You are on page 1of 12

pubs.acs.

org/ac Perspective

Selection of Analytical Technology and Development of Analytical


Procedures Using the Analytical Target Profile
Phil Borman,* Cristiana Campa, Ghislain Delpierre, Elliot Hook, Patrick Jackson, Wayne Kelley,
Michel Protz, and Olivier Vandeputte

Cite This: https://doi.org/10.1021/acs.analchem.1c03854 Read Online

ACCESS
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Metrics & More Article Recommendations

ABSTRACT: A structured approach to method development can help to ensure an


analytical procedure is robust across the lifecycle of its use. The analytical target profile
Downloaded via 191.101.136.143 on December 21, 2021 at 03:20:40 (UTC).

(ATP), which describes the required quality of the reportable value to be produced by the
analytical procedure, enables the analytical scientist to select the best analytical technology on
which to develop their procedure(s). Once the technology has been identified, screening of
potentially fit for purpose analytical procedures should take place. Analytical procedures that
have been demonstrated to meet the ATP should be evaluated against business drivers (e.g.,
operational constraints) to determine the most suitable analytical procedure. Three case
studies are covered from across small molecules, vaccines, and biotherapeutics. The case
studies cover different aspects of the analytical procedure selection process, such as the use of
platform method development processes and procedures, the development of multiattribute
analytical procedures, and the use of analytical technologies to provide product
characterization knowledge in order to define or redefine the ATP. Challenges associated
with method selection are discussed such as where existing pharmacopoeial monographs link acceptance criteria to specific types of
analytical technology.

■ INTRODUCTION
Predefining what constitutes success for the measurement of a
To enable the identification of optimal procedure con-
ditions, a broad initial screen of all plausible conditions is
recommended. This screen should be structured such that the
quality attribute1 is a key input for a sound method
testing identifies potential sets of procedure conditions that are
development strategy. In the pharmaceutical industry,
likely to meet the ATP, which can subsequently be ranked, and
predefined acceptance criteria associated with the analytical the most preferable ones checked to see whether there is a
measurement can be summarized using the analytical target likely robust region which will result in acceptable operation of
profile (ATP).2−6 the procedure.
The ATP has been defined by USP ⟨1220⟩ as “a prospective The output of a successful method development strategy
description of the desired performance of an analytical should lead to one or more sets of method conditions which
procedure that is used to measure a quality attribute, and it satisfies the ATP, ensures optimal method performance, and
defines the required quality of the reportable value produced identifies a region around the selected method conditions
by the procedure”.6 The ATP is informed by product/process which is likely to be robust over the analytical procedure
requirements (e.g., specification acceptance criterion for a lifecycle. Other potential method conditions may also be
quality attribute) and is not linked to any specific analytical available for analysts to fall back on if unexpected failures arise
technology. The ATP should enable the analyst to select the for the chosen procedure.
best analytical technology for developing their procedure. This It is advisable to monitor the availability of new technologies
will be driven by prior knowledge and/or experimental during product development and across the lifecycle, to ensure
verification relating to the performance of available analytical the most up-to-date technology is being used. Following the
technologies. Where more than one technology has been
demonstrated to meet the ATP, a review of business Received: September 6, 2021
requirements (e.g., throughput, automation, availability) Accepted: December 1, 2021
should be performed to aid selection. Once the technology is
selected, procedure specific performance indicators can be
defined which will support method development and ensure
the performance criteria in the ATP are achieved.
© XXXX The Authors. Published by
American Chemical Society https://doi.org/10.1021/acs.analchem.1c03854
A Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry


pubs.acs.org/ac Perspective

above approach ensures a continued focus on product and METHOD DEVELOPMENT: GENERAL CONCEPTS
process needs and enables robust bridging between new and The application of quality by design (QbD) principles to
existing analytical procedures, in case of change. method development provides a systematic approach for

■ SCREENING AND SELECTION OF


FIT-FOR-PURPOSE ANALYTICAL TECHNOLOGIES
building in quality from the onset of development by
establishing method requirements (via the ATP) against
identified product quality attributes. The analytical QbD
The process leading to selection of a fit-for-purpose analytical framework2 allows decision making using a science- and risk-
based approach. These risk-based method development steps
procedure may start as soon as the performance expectations
involve (I) preliminary risk assessment, (II) risk management
(ATP) and operating environment are defined. The first step if/where appropriate for high-risk analytical procedures. This
of method development aims at identifying all available could involve improving method performance understanding
technologies that can potentially be used to meet the ATP via modeling (e.g., multivariate experimentation) and (III) risk
requirements. This preliminary step is crucial to minimize the confirmation, implementing method robustness and rugged-
risk of using routine or general-use analytical procedures that ness. The ATP enables the analyst to select the most
are unable to meet the performance criteria. Technology appropriate analytical outputs to monitor during development.
assessment also allows for the evaluation of any existing The definition of surrogate performance criteria (also known
platform analytical procedures or new technologies. Special as analytical procedure attributes (APAs) linked to the ATP
attention should also be given to orthogonal technologies acceptance criteria) underpins the method development
which can be used to analyze the same attribute but based on process. The APAs defined for a given method are technology
different principles of measurement. Such technologies are specific properties (for example, attributes for chromatography
useful as back-up options in case of failure during development measurements may include peak symmetry factor and
of the selected technology. Orthogonal tests can also provide resolution). The expert analyst should identify the most
additional product and analytical knowledge, thereby increas- appropriate APAs used to study the performance during
development and to meet the ATP requirements. APAs are
ing the confidence in the selected method.
also used to identify critical analytical procedure parameters
After this first step, a roadmap leading to method selection
(CAPPs) through modeling studies, the output of which can
can be described as follows: be used to define a method operable design region4,7 (MODR)
1. Scoring of the analytical technologies against both for the analytical procedure. CAPPs are analytical procedure
required performance (ATP) and operational con- parameters whose variability have an impact on APAs and
straints (business drivers). consequently on the measured result.
Three examples of technology screening and method
2. Experimental testing/screening of potentially fit-for- development are covered below from across small molecules,
purpose methods. vaccines, and biotherapeutics. The above outlined concepts
will be covered for each example.
3. Final scoring and ranking leading to method(s) selection In the first example, HPLC specific method development
and start of formal development. The approach and screening principles are described using APAs linked to
described for method screening and selection is the ATP acceptance criteria. The second example demon-
illustrated in Figure 1. strates the method screening and selection process with an
evaluation of innovative approaches to address complex
biological CQAs during vaccine development. Example three
discusses how CQA knowledge gained during development
can drive revisions to the ATP and an evolving method
selection process in preparation for a commercial control
strategy.

■ EXAMPLE 1−SMALL MOLECULES: METHOD


DEVELOPMENT APPROACHES/PLATFORMS
Development of analytical procedures for the analysis of small
molecule pharmaceutical products or ingredients, following
quality by design principles, is well established.7−9
The structured analytical QbD method development
approach driven by the requirements of the ATP can be
applied to any analytical technology and has been applied to a
range of analytical techniques, such as HPLC,9 Karl Fischer,10
and colorimetry.11 Further technique specific method develop-
ment strategies have been developed and applied. For example,
in the case of developing an analytical procedure for the
determination of related organic impurities of an active
pharmaceutical ingredient, the ATP will direct the develop-
ment to an analytical technique (such as liquid chromatog-
raphy) that is capable of separating and quantifying the
Figure 1. Method screening and selection approach. impurities which are critical quality attributes (CQAs) that
B https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry pubs.acs.org/ac Perspective

may impact the quality of the active pharmaceutical ingredient


(API). Once the technique is selected, a number of technique
specific method attributes together with their performance
criteria can be used to ensure the analytical procedure is likely
to meet the ATP, meet ICH Q2(R1)12 validation require-
ments, and to reliably be used across the entire method
lifecycle. The technique specific method attributes and their
performance criteria will be common to the majority of small
molecule HPLC impurity methods and so can be usually used
for all HPLC analytical procedures.
With the APAs defined, experimental scouting of analytical
procedure parameters can begin, typically starting for HPLC
with mobile phase pH, organic modifier, and column.9,13 See
Figure 2 for the number of mobile and stationary phases that

Figure 2. Mobile and stationary phases used in HPLC scouting.

are typically used. These are from a predetermined subset of


solvents/column, selected based on prior knowledge of
performance against the APAs, affording a degree of control
and commonality when this process is applied across multiple Figure 3. HPLC method development strategy.
assets.
This screening process is supported by data evaluation and
in-silico modeling to aid the initial method selection, before vaccines under development and available on the market.16 A
undergoing iterative risk assessment and optimization to meet refinement of the recombinant DNA technology consists in
the requirements of the ATP and APAs. As this process will be delivering the gene of interest directly into the host and in
common to the majority of small molecule HPLC methods, it using the host cell machinery to produce the corresponding
can be defined into a structured, technique specific method protein/antigen which, hence, induce the desired immune
development strategy (see Figure 3 for a simplified version of response.16−18 Different virus vectors can be used to achieve
what has previously been published by Vogt and Kord8) to this strategy18−21 and include replication-defective adenovi-
ensure consistent application of the QbD principles during rus.22−24
analytical procedure development. These adenovirus vectors are defective for viral replication,
Use of risk assessment is key throughout this strategy to thereby ensuring the patient’s safety but are fully capable of
understand how the method is performing against the method hijacking the host cell machinery to produce the protein/
attributes and the ATP. Borman et al.14 demonstrates the antigen of interest that will induce the desired immune
application of risk assessment and experimental design tools to response.22−24 As for any other vaccines, several attributes are
evaluate the final method robustness and identify method part of the comprehensive understanding of the product. More
controls as part of the method control strategy.


importantly, they are cornerstones of the control strategy since
they are key indicators of the efficacy, stability, and consistency
EXAMPLE 2−VACCINES: ANALYSIS OF of the vaccine as well as of the process robustness. In this
ADENOVIRUS VECTORS example, three specific quality attributes will be discussed: two
Many physicochemical methods have been designed using the related to the mode of action of replication-defective
QbD approach,15 but very few examples highlight how adenovirus and one that is common to all vaccines.
biological methods can benefit from this iterative and To be efficacious, the replication-defective adenovirus vector
structured mindset. Using adenovirus vectors developed for must enter the host cells where it can trigger the expression of
vaccination, this example will describe how QbD principles can the gene of interest that will induce the desired immune
improve and foster the selection of fit-for-purpose methods, response.24 Therefore, two critical quality attributes of the
the identification of multiattributes methods, and the deploy- replication-defective adenovirus vectors that must be moni-
ment of innovative analytical methods addressing biological tored are the “infectivity” (i.e., capability to enter and to deliver
attributes. the gene of interest into the host cells) and the “transgene
Virus-based vaccines and antigens produced using recombi- expression” (i.e., control the expression of the gene of interest
nant DNA technology represent an important part of the in the recipient cells).24−27 Both attributes are key to
C https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry pubs.acs.org/ac Perspective

Table 1. List of the Potential Fit-for-Purpose Technologies to Address the CQAs “Infectivity”, “Expression of the Gene of
Interest”, and “Identity”

demonstrate the functionality and the stability of the were discussed to address the “transgene expression” attribute,
nonreplicative adenovirus vector capsid components, the including nucleic-acid based technologies (NAT), antibody-
efficient packaging of the full-length genomic DNA, and the based methods, chromatography, electrophoresis, and spectro-
integrity of virus particles among other attributes.27−29 Being metric methods. Typically, cell-based methods (like CCID50,
produced through amplification in specific cell lines, any TCID50, and IFU but also flow cytometry) were considered
change in process parameters can impact the functionality of for the infectivity attribute. Even though these methods meet
the replicative-deficient adenovirus vector since misfolded many analytical requirements (see Table 1) and could be used
proteins will result in nonrecognition by cell receptors, as such to address the three CQAs, the technology evaluation
misassembled capsid will result in unstable particles and/or exercise with all SMEs led to a comparison of the “pros” and
premature degradation, and incomplete genomic DNA will “cons” of all technologies.
result in decreased gene expression among other biological As shown in Table 1, some methods were identified as being
misfunctions.30,31 As for any other vaccines, the identity of the release-ready (such as electrophoresis, chromatographic
gene of interest must be verified to ensure that the right methods, antibody-based methods, and NAT), accepted by
antigen is used in the vaccine.32 the regulatory agencies (such as antibody-based methods and
After definition of the ATP for the identified CQAs, different NAT), ready for transfer to quality control (such as
technologies were then evaluated with analytical subject matter electrophoresis, chromatographic methods, antibody-based
experts (SME). “Obvious” methods were first considered methods, and NAT), or specific and sensitive (e.g.,
before looking at multiattribute and innovative methods. The spectrometric methods and NAT). Drawbacks were also
identity of the antigen can typically be addressed by antibody- identified, such as the need for control materials (e.g., peptides
based methods (e.g., ELISA or Western blot). Several methods for mass spectrometry or characterized protein standards for
D https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry pubs.acs.org/ac Perspective

Table 2. Summary of the Method Scoring for the Three Attributes (Infectivity, Identity, and Transgene Expression)a

a
For each ATP requirement, a score is given as follows: “1”, does not meet; “4”, partially meets; “7”, meets; and “10”, exceeds the requirements.

electrophoresis, chromatography, and antibody-based meth- performance (ATP) versus operational (business drivers)
ods), the need for additional data (such as total protein constraints. It is important to keep in mind that this scoring
content for antibody-based methods), or the need for specific is made to help in selecting what methods will go into further
reagents (i.e., monoclonal antibodies). A major outcome of the development and is not, as explained later, a “score-based”
method screening exercise was that all these methods were decision tool. Using this way of working also helps in capturing
identified as being able to address at least two attributes. More the knowledge and rationales behind a go/no go choice.
particularly, the cell-based assays as well as the NAT methods Following the rationale shown in Table 2, three groups of
were identified as being able to address the three CQAs in a methods (i.e., electrophoresis/chromatography, spectrometry,
single assay. and antibody-based methods) out of five were not further
A second important step in the method selection is to ensure considered because their performance was not able to meet the
that they meet the requirements defined in the ATP and the ATP requirements with regards to LOD/LOQ and combined
associated business drivers. Table 2 shows how the method uncertainty.2 NAT-based and cellular-based assays were ranked
screening was implemented to address the “infectivity” CQA with the highest scores regarding performance and business
and how the performance of the different methods can be requirements. Besides, these groups of methods were also
ranked according to the expectations of the ATP. Although the identified as being able to target the three attributes at the
“infectivity” CQA is taken because of its criticality for vaccine same time, which is an additional significant advantage for
efficacy, it is important to note that a similar screening was downstream quality control. NAT-based assays were slightly
performed for the other CQAs considered in this case study. ahead in terms of industrialization because they were already
The scoring/ranking is performed with all analytical SMEs to implemented in the quality control department. However,
foster brainstorming and innovation. The most promising since NAT methods target nucleic acids and not the protein/
methods that will go into further practical assessment and antigen, the decision was made to move forward with the cell-
development were identified by comparing the capability of the based assay even though flow cytometry was a new technology
analytical methods to meet the ATP requirements defined for for quality control. It is important to note that the main part of
all CQAs. Summing all the scores allows to provide a global the cellular-based assay method relies on cell-based technology
score for each method but also to discriminate between already implemented in quality control. As shown in Table 3,
E https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry pubs.acs.org/ac Perspective

Table 3. Comparison between Flow Cytometry and NAT

Figure 4. All attributes in one single assay: (A) identity, (B) potency, and (C) transgene expression.

Table 4. Critical Analytical Procedure Parameters Evaluated

the need for training and budget to deploy the new technology during the method screening helped alleviate these risks and
in the final lab (i.e., quality control (QC)) were identified as allowed QC to anticipate the deployment of a new technology
the main risks associated to the use of a new cell-based assay. in a regulated environment, facilitating the deployment of the
Nevertheless, the priming and inclusion of the QC teams new assay in a short period of time in all involved laboratories.
F https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry pubs.acs.org/ac Perspective

Notably, the screening process described here does not overall testing strategy. Over the course of development,
necessarily lead to a clear-cut decision about the method to be information from analytical structure−function studies as well
further developed. Instead, the screening is a tool which fosters as the clinical experience can lead to a reassessment of a CQA
innovation, triggers the selection of multiattribute methods, and the identification of related attributes, which will in-turn
causing analytical experts and project leaders to leverage, in a drive refinements of the initial ATP. This example for a mAb
simple and visual way, the pros and cons of each considered will demonstrate how QbD principles can help to focus this
method and to make the best decision in a given context. evolving testing strategy.
The next step in the development of the method consisted Therapeutic mAbs are complex proteins (immunoglobulins)
of defining critical method parameters to establish an produced from a single clonal cell that have a high degree of
optimized MODR. In a nutshell, the method consisted of specificity for an antigen related to a disease (e.g., a tumor cell,
incubating permissive eukaryotic cells with drug substance, viruses, or bacteria) or an adverse immune response (e.g.,
drug product, or reconstituted vaccine samples to let the virus asthma). Therapeutic mAbs work, in general, by binding to
grow. After 24 h of incubation, the cells were permeabilized antigens on the membrane or surface of a pathogen, such as a
and a specific monoclonal antibody (labeled with a cancer cell or virus, interfering with the disease pathway. This
fluorochrome) was used to detect and quantify the protein is achieved by inhibiting absorption of a virus or replication of
expressed by the adenovirus-vectored transgene. As shown in a cancer cell. Additional mechanisms can include binding to
Figure 4, the three attributes (identity, potency, transgene effector cells of the immune system, which will affect a
expression) were addressed in a single experiment. Indeed, the complement response or signal specific cells such as natural
presence of antibody-responsive cells and the specificity of the killer cells or macrophages which mediate the destruction of
antibody ensure that the identity of the product is verified. The the pathogen.
content of infective/functional virus present in the sample is Antibodies are categorized by five different isotypes.
determined by the number of antibody-positive cells (each Therapeutic mAbs are an immunoglobulin G isotope (IgG)
infective/functional virus will infect one cell that will be where the structure consists of four polypeptides composed of
detected by flow cytometry). Finally, the transgene expression two light chains and two heavy chains linked by disulfide
level is obtained by comparing the median fluorescence bridges and arranged in a Y shape configuration. The IgG
intensity to the one obtained with a reference lot, thereby isotype is further defined by subtypes that differ by their
ensuring the consistency between batches. disulfide bond arrangement. IgG1, IgG2, and IgG4 are
The robustness of the cell-based flow cytometry multi-CQA commonly employed as therapeutic mAbs with the IgG4
assay was evaluated through a QbD approach where the design typically used for immunotherapies.36 Therapeutic mAbs can
of experiments (DoE) with different CAPPs was analyzed. As be described as having two characteristic domains: the variable
shown in Table 4, these parameters are grouped into three and the constant. The variable domain contains the
categories: parameters related to the cell culture (such as complementary determining regions (CDR) where the amino
subpassages, percentage of confluence, and dilution of acid sequence varies significantly from antibody to antibody
inoculum), the composition of the growth medium (such as and is what makes the antibody highly specific for a particular
fetal bovine serum (FBS), glutamine, and lots of reagents), and antigen. Therapeutic mAbs are further characterized by the
the method of infection (such as operator and method of antigen binding fragment (Fab), involved in target binding,
dilution). As shown in Table 4, many of these parameters had and the crystallizable fragment (Fc) in the constant region.
no impact on the performance of the method for determining The latter can play a role in the mechanism of action and
the potency and the transgene expression. pharmacokinetics (PK) by binding to Fc receptors on innate
For the further development of the method, the DoE immune cells and activating the immune system. These
approach allowed effort to be focused on the most critical structural characteristics and some of the relevant physi-
parameters of the assay, such as passage before culture, batch ochemical and biological quality attributes to be considered in
of critical raw materials (e.g., glutamine, trypsin, and FBS), and a full product assessment are depicted in Figure 5.
operator impact.


The CQA assessment process follows the ICH Q11
guideline and involves the identification and prioritization of
EXAMPLE 3−BIOTHERAPEUTICS: AN EVOLVING
the potential risks to patient safety or the product quality that
ATP FOR MONOCLONAL ANTIBODY
could impact the efficacy, consistent manufacture, or stability
AGGREGATION of the product. The impact of each product quality attribute is
Therapeutic monoclonal antibodies (mAbs) have become a
significant part of the pharmaceutical landscape, providing
powerful treatments for patients spanning a wide range of
diseases in the fields of oncology, cardiology, hematology,
neurological and autoimmune system related disorders, and
infectious disease.33−35 As with other therapeutic modalities,
biotherapeutics can benefit from the use of a systematic, risk-
based, and data driven approach for the development and
implementation of analytical methods. Understanding the
CQAs of a mAb is an important step in a QbD approach to
selecting the appropriate analytical technology and methods
used to establish a comprehensive product testing and control
strategy. Attributes that are identified as CQAs from the risk
assessment and attributes that need to be assessed further are Figure 5. Structure of an mAb and its site-specific physiochemical and
included in the method selection process establishing the biological attributes.

G https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry pubs.acs.org/ac Perspective

typically assessed against biological activity, PK, immunoge- establishing characterization and structure−function studies.
nicity (specific to antidrug antibodies), and safety. Information For example, during development, it will be critical to further
critical to the assessment includes the biology and mechanism characterize the type of aggregation present because higher
of action (MoA) of the molecule, therapeutic dose, process order aggregates (e.g., greater than dimer) have an increased
and product-related variants, PK characteristics, biochemical potential to elicit adverse immunogenic responses in patients
and biophysical characterization, and the stability and safety and can, similar to the dimer, impact biological activity and
profile. Additionally, in vitro (e.g., immunogenicity), non- PK. 42 This approach will establish an evolving, but
clinical, and safety studies are leveraged to guide the CQA comprehensive, ATP based on a clear assessment of the
assessment and focus the scope of testing required. structural features to monitor and the analytical controls
Higher-order molecular weight species or aggregates are required for the commercial control strategy.
typically assessed as a high risk CQA37 as they can impact An example comparison of pros and cons for two techniques
product quality and efficacy and pose a potential safety risk to considered for the initial method selection to detect aggregates,
patients through enhanced immunogenicity.38 Aggregation is a and for the revised method selection when considering
general term for the self-association of proteins outside of the additional characterization, is provided in Table 5. Addition-
native quaternary structure.39 The formation of protein ally, business drivers for the two techniques, such as how
aggregates is a common, yet challenging, problem for quickly the method can be established in various laboratories,
biotherapeutics. During the downstream manufacturing steps, the availability of technology, throughput, and speed of analysis
the expressed protein is purified through various process are provided in Table 6. While size exclusion chromatography
column steps (e.g., protein A chromatography, ion exchange (SEC) is a common and readily available approach to detect
chromatography) and formulated using ultrafiltration/diafiltra- aggregates that has been reviewed elsewhere,43,44 it is
tion. Most aggregates are removed during these processing important to have a systematic evaluation using up-to-date
steps. However, aggregation occurs through a complex process technical specifications and analysis requirements. The “pros”
that can originate during the cell culture and downstream and “cons” for particular technologies may be weighted
process and product manufacturing, fill, and storage steps. differently depending on the CQA details. Aggregation that
The formation of protein aggregates can be influenced by is suspected to be driven by different mechanisms may indicate
post-translational modifications (PTMs), such as deamidation, concerns about the stability of the aggregate for analysis and
oxidation, iso-aspartic acid formation, disulfide bond scram- lead to the selection of an orthogonal approach during
bling, cross-linking, and by the presence of host-cell proteins development of the process or even characterization of the
(residual proteins other than the intended product).40 Various product.
process manufacturing conditions can also influence the In this example, the first goal for analysis is to establish a
formation of aggregation including pH, temperature, surface method to quantitatively monitor the total % aggregate
area, and physical stress as well as extrinsic factors such as throughout the various clearance steps and at release of the
storage conditions.41 The relationship between quality product for clinical use. SEC with UV detection is most often
attributes that can influence the amount and type of selected for process and quality control testing.45 The
aggregation and the impact of aggregation on other CQAs separation and quantitation of aggregate species is achieved
related to safety and efficacy is depicted in Figure 6. based on the permeation of the analyte into and out of the
stationary phase pores (molecular sieving). There are benefits
with this as opposed to reverse phase and ion exchange
chromatography where analytes are retained by chemical
interactions with the stationary phase. SEC provides a robust,
rapid, quantitative approach to detecting aggregation with
good sample throughput. Equipment is readily available, low
cost, and already deployed in GMP QC laboratories. SEC is
relatively easy to train on and operate, meaning that
transferring the methodology can be a relatively uncomplicated
process. SEC can be coupled with fraction collection to assess
binding and bioactivity as part of critical structure−function
experiments and is compatible with a variety of detectors
including multiangle laser light scattering (MALS), expanding
SEC’s utility in providing characterization information for
aggregate species. Other separation techniques such as
hydrophilic interaction chromatography (HILIC) and RP-LC
may denature the protein. Many of these reasons also make
SEC ideal to deploy as in-process testing to monitor
Figure 6. CQA relationships to aggregation.
manufacturing steps. For these reasons, SEC with UV
detection, as is the case here, is typically the appropriate
choice to fulfill the early phase development testing require-
While PTMs are monitored through characterization studies ments. Figure 7 shows a chromatogram for a protein
and clearance of aggregates is monitored during the down- therapeutic using a high-pressure, platform, SEC method that
stream process, the initial ATP would include the monitoring provides faster analysis time with good resolution between the
of total aggregates and fragments in the drug substance, drug monomer and higher molecular weight aggregate forms.
product, and on stability. As development proceeds, this As development proceeds, it is critical to the commercial
selection process will be refined, linking related CQAs and control strategy to understand the impact of aggregation,
H https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry pubs.acs.org/ac Perspective

Table 5. Pros and Cons of Methodology Selected to Detect and Characterize mAb Aggregation

particularly when significant levels of aggregate remain in the approach to characterizing the aggregate species, particularly
final product or occur on stability. Understanding aggregation confirming the accuracy and range of higher order aggregate
involves not only separating and quantifying the total amount species, is sedimentation velocity analytical ultracentrifugation
of aggregates but also characterizing the aggregate forms (SV-AUC). SV-AUC measures the sedimentation coefficient of
present and the potential impact on safety and efficacy. The the protein particles in a gravitational field. Radial scans of the
ATP was revised to select additional methods (complementary concentration profile are collected sequentially guided by UV
and orthogonal to SEC-UV) that could confirm the accuracy absorbance detection (typically 280 nm wavelength).48,49 The
and range of higher order aggregate species and methods that distribution profile is used to provide an estimated molecular
could collect samples of the observed peaks for character- weight and % abundance for each aggregate species. While SV-
ization and binding studies. AUC is sensitive to analyst control, has low throughput, and
MALS, in addition to UV absorbance, can be used as a can demonstrate relatively high measurement variability, SV-
complementary detection scheme to determine the molecular AUC can also characterize insoluble or soluble aggregates. In
mass of the SEC fractions as seen in Figure 8. The molar mass addition, SV-AUC avoids the issues with SEC previously
is calculated using light scattering intensity as a function of the described that can impact quantitation and detection of the
concentration at elution. While SEC coupled with MALS can range of higher order aggregate species.47,50 SV-AUC is an
provide sensitive detection of higher order aggregates,46 SEC orthogonal approach to confirm the SEC and SEC-MALS
has disadvantages that can challenge accurate characterization observations.48,49
of the full range of aggregate species in the product, and it is The ATP was revised using the additional requirements to
generally accepted that additional techniques are needed for confirm the identification and range of higher order aggregates
the assessment of aggregates.47 Some of these disadvantages in the product and the quantitative accuracy observed by SEC-
include dilution effects where preparation of the sample can UV. Methods were screened against these requirements,
induce the formation or dissociation of aggregates. Addition- general method performance characteristics, and business
ally, adsorption of the protein to column or exclusion of requirements. In addition to using SEC-UV, SEC-MALS and
aggregates by the frit into the void can impact quantitation and SV-AUC were selected. Aggregate fractions from SEC were
identification of higher order species.47 An orthogonal collected and subjected to SEC-MALS demonstrating that
I https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry pubs.acs.org/ac Perspective

Table 6. Evaluation of Business Considerations for Selected Aggregate Detection and Characterization Methodologya

a
Scoring = (++) high agreement with requirements, (+) meets requirements, (−) does not meet basic requirements or is not able to be evaluated.

Figure 7. Example SEC chromatogram (UHPLC). Inset showing the Figure 8. Example of SEC-MALS of collected aggregate fractions.
zoom area. The dotted line indicates the molecular mass in Daltons as a function
of time.
aggregate species were predominately a dimer (Figure 8). SV-
AUC (not shown) confirmed that the drug product is studies revealed that binding to the antigen is impacted by
predominately a monomer, with noncovalently bound dimer aggregates, reducing the specific binding activity of the
being the significant aggregate impurity. product. However, even larger percentages of dimer than
Additionally, the aggregate fractions were tested in binding were observed (up to 5%) would have a negligible overall
studies using surface plasmon resonance (SPR). The SPR effect on the potency of the product. The aggregate
J https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry


pubs.acs.org/ac Perspective

characterization work described here links directly to patient CONCLUSION


safety. This, coupled with binding studies addressing efficacy, A well-constructed ATP enables all potential suitable analytical
could result in defining patient-centric specification limits and technologies to be evaluated in an objective way. Consid-
avoiding limits derived solely by the process capability.37 The eration of business drivers at the same time enables the
result is further refinement of the ATP and a simpler screening of methods to focus on the most preferred
commercial analytical control strategy. technology that is likely to be robust and rugged. Multiple


ATPs can be considered together to identify a single
DISCUSSION technology which can be used to develop a multiattribute
analytical procedure. In early development, for quality
In small molecules, well-established pharmacopoeial mono- attributes requiring assessment of impact on safety/efficacy,
graphs exist which link acceptance criteria to well-established analytical technologies can be used to develop structural
types of analytical technology. This makes method develop- understanding which is required to establish an ATP in the
ment approaches for novel/different analytical technology context of the overall control strategy.


problematic. For example, Goodwin et al.51 describe the
situation for end product testing of tablet content, where the AUTHOR INFORMATION
traditional approach involves taking a composite sample of 10 Corresponding Author
tablets and performing a bulk assay by HPLC of 5 tablets in Phil Borman − Product Development and Supply, GSK,
duplicate. The mean of the two determinations is typically Medicines Research Centre, Stevenage SG1 2NY, U.K.;
reported with a limit of ±5% label claim in Europe and ±10% orcid.org/0000-0002-6742-5826;
in the U.S. Email: phil.j.borman@gsk.com
The application of at-line near-infrared (NIR) can be used
directly on intermediate materials during the manufacture of Authors
final products without the need for any sample preparation and Cristiana Campa − Technical Research & Development,
has obvious benefits as discussed by Goodwin et al.51 For NIR Vaccines, GSK, 53100 Siena, Italy
(and other spectroscopic analytical procedures), tablet content Ghislain Delpierre − Analytical Research and Development,
is provided on individual tablets, and there is an obvious GSK, 1330 Rixensart, Belgium
potential for increasing the sample size well above 10 tablets Elliot Hook − Global Pharma Analytical Science and
per batch for a more accurate determination of the batch mean. Technology, Pharma Supply Chain, GSK, Ware SG12 0DJ,
Rather than rely on technology specific pharmacopoeial U.K.
guidance, an ATP can be used to ensure true technology Patrick Jackson − Product Development and Supply, GSK,
Medicines Research Centre, Stevenage SG1 2NY, U.K.;
independent criteria can be established for the measurement of
orcid.org/0000-0002-0322-040X
quality attributes. Jackson et al.2 define an ATP for tablet
Wayne Kelley − Product Development and Supply, GSK, King
content where criteria for specificity, accuracy, and precision of Prussia, Pennsylvania 19406, United States
are defined up front which can be adapted to any technology. Michel Protz − Analytical Research and Development, GSK,
The vaccines example (Example 2: development of a cell- 1330 Rixensart, Belgium
based assay) demonstrated how a systematic approach for Olivier Vandeputte − Analytical Research and Development,
technology evaluation and method screening enabled an GSK, 1330 Rixensart, Belgium
efficient analytical method which was aligned with regulatory
Complete contact information is available at:
expectations to be developed which could analyze three very
https://pubs.acs.org/10.1021/acs.analchem.1c03854
different CQAs. This “multi-CQA” method could potentially
be converted into a platform analytical method (provided the Notes
availability of specific monoclonal antibodies). Several backup The authors declare no competing financial interest.


methods (including NAT assays) were also identified.
Unlike examples 1 and 2 where ATPs were already ACKNOWLEDGMENTS
established, the method development approach in example 3
for the aggregation of a monoclonal antibody in early The authors wish to acknowledge Troy Adams, Marc Fiorucci,
Claudia Magagnoli, and Margarita Orticochea for their
development was used to generate the structural understanding
contributions.


required to establish a comprehensive analytical target profile.
A method was initially required to characterize and quantify REFERENCES
any resultant aggregation in the drug substance, drug product,
(1) ICH Q8(R2) Pharmaceutical Development, 2009.
and on stability. As development proceeded, the method (2) Jackson, P.; Borman, P.; Campa, C.; Chatfield, M.; Godfrey, M.;
selection process was refined linking related CQAs and Hamilton, P.; Hoyer, W.; Norelli, F.; Orr, R.; Schofield, T. Anal.
establishing characterization and structure−function study Chem. 2019, 91 (4), 2577−2585.
goals. An ATP could only be developed once this character- (3) Barnett, K. L.; McGregor, P. L.; Martin, G. P.; Le Blond, D. J.;
ization work was complete. Weitzel, M. L. J.; Ermer, J.; Walfish, S.; Nethercote, P.; Gratzl, G. S.;
Fit-for-purpose analytical procedures were developed for Kovacs, E.; Pappa, H., Analytical target profile: Structure and
examples 1 and 2, which formed part of the overall control application throughout the analytical lifecycle. Pharmacopeial Forum
2016, 42 (5).
strategy. Example 3 demonstrated how an ATP was revised (4) Rignall, A.; Borman, P.; Hanna-Brown, M.; Grosche, O.;
based on the knowledge gained from using a battery of Hamilton, P.; Gervais, A.; Katzenbach, S.; Wypych, J.; Hoffmann, J.;
analytical technologies to better define the requirements for Ermer, J.; McLaughlin, K.; Uhlich, T.; Finkler, C.; Liebelt, K. Pharm.
measuring a CQA. Technol. 2018, 42 (12), 18−23.

K https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX
Analytical Chemistry pubs.acs.org/ac Perspective

(5) Elder, D. P.; Borman, P. Pharmaceutical Outsourcing 2013, 14 Winkel, J. G.J.; Aalberse, R. C.; Parren, P. W. H. I. Science 2007, 317
(4), 14−19. (5844), 1554−1557.
(6) USP ⟨1220⟩ Analytical Procedure Lifecycle Pharmacopeial (37) Ruesch, M. N.; Benetti, L.; Berkay, E.; Cirelli, D. J.; Frantz, N.;
Forum 2020, 46 (5). Gastens, M. H.; Kelley, W. P.; Kretsinger, J.; Lewis, M.; Novick, S.;
(7) Hanna-Brown, M.; Borman, P.; Bale, S.; Szucs, R.; Roberts, J.; Rellahan, B.; Pack, L.; Stroop, C. J. M.; Subashi, A.; Yin, P.; Zeng, M.;
Jones, J. Sep. Sci. 2010, 2, 12−20. Stults, J. J. Pharm. Sci. 2021, 110 (2), 771−784.
(8) Vogt, F. G.; Kord, A. S. J. Pharm. Sci. 2011, 100 (3), 797−812. (38) Den Engelsman, J.; Garidel, P.; Smulders, R.; Koll, H.; Smith,
(9) Li, Y.; Terfloth, G. J.; Kord, A. S. Am. Pharm. Rev. 2009, 12 (4), B.; Bassarab, S.; Seidl, A.; Hainzl, O.; Jiskoot, W. Pharm. Res. 2011, 28
571−583. (4), 920−933.
(10) Zhou, L.; Socha, J. M.; Vogt, F. G.; Chen, S.; Kord, A. S. Am. (39) Narhi, L. O.; Schmit, J.; Bechtold-Peters, K.; Sharma, D. J.
Pharm. Rev. 2010, 13 (1), 74−84. Pharm. Sci. 2012, 101 (2), 493−498.
(11) Zhou, L.; Vogt, F. G.; Overstreet, P. A.; Dougherty, J. T.; (40) Le Basle, Y.; Chennell, P.; Tokhadze, N.; Astier, A.; Sautou, V.
Clawson, J. S.; Kord, A. S. J. Pharm. Innov. 2011, 6 (4), 217−231. J. Pharm. Sci. 2020, 109 (1), 169−190.
(12) ICH Q2(R1) Validation of Analytical Procedures, 1994. (41) Philo, J. S.; Arakawa, T. Curr. Pharm. Biotechnol. 2009, 10 (4),
(13) Borman, P.; Roberts, J.; Jones, C.; Hanna-Brown, M.; Szucs, R.; 348−351.
Bale, S. Sep. Sci. 2010, 2, 2−8. (42) Ratanji, K. D.; Derrick, J. P.; Dearman, R. J.; Kimber, I. J.
(14) Borman, P.; Chatfield, M.; Jackson, P.; Laures, A.; Okafo, G. Immunotoxicol. 2014, 11 (2), 99−109.
Pharm. Technol. Eur. 2010, 4 (22), 38−46. (43) Brusotti, G.; Calleri, E.; Colombo, R.; Massolini, G.; Rinaldi, F.;
(15) Tome, T.; Ž igart, N.; Č asar, Z.; Obreza, A. Org. Process Res. Temporini, C. Chromatographia 2018, 81 (1), 3−23.
Dev. 2019, 23 (9), 1784−1802. (44) Hong, P.; Koza, S.; Bouvier, E. S. P. J. Liq. Chromatogr. Relat.
(16) Pollard, A.; Bijker, E. Nat. Rev. Immunol. 2021, 21, 83−100. Technol. 2012, 35 (20), 2923−2950.
(17) Brisse, M.; Vrba, S. M.; Kirk, N.; Liang, Y.; Ly, H. Front. (45) Philo, J. S. AAPS J. 2006, 8 (3), E564−E571.
Immunol. 2020, 11, 2578. (46) Ahrer, K.; Buchacher, A.; Iberer, G.; Josic, D.; Jungbauer, A. J.
(18) Sasso, E.; D’Alise, A. M.; Zambrano, N.; Scarselli, E.; Folgori, Chromatogr. A 2003, 1009 (1−2), 89−96.
A.; Nicosia, A. Semin. Immunol. 2020, 50, 101430. (47) Gandhi, A. V.; Pothecary, M. R.; Bain, D. L.; Carpenter, J. F. J.
(19) Humphreys, I. R.; Sebastian, S. Immunology 2018, 153 (1), 1− Pharm. Sci. 2017, 106 (8), 2178−2186.
9. (48) Gabrielson, J. P.; Arthur, K. K. Methods 2011, 54 (1), 83−91.
(20) Voellmy, R.; Bloom, D. C.; Vilaboa, N. Vaccines 2020, 8 (2), (49) Philo, J. S. Curr. Pharm. Biotechnol. 2009, 10 (4), 359−372.
230. (50) Arthur, K. K.; Kendrick, B. S.; Gabrielson, J. P. Methods
(21) Barry, M.; Rubin, J.; Lu, S. FEBS Lett. 2020, 594 (12), 1918− Enzymol. 2015, 562, 477−500.
1946. (51) Goodwin, D. J.; van den Ban, S.; Denham, M.; Barylski, I. Int. J.
(22) Afrough, S.; Rhodes, S.; Evans, T.; White, R.; Benest, J. Vaccines Pharm. 2018, 537 (1−2), 183−192.
2020, 8 (1), 131.
(23) Fedosyuk, S.; Merritt, T.; Peralta-Alvarez, M. P.; Morris, S. J.;
Lam, A.; Laroudie, N.; Kangokar, A.; Wright, D.; Warimwe, G. M.;
Angell-Manning, P.; Ritchie, A. J.; Gilbert, S. C.; Xenopoulos, A.;
Boumlic, A.; Douglas, A. D. Vaccine 2019, 37 (47), 6951−6961.
(24) Vitelli, A.; Folgori, A.; Scarselli, E.; Colloca, S.; Capone, S.;
Nicosia, A. Expert Rev. Vaccines 2017, 16 (12), 1241−1252.
(25) Kozlowski, D. A.; Bremer, E.; Redmond, D. E., Jr; George, D.;
Larson, B.; Bohn, M. C. Mol. Ther. 2001, 3 (2), 256−261.
(26) Suzuki, M.; Kondo, P.; Pei, Z.; Maekawa, A.; Saito, I.; Kanegae,
Y. Gene Ther. 2015, 22, 421−429.
(27) Takahashi, Y.; Vikman, E.; Nishikawa, M.; Ando, M.;
Watanabe, Y.; Takakura, Y. J. Gene Med. 2010, 12 (9), 739−746.
(28) Nambiar, B.; Cornell Sookdeo, C.; Berthelette, P.; Jackson, R.;
Piraino, S.; Burnham, B.; Nass, S.; Souza, D.; O’Riordan, C. R.;
Vincent, K. A.; Cheng, S. H.; Armentano, D.; Kyostio-Moore, S. Hum.
Gene Ther: Methods. 2017, 28 (1), 23−38.
(29) Wang, Z.; Zhao, J. Viruses 2019, 11 (8), 741.
(30) Brescia, M.; Janssen, J.; Liu, J.; Goncalves, M. Cells 2020, 9 (4),
869.
(31) Lee, C. S.; Bishop, E. S.; Zhang, R.; Yu, X.; Farina, E. M.; Yan,
S.; Zhao, C.; Zeng, Z.; Shu, Y.; Wu, X.; Lei, J.; Li, Y.; Zhang, W.;
Yang, C.; Wu, K.; Wu, Y.; Ho, S.; Athiviraham, A.; Lee, M. J.; Wolf, J.
M.; Reid, R. R.; He, T. C. Genes Dis. 2017, 4 (2), 43−63.
(32) FDA. Chemistry, Manufacturing, and Control (CMC) Information
for Human Gene Therapy Investigational New Drug Applications
(INDs), 2020.
(33) Kimiz-Gebologlu, I.; Gulce-Iz, S.; Biray-Avci, C. Mol. Biol. Rep.
2018, 45 (6), 2935−2940.
(34) Teo, E. C. Y.; Chew, Y.; Phipps, C. Crit. Rev. Oncol./Hematol.
2016, 97, 72−84.
(35) Mahmuda, A.; Bande, F.; Kadhim Al-Zihiry, K. J.;
Abdulhaleem, N.; Majid, R. A.; Hamat, R. A.; Abdullah, W. O.;
Unyah, Z. Trop. J. Pharm. Res. 2017, 16 (3), 713−722.
(36) van der Neut Kolfschoten, M.; Schuurman, J.; Losen, M.;
Bleeker, W. K.; Martinez-Martinez, P.; Vermeulen, E.; den Bleker, T.
H.; Wiegman, L.; Vink, T.; Aarden, L. A.; De Baets, M. H.; van de

L https://doi.org/10.1021/acs.analchem.1c03854
Anal. Chem. XXXX, XXX, XXX−XXX

You might also like