cause of the failure is also worthwhile. For example, Junker [16] showed that 65–70% of fermentation contaminations arose <48 h into the batch, and 55% were equipment related. This suggests that focusing on equipment problems resulting in early contaminant introduction might have the greatest impact on further contam­ ination rate reduction. These facility metrics should be highlighted to all, especially operators and mechanics, on a regular basis. 2.68.7 Summary Schedule delay and loss of production capacity from contamination have been observed across the fermentation/cell culture facilities of the industry. Literature examples of unresolved contaminations include facilities switching to different products and specific vessels only being used for a particular product [14, 46]. This can be particularly challenging in today’s need for multiuse facilities using different expression systems and platform technologies. The control of low contamination rates from a proactive rather than reactive approach results in less time and expense spent on investigations and fewer interruptions to the facilityprocessing schedule. The continued control of low contamination rates requires the sustained effort from a multidisciplinary team. Diligent attention to detail is required along with effective training and constant communication of contamination awareness. As we move forward, there will be continued momentum to reduce contamination rates with the increasing pressure on the industry to control costs and improve efficiency. The expansion of disposable single-use equipment will increase as scalability and standardiza­ tion of materials of construction improve. The use of Q-PCR and DNA detection methods such as DNA chip arrays will likely become common industrial practice. This will lead to rapid identification of contamination (hours instead of days) and should ultimately lead to improvement of identifying the sources of unknown contaminations especially in regard to raw material quality. The use of risk assessment tools will likely increase as we move toward a risk-based regulatory scrutiny. This will challenge the level of understanding of scientific knowledge/understanding of how the environment and the process impact product quality. References [1] US Food and Drug Administration (FDA) (1993) FDA Guide to Inspections of Validation of Cleaning Processes. Rockville, MD: FDA. [2] US Food and Drug Administration (FDA) (2004) FDA Guideline on Sterile Drug Products Produced by Aseptic Processing. Center for Drugs and Biologics, Division of Manufacturing & Products Quality (HFN-320). Rockville, MD: FDA. [3] Agalloco J and Akers J (2008) Sterile product manufacturing. In: Cox S (ed.) Pharmaceutical Manufacturing Handbook, Production and Processeses, 1st edn., pp. 99–135. [4] Hamid M (2008) Cleaning validation for biopharmaceutical manufacturing at Genentech. BioPharm International 21(3): 68. [5] Whyte W and Eaton T (2004) Microbiological contamination models for use in risk assessment during pharmaceutical production. European Journal of Parenteral and Pharmaceutical Sciences 9: 1–10. [6] International Conference on Organization (ICH) (2005) Draft Consensus Guidelines Quality Risk Management Q9. Geneva, Switzerland: ICH. [7] Lubiniecki A (1997) Potential influence of international harmonization of pharmaceutical regulations. Current Opinion in Biotechnology 8: 350–356. Aseptic Operations [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] 945 Sharma MC and Gurtu AK (1993) Asepsis in bioreactors. In: Neidleman S and Laskin AI (eds.) Advances in Applied Microbiology, pp. 42–57. New York, NY: Academic Press. Corbett K (1985) Design, preparation and sterilization of fermentation media. In: Moo-Young M (ed.) Comprehensive Biotechnology, vol. 1, pp. 127–139. Oxford: Pergamon Press. Soderberg AC (1983) Fermentation design. In: Vogel HC (ed.) Fermentation and Biochemical Engineering Handbook, pp. 77–118. Park Ridge, NJ: Noyes Publication. Bartholomew WH, Engstrom DE, Goodman NS, et al. (1974) Reduction of contamination in an industrial fermentation plant. Biotechnology and Bioenginnering 16: 1005–1013. Spier R (1998) Animal cells in culture: Moving into the exponential phase. Trends in Biotechnology 6: 2–6. Wu G and Liu X (2007) Characterisation of predominant bacteria isolates from clean rooms in a pharmaceutical production unit. Journal of Zheijang University 8(9): 666–672. Perkowski CA, Daransky GR, and Williams J (1984) Detection of microscopic leaks in fermentor cooling coils. Biotechnology and Bioengineering 26: 857–859. Junker B (1999) Good manufacturing practice (GMP) and good industrial large scale practices (GLSP). In: Flickinger MC and Drew SW (eds.) Encyclopedia of Bioprocess Technology: Fermentation, Biocatalysis and Bioseparation, pp. 1355–1367. New York, NY: John Wiley and Sons. Junker B (2006) Sustainable reduction of bioreactor contamination in an industrial fermentation pilot plant. Journal of Bioscience and Bioengineering 102(4): 251–268. Aleman M (2000) Some methods for contamination testing of a master cell bank. BioPharm International June: 48–52. Bruchmuller I and Pirkl E (2006) Introduction of a validation concept for a PCR-based Mycoplasma detection assay. Cytotherapy 8(1): 62–69. Stacey A and Stacey G (2000) Routine quality control testing for cell cultures. In: Kinchington D and Schinazi R (eds.) Antiviral Methods and Protocols, Methods in Molecular Medicine, pp. 27–40. New Jersey, NJ: Humana Press. Papavasileiou V, Siletti C, and Petrides D (2009) The role of single – use systems (disposables) in the development and manufacturing of biopharmaceuticals. Biopharmaceuticals Chemistry 8(1–2): 14–20. Garnick R (1996) Experience with viral contamination in cell culture. Developmental Biology 88: 49–56. Rabeneau H and Ohlinger V (1993) Contamination of genitically engineered CHO cells. Biologicals 21: 207–214. Pollard DJ, Telari K, Lane J, et al. (2006) Asymmetric reduction of α, β-unsaturated ketone to (R) allylic alcohol by Candida chilensis. Biotechnology and Bioengineering 93(4): 686–692. Whyte W, Green G, and Albisu A (2007) Collection efficiency and design of microbial air samples. Aerosol Science 38: 101–114. Brooks CH and Russell PD (1986) A unified design approach to fermentor sterility and containment. Process Biochemistry 21: 77–80. Guy M (2008) Fermentation equipment selection. In: McNeil B and Harvey LM (eds.) Practical Fermentation Technology, pp. 4–33. New York, NY: Wiley & Sons. Eibl R and Eibl D (2007) Disposable bioreactors for inoculum production and protein expression. Methods in Biotechnology 24: 321–335. McLeod L (2009) The road to a fully disposable protein purification process. BioProcess International 7(5): 4–7. Haughney H (2003) Integrating single use filtration systems in biopharmaceutical applications. Biotechnology 99(1): 43–48. Potgieter TI, Cukan M, Drummond JE, et al. (2009) Production of monoclonal antibodies by glycoengineered Pichia pastoris. Journal of Biotechnology 139: 318–325. Davis C (2007) Squeaky clean. BioPharma 794: 33–38. Anderson S (2008) Filtration of liquids using cellulose-based depth filters. PDA Journal of Pharmaceutical Science and Technology 62(2): 1–40. Sundaram S, Auriemma M, Howard G, et al. (1999) Application of membrane filtration for removal of bioburden organisms. PDA Journal of Pharmaceutical Science and Technology 53(4): 186–201. Christy C and Vermant S (2002) The state of the art of filtration in recovery processes for biopharmaceutical production. Desalination 147: 1–4. Pflug I and Evans K (2000) Carrying out biological qualification for producing sterile pharmaceuticals and medical devices. PDA Journal of Pharmaceutical Science and Technology 54(2): 117–135. Adamson SR (2000) Process validation and characterization: Animal cell culture processes. In: Sofer G and Zabriskie D (eds.) Biopharmaceutical Process Validation, 1st edn., pp. 101–128. New York, NY: Marcel Dekker, Inc. Valdes-Mora L (2004) Proper use and validation of disinfectants. Drugs and Pharmaceutical Science 142: 251–281. Bruckner J and Venkateswaran K (2007) Overview of methodologies to sample and assess microbial burden in low biomass environments. Japan Journal of Food Microbiology 24(2): 61–70. Agranovski IE and Agranovski V (2002) Collection of airborne microorganisms into liquid by bubbling through porous medium. Aerosol Science and Technology 36: 502–509. Pina R (2006) Testing compressed air lines for microbiological contamination. Controlled Environments 9(5): 11–16. Lappalainenen J and Loikkanen S (2000) Microbial testing methods for detection of residual cleaning agents and disinfectants. Journal of Food Protection 63(2): 210–215. Raymond N (2008) Detection of cache valley virus in biologicals manufactured in CHO cells. Biopharm International 21(10): 89–93. Xu Y and Brorson K (2003) An overview of Q PCR assays for biologicals. Development in Biologicals 113: 89–99. Chiu CY and Urisman A (2008) Utility of DNA microarrays for diction of viruses in acute respiratory tract infections in children. Journal of Pediatrics 52: 76–83. Kolman JL (2009) New technologies for the rapid detection of adventitious viral infections. PDA Cell Substrate Workshop. http://www.pda.org/Presentation/PDA-Cell-Substrate­ Workshop/John-Kolman.aspx Perkowski CA (1990) Operational aspects of bioreactor contamination control. Journal of Parenttral Science and Technology 44: 113–117.