BioProcess Theater: Analytical Services

Cheryl Scott

May 12, 2015

13 Min Read

DigitalVision-332004-244x300.jpgBioProcess International launched about the same time as a major FDA regulatory announcement that has radically altered biopharmaceutical development: The quality by design (QbD) initiative is an important part of the agency’s 21st-century good manufacturing practice (GMP) approach, which is changing how regulators review product applications and thus how companies must approach them (1). It has placed increasing pressure on analytical laboratories, whose work is more important to the success of biotherapeutic products than ever before.

Backed by harmonized tripartate guidelines from the International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH), the guiding principles of QbD include risk management, science-based policies and standards, and integrated quality systems. International cooperation brings Europe, North America, and parts of Asia together in agreement on the value of this approach to public health protection.

QbD particularly emphasizes the value of scientific analysis in characterizing products and processes, identifying critical quality and process attributes (CQAs, CPAs) and critical process parameters (CPPs), setting specifications, and defining a “design space” within which unit operations can function with guaranteed results. Analytical strategies and methods form the vital foundation of quality assurance and control.

Bioprocess development involves a large amount of analytical work:

  • cell line engineering and characterization, optimization of culture media, and leachables and extractables studies for single-use technologies in upstream/production

  • studies of membrane and resin lifetimes, in-process and buffer hold times, protein load limits for liquid chromatography columns, pH and conductivity specifications, extractables and leachables, virus removal and/or inactivation, and impurity removal for downstream processing of drug substances

  • preformulation and product characterization, immunogenicity and stability testing, and formulation optimization at the drug-product stage.

Bio/Analysis: Industry consultant Nadine Ritter points out that according to FDA guidance, analytical and bioanalytical methods are not the same thing (2). The latter are not used for elucidating quality parameters (e.g., identity, purity) of a biologic; they’re for determining the quantity of a drug (or the presence of induced antibodies) in biological samples. So their applications in pharmacology, bioavailability, bioequivalence, pharmacokinetic, and toxicology studies are a very important but separate part of product development.

Analytical Strategy

Advancing analytical technologies have made QbD possible. Large companies led the way in developing strategies for implementing the new paradigm (7–9). The A-MAb Case Study on applying QbD principles in development of a monoclonal antibody (MAb) resulted from a two-year effort by a consortium of biotechnology companies collectively known as the CMC-Biotech Working Group (3). The consortium brought together Abbott Laboratories, Amgen, Genentech, GlaxoSmithKline, Eli Lilly and Company, MedImmune, and Pfizer to identify ways that QbD could help in MAb development. To ensure free public access and further promote the industry-wide discussions that led to its creation, they provided their results to two prominent industry organizations: the California Separation Science Society (CASSS) and the International Society of Pharmaceutical Engineers (ISPE).

“The analytical program for a given biotherapeutic has a life cycle analogous to that of a manufacturing process used to prepare material for clinical and commercial use” (4). That life cycle is divided into two distinct phases (5): a developmental phase (initial method development through clinical studies) and a validated phase (late-stage activities for commercial settings). New products that have been in development during the QbD period may have an advantage because they weren’t subject to major strategic changes partway through this life cycle (6).

The FDA’s newest process validation guidance both clarified and complicated the issue (7). It changed the language of discussion. For example, the traditional “4Q” qualification approach famously appears nowhere in the guidance (7, 8). Industry consultant Peter Watler explains the new paradigm this way (7): “The focus should be on what a study says, not about what it is called. And the FDA is clear on this. It has caused some in industry to divert validation efforts away from a scientific understanding to strict adherence of terminology and protocol. Fortunately the new guidance does away with that nonsense, so industry can put resources into science and understanding rather than protocol and definitions.”

Analytical Services: Clearly, it helps to have experts at your disposal when it comes to implementing QbD analytical strategies. And the smaller the biopharmaceutical development company, the less likely it is to have such experts on staff. Enter the service providers. Contract research organizations (CROs) assist biomanufacturers in clinical and preclinical testing of their products. Traditionally, they have gotten involved at the investigational new drug (IND) stage of innovator products. Lately, biosimilars have given their business a boost (9). Other contract testing organizations provide viral-safety, product-characterization, and immunogenicity services. Most contract development and manufacturing organizations (CDMOs) provide laboratory services as well: validation, scale-up/scale-down, product and process characterization, for example. Through varied work with many clients, many of these companies are amassing a great amount of expertise in QbD, risk management, quality systems, and analytical technologies and trends.

Recent Trends

Biomolecular analysis falls into three categories: chemical or biochemical, immunological, and biological assays (10). Biochemical and chemical assays account for 66–75% of those performed in product characterization, and they can take 400–800 work-hours to develop and validate. Bioassays require more time and make up about 15% of the total number of assays performed. Overall assay development and validation can cost around US$1.5 million — $150,000 for three immunological assays, $1 million for maybe a dozen chemical and biochemical assays, and nearly $500,000 for three bioassays.

If you’ve noticed an increasing number of BPI articles that focus on analytical methods and approaches over the years, you’re not mistaken. Although we’ve emphasized the importance of analytical work in biopharmaceutical development from the beginning (11), the industry we cover has become increasingly aware of product and process analysis in recent years. As new analytical methods are developed and validated, companies are finding new ways to solve old problems with them. And as new problems present themselves, the methods are there for companies to turn to for help.

In the past few years, I’ve delved into these topics myself through a series called BPI Lab (12–22). Throughout 2013 and 2014, I highlighted methods such as liquid chromatography with mass-spectrometry (LC–MS), polymerase chain reaction, electrophoresis, flow cytometry, calorimetry, and so on. Meanwhile, BPI authors were contributing more in-depth reports describing applications of such technologies in biopharmaceutical development. Topics that have been of particular interest include

  • Sensors and other methods of process monitoring and control

  • Process modeling, design of experiments (DoE), and statistics

  • Biosimilars and product comparability

  • Bioassays and immunogenicity

  • Leachables and extractables testing for disposable components and systems

  • Process optimization, validation, and scale-down studies

  • Product structure and characterization.

A number of educational program tracks at the 2015 BIO International Convention
might appeal to those interested in analytical methods and services: Next-Generation
Biotherapeutics, Oncology, Personalized Medicine and Diagnostics, Primary Care and
Chronic Diseases, Infectious Disease, Regulatory Science, and Translational Research.
Some sessions stand out in particular:
Next Generation Biotherapeutics
“The Promise of Gene Editing” (9:00–10:00 am, 16 June 2015, Room 107AB)
“Nucleic Acid and Cell Therapy: Insights, Successes, and Challenges” (10:15–11:45 am,
16 June 2015, Room 107AB)
“Harnessing the Power of Antibodies” (2:00–3:15 pm, 16 June 2015, Room 107AB)
“Nanomedicine: The Next Big Thing?” (3:30–4:45 pm, 16 June 2015, Room 107AB)
Oncology
“Leveraging the Immune System to Cure Disease: Emerging Approaches”
(10:30–11:45 am, 18 June 2015, Room 108A)
“Biotech–Pharma Collaborations in Oncology Clinical Trials” (1:00–2:00 pm, 18 June
2015, Room 108A)
Personalized Medicine and Diagnostics
“The Future Is Now in Precision Medicine: Developing Targeted Oncology
Therapeutics” (10:15–11:30 am, 17 June 2015, Room 105AB)
“Transforming Diagnostics With Innovative Technologies: Potential and Pitfalls”
(2:00–3:15 pm, 17 June 2015, Room 105AB)
“FDA Takes Charge: The Changing Regulatory Landscape for Molecular Diagnostics”
(3:30–4:45 pm, 17 June 2015, Room 105AB)
Primary Care/Chronic Diseases
“The Human Microbiome: Translating Research into Therapeutics” (9:00–10:00 am,
16 June 2015, Room 108A)
“Preventing Death from Cardiovascular Disease — Is It All About Diabetes and
Obesity?” (10:15–11:30 am, 16 June 2015, Room 108A)
“The Revitalization of Neuroscience R&D” (2:00–3:15 pm, 16 June 2015, Room 108A)
Infectious Disease
“Next-Gen Antibodies: Unlocking a Treasure Trove of Antigen Targets” (2:00–3:15 pm,
17 June 2015, Room 113A)
Regulatory Science
“Enabling Future Medical Advances: A Review of Global Initiatives to Increase Clinical
Data Transparency” (3:00–4:15 pm, 15 June 2015, Room 104AB)
“Putting Patients in the Center: Advancing the Science of Patient Preference
Assessment” (2:00–3:15 pm, 16 June 2015, Room 106AB)
“Global Regulatory Trends for Biosimilars and Biotherapeutics” (10:15–11:30 am,
17 June 2015, Room 106AB)

New Products, New Challenges: Meanwhile, biotechnology itself continues to advance and bring forth novel therapeutic approaches: e.g., cell, gene, and tissue therapies; antibody– drug and other conjugates; and novel vaccines and immunotherapies. The age of biosimilars is upon us, as well. All these “nonantibody” and novel product modalities pose analytical questions that are different from those familiar to developers of MAb and other traditional protein products.

For example, ADCs combine antibodies with highly potent small-molecule drugs. The process of conjugation is not yet fully controllable. So analytical laboratories must be able to characterize the results and help manufacturing groups refine their product streams to get the best possible drug substance ready for formulation.

Biosimilars must be demonstrated to replicate the structure, quality, and effects of the innovator products they purport to replace. If analytical proof shows it to exceed the safety, quality, or efficacy of the original, it might be classed as a “biobetter” instead.

The value of stem cells is in their ability to differentiate into a range of cell types that can provide patients with a therapeutic benefit. So that differentiation must be characterized, as are the means of inducing it.

Those are just some of the new questions being asked an answered in modern biotechnology laboratories.

Analytical Service Companies

Here are some companies that offer analytical services and will be part of the
BioProcess Zone at the 2015 BIO International Convention. They may be
contract testing laboratories, R&D and contract research organizations, or
discovery services companies. See the “Cell Line and Produce Development”
chapter for contract development and manufacturing organizations that perform
validation and other testing as part of their process development services.
Aragen Bioscience
Aseptic Technologies
Boston Mountain Biotech
Charles River Laboratories International
DV Biologics
Masy BioServices
Nitto Denko Technical Corporation
Phage Consultants
Selexis
Toxikon
Thomas A. Little Consulting
VGXI

Here are some companies that offer analytical services and will be part of the
Contract Services Zone at the 2015 BIO International Convention. They include
contract research and testing laboratories, or other specific analytical service
providers. See the “Formulation, Fill and Finish” chapter for contract organizations
that focus on preformulation and drug product analysis.
AMRI
BRI Biopharmaceutical Research Inc.
HiQScreen
Institute of Industrial Organic Chemistry
Branch Pszczyna (Poland)
Lab Support (a division of On
Assignment)
MultiLing
Neuroservice

Here are some companies that offer analytical services and will be part of the
Discovery Services Zone at the 2015 BIO International Convention:
Abzena
BioXcel Corporation
BRTI Life Sciences
CASIS
Eurofins Pharma Discovery Services
NDRI

Modern Approaches

Quality cannot be left up to testing or inspecting a finished product. Instead, biopharmaceutical quality, safety, and effectiveness must be designed and built into a biomanufacturing process — and that can happen only with the right information at hand. A typical bioprocess involves a complicated matrix of input and output parameters, which can be interlinked with or independent of one another. Sources of variability include changes in raw materials, operators, facilities, and equipment, and it is difficult to understand all their possible permutations and effects on the quality of a final drug product.

Statistical DoE methods are helping process engineers understand the effects of possible multidimensional combinations and interactions of various parameters on final drug quality (23, 24). Application of a DoE strategy provides scientific understanding of the effects of multiple process parameters and raw material attributes on product CQAs and leads to establishment of a design space and manufacturing control strategy.

A company’s quality control unit is part of its quality system, one of six manufacturing systems in a biotech facility that are subject to inspection by the FDA: quality, facilities and equipment, materials, production, packaging and labeling, and laboratory controls. If a manufacturing system is in compliance, then all products of that system should be compliant as well. Documentation continues to play a vital role in making the case — especially for laboratory personnel who must be familiar with both GMPs and good laboratory practice (GLP), and in some cases, even good clinical practice (GCP) as well.

References

1 Pharmaceutical cGMPs for the 21st Century — A Risk-Based Approach: Final Report. US Food and Drug Administration: Rockville, MD, September 2004.

2 Ritter NM. Distinctions Between Analytical and Bioanalytical Test Methods. BioProcess Int. 9(3) 2011: 80.

3 Koslowsky S, et al. QbD for Biologics Learning from the Product Development and Realization (A-MAb) Case Study and the FDA OBP Pilot Program. BioProcess Int. 10(8) 2012: 18–29.

4 Apostol I, Kelner DN. Managing the Analytical Lifecycle for Biotechnology Products: A Journey from Method Development to Validation, Part Two. BioProcess Int. 6(9) 2008: 12–19.

5 Apostol I, Kelner DN. Managing the Analytical Lifecycle for Biotechnology Products: A Journey from Method Development to Validation, Part One. BioProcess Int. 6(8) 2008: 12–19.

6 Capen R, et al. Establishing Potency Specifications for Antigen Vaccines: Clinical Validation of Statistically Derived Release and Stability Specifications. BioProcess Int. 5(5) 2007: 30–42.

7 Scott C. Quality By Design and the New Process Validation Guidance. BioProcess Int. 9(5) 2011: 14–21.

8 Winter W. Analytical Instrument Qualification: Standardization on the 4Q Model. BioProcess Int. 4(9) 2006: 46–50.

9 Galbraith D. Biosimilars Awaken CROs. BioProcess Int. 12(6) 2014: S24–S29.

10 Lundblad RL, Price NC. Protein Concentration Determination: The Achilles Heel of GMP? BioProcess Int. 2(1) 2004: 38–47.

11 Scott C. A Decade of Characterization: Products, Processes, and Quality Control. BioProcess Int. 10(6) 2012: S58–S61.

12 Scott C. Biophysical Analysis of Living Cells: Not Just for Basic Research Anymore. BioProcess Int. 11(1) 2013: 24–27.

13 Scott C. Antibodies, Bioassays, and Cells: The ABCs of Immunochemistry. BioProcess Int. 11(2) 2013: 24–28

14 Scott C. A Powerful Pairing: Using Mass Spectrometry with Analytical Chromatography. BioProcess Int. 11(3) 2013: 28–33.

15 Scott C. Amplifying the Possibilities: Using the Polymerase Chain Reaction in Biodevelopment. BioProcess Int. 11(4) 2013: 24–29.

16 Scott C. “Transformation By Infection”: Genetic Engineering for Biodevelopment. BioProcess Int. 11(5) 2013: 20–23.

17 Scott C. Enlightening Results: Spectroscopic Analysis of Cells, Molecules, Solutions, and More. BioProcess Int. 11(8) 2013: 24–27.

18 Scott C. Cellular Communications: How Cultures and Tissues React to Their Environments. BioProcess Int. 11(9) 2013: 40–43

19 Scott C. Robots in the Laboratory: Modern Pipetting Enables High-Throughput Investigations. BioProcess Int. 11(10) 2013: 22–25.

20 Scott C. Analysis By Size and Charge: SDS-PAGE, Capillary, and Isoelectric Focusing Techniques Anchor the Biopharmaceutical Laboratory. BioProcess Int. 12(1) 2014: 26–29, 40.

21 Scott C. North, South, East, and West: Blotting Techniques Expand the Utility of Electrophoresis. BioProcess Int. 12(2) 2014: 16–18.

22 Scott C. The Heat of the Moment: Analyzing Reactions in Action. BioProcess Int. 12(4) 2014: 24–26.

23 Scott C. Analytical Methods for Biologics. BioProcess Int. 5(2) 2007: S35–S38.

24 Montgomery SA. Operations and Quality Systems: Building in Success. BioProcess Int. 4(3) 2006: S58–S71.

Further Reading

Dong D, Kang J. Comparing SDS-PAGE and CE-SDS for Antibody Purity Analysis. BioProcess Int. 12(4) 2014: 28–31.

Girard FC. Process Optimization of Biosimilars Production Using NMR Profiling. BioProcess Int. 11(1) 2013: 52–56. Hulse J, Cox C. In Vitro Functional Testing Methods for Monoclonal Antibody Biosimilars. BioProcess Int. 11(6) 2013: S24– S27, S41.

Mire-Sluis A, et al. Reference Standards for Therapeutic Proteins: Current Regulatory and Scientific Best Practices and Remaining Needs, Part 1. BioProcess Int. 12(3) 2014.

Mire-Sluis A, et al. Reference Standards for Therapeutic Proteins: Current Regulatory and Scientific Best Practices and Remaining Needs, Part 2. BioProcess Int. 12(4) 2014.

Stephens E. Site-Specific Characterization of Glycosylation on Protein Drugs. BioProcess Int. 12(6) 2014: 46–53.

Toward Industry Standardization of Extractables Testing for Single-Use Systems: A Collective BPSA Perspective. BioProcess Int. 13(3) 2015: S4–S9.

Zhang Y, et al. Improved Fluorescent Labeling Efficiency of N-Linked, High- Mannose Oligosaccharides: Using APTS for Analysis of Glycoproteins. BioProcess Int. 13(3) 2015: 26–31. c

Cheryl Scott is cofounder and has been senior technical editor of BioProcess International since the first issue.

You May Also Like