As an industry, biopharmaceutical manufacturing is in transition and is facing challenges borne out of success. It was in the early 1980s that the first products were commercialized, namely the replacement hormones insulin and human growth hormone. The industry initially grew rapidly in the early ‘80s against a background of an immature supply industry: there were no large-scale columns (>10 cm diameter), column controllers, limited resin supplies and no established ultrafiltration technology. The position today is that the industry has achieved sales of around $85 billion in 2007 ( 1 ). This has been accompanied by a huge investment in capacity; for example, cell culture capacity has grown from less than 300,000 L pre-2000 to around 3 million litres today, driven by the requirements for Monoclonal Antibodies (MAbs). Although we often talk about the biopharm industry as a homogeneous entity, it is in fact highly heterogeneous, with mature segments such as the replacement hormones and traditional vaccines,...
In June 2007, Sartorius AG acquired the French company, Stedim S.A., and a global solution provider for the biopharmaceutical industry was created. Since then, the new company — Sartorius Stedim Biotech (SSB) — has entered into collaborations with a number of other key industry players, including Metroglas, Bayer Technology Services GmbH and the recently acquired Wave Biotech AG to bring technologies such as single-use bioreactors, UVC irradiation and the first disposable–integrated electrochemical pH sensor into its rapidly expanding product portfolio. Since the Stedim acquisition, SSB has developed a broad range of turnkey single-use technologies, anticipating market trends and keeping one step ahead of the competition. As the biomanufacturing industry moves ever closer to the concept of fully integrated, totally disposable processes, Dr Kevin Robinson spoke with Dr Joachim Kreuzburg, CEO and Chairman of the Board of SSB, to find out more about the company’s recent activities, what’s driving the sector ...
Biopharmaceutical drug products are not only well established but also contribute to a large degree to new drug entity filings. Currently approved biopharmaceuticals and proteins are now widely used to treat diseases as diverse as cancer, autoimmune disorders, myocardial infarction and various growth factor deficiencies. The unmet medical need can be so essential, such as a novel approach to cancer treatment, that biotech companies will choose to defer the optimal design of the production process to reduce the time-to-market. In other instances, the manufacturer may choose to defer investment in large-scale production facilities until a real-world understanding of the market potential of the new product is determined. The cost and start up times for large-scale protein production facilities is substantial, typically ranging from $500 million to $1 billion and taking approximately 3–5 years to achieve full regulatory approval. In addition, the timelines for modifications to existing facilities are also sig...
During the last 10 years, cost pressures and the changing requirements for bioreactors in the modern pharmaceutical industry have resulted in the increased use of disposable bioreactors in both R&D and manufacturing. Numerous studies have demonstrated their efficiency in cell culture-based upstream processing at small- and middle-volume scales. As shown in Figure 1, disposable bioreactors with culture volumes between 10 mL and 2 m 3 are most widely used for cell proliferation, screening experiments, the production of therapeutic agents (recombinant proteins, antibodies, secondary metabolites) and the production of viruses for gene therapies as well as veterinary and human vaccines ( 1 , 2 ). Figure 1: () The key element of every disposable bioreactor is its presterile cultivation container, which is made of FDA-approved plastics and is only intended for single-use. The cultivation containers (consisting of a multiwell plate, a tube, a flask, a cartridge or a bag) vary in design, polymer type, scale, inst...
The use of disposables in biopharmaceutical manufacturing has increased significantly during the last few years and is expected to grow substantially in the near future. Bioplan Associates report in their 2007 “Report and Survey of Biopharmaceutical Manufacturing Capacity and Production” that the main drivers for this ongoing trend continue to be the elimination of cleaning followed by the reduction of capital investment in facilities and the required equipment. However, with the further penetration of disposables into larger volumes and more critical areas, companies involved in biopharmaceutical development and the production of new molecules started to investigate more closely the advantages, limitations and facility implications of single-use manufacturing scenarios. Whereas in the past, disposables where recognized as useful components in the production of biopharmaceuticals, today a new buzzword has found its way into the biopharmaceutical lexicon. Disposable Factory As no clear definition and no co...
This paper describes the design, development and validation procedure for a novel, single-use gamma-stable electrochemical pH probe jointly developed by Sartorius Stedim Biotech SA and Metroglas. This new, single-use pH sensor offers a range of pH measurements (from 0 to 11 with ±0.1 precision) and features a one-point calibration process in its storage solution that provides a fast and easy pre- and post-use sensor performance check. Also described is a specific encapsulation device designed to integrate the pH probe into single-use containers used to hold, mix, store, transfer and transport biopharmaceutical fluids and drug products. Finally, a case study is detailed that outlines the potential application of a combined single-use pH probe and mixer technologies for buffer formulation applications. Single-use technologies have been widely adopted by the biopharmaceutical industry for the management of buffers, media, cell culture, harvest, cleaning agents and intermediate products. As single-use technol...
Evaluating single-use systems for extractables and leachables is new territory for many end-users. This paper presents a risk-based approach for evaluating extractables and leachables from single-use systems. Approaches are described that have been accepted by various regulatory agencies utilizing risk assessments and sound scientific principles. Biopharmaceutical processing materials must be evaluated to determine whether they impact the final drug product with regard to safety and efficacy. For single-use and reusable systems, this usually leads to evaluations that demonstrate compatibility with the process formulations. Many of these evaluations can be performed by the vendor and do not need to be addressed by an end-user. Examples of these are as follows: A comprehensive list of tests that may be performed for single-use systems has been published by the Bio-Process Systems Alliance (BPSA) ( 3 ). Regulatory Requirements for Single-Use Systems: Extractables and Leachables Single-use systems, also refer...
The combination of innovative and traditional process technologies has resulted in major advancements in the antibody industry, such as accelerated process development and time-to-market. In addition, this paper examines the avenues that have opened as a result of exploring established process technologies for new applications, as in the case of perfusion cell cultures to amplify dhfr-based expression cell lines by incrementally increasing selection markers in the perfusion medium for the faster generation of stable and high-productivity clones. Furthermore, performing perfusion processes in single-use bioreactors has provided a convenient means of generating large amounts of cells for high density freezing in cGMP cell banking operations. High cell density freezing minimizes the inoculum expansion time from vial thaw to seed bioreactor and leads to cost-effective facility utilization. All phases of cell line development have been integrated using a single medium — from host cell bank adaptation to high l...
This paper describes how RFID (Radio Frequency Identification) technologies can be implemented into single-use systems to generate electronic records for both bag manufacturers and bag end-users. RFID technology will enable the user to both read and write all relevant product and process information directly onto the single-use bag, providing instantaneous data recall. The tag can also provide the user with immediate access to the bag’s original part number, lot number, date of manufacture, expiration date and other critical item-level data that is normally contained in the product certificate of quality. Gamma-stable RFID tags attached to disposable bag systems, writing and reading software and handheld data reader systems can now be used to offer customers a unique paperless record, improve traceability, retrieve release documentation and leverage the capabilities of RFID in their facilities. The system presented in this paper utilizes Radio Frequency Identification technology to write identification in...
The most important contributions to high-yield manufacturing processes for the production of recombinant proteins from cultivated mammalian cells have come from the identification of highly enriched and well-balanced media formulations, and fine tuning the process conditions that support high cell culture densities with high specific productivity. The industry standard yield for immunoglobulins or similar molecules derived from suspension-cultivated mammalian cells in bioreactors has risen during the past 20 years from the tens of mg/L to g/L. The more favourable conditions used today for high-yield, large-scale protein expression have typically been identified in small-scale bioreactor systems. Using these “scale-down” systems, attempts were made to match the physicochemical constraints, as closely as possible, to those found in very large bioreactors (up to 25,000 L). But the question remained, how was it possible to establish and use high-throughput, scale-down systems that successfully mimicked such c...
Disposable bioprocessing has come of age. Economic and regulatory conditions are driving the widespread adoption of disposable equipment at all stages of bioprocessing. This review considers the entire bioprocess chain and assesses the status of disposables. In particular, we focus on the current availability and need for additional sensors that will enable the disposable process to be integrated — in compliance — with the latest Process Analytical Technologies. Traditional bioprocessing is highly compartmentalized into upstream and downstream operations. Within these broad divisions, one finds early stage, pilot and manufacturing units as further subdivisions. In an ideal world, there should be a seamless handoff from discovery through to manufacturing. In reality, many companies operate with departments that end up being vertically integrated, but not horizontally. The rising costs of drugs and economic pressures are creating a “perfect storm.” Coupled with the sequencing of the human genome, there is n...
Drug manufacturers face the very real challenge of being both innovative and efficient — having to get products to market quickly — whilst at the same time facing existing hurdles that can limit both of these goals. To manufacture products innovatively and quickly, while at the same time reducing costs and ensuring quality, drug manufacturers must find ways to build quality into their processes. Doing so will aid in product approvals, cut down on poor design issues and ultimately lead to a reduction in costs as, currently, quality assessments rely heavily on end-product testing. The pharmaceutical industry and regulators understand the need to move from end-product testing to a more enhanced quality model. The ideal model would utilize the concepts of quality by design, risk management and quality systems for identification, prioritization, design and product quality control. The focus should now be on how to overcome the hurdles to implementing these key concepts. Traditional Pharma Compared with many in...
Generic validation is conceivable only through a thorough understanding of the parameters affecting the performance of a process step. In this paper, we provide a detailed example demonstrating the robustness of a virus filtration step. As a first step towards the establishment of a generic validation package for a monoclonal antibody, the robustness of clearance of PP7 across the ViroSart CPV filter was evaluated by changing several critical operational parameters using a simple one-off experimental design. Two different validation approaches were used during this study: the classical validation approach and the “run-and-spike” approach. This first screening experiment, combined with data accumulated for several other products, provides valuable insights for the future development and validation of robust virus filtration steps. Virus filtration has become a standard feature of modern biomolecule purification processes. Indeed, this step is generally capable of providing robust clearances of small viruse...
The biomanufacturing industry has issues. From key drivers and hurdles, to the spectre of leachables legislation and the need for greater harmonization between suppliers, the biotech sector is experiencing a period of growing competition and increasing pressure. But, it is also a market with a future; the commercial success of more than 350 approved biologics has prompted the biotechnology industry to accelerate discoveries in further protein-based therapeutics, placing greater emphasis upon the importance of biomanufacturing. In addition, it is predicted that pharmaceutical biomanufacturing could account for more than 20% of the global contract manufacturing market in 2012. New disposable technologies, build-versus-buy decisions and opportunities to collaborate and co-operate were high on the agenda when Sartorius Stedim Biotech invited some of the industry’s elite to attend a roundtable discussion and debate the pros and cons of single-use technologies to produce cost-effective biomanufacturing services...
In my opening editorial, I referred to an industry in transition. This is being driven by challenges that are by no means unique to biotech: all other industry sectors are experiencing similar pressures. We are seeing the impact in terms of factory design, manufacturing technologies, validation and business processes; these are all themes that have been addressed in this supplement. The big innovation in terms of manufacturing systems has been the wholesale acceptance of disposable manufacturing technologies during the last 10 years. As the industry moves to a strategically significant level of single-use technology integration, then we have to rethink our approach to manufacturing. Why? Well, for the stainless steel facility, the manufacturing assets are physical, owned and installed by the operator; however, in the case of the facility that is largely single-use, the operator is dependent on the suppliers and the whole supply chain delivering the manufacturing capacity when needed. If we go for a lean m...
The contamination of microbiological media by mycoplasmas such as Acholeplasma laidlawii is not a recent phenomenon. It has been a major problem with animal-derived sera since the 1980s and has been a concern in the management of cell cultures for decades. The main culprit of serum contamination was the inadequate blood collection methodology and was eliminated with the introduction of hollow collection needles. In addition to the introduction of an improved collection method, serum was filtered with 0.1 µm-rated membrane filters to isolate potential mollicute contaminants, albeit with varying degrees of success ( 1 , 2 , 3 , 4 ). Improved collection and filtration technologies mitigated the mycoplasmas contamination problem, until the threat of spongiform contaminants in animal-originated media initiated a switch to plant peptone-derived cell culture media. One contaminant was exchanged by another, which, once again, turned out to be mycoplasmas. This time, however, to varying degrees, the contaminat...