Monday, September 30, 2013

What is the value of antibody engineering in immunotherapy?

We at the Future of Biopharma blog recently had some time to sit down and speak with speakers from the Antibody Engineering and Therapeutics event. Today speaker Dr. Holbrook Kohrt, Associate Professor in the Department of Medicine in the Division of Oncology at Stanford University shares with us what he thinks the value of antibbdy engineering in immunotherapy is.
I actually think that antibody engineering is critical in the area of immunotherapy because of the interaction of a novel antibody that we are engineering today and applying for patients with cancer. 
What we’ve identified over the past 20 years, since the initial development of Rituximab, is an antibody that targets CD20 on the surface of non-Hodgkin’s lymphoma. Now, this antibody – Rituximab – was put into Phase I testing in 1994, FDA approved in 1997 and today is approximately an $8 million worldwide market in immunotherapy and induces responses anywhere between 60 and 90% of non-Hodgkin’s lymphoma patients and has been demonstrated to improve survival. 
Since the development of Rituximab, there have been multiple other antibodies developed, such as Trastuzumab, which targets HER2 on the surface of breast cancer, as well as Cetuximab, which targets EGFR on the surface of head and neck, as well as in colorectal and potentially in lung and other tumor types. 
Unfortunately, these monoclonal antibodies have failed to be the magic bullet that we all once hoped. So today the major additional effort is trying to identify how we can improve the efficacy of these monoclonal antibodies and thus the importance of antibody engineering.

For a long period of time we have considered antibody engineering really just as enhancing a single monoclonal antibody to improve its efficacy. But what I argue is that actually there is an alternative paradigm changing approach by antibody engineering to two targets simultaneously. Now this may sound similar to biospecifics or diabodies. But in fact, what I’m referring to is targeting one target on the tumor itself and a second target on a completely different cell type, specifically on an infector cell within the immune system. 
So, how do monoclonal antibodies work? Well, the majority monoclonal antibodies, such as Rituximab, work by ADCC – Antibody Dependent Cell-Mediated Cytotoxicity. In this process, a monoclonal antibody binds to the surface of the cell of interest, such as the lymphoma cell or breast cancer cell or the head and neck cancer cell. Once that first antibody binds, the Fc portion of that antibody recruits and binds to parts of the immune system, specifically natural killer cells or any cells bearing an Fc receptor. Now the affinity of that interaction is very important. So, if we can improve the first monoclonal antibody by making that affinity even higher, that’s one success. But the second success really comes by targeting the immune system. How can we do that? When these cells – the natural killer cells – bind to the Fc portion of a monoclonal antibody, that interaction triggers stimulation of them. That stimulation helps regulate things on their cell surface.

For the rest of Dr. Holbrook's answer, download the full podcast and PDF here.

Dr. Kohrt will be presenting Stimulation of Natural Killer Cells with an Anti-CD137 Antibody Enhances the Efficacy of Trastuzumab, Cetuximab, and Rituximab in HER2-expressing Breast Cancer, EGFR+ Head and Neck Cancer, and CD20+ Lymphoma on Wednesday, December 11 at the Antibody Engineering and Therapeutics Event.  For more information on his session and the rest of the program, download the agenda.  If you'd like to join us, as a reader of this blog, when you register to join us and mention priority code XD13172BLOGJP, you can save 20% off the standard rate!


Share this article with your social network, just click below to share now!


Friday, September 27, 2013

FDA Perspective - Changing The Manufacturing Process to Remove Impurities

As the complexity of therapeutic protein increases, so do the challenges of controlling variants and impurities during process and product development, for scientists must continue to implement new methods and strategies to mitigate everything from quality control an assurance issues to immunogenic consequences.

Attend IBC's Product and Process Variants & Impurities conference to hear comprehensive updates on the latest approaches, methods, and technologies. to overcome these issues. An eclectic mix of regulatory, industry and academic speakers provide a diverse perspective on the topic, including:

• FDA perspective on changing the manufacturing process to remove impurities

• Industry panel discussion on biophysical methods to detect impurities

• Pfizer's technology roadmap to navigate the characterization landscape for increased product understanding

• University of Kent's list of challenges of product- and process related impurities to an evolving biopharmaceutical industry • Genentech's use of nucleic acid technologies to for early mutation detection

• NIST's sub-visible particle round-robin comparison on the level of agreement on sizing and counting

• Amgen's aggregation control strategy in manufacturing, QbD expectations and other process specifications

Want to learn more? Download our agenda.

As a member of the Bioanalytical Method Development group, you’ll receive 15% off the standard rate when using code XB13194BLOG to register. If you have questions about the event, contact Kate Devery (Kdevery@iirusa.com), or visit our webpage.

We look forward to seeing you October 21-23 in Washington, D.C!

Cheers,
The PPVI Team

PPVI Webpage
Follow us on Twitter
Join PPVI on Linkedin

P.S. When you register for IBC’s Produce and Process Variants & Impurities conference, you’ll also have access to all sessions for our co-located event, Well Characterized Biologicals.


Share this article with your social network, just click below to share now!


Thursday, September 26, 2013

Antibody Engineering Presentation: Exploiting light chain diversity to create a fully human bispecific IgG platform technology

Over the next few weeks, we'll be looking at some of the best presentations of last year's Antibody Engineering & Therapeutics event.  Today, we feature the presentation Exploiting light chain diversity to create a fully human bispecific IgG platform technology presented by Nicolas Fischer, Ph.D., Head of Research, Novimmune, Switzerland.

During his presentation, Dr. Fischer discussed how kλ-bodies are unmodified fully human bispecific IgGs that contain two different light chains. In contrast to existing engineered formats, kλ-bodies are unique in offering the typical functional and biochemical characteristics of a human antibody. A platform for the isolation of specific and neutralizing antibodies based on light chain diversification as well as a streamlined approach for production and characterization of kλ-bodies will be demonstrated.


Download the full presentation here.


This year, Dr. Fischer will be back to present Exploiting Light Chain Diversity Supports the Development of Multiple Therapeutic Antibody Formats.  For more information on this session and the rest of the program, download the agenda.  If you'd like to join us December 8-12, 2013 in Huntington Beach, California, as a reader of this blog, when you register to join us and mention priority code XD13172BLOGJP and save 20% off the standard rate!


Share this article with your social network, just click below to share now!


Wednesday, September 25, 2013

What are the techniques used to better understand polyspecificity?

Leading up to the Antibody Engineering & Therapeutics Event, podcast, we'll have interviews with a few of the speakers from the event.  Today we feature a portion our interview with James Crowe, Jr., M.D., Director, Vanderbilt Vaccine Center, Vanderbilt University Medical Center.

Today, he answers the question:
What are the techniques that your group has used to better understand polyspecificity?
Dr. Crowe: In order to sample the interacting energies of a large number of antibodies and antigens, it’s currently not possible to express thousands or millions of antibodies, at least not in an economic manner to measure energies. So, we’ve been focusing on using computational modeling experiments to determine the predicted interacting energies. In particular, we’ve been using a computational suite called Rosetta that was developed in Seattle. They have a Baker Lab and he’ll be talking at the IBC Conferences and going over the impact of Rosetta and structural modeling.

We’ve used Rosetta to ask: “What are the ideal or the optimal framework residues that can make antibodies have the ability to bind more than one antigen?” So, within Rosetta you cannot only model the binding to one antigen, but you can do a process called “multi-state design” in which you are asking the antibody to have more than one state – more than one bound state – which is binding to more than one antigen. By doing this, we asked Rosetta at many residues in the framework that we saw mutated in naturally occurring antibodies to just --- of the 20 amino acids that are available to predict which amino acids would allow binding to more than one antigen. Very remarkably, Rosetta predicted amino acids that are the germline encoded residues at those positions, which it is highly unexpected that a computer could pick at multiple positions the correct amino acid of 20 – an ensemble that would allow binding to more than one antigen.

So, the computational modeling is done on supercomputing. It is very computationally intensive. It predicted what the optimal sequences are and lo and behold, the optimal sequences for binding more than one antigen turned out to be the very germline sequences that we’re using in our genome. So, it looks like germline sequences are already in a state. It’s as if they are designed perfectly to be polyspecific and it may be that many – if not most – antibodies are polyspecific when they start.

The process of somatic mutation is, in essence, a two-step process. One in which the framework residues are mutated to reduce flexibility and rigidify the framework to orient the loops in a proper configuration. Then somatic mutations occur in the tips of the loops to adjust the interface so that their shape is complementarity and other interactions. So, really reducing flexibility achieves specificity and then you get some optimization of surface by somatic mutation.

So, we’ve focused on computational modeling, but the validation of the results of those computational experiments was the observation that the residues that are predicted are, in fact, the ones that are encoded by the genome. Even though the computer program didn’t know what those sequences were, it predicted them and they are the naturally occurring ones. So, we’ve been using Rosetta, trying to develop new modeling techniques within Rosetta and furthermore developing statistics to ask how expected or unexpected are the findings. It’s really quite remarkable that the computer can pick out the naturally occurring residues.

Dr. Crowe will be presenting Deep Sequencing the Human Antibody Response to Viral Infections and Human Germline Antibody Gene Segments Encode Polyspecific Antibodies. For more information on these sessions and the rest of the program, download the agenda. The Antibody Engineering & Therapeutics Event will take place December 8-12, 2013 in Huntington Beach, California. If you'd like to join us, as a reader of this blog,when you register to join us and mention priority code XD13172BLOGJP to save 20% off the standard rate.


Share this article with your social network, just click below to share now!


Tuesday, September 24, 2013

Biorepositories Preview: Where can you continue your Biorepositories experience after the hall is closed?

About the author: Alex Gelman is the Managing Partner of Poplar Partners, an investment firm looking to acquire a company in the biorepository space. Before founding Poplar Partners, he worked at KKR Capstone, the operating arm of private equity firm Kohlberg Kravis and Roberts, and at McKinsey &Co. He has an MBA from the Stanford Graduate School of Business and a BA from Dartmouth College.


6th Annual BioRepository and Sample Management Conference Preview:
Where can you continue your Biorepositories experience after the hall is closed?

I hope everybody is as excited about this week’s conference as I am. As my last pre-conference post before Biorepositories 2013, I thought it’d be helpful to highlight some attractions in the surrounding Seaport District area.

My first piece of advice would be to go by the Seaport Hotel Bar and try their new golden honey crafted beer from partner Long Trail Brewing Company. The excitement around this new addition is that the honey used in crafting this beer comes directly from the source, the Seaport Hotel. As an eco-friendly hotel, their 11 rooftop hives have gathered several hundred pounds of honey in their most recent harvest, which was sent up to Long Trail Brewing’s production facility. If you’re looking for the perfect fall beer, the Seaport Hotel Bar is a great spot.

Another highlight in the up and coming Seaport District is Sam’s located above the high-end department store Louis and just around the corner from the Seaport Hotel on 60 Northern Ave. With a huge wraparound deck and floor to ceiling windows, the views of the Boston Harbor are too good to resist. Offering a creative twist to the American classics, Sam’s is a highly coveted restaurant in the area.

The main draw to the Seaport District is its restaurants, but if you’re looking to go to the heart of Boston, I suggest you make your way towards the Faneuil Hall Marketplace. Over 70 retailers and 40 office tenants occupy the 200,000 square feet of retail and 160,000 square feet of space on Boston’s iconic mixed-use festival marketplace. There you can enjoy unique shops and cuisine as well as music and street performers. The world-famous Quincy Market Colonnade is an obvious attraction but you can’t go wrong with any of the diverse experiences found throughout the marketplace. Despite the Red Sox being out of town this week, the Faneuil Hall Marketplace is guaranteed to provide fun entertainment.


Share this article with your social network, just click below to share now!


What are some of the sessions that highlight this year's Cell Therapy Bioprocessing Event?

The Cell Therapy Bioprocsesing event taking place October 21-22, 2013 in Bethesda, Maryland.  For more information on the event, download the agenda.  If you'd like to join us, as a reader of this blog, you're can save 20% off the standard rate when you register to join us and mention code B13188JP20!

Leading up to the Cell Therapy BioProcessing event, we've interviewed several of the key stakeholders who helped shape this year's event.  We'll start by looking at the recent interview we conduced with Event Chair Lee Buckler, the Founder and Managing Director of the Cell Therapy Group.

Today, Buckler answers the question:
What are some of the sessions you are particularly looking forward to in this year’s program? And who are you looking forward to meeting while there?

Lee: Well, I always look forward to meeting my friends, of course, and I’ve been in this sector for a lot of years. So, it’s always good to see people I know and hang out with them. But, inevitably, you meet new, fresh faces as well and I think there’s any of a number of them this year that I’m looking forward to meeting and hearing from at the different companies. There have been a couple of important additions this year, which is going to result in some interesting sessions and be the infusion of new people who I look forward to meeting. One of those is around the production of iPS cells, which is not an area that I have a tremendous amount of experience or exposure to. So, I’m always keen to hear about what is happening on that front with the potential clinical production of human-induced pluripotent stem cells. Then, also, there is a fair amount of focus this year on the production of immunotherapies. I’m anxious to hear what Novartis is doing, for instance, with a program that they picked up at the University of Pennsylvania. They bought the old Dendreon Facility in New Jersey and are looking to replicate and do some process improvements on that process. I’m keen to hear about that. 
And then the tissue in bioengineering track, as well, is a new addition to this year’s Conference and is always interesting to hear about the intersection between cells and scaffolds and constructs because I think that’s where a lot of innovation is going to take place where we can combine cells either with genetic manipulation or with biologically inerts or non-biological material.


You can download Lee's full podcast here. Next week, Lee will look at what is unique about this conference.


Share this article with your social network, just click below to share now!


Monday, September 23, 2013

#BPIConf: Thursday’s Talks

Author: Brandy Sargent, Editor, Cell Culture Dish, www.CellCultureDish.com

The last day of talks at BioProcess International featured more great speakers and topics. I chose to spend most of my time in the manufacturing track and I selected the following three talks to highlight.

Michael Goodwin, Associate Director, Development Engineering, Thermo Fisher Scientific, gave a talk titled “Development, Qualification and Application of Single-Use Technologies for Bioprocessing.”

Mr. Goodwin did a great job providing an example of how important it is for vendors and end users to work together to understand new technology and mitigate risk. The example he provided clearly demonstrated that implementation of single-use systems requires a close partnership between both parties. He stated that each step in the disposable supply chain is complicated and each step can impact quality and control.

The example Mr. Goodwin provided was a case study done in conjunction with Amgen. Amgen had seen poor cell growth for two sensitive cell lines where media was being used that had been held in bags.

After analyzing results, one extractable compound stood out as the issue. The leached compound was antioxidant related and had come from one of the stabilizers used in the bag. They later narrowed it down to bDtBPP that had an impact on both viable cell density and cell growth at very low concentrations. For this particular clone impact was seen as low as 30 ppb.

After the compound was identified and it was traced back to a particular stabilizer, the question then became how to solve the problem and how much stabilizer is actually needed. bDtBPP was linked to an antioxidant connected to the polyethylene resin. The typical prevailing thought across most industries where polyethylene is used is that more antioxidants are better, however this may not be the case in biomanufacturing where cell lines can be particularly sensitive.

The solution was to reduce the polyethylene antioxidant and then test to be sure that the reduction did not have a negative impact on the bags. After the reduction, the bags were tested across a number of different performance variables and it was found that there was no difference between the test bag and the control. There was a difference between the films at a molecular level, but it did not impact performance on a macro level. This solution was the result of both companies working closely together to identify then correct a problem. This demonstrated the importance of transparency between supplier and end user.

Mr. Goodwin presented the following conclusions:
  • - Science based understanding of the single-use system is a combined effort between supplier and end user.
  • -Risk of use is reduced with transparency.
  • -Control along the entire supply chain must be clearly defined
  • -Understanding variability in raw materials and processes along the entire supply chain is foundational to ensuring form, fit and function of single-use systems

Rajesh Krishnan, Ph.D., Associate Director, Cell Line and Upstream Process Development, Gilead Sciences, gave a talk titled “Investigation and Reduction of Performance Variability in Single-use Cell Culture Bioreactors.”

In the talk Dr. Krishnan provided a very interesting case study in which Gilead was seeing performance variability in two cell lines in their single-use bioreactors. Both lines were from the same CHO parental host and were producing antibodies. Gilead was working on scale translation of the two monoclonal antibody processes (mAb 1 and mAb2).

Pilot scale for both mAb 1 and mAb2 went well but scale translation to pilot single-use bioreacators resulted in significant decrease in performance. After additional testing they discovered that performance was partially restored in bags conditioned with media and cells prior to production, but a media rinse alone with no cells had no impact.

They initially thought that it must be the single-use bag causing the poor performance. Gilead then began examining potential root causes, but nothing jumped out as a cause. They did notice that there had been a great deal of variability in set up and operation of single-use bags during normal operation. So they decided to set up a modified operational strategy for single-use bioreactors including delay sparging and pH control after media change. In mAb 1 the optimized gassing and agitation strategy restored the single-use bioreactor performance. In mAb 2 they followed the same process but also added an exogenous stabilizer which conferred extra protection to cells.

When they ran the process with the old strategy they got similar failures to the original failure prior to process failures. So even though they initially thought that the bag was the problem and there was bag lot variability found, they were able to restore performance by optimizing their process.

Ying Zhu, Ph.D., Lab Head, Cell Culture Technology, Boehringer Ingelheim, gave a talk titled “Implementing Fully Disposable Upstream Bioprocessing Systems for GMP Clinical Supply.”

Dr. Zhu gave an interesting talk about Boehringer Ingelheim’s experiences using single-use bioreactors. In both their Freemont and Germany facilities they have a cell culture train up to 500 liters, a GMP cell culture train at 100 liters and 500 liters (both single-use bioreactors) and a GMP downstream train. In their China facility they are establishing equivalent systems.

For inoculum and fermentation they follow the process below:
Vial to Shake Flask to WAVE 10L to WAVE 50L to Single-use 100 liters to Single-use 500 liters

Dr. Zhu did present some of the challenges and solutions to their use of single-use bioreactors. In one case they experienced a deformed bag during a GMP run due to back pressure in the exhaust line. This was not an issue in non-GMP because there was no back pressure in the non-GMP exhaust line.

In another similar case the vent filter clogged and caused high pressure in the bag. The bag became increasingly inflated due to increasing gas flow. As a result the bag became deformed around the motor and agitator seal where the bag then became thinned and ruptured causing a small break. The vent filter clogging caused the bag leakage. The resolution was to increase the height of the filter holders and improve the setting with critical process interlocks for shutting down at high pressure.

In another case they found that cell growth and viability in bags was impacted by the age of the bags. They found better growth in bags older than six months olds. Their hypothesis is that volatile substances generated during gamma irradiation diffuse out concentrations after storage.

They also found in one instance that media/solution storage in bags was causing low cell growth. This was identified as the leachable compound bDtBPP. The resolution was to change to an alternative vendor for media solution storage that don’t contain bDtBPP.

Overall in their production and product studies they found that in a disposable upstream process the scale-up growth, cell viability and production are all comparable to using stainless steel systems. In addition, product quality was comparable in all bioreactor systems.

Even though there have been some challenges, they still believe that single-use systems are important to getting a GMP facility commissioned quickly (within 5 months). Also key in multiproduct manufacturing, clinical trial material supply, and having global facilities due to the easy technology transfer across multiple locations.


Share this article with your social network, just click below to share now!


Friday, September 20, 2013

Biorepositories Preview: An Interview with Mark Collins, PhD. Director of Marketing at BioFortis

About the author: Alex Gelman is the Managing Partner of Poplar Partners, an investment firm looking to acquire a company in the biorepository space. Before founding Poplar Partners, I worked at KKR Capstone, the operating arm of private equity firm Kohlberg Kravis and Roberts, and at McKinsey &Co. He has an MBA from the Stanford Graduate School of Business and a BA from Dartmouth College.

6th Annual BioRepository and Sample Management Conference Preview:
An Interview with Mark Collins, PhD. Director of Marketing at BioFortis

Today I had the pleasure of interviewing Dr. Mark Collins, Director of Marketing at BioFortis. As Dr. Collins told us, “I’m passionate about helping researchers use computers to help discover new drugs”. We had a fascinating conversation about just how researchers can do that and the future of bioinformatics for biorepositories.

Us: Why don’t we start off by learning a little bit about BioFortis. What exactly do you guys do?
Dr: Collins: We were a spinout of Johns Hopkins, about 10 years ago. We work in the translational research space – the idea that you can translate research from the lab or pre-clinical studies into new therapies. We are passionate about using informatics to help our customers, mainly pharma companies and biotechs, leverage their data to drive better outcomes for patients. Our software gets used in biobanking, translational and clinical studies as well as biomarker research.

Scientists now have this unprecedented ability to create data, which they’ve never had before, and research has fast become a “big data” problem. They need to think very carefully about how they are going to analyze and manage it, and that’s what we help them do, with informatics software that allows a secure, compliant and holistic view of that data, coupled to decision support tools so they can generate scientific insights.

Us: So from your perspective, what are the biggest challenges facing the biorepository industry today?
Dr. Collins: Well that depends on where you sit.

If you are a biobank, your challenges are about the ethics of biobanking and best practices for sample acquisition, storage and quality.

If you are a researcher the challenge is often access to samples. There is no real “one-stop-shop” for samples, or information about them.

If you are a pharma company, the biggest challenge is managing all the samples and their data that come in from clinical trials because more and more clinical trials are using biomarkers. Data collected about samples in clinical trials is becoming a very valuable resource for research and the highest value sample is one that you know has been collected well, is consented to, and is annotated with clinical and molecular data.

Consent information is especially important, the last thing you want as a pharma company is to submit data on samples to the FDA where patient consent isn’t clear. Our software not only helps with the operational side of tracking samples and data but also provides a harmonized way to collect disparate data and annotate it so you can query and ask questions across trials in a common kind of way.

Us: Conversely, what are the biggest opportunities?
Dr. Collins: The promise of biobanking is that by banking samples, analyzing those samples, and linking it with patient data we will be able to deliver on the promise of personalized medicine. That’s the reason people are excited about biobanks.

In the drive to personalized medicine you have to find a biomarker. And in order to find biomarkers you need to collect samples from patients. After that you need to run different analyses on the samples you collect. But it’s not just analysis on the sample. You must marry the lab analysis with patient clinical information to begin to understand and validate the biomarker.

Us: You work at a data software company. What are the biggest blindspots people have in terms of their data management and existing collection efforts?
Dr. Collins: They often underestimate the effort required for compliance. It’s very important ethically that the sample and patient data are kept secure and that patient has given consent. If you don’t have a compliant and secure system then there are a lot of tedious, manual processes, to ensuring that you meet regulatory guidelines. Additionally, if you don’t have a comprehensive, holistic view, it’s quite difficult to query the data to generate scientific insights. For example, let’s say you wanted to know “How many women in the 50-55 age group with a certain therapy have a certain biomarker”. Well to figure that you out you need to query both the sample, molecular and clinical data sources. If you don’t have a holistic view of all your data then that is very difficult to do.

The other challenge is that we live in a very connected world. Samples and processes don’t just happen inside your four walls anymore. Managing data and tracking samples when you are collaborating with 20 to 30 researchers around the globe is difficult to do with Excel and ad hoc tools that you put together in-house. You need a comprehensive system that’s secure, encourages collaboration and accessibility – a “next generation biobank” if you will!

Us: What are you most excited about at the Biorepository and Sample Management Conference in Boston?
Dr. Collins: We are excited to listen and learn more about people’s challenges in this space.

Us: Thanks Dr. Collins! See you in Boston.


Share this article with your social network, just click below to share now!


Thursday, September 19, 2013

#BPIConf: Wednesday’s Talks

Author: Brandy Sargent, Editor, Cell Culture Dish, www.CellCultureDish.com

Another great day of talks at BioProcess International 2013, the sessions this year have been excellent and I am not the only one who thinks so. Many of the sessions have been completely full. Today I chose the following three talks to highlight.

James Brooks, Ph.D., R&D Manager, BD Biosciences-Advanced Bioprocessing, gave a talk titled “Chemically Defined Media Optimization: Challenges and Solutions.”

Dr. Brooks provided an interesting background as to where we have been with media and where we are at now. In addition he provided two interesting case studies from media optimization projects at BD Biosciences including, steps taken, challenges faced and solutions discovered. I will summarize one of the case studies below.

Dr. Brooks presented his thoughts on a new or re-emerging paradigm of optimizing media for a particular cell line, clone and process to meet the increased need for reduction in process times and to increase speed to market.

In the talk he identified the following reasons for optimizing media:
  • - No universal medium – each cell line/clone has different nutritional requirements and genomic diversity
  • - Removal of components – continued desire to remove animal derived and hydrolysate components
  • - Enhanced culture performance – product yield and product quality
  • - Consistency
Case Study 1
In the first case study presented by Dr. Brooks, the goal was to develop a chemically defined medium and feed strategy to enhance protein production two-fold. The cell line was CHO K1. BD Biosciences then performed the following steps for optimization.
  • - Chemically defined library media screen to select media to identify media with at least 150% or more compared to control production.
  • - Chemically defined library media scale up of selected media, looking at growth production and viability. They selected two to carry forward.
  • - Bioreactor optimization and feed evaluation. With the addition of a feed strategy they were able to increase production six-fold. Based on these results they selected one media to move forward.
  • - Conducted “CHO Flow” Viability Evaluation to check for apoptosis.
  • - Bioreactor performance confirmation with a benchtop bioreactor where they met goal of two-fold increase without feed and when feeds were added achieved six-fold increase in production.
  • - mAb quality analysis – glycosylation profile and confirmation that quality was maintained.
Veronique Chotteau, Ph.D., Principal Investigator, KTH – Royal Institute of Technology, Sweden, gave a talk titled “Perfusion of IgG Producing Chinese Hamster Ovary Cells by Alternating Tangential Flow Filter at Very High Cell Density.”

In the talk Dr. Chotteau discussed a study in which the goal was to develop perfusion in a disposable WAVE bioreactor and a stirred tank bioreactor to evaluation ATF and TFF and test the limits of the system with respect to cell density. The study used CHO cells producing IgG and the cell densities achieved were quite remarkable. These high cell density cultures have many applications in bioprocessing.

The WAVE bioreactor was tested in perfusion mode with both ATF and TFF and very high cell density was achieved in both. Max cell density of 132 x 106 cells/ml was reached using ATF. However, WAVE with ATF was challenged at very high cell densities due to pressure limitation to push highly viscous fluid. Max cell density of 200-230 x 106 cells/ml was reached using TFF. Cell viability was very good in both with viability equal to or greater than 90%, mostly around 95%. There was comparable cell growth between ATF and TFF.

Next the study was conducted in a stirred tank bioreactor with working volume of 1 liter, comparing ATF and TFF. Cell density was stabilized at 20 x 106 cells/ml by daily cell bleeds.

One interesting finding of the study was that there was smaller cell diameter at such high cell density and when distance between cells becomes too small i.e. 2 micrometers, cells shrink. They found that 250 x 106 cells/ml was the limit for 16 micrometer diameter cells.

Conclusions included:
  • - Very high cell density of 100 x 106 cells/ml were stable and maintained
  • - 200 x 106 cells/ml in stirred tank with ATF or in WAVE with TFF were successfully achieved
  • - WAVE with ATF was limited by high viscosity
  • - Applicable limit for cell density in suspension depends on cell diameter and equipment
  • - Not clear on the impact of cell shrinking so perhaps best to avoid shrinking cells
Applications for use of high cell density cultures include:
  • - Seed bioreactor
  • - Production bioreactor
  • - Rapid non-optimized production of protein for exploratory research
  • - Cell expansion for cell banking
Mireli Fino, Vice President, Manufacturing Operations, Protein Sciences Corporation, gave a talk titled “Flublok: Developing the World’s First Recombinant, Highly Purified, Egg-Free Influenza Vaccine

Ms. Fino gave a very interesting talk about the Flublok technology and also the journey to receiving approval. Protein Sciences uses a Baculovirus Expression Vector System (BEVS) to produce Flublok. They have a pilot facility in Meriden, Connecticut, which runs a 500 liter bioreactor and can produce 250,000 doses of Flublok per year. They also have a large scale manufacturing facility in Pearl River, New York where they use 2,000 liter bioreactor that can produce 2-5 million doses/year of Flublok. Their technology enables them to manufacture 50 million doses of pandemic flu vaccine within six months.

BEVS begins with engineering the baculovirus with the gene of interest using a powerful promoter that generates high yields. Insect cells are then grown in fermenters and infected with engineered virus. A serum-free media is used for culture. Then protein is purified and formulated with PBS into influenza vaccine.

During the talk the following advantages were given for use of BEVS:
  • - Safe eukaryotic cells
  • - Fast and flexible manufacturing
  • - High expression level particularly with large proteins
  • - Scale proven up to 20,000 liters


Share this article with your social network, just click below to share now!


Attend the #1 Forum for Scientific Exchange in Antibody Engineering & Therapeutics

Accelerate your antibody research by joining us at IBC's Antibody Engineering & Therapeutics meeting, taking place December 8-12, 2013 in Huntington Beach, CA. Join top antibody researchers from across the globe at the #1 forum for academia and industry to connect for scientific exchange in antibody engineering & therapeutics, immunobiology and next-generation binders.

Our keynotes this year offer the latest advances in systems biology and computation design for protein therapeutics. New session topics this year cover the latest developments in antibodies in cardiometabolic medicine, polyreactive antibodies, knowledge-based design and effects of antibody gene variation and usage on antibody response.

New to this conference? Preview three complimentary past presentations from our 2012 event:
  • - Exploiting light chain diversity to create a fully human bispecific IgG platform technology
    • Nicolas Fischer, Ph.D., Head of Research, Novimmune, Switzerland
  • - Improved antibody therapies for the treatment of Clostridium difficile infection
    • David P. Humphreys, Ph.D., Director, Antibody Biology, UCB Pharma, United Kingdom
  • - Multi-epitopic anti-EGFR antibodies: enhancing surface downregulation by clustering
    • K. Dane Wittrup, Ph.D., Professor, Massachusetts Institute of Technology

Antibody Engineering and Therapeutics will take place December 8-12, 2013, in Huntington Beach, California. If you’d like to know more about the event, download the agenda. If you’d like to join us, as a member of this LinkedIn group, you can save 20% off the starndard rate when you register to join us and mention code XD13172BLOGJP.*

Have any questions? Feel free to reach out to Jennifer Pereira at jpereira@iirusa.com.


Share this article with your social network, just click below to share now!


Wednesday, September 18, 2013

Developments in understanding polyspecificity

Leading up to the Antibody Engineering & Therapeutics Event, podcast, we'll have interviews with a few of the speakers from the event.  Today we feature a portion our interview with James Crowe, Jr., M.D., Director, Vanderbilt Vaccine Center, Vanderbilt University Medical Center.

Today, he answers the question:
What are recent developments in understanding polyspecificity?

His answer:
Well, the recent development of repertoire sequencing technologies has allowed us to start getting a sense of how big antibody repertoires are in human beings. There is a theoretical number of something like ten to the eleventh antibodies that can be generated by stitching variable gene segments together in recombination and adding injunctional diversity. So, these are the normal mechanisms by which you get your naïve repertoire. But, when people look at the repertoires of sequencing they seem to be smaller than predicted.

This leaves us with a conceptual problem – how, with a relatively small repertoire, can you recognize all of the antigens that you have to deal with? In fact, in something like the flu there are many closely related variant antigens. Or for HIV there are variants occurring in each person. For Hepatitis C there is extreme variability with point mutations going on in the virus antigens and it’s not entirely clear that the antibody repertoires are large enough to commit individual clones in the repertoire in each antigen that’s encountered. So, just mathematically you have a problem and it appears that what the B-cell repertoire system is doing is reusing B-cell receptors – which are also later the secreted antibody. So, they are reusing specificities for more than one antigen and when an antibody binds multiple antigens, it is termed “polyspecificity,” generally.

So, we’re certain that this does occur and now that people are able to generate human monoclonal antibodies in great numbers, it’s increasingly apparent that many antiviral antibodies also bind to host proteins or some antiviral antibodies by more than one viral protein or at least by more than one variant. So, the question is how do they do this? We had this concept that monoclonal antibodies are very specific. So, how do they bind to two or more antigens? I think recent developments using these higher throughput sequencing and computational techniques have given us some insight into what’s going on.

In the past we thought that most of the interaction between an antibody and an antigen was driven by the residues that are touching on both sides of that protein/protein interaction. So, very close within several Ångström relationships of the surface of an antibody combining site with the epitope on the virus. But more recently we’ve realized that it’s not just the contact residues that drive the interaction, but rather the contact residues are typically on the tips of hypervariable loops called the “complementarity determining regions”. These loops arrayed on a scaffold, which is the framework of the heavy chain or light chain. So, the framework loops and residues are holding up the variable loops and when you think about it, it becomes fairly intuitive to realize that if you altered the scaffold at the base of the antibody structure dramatically, then the orientation or placement of the hypervariable loops could change very dramatically on the interface. This appears to be one of the very important physical principals in antibodies that underlie polyspecificity or the molecular basis for polyspecificity.

It looks like the framework residues that allow some flexibility confer to those antibodies the ability to move variable loops around in space and, therefore, the antibody combining site is not fixed and rigid, but rather it is moving around and dynamic and it’s able to achieve various conformations. This may be how a single antibody can bind to more than one thing because that antibody exists in sort of a fluid state of multiple conformations. While it is exploring those various states, it may have the capacity to bind antigen A and when it’s in an unbalanced state when it’s in a different conformation – sampling a different part of its conformation space – it may be able to bind antigen B. So, that’s kind of the most existing recent development. It looks like the framework residues, rather than contact residues, may be driving flexibility in the frameworks and thus confer to those antibodies a capability to bind very diverse antigens.

I think this is a new notion that most of us have thought about. The contact residues (are) driving it, but instead the framework residues may be very important. That’s the principal breakthrough in the last year or so, I think, in understanding polyspecificity.

Dr. Crowe will be presenting Deep Sequencing the Human Antibody Response to Viral Infections and Human Germline Antibody Gene Segments Encode Polyspecific Antibodies. For more information on these sessions and the rest of the program, download the agenda. The Antibody Engineering & Therapeutics Event will take place December 8-12, 2013 in Huntington Beach, California. If you'd like to join us, as a reader of this blog,when you register to join us and mention priority code XD13172BLOGJP to save 20% off the standard rate.


Share this article with your social network, just click below to share now!


#BPIConf 2013: Producing High Quality, Low Cost Biotherapeutics in the Century of Biology

One of our first keynote of the BioProcess International Conference was James Thomas, Ph.D., Vice President, Process and Product Development and WA Site Head, Amgen, Inc.

He began by sharing that bioprocess professionals set out to do is develop a molecule that can target disease effectively. In the protein modality space there will be a lot of varieties. We know that protein therapeutics are complicated. The antibodies have attributes and it’s important to figure out what factors into the therapeutics.

Process development improvement is working towards enhancing expression level of the clones.  It depends on the facilities and what they can hold. If in a less fixed environment, there is space to expand. In the future, we’ll see more mass maid product volume. We need approaches to be able to exploit the ability to generate more mass. The system biology tools in the age of the computer will allow more sophistication. It’s likely that those experts in the modeling space will contribute to this.   As it stands, the healthcare industry can’t afford Pharma’s current methods of innovation.  How can this industry work to improve that?  Facilities must be designed to be flexible enough to manufacture a lot of molecules effectively. Raw materials and plants will be commodities. This is one way to drive down the cost of medication. A plant should be as flexible as possible. You could design the plant to produce as many therapeutics as needed.

Abbie Celniker, Ph.D., CEO, Eleven Biotherapeutic also joined us to present Transforming the Development of Biotechnology Drugs for the 21st Century: A CEO’s Perspective from a Small Biotech Company  During her presentation, she looked at some of the life of a small biotech company. Small biotech companies are dependent on funding and fundraising is constantly going on. There were virtually no IPOs in biotech between 2008-2011. This is changing because new investing in biotech in 2013 has grown rapidly. It could open up investing tremendously – she uses the 1990s as an ideal point in time. Most biotechs in this time frame knew that M&A was a way out so they had to develop products that were desired by Pharma. Less productive companies were driven out of the market. Companies had to have great intellectual property – it’s the formulation, manufacturing, and configuration. A biotech company is faced with Value Creation and charged with with the creation of new medicines have to matter a lot to their potential purchasers. Biotech companies know they have science and technology to develop the new transformative medications. These companies are positioned to fill this need for Pharma. From the
beginning of drug creation, a company has to think of all stake holders – patients, payers, regulators, investors and more.

We finished our evening in the Poster and Exhibit Hall for the Oktoberfest celebration - networking and meeting colleagues.  We hope to see you tonight in the exhibit hall at 5:45PM again for more great networking with the 2nd Annual Mixed-Mode Mixer!


Share this article with your social network, just click below to share now!


#BPIConf – Tuesday’s Talks

Author: Brandy Sargent, Editor, Cell Culture Dish, www.CellCultureDish.com.

Today was a great day full of interesting talks at the Bioprocess International Conference. It was difficult to choose only a few, but I have highlighted three below.

Thomas Ryll, Ph.D., Senior Director, Cell Culture Development, Biogen Idec, gave a talk titled “Impact of Single-Use Bioreactors on Upstream Processing.”

Dr. Ryll did an excellent job highlighting both the advantages of single-use bioreactors and also opportunities for improvement.

In the talk single-use bioreactor advantages were identified as:
  • - Requiring reduced infrastructure
  • - Lower capital investment
  • - Ease of use and labor savings
  • - Enables scaling down production culture volume
  • - Important to goals of increased globalization
Dr. Ryll also discussed how Biogen Idec implemented single-use bioreactors at their facility in Research Triangle Park after validating single-use bioreactors on a number of points and across different size bioreactors. They converted warehouse space to “gray space” where they could use single-use bioreactors at different sizes to scale up process development in a non-clinical manufacturing setting, which eased technology transfer of the perfected process to clinical manufacturing. Another interesting comparison was shared by Dr. Ryll that looked at the cost for single-use bioreactors vs. stainless steel tank bioreactors.

It was found that single-use bioreactors:
  • - Had higher consumables cost
  • - The same raw material costs
  • - Significantly lower utilities
  • - Lower labor cost
  • - Overall single-use bioreactors costs were found to be comparable
Dr. Ryll then pointed out the following areas as opportunities for improvement in single-use bioreactors:
  • - Find balance between mass transfer and bubble size
  • - Reduce lot to lot variability in bag lots
  • - Address toxic leachable from polymer films
Richard Ferraro, Business Leader WAVE Products Group, GE Healthcare, gave a technology workshop titled “Efficient and Intelligent Process Control for Animal Cell Culture.”

In the talk Mr. Ferraro discussed the high cost of drug development and possible platform and toolbox approaches to address these issues. One of the platforms covered was an intensified process approach, for example perfusion technology.

When compared with fed batch processes intensified processes have advantages, but they can also create challenges

The talk identified the following intensified process challenges:
  • - More training required for operation because you are working with more than just the bioreactor
  • - Ancillary equipment adds complexities – including connections and monitoring
  • - Increased logistics
  • - Lots more data
GE Healthcare has taken what they have learned about challenges in intensified processes and has worked to address these challenges by implementing new solutions. They have taken their comprehensive experience in downstream systems and have applied some of the same concepts to upstream. These ideas for improvements were important in the development of the new ReadyToProcess WAVE 25 Bioreactor, particularly in the area of process control.

New Tools for Process Control:
  • - Application specific PID parameters
  • - Method editor function for scheduled changes of process conditions
  • - Accessories to facilitate process intensification; pumps, load cells & auto-calibration function
  • - Intuitive user interface, help screens, set up support
  • - Remote monitoring – WAVE25 App

GE Healthcare launched the new ReadyToProcess WAVE 25 Bioreactor at their booth Tuesday evening, please see picture above. The new WAVE has many new and improved features and it is interesting to see the advancements after so many years of WAVE bioreactors being in use.

Sandra Poole, Senior Vice President Biologics Operations, Genzyme, gave a Keynote Presentation titled “A Systems Approach to Managing Biomanufacturing Complexity – Genzyme’s Allston Plant Case Study.”

Sandra Poole gave an excellent talk describing the challenges that Genzyme faced during their viral contamination at their Allston facility and how they have implemented a systems approach to recover and again function at capacity. I appreciated Ms. Poole’s candor and sharing a real experience as a learning tool for the industry. In the talk she discussed the importance of taking a systems approach and really looking at the ripple effect that a change in one area can have on several other areas of manufacturing. These effects often aren’t expected, but it doesn’t mean that they can’t be anticipated with careful examination. She also stressed the importance of looking at system feedback loops and determining what the limiting factor is.

Ms. Poole discusses the manufacturing competency curve and how after competency improves additional tasks are regularly added, which as a result dilutes skills. In the Genzyme example, she describes how the Allston facility was originally designed just to manufacture Cerezyme, after their capability increased, manufacturing of Fabrazyme was added and then a third product, Myozyme, was also added. These additional tasks created challenges in manufacturing and did not lend itself to a systems approach.

Since the contamination, a systems approach has been implemented and the facility is running at capacity, under consent decree at 100% batch certification. They are also implementing a work plan plus improvement program targeting their entire manufacturing system.

To close the talk, Ms. Poole described the following key learnings:
  • - Understanding how a change to manufacturing can impact complexity, capability and performance
  • - Understanding balance between a plant’s complexity and the plant’s capacity to manage that complexity
  • - Choosing the right tools and metrics to judge performance and health of the system are key
  • - Need for new skills and education in systems thinking


Share this article with your social network, just click below to share now!


Tuesday, September 17, 2013

#BPIConf 2013: A Look at Manufacturing Strategy

Conference chair Ran Zheng, Executive Director, Plant Manager, Amgen Inc. kicked off the morning with sharing that in 2012, there were 38 US FDA approvals, 50% were biologics.  We must pursue facility changes and fundamental capabilities to enable the production. She encouraged companies to embrace the culture of innovation and anticipate changes in market.

In our next presentation, Chris M. Brodeur, Associate Director, Commercial Operations, BioMarin
presented Implementing and Improving Disposable Systems in BioMarin’s Manufacturing Environment to Meet Growing Capacity.  When creating and updating their facilities, they have design for flexibility, minimize filling impact to approved products, allow for future process introductions and use disposables to enhance flexibility and efficiently. Brodeur states that they’ve added disposables wherever possible in their production plant. He encourages attendees that when they consider building a facility, they should start with the end in mind. He also cautioned that when creating a facility with disposables, much of facilities are no longer hidden in the walls, but all out in the open.

Our next presentation was Maximizing Efficiencies and Capacity Utilization in a Multi-Product Clinical and Commercial Facility from John Kenneally, Director, Drug Substance Manufacturing, Genentech, Inc.  He starts out with question: Flexibility versus Throughput: Do you really have to choose between the two? Both can be accomplished with speed but a company  has to consider their priorities.  For example: is it more important to get 3% more out of process or get there six months earlier?

Our next speaker Naveen Pathak, Associate Director, Technical Strategy, Manufacturing Science & Technology, Genzyme, a Sanofi Company presented A Case Study in Creation of a Strategic Technical Lifecycle Plan (TLCP) for Continuous Improvement of a Legacy Biological Process.  It is important to enable infrastructure to chart the TLCP Course. In this example, a governance model was created to dictate the flow of communications between teams. You need to have good technical solutions. This is the pathway to achieving a successful plan. All solutions should be strategic, sound and right sized. For every project, it was evaluated through a grid that looked at regulator/quality/compliance, Process understanding, product supply and business efficiency. After you have programs and have grouped them, you now have to create the plan. Resources and timeline knowledge are needed to create the model. It should look forward to the future state and will help you figure out the model you can use to get this TLCP completed. Both projects and programs can overlap. There should be touch points that can be checked to make sure that all programs work across processes.  Naveen also participated in a podcast we recorded for BioProcess International Magazine.  Listen to it and other presentations from this year's speakers here.

In the presentation from Jolene Ignowski, Staff Development Scientist, Bayer Healthcare,emphasized that in the right situation, perfusion must be done continuously. They must move the product from the bioreactor. Efficiencies in the process to develop this product that must be considered: process design, equipment, people. To increase process design, they eliminated unite operations and took advantage of new technologies. They use disposables for quicker turnaround time. They also use interchangeable disposables to save time that comes with disposables. Perfusion is not an efficient manufacturing method, but unstable products need them. By improving manufacturing processes and standardizing the flow and staff to make this happen difficult process happen.

In our final session of the morning, Sourav Kundu, Ph.D. Director, Biopharmaceutical Development, Teva Biopharmaceutical USA looked at the Biosimilars Development Pathway and began with a basic question - Why biosimilars? Worldwide the population is growing and aging. There are more chronic diseases along with an increase in the number of patients. The emerging markets are also expanding and looking into innovative products. Populations are demanding good quality healthcare. On the supply side – companies and governments – are mandated to control costs. For manufacturers, they’re seeing an increasing cost to manufacture these drugs. The annual growth rate for biologics has been 20% and isn’t showing signs of slowing. Almost all biologics are being worked on, looking for biosimilars or biobetters. Recent approvals have taken place. Biosimilars present a front-loaded effort. The amount of work upfront is massive – create a reference product database, reacting the molecule, in-depth characterization of assay development then follows a path of clinical studies eventually followed by a launch.

A critical decision point for any molecule is deciding if the product a biosimilar. Other considerations include: are there any product differences? Decision points are if the product is safe and effective. Maintaining biosimilarity is a challenge. It must be done and maintained through the process development, scale up and manufacturing. Change of scale can complicate biosimilarity.

Stay tuned for updates this afternoon focusing more on manufacturing strategy and highlights from our keynote speakers including James Thomas of Amgen, Sandra Poole of Genzyme and Dr. Abbie Celniker of Eleven Biotherapeutics.


Share this article with your social network, just click below to share now!


What is the value of the IBC Cell Therapy Bioprocessing Event?

The Cell Therapy Bioprocsesing event taking place October 21-22, 2013 in Bethesda, Maryland.  For more information on the event, download the agenda.  If you'd like to join us, as a reader of this blog, you're can save 20% off the standard rate when you register to join us and mention code B13188JP20!

Leading up to the Cell Therapy BioProcessing event, we've interviewed several of the key stakeholders who helped shape this year's event.  We'll start by looking at the recent interview we conduced with Event Chair Lee Buckler, the Founder and Managing Director of the Cell Therapy Group.

Today, Buckler answers the question:
What is unique about the value of this Event when compared to other cell therapy conferences in the calendar?
Lee: So, I think when you look at other cell therapy conferences, cell therapy itself is a wide topic representing a number of different modalities and technologies. You’ve got the autologous vs. allogeneic, which themselves represent vastly different bioprocessing kind of platforms and technologies. You’ve got the different kinds of indications and you’ve got the different kinds of clinical markets. Most cell therapy conferences try to cover a wide rubric of clinical vs. technical vs. regulatory, reimbursement, commercial. So, the bioprocessing elements of any of those conferences gets fairly watered down or is fairly discreet, you know, restricted to one or two sessions. So, for us to be able to really get in a room together for a couple of days – people whose sole focus or who are very focused on the issues related to bioprocessing – is a unique opportunity to really focus on the topic that needs a fair amount of attention and has been identified as one of those that needs a fair amount of focus in order to make cell therapies commercially viable.

You can download Lee's full podcast here. Next week, Lee will look at some of the key challenges facing the cell therapy industry.


Share this article with your social network, just click below to share now!


Monday, September 16, 2013

#BPIConf 2013: Antibody-Drug Conjugates: Current Best Practices an Considerations for Processes and Facilities

Today, the 10th Annual BioProcess International event began in Boston.  Here are a few of the highlights form the Antibody-Drug Conjugates track.

During Dr. Leo Letendre of Pfizer's presentation Impact of 2011 FDA Process Validation Guidance on Process Development Strategy, he looked at the fact that Process Validation began as the collection and evaluation of data from the process design stage to the throughout the production, which establishes scientific evidence that a process is capable of consistently delivering quality products.  Over the years it has evolved to a life cycle with these steps, to name a few: 1) Process design 2) Process performance qualification 3) Continued process verification.  One key change with the new definition is that after a validation has occurred with an antibody drug conjugate, a company has to continue to monitor as we develop more information. This allows a company to potentially optimize more in the long run.  One key change to this is that companies in the process are now looking for optimization. Life cycle approach is an acknowledgement that we will continue to learn about our processes and continue to apply new data in a rational way.

In the presentation from Deborah Meshulam, MS, Immunogen, titled Assembling Teams to Manufacture ADCs: Realities vs Expectations, she looked at how to develop a team that can fully oversee antibody drug conjugates.  As an overview, benefits of antibody drug conjugates, there is a selective binding to tumors and longer half life. Cytotoxic agents are released into the cancer cell after it has attached. The type of linker on the ADC will tell what kind of tumor it should be used for. Antibody, linker, toxin: conjugated into substance then a drug product.

There are numerous groups involved in the manufacturing of antibody drug conjugates. How can a company keep all party members involved and engaged in the full production cycle?  Work to improve governance to ensure execution of decisions. Roles are clarified, responsibilities and the right parties are accountable. Each team should have goals and feel empowered to reach them. Across the board, teams should be prepared, participate and be proactive. Good teams feel empowered to manage themselves tactically Efficient and clear communicators; better faster cross functional decisions, consistent and timely data for better decision making are all benefits to this process. 

Project charters give each team an overview of the various products. They allow the teams to look at what decisions they should be making and what information can be factored in. Each antibody has a different team.

Tomorrow, we kick off the main conference.  Registration begins at 7:00AM, followed by a full day of presentations focused on Cell Culture, Rediscovery and purification, Formulation and Delivery, Analytical and Quality and Manufacturing Strategy.  Our Poster and Exhibit hall will open at 3:15 followed by our kickoff keynotes from 4:15-5:25.  We'll conclude our day with Oktoberfest, our evening networking reception in the exhibit hall from 6:00PM-7:15PM.


Share this article with your social network, just click below to share now!


Friday, September 13, 2013

What posters can you see in the BioProcess International Exhibit Hall?

Over the past few months, we've been profiling some of the posters that will be featured next week in the BioProcess International Exhibit Hall. Would you like to hear more about any of these posters or discuss them with their presenters? Join us in Boston! As a reader of this blog, when you register to join us and mention priority code  BLOG13JP, you'll save 20% off the standard rate!

Here is a comprehensive list of the posters we have previewed here at the Future of Biopharma Blog:




Share this article with your social network, just click below to share now!


Thursday, September 12, 2013

Single-Use Stirred Tank Bioreactors: Enabling Flexible Biomanufacturing

Author: Brandy Sargent, Editor, Cell Culture Dish, www.CellCultureDish.com.  Brandy will be joining us this year at BioProcess International as a guest blogger on September 16-19, 2013 in Boston, MA.  If you'd like to join us at BioProcess International, as a reader of this blog, register to join us and mention code BLOG13JP to save 20% off the standard rate!

Background
Stainless Steel Stirred Tank Bioreactors have been used for several years in biomanufacturing and have proven their effectiveness across several biomanufacturing platforms. Of all production platforms, CHO cells are the most prevalent in stirred tank reactors, but they have also been successfully used with other platforms including insect cells, yeast, E. coli, plant cells, Pseudomonas, hybridoma, vero and others. While stirred tank bioreactors are traditionally associated with suspension culture, the use of microcarriers and associated technologies have allowed adherent cell lines to be effectively cultured in stirred tanks and stirred tanks have also had some success in stem cell manufacturing.

The traditional model for manufacturing biopharmaceuticals is to build a manufacturing facility focused on fixed stainless steel stirred bioreactors usually in sizes ranging between 100 – 25,000 liters. These facilities require high initial capital investment and large overhead. Initial capital investment includes cost of setting up the bioreactors, detailed facility design including piping and infrastructure for media delivery, plus water and steam necessary for Clean in Place (CIP), Steam in Place (SIP) and Water for Injection (WFI) requirements. The challenge with this traditional design is that these facilities have large manufacturing capacity, but little flexibility. They usually focus on manufacturing a handful of biopharmaceuticals requiring large volumes. However if demand changes, cell culture improvements result in higher titer, or any other alterations to volume requirements, excess capacity can quickly become a problem.

Due to some of the challenges experienced with traditional fixed biomanufacturing facilities there has been a desire to create more flexible spaces. Improvements in cell culture and downstream processing have made manufacturing more productive resulting in higher titers and product quality. In some cases reducing the overall number of cell culture liters required to meet a biopharmaceutical’s demand. As a result, facilities that were used to operating at capacity could find they now have costly capacity excess to fill.

Single-use Stirred Bioreactors Designed for Flexibility
The development and subsequent improvements to single-use stirred bioreactors have offered a more flexible and cost effective alternative to their stainless steel fixed counterparts. Single-use stirred bioreactors offer several advantages that makes them an attractive option in designing flexible facilities:

Reduced Start Up Time and Capital Investment
Single-use stirred bioreactors offer a reduction in installation time. They don’t require extensive piping systems or infrastructure to support media delivery, clean in place, or steam in place requirements. The overall capital investment is also reduced due to the simpler facility design, smaller facility footprint and lower cost for equipment. In addition, many single-use stirred bioreactors offer “ready out of the box,” efficiency, which provides lower set up hurdles prior to use.

Process Flexibility
Single-use stirred bioreactors offer optimal process flexibility, particularly when combined with other single-use technologies. The ability to move these bioreactors around offers tremendous flexibility in facility layout and process flow. This allows the bioreactors to move to other areas within one facility based on need or even to other manufacturing locations. Because these are mobile and disposable, they do not need to be dedicated to one product type. For example, they could be used to manufacture one product for a clinical trial material campaign in one area, then moved to another area and used for commercial product manufacturing. Each different product may utilize different cell lines, media or other manufacturing requirements, but single-use bioreactors can be repurposed and utilized in different scenarios.

Single-use stirred bioreactors also support different production modes including fed batch, perfusion or continuous processing. Just like their stainless steel counterparts, they have been proven across many different biomanufacturing platforms and have consistently performed comparably with fixed stainless steel bioreactors.

Manufacturing Efficiency
One of the biggest advantages for single-use stirred bioreactors is the reduction or elimination of cleaning, sterilization and validation requirements. This reduces product changeover time and thus promotes increased manufacturing efficiency and improves scheduling of product manufacturing. Disposable systems also reduce the risk of product cross contamination, thus eliminating costly manufacturing shut downs. As mentioned in process flexibility, because these bioreactors are not fixed or dedicated, they can be moved around to create a manufacturing configuration that is most efficient.

Single-use stirred bioreactors coupled with new automation systems, offer a truly all in one solution. With the ability to have advanced control over processes, including data management, temperature, RPM, pH, DO, gas and liquid flow rates, weight and pressure, these systems offer a number of parameters to be set automatically and can even aid in troubleshooting. They also make evaluating data convenient with export options.

Flexible Scaling to Meet Product Demand
Single-use stirred bioreactors offer many choices as to size and scale (<1L - 2,000L), so that lot sizes can be matched with the current product demand. If product demand changes, then the scale of the single-use bioreactor can also be adjusted to keep pace, which greatly reduces costly capacity excess or drug shortages. This also enables the move toward smaller market or orphan drug products that have smaller product demand.

Cost Savings
Single-use stirred bioreactors can offer a reduction in cost of goods by reducing overall utility costs, reducing or eliminating manufacturing steps (cleaning and validation, for example), reduction in labor requirements, and as mentioned earlier, scalability to more closely match demand.


Single-use Bioreactors Offer Solutions to Manufacturing Challenges
There are several specific manufacturing challenges to which single-use bioreactors and even entire single-use systems are well suited. Below I have listed some examples.

More Products, Smaller Volumes
There has been increasing need to produce more products at smaller volumes. This has occurred for several reasons. First, product yield increases has reduced the need for 10,000 – 25,000 liter batches and allowed for the opportunity to manufacture multiple products in the same facility. Traditionally turnaround time and cleaning, sterilization, and validation would have made it challenging to produce many products at the same facility. However with single-use systems, cleaning, sterilization and validation are greatly reduced or eliminated, thus enabling multiple products produced in the same facility at smaller lot sizes.

Biosimilars
Another industry area where single-use bioreactors can be very effective is in biosimilar manufacturing. Biosimilars will have new technologies available that the original biopharmaceutical manufacturer did not, including single-use systems. Speed to market plus reduced cost will be key drivers in the success of biosimilars. In terms of speed, single-use bioreactors clearly have the advantage by offering reduced installation time, no piping infrastructure required, and easy set up with “ready out of the box” type operations. In addition they offer more attractive economics with lower start up costs and lower water for injection use requirements.

Clinical Trial Material
There has also been a great deal of interest in the ability to manufacture clinical trial material with limited investment. When drugs enter clinical trials it is unclear at that time how the drugs will do or if they will eventually earn approval. The attrition rate for investigational drugs is quite high, so logically companies do not want to make significant capital investments in manufacturing these products prior to approval. The flexible manufacturing offered by single-use bioreactors is perfectly suited to address these needs. Clinical trial manufacturing campaigns can be initiated to manufacture the necessary product and then can be switched to manufacture a different drug or could be taken offline until future need arises.

Globalization
With product globalization, there is a desire to manufacture medicines closer to the populations that they will be supplied to. This also provides some security with respect to facilities going offline. If one facility goes offline due to contamination, natural disaster, etc., the entire product supply chain will not be disrupted. With flexible manufacturing options, more single-use bioreactors could come online to address any temporary shortages. Ideally model single-use system facilities could be designed with the goal of replicating these facilities in multiple countries or regions.


Employing Single-use Systems Facility Wide
While single-use stirred bioreactors are a primary enabling strategy to achieving the goal of flexible manufacturing, there are a variety of single-use technologies, which when combined, create entire single-use systems that can support the entire biomanufacturing process from upstream to downstream. Single use products like disposable mixers and filters can aid upstream preparation of cell culture media. In manufacturing, single-use bioreactors and single-use mixers offer many advantages in terms of reduced cleaning, sterilization and validation up to the 2,000 liter scale. Finally downstream can take advantage of single-use filtration, centrifugation and chromatography options, many of which are still in development and improving rapidly.

A recent study titled “Fast Track API Manufacturing from Shake Flask to Production Scale Using a 1000-L Single-Use Facility” by B. Minnow, et al., and published in Chemie Ingenieur Technik, describes how Rentschler Biotechnologie set up a “fast and flexible multipurpose manufacturing facility in the 1,000L scale”. The completely single-use facility was awarded Facility of the Year in 2012 by the International Society for Pharmaceutical Engineering. The facility was able to reduce manufacturing costs, timelines and experienced increased flexibility for clinical manufacturing. Scale up was from shake flasks to 1,000L production scale.

In the study, a CHO DG44 DHFR line producing a monoclonal antibody that was optimized for high titer was thawed and initially expanded in shake flasks. Further cell expansion was performed in 50L and 200L XDR single-use stirred bioreactors (Xcellerex). The final production reactor was a 1000L XDR single-use stirred bioreactor (Xcellerex). A 28 day (vial thaw to harvest) fed batch process was used in the study.

The scale-up was performed based on calculating the specific volumetric power input, which allowed a direct transfer from small culture volumes to the production scale. A seeding volume of 700L was used.

The study demonstrated that fast track protein production could be achieved in less than 12 weeks from cell bank to final bulk drug product. The authors of the study stated the following in the conclusion. “With these experiments a fast track concept for the manufacturing of active pharmaceutical ingredients in single-use bioreactors combined with a robust and reliable process was shown. This led to a short manufacturing timeline from clone to product. A well-known and also simple scale-up strategy was applied and the calculated specific volumetric power input was used as key scale-up parameter. Additionally, results regarding product quality and titer as well as reproducibility in key process parameters are highly comparable. Finally, it was concluded that the applied scale-up method is feasible for single-use bioreactors in production scale as well as for non single-use bioreactors in laboratory scale.”

Conclusion
With the many pressures facing biomanufacturing today including cost effective manufacturing, stringent regulatory requirements and effective capacity management coupled with the many different product demands, it is not surprising that companies are looking for more flexible manufacturing options. In particular, single-use bioreactors and platforms enable fast-track manufacturing strategies. Single-use stirred bioreactors and single-use system based facilities are key in enabling these processes and moving manufacturing forward to meet new biopharmaceutical manufacturing challenges.


Share this article with your social network, just click below to share now!


BioProcess International Poster Preview: Small Scale Protein Purification for Rapid Optimization of Biotherapeutic Protein Production

Leading up to the BioProcess International Event, we'll be highlighting many of the posters you'll be able to find in the hall at the event taking place September 16-19, 2013 in Boston, MA. If you'd like to join us at the event, as a reader of this blog, when you register to join us and mention code BLOG13JP, you'll save 20% off the standard rate!

Poster: Small Scale Protein Purification for Rapid Optimization of Biotherapeutic Protein Production

Presenter: Andrew Barry, PerkinElmer

About: Optimization of expression and purification conditions presents a key challenge towards the development of processes to efficiently produce biotherapeutic proteins. The ability to screen matrices of conditions critical to protein identity, titer, structure, and function requires systems for rapid purification and analysis. Small scale protein purification enables increased sample throughput and reduced purification time, resulting in faster, more efficient optimization for biotherapeutic process development. These advances enable the use and scaling of novel resins through functional dynamic inding capacity testing for ion exchange and affinity chromatography. Here, we will discuss automated purification using three different modalities, each with benefits of loading mass, speed of purification, and flexibility of resin choices and sample throughput.


Share this article with your social network, just click below to share now!


Only a few discounted passes left for IIR’s 6th Annual Biorepositories and Sample Management Conference. How can you save 25%? Details here!

The 6th Annual Biorepositories and Sample Management Conference is right around the corner, coming up September 25-27, 2013 at the Seaport Hotel in Boston. The excitement for this event has blown us away and exceeded all of our expectations! In fact, we are actually almost out of discounted passes BUT we wanted to let you know this Thursday and Friday we are giving out the last few left.

For the next two days, we're offering you - the Biorepositories Community - a chance to attend IIR’s 6th Annual Biorepositories and Sample Management Conference in Boston for 25% off the standard registration rate!

How can you take advantage of this offer? Email klentini@iirusa.com with your full contact information and the code XP1898SUPER2* to receive the discount. This limited-time offer expires Friday, September 13, 2013 at 11:59PM ET.

To see what all the hype is about, download the agenda if you'd like to know more about the program. If you have any questions, email Jennifer Pereira.

*Rules and Regulations Apply:
• This offer is only available for biobank and biorepositories executives, and is subject to IIR approval
• Registrants will receive 50% off the standard registration rate
• This offer only applies to passes purchased on Wednesday, August 28 and Thursday August 29
*This is a non-transferrable offer and cannot be retroactively applied nor combined with other offers, discounts or promotions. Offer only valid between September 12-13, 2013.


Share this article with your social network, just click below to share now!


Wednesday, September 11, 2013

Technology and it's impact on understanding the biology of the antibodies

Leading up to the Antibody Engineering & Therapeutics Event, podcast, we'll have interviews with a few of the speakers from the event.  Today we feature a portion our interview with James Crowe, Jr., M.D., Director, Vanderbilt Vaccine Center, Vanderbilt University Medical Center.

Today, he answers the question:
Are there examples of where technology is already having an impact on understanding the biology of the antibodies?

Dr. Crowe's answer:
Yes. I think we are starting to see publications in the immunology field and some of the highest impact journals that are bringing high throughput sequencing of antibody replicons into the literature. So, Andrew Fire’s group at Stanford and Scott Boyd have put out papers in a way looking at repertoires – sort of like a micro alert ray or using patterns of repertoires as biomarkers for responses in cancer or autoimmunity in various conditions. They’ve been starting to describe methods for looking at entire repertoires as correlating with clinical phenotypes. Our own group has used these technologies to couple repertoire sequencing with individual monoclonal antibody technologies derived from antibodies from hybridomas or similar technologies. So, we know the function of influenza or dengue or pox virus specific monoclonal antibodies and we’ve been able to find siblings of those monoclonal antibodies in the whole repertoires and we started publishing papers in this area. 
Others have been the same. The NIH Research Group has correlated development in human antibody phylogenies of sequences with HIV specific neutralization fina types and development of repertoires. In fact, they are starting to use those phylogenies to suggest that we could design immunogens that interact with various stages of antibody development in a predictive phylogeny. So, we’re not only seeing papers that describe repertoires and link to function, but these studies are already leading to rational vaccine design strategies that play up on the information we’ve gained from the sequencing. So, it is a very exciting time. A lot of papers are going to come out in the next couple of years and it’s just very exciting to learn more about antibody repertoires.

Dr. Crowe will be presenting Deep Sequencing the Human Antibody Response to Viral Infections and Human Germline Antibody Gene Segments Encode Polyspecific Antibodies. For more information on these sessions and the rest of the program, download the agenda. The Antibody Engineering & Therapeutics Event will take place December 8-12, 2013 in Huntington Beach, California. If you'd like to join us, as a reader of this blog,when you register to join us and mention priority code XD13172BLOGJP to save 20% off the standard rate.


Share this article with your social network, just click below to share now!


BioProcess International Poster Preview: Use of a Simple Visual Tool to Monitor Commercial-Scale Purification Process Performance on the Manufacturing Floor

Leading up to the BioProcess International Event, we'll be highlighting many of the posters you'll be able to find in the hall at the event taking place September 16-19, 2013 in Boston, MA. If you'd like to join us at the event, as a reader of this blog, when you register to join us and mention code BLOG13JP, you'll save 20% off the standard rate!

Poster: Use of a Simple Visual Tool to Monitor Commercial-Scale Purification Process Performance on the Manufacturing Floor

Presenter: Lakshmi Pathange, Bayer HealthCare

About: Tools such as standard work and daily weekly operational reports are used in the manufacturing to manage floor operations. These tools play an important role in facilitating communication between floor operators and management. They are used by operators to track, monitor and improve floor operations. Though these tools are excellent in analyzing operational efficiency, they rarely contain details to monitor purification process performance. In order to enhance floor operator involvement in process performance issues, a simple visual tool was developed. Chromatographic data such as UV peak height, conductivity and pressure were used to generate a process monitoring tool. The data entry and compilation are performed by operators on the floor after each process step. The data is continuously reviewed by operators and management to identify any emerging issues that could impact process performance. Case studies will be presented to demonstrate its use by operators and management as simple and effective tool to communicate and evaluate commercial-scale purification process performance.


Share this article with your social network, just click below to share now!