Cheryl Scott

September 1, 2010

13 Min Read

With infusions of public and private venture capital as well as technological advances, vaccine development is entering a new golden age as one of the fastest growing sectors in the biotechnology industry. In the 19th and 20th centuries, immunization programs eliminated or controlled infectious diseases including smallpox, polio, measles, mumps, and rubella. The biotech era has made significant changes both in the number of companies involved in vaccine manufacture and the production systems they use.

BPI CONFERENCE SESSIONS

Wednesday, 22 September 2010
Keynote Presentation: Global Vaccine Production Challenges — Emerging Immunotherapeutics, Manufacturing Flexibility, and Reducing COGS

Lessons Learned from the 2009 Flu Season to Guide Rapid Vaccine Development and Manufacturing Scale-Up

Process Development for Novel Vaccines and Immunotherapeutics

Defending Biosimilar Competition: Bioprocess IP Protections for Next-Generation Vaccines and Immunotherapeutics

Process Development and Analytical Characterization for Vaccine Production

BPI_A_100808SUPAR05_106242b.jpg

The benefits of cell-culture based vaccine production are clear to most everyone. Insect cell culture is of particular interest to flu vaccine companies because of its baculovirus expression system (BVES) — an obvious method for producing viral subunits and virus-like particles (VLPs). VaxInnate Corporation uses a microbial expression system to make influenza vaccines and will present on them at the 2010 BPI Conference.

According to René Labatut (vice president of global manufacturing technology for Sanofi Pasteur), with “last strain selection in February, first clinical doses in May, and vaccine delivery in September,” traditional egg-based and mammalian cell culture flu vaccine production both operate on about a six-to eight-month timeline. BVES-based insect cell production can be done in five to six months. Even small improvements can make a big difference in a pandemic situation. Labatut’s colleague Pierre Fourier (associate vice president of manufacturing technology at Sanofi Pasteur) will present a keynote address on Wednesday 22 September 2010 at the BPI Conference on global vaccine production challenges.

But influenza isn’t the only application of biotechnology to vaccine manufacturing. Norovirus infection is the most common cause of acute gastroenteritis in the United States, afflicting an estimated 23 million people every year. To generate a bivalent vaccine that can confer broad protection against this disease state, LigoCyte Pharmaceuticals is using the baculovirus expression system to make two distinct norovirus VLPs. They are purified by chromatography steps in downstream processing. At the BPI Conference, Ross Taylor (the company’s process development director) will present results from CGMP manufacturing to support phase 1 clinical trials.

Vaccines present special issues for manufacturers, particularly in scale-up, cost, and the pace of pathogen mutation. Good laboratory and manufacturing practices must be ensured, and product safety, efficacy, and stability must be maintained. In the new economic environment, time-to-market and flexibility are becoming increasingly important to improving cost-effectiveness. New support technologies can simplify development and manufacturing.

Although the vaccine market is still dominated by a select group of key players, several biotechnology companies are developing new ideas and product approaches such as cancer vaccines and immunotherapies to expand the vaccine market even further. And emerging geographic regions are poised to dominate both vaccine manufacturing and use in the not-too-distant future. Globalization is offering new routes to developing products and increase market access.

Q&A WITH FEATURED PRESENTER

Andrew Clutterbuck (purification development team leader for Eden Biodesign Ltd.) will present in the “Rapid Vaccine Development and Production” session at 5:30 on Wednesday 22 September 2010: “From Bench to Bag: Deployment and Implementation of Novel Solutions for Vaccine Production.”

BPI: In your presentation abstract, you called traditional adenoviral purification process (e.g., centrifugation) “laborious, time-consuming, and scale limiting.” Can you be a little more specific? how fast, how slow, and at what scales? and what about cost?

AC: Typically, the scale-up of centrifugation processes is done in a modular way — increasing the number of centrifuges to gain capacity, each of which required not only significant capital investment but also the utilities and infrastructure and experienced personnel to support the process. A chromatography-based approach allows for linear scale-up by proportionally increasing column sizes to accommodate the process stream. This is far easier, faster, and cheaper.

BPI: You also mentioned disposables “and novel technologies.” What other concepts will you highlight beyond single-use technology?

AC: There are a number of technologies being exploited for the production of viruses and vaccines, the most notable of which are biomonolith technology (from BIA Separations) and fully disposable solutions. I can envision a time in the not-too-distant future when a full production process from bioreactor through to fill finish can be performed using a fully disposable and integrated production process. This is something we are currently actively exploring at Eden Biodesign.

BPI: We recently published a special report on the globalization of biomanufacturing — focusing on how technological advances are making it possible. do you see a rise in capabilities for bioprocessing outside Europe and North America? Is this more prevalent for vaccine production than for other biopharmaceuticals?

AC: I would say so, yes. There is a large and emerging market in Asia, South America, and the Middle East. It would be the next logical evolution of vaccine manufacturing to move into these markets, which would include significant investment in production facilities, support functions (e.g., cold chain logistics), and local training.

Lessons Learned from 2009

Pandemic planning enables nations to prepare for an influenza (or other) pandemic outbreak. It’s intended to help reduce virus transmission, decrease infection cases overall, maintain essential services, and reduce the economic and social impacts of such outbreaks. The 2009–2010 H1N1 “swine flu” outbreak may have served as a sort of dress rehearsal for such planning systems.

Vaccine manufacturers are unique among biotechnology companies in their need for rapid response to mutant versions of existing pathogens or sudden emergence of new, highly infectious ones. New vaccines must hit the ground running, scaling up either through larger and larger batches or through increasing numbers of batches. Sometimes a manufacturing process and scale-up strategy must be developed simultaneously. Risk analyses help companies determine the candidates most lik
ely to be effective and get them into production as soon as possible — because these products may go to market without as much testing as other biologics undergo.

Considering the uncertainties of product development and the capital expenditure required to develop a facility for a 10,000-L bioreactor process — as well as the biosafety containment levels necessary for viral vaccine manufacturing — vaccine companies are following the lead of their biopharmaceutical colleagues toward process intensification. At the BPI Conference, Ciska Dalm (a senior scientist in upstream process development at Crucell NV) will highlight her company’s progress in upstream development, showing a tenfold intensification at bench scale and in 50-L single-use bioreactors for rAd35 manufacturing.

Q&A WITH FEATURED PRESENTER

Michael Moussourakis (technical manager at Pall Life Sciences) will present a case study at 2:45 PM during the “Process Development and Analytical Characterization for Vaccine Production” session on Wednesday 22 September 2010: ”Particle and Endotoxin Control in Form/Fill Tubing Manifolds for Vaccines.”

BPI: What analytical techniques (and why) did you use in quantifying/qualifying particles and endotoxins involved in vaccine fill/finish? How do they complement one another?

MM: Single-use disposable systems for vaccine fill/finish operations were filled and agitated with particulate-and endotoxin-free reagent water. A single water sample could be obtained and analyzed for both particulate and endotoxin content. The analytical technique used for endotoxin detection was the Limulus amoebocyte lysate (LAL) kinetic chromogenic method. For particles, the system rinse was passed through an analytical membrane filter and examined microscopically. These methods are routinely used for analysis of pharmaceutical fluids.

BPI: What regulatory/guidance documents did you refer to in developing your methods?

MM: For endotoxins, USP Bacterial Endotoxins; for particles, USP Particulate Matter in Injectables.

BPI: Can you describe what the form/fill tubing manifolds are and how they work within a fill/finish system? Are they applicable to other biopharmaceutical products?

MM: Formulation and filling systems are essentially multiple-port tubing manifolds used to either combine several formulations into a single combined formulation (e.g., for a polyvalent vaccine) or dispense a single formulation into multiple smaller containers. There are many different variations of these functions. The single-use disposable system tubing manifolds contained various arrangements of Pall Allegro™ biocontainers (bags), Pall Kleenpak™ sterile connectors, Pall Kleenpak filter capsules, and associated silicone tubing and polypropylene fittings and adapters. By combining and building different single-use/disposable manifold systems, they can be applicable to formulating or filling of nearly any biopharmaceutical product.

BPI: Will this be your first time as a BPI Conference presenter? What do you think of the location this year? is there anything you’re particularly looking forward to?

MM: Yes, this is my first time as a speaker at the BPI conference. I am particularly looking forward to reacquainting with familiar faces and networking with others throughout the industry. The location this year places it at a central northeastern point and provides convenience for many attendees including those in the New York, Massachusetts, Connecticut, and New Jersey areas.

Knowledge Is Power

Even responding to the natural genetic drift in seasonal influenza viruses necessitates yearly process modifications by flu vaccine manufactures. Changes made to analytical techniques and process steps must happen quickly (within as little as eight weeks) and satisfy the regulatory requirements of a licensed vaccine. At the BPI Conference, Robert Boulanger (production manager at Protein Sciences Corporation) will outline how those requirements have evolved over the years for recombinant hemagglutinin-based vaccine candidates.

As with antibodies and other recombinant proteins about a decade ago, vaccine makers are discovering the value of powerful new analytical techniques to their work. Especially for products that don’t spend as a lot of time in clinical testing, good characterization work can be the key to success. “Efficiency is the key issue around vaccines,” explained René Labatut in a recent email exchange. That’s unlike the activity question that’s essential to so many therapeutics.

Even as new technologies improve production and processing, however, they can introduce new questions into the analytical mix. For example, single-use tubing manifolds used in sterile formulation and filling of vaccines can be a potential source of particles and endotoxins. Vendors of such products are working to help their customers characterize the associated risks. At the BPI Conference, Michael Moussourak is (technical manager for Pall Life Sciences) will describe test-method development and qualification, presenting data collected over time from multiple system designs and lots, for determining particulate matter and endotoxin content in radiation-sterilized single-use tubing manifold systems.

Of course, better characterization methods have made biosimilar competition inevitable, and recent legislation in different regions has brought the prospect of “well-characterized vaccines” closer to reality. One key to forestalling “generic” competition for biologics is the ability to develop a comprehensive IP portfolio that includes not only a biologic and its method(s) of administration, but its method of manufacture as well. At the BPI Conference, George A. Xixis (a partner in the law firm of Nutter McClennen & Fish) will present a hypothetical case to examine issues involved in putting together such an IP strategy, with examples of some useful techniques in light of recent case developments.

Process Development Case Studies

Part of the US National Institute of Allergy and Infectious Diseases, the Vaccine Research Center (VRC) develops vaccines against a broad range of viral infections. For example, chikungunya is a mosquito-borne disease that is growing in significance as its host species expands in range and numbers. So the VRC has a VLP-based chikungunya vaccine in phase 1 clinical testing. Richard M. Schwartz (chief of the center’s vaccine production program laboratory) will be at the BPI Conference to report on optimization of the early phase manufacturing process based on transient transfection in cell culture.

VaxInnate’s proprietary approach to influenza vaccines is based on genetic fusion of a Toll-like receptor 5 agonist to a protective subunit of the hemagglutinin protein. Resulting chimeric proteins can be produced rapidly at low cost and high volumes using standard bacterial expression systems and protein purification techniques. At the BPI Conference, Bruce Weaver (the company’s director of process development and formulations) will describe a strategy to manufacture US pandemic stockpiles in three months based on VaxInnate’s experience dealing with avian and swine influenzas.

Traditional purification processes for adenoviral viral vectors (e.g., density gradient centrifugation) are considered laborious, time consuming, and scale limiting. Recent interest in rapid, flexible, and scalable vaccine production has demanded a radical rethink, according to Andrew Clutterbuck (purification development team leader at Eden Biodesign Ltd.). He will present a case study at the BPI Conference of a manufacturing process that could offer a general approach for other vaccine platforms as well. From small-scale process development to CGMP manufacturing, his team u
sed disposables and other new technologies in both production and purification of an Ad5 product. For the industry at large, single-use bioreactor and purification technology could enable production in multiple facilities without affecting product quality.

Cancer vaccines and immunotherapies present their own challenges. For example, autologous RNA and dendritic cell processing methods are complicated by variability in their biological starting materials. Despite that variability, Argos Therapeutics has been able to develop robust processes for manufacturing autologous dendritic cell immunotherapies both for oncologic and infectious diseases. At the BPI Conference, Tamara Monesmith (the company’s director of manufacturing and process development) will describe how such processing methods have been used to produce materials for use in phase 2 clinical trials of renal cell carcinoma and human immunodeficiency virus indications.

Identifying suitable antigens, adjuvants, and delivery methods are just the beginning of vaccine development. Regulatory, technical, and manufacturing hurdles stand in the way of companies seeking to take a candidate product to the clinic — and eventually to market. Process development provides a technological foundation for manufacturing. And as antibody and other protein developers can tell you, analytical method and assay development for characterization and potency determination must be part of it. This lays the groundwork for successful commercialization.

About the Author

Author Details
Cheryl Scott is senior technical editor of BioProcess International.

REFERENCES

1.) Ball, P, C Brown, and K. Lindström. 2009. 21st Century Vaccine Manufacturing: Examining the Potential of Rapid Analytical Methodologies and Worldwide Supply Chains. BioProcess Int. 7:18-28.

2.) Capodici, J. 2006. Large-Scale Beta-Propiolactone Inactivation of HIV for Vaccines. BioProcess Int. 4:36-41.

3.) De Boer, E. 2008. Optimizing Vaccine Supply Chains Through Quality Management in Manufacturing. BioProcess Int. 6:S38-S40.

4.) Foulon, A. 2008. Using Disposables in an Antibody Production Process: A Cost-Effectiveness Study of Technology Transfer Between Two Production Sites. BioProcess Int. 6:12-17.

5.) Gerson, DF, and B Mukherjee. 2005. Manufacturing Process Development for High-Volume, Low-Cost Vaccines. BioProcess Int. 3:42-47.

6.) Gombold, J. 2006. Lot Release and Characterization Testing of Live-Virus–Based Vaccines and Gene Therapy Products, Par t 2: Case Studies and Discussion. BioProcess Int. 6:56-65.

7.) Hitchcock, T. 2009. Production of Recombinant Whole-Cell Vaccines with Disposable Manufacturing Systems. BioProcess Int. 7:36-43.

8.) Kowolenko, MD. 2009. The Vaccine Renaissance: One Company Rises to the Manufacturing Challenge. BioProcess Int. 7:72.

9.) McLeod, L. 2008. From Pandemics to Bioterrorism: The Role of Bio-Manufacturing in Global Healthcare. BioProcess Int. 6:26-32.

10.) McLeod, LD. 2009. Novel Vaccines and Virology. BioProcess Int. 7:S34-S38.

11.) Mørtz, E. 2008. Proteomics Technology Applied to Upstream and Downstream Process Development of a Protein Vaccine. BioProcess Int. 6:36-43.

12.) Nechaeva, E. 2004. New Technology for Producing Live Influenza Vaccine: Using a Serum-Free Medium Supplemented with Additives and Stabilizers of Plant Origin. BioProcess Int. 2:52-55.

13.) Offit, PA. 2005. Vaccine Roadblocks: What Can We Learn from History?. BioProcess Int. 3:72.

14.) Smith, M. 2009. Rapid Assessment of Vaccine Potency: How Automation Can Help. BioProcess Int. 7:52-55.

15.) Wassard, K, and M. Monge. 2007. Rapid Implementation of a Smallpox Vaccine Facility: A Case Study in Single-Use Technology. BioProcess Int. 59 7:32-37.

16.) Whitford, WG. 2010. Using Disposables in Cell-Culture –Based Vaccine Production. BioProcess Int. 8:S20-S27.

You May Also Like