You are on page 1of 102

CONTINUED PROCESS

VERIFICATION
AN INDUSTRY
POSITION PAPER
WITH EXAMPLE PLAN

CONNECT
COLLABORATE
ACCELERATE TM

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 1
Contributors
The following people were lead contributors to the content of this document, writing sections, editing and
liaising with colleagues to ensure that the messages it contains are representative of current thinking across
the biopharmaceutical industry. This document is a consensus view of a model CPV Plan, but it does not
represent fully, the internal policies of the contributing companies.

AbbVie GSK
Mike Doremus Dan Baker
AstraZeneca Lonza
Cynthia Ball Rajesh Beri
Baxter Merck
Joerg Gampfer Julia O’Neill
Bayer Novartis
John Grunkemeier Abe Germansderfer
Gallus Pfizer
Madeline Roche Jeff Fleming
Genzyme Regeneron
Lada Laenan Jenny McNay

Additionally, excellent editorial support and constructive criticism was provided by:

AstraZeneca Pfizer
Ranjit Deschmukh Eric Hamann
Paul McCormac
Genentech/Roche
Mark Smith Regeneron
Rajesh Ahuja
Merck
Beth Junker Shire
Bert Frohlich
Novartis
Christelle Pradines

The work was facilitated by Darren Whitman and Robin Payne of BioPhorum Operations Group.

Though this paper is issued under copyright, © 2014, Biophorum Operations Group, it is intended to be readily accessed
across the industry, free of charge and can be accessed from the BioPhorum website at the following address:

www.biophorum.com/download/cvp-case-study-interactive-version/

When citing this paper, please use the following form:

BioPhorum, 2014, Continued Process Verification: An Industry Position Paper with Example Plan
© 2014, Biophorum Operations Group

Redesigned in line with new brand guidelines April 2020

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 2
SUMMARY

Executive summary
This paper is a response to US Food and Drug Administration (FDA)
2011 process validation guidance on Stage 3, ‘Process Validation:
General Principles and Practices’5. It describes the approach commonly
referred to as ‘Continued Process Verification’ (CPV). As one might
expect, manufacturers in the biopharmaceutical sector all wish to
respond to this guidance appropriately. A group of 20+ companies felt it
would be valuable to work on this topic together, using the facilitation
services of the BioPhorum Operations Group (BPOG) (www.biophorum.
com). This paper is one of the results of the collaborative effort. It is
written as a consensus view of an acceptable CPV program, but it does
not fully represent the internal policies of the contributing companies.
It is a basis upon which to build and share knowledge further across
the industry. The authors believe this is one of the first comprehensive
papers on this topic.
The paper seeks to provide practical developments on the themes: what is CPV, why is it
important, and how might it be implemented. It offers some specific recommendations on the
content of a CPV Plan, along with associated rationale. These recommendations are based on
a typical cell culture production process for making a fictitious monoclonal antibody product,
described in the ‘A-Mab Case Study’3. Consequently, not all of the details contained in this
paper are going to apply directly to actual products or processes. The authors recognize that
the A-Mab Case Study represents only one industry archetype, and that there are a number
of others that are important. However, the concepts and principles upon which the content
of this paper was derived should help with CPV implementation for a real product. Some of
the complications of implementation are addressed, with recommended approaches, but the
issue of information technology (IT) systems is not dealt with directly here. The case for IT
systems, their design and introduction, is the subject of other collaborative efforts facilitated
by BPOG and some of the results of that work may be published in the future.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 3
CPV is fundamentally a formal means by which a robust set of data at the scale of commercial manufacture.
commercial manufacturing process is monitored to To manage this situation in practice, it is recommended
ensure product quality. It encompasses a written plan for that short term control criteria are set initially, based on
monitoring a licensed biopharmaceutical manufacturing prior process experience and including data acquired at
process, as well as regular reporting and actions based the laboratory and clinical scales of manufacture. This
on the results of monitoring the process. CPV reporting initial period of production would then be used to establish
provides a basis from which to improve process longer term criteria that are more statistically appropriate.
understanding, risk assessment, the control strategy
The implementation and ongoing execution of a CPV
(CS) [9], and ultimately the process itself. In general, the
Plan is likely to require additional effort, beyond what is
nature and extent of CPV is aligned with the outcomes of
typically needed to prepare for the Annual Product Review
process qualification. Whilst a CPV Plan is likely to include
(APR), because significant amounts of additional data
data related to Batch Release (BR), and so may be useful
are collected and analyzed to improve understanding of
in supporting BR decisions, CPV operates independently
process variability. However, it is likely that the benefits
from the BR process and is not expected to have any
accruing from the improved information available for
impact on batches that have been previously released.
process improvement will outweigh any additional costs.
Adopting or building on an existing system of monitoring The actual additional cost depends on the amount of data
manufacturing process performance means more data to be analysed which in turn depends on the outcomes
will be collected over the lifetime of the product. CPV of quality risk assessments that define data collection
execution may involve examination of existing process scope and frequency. The frequency of collection depends
control measurements and improved methods for data on several factors, including: whether production is
tracking and analysis. Enhanced monitoring of process campaigned or continuous; the extent of variability
performance provides the opportunity to identify and apparent in the process; whether risks to product quality
control sources of variation and hence improve process (and thus product disposition) and process consistency are
robustness, offering the major benefit of reliable supply to sufficiently mitigated, and the intended use of the reported
the market. data (for example, use in continuous improvement may
One of the main technical issues to resolve when mean collecting and analyzing certain data on a daily
implementing CPV relates to the quantity of data required basis).
before product commercialization. In a sense, CPV Given the importance of CPV in both compliance and
complements the ‘Quality by Design’ (QbD) framework process improvement terms, the authors encourage
that manufacturers have developed to license and executives to read and share this paper with their
commercialize the product, though a CPV Plan may be colleagues. The authors also welcome any comments or
constrained to data available in manufacturing. It should questions arising which can be submitted via the following
be noted that not all products will have a QbD framework email address: cpv@biophorum.com.
but all need a CPV Plan. Also, at the time of commercial
product introduction, there is unlikely to be a statistically

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 4
Contents
Contents............................................................................................................................................................................................................. 5

1.0 Purpose................................................................................................................................................................................................. 7

2.0 Scope..................................................................................................................................................................................................... 9

3.0 Roles and responsibilities..............................................................................................................................................................10

4.0 CPV plan references ......................................................................................................................................................................12

5.0 Product and process description .................................................................................................................................................13

5.1 Brief description of the general approach used in the A-Mab case study.................................................................... 14

5.2 Parameters to be included in CPV........................................................................................................................................... 15

5.3 Upstream process overview...................................................................................................................................................... 16

5.4 Downstream process overview................................................................................................................................................ 17

5.5 Identification of CQAs and acceptance ranges.................................................................................................................... 18

5.6 Process parameter characterization....................................................................................................................................... 20

5.7 Control strategy CQAs and CPPS ............................................................................................................................................ 22

6.0 Developing a monitoring strategy...............................................................................................................................................23

6.1 Rationale and background ......................................................................................................................................................... 23

6.2 Hypothetical scenarios and planned process changes....................................................................................................... 24

7.0 CPV plan recommendations for the A-Mab process ..............................................................................................................28

7.1 Step 1, seed culture expansion in disposable vessels – CPV recommendations........................................................ 29

7.2 Step 2, Seed Culture Expansion in Bioreactors – CPV Recommendations................................................................. 30

7.3 Step 3, Production Culture Bioreactor – CPV Recommendations................................................................................ 31

7.4 Step 4, Clarification (centrifugation and depth filtration) – CPV recommendations............................................... 35

7.5 Step 5, Protein A Chromatography – CPV recommendations......................................................................................... 36

7.6 Step 6, Low pH treatment – CPV recommendations.......................................................................................................... 37

7.7 Step 7, Cation Exchange Chromatography (CEX) – CPV recommendations.............................................................. 39

7.8 Step 8, Anion Exchange Chromatography (AEX) – CPV recommendations................................................................ 40

7.9 Step 9, Small Virus Retentive Filtration (SVRF) – CPV recommendations.................................................................. 42

7.10 Step 10, Ultrafiltration and Diafiltration (UF/DF) – CPV recommendations.............................................................. 43

7.11 Step 11, Final Filtration and Freezing of BDS – CPV recommendations...................................................................... 45

7.12 Bulk Drug Substance Lot Data – CPV recommendations.................................................................................................. 47

8.0 Frequency and scope of CPV analysis ........................................................................................................................................49

8.1 Scope of CPV Analysis ............................................................................................................................................................... 49

8.2 Frequency of Analysis.................................................................................................................................................................. 50

9.0 Establishing control limits ............................................................................................................................................................51

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 5
10.0 Example CPV execution plan for drug substance ....................................................................................................................53

10.1 Step 1, Seed culture expansion in disposable vessels – CPV variables........................................................................ 57

10.2 Step 2, Seed culture expansion in bioreactors – CPV variables..................................................................................... 58

10.3 Step 3, Production culture bioreactor – CPV variables..................................................................................................... 59

10.4 Step 4, centrifugation and depth filtration – CPV variables............................................................................................ 62

10.5 Step 5, Protein A chromatography – CPV variables........................................................................................................... 63

10.6 Step 6, Low pH treatment – CPV variables........................................................................................................................... 64

10.7 Step 7, Cation exchange chromatography – CPV variables............................................................................................. 65

10.8 Step 8, Anion exchange chromatography – CPV variables............................................................................................... 66

10.9 Step 9, Small virus retentive filtration – CPV variables.................................................................................................... 68

10.10 Step 10, Ultrafiltration and diafiltration – CPV variables................................................................................................. 68

10.11 Step 11, Final filtration/Bulk fill and freezing of BDS – CPV variables......................................................................... 70

10.12 CPV monitoring of bulk drug substance lot data................................................................................................................. 71

11.0 CPV sampling plan ..........................................................................................................................................................................73

11.1 Template for specific process steps ....................................................................................................................................... 76

12.0 How data will be analyzed ............................................................................................................................................................80

12.1 Identifying software..................................................................................................................................................................... 80

12.2 Description of tools to trend and evaluate data.................................................................................................................. 81

12.3 Process capability index............................................................................................................................................................. 82

12.4 Control charts................................................................................................................................................................................ 84

12.5 Multivariate data analysis.......................................................................................................................................................... 86

12.6 Responses to shifts and trends.................................................................................................................................................. 87

12.7 Establishing initial limits............................................................................................................................................................. 88

12.8 Establishing long-term limits..................................................................................................................................................... 88

12.9 Finding signals of special cause variation.............................................................................................................................. 89

13.0 Change management .....................................................................................................................................................................90

14.0 Data verification...............................................................................................................................................................................93

Discretionary elements of a CPV program .............................................................................................................................................95

References.......................................................................................................................................................................................................96

Glossary............................................................................................................................................................................................................97

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 6
1.0
Purpose
This document is written with the aim of providing a technical, non-binding, industry consensus response to
regulatory guidance. It is not in itself guidance. The objective of this paper is to provide:
(1) an example of key portions of a Continued Process Verification (CPV) plan for a biologics process; (2) relevant
industry thinking on CPV plan development and implementation.

This document is different from others on this subject1, 2 control over the manufacturing process. N.B. at the time
because it is specific to a biologics manufacturing process of writing, the European Medicines Agency (EMA) draft
and provides a comprehensive case-study lifecycle guidance on Process Validation is out for consultation,
view that leverages antibody manufacturing process referring to KPAs as 'performance indicators'. The thought
development, as described in the A-Mab Quality-by- processes and examples presented in this document
Design case study3. It is worth the reader being familiar are backed by biotech industry experience with, subject
with the A-Mab case study and perhaps having a copy matter expertise in process monitoring for monoclonal
available for reference. It should be recognised that antibody and similar manufacturing processes.
the monoclonal antibody process is just one archetype
Furthermore, this document describes the thought
in the industry, though it is a useful one upon which to
processes that determine the content for a CPV plan. The
demonstrate principles, as it is known to many.
plan serves as the procedure governing document for the
The example of a CPV plan shown in this paper describes implementation and maintenance of CPV for a licensed
how to meet expectations5 for routine monitoring manufacturing process. Various parts of the plan are
of critical process parameters (CPPs), critical quality described in the following sections of the document, as
attributes (CQAs), key process attributes (KPAs) and key noted in Table 1.1 overleaf:
process parameters (KPPs) to demonstrate the state of

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 7
Table 1.1: Plan parts referenced by section number

General topic section Section Description


number/title

Manufacturing Process 5 Summary of the A-Mab manufacturing process and the A-Mab product description.

6 Selection of the process monitoring sampling plan backed by the process validation Phase I and II
data and the updated risk assessment.

7 The rationale for classification of quality-linked process parameters summarized in the A-Mab
case study is reviewed and summarized in the table that presents process performance
consistency and robustness. Rationale for what to include in CPV is provided, based on a
review and analysis of quality-linked process parameters from the A-Mab case study that
affect process performance, consistency and robustness.

Verification process 8 The frequency of CPV data analysis and trend review is discussed. The concept of an initial
or short-term CPV phase is introduced, where sufficient process experience is collected
to establish the manufacturing control limits for the process attributes identified during
validation. A subsequent phase of CPV implementation; that of steady state or long-term
process monitoring is also discussed.

9 Statistical and general methods for establishing CPV trend limits are presented.

10 The summary of the monitored attributes and parameters within the scope of the CPV program
are presented. The monitoring method and periodicity associated with specific attributes and
parame ters are also specified.

11 The sampling plan derivation with tabulated examples.

12 Aspects of data analysis and evaluation of results are discussed in this section. The emphasis is on
the possible outcomes of routine monitoring.

13 Change management and the impact of CPV on this process.

14 The specific need for data verification.

15 Elements of CPV that are considered discretionary.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 8
2.0
Scope
Consistent with the FDA’s 2011 Process Validation guidance document5 describing three stages of the product
lifecycle, CPV implementation discussed in this paper is limited to Stage 3, commercial manufacture of a drug
substance, following process design (Stage 1) and process qualification and qualification of the equipment and the
facility (PQ, Stage 2, see FDA 2011 Guidance Stage 24) 12.

Note: Whilst this paper focuses on the drug substance However, the proposed approach for CPV model CPV
manufacturing process, CPV should be applied all areas of Plan. implementation is also applicable to legacy products
Operations including formulation, fill and finish. where quality attributes and parameters for monitoring
The application of the principles discussed in this can be determined based on a combination of process
document BioPhorum is initiating collaborative work, knowledge and historical performance data.
specifically focused for new products relies on product BioPhorum is initiating collaborative work, specifically
and process development on CPV for established, licensed focused on CPV for established, licensed (or legacy)
(or legacy) products and the and characterization studies products and the resulting recommendations may be
(Stage 1) to define the scope resulting recommendations published in the future. An ISPE group produced an article
may be published in the future. of the CPV program. This covering this broadened scope in 201224; here we believe
document is based on the CS An ISPE group produced an we address a reduced scope in greater detail, providing
article covering this broadened presented in the A-Mab deeper development of a model CPV Plan.
bioprocess development case scope in 201224; here we
believe we address a reduced study and is primarily
focused on the commercialization of scope in greater
detail, providing deeper development of a a new product.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 9
3.0
Roles and responsibilities
The roles and responsibilities suggested here as examples, are based upon a typical organizational structure of a
biopharmaceutical manufacturing company (Table 3.1.).

Several primary functional areas have important Outputs of the CPV program can be used by the
responsibilities required to successfully execute the CPV Regulatory Affairs and Quality organizations for annual
program. These areas are: Development, Validation, agency updates, such as the Annual Product Review (APR)
Operations, Quality Control, Quality Engineering and and Product Quality Review (PQR). Terminology for each
Quality Assurance. Operations, a function which may also function may vary by organization.
be known as Technical Operations, is assumed to contain
Note: The responsibilities for continued process monitoring
Manufacturing as well as Manufacturing Science and
should be clearly defined within the organization and recorded
Technology personnel. Mathematical sciences or
in the CPV Plan. Responsibilities can be tailored to a specific
non-clinical statistics support is of paramount importance
organizational structure, given its maturity and size.
in achieving sound data interpretation. Each functional
area has responsibility for specific activities, as shown in
Table 3.1.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 10
Table 3.1: Roles and Responsibilities for a CPV Program:

Functional area Responsibility

Management • Ensure that adequate resources are available to carry out the CPV program and to regularly
perform a review of CPV plan summaries or reports.

Development • Provide documentation that defines current process knowledge, quality attributes, process
parameters and elements of the overall CS that forms the basis for the CPV program.
• Provide documented scientific justification for parameters, limits, ranges and elements of the
CS, based upon development studies or other prior knowledge.
• Provide technical input to develop response actions, including input in prioritization of
continuous improvement activities.
• Consider application of CPV outcomes to new processes in development.

Validation/ Quality functions • Provide internal advice on current validation principles and ensure that validation protocols,
interim and final reports meet applicable standards.
• Participate in cross-functional teams to review production and QC data as part of the CPV
program.
• Review the data, pursue appropriate investigations and make decisions on how to proceed.
• May generate CPV plans and summary reports.
• Review and approve CPV plan, CPV reports and any changes to the CPV plan.

Operations/Manufacturing • Own the manufacturing process and take responsibility to ensure that it is maintained in a
Science and Technology state of control throughout the product lifecycle in manufacturing.
(N.B. It is not unusual for a Manufacturing Science • Ensure that all required production and process data are collected as part of executing the
and Technology function to be independent CPV plan for the product.
of Operations and Quality organisations. An • Performs continued process monitoring activities, including collecting, entering, verifying,
alternative arrangement may be reviewing and analyzing process data.
reporting into Process Sciences.) • Generate control charts and document CPV analysis for process data.
• Regularly participate in cross-functional teams in order to review production and QC data
as part of the CPV program.
• Maintain the process commercial master batch production and control records up to date,
capturing continuous improvements resulting from CPV in documentation as necessary.

Quality Control • Perform quality control testing and document results that are used in CPV evaluations.
• Perform continued process monitoring activities, including collecting, entering, verifying, reviewing
and analyzing QC data.
• Generate control charts and document CPV analysis for QC data.
• Participate in cross-functional teams to review production and QC data as part of the CPV program.

Quality Engineering/Mathematical Sciences/ • Provide internal advice on statistical analyses needed to successfully complete CPV activities.
Non Clinical Statistics • Act as a Subject Matter Expert (SME) and train personnel in other groups on statistical data
analysis techniques used in CPV.
• Provide internal advice on how to develop the data collection plan and help select suitable
statistical methods and procedures that are used to measure and evaluate the process
stability and capability.
• Generate procedures that define the way statistical tools and approaches are to be used in
routine process monitoring.
• Provide guidance on how to set control limits and define and interpret signal criteria.

Quality Assurance • Review and approve CPV plans and reports.


• Review and approve the list of attributes and parameters to be monitored, and control
chart limits.
• Participate in cross-functional data review to review production and QC data as part of
the CPV program.
• Review CPV reports and establish where signals require formal non-conformance
investigations.
• Coordinate assembly of CPV program reports.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 11
4.0
CPV plan references
The following references are expected to be created in the quality management system and are important when
constructing a CPV plan, providing background and critical internal interpretation of regulatory guidance. They
should be referenced accurately in a CPV Plan document. Note, a CPV Plan is expected to be product and process
specific. It may be advantageous to develop corporate policies and this forms the basis for some of the list of
references that follows.

• Quality Policy, Manual or Master Plan on CPV Technical references relevant to the detailed sections of
this paper are provided in section 16. References 1 to 9 are
• 
Company Standard/Guideline for CPV
recommended as initial texts when creating or updating a
(requirements for CPV, for e.g. timing, relationship
CPV plan.
to APRs, etc)
*The authors recognize that the plan illustrated in this paper
• 
SOP on CPV (Definitions, Abbreviations, responses
is written largely with CPV for new products in mind and that
to deviations, report generation, etc)
there would not be APRs available at the point of product
• 
SOP on Statistical Methods for trending, statistical licensure. This bullet point is included as a reminder that
analysis and identifying special cause variations historic APRs would provide data for the creation of a CPV plan
• 
Template for CPV Plan where established, licensed or legacy products are concerned.

• 
Template for CPV Charts & Graphs
• 
Template for CPV Report
• 
Manufacturing process description
• 
Control Strategy for the process (version number)
• 
Process risk assessment (version number)
• 
Applicable Risk assessment(s) (version number)
providing basis for rationale of CPV monitoring
selection
• 
Previous annual product report(s) if available,
otherwise consider evidence for a similar product*.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 12
5.0
Product and
process description
The A-Mab case study describes a model Quality
by Design (QbD) approach for development of a
monoclonal antibody (A-Mab)3,6. Considering the FDA
process validation guideline5, the case study includes
work covered during Stage 1 (Process Design) but
does not include information on Stage 2, Process
Performance Qualification (PPQ)5.
In preparing this CPV example plan, it is assumed that
Stage 2, was completed successfully for the A-Mab
process. The plan described applies to Stage 3 of the
process validation lifecycle.
Note: Whilst a QbD approach could be said to provide was
completed successfully for the A-Mab process. The plan
advantages in terms of process understanding, it is not an
described applies to Stage 3 of the process validation lifecycle.
approach that has to be applied. However, it is necessary to
have a CPV Plan for each product, even if a QbD approach has
not been applied.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 13
5.1 Brief description of the general
approach used in the A-Mab case study
Principles outlined in the ICH guidelines Q8, Q9, Q10 and • 
Univariate and multivariate approaches to define
Q117-9, 22 provide the basis for the methodology used for Proven Acceptable Range (PARs) or limits;
this case study, even though Q11 was published after the
• 
Rational approach to define a CS that reflects
A-Mab case study.
product/ process knowledge and risk mitigation;
One principle of a QbD approach is to develop a Target
• 
Process (and Equipment) Performance
Product Profile (TPP). As a natural extension of a TPP, a
Qualification to verify the CS established in Stage 1
Quality Target Product Profile (QTPP) is built to describe
of development.
quality characteristics (attributes) of the drug product.
• 
Facility design qualification of Stage 25.
The process of systematic development follows a general
roadmap that includes the following steps: In creating this CPV plan it is assumed that all deliverables
up to establishment of a CS and PQ are available based on
• 
Identification of Quality Attributes (QA)
the work described in the A-Mab study (see Figure 5.1.2 /
based on a QTPP;
green boxes). For the A-Mab process, it is assumed that
• 
Risk Evaluation to identify CQAs; PPQ was completed successfully, after investigating and
• 
Upstream/ downstream/ drug substance and resolving deviations.
product development; PPQ and Equipment Qualification (EQ) are part of Stage
• 
Risk based approaches and potentially, 2 and are therefore presumed to have been completed
multivariate analyses25 (see Section 12.5 for a before Stage 3 where CPV guidance applies. They are a
description of multivariate analysis), to classify pre-requisite for Stage 3 CPV. See guidance for Industry5.
process parameters and other variables linked
to product quality (e.g. identification of Critical
Process Parameters, CPPs);

Figure 5.1.2. Process flow of a QbD based product development according to ICH Q8, 9, 10, 11 and FDA PV guideline January 2011.

PPQ
Covered in A-Map Study Proven CPV CPV
Acceptable
TTP QTTP CQA CCP Ranges
(Design Space) Short-term Long-term
Development of Control Strategy Plan Plan
EQ

PV Stage 1 PV Stage 2 PV Stage 3

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 14
5.2 Parameters to be included in CPV (4) General Process Parameter (GPP): An adjustable
parameter (variable) of the process that does not have
All types of parameters should be considered for inclusion
a meaningful impact on product quality or process
in CPV. Typically those included will be weighted more in
performance.
favor of CPPs and WC-CPPs because of their importance
to the control strategy, but non-critical “key” and general Typically the parameters included in CPV will be weighted
parameters should not be overlooked if they are indicative more in favor of CPP and WC-CPP because of their
of process performance and/or measurably impact process importance to the control strategy. But, non-critical “key”
variation. Parameters to be included should be based on and general parameters should not be overlooked as
the current understanding of the manufacturing process they may be indicative of process performance and/or
and may be subject to change over time. measurably impact process variation. Definitions of A-Mab
terms used to define categories of process parameter are
Parameter types described in A-Mab study are as follows:
provided in a Glossary at the end of this document.
(1) Critical Process Parameter (CPP) and (2) Well-
Controlled Critical Process Parameter (WC-CPP): CPPs Note: Throughout this paper the A-Mab classification of
and WC-CPPs are process parameters whose variability process parameters is used for consistency with the structure
impact a critical quality attribute and should be monitored of that case study, but it must be recognised this is not the
or controlled to ensure the process achieves the required only scheme used in the industry; a situation arising in part
product quality. no standard approach is recommended by the regulators.
Consistency with ICH Q8 and Q11, where definitions exist
• 
A WC-CPP has a lower risk of falling outside the
seems prudent. A recent informal communication by FDA/
specified limits.
EMA counseled against using “key parameter” for describing
• 
A CPP has a higher risk of falling outside the lower levels of criticality in formal submissions and stated that:
specified limits. ‘all parameters potentially impacting product quality should be
The assessment of risk is based on a combination of factors classified as critical process parameters’23. The use of KPPs in
that include severity of impact to quality, equipment internal systems and documentation seems not to contravene
design considerations, process control capability this statement.
and complexity, the size and reliability of the proven In general, it is the responsibility of the biopharm company
acceptable range and/or design space, ability to detect/ to establish a categorization and nomenclature fitting with
measure a parameter deviation, etc. their development approach and risk evaluation tools. The
(3) Key Process Parameter (KPP): An adjustable company’s approach should be clearly explained and followed
parameter (variable) of the process that ensures over the life cycle of the product.
operational reliability when maintained within a
narrow range. A key process parameter does not affect
critical product quality attributes but rather impacts
process consistency.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 15
5.3 Upstream process overview
The upstream commercial manufacturing process for A-Mab comprises 4 steps and is summarized below and in Figure 5.4.
The A-Mab cell culture process uses a proprietary, chemically defined, basal medium formulation. The medium is essentially
protein free with recombinant human insulin (1 mg/mL) being the only protein component added. The growth medium also
contains 1 g/L pluronic and 50 nM methotrexate, which are added up to the N-2 seed bioreactor. The N-1 and production
bioreactor steps do not contain methotrexate.

Figure 5.4: Upstream process flow diagram. [Adapted from A-Mab case study, Page 62 (Figure 3.1)]

Thaw
Working Cell Bank

Step 1
Seed cultures are expanded through multiple passages
Seed Seed Culture Expansion by increasing the volume and/or number of disposable
maintenance in disposable shake flasks culture vessels. Seed cultures may be maintained for
and/or bags additional culture passages or used to generate
additional inoculums trains.
Step 2
Additional seed expansion in fixed stirred tank
Seed
Seed Culture Expansion bioreactors. Cultures obtained from Step 1 are
maintenance
in fixed stirred tank reactors expanded to increase the volume of culture to meet the
target initial cell density for the production bioreactor.

N-1 Seed Culture Bioreactor


3000L WV

Step 3
Production bioreactor is inoculated with the seed
Nutrient feed
Production Bioreactor culture prepared in Step 2 to achieve an initial Viable
15,000 L WV Cell Concentration (VCC) and is cultivated at
Glucose feeds controlled conditions for temperature, pH and
dissolved oxygen (DO). A bolus addition of nutrient
feed (NF-1) and multiple discrete glucose feeds are
used to maintain the glucose concentration at > 1.0
g/L. Antifoam solution is used for foam control of
the agitated mixture. VCC, culture viability and
residual glucose concentration are monitored
periodically. The fermentation reaction produces a
mixture containing the A-Mab drug substance.

Step 4
Cultures are clarified by a primary continuous
Harvest
centrifugation step using a disk-stack centrifuge to
Centrifugation & Depth Filtration
remove the bulk of suspended cells and cell debris.
A secondary clarification using a depth filtration
system removes remnant solids and smaller debris to
Clarified bulk provide a clarified bulk solution of A-Mab.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 16
5.4 Downstream process overview
The downstream manufacturing process for A-Mab comprises 7 steps which are summarized in Figure 5.5.
The downstream process captures A-Mab from the clarified bulk and purifies the antibody by a combination
of chromatography unit operations11. Also included in the process are two orthogonal steps dedicated to virus
inactivation and removal. The antibody is formulated through an Ultra-Filtration/Dia-Filtration (UF/DF) step
to a composition and concentration suitable for drug product manufacturing. The formulated product is 0.2 μm
filtered, filled into the appropriate storage containers and stored frozen.

Figure 5.5: Downstream process flow diagram. [Adapted from A-Mab case study, Pages 113 (Figure 4.1) and 114 (Table 4.1)]

Purpose of step

Clarified Purpose

• Capture monoclonal antibody from


Step 5 clarified harvest liquid
Protein A Affinity Chromatography • Removal of process-related impurities
(HCP, DNA and small molecules)

• Inactivate enveloped viruses that are


Step 6 potentially present in therapeutic
Low pH Incubation protein products derived from
mammalian cell culture

• Reduce aggregate to acceptable levels


for drug substance
Step 7
• Reduce HCP to acceptable levels for
Cation Exchange Chromatography
subsequent processing by AEX
chromatography

• Remove HCP, DNA, Protein A and


Step 8 endotoxins to levels that meet drug
Anion Exchange Chromatography substance acceptance criteria
• Virus removal

• Removal of small parvoviruses such as


Step 9 minute virus of mice (MVM) and larger
Small Virus Retentive Filtration viruses such as murine leukemia virus
(MuLV), potentially present in product
derived from mammalian cell culture

Step 10 • Formulation and concentration of mAb


Formulation: to drug substance specifications (e.g.
Ultrafiltration and Diafiltration A 75 g A-Mab/L)

Step 11 • Sterilize filtration and dispensing for


Final Filtration, Fill and Freeze drug substance storage

A-Mab drug substance

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 17
5.5 Identification of CQAs and acceptance ranges
Table 5.6.1 provides the QTPP of the A-Mab drug product, as defined in the A-Mab case study. The QTPP describes
quality characteristics (attributes) that the drug product should possess in order to reproducibly deliver the therapeutic
benefit promised in the label. Attributes in the red box are determined during Drug Substance (DS) manufacturing.
Therefore, these attributes guide determination of DS CQAs22 relevant for establishing a CPV strategy.

Table 5.6.1: QTPP for A-Mab (reference 3, Page 180). DS relevant product attributes are marked with a red box

Product attribute Target

Dosage form Liquid, single use

Protein content per vial 500mg

Dose 10mg/kg

Concentration 25mg/mL

Mode of administration IV, diluted with isotonic saline or dextrose

Viscosity Acceptable for manufacturing, storage and delivery without the use of special devices (for example, less
than 10 cP at room temperature

Container 20R type 1 borosilicate glass vials, fluro-resin laminated stopper

Shelf life ≥ 2 years at 2–8°C

Compatibility with manufacturing process Minimum 14 days at 25°C and subsequent 2 years at 2–8°C, soluble at higher concentration
during UF/DF

Biocompatibility Acceptable toleration on infusion

Degradants and impurites Below safety threshold, or qualified

Pharmacopeial compliance Meets pharmacopoeial requirements for parental dosage forms, colorless to slightly yellow, practically
free of visible particles and meets USP criteria for sub-visiable particles

Aggregate 0–5%

Fucose conent 2–13%

Galactosylation (%G1+%G2) 10–40%

HCP 0-100 ng/mg

The DS QAs related to the QTPP are identified as summarized in Table 5.6.2. Criticality Analysis was performed using a risk
ranking approach (as in ICH Q98) and CQAs were identified as attributes of high or very high risk regarding their potential
impact on patient safety.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 18
Step 4:
freezing

Controls
this CQA?

treatment

Bioreactor
Step 7: CEX
Step 8: AEX
“Product Quality Attributes”

Clarification
filtration and

Raw Material
Step 9: Nano-
Step 11: Final

Step 10: Ultra-

Step 6: Low pH
filtration (SVRF)

chromatography
chromatography
chromatography

Step 5: Protein A

Steps 1 & 2: Seed


filtration (UF/DF)

Culture Expansion
Step 3: Production
Centrifugation and
BDS or DP testing for
Identity Form Form BDS, DP
Protein concentration Form Alter Alter Alter Alter Alter DP IPC
ADCC activity Form DP
SEC monomer Form BDS, DP
SEC Aggregates Form Remove Form Remove Remove Form Form Form BDS, DP

©BioPhorum Operations Group Ltd | April 2020


summarized in the Table 5.6.2 below.

Color Introduce Alter Alter DP


Clarity & sub-visible particles
Introduce Alter Remove Remove DP

Deamidated isoforms Form Remove Remove Remove BDS, DP


other Acidic variants Form Remove Remove Remove BDS, DP
Charge variants Form Remove Remove Remove BDS, DP
“Oligosaccharides:
afucosylated glycans Form
galactosylated glycans”
Page 29). BDS is Bulk Drug Substance, DP Drug Product and IPC in-process control.

“Glycosylation related:
sialic acid content,
Form
mannose content,
non-glycosylated heavy chain”
Osmolality Alter DP IPC
pH Alter Alter Alter Alter Alter Alter DP IPC
Methotrexate non-
Introduce Introduce Remove Remove Remove
routine
Antifoam C non-
Introduce Introduce Remove Remove
routine
Protein A ligand Introduce Introduce Remove Remove Remove
Host Cell Protein (HCP) non-
Form Form Remove Remove Remove Remove
routine
DNA Form Form Remove Remove
Bioburden Introduce Introduce Introduce Introduce Introduce Introduce Introduce Introduce Introduce Remove DP
Endotoxin Introduce Introduce Introduce Introduce Introduce Introduce Introduce Introduce BDS, DP
“Adventitious viral agents step 3
The product quality attributes and the points where they are impacted in the A-Mab drug substance process are

(AVA)” Introduce Introduce Introduce Inactivation Remove Remove IPC, BDS


release
Table 5.6.2: A-Mab drug substance Product Quality Attributes and the points where they are impacted in the process (see A-Mab Case Study (3), Section 2.3.2,

impacted by CPP= impacted by WC-CPP= impacted by KPP= no key impact claimed= entry test or prep control=

Continued Process Verification : An industry position paper with example plan


19
5.6 Process parameter characterization
In reviewing the A-Mab process information while preparing the CPV plan, members of BioPhorum team questioned
thecompleteness of the CPPs, KPPs and Key Process Attributes (KPAs) identified in the case study. Specifically, it was
felt two steps of the downstream process (step 10 UF/DF, and step 11 final filtration and freezing of the Bulk Drug
Substance, BDS) were not addressed in sufficient detail in the case study for the purpose of developing a CPV Plan, so
typical characterization outcomes for these steps were assumed and CPPs, KPPs and KPAs were identified based on
that characterization10. In addition, two more KPPs and KPAs were identified for process steps 3 and 7, based on typical
outcomes for similar monoclonal antibody processes. The following table summarizes all CPPs, in-process quality attributes
(IPQAs), KPPs and KPAs identified for the process in preparation for CPV.

Figure 5.7: Critical and key process parameters and key process attributes identified during process characterization. Lists were amended during
planning for CPV (bold entries)

Process Step Critical Process Parameters In-process Controls Key Process Parameters Key Process Attributes

Step 1: Seed culture None None Temperature, VCC (viable cell conc), Culture
expansion in disposable Culture duration, viability
shake flasks and/ or bags Initial VCC/ split ratio

Step 2: Seed culture None None Temperature, VCC,


expansion in fixed stirred pH, Dissolved oxygen, Culture viability
tank reactors Culture duration,
Initial VCC/ split ratio

Step 3: Production Temperature, Bioburden, Antifoam conc., Product yield (titer),


bioreactor 15,000l wv pH, Mycoplasma, Time of nutrient feed, Viability at harvest,
Max partial pressure of CO2 MMV and AVA Volume of nutrient feed, Turbidity at harvest,
(pCO2), (murine minute virus and Time of glucose feed, Peak VCC, Remnant glucose
Culture duration, adventitious viral agents) Volume of glucose feed, concentration
Medium Osmolality Dissolved oxygen

Step 4: Harvest None None Flow rate, Pressure, Step yield,


centrifugation & depth Duration of clarification Turbidity
filtration

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 20
Figure 5.7: Critical and key process parameters and key process attributes identified during process characterization. Lists were amended during planning for CPV
(bold entries)

Process Step Critical Process Parameters In-process Controls Key Process Parameters Key Process Attributes

Step 5: Protein a affinity Protein load ratio, Bioburden, End collection, Step yield
chromatography Elution buffer pH Endotoxin Step duration

Step 6: Low ph incubation pH, Bioburden, Quantity of acid added


Time, Endotoxin
Temperature

Step 7: Cation exchange Protein load ratio, Bioburden, Step duration Step yield,
chromatography Wash conductivity, Endotoxin Eluate volume
Elution pH,
Elution stop collect

Step 8: Anion exchange Equilibration/ Wash1 Bioburden, Step duration Step yield
chromatography buffer conductivity, Endotoxin
Protein load ratio,
Load conductivity,
Load pH,
Flow rate

Step 9: Small virus Operating pressure, Bioburden, None Step yield


retentive filtration Filtration volume Endotoxin

Step 10: formulation: Number of Bioburden, Protein conc. prior to Step yield,
ultrafiltration and dia-volumes, Endotoxin Diafiltration, Permeate flow rate
diafiltraion pH, Recirculation flow rate
Step processing time,
Protein conc. prior to fill

Step 11: final filtration, fill None Bioburden, Filtration volume, Bulk fill step yield
and freeze Endotoxin Filtration time,
Maximum (inlet) pressure

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 21
5.7 Control strategy CQAs and CPPS
Risk-based criticality assessment, along with process
characterization studies, allows a CS to be established
which is subsequently verified during PPQ. Table 5.7
summarizes the CS established for the A-Mab upstream
and downstream process steps for A-Mab production.
The CS consists of CPPs and WC-CPPs, KPPs, KPAs and
IPQAs. The CS should ensure required product quality and
a consistent and robust process.
Here, CPPs must be controlled within limits and in-process
controls (specifically microbial and viral safety) must be
within specified ranges to ensure drug safety and efficacy.
Although KPPs and KPAs have been shown not to impact
product quality, they are included in the CS because
their monitoring and control ensures that the process
is operated in a consistent and predictable manner. The
control of KPPs and KPAs also ensures that commercial
success criteria such as cycle time and yield are met.
Product quality and safety are ensured by controlling all
quality-linked process parameters (CPPs and WCCPPs)
within the limits. Process consistency is ensured by
controlling KPPs within established limits and by
monitoring relevant process attributes.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 22
6.0
Developing a monitoring strategy
6.1 Rationale and background
CPV is a formal activity enabling the detection of variation in the manufacturing process that might have an
impact on the product quality or process consistency. CPV should evaluate whether the process consistently
delivers product with acceptable QAs and continues to operate robustly, within the validated state. It should
also identify any new sources of variability in the process that may have arisen since the initial Stage 2 PQ
was performed. For this case study it has been determined that PPQ batches will be included in CPV data
collection and analysis; indeed, all appropriate batches should be considered. CPV efforts should, where
appropriate, also focus on areas that have proved challenging or may have shifted since the initial validation.
A risk based approach to process monitoring should be used to direct these efforts. For products with a
legacy history, a defined time period or number of batches should be set to determine how much of the
historical experience will be considered. The assessment interval chosen should be sufficient to establish a
solid production history and also reflect the frequency of production. For example a product that is produced
frequently may permit a shorter time period to be used relative to a product that is produced infrequently.
In general, the points in the process to be monitored (3) The control strategy should be updated as necessary
during CPV should be comparable to, but not necessarily and hence the CPV Plan.
include all of those selected during the initial validation.
The selection of points in the manufacturing process
If limited data results are available at the time of PPQ
that are to be monitored for CPV purposes may be
completion, prior to execution of the CPV plan, a short
either a subset of those selected during PPQ or include
term sampling plan may be established to continue
additional monitoring points beyond those included in
sampling based on the PPQ protocol until sufficient data
the initial PPQ to reflect new learning obtained since
results are gathered in preparation for CPV. Additional
the initial validation was conducted.
considerations that influence the determination of which
points in the process are monitored during a CPV exercise This includes but is not limited to:
are summarized below. • New CS elements
(1) The final classification of attributes should be revisited. • 
Process elements that have proved challenging
(2) The process risk assessment, which is typically but may not have been covered during the initial
performed prior to the initial PPQ, should be revisited process validation
and updated to develop the CPV plan. The revised risk • 
Changed or additional analytical capabilities,
assessment should reflect learning obtained during including the availability of online data
PPQ, any additional laboratory process characterization collection systems and improvements in assay
information, and key findings from historical or instrument capabilities
manufacturing experience. In revisiting the process
• 
If a parameter has been shown to have good
risk assessment prior to commercial manufacture, late
control and consistency, it may not be necessary to
stage clinical manufacturing knowledge is particularly
continue monitoring this parameter in subsequent
important. Levels of risk, and indeed the range of risks,
CPV evaluations.
that apply in the manufacturing environment might be
quite different to those anticipated from the early stage
development environment.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 23
(4) CPPs, WC-CPPs, KPPs and GPPs should be clearly 6.2 Hypothetical scenarios and
specified. These parameters and established release
specifications, additional product characterization
planned process changes
testing, and KPAs should be appropriately considered Five hypothetical scenarios and planned changes are
during CPV. Any changes since the initial validation provided below to illustrate how the CPV monitoring
should be explained and justified. plan might be affected by events encountered during
commercialization of a product such as A-Mab. In
(5) All changes implemented should be assessed in
this example it is assumed that the PPQ campaign
the context of potential impact on process validation. proceeded smoothly and that the expected results
Process changes which may have occurred after the were achieved. In particular, CPPs, WC-CPPs, KPPs and
PPQ, such as vendor initiated change in a raw material, GPP are defined and achievable and the process CS is
should be handled a change control process including appropriately established. The process CS is assumed
but not limited to data trending and risk assessments, to include input raw material controls, procedural
to determine if the change has any impact on process controls, process parameter controls and monitoring,
performance and/ or product quality. These changes in-process testing, and product specification testing
may potentially require additional testing beyond that (see Figure 5.1.1). These scenarios are accounted for in
performed as part of PPQ to ensure full characterization. the CPV plan:
Such testing may be incorporated as part of CPV or may Scenario 1: Supplier change notification - culture
be handled separately as part of the company’s change medium change.
control process, depending on the nature of the change
A supplier converted to a new process to manufacture a cell
and the potential for product impact.
culture medium ingredient that may alter its performance
(6) Appropriate regulatory reporting of CPV outcomes, in the A-Mab process without impacting the material
such as inclusion in the Annual Product Review (APR), procurement specifications. No intentional changes to
must be made for any conclusions related to process composition, test requirements or certificate of analysis
assessment conducted during CPV. The CPV reports were made. The following justification for the change was
should be consistent with regulatory reporting provided:
standards, so that CPV charts may be copied and pasted
(1) Improved control of temperature during blending reduces
directly into the regulatory submissions or included
potential for degradation of the heat labile components;
as an attachment. The regulatory submissions then
provide context and unify the information presented in (2) Equipment cleaning will use robust validated cycles to
the attached CPV reports. reduce ingredient carryover risks;

(7) Other elements of Good Manufacturing Practice (3) Equipment is located in an Animal Origin Free area to
(GMP) applicable to biopharmaceutical production reduce cross contamination risks.
operations are assumed to be handled by appropriate
quality systems and are therefore outside the scope
of this document, and will not be discussed further
in the context of process validation. In particular,
acceptable microbial control is a critical element for any
biopharmaceutical process and is typically demonstrated
via initial validation efforts and then monitored as part of
routine operations.
6.2 Hypothetical scenarios and planned process changes
Five hypothetical scenarios and planned changes are
provided below to illustrate how the CPV monitoring
plan might be affected by events encountered during
commercialization of a product such as A-Mab. In this
example it is assumed that the PPQ campaign proceeded
smoothly and that the expected results were achieved.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 24
The following strategy was employed to introduce the revised Scenario 2: High Protein A leachate observed in
cell culture medium: chromatography eluate, Step 5.

• 
Determining process and quality impact for the material A PPQ batch contained 123 mg of protein A/g A-Mab in the
change through the change control process was electively Protein A pool, which exceeded the control limit for this
agreed to by process experts and quality representatives process-related impurity. Investigation revealed that:
via verification testing of culture performance and the • 
Protein A ligand released from the chromatography resin
ability to operate within the established parameters and (‘Resin A’ from Supplier A) and entered the process stream
attributes; during product elution. R&D and Supplier A confirmed
• 
A study was thus completed in the small-scale model that elevated amounts of Protein A can leach from the
from thaw through the production bioreactor to provide bead surface during an initial elution after extended
additional process characterization data and establish resin storage, even when storing under recommended
confidence in expectations of process control when the conditions;
new material lot is introduced into the commercial scale • 
Extended storage can cause increased Protein A leaching
process; in the next use cycle. The resin storage time of more
• 
Minor but statistically significant differences for KPPs than 12 months between the last clinical manufacturing
normal operating ranges and attributes (e.g. VCC, and cell batch and first PPQ batch was longer than previously
density, titer and turbidity at the end of the bioreactor experienced and was not represented in small scale trials
production) were identified at small scale; used to establish PPQ limits;

• 
Medium qualification attributes should be assessed in • 
In-process testing of the Protein A clearance will be
the change control evaluation to determine if/ how these performed to further demonstrate downstream process
attributes may be impacted. The supplier was requested capability of control of this product quality attribute (AEX
to demonstrate if a detectable mean shift in any of their Table 7.8, 10.8);
output tests could be identified with respect to their • 
The level measured in the Protein A step eluate for the
change. batch implicated by this scenario was orders of magnitude
Small scale production bioreactor material was purified below the impurity safety limit for final drug product.
downstream. No structural modifications to the protein, Also, at full scale in the affected PPQ batch, downstream
or shifts in CQAs were observed. Based on the outcome clearance of Protein A below the detectable level was
of the small scale studies, a comparison should be made to demonstrated which is consistent with small-scale
evaluate the product quality obtained at full scale, to verify observations that the subsequent chromatography steps
that no unexpected quality change has occurred and to are capable of removing Protein A (The possibility that
provide further verification of process control ranges and limits or controls on extended storage time, conditions,
performance outcomes. and/ or resin treatments may need to be considered if
data indicates the clearance capability of the process is
A CPV plan is expected to take account of this type of
not sufficiently high enough for the reader’s situation).
scenario, providing the internal policies and procedures upon
which decisions related to changes in process verification An additional Design of Experiments (DOE) study was
should be based. The change described in this scenario can be conducted after PPQ to determine the potential for Protein
addressed through the change management system and does A leaching relative to storage time, resin age (use cycles) and
not require additional sampling in the CPV plan, as routine storage conditions. Spiking study confirmation of clearance
sampling is already in place to monitor the upstream cell capabilities in the downstream process steps was achieved
growth impact of this scenario (Tables 7.1, 7.2, 7.3 and 10.1, and is discussed in the amended CS revision completed
10.2, 10.3). Potential downstream impact could be included after the PPQ experience, where the new CPPs to control
in the monitoring plan, e.g. the KPAs of inlet pressure to clearance are clearly identified. Within CPV, results will
depth filters and duration of the broth clarification, which are be monitored to detect any departures from the expected
suggested as optional items for CPV in Tables 7.4, 10.4. behavior observed during development; monitoring tools
such as ‘tool wear charts’ or ‘residuals charts’ may be useful,
Note: Attributes should only be considered optional after their
and consultation with a statistician is recommended. These
impact on the process has been risk assessed and any lack of
tools are mentioned again in Section 12.4.
monitoring fully justified.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 25
Scenario 3: High elution volume from CEX, CPV recommendations for this step (see Tables 7.7,
Step 7. 10.7) because it has demonstrated variability and there
Because of resin capacity limitations, elution of the is a theoretical potential for increased aggregates with
product stream through the CEX resin requires processing extended processing time (not observed in any studies as
a batch in multiple portions (sub-batches). The column of yet) that may result from the need for additional elution
eluate streams are then pooled. With one PPQ batch, to recover A-Mab from the CEX resin.
an unexpected additional volume of buffer solution was Scenario 4: UF/DF measurements exceeded action limits.
required to complete the elution of A-Mab from the CEX
During preparation of one PPQ batch, the starting UF/
column resin for one sub-batch. The prior wash of process
DF concentration measurements did not meet the PPQ
impurities from Cation Exchange (CEX) Chromatography
control limits and step yield was above the expected PPQ
resin proceeded without incident but there was a delay
range. The starting UF/DF concentration has not been
while the additional elution buffer was prepared (during
classified as a KPA in the A-Mab case study.
which the product loaded column was idle) before
proceeding to complete the product elution operation to • 
A change prior to PPQ revised the in-process UV
recover all the A-Mab from the resin. absorbance (A280) test method, which led to an
apparent upward shift in yield results. While a
• 
No impact on A-Mab quality was detected, which
bridging study was conducted to determine the
involved a deviation for a KPA (elution buffer
suitability of the revised test method, evaluation
volume);
of the change did not consider the impact to the
• 
The investigation did not reveal a definitive root limits used during PPQ that were calculated based
cause. Performance of the flow meter was not on earlier experience. Limits in place during
implicated as a cause of the unusual observation PPQ were based on measurements from the
from review of GPPs and instrument calibration previous version of the method used for in-process
checks; monitoring.
• 
Flow channeling through the resin was the initial • 
Change control improved the accuracy of the
suspected cause, but no similar observation was measurement and also removed a bias error
made during earlier or later PPQ sub-batches; when compared to the final bulk drug substance
• 
Delay in starting the elution operation may have concentration which uses a different method
played a role, but this could not be confirmed performed in the QC release testing laboratory.
because it had not been specifically studied, nor did • 
The implemented change in the test method
delays after load prior to elution occur in historical involved improvements to both the precision and
small-scale studies; accuracy of the in-process measurement system;
• 
Similar incidents have not been observed with there has been no change to the UF/DF process.
other A-Mab batches at any scale studied; A Analytical SME’s decided it would be inappropriate
change in the buffer (e.g. conductivity which is not to compare new results to a set of limits based
a CPP, or pH which is a CPP) as a result of the delay on data measured using a different/ altered
has not been conclusively eliminated as a cause, procedure, or simply adjust previous results for a
but no deviation associated with the buffer was fixed bias correction (due to potential proportional
apparent from careful scrutiny of the batch record variance, see section 12.4).
(BRc) and interview with process operators. • 
The corrective action being implemented will
supersede the original PPQ limits with new CPV
Investigation of elution buffer stability data is also
limits calculated using data from the revised test
suggested. If insufficient hold time and buffer attribute
method procedure.
data exists to determine the potential for buffer stability
to be a contributing cause, this may be pursued as an No monitoring recommendations for CPV are proposed
independent study, rather than including buffer chemical as a result of this scenario. Care should be taken not to
stability in the CPV Plan. Tracking of buffer volume used include data generated prior to the method change in
to elute A-Mab from the CEX column is included in the calculating long-term limits.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 26
Scenario 5: Environmental monitoring during Bulk Drug
Substance (BDS) fill, Step 11.
During environmentally controlled open system filling
of one BDS batch, the routine sentinel plates indicated
environmental monitoring (EM) bioburden was above the
PPQ action limits. Investigation determined that:
• 
Based on organism identification, the likely source
was skin flora shed by an operator who conducted
the final filtration and filling of the BDS;
• 
The bioburden samples of each post-filtration
product container (for the PPQ) met the
acceptance criteria with results of 0 CFU/ 10 mL.
This confirmed that the 0.2 μ m filtered BDS was
not impacted and the routine criteria were met for
batch release (BR);
• 
Following the filling operation, BDS is frozen
within 24 hrs, and once thawed, the material is
pooled, mixed, and sampled for bioburden prior
to sterile filtration when initiating drug product
manufacturing;
• 
Corrective and preventive actions have been
implemented, including a review of personnel
practices, skills and training, and changes to
operating procedures to alert operators to
use appropriate practices when working in the
controlled filling environment.
No additional monitoring recommendations for CPV
are proposed as a result of this scenario because, even
with this incident, no impact to the BDS was found and
corrective actions have been implemented to prevent
its recurrence. Routine monitoring is sufficient. No
addition to the enhanced monitoring plan is needed
because it is not reasonable to expect from a single
incident that there will be variability in bioburden results
due to the processing of this step. Note: Whilst attributes
and parameters that are included in a CPV Plan are likely
to include some that are relevant to BR, a CPV program
is expected to operate independently of BR processes and
procedures. Analysis of data within the CPV program is
not expected to have an impact on product that has been
previously released. The release of batches compares batch
quality and performance to a specific set of pre-determined
specifications and other measures. In contrast, the focus
of CPV is to reveal trends and sources of variation in
batch quality and performance that already fall within the
predetermined criteria for BR.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 27
7.0
CPV plan recommendations for the
A-Mab process
This section describes, for each of the A-Mab process steps, what to include in the CPV plan and the
justification for its conclusion. This justification is primarily based on process knowledge and process
experience. A table is provided for each step to summarize the recommendations for CPV. Discretionary
items are also included that may be needed in a CPV program depending on the assurance of process
understanding or that provide additional depth to the monitoring plan.
No recommendation for including in-process product Steps that have in-process quality attributes related to
pool hold times in CPV is proposed, because the hold microbial control (bioburden, endotoxin) are sampled and
times were validated as part of the basis for controls tested as routine in-process controls. The nature of test
within the Master BRc. In the event that a hold time is results in this case (approximately 0 cfu/ sample, and ≤
exceeded this one-off event would trigger a deviation Limit of Quantification, LOQ, respectively) do not permit
within the Quality System, under which impact to meaningful Statistical Process Control (SPC) analysis in
product quality would be determined. CPV. QC microbiology laboratory review of these results
In the steps with elution of product from resin beds against action and alert limits will provide appropriate
(i.e. steps 5 and 7), several resin loading/ elution cycles monitoring for drift in microbial control of the process and
are used to process each batch. No controls have been management of deviations, so monitoring, data analysis
identified for resin regeneration operations in either of and any response to bioburden and endotoxin results are
these steps. For these steps, concurrent validation of not included under this CPV plan.
the resin use lifetime includes periodic sample testing of Note: It could be seen as best practice that the quality system
appropriate quality attributes for continued verification for bioburden and endotoxin monitoring and the CPV system
of packed resin effectiveness during its use lifetime. are connected, so that any deviations would be reflected in
Effectiveness of resin regeneration conditions is included CPV Reports.
in the ongoing resin use validations. Therefore monitoring
Statistical criteria that may be applied to analyses of data
of CQAs for this purpose need not be included in the CPV
are discussed in section 12.
plan. Continued monitoring, and further verification of
effective process controls, should be considered for CPV The A-Mab case study did not identify any critical raw
when resin use lifetime monitoring ceases, if further data materials or address CS or risk assessment for input
are needed for understanding of impurity clearance. material controls. However, as a result of a hypothetical
culture medium change described in section 6, one
No recommendation for including in-process hold times
monitoring recommendation related to material variability
in CPV is proposed because ongoing study of hold times
is provided as a recommendation for the CPV plan.
during commercial manufacturing is conducted using a
Additional monitoring of materials used in the bulk drug
separate hold time qualification study.
formulation is also included as an option.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 28
7.1 Step 1, seed culture expansion in disposable vessels –
CPV recommendations
The process risk assessment established that steps 1 and 2 the seed expansion steps are routinely monitored and
of the A-Mab process do not entail risk of impact to product operated within established limits. Therefore, monitoring
quality in the production bioreactor because no product of non-critical parameters in this step such as temperature,
is accumulated at these stages. Specifications for raw pH, and dissolved oxygen need not be included in the CPV
materials, such as cell banks and media components, assure plan. This is shown in Table 7.1 below.
use of the intended genetic cell line to produce A-Mab and
Environmental Monitoring (EM) is routinely performed
control introduction of endotoxins which could affect cell
for open (under appropriate ISO classified conditions)
metabolism.
process manipulations (including use of Rodac and settling
CPV for this step should focus on process consistency and plates) to demonstrate microbial control and the existing
obtaining sufficient data to calculate long-term control QC laboratory program is established for reporting results
limits (see Sections 9.0 and 10 for further discussion and and assessing trends. Therefore inclusion of this EM
examples of control limits, and Section 12 for information monitoring plan in the CPV plan is unnecessary. As noted
on the statistical basis for control limits. which account for previously, these systems need to connect as it would
normal process variability. As stated in the A-Mab case be best practice to ensure deviations are present in CPV
study and demonstrated in the PPQ, BR procedures, SOPs, Reports.
automated process controls and use of alarms all ensure

Table 7.1. Step 1 CPV recommendations

Variable Class CQAS CPV recommendation & Determination method Type of data expected/
impacted justification and/or source analytical approach

VCC — Include, to verify process Routine batch documentation for each Discrete value,
KPA
(each passage end) consistency passage univariate

Optional elements to include in CPV

Initial VCC/split ratio — Optional, to verify process Calculation from routine batch Discrete value,
KPP
(each passage) consistency documentation for each passage, ratio of multivariate
passage ending cell density over initial cell
density of next passage.

Culture duration — Optional, to verify process Routine batch documentation for each Discrete value,
KPP
(each passage) consistency passage. univariate

Culture viability Optional, to verify process Routine batch documentation for each Discrete value,
KPA
(each passage end) consistency passage. multivariate

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 29
7.2 Step 2, Seed Culture Expansion in Bioreactors – CPV Recommendations
As noted in section 7.1 for step 1, cell growth is complex and it is difficult to comprehensively define or predict all sources
of variability. Expansion culture conditions may impact cell biology which in turn can impact product quality during product
expression. CPV for this step should focus on process consistency and obtaining sufficient data to resolve long-term control
limits which account for normal process variability.
Inclusion of EM in the CPV plan is unnecessary because an existing QC program is established for reporting and trending of
EM results.

Table 7.2. Step 2 CPV recommendations

Variable Class CQAS CPV recommendation & Determination method Type of data expected/
impacted justification and/or source analytical approach

VCC — Include, to verify process Routine batch documentation for each Discrete value,
KPA
(each passage end) consistency passage univariate

Optional elements to include in CPV

Initial VCC/split ratio — Optional, to verify process Calculation from routine batch Discrete value,
KPP
(each passage) consistency documentation for each passage, ratio of multivariate
passage ending cell density over initial cell
density of next passage.

Culture duration — Optional, to verify process Routine batch documentation for each Discrete value,
KPP
(each passage) consistency passage. univariate

Culture viability Optional, to verify process Routine batch documentation for each Discrete value,
KPA
(each passage end) consistency passage. multivariate

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 30
7.3 Step 3, Production Culture Harvest attributes of titer, viability, and culture duration
are also included for trend monitoring to verify process
Bioreactor – CPV Recommendations outcome consistency.
The process risk assessment established the BDS
The Partial Least Squares (PLS) multivariate model
CQAs that may be impacted by this step. As stated in
generated during process characterization in the A-Mab
the A-Mab case study and demonstrated in PPQ, BR
case study [3, Section 3.10, Page 108]25, includes other
procedures, other SOPs, automated controls, and an
CPPs (e.g. dissolved oxygen, pressure, gas addition rates)
alarm system all ensure the production step is routinely
and KPAs (e.g. VCC, viability), noted in Table 7.3.
monitored and operated within established limits for
many of the related parameters. The CPV plan may optionally include selected KPPs and
KPAs to provide additional measurements of robust
Turbidity at harvest (a KPA known to vary in response
process consistency and to obtain sufficient data to
to bioreactor culture conditions) is not included in the
resolve long-term control limits that account for normal
CPV plan because of the confidence that centrifugation
process variability. Two suggested discrete KPAs, peak
and depth filtration can accommodate variability in
VCC and culture viability at harvest, are optional in
the harvest material (low differential pressure across
Table 7.3 for Step 3. Other KPAs (glucose and lactate
depth filters). However, if there is a filter change, or the
concentrations) are also included as part of the PLS model
medium component change introduced between PPQ
described in the A-Mab case study.
and commercial manufacturing indicates a shift in other
monitored variables for this step or process performance In-process quality attributes for this step, namely
of the next step, establishing control limits for turbidity at bioburden, Murine Minute Virus (MMV), mycoplasma,
the end of the production bioreactor should be added to and Adventitious Viral Agents (AVA) are controlled
the CPV plan. as routine in-process specifications linked to drug
substance BR. Their binary pass/ fail nature does not
The CPPs for medium osmolality and culture duration
permit meaningful SPC trend monitoring, and does
are included in the CPV plan. For these CPPs, a large
not provide prospective warning of pending batch
tolerance for variation has been shown in development
failures. These routine control measures are sufficient
during process characterization studies. Maximum pCO2,
for maintaining the process in its validated state and
bioreactor temperature and bioreactor pH are other
deviations detected will trigger investigations for out of
identified CPPs to be included in the CPV plan.
control situations/events.
At the time this protocol is initiated, the PLS model
Regarding the productivity of the production culture step,
is classified as a KPA; it is a predictor of A-Mab
whether to include the Antibody-Dependent Cellular
oligosaccharide structure CQAs and acidic variants. Model
Cytotoxicity (ADCC) bioassay in the CPV plan or not,
input parameters of temperature and pH, and model
is an interesting and somewhat complicated question.
input attributes of titer, VCC, and viability are separately
ADCC is correlated with afucosylation in vitro. Thus
included in the initial monitoring while the bioreactor
measurement of potency by ADCC is an indicator for this
model is qualified.
quality attribute that might impact Fc effector function.
Remnant glucose concentration is not included in the CPV However, this bioassay is not qualified to test crude
plan because it is assumed to be a fixed value CPP which production bioreactor material just prior to harvest due to
triggers additions of glucose feed. However, as an attribute broth interference. Fundamentally, that would not prevent
of the culture, it is measured daily and when the glucose reliable results that correlate with the potency of the
concentration drops below a particular level, a discrete purified material. But, confirmation of functional activity
volume (assumed to be a fixed KPP) of a glucose solution is relevant to the finished dosage form since it is the drug
is added as a bolus to ensure the glucose concentration product that is provided to the patient. So, monitoring of
remains ≥ 1.0 g/L. A fixed volume of nutrient feed is added bioreactor harvest for potency is not recommended since
at a defined time under automation and routine batch ADCC activity has a drug product release specification for
document controls, therefore trending of the KPPs nutrient CPV trending and is a stability indicating assay included
feed volume and timing of nutrient feed does not provide in routine stability testing protocols [derived from A-Mab
value because they are not subject to random variation. case study section 6.4.2, Page 247].

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 31
The CS categorized the antifoam ingredient to be a CPV trend monitoring of afucosylated and galactosylated
critical raw material (CRM), probably because it is a glycans in the bioreactor for step 3 is recommended to
process residual CQA. However, there is no particular build confidence in process consistency (see Table 7.12,
critical material attribute (CMA) that requires enhanced 10.12). Note that sialylation, high mannose content (also
monitoring. The addition of antifoam varies as needed up afucosylated) and non-glycosylated heavy chain were
to a maximum 100 mg/L concentration (section 5). As a also determined to be CQAs but recommended only as
single antifoam lot is used for multiple bioreactor batches, optional elements to include in CPV monitoring (see Table
lot change points in the batch genealogy will be traceable 7.12, 10.12). The frequency of lifecycle monitoring of
to correlate with any process shifts in trends for this step, glycans will be reviewed and adjusted based on trends.
the clarification step, or the Protein A chromatography Characterization of the oligosaccharide profile will be
step (steps 3, 4 and 5 in the process). Because only one conducted to confirm comparability when needed to
lot of antifoam was introduced in the PPQ, three BDS support process changes [derived from A-Mab case study
batches during the initial CPV period, which employ section 6.4.5, Page 250 and section 6.6.1, Page 251-253].
different antifoam lots in the upstream process, are tested
The mechanism and conditions conducive to formation
to provide evidence of robust clearance of the process
of deamidated isoforms are widely known and well
residual. Due to the low turnover in antifoam lots, routine
understood. This knowledge, in conjunction with the
but periodic batches being tested for stability may also
level of risk associated with the quality attribute in the
be selected for this extra testing in BDS, i.e. at ‘time zero’.
post-PPQ risk assessment, negates the need for in-process
This does not suggest that clearance of antifoam is a
CPV testing. Process control includes testing with a
stability indicating attribute.
routine CEX HPLC method at lot release, of both drug
The oligosaccharide profile (a CQA) is solely influenced substance and drug product, to confirm the identity of
by the production bioreactor. Input material and A-Mab, monitor charge heterogeneity and detect shifts
procedural controls are in place to ensure the quality of in deamidated isoforms [derived from A-Mab case study
raw materials and the cell line. Control of step 3 CPPs section 6.6.4, Page 259]. The method separates the main
(temperature, pH, dissolved carbon dioxide, culture charged isoforms of A-Mab that are considered to be
duration, and medium osmolarity) within their limits product-related substances as defined in ICH Q6B. The
ensures consistent glycosylation. No process clearance resulting chromatographic profile is specific to A-Mab and
or further glycan modification occurs in downstream unambiguously distinguishes it from other monoclonal
processing, and the oligosaccharide profile is not regarded antibodies. The spectrum of isoforms contained in the
as stability indicating. Routine testing is not part of reference chromatogram for A-Mab represents acidic and
the drug substance lot release specification based on basic isoforms. The chromatogram is inspected to ensure
the development process design history, process risk a consistent profile with the reference standard and the
assesments, CS, and PPQ. The risk that exists is that no absence of any new peaks. A quantitative definition of
process clearance or further modification is expected in new peaks is included in the CEX test method. Charged
downstream processing. An oligosaccharide profiling isoforms of A-Mab do not increase when stored at
method utilizing Capillary Electrophoresis-Laser Induced recommended conditions; therefore, the attribute is not
Fluorescence (CE-LIF) was developed and qualified for monitored on stability [derived from A-Mab case study
characterization of the oligosaccharide profile. There section 6.4.2, Page 247].
is also an in vitro cell-based bioassay qualified to enable
The KPA of titer (yield) is included in CPV for trend
collection of biological activity data related to ADCC
monitoring for process consistency.
functions, as a means to assess Fc-oligosaccharide
structure-function relationships.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 32
Table 7.3. Step 3 CPV recommendations

Variable Class CQAS impacted CPV recommendation & Determination method Type of data expected/
justification and/or source analytical approach

Culture duration Aggregates, glycosylated Include, to establish Routine batch documentation Discrete value,
CPP
glycans, HCP, DNA; can SPC capability and large univariate
also impact turbidity at tolerance for variation
harvest, yield variation

Maximum Glycosylated glycans, Include, to establish SPC Routine batch documentation Discrete value,
CPP
(dissolved) pCO2 deamidated isoforms; capability and correlate with univariate
also product yield in-vitro cell age (IVCA)

(Bioreactor) Glycosylated glycans, Include, to demonstrate Routine batch documentation Continuous datastream,
CPP
temperature deamidated isoforms appropriate range is univariate
established

(Bioreactor) pH Glycosylated glycans, Include, to demonstrate that Routine batch documentation Continuous datastream,
CPP
deamidated isoforms appropriate monitoring and univariate
automated adjustments are
established

Afucosylated — Include, to verify process Will require non-routine test, Discrete value,
CQA
glycans consistency record results in Laboratory univariate
Information Management
System (LIMS)

Galactosylated — Include, to verify process Will require non-routine test, Discrete value,
CQA
glycans consistency record results in LIMS univariate

PLS model Isoforms, variants, DNA, Include, to verify process Routine batch documentation Continuous datastream,
KPA
employing pH, monomer, aggregates, consistency multivariate
DO, temperature, HCP oligosaccharides
pressure, gas
rates, weight, VCC,
viability, titer,
glucose, lactate

Product yield (titer — Include, to verify process QC ELISA results in LIMS Discrete value,
KPA
at harvest) consistency univariate

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 33
Table 7.3. Step 3 CPV recommendations (continued)

Variable Class CQAS impacted CPV recommendation & Determination method Type of data expected/
justification and/or source analytical approach

Antifoam lot Residual antifoam C Include, to track lot Routine batch document Qualitative text/label,
CMA
changes. Test clearance at genealogy univariate
BDS for 3 different lots

(Medium) osmolality Glycosylated glycans, Include; large tolerance for Routine batch documentation Discrete value,
CPP
deamidated isoforms variation has been shown. univariate
Monitor by exception a

Mannose content — Optional, to verify Will require non-routine test, Discrete value,
CQA
process consistency record results in LIMS univariate

Sialic acid content — Optional, to verify Will require non-routine test, Discrete value,
CQA
process consistency record results in LIMS univariate

Non-glycosylated — Optional, to verify Will require non-routine test, Discrete value,


CQA
heavy chain process consistency record results in LIMS univariate

Time of glucose feeds — Optional, to verify Routine batch documentation Discrete value,
KPP
(hrs since inoculation) process consistency univariate

Peak VCC — Optional, to verify Routine batch documentation Discrete value,


KPA
process consistency univariate

(Culture) viability — Optional, to verify Routine batch documentation Discrete value,


KPA
at harvest process consistency multivariate

Key: a: The term “monitor by exception” means that reported data outside of established alert or action limits will be reported as incident(s); for CPV, a review of
reported incidents will examine the occurrence of any events outside of established limits and determine the collective impact of these events.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 34
7.4 Step 4, Clarification (centrifugation and depth filtration) –
CPV recommendations
The process risk assessment established that the clarification step is unlikely to impact product quality. Monitoring for CPV
is limited to process consistency for the purpose of examining data to establish long-term control limits that account for
normal process variability (per statistical confidence criteria stated in section 12). Therefore, the KPA of yield is included in
CPV for trend monitoring as a process performance indicator. One KPA, turbidity of filtrate, is also recommended to confirm
process consistency following the culture medium change (see section 6.2, scenario 1).
The PPQ demonstrated that BR procedures, SOPs, automated process controls and alarming ensure the centrifuge and
filtration step are routinely monitored and operate within established limits. Temperature, centrifuge feed rate and rpm, and
filter flow rate are not CPPs and are tightly controlled engineering or fixed design parameters that are not subject to random
variation and therefore do not merit inclusion in CPV.
Evaluation of a change in the manufacturing method of the culture medium used upstream (see Section 6, Scenario 1) could
include additional monitoring of downstream KPAs of inlet pressure to depth filters and duration of the broth clarification,
which are noted in the Table 7.4 as optional items for CPV. The small scale model evaluation of new lots of the medium
material showed that product quality attributes of Host Cell Protein (HCP), DNA, and product structural characteristics are
not impacted, so monitoring of these KPAs is not included in the CPV recommendations. The decisions not to include these
KPAs in CPV could be re-examined pending the results of the change management evaluation of the culture medium change.

Table 7.4. Step 4 CPV recommendations

Variable Class CQAS impacted CPV recommendation & Determination method Type of data expected/
justification and/or source analytical approach

Turbidity (of filtrate) Glycosylated glycans, Include, to confirm process Non-routine testing needed Discrete value,
KPA
deamidated isoforms consistency following medium univariate
change

Step yield — Include, to verify process QC ELISA test results in LIMS Discrete value,
KPP
(product in filtrate) consistency univariate

Optional elements to include in CPV

Duration of broth — Optional, to confirm process Routine batch documentation, Discrete value,
KPP
clarification consistency following medium elapsed time from start of univariate
change harvest (opening of bioreactor
bottom valve) to end of
filtration (closing or filtrate
vessel inlet valve)

Inlet Pressure to — Optional, to confirm process Routine batch documentation Continuous datastream
KPP
filters consistency following medium or discrete value,
change univariate

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 35
7.5 Step 5, Protein A the expected control and minimal variation for these key
parameters. Operating temperature and other GPPs
Chromatography – were shown in characterization studies to not impact
CPV recommendations product quality or process consistency when controlled
The process risk assessment established that protein within easily achieved design ranges. The automated
A purification may impact product quality (aggregate; continuous process controls and alarm system, as well
charge variants; leached Protein A; clearance of HCP, as BR sequencing and SOPs, ensure the step is routinely
DNA, and methotrexate) and links to performance of monitored and operated within its established limits.
chromatography steps 7 and 8 (CEX and AEX). Platform A linkage model study was proposed in the A-Mab case
and prior process knowledge negate the need for specific study to examine HCP levels at different points within
process studies except as noted below for HCP and the process (after each chromatography step – steps 5, 7
leached Protein A. and 8). This is included in the CPV plan and involves non-
CPV for step 5 (the first of the downstream DS process routine analysis to provide data on measured HCP levels
steps) should focus on process consistency to obtain at the final point in the process covered by the multivariate
sufficient data to establish long-term control limits that model (after AEX chromatography, step 8) against the
account for normal process variability (see section 12 for predicted outcome.
statistical confidence criteria). Variables recommended Storage of the Protein A resin (section 6, scenario 2) is
for inclusion in the CPV plan are shown in the Table 7.5 expected to potentially introduce a variable amount of
below, including CPPs identified for this step (protein load leached Protein A into the product stream. This will be
ratio and elution buffer pH). Elution buffer pH is closely monitored via residual protein A (leached from the resin)
controlled by batch procedure and buffer is not released testing and trending of the results to establish process
for use if pH is out of range. This variable is included in the capability for controlling this CQA.
CPV plan to monitor the extent of buffer pH variability
Process control deviations for this step should evaluate
incorporated in the HCP model prediction. Step duration,
the case-by-case potential impact on these attributes
a KPP, is included in the CPV recommendations to
(and viral clearance), as process streams continue
establish capability on processing time for this step.
further downstream for purification. For deviation
The key process attribute of yield is included in investigations, it may be appropriate to review the risk
the CPV recommendations for trend monitoring of assessment justification for any low risk CPPs. Note that
process consistency. the process has high Impurity Safety Factor (ISF) (for a
CPV need not include monitoring of flow rate through definition, see A-Mab Case Study3 Section 4.10.3, Page
the resin, nor the end collection point (column volume or 167) clearance (>5x104) for all process related impurities
A280 absorbance) for the eluate because the PPQ verified for normal processing.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 36
Table 7.5. Step 4 CPV recommendations

Variable Class CQAS impacted CPV recommendation & Determination method Type of data expected/
justification and/or source analytical approach

Protein load (ratio) HCP, DNA, process Include, to establish SPC Routine batch documentation, Discrete value,
CPP
(in HCP model), each impurities capability calculated using packed resin univariate
sub-batch volume

Elution buffer pH (in HCP, DNA, process Include, to verify process Routine batch documentation Discrete value,
CPP
HCP model) impurities consistency univariate

Residual Protein A in Protein A Include, to establish SPC Non-routine testing needed, Discrete value,
CQA
eluate pool capability results recorded in LIMIS univariate

Step duration — Include, to verify that Routine batch documentation, Discrete value,
KPP
process can capably control elapsed time from closing of univariate
this CQA vessel inlet valve to eluate
pooling is completed

Step yield — Include, to confirm process Routine batch documentation, Discrete value,
KPA
consistency calculation using in-process multivariate likely
A280 test result to exhibit normal
distribution

7.6 Step 6, Low pH treatment – manually controlled and susceptible to variation within
their PARs. One additional test, for aggregates, is also
CPV recommendations recommended for CPV to establish process capability for
The process risk assessment established that the low pH this CQA.
treatment step for viral inactivation impacts two product
CPV need not include inactivation temperature and
CQAs (aggregate and viral inactivation). There is no claim
agitation mixing because PPQ verified the expected
for removal of process related impurities (HCP, DNA,
control and minimal variation for these key parameters.
methotrexate or leached Protein A) but some incidental
BR sequencing, automated process controls and the alarm
reduction in these impurities may be achieved in this
system will ensure the step is routinely monitored and
step, which includes precipitation and downstream filter
operated within its established limits for these parameters.
clearance. In general, CPV for this step should focus on
obtaining sufficient data to resolve long-term control The limit for maximum protein concentration in the
limits related to viral inactivation, so CPPs for inactivation Protein A pool is bound by the pH inactivation step
time and pH should be included in CPV. The viral safety requirements, but trending of the protein concentration
risk CQA (inactivation of particular AVA) for the A-Mab is not recommended as the information will provide little
process has been validated in the small scale model during benefit in process understanding. However, a related
stage 1 process validation. Inactivation time and pH are optional inclusion for CPV is to trend the amount of acid
readily controlled within desired limits for the process added, to ensure the A-Mab process does not drift or shift
as shown by PPQ. However, inclusion of both these toward the edge of the qualified conditions of the platform
parameters in CPV is recommended, because they are process without this being recognised.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 37
Step yield is not included as a CPV recommendation because yield is not expected to be impacted at this point in the
process and variability around the expected 100% has been associated with measurement uncertainty rather than process
variability, therefore it does not merit CPV trending or monitoring. The basis for yield for the next step (7, CEX) begins from
the eluate pool of the previous step (5, Protein A).

Table 7.6. Step 6 CPV recommendations

Variable Class CQAs CPV Recommendation & Determination Method and/ Type of data expected/
impacted Justification or source Analytical approach

pH (during inactivation) AVA, Include, to confirm process Routine batch documentation, Continuous datastream,
CPP
aggregates consistency integrated average of online pH univariate
values during inactivation time

Post-inactivation — Include, to establish SPC Non-routine testing needed, Discrete, multivariate


CQA
aggregates capability results recorded in LIMS

Optional elements to include in CPV

(Inactivation) time AVA Optional, to establish SPC Routine batch documentation, Discrete value, univariate
CPP
capability calculate from completion
of acid addition to start of
titration

Quantity of acid added — Optional, to establish SPC Routine batch documentation, Discrete value, univariate
KPP
capability change in supply vessel weight

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 38
7.7 Step 7, Cation Exchange volume, nor the starting or end collection point
(A280/A320) for the eluate because PPQ verified
Chromatography (CEX) – CPV the expected control and minimal variation for
recommendations these key parameters. BR sequencing, automated
The process risk assessment established that the CEX process controls, and the alarm system will ensure
step primarily impacts two product CQAs, aggregate the step is routinely monitored and operated within
and residual HCP. Characterization studies showed that its established limits for the independent parameters.
DNA and protein A clearance were not impacted by this Development studies concluded a wide operating
step, and there is no claim of viral clearance for this step. temperature range had no impact on product quality
Trending of CPPs identified as potentially impacting or performance/ process consistency, so monitoring
these CQAs are included as recommendations for CPV. of temperature beyond that routinely done for each
batch is not included in CPV recommendations.
CQAs for HCP (as supporting evidence for the linkage
model prediction) and aggregate are included in CPV CEX eluate volume (each sub-batch) is included in CPV
to establish process capability, and the KPA of yield to determine a higher confidence range of the normal
is included in CPV for trend monitoring as a process variation due to the special cause event that occurred
performance indicator. during PPQ (see section 6.2). Data obtained will be used to
show process consistency with respect to this parameter.
Univariate monitoring is not required for flow rate
through the resin, elution buffer pH, load buffer pH, The basis for yield for this step begins from the step 5
wash buffer pH, re-equilibration buffer pH, eluate eluate pool.

Table 7.7. Step 7 CPV recommendations

Variable Class CQAS CPV recommendation & Determination method and/ Type of data expected/
impacted justification or source analytical approach

Protein load (ratio) (in Aggregates, Include, to establish SPC Routine batch documentation, Discrete value, multivariate
CPP
HCP model) HCP capability calculation using packed resin
volume

Wash conductivity (in HCP Include, to confirm process Routine batch documentation Discrete value, univariate
CPP
HCP model) consistency

Elution pH HCP, DNA, Include, to confirm process Routine batch documentation Continuous datastream,
CPP
Protein A, consistency univariate
aggregates

Aggregates in CEX eluate – Include, to verify process Will require non-routine in- Discrete value, multivariate
CQA
pool performance process test, results recorded
in LIMS

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 39
Table 7.7. Step 7 CPV recommendations

Variable Class CQAS CPV recommendation & Determination method and/ Type of data expected/
impacted justification or source analytical approach

HCP content in CEX – Include, to verify process Will require non-routine Discrete value, univariate
CQA
eluate pool performance in-process test, record results
in LIMS

CEX eluate volume (each – Include, to confirm process Routine batch documentation Discrete value, univariate
KPA
sub-batch) consistency

Step yield – Include, to confirm SPC Routine batch document Discrete value, multivariate
KPA
capability calculation using field A280
test result

Optional elements to include in CPV

Step duration – Optional, to confirm SPC Routine batch documentation; Discrete value,
KPP
capability elapsed time from end of step 6 univariate
(vessel inlet valve closes)

7.8 Step 8, Anion Exchange Monitoring of step duration (a KPP) is suggested as an


optional inclusion for measuring process capability.
Chromatography (AEX) – Inclusion of other process parameters including flow
CPV recommendations rate (a CPP) and KPPs such as starting or end collection
The process risk assessment established that the AEX UV for the eluate, or pH of the prepared equilibration/
step impacts several CQAs (viral clearance, aggregate, wash 1 buffer are not suggested because PPQ verified
endotoxin, and clearance of protein A, charge variants, the expected control and minimal variation for these
HCP, DNA, and methotrexate). Trending of three CPPs parameters. BR sequencing, automated process controls,
identified as potentially impacting these CQAs (protein and the alarm system will ensure the step is routinely
load ratio, equilibration buffer conductivity and load pH) monitored and operated within its established limits for
are included as recommendations for CPV. these independent parameters. Development studies
concluded that a wide protein concentration range had
Monitoring of other CQAs impacted by this step is not
no impact on product quality or performance/process
recommended because trending for HCP and Protein A are
consistency, so trending of protein concentration is also
sufficient to represent the performance and establish the
not included in CPV recommendations.
capability of this step. Since the step 5, 7, 8 linkage model
(see Section 12) is for predicting an impurity CQA (residual Monitoring of step yield will serve as an indicator of any
HCP), the output of the model is classified as a KPA, and drift in process control for this step.
is also included for monitoring against CPV control limits
(not BR acceptance criteria).

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 40
Table 7.8. Step 8 CPV recommendations

Variable Class CQAS CPV recommendation & Determination method and/ Type of data expected/
impacted justification or source analytical approach

Protein load (ratio) HCP, viral Include, to establish SPC Routine batch documentation, Discrete value, univariate
CPP
clearance capability calculation using packed resin
volume

Load conductivity Viral Include, to confirm process Routine sample and test for Discrete value, univariate
CPP
clearance consistency buffer use

Load pH (in HCP model) HCP, viral Include, to confirm process Routine batch documentation, Discrete value, univariate
CPP
clearance consistency sample test

Equilibrium/ Wash 1 HCP, viral Include, to verify process Routine sample and test for Discrete value, multivariate
CPP
buffer conductivity (in clearance performance buffer use
HCP model)

Linkage model output – Include as outcome of HCP Calculated from six variable Discrete value, multivariate
KPA
for HCP content in AEX linkage model, to demonstrate terms logged in batch
eluate (predicted) understanding of HCP documents for step 5,7,8
clearance through multiple
processing steps

HCP content in AEX – Verify model of HCP clearance Non-routine test, results Discrete value, univariate
CQA
eluate (measured) through multiple processing recorded in LIMS
steps

Residual Protein A in – Include, to confirm process Will require non-routine Discrete value, univariate
CQA
eluate consistency in-process test, record results
in LIMS

Step yield – Include, to confirm SPC Routine batch document Discrete value, multivariate
KPA
capability calculation using field A280
test result

Optional elements to include in CPV

Step duration – Optional, to confirm SPC Routine batch documentation; Discrete value, univariate
KPP
capability elapsed time from end of step 7
(vessel inlet valve closes)
until product elution completed
in step 8

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 41
7.9 Step 9, Small Virus Retentive impact on processing time for this step. Verification of
the filter integrity testing was included in PPQ and will be
Filtration (SVRF) – included upon completion of the filtration of every process
CPV recommendations batch. Re-filtration has also been validated and details
SVRF is a physical size separation step that is critical for are registered in regulatory licenses. Detectable impact
viral clearance. Filter function is confirmed after each batch of re-filtration is a decrease in the measured protein
by standardized integrity testing. Introduction of leachate concentration due to dilution by a hold-up recovery flush
from the filter is minimized by a routine pre-use rinse of the after filtration to optimize step yield. Incidents of failed
filter with a validated quantity of AEX elution buffer. filter integrity and/ or when re-filtration is performed are
tracked with the change control system as incidents and
Operating pressure is a WC-CPP recommended for
are trended as part of Annual Product Review, so will not
including in the CPV plan. Some variation in pressure has
be included in CPV.
been observed; trending of pressure data will increase
predictability and confidence in knowledge of the natural Step yield is not included as a CPV recommendation
variation and what, if any, impact this variation may have because yield is not expected to be impacted by this step.
on process consistency. Correlation of operating pressure Yield after step 10 will include step 9.
with filtration volume (the other CPP to be monitored) Rinse or processing flow rate through the filter and the
and protein concentration will also ensure consistent flush volume used are not recommended for inclusion
viral Log Reduction Value (LRV) and serve as a basis for in CPV, because PPQ demonstrated tight control and
future process improvements/change controls. Small- minimal variation of these KPPs. Although these variables
scale studies have shown that the likely variation in these are manually controlled, the BR instructions and sequence
parameters does not represent a BR risk for product safety will ensure the step is routinely monitored and operated
or quality. Including filtration load volume in CPV provides within its established limits.
an alternate measure of process consistency, given its

Table 7.9. Step 9 CPV recommendations

Variable Class CQAs CPV recommendation & Determination method and/ Type of data expected/
impacted Justification or source Analytical approach

Operating (inlet) pressure Viral Include, to confirm process From online data acquisition, Continuous datastream,
CPP
clearance consistency plot results with range of univariate
acceptable standard profiles.
Correlate these data with
filtration volume and protein
concentration.

Filtration (load) volume Viral Include, to confirm process Routine batch documentation, Discrete value, univariate
CPP
clearance consistency vessel weight change from pre-
rinse tare to filled weight

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 42
7.10 Step 10, Ultrafiltration and The number of dia-volumes needed to complete the buffer
exchange, pH of the AEX eluate solution to be processed
Diafiltration (UF/DF) – in this step, and UF/DF processing time are additional
CPV recommendations CPPs to include in CPV, because of their potential impact
The A-Mab case study does not provide sufficient detail on product concentration and dia-volumes (affects
to trace CPV rationale back to a CS, risk assessment, or osmolality), the potential formation of aggregate (from
process development for this step. Therefore, six well lengthy processing time and/or incorrect pH), and because
known process parameters typical for the operations of a process validation has not yet provided sufficient data to
UF/DF step are considered for CPV: demonstrate process capability for these parameters.

• 
Trans-Membrane Pressure (TMP); It is assumed that a risk assessment established that
the UF/DF step has potential to affect various product
• 
Temperature of the product containing stream;
quality attributes. Variation in protein concentration
• 
Permeate and recirculation flow rates; prior to BDS freeze and fill (step 11, post-filtration)
• 
Number of dia-volumes to complete the may affect downstream drug product manufacturing
buffer exchange; controls/ capability, so trending of this CQA is included
in CPV recommendations. These data will also provide
• 
Product concentration (prior to and after
evidence for any correlation with other variables (e.g.
buffer exchange);
dia-volumes needed for buffer exchange). Optionally, CPV
• 
Step processing time. may include selected product CQAs, chosen because of
Platform knowledge was leveraged to define an initial knowledge that they may reveal the impact of variability
membrane life limit controlled via batch documentation in the process or provide useful information about process
and equipment logbooks. A specific membrane lifetime capability. The product solution identity, composition, and
monitoring protocol is expected to be in place alongside aggregation could be altered by either post-diafiltration
the CPV plan, to verify filter performance. pH or osmolality (or by a trace contaminant in compendial
grade raw material), so inclusion of these CQAs should
Flow rates are response variables that automatically
be considered for CPV. The A-Mab case study CS
adjust to maintain a fixed TMP set point (pressure
established that impurity clearance capability for residual
controlled operation). Because flow rate profiles tend
methotrexate is very high and does not require further
to vary over the re-use lifetime of the membranes,
verification. The genealogy link between the culture media
an optional choice for CPV includes monitoring of
used in the upstream process batches (impurity clearance
permeate and recirculation flow rates via a trajectory
of antifoam and methotrexate) and the UF/DF membrane
profile of the continuous dynamic data. Reference
lot will be logged in the enterprise resource planning
standard profiles (3SD tunnels, i.e. control charts with + 3
system for use in investigations.
standard deviation acceptability limits) will be shown for
comparison (sourced from the initial use cycles for the Protein concentration prior to diafiltration is suggested as
membrane and from the PPQ batches). an option for inclusion in CPV, to provide data linking this
in-process CQA measure to the final protein concentration
It is assumed that PPQ demonstrated that BR procedures,
at completion of this step. Another suggested option is to
automated process controls, and alarming ensure
trend aggregates in the final retentate with the intent of
the UF/DF step is routinely monitored and operated
providing additional data to trend this step for ability to
within established limits characterized in small scale
control formation of this process-related impurity.
development DOE studies. Therefore temperature and
TMP are dismissed as non-CPPs and are not recommended
for inclusion in CPV.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 43
Diafiltration and final formulation buffer ingredients are CPV as a risk mitigation action for the CS, so this has been
excipients of the drug product and therefore are critical included as an option for CPV.
raw materials. Supplier quality management includes
The key process attribute of yield is included in CPV for
specifications for material purity, content of particular
trend monitoring as a process performance indicator.
elemental impurities, and endotoxin from inspection of
Certificate of Assay (CoA) summaries and/or in-house Normalized Water Permeability (NWP) and average
verification testing. Reserve samples and CoA summaries filtrate flux are monitored and verified by a membrane
for each excipient lot are preserved until drug product lifetime protocol and new membrane installation
expiration to enable investigations as needed. Although SOP. Sampling for lifetime monitoring, verification,
not specifically identified here, an un-named Critical and potentially extension of the number of reuses is
Material Attribute (CMA) for ‘excipient 1’ may require managed under this separate protocol and is therefore
characterization of the variability of ‘attribute A’ during not considered here for CPV.

Table 7.10. Step 10 CPV recommendations

Variable Class CQAs CPV recommendation & Determination method and/ Type of data expected/
impacted justification or source analytical approach

UF/DF processing time Aggregates Include, to verify process Routine batch documentation, Discrete, univariate
CPP
capability elapsed time from UF start until
defined UF end

Number of dia-volumes Product con- Include, to establish process From online data acquisition, Discrete, univariate
CPP
centration consistency include in batch documentation
and several
others

UF/DF retentate final pH Aggregates Include, to establish process Routine batch documentation Discrete, univariate
CPP
consistency

Protein concentration Protein Include, to establish process Routine batch document Discrete, univariate
CPP
prior to BDS fill step conc. of BDS consistency recording of field A280 test
results

Yield (final retentate) – Include, to confirm SPC Routine batch document Discrete value, multivariate
KPA
capability calculation using field A280
test result

Optional elements to include in CPV

Excipient ”1” Attribute – Optional, to examine variability Released by compendia testing Discrete, univariate
CMA
“A” of materials used or COA, results recorded in
LIMS

SEC aggregates in final – Optional, to confirm Will require non-routine Discrete value, multivariate
CQA
retentate consistency of mixing and foam in-process test, record results
control in LIMS

Protein concentration – Optional, to establish process Routine batch document Discrete, univariate
KPP
prior to dia-filtration consistency recording of field A280 test
results

Recirculation flow rate – Optional, to establish process From online data acquisition, Continuous datastream,
KPP
consistency include in batch documentation univariate

Permeate flow rate Optional, to establish process From online data acquisition, Continuous datastream,
KPA
consistency include in batch documentation univariate

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 44
7.11 Step 11, Final Filtration and
Freezing of BDS –
CPV recommendations
Final filtration performed while filling sterile containers
provides assurance of microbial control of the drug
substance intermediate, but is otherwise unlikely to
impact product quality. No provisions are assumed for a
validated re-filtration option. Filter function is confirmed
after each batch by standardized filter integrity testing.
Introduction of filter leachates are minimized by process
design and leachable studies, which include a pre-use
rinse of the filter with a qualified fixed amount of final
formulation buffer.
The KPA of yield was chosen for monitoring this step in
CPV, because trend monitoring of yield will provide a good
process performance indicator.
Although it is not a CPP, maximum inlet pressure (filter
pressure) is known to exhibit product specific batch
variation from platform process knowledge and so is
suggested as an optional inclusion in CPV. Filtration
volume is another KPP that would be a reasonable
optional choice for CPV, providing a different measure
for assessing processing capability. Correlation of filter
pressure with filtration volume, protein concentration,
and filtration time are other optional considerations that
could be included in the CPV plan, to characterize normal
performance and variation for the A-Mab process for
future predictability.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 45
The BDS freezing rate profile and container seal integrity freezing temperature. BR sequence and instructions,
has been validated at commercial scale and the freezing automation monitoring and alarm systems will ensure
time and conditions are well controlled and documented the step is routinely monitored and operated within its
to support any investigations. The freezing equipment established limits. The time the intermediate is stored
is included in the periodic validation maintenance and frozen prior to shipment for drug product manufacturing,
instrument preventative maintenance programs. CPV could be considered as a means of identifying any potential
monitoring is not proposed for several related operating correlation with data from the stability program, but this is
variables because PPQ demonstrated tight control and not included in the CPV recommendations here.
minimal variation of these variables, including: bulk
EM is a supporting quality system subject to periodic
mixing after UF/DF and during the fill, the fixed flow rate
monitoring, so it is not included in CPV recommendations,
through the filter, the flush volume used, verification
despite a related incident report for this step (see Section
of the filter integrity testing and product intermediate
6, scenario 5).

Table 7.11. Step 10 CPV recommendations

Variable Class CQAs CPV recommendation & Determination Method and/ Type of data expected/
impacted justification or source Analytical approach

Bulk Fill step yield – Include, to establish process Routine batch document Discrete value, multivariate
KPA
consistency determination using field A280
test result

Optional elements to include in CPV

Filtration volume – Optional, to verify process From online data acquisition, Discrete value, univariate
KPP
capability include in batch documentation

Maximum (inlet) pressure – Optional, to establish process Routine batch documentation, Discrete value, univariate
KPP
consistency max. inlet pressure from online
data acquisition during fill

Filtration time – Optional, to define normal Routine batch Discrete value, univariate
KPP
documentation, elapsed range
time

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 46
7.12 Bulk Drug Substance Lot Data – chromatography parameters25. No particular deamidated
isoforms (which incidentally, were not designated as
CPV recommendations CQAs) or other charge variants are required for CPV
The QC group ensures each drug substance batch monitoring. The routine drug substance specification
meets specifications for lot release to drug product confirmation of A-Mab identity includes a CEX HPLC
manufacturing. Some specifications, such as the identity method which separates isoforms (product-related
attribute of consistency with reference standard and substances as defined by ICH Q6B) and both a consistent
inspection for new peaks, are not amendable to trend profile with the reference standard and absence of new
analysis for CPV. The QC microbiology laboratory peaks are part of the acceptance criteria, so this would not
monitors all drug substance lot data for endotoxin and be included in the plan.
bioburden against alert and action limits to provide
It is recommended for CPV, that the routine lot release
appropriate monitoring of the process and management of
Size Exclusion Chromatography (SEC) results for percent
microbial control deviations.
monomer and aggregates be trended and long-term
In the A-Mab case study, routine BR specifications control (alert) limits defined within their release and
proposed for the drug substance were intentionally stability specifications. The data for these parameters
minimized to show a potential application of QbD will not form a normal distribution, so control will involve
development for process validation Stage 1. Endotoxin QC review of the results against action and alert limits.
testing, a BDS and drug product release requirement is Note that samples for SEC testing are collected from
reviewed as per the QC laboratory SOP, with SPC based the product intermediate prior to bulk freezing and the
alert limits which are assessed for suitability during each effect of freezing, storage, and shipping conditions on
annual product review. For the CPV plan, additional aggregation should be considered for inclusion in CPV as
BDS CQAs were selected for continued verification and inputs to the DP process.
enhanced monitoring, to demonstrate consistency over a
Additional CQAs are listed here as optional for inclusion
longer period during which more process variation may be
in the CPV plan. Those CQAs that should perhaps be
observed. Content of various oligosaccharide structures
included in trending more often than annually for the
were selected as high risk CQA examples from the A-Mab
Annual Product Review, as a result of relatively frequent
case study (refer to section 7.3 and 10.3). Control limits
manufacture, should be included in CPV. Content of
are set inside the claimed acceptable range (see section
three oligosaccharide structures (sialic acid, mannose
5) based on statistical analysis of data to provide early
and non-glycosylated heavy chain) and two process
warning during trend monitoring.
impurities (DNA and methotrexate) were selected as
Some additional process and product related impurity options for CQAs to be added to CPV trending. As noted
parameters are included in the CPV plan for this process earlier, trending of results for two other oligosaccharide
step. For example, monitoring of antifoam C rather than structures will be done at step 3. Methotrexate is a raw
methotrexate clearance was chosen. Antifoam additions material used in steps 1 and 2 and there are no specific
vary batch-to-batch to control foam and clearance is controls for its removal but since there is a high safety
combined over steps 4 and 5. In contrast, methotrexate is clearance limit for this residual process impurity, testing
a fixed addition prior to the N-1 seed bioreactor, resulting for it in the BDS is optional.
in significant dilution as the process scales up to 15,000L
Shipping of the drug substance has been validated.
and a high log reduction factor was demonstrated for the
Monitoring of in-shipment time and maximum
Protein A step 5 alone. HCP is not included for CPV at
temperature during shipment is routinely verified to be
BDS because it is monitored for CPV at step 8 (AEX), as
within qualified limits. Trending of shipping conditions
both a special sample test with a control limit well inside
should be considered for monitoring of the shipping
the 0 to 100 ng/mg acceptable range (based on the similar
process, though these parameters are considered out of
X-mAb process) and via the multivariate model for linked
scope for Stage 3 CPV, so they are not included in the plan.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 47
Table 7.12. Step 10 CPV recommendations

Variable Class CQAs CPV recommendation & Determination method and/ Type of data expected/
impacted justification or source analytical approach

Monomer (by SEC) – Include, to establish SPC LIMS results from routine Discrete value, univariate
CQA
testing of BDS

Aggregates (by SEC) – Include, to establish SPC LIMS results from routine Discrete value, multivariate
CQA
testing of BDS

Galactose content – Include, to establish SPC LIMS results from routine Discrete value
CQA
testing of BDS

Afucosylation – Include, to establish SPC LIMS results from routine Discrete value
CQA
testing of BDS

Optional elements to include in CPV

DNA Include, to establish SPC LIMS results from routine Discrete value
CQA
testing of BDS

Methotrexate and/or Include, to establish SPC LIMS results from routine Discrete value
CQA
antifoam C testing of BDS

Sialic acid content Include, to establish SPC LIMS results from routine Discrete value
CQA
testing of BDS

Mannose content Include, to establish SPC LIMS results from routine Discrete value
CQA
testing of BDS

Non-glycosylated heavy Include, to establish SPC LIMS results from routine Discrete value
CQA
chain testing of BDS

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 48
8.0
Frequency and scope of CPV analysis
CPV analysis commences with commercial production, following successful completion of the PPQ batches.
The start of data collection and analysis begins with the first representative commercial batches produced at the
commercial scale facility. Due to potential scale and facility differences, as well as modifications in the process
control or adjustments to test methods prior to PPQ, CPV monitoring will not include data from clinical batches,
though experience gained in these project phases are likely to help in assigning initial control limits. As a result,
the amount of directly relevant data available to set appropriate monitoring limits will be limited at this point.
This poses a problem, at least until significant quantities of data have been gathered.

8.1 Scope of CPV Analysis


To address the problem of limited data when commercial description of a Normal distribution. But, the actual
production starts, it is recommended that CPV analysis sample size needed to establish variation with a good level
is performed in two phases, the initial CPV phase and the of confidence could involve a larger number of batches.
long-term CPV phase. It is recommended that a statistician is consulted in the
Phase 1: Initial CPV Phase context of a particular data set.
The initial CPV phase is considered pre-SPC and provides At the conclusion of the initial CPV phase, alert limits for
the ability to analyze process performance based on the monitored parameters should be established where
a limited data set to gain understanding of the normal applicable, if they do not already exist, or to justify the
process variability in the commercial facility. This phase alert limits that have been set. Additionally, the risk
should include enough batches to provide data to reflect assessment performed following completion of the PPQ
the range of potential variability and allow statistical batches should be reviewed to determine whether the
process ranges to be established. During this phase, charts additional process experience has changed the risk score
are run using the specifications based on PPQ, clinical and for the monitored parameters. Trends in process related
process characterization information. Data collected will non-conformances should also be included in the review
be used to identify possible trends and to demonstrate of the risk assessment, and this should involve considering
that the process remains in a state of control. For A-Mab, whether parameters not originally included in the plan
the initial CPV phase will continue until at least 30 batches for the initial CPV phase ought to be added. Should there
have been produced (this is assuming one upstream cell be an increase or decrease in risk for the monitored
culture batch feeds one downstream purification batch). parameters, or a noted non-conformance trend for a
It is worth noting that, though 30 batches are suggested parameter which was not previously monitored for CPV
as the minimum number to form a representative data set, analysis, the plan may be revised to reflect the updated
this should not be regarded as a ‘magic number’. Many process understanding and risk analysis prior to initiation
introductory statistical texts cite 30 as a reasonable of the long-term CPV phase.
start for independent data that fit approximately the

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 49
Phase 2: Long-term CPV Phase 8.2 Frequency of Analysis
The long-term CPV phase is the statistical process control
A documented analysis and conclusion as to whether
phase. This phase has the following objectives:
the process remains in a state of control (a CPV Report)
• 
Ongoing verification of the process over the may be performed based on the production schedule.
lifetime of the product to demonstrate the process For example, the CPV plan might include the following
remains in control; conditions for a particular product like A-Mab:
• 
Identify trends which may be within the normal • 
Campaign (< 10 batches) – Minimally at the
process variability, but indicate a potential to trend conclusion of the campaign;
outside the alert limits;
• 
Campaign (> 10 batches) – Minimally every 10
• 
Continue to build understanding of the sources of batches, and at the conclusion of the campaign, or
variability in the process and their impact. at a predetermined time interval (e.g. quarterly);
Section 10 provides detail proposing how monitored items • 
Continuous – Minimally every 10 batches, or at a
fit into the plans for short-term and long-term CPV. predetermined time interval;
• 
A frequency preference of every 10 batches
has been selected to enable trend identification
via typical tests for special causes of variation
in control charts. Note that analysis will be
performed as described in section 9 per the
requirements of the phase of CPV analysis.
Frequency of documented analysis and conclusion
may be increased when greater than desired
process variability is noted or if conclusions are
needed to support product disposition.
It is important to note that these statements are given as
an example for a product like A-Mab, being manufactured
at a frequency of the order of two to ten batches a month.
Even so, formal CPV Reports are only likely to be created
up to four times a year. For products where the frequency
of batch manufacture is low e.g. once a year, it wouldn’t
make sense to have more than one CPV Report a year.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 50
9.0
Establishing control limits
Note: Control limits (for parameters and attributes selected for the CPV Plan) need to be established initially. However,
they are likely to be re-established at some point and this may require change control (see Section 13). This section focuses
on the principles involved in establishing initial and long-term control limits, in preparation for the example of a CPV
execution plan (Section 10). More detailed, mathematical considerations are covered in Section 12.

To initially establish control limits a documented such indications may need timely intervention to drive
business process should be in place to address process consistency. Initial control limits in a CPV plan
collecting, analysing, reporting and storing of data for should not be interpreted as acceptance limits (i.e. a
the process at the manufacturing scale. Additionally, specification for the product).
data generated during development, scale up, as well
Through the initial control limits evaluation, the
as small scale data, can be used to set control limits.
strategy for process control should be identified and
By evaluating process performance, the initial control
applicable limits established based on process and
limits would help provide an early indication of a lack of
measurement system capability. The Process Capability
control in the process for certain process parameters or
Index (Cpk) and Process Performance Index (Ppk)
quality attributes, by establishing the anticipated range
provide useful indicators of the level of control likely to
of expected variation. During the evaluation process,
be achievable for the process.

Figure 9.1. A Mab case study example indicating different limits during monitoring of certain process parameter

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 51
The means of establishing SPC limits for an initial period transformation can be helpful in making data meet the
and for the long-term, is described in detail in Section assumptions of normality. Process knowledge may help
12.4. Additionally, an SPC chart showing different limits: in transforming data to a more SPC-amenable form. For
upper and lower control limits (UCL, LCL), being the example when a known and codified relationship exists
most commonly used and arguably important for many between process parameters and QAs, normalizing the
data sets, is presented in the Figure 9-1. If insufficient data (taking into account the available process knowledge)
data are available, either with a new process or after a can lead to a more relevant and reliable value for trending.
major process/assay change, initial, temporary limits
Data distribution should be considered when selecting
may be proposed, based on available development,
analysis tools. The method of reporting each data set
initial small scale data and process knowledge. If so,
should be defined and approved in applicable GMP
small scale models should be appropriately developed
documentation. All excursions outside approved
and qualified in order to guarantee the scale process is
documentation should be further investigated, justified
representative and predictable.
and documented in appropriate GMP documentation.
Statistical and scientific rationale should pre-determine
When more data are available, calculated SPC limits
what data set is required. Once sufficient at-scale data are
can be identified. The SPC limits should be periodically
available long-term SPC limits can be established.
reviewed to capture process variability and be brought
Understanding which elements (e.g. raw materials, into line with any new regulatory or quality guidance or
operators, facility etc.) contribute to common-cause additional CPV Plan requirements. Established SPC limits
variation may depend on the relevance and knowledge should be reviewed in light of process changes to confirm
of the specific process, and will help to set relevant their continuing validity and may be adjusted in response
and appropriate SPC limits. This requires the inclusion to generation of additional data. The process monitoring
of sufficient data (initially determined or statistically procedure, as well as process capability review, should
relevant) to capture long-term common-cause variation. be established in applicable documentation (e.g. the
Factors that may lead to variation include for example: CPV Plan). With a given frequency of analysis, further
pack-to-pack variation in chromatography columns, statistical examination is required to determine if the
measurement system recalibrations, raw material lot-to- results suggest a potential impact on the product.
lot variability, etc. This is described further in Section 10. Multiple data
The calculation of control limits depends on an assumption sources and applicable analysis should be organized and
that data is normally distributed and each datum point is integrated in appropriate process data analysis tools.
independent. This may not always be the case, and data Subsequent statistical tools should be appropriate for the
data to be analyzed (see Section 12).

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 52
SECTION 10.0

10.0
Example CPV execution plan
for drug substance
After completion of the PPQ, continued process verification should demonstrate that the process is in control.
In the words of the FDA guidance from 2011, it should offer: ‘assurance that the process is reasonably protected
against sources of variability that could affect production output, cause supply problems, and negatively affect
public health’5. A monitoring and trending program for the A-Mab drug substance process parameters and
attributes is outlined in this section, but the reader should view the discussion and content of the tables as
recommendations and ensure that the parameters and values they use are appropriate for their product and
process. It is applicable for both initial and long-term monitoring of the drug substance manufacturing process.
Selection of variables for monitoring is based on information and rationale in Sections 5 and 7.

Note: This plan sub-section is neither a minimalistic nor Continued assurance of consistent process performance
comprehensive listing of variables expected for CPV and identification of potential out of trend results is
monitoring. Rather we attempt to maintain a reasonable achieved by applying SPC rules and capability analysis
consistency with the A-Mab case study to provide an example (Ppk) as discussed in Section 12. CPV datasets enable
of likely CPV variables associated with a product launch process capability predictions with higher confidence,
(where in this case, understanding of variability is not evident deepen process understanding, and improve process
immediately after completing a platform-based Process robustness by increasing the likelihood of detecting
Validation (PV) Stage 2 PPQ with only two commercial scale sources of process variability before they cause batch
batches of the particular A-Mab molecule). This is meant to failures. The CS is updated based upon reviews of related
demonstrate reliable process control and ability to detect risk assessments, as a part of assessing accumulated CPV
process drift. Commercial scale process data for legacy data in summary CPV Reports. CPV should be integrated
processes would likely be available and may justify a smaller into the organization’s development process and quality
set of CPV monitored variables. It is emphasized that this is system. A CPV Master Plan may be used across a
an untested example package for consideration, not general corporation, to guide development of product specific CPV
guidance or proven best practice approach. procedures including the incorporation of outputs from
Stage 1 and Stage 2 (e.g., CQA, CPP). CPV output (from the
executed plan) will be documented and summarized at a
frequency defined by the plan. Figure 10.1 is a schematic
showing the continuity of review in the product lifecycle.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 53
Figure 10.1: Continued review as part of the product lifecycle.

1
Process Understanding
CPP/CQA’s
Risk Assessment Review
Process Knowledge Report

2
Continuous
Continuous Quality
Process Monitoring
Improvement Product and Feedback
Change Quality Process Control
Strategy
Management
Documentation Batch Record Data
Specifications

4 Process Analysis

3
Initial Process Performance
Evaluation Acceptance & Release
Ongoing Process Monitoring
CpK Statistics Database
Annual Product Review

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 54
The A-Mab case study (and assumed post-PPQ CS) proposed control limits; ‘EWMA chart’ (Exponentially
contains information on the linkage between product Weighted Moving Average control chart) (see Section
quality attributes and CPPs across the eleven process 12 for application); ‘3SD tunnel’ (a control chart with
steps. These relationships and the relationships between +3 standard deviation control limits) for data that has a
KPPs and KPAs are specific to each step. Therefore, this dynamically changing mean during the batch processing
section is divided into tables for each (Steps 1 through time (such as a VCC profile, UV chromatogram, or UF/DF
11, plus BDS) in order to list process parameters and flow rate); or ‘exception flag’ which uses routine process
attributes to be monitored to verify process control over monitoring for process parameters and reporting of any
the entire lifetime of manufacturing the drug substance. out of range result (exception). Any custom correlations
Non-routine sampling and specific data gathering will that are developed during CPV would also be shown in this
augment routine sampling and data recording to generate column (e.g. VCC versus dCO2, or step processing time
the data for trending and monitoring under this CPV plan. versus step yield).
For the tables presented in this section, CPV process Column E identifies plan monitoring requirements for
variables and their classification are listed in Columns an initial short-term CPV period of manufacturing which
A and B, as recommended earlier in Section 7. Column follows completion of PPQ in order to obtain sufficient
C includes information on any data treatment required data to set long-term control limits (unless limits already
before graphing to monitor trends. ‘Unadjusted’ raw data exist with sufficient confidence and understanding of the
are measurement results (source data) that are directly expected long-term normal variation). This period is based
charted. ‘Converted’ data indicate that monitoring the on a minimum number of independent batch experiences,
process variable involves treatment of measured results e.g. 30 as mentioned in Section 8.1 (with a reminder to
and either combining with other process data (e.g. yield is consider that raw material lot impact experience may
a ratio of combined raw data) or standardizing to match lag process lot experience), or achieving a target Ppk for
the intent of control limits (e.g. weight measurements the variable’s range of control. Proposed initial control
converted to volume or converting totalized flow limits to use when starting CPV baseline monitoring are
through volume to column volumes). Raw (or converted) given in column F and are based on assumed PPQ criteria
data that is mathematically ‘transformed’ is a third type for A-Mab, or a fictitious control range proposed after
of data treatment that may be required before charting completing the process validation Stage 2 effort. See
for trend monitoring. Section 9 for more information on establishing initial
Column D specifies the recommended SPC tool for control limits.
monitoring performance trends against control limits. Note: Due to various strategies for combining batches in
The tool listed is selected based on subject matter expert manufacturing, 30 completed batches may not necessarily
experience with the process development history. The be sourced from 30 independent vial thaws; or use 30
chosen tool provides a means to visually review the data uniquely prepared lots of the involved solutions; or employ
and may be revised when the nature of the data is better 30 different raw material lots (which could be sub-lots
understood. The options included in the plan include: of fewer supplier bulk lots); or may not produce 30 drug
‘individual run chart’ for data without initial control limits; substance or drug product batches. Awareness and
‘individual measurement chart‘ (a control chart) for data tracking of different lot counts for different variables is
that can be plotted with an expected fixed mean and important information during CPV.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 55
Column G is dedicated to the CPV plan after the initial monitoring data may verify that the expected control is
baseline period ends and the frequency of sampling and/ routinely achieved, and that there are select CQAs for
or trending is subject to long-term monitoring during which monitoring data shows little variability. Responding
commercial manufacturing. At this future point in the CPV to signals in the data in this way allows adjustment of the
lifetime, it is assumed that sizeable specific commercial long-term monitoring plan to tailor it for monitoring those
manufacturing experience and knowledge of the A-Mab elements of the process most likely to exhibit variability
process exists and sources of variation are understood and hence need the greatest attention.
well enough to support a reduced frequency of testing or
Besides time-based risks to maintaining the validated state
review of data trends with lowered risk for undetected
of process control, the other type of risk that requires
process drift. In some cases it may be possible to gain
verification and monitoring involves change-based risks.
enough confidence in the behavior of certain parameters
and their relationship to the process, that they may be These assumed known ‘for-cause’ events are shown in
removed from the CPV Plan and only reconsidered for column I and new SME knowledge can be added as it
enhanced monitoring to evaluate future process changes. is gained. These changes (e.g. critical raw material lot
In column H, ‘Initial limits’ is an abbreviated placeholder changes or process improvements) may have an impact
term for documenting the dates that particular life-time that extends beyond the change implementation and may
control limits apply which would be documented as make previous data and trend characteristics obsolete and
‘Range1’. This information would be populated after the invalidate previous short-term or long-term control limits.
initial baseline monitoring is complete and short-term When available, collected monitoring data should be
control limits are superseded by long-term limits. Long- provided with the resolution recorded in its raw data form,
term limits and ‘Range2’ are included in the early tables rather than reflecting any rounding to the significant figures
to demonstrate the historical nature of CPV lifecycle included in the control limits. This enables more accurate
management. Control limits may change at a given time statistical analysis and determination of capability.
for a particular justified reason (such as appearance of
Note: Situations that would result in duplicating information
very long-term variation or change factors), and past data
across a table are occasionally presented with alternative
profiles should not necessarily be assessed (or displayed)
proposals to offer the reader different options to consider for
against more recent control limits. However, the ability to
CPV. Various charting options are presented as examples and
review historical data ranges along with changes in more
different life-time plans shown for the variables. Rationale
recent predecessor control limits can enhance process
would be subject to SME justification for each individual
understanding over the product lifespan, especially if these
variable, and as not shown below, may actually result in the
changes are associated with a set of diagnosed root causes.
same monitoring tool and life-time monitoring plan.
Collection of data may at some point provide sufficient
Note: Since the contents of column H are subject to more
demonstration of control of a variable that it may be
frequent updates than the other plan elements, the reader
removed from the long-term monitoring plan, or that the
could consider migrating or referencing the column H lifetime
frequency with which a particular variable is monitored
control limits for each variable (as they become available) in a
can be reduced to an occasional (audit) basis. Examples
separate document for efficient review and approval of revised
of variables that might be removed from long-term
ranges to maintain both the historical control ranges with
monitoring include CPPs that have been identified as
current control ranges.
being well-controlled (WC-CPP). Initial (short-term)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 56
10.1 Step 1, Seed culture expansion
in disposable vessels – CPV variables
The CPV plan for process variables in this step are shown in medium component presented earlier are included in this
the following table and align with the rationale in Section 7. example. In this example, it was assumed the split ratio
The qualification of new cell banks is beyond the scope of for this step did not have PPQ acceptance criteria (VCC, %
this CPV plan and subject to change control management viability, and duration ranges employed in PPQ) nor was
by registered regulatory agreements. Monitoring and there sufficient process data from the A-Mab working cell
verification of the commercial scale impact of a change in a bank to adopt initial CPV control limits.

Table 10.1. Step 1 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-based)
limits based) (Long-
(short- term)
term)

Viable Cell Conc., each Unadjusted Individual chart Every batch until 0.7 to 2.8 Every batch LT range1 Cell bank or
KPA
passage end (raw data) with long-term long-term limits x 106 (vc/ TBD (from growth medium
3-sigma limits set mL) date X changes
to Y) LT
range2
(from date
Y to Z)

Optional elements

Initial VCC split ratio, Converted Individual Run Every batch until Character- Once annually LT Period1 Cell bank or
KPP
each passage (ratio) chart long-term limits ize (No Range1 growth medium
set PPQ limits) TBD LT changes
Period2
Range2

Culture duration, each Raw data Individual chart Every batch until 3 to 4 While PpK < LT range Cell bank or
KPP
passage with long-term long-term limits (days) 1.0, Otherwise TBD growth medium
3-sigma limits set not required (dates: changes
TBD)

Culture viability, each Converted EWMA chart Every batch until 88 to 98 not required LT range Cell bank or
KPA
passage end (ratio) long-term limits (%) TBD (from growth medium
set date X changes
to Y)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 57
10.2 Step 2, Seed culture expansion
in bioreactors – CPV variables
The CPV plan for process variables in this step is shown earlier is included in this example. It was assumed the
in the following table and follows the rationale in Section cell culture split ratio for this step had sufficient data for
7. Monitoring and verification of the commercial scale the expected bioreactor expansion performance to adopt
impact of a change in a medium component presented initial control limits.

Table 10.2. Step 2 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-based)
limits based) (Long-
(short- term)
term)

Viable Cell Conc., each Unadjusted Individual chart Every batch until 3.9 to 6.0 x Every batch LT range Cell bank or
KPA
passage end (raw data) with long-term long-term limits 106 TBD growth medium
3-sigma limits set (vc/mL) (from date changes
X to Y)

Optional elements

Initial VCC split ratio, Converted Individual Run Every batch until 3.0 to 4.1 Once annually LT range Cell bank or
KPP
each passage (ratio) chart long-term limits TBD growth medium
set (from date changes
X to Y)

Culture duration, each Unadjusted (x.x Individual chart Every batch until 3 to 5 While PpK LT range Cell bank or
KPP
passage resolution) with long-term long-term limits (days) < 1.0, TBD growth medium
3-sigma limits set Otherwise not (from date changes
required X to Y)

Culture viability, each Converted Individual chart Every batch until 90 to 99 not required LT range Cell bank or
KPA
passage end (ratio) with long-term long-term limits (%) TBD growth medium
3-sigma limits set (from date changes
X to Y)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 58
10.3 Step 3, Production culture
bioreactor – CPV variables
To be consistent with the status of the A-mab case A fixed volume of nutrient feed is added at a
study when this plan is initiated, the option of creating a defined time under automation and routine batch
time-dependent multivariate PLS (partial least squares) document controls, therefore trending the amount
bioreactor model is a CPV objective, based on previous and timing of the nutrient feed addition does
successful experiences25. The A-mab case study [3, not provide value because they are not subject
Section 3.10, Page 107-109] describes a principle to random variation. Production cultures are
components bioreactor model as a predictor of acceptable harvested within an acceptable duration based on
oligosaccharide structure and acidic variant CQAs. The viability and titer considerations.
parameter inputs to the model include temperature and Temperature and pH are continuously feedback
pH, and attribute inputs to the model include titer, VCC, controlled to set points, during the 16 to 18 day
and viability. All these variables are included individually culture but measured values are dynamic over that
for CPV monitoring while the bioreactor model is qualified. time period. Therefore, 3SD tunnels (the range
The output of the model for each batch is classified as a defined by the mean + 3 standard deviations) for the
KPA. The potential added value in using the model is in parameter profiles will be developed during the initial
ensuring internal correlations among different variables CPV period, to generate an expected ’conduit’ for
are considered. In the future, the values generated from results when tracking consistent control of the CPP.
this model may provide a multivariate output for trend
monitoring that is predictive of process performance, with
its own alert and action limits.

Table 10.3. Step 3 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-based)
limits based) (Long-
(short- term)
term)

Culture duration Unadjusted Individual chart Every batch until 16 to 18 Every batch LT range Change in cell
CPP
(x.x resolution) with long-term long-term limits (days) TBD bank, culture
3-sigma limits set (dates: medium,
TBD) or process
setpoint

Maximum pCO2 Unadjusted Individual chart Every batch 45 to 140 Every batch LT range Change in cell
CPP
(raw data) with long-term until long-term (mmHg) TBD bank, culture
3-sigma limits limits set, also (dates: medium,
correlate w/ TBD) or process
IVCA setpoint

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 59
Table 10.3. Step 3 CPV variables

A B C D E F G H I

Variable Class Data Monitoring Initial baseline Initial Periodic Lifetime For cause
treatment tool monitoring baseline monitoring control monitoring
prior to (short-term) control limits (time/cycle- limits (change-based)
analysis (short-term) based) (Long-term)

Culture duration Unadjusted Individual chart Every batch until 16 to 18 Every batch LT range Change in cell
CPP
(x.x with long-term long-term limits (days) TBD (dates: bank, culture
resolution) 3-sigma limits set TBD) medium,
or process
setpoint

Bioreactor pH Unadjusted 3SD tunnel Every batch until 6.75 to 6.95 Not required, See PLS Change in cell
CPP
(raw data) long-term limits (-log [H+]) included in model, LT bank, culture
set for reference PLS model range TBD medium,
(dates: or process
TBD) setpoint

Afucosylated glycans Unadjusted Individual chart Required 5 to 10 (%) Once annually LT range 5 Change in cell
CQA
(raw results) with long-term for model (time 0 to 10 Initial bank, culture
3-sigma limits qualification only of annual to current medium,
stability date or process
batch) setpoint

Galactosylated glycans Unadjusted Individual chart Required 15 to 35 (%) Once annually LT range 15 Change in cell
CQA
(raw results) with long-term for model (time 0 to 35 Initial bank, culture
3-sigma limits qualification only of annual to current medium,
stability date or process
batch) setpoint

PLS model employing Converted & Custom Every batch until Trajectory Every batch LT range Change in cell
KPA
pH, DO, temperature, transformed PLS model model is > 95% versus time TBD bank,
pressure, gas rates, PCA t1 predictive ± 3 StDev (dates: TBD culture medium,
weight, VCC, viability, or
titer, glucose, lactate process
setpoint

Product yield (titer) at Unadjusted Individual chart Every batch until 4.0 to 5.5 Not required, See PLS Change in cell
KPA
Harvest (raw data) with long-term long-term limits (g/L) included in model, bank,
3-sigma limits set PLS model LT range culture medium,
and PpK >1.0 TBD or
(dates: process
TBD) setpoint

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 60
Table 10.3. Step 3 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-
limits based) (Long- based)
(short-term) term)

Optional elements

Antifoam lot Unadjusted Exception Test BDS < LOD Not required < LOD Change in
CMA
(raw data) Flag (new lot) clearance material
for 3 different ID or supplier
lots

Medium Unadjusted Individual chart Track OOR 365 to 435 Track OOR 375 to 425 Change in
CPP
osmolality (raw data) with long-term Exception flags (mOsm) Exception mOsm medium prep
3-sigma limits flags (dates:
current)

Mannose Unadjusted Individual chart Required 5 to 8 Not required LT range Changes in cell
CQA
content (raw data) with long-term for model (%) at this time TBD bank
3-sigma limits qualification only (dates: or step 3
TBD) setpoints

Sialic acid Unadjusted Individual chart Required NMT 1.6 Not required LT range Changes in cell
CQA
content (raw data) with long-term for model (%) at this time TBD bank
3-sigma limits qualification only (dates: or step 3
TBD) setpoints

Non-glycosylated Unadjusted Individual chart Required 0 to 2.4 Not required LT range Changes in cell
CQA
heavy chain (raw data) with long-term for model (%) at this time TBD bank
3-sigma limits qualification only (dates: or step 3
TBD) setpoints

Time of glucose feeds Unadjusted Individual chart Every batch until Feed 1: a to Not required LT range Change in cell
KPP
(hrs since inoculation (raw data) with long-term long-term limits b hrs at this time TBD bank,
3-sigma limits set Feed 2: c to (dates: culture
d hrs TBD) medium, or
Feed 3: e to process
f hrs setpoint

Peak VCC Unadjusted Individual chart Every batch until 20 to 30 x Not required, See PLS Change in cell
KPA
(Viable Cell Conc.) (raw data) with long-term PLS qualified 106 included in model, bank,
3-sigma limits (vc/mL) PLS model LT range culture
TBD medium, or
(dates: process
TBD) setpoint

Culture viability Converted Individual chart Every batch until 40 to 61 Not required, See PLS Change in cell
KPA
at Harvest (ratio) with long-term PLS qualified (%) included in model, bank,
3-sigma limits PLS model LT range culture
TBD medium, or
(dates: process
TBD setpoint

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 61
10.4 Step 4, centrifugation and depth
filtration – CPV variables
For Step 4, Critical Raw Materials (CRM) that impact the process can attenuate upstream process variability
the step include the working cell bank, upstream growth prior to purification. If turbidity or the duration of depth
medium components, and depth filters. Changes to these filtration shifts upward, monitoring the inlet pressure
items would require a period of enhanced monitoring parameter or the attribute of differential pressure across
of filtrate turbidity and step yield, to demonstrate that the filters may need to be added to CPV.

Table 10.4. Step 4 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-based)
limits based) (Long-
(short-term) term)

Turbidity Unadjusted Individual Every batch until <2 Not required LT range CRM or process
KPA
of filtrate (raw data) chart with long-term limits (NTU) TBD change to this
long-term set (dates: or previous
3-sigma limits TBD) step

Step yield Converted Individual Every batch until Characterize Every batch LT range CRM or process
KPA
Filtrate (ratio) Run chart long-term limits (No PPQ TBD change
set limits) (dates: to this or
TBD) previous step

Optional elements

Duration of Broth Converted Individual Every batch until Characterize Once annually LT range Upstream
KPP
Clarification (end minus Run chart long-term limits (No PPQ TBD scale-up,
start time) set limits) (dates: Centrifuge feed
TBD) or flow
rate setpoint
changes

Inlet pressure, Unadjusted Individual Not required None Not required ± 3 StDev Upstream
KPP
depth filters (raw data) chart with of most scale-up,
long-term recent 30 filter changes,
3-sigma limits batches Centrifuge feed,
(pre- or filter flow
change) rate setpoint
changes, shifts
in turbidity or
filtration time.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 62
10.5 Step 5, Protein A
chromatography – CPV variables
Initial control limits for load ratio and elution buffer pH count AEX resin to clear protein A (spiking study) and,
are assumed, based on a simulated normal range within characterization of potential Protein A leachate from both
the DOE PAR provided in the A-Mab case study. Likewise, high and low use cycle counts with respect to resin storage
the short-term residual protein A control limits are based time. Note that elution buffer pH data is included while
on the outcome of a fictional study done after PPQ at the limits for the HCP model are generated.
small scale, which examined the capacity for high cycle

Table 10.5. Step 5 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-
limits based) (Long- based)
(short-term) term)

Protein Load Ratio Converted Individual chart Every batch until 15 to 40 Every batch LT range Resin, or
CPP
(in HCP model) (ratio) with long-term long-term limits (g A-mAb/L TBD process
Each sub-batch 3-sigma limits set resin) (dates: change to this
TBD) or previous
step

Elution buffer Unadjusted Individual chart Every batch until 3.4 to 3.8 Track OOR LT range Buffer
CPP
pH (in HCP model) (x.xx resolution) with long-term HCP model (-log [H+]) Exception 3.4 to 3.8 formulation
3-sigma limits limits set flags (dates: scale-up
initial to
current)

Residual Protein A Unadjusted w/ upper limit, Every batch until ≤ 1234 Per resin/ LT range First two
CQA
in eluate pool (raw data) & correlation long-term limits (ng/mg column TBD cycles after
vs. storage age set A-mAb) lifetime (dates: extended
protocol TBD) storage
(≥ 3 months)

Step duration Converted Individual Every batch until Characterize Once annually LT range Scale
KPP
(elapsed) Run chart long-term limits (No PPQ TBD increases
set limits) (dates:
TBD

Step yield Converted Individual chart Every batch until 68 to 88 Every batch LT range Reset range
KPA
(ratio) with long-term long-term limits (%) TBD for process
3-sigma limits set (dates: change to
TBD) this or
previous step

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 63
10.6 Step 6, Low pH treatment –
CPV variables
The viral safety risk CQA (inactivation of particular AVA) as a single point measurement. Aggregate results are
for the A-Mab process has been validated in the small scale assumed to include a sum of all quantifiable non-monomer
model during Stage 1 process validation. Initial control SEC peaks (dimer, trimer, etc). Yield is not expected to
limits for inactivation time and pH are assumed, based on be impacted by this step and variability around 100% has
a fictitious normal range within the DOE PAR provided in been associated with measurement uncertainty, therefore
the A-Mab case study. 3SD tunnel monitoring of pH during it does not merit CPV trending or monitoring. The basis of
inactivation ensures the parameter remains in range yield for the next step (7, CEX) begins from the eluate pool
throughout the inactivation time and it is not monitored of the previous step (5, Protein A).

Table 10.6. Step 6 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-
limits based) (Long- based)
(short-term) term)

pH during Unadjusted 3SD tunnel Every batch until 3.4 to 3.8 Every batch LT range Scale
CPP
inactivation (raw data) long-term tunnel (-log [H+]) TBD increases
set (dates:
TBD)

Post inactivation Converted Individual chart Every batch until ≤ 3.0 Once annually LT range Process
CQA
aggregates (additive) with long-term long-term limits (%) TBD change
3-sigma limits end and PpK (dates: to this or
>1.3 TBD previous step

Optional elements

Inactivation Converted Individual chart Every batch 80 to 120 Track OOR LT range No known risk
CPP
time (elapsed) with long-term until a (minutes) Exception TBD events
3-sigma limits long-term sigma flags (dates:
set TBD)

Quantity of Unadjusted Individual chart Every batch Characterize Not required LT range Scale
KPP
acid added (raw data) with long-term until a (No PPQ TBD increases
3-sigma limits long-term sigma limits) (dates:
set TBD)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 64
10.7 Step 7, Cation exchange
chromatography – CPV variables
Initial control limits provided in the table below are platform experience, but for A-Mab CPV a typical range
assumed, based on a fictitious normal range within will be determined for historical reference. Aggregate
the DOE PAR provided in the A-Mab case study. Step results are assumed to include all quantifiable non-
duration has a batch document control limit based on monomer SEC peaks (dimer, trimer, etc).

Table 10.7. Step 7 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-
limits based) (Long- based)
(short-term) term)

Protein Load Ratio Converted Individual chart Every batch until 15 to 25 Every batch LT range Process
CPP
(in HCP model) (ratio) with long-term long-term limits (g A-mAb/L TBD change
3-sigma limits set resin) (dates: to this or
TBD) previous step

Wash Conductivity Unadjusted Individual chart Every batch until 4 to 6 Track OOR LT range Buffer
CPP
(in HCP model) (x.x resolution) with long-term long-term limits (mS/cm) Exception TBD formulation
3-sigma limits set flags (dates: scale-up
TBD)

Elution pH Unadjusted 3SD tunnel Every batch until 5.9 to 6.1 Every batch LT range Process
CPP
(x.xx resolution) long-term tunnel (-log [H+]) TBD change to
set (dates: this or
TBD) previous step

Aggregates Converted Individual chart Every batch until ≤ 1.0 Per resin/ LT range Process
CQA
in CEX eluate pool (additive) with long-term long term limits (%) column TBD change to
3-sigma limits set lifetime (dates: this or
and PpK >1.3 protocol TBD) previous step

HCP content Unadjusted Individual chart Every batch ≤ 130 Per resin/ LT range Process
CQA
in CEX eluate pool (raw data) with long-term until a (ppm) column TBD change to
3-sigma limits long-term sigma lifetime (dates: this or
set protocol TBD) previous step

CEX eluate volume Unadjusted Individual chart Every batch until 3.7 to 4.7 Not Required LT range Column pack
KPA
(each sub-batch) (raw data) with long-term long-term limits (CV) TBD or repack
3-sigma limits set (dates:
TBD)

Step yield Converted Individual chart Every batch 83 to 97 Every batch LT range Process
KPA
(ratio) with long-term until a (%) TBD change to step
3-sigma limits long-term sigma (dates:
set TBD)

Optional elements

Step duration Converted Individual Every batch until Characterize Not Required LT range Process
KPP
(elapsed Run chart long-term limits (No PPQ TBD change to step
set limits) (dates:
TBD)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 65
10.8 Step 8, Anion exchange
chromatography – CPV variables
Initial control limits provided in the table below are The fictitious HCP clearance linkage model for the three
assumed, based on a fictitious normal range within a chromatography steps (Protein A, CEX, and AEX) is
DOE PAR provided in the A-Mab case study. The CEX included here for monitoring the trend in the algorithm
eluate is adjusted to a target pH and conductivity (CPPs) output, using the 6 parameters (two from each step) for
before loading for flow-through mode chromatography. a given batch.

Table 10.6. Step 6 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-based)
limits based) (Long-
(short- term)
term)

Protein Converted Individual chart Every batch until 100 to 200 Every batch LT range Process change
CPP
Load Ratio (ratio) with long-term long-term limits (g A-mAb/L TBD to this or
3-sigma limits set resin) (dates: previous step
TBD)

Load Conductivity Unadjusted Individual chart Every batch until 4.0 to 7.0 Every batch LT range Process change
CPP
(raw data) with long-term long-term limits (mS/cm) TBD to this or
3-sigma limits set (dates: previous step
TBD)

Load pH Unadjusted Individual chart Every batch until 7.4 to 7.7 Track OOR LT range Process change
CPP
(in HCP model) (x.xx resolution) with long-term long-term limits (-log [H+]) Exception TBD to
3-sigma limits set flags (dates: previous step
TBD

Equilibration/ Wash 1 Unadjusted Individual chart Every batch until 2.0 to 3.2 Track OOR LT range Buffer
CPP
Buffer Conductivity (in (raw data) with long-term long-term limits (mS/cm) Exception TBD formulation
HCP model) 3-sigma limits set flags (dates: scale-up
TBD)

Linkage model Transformed Individual chart Every batch until 99.5% Every batch LT range Re-qualify
KPA
output for HCP A-mAb case with long-term AEX prediction TBD model for
(predicted) study 3-sigma limits eluate HCP interval for (dates: process
model equation is predictive with mean TBD) changes in
6 95% confidence of HCP ProA, CEX, or
(Pg 158) output AEX steps

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 66
Table 10.8. Step 8 CPV variables

A B C D E F G H I

Variable Class Data Monitoring Initial baseline Initial Periodic Lifetime For cause
treatment tool monitoring baseline monitoring control monitoring
prior to (short-term) control (time/cycle- limits (change-based)
analysis limits based) (Long-
(short-term) term)

HCP content Unadjusted Individual chart Every batch ≤ 15 Per resin LT range Process
CQA
in AEX eluate (raw data) with long-term until a (ng/mg) column TBD change to any
(measured) 3-sigma limits long-term sigma Lifetime (dates: chromatography
set protocol TBD) step

Residual Protein A Unadjusted Individual chart Not Required ≤ 10 Per resin LT range Align monitoring
CQA
in eluate (raw data) with long-term (ng/mg column TBD with
3-sigma limits A-mAb) Lifetime (dates: ProA process
program TBD changes

Step yield Converted Individual chart Every batch ≥ 87 Trend every LT range Reset range for
CQA
(ratio) with long-term until a (%) batch vs. long- TBD Process change
3-sigma limits long-term sigma term limits (dates: to step
set TBD)

Unadjusted Individual chart Every batch until Feed 1: a to Not required LT range Change in cell
KPA
(raw data) with long-term long-term limits b hrs at this time TBD bank, culture
3-sigma limits set Feed 2: c to (dates: medium,
d hrs TBD) or process
Feed 3: e to setpoint
f hrs

Optional elements

Converted Individual Every batch until Characterize Not LT range Reset range for
KPP
(elapsed) Run chart long-term limits (No PPQ Required TBD Process change
set limits) (dates: to step
TBD)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 67
10.9 Step 9, Small virus retentive
filtration – CPV variables
3SD tunnel monitoring of the operating inlet pressure range within a DOE PAR, provided in the A-Mab case
ensures the parameter (and filtration flow rate) remains study. Platform knowledge of SVRF operations indicates
consistent throughout the filtration; it is not monitored as this step does not impact yield, therefore consideration is
a single point measurement. Initial control limits provided limited to any future qualification of re-filtration.
in the table below are assumed based on a fictitious normal

Table 10.9. Step 9 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-based)
limits based) (Long-
(short- term)
term)

Operating Unadjusted 3SD tunnel Every batch until Tunnel Trend every LT range Process change
CPP
Inlet pressure (raw data) long-term limits derived batch TBD to this or
set from vs. long-term (dates: previous step
PPQ and limits TBD)
platform
batches at
same scale

Filtration load Unadjusted Individual chart Every batch 640 to Not required LT range No known risk
CPP
volume (raw data) with long-term until a 1040 TBD events
3-sigma limits long-term sigma (L) (dates:
set TBD)

10.10 Step 10, Ultrafiltration and


diafiltration – CPV variables
To address the risk that future incoming buffer component for use in this process step. The A-Mab case study
lots test within specification, but outside a typical includes IPC tests for pH and osmolality in drug product
experience to cause an undesired or unpredicted variation manufacturing, but does not include BDS specifications
not controlled by the process, we propose monitoring a for these attributes. It will be assumed, based on the case
trend of the CoA reported attribute for at least 30 batches study information, that the process stream after UF/DF
from each approved supplier (manufacturer/ packager/ (rather than after Step 11) is subject to probe monitoring
distributor/ vendor) as an optional item (see ‘Excipient 1’ in for pH (PAT), to show performance prior to final filtration,
Table 10.10). If the actual range for this material attribute as an input to the drug product CPV effort (which occurs
does not exhibit a Ppk > 1.3 against its ICH Q3D limit, after BDS storage, shipping, and thawing).
further monitoring and control actions may be necessary.
TMP is a design parameter with a fixed setpoint even as
The initial control limits provided, are based on fictitious membrane use cycles increase. The permeate flow output
normal ranges to serve as early warning triggers. Buffer attribute varies and the input retentate flow parameter
pH, osmolality, and conductivity are controlled (and responds to achieve and maintain the TMP setpoint.
adjusted as necessary), before the solutions are released

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 68
Table 10.11. Step 10 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-
limits based) (Long- based)
(short-term) term)

UF/DF Converted Individual Every batch until ≤7 Trend every LT range Scale or
CPP
processing time (elapsed) Run chart long-term limits (hours) batch TBD process
set vs. long-term (dates: change to step
limits TBD)

Number of Converted Individual Every batch until 9.5 t 10.4 Not Required LT range Buffer
CPP
dia-volumes (ratio) Run chart long-term limits (DV) TBD formulation
set (dates: or protein
TBD) conc. changes

UF/DF retentate Unadjusted Individual chart Every batch until 5.2 to 5.4 Once annually LT range Step 10 or 11
CPP
final pH (raw data) with long-term long-term limits (-log [H+]) TBD process or
3-sigma limits set (dates: raw material
TBD) changes

Protein Conc. A280 Unadjusted Individual chart Every batch until 65 to 85 Trend every LT range Process
CPP
Prior to BDS fill step (raw data) with long-term long-term limits (mg/mL) batch TBD change
3-sigma limits set vs. long-term (dates: to this step
limits TBD)

Product yield, Converted Individual chart Every batch Per Trend every LT range When
KPA
Final Retentate (ratio) with long-term until a membrane batch TBD extending
3-sigma limits long-term sigma lifetime vs. long-term (dates: re-use cycles
set program limits TBD)

Optional elements

Excipient1 Unadjusted Individual Each new lot Characterize Not Required LT range Qualify new
CMA
AttributeA (raw data) Run chart until (No PPQ TBD suppliers,
30 lots tested limits) (dates: RM or CoA
TBD changes

SEC aggregates in Unadjusted Individual chart Every batch until ≤ 1.4 Not Required LT range Process
CQA
final retentate (raw data) with long-term long-term limits (%) TBD change to
3-sigma limits set (dates: this step,
TBD) extensions of
membrane
lifetime

Protein Conc. A280 Unadjusted Individual chart Every batch until 40 to 60 Not Required LT range No known risk
KPP
Prior to diafiltration (raw data with long-term long-term limits (mg/mL) TBD events
3-sigma limits set (dates:
TBD)

Recirculation Unadjusted 3SD tunnel Every batch until Profile Not Required LT range When
KPP
flow rate (raw data) long-term limits within TBD extending
set tunnel limits (dates: re-use cycles
TBD)

Permeate Unadjusted 3SD tunnel Every batch until Profile Verify LT range When
KPA
flow rate (raw data) long-term limits within when new TBD extending
set tunnel limits membranes (dates: re-use cycles
installed TBD)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 69
10.11 Step 11, Final filtration/Bulk fill
and freezing of BDS – CPV variables
The initial control limits provided in the next table are
assumed, based on a fictitious normal range and PPQ
consistency limits, or in the case of yield, an assumed limit
from fictitious platform knowledge.

Table 10.10. Step 10 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to analysis tool monitoring baseline monitoring control monitoring
(short-term) control (time/cycle- limits (change-based)
limits based) (Long-
(short- term)
term)

Bulk Fill Converted Individual chart Every batch ≥ 98 Once annually LT range No known risk
KPA
Step yield (ratio) with long-term until a (%) TBD events
3-sigma limits long-term sigma (dates:
set TBD)

Optional elements

Filtration Unadjusted Individual Every batch 200 to 300 Trend every LT range Reset range for
KPP
volume (raw data) Run chart until a (L) batch TBD scale or
long-term sigma vs. long-term (dates: process change
set limits TBD) to this
or previous step

Maximum Unadjusted Individual chart Every batch ≤2 Not Required LT range No known risk
KPP
Inlet pressure (raw data) with long-term until a (psig) TBD events
3-sigma limits long-term sigma (dates:
set TBD)

Filtration Converted Individual Every batch until ≤3 Trend every LT range Scale or process
KPP
time (elapsed) Run chart long-term limits (hours) batch TBD change to step
set vs. long-term (dates:
limits TBD)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 70
10.12 CPV monitoring of bulk drug substance lot data
As with the process steps, BR depends on a rational assessment of lot data. This is shown in the next table.

Table 10.12. Step 12 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-based)
limits based) (Long-
(short- term)
term)

Monomer Peak Individual chart Every batch NLT 98 Trend every LT range Per BDS
CQA
by SEC integration with long-term until a (%) batch TBD stability
(raw data 3-sigma limits long-term sigma vs. long-term (dates: monitoring
set limits TBD protocol

Aggregates Converted Individual chart Every batch NMT 2 Trend every LT range Per BDS
CQA
by SEC (additive) with long-term until a (%) batch TBD stability
3-sigma limits long-term sigma vs. long-term (dates: monitoring
set limits TBD) protocol

Galactosylated Unadjusted Individual chart Every batch until 15 to 35 Not required, LT range Confirm
CQA
Glycans (raw data) with long-term long-term limits (%) Not TBD comparability
3-sigma limits set stability (dates: for
and PpK >1.0 indicating TBD changes in cell
bank or
step 3 setpoints

Afucosylated Unadjusted Individual chart Every batch until 5 to 10 Not required, LT range Confirm
CQA
Glycans (raw data) with long-term long-term limits (%) Not TBD comparability
3-sigma limits set stability (dates: for
and PpK >1.0 indicating TBD) changes in cell
bank or
step 3 setpoints

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 71
Table 10.12. Step 12 CPV variables

A B C D E F G H I

Variable Class Data treatment Monitoring Initial baseline Initial Periodic Lifetime For cause
prior to tool monitoring baseline monitoring control monitoring
analysis (short-term) control (time/cycle- limits (change-based)
limits based) (Long-
(short- term)
term)

Optional elements

DNA Unadjusted Individual Every batch for None Per column LT range Per column
CQA
(raw data) Run chart 30 BDS batches detected resin Lifetime TBD resin Lifetime
protocols (dates: protocols
TBD)

Methotrexate and/or Unadjusted Individual Every batch for None Per column LT range One BDS batch
CQA
Antifoam C (raw data) Run chart 30 BDS batches detected resin Lifetime TBD for
protocols (dates: 3 raw material
TBD) lots

Sialic acid Unadjusted Individual chart Not required NMT 1.6 Not required, LT range Confirm
CQA
Content (raw data) with long-term at this time (%) Not TBD comparability
3-sigma limits stability (dates: for
indicating TBD cell bank
or process
changes

Mannose Unadjusted Individual chart Not required 5 to 8 Not required, LT range Confirm
CQA
Content (raw data) with long-term at this time (%) Not TBD comparability
3-sigma limits stability (dates: for
indicating TBD) cell bank
or process
changes

Non-glycosylated Unadjusted Individual chart Not required 0 to 2.4 Not required, LT range Confirm
CQA
heavy chain (raw data) with long-term at this time (%) Not TBD comparability
3-sigma limits stability (dates: for
indicating TBD changes in cell
bank or
step 3 setpoints

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 72
11.0
CPV sampling plan
A sampling plan makes it clear which samples need to be taken to meet the data acquisition requirements of
CPV. This section shows how this might be done giving examples and using templates. A sampling plan matrix
is presented in Table 11.0.1. Please note, this table is not intended to be complete, or entirely consistent with
information contained previously. It is an example of a visual tool that can usefully summarize an otherwise
complex plan, making it easier to develop and communicate.

Considerations for non-routine sampling • 


Sample container and container size (e.g.
and testing include: polypropylene tubes);
• 
Sample plan for routine monitoring, baseline • 
Sample volume, number of aliquots, and retains;
monitoring, time-based periodic monitoring, or
• 
Sample labelling (may be driven by site SOP);
special event / change based monitoring;
• 
Sample storage temperature and transport
• 
Sample frequency (e.g. every day during a reactor
conditions;
run, every batch, or every 5 batches, etc);
• 
Tests to be performed (including any additional
• 
Specific sampling location within the process.
sampling due to assay needs (e.g. a blank solution
Provide specific and clear instructions for collection
as reference for the test);
of the required sample, e.g. BRc, BRc step number,
sampling device such as manual sample valve or • 
Testing acceptance criteria.
automated sample valve. It is critical to ensure that
samples collected at the selected sampling points
are representative of the drug substance or process
intermediates;

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 73
Table 11.0.1. Sampling Plan Matrix

Sampling plan for process attributes

On-floor QC Product quality testing


tests Micro

Truncated impurity

Product variant
Affinity ligand

Methotrexate
Product conc.
Conductivity

Endotoxon
A280 conc

Bioburden

RP-HPLC

Antifoam
Bioassay
Retain

DNA
CIEF

HCP
CGE

SEC
pH
Vol req'd for assay

Storage temp <-40 °C

Testing offsite

Testing onsite

Method SOP

Stage 1: Cell Expansion

After last open manipulation prior


to transfer to production stage

Stage 2: Product Expression

EOR prior to harvest

Stage 3: Clarification

Centrate

Clarified filtered broth •

Step 4: Affinity chromatography

Load post hold period

Load flow-through

Elution pool • • • ο ο ο ο ο ο

Strip flow thru

Symbols are used to indicate which sampling is relevant to each A-Mab process step so the intention of sampling is clear:
• (Routine test), ο (CPV test), ∆ (Retain),  (Stability),  (Reuse Lifetime Performance),  (Cleaning Verification)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 74
Table 11.0.1. Sampling Plan Matrix

Sampling plan for process attributes

On-floor QC Product quality testing


tests Micro

Truncated impurity

Product variant
Affinity ligand

Methotrexate
product conc.
Conductivity

Endotoxon
A280 conc

Bioburden

RP-HPLC

Antifoam
Bioassay
Retain

DNA
CIEF

HCP
CGE

SEC
pH
Step 5: CEX chromatography

Load post hold period

Load flow-through

Elution pool • • • ο ο

Strip flow thru

Step 6: IEX chromatography

Load post hold period

Load flow-through

Elution pool • • • ο ο ο

Strip flow thru

Step 9: UFDF

Load pool post hold period

Permeate during concentration

Retentate pool post diafiltration • • ο

Step 10: BDS filtration and Freezing

Post hold period, prior to filter

BDS sample prior to freeze • • • • • • •


Symbols are used to indicate which sampling is relevant to each A-Mab process step so the intention of sampling is clear:
• (Routine test), ο (CPV test), ∆ (Retain),  (Stability),  (Reuse Lifetime Performance),  (Cleaning Verification)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 75
11.1 Template for specific process steps
For each sampling activity, it can be helpful to have a breakout template which shows what needs to occur (thus
a symbol on Table 11.0.1 may become a template table in itself. Examples of templates for sample collection
and testing are presented (Table 11.1.1 and 11.1.2). The rows represent the considerations for sampling and
testing and suggested alternatives. The yellow highlighted cells in Table 11.1.1 are the choices for the peak viable
cell concentration tested at Step 3 in the process (Production Culture Bioreactor. In Table 11.1.2, the yellow
highlighted cells relate to aggregates tested at process Step 7, CEX. It is worth noting that, though justification for
the choices made is not given fully in this example, justification would be expected as part of the CPV plan.
Note: Whilst a sampling template may be regarded as good practice, it is not mandatory. Readers may feel it is worth creating such
plans for CPV as a priority and extending the activity to elements of process monitoring not included in CPV on a risk managed basis.

Table 11.1.1. Template for Sampling and Testing (CPP). Completed example is for Step 3, Production Culture Bioreactor

Options

Step 3: Production
Process Step
Culture Bioreactor

Peak Viable Cell


Variable
Conc.

Classification
(Product Quality
CQA CPP KPP KPA
Attribute Or
Process Parameter)

Capacitance Probe,
Assay Method
Per Sop

Special Event Or
Time-Based
Sample Plan Routine Monitoring Baseline Monitoring Change Based
Periodic Monitoring
Monitoring

Multiple Times In A
Sample Frequency Batch (E.G., Every Once Per Batch Every X Batches
Day For Bioreactor)

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 76
Table 11.1.1. Template for Sampling and Testing (CPP). Completed example is for Step 3, Production Culture Bioreactor (continued)

Options

Sample Collection Sample Location Bioreactor

Manual Sample Automated Sample Automated


Sampling Device In-Line Sensor
Valve Valve Sampling Device

Container Materials
No Sample - On-
Of Construction Pp Tube, Sterile
Line Instrument
(Moc)

Container Size 10 Ml N/A

Sample Volume 5 Ml

Sample Replicates No 2

Sample Retain No Yes

Prepared For
Sample Handling No Aliquoted
Shipping

Sample Labeling Driven By Sop N/A

Sample Storage Temperature Ambient 2-8 C -20 C -70 C N/A

Location Manufacturing Qc Lab Development Lab Sample Control N/A

Sample
Transport Yes (Per Sop) No
Transportation

In-Situ (On-Line
Assay Testing Who Manufacturing Qc Dept Development Dept External Lab
Sensor)

Reference/Blank Reference Blank Solution Control No

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 77
Table 11.1.2. Template for Sampling and Testing (CQA). Completed example is for Step 7, Cation Exchange Chromatography (CEX)

Options

Step 7: Cation
Process Step
Exchange Chrom.

Variable Aggregation

Classification
(Product Quality
CQA CPP KPP KPA
Attribute Or
Process Parameter)

Assay Method SEC

Special Event Or
Time-Based
Sample Plan Routine Monitoring Baseline Monitoring Change Based
Periodic Monitoring
Monitoring

Multiple Times In A
Sample Frequency Batch (E.G., Every Once Per Batch Every X Batches
Day For Bioreactor)

After Eluate Is Well


Sample Collection Sample Location Mixed At The Eluate
Collection Tank

Manual Sample Automated Sample Automated


Sampling Device In-Line Sensor
Valve Valve Sampling Device

No Sample - On-
Container Moc Pp Tube, Sterile
Line Instrument

Container Size 10 ml

Sample Volume 5 ml

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 78
Table 11.1.2. Template for Sampling and Testing (CQA). Completed example is for Step 7, Cation Exchange Chromatography (CEX)

Options

Sample Collection Sample Replicates No 2

Sample Retain No Yes

Prepared For
Sample Handling No Aliquotted
Shipping

Sample Labeling Driven By Sop

Sample Storage Temperature Ambient 2-8 C -20 C -70 C N/A

Location Manufacturing Qc Lab Development Lab Sample Control N/A

Sample
Transport Yes (Per Sop) No
Transportation

In-situ (on-line
Assay Testing Who Manufacturing Qc Dept Development Dept External Lab
sensor)

Reference/Blank Reference Blank Solution Control No

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 79
12.0
How data will be analyzed
Fundamentally, the aim of setting values for capability criteria and analyzing data against those criteria is to
establish a plan that provides sound rationale for decision making. This section provides a basic introduction to
the statistical tools likely to be used in support of analytical elements of a CPV plan.

Statistics is a mature though complex mathematical 12.1 Identifying software


discipline and it is recommended that specialists are
Software supporting CPV should make it as easy as
consulted when applying the approaches described in
possible for CPV reports to be consistent with the APR,
this section. A number of references are provided, but
in order to avoid redundant effort by the technical and
given the maturity of the subject, the authors recognize
quality staff. The charts of quantitative results included in
this list is far from exhaustive. The focus here is on recent
CPV should meet all requirements of the APR, such that
regulatory guidance and what might be regarded as a few
the CPV charts may be copied and pasted directly into the
useful, standard texts.
APR or included as attachments. The APR would provide
Note: In this paper we are describing some ways in which data context and tie together the information brought forward
can be analysed, but these are not intended to be exclusive of from the CPV Reports.
any others. Other analytical methods may be better suited to
The CPV analysis may be performed using well established
particular data sets.
software such as MINITAB, JMP, Discoverant or Statistica.
CPV Reports focus on quantitative attributes and The software vendor should provide documentation of
parameters which can be monitored using statistical their quality and validation procedures if data is used to
process control charts, to identify shifts, trends, and support GMP functions18-20.
unusual results. Typical practice is that the attributes and
‘Homegrown’ analysis routines should be similarly
parameters in a CPV Report include all the quantitative
validated if used to support GMP functions. If specific
attributes and parameters which are included in the
calculations outside the base software packages are used
APR. CPV Reports can also include additional attributes
to support GMP functions, they should be validated also.
or parameters, which are useful for building process
understanding and identifying sources of variation. Whilst
the scope of quantitative attributes included in CPV is
equal to, or broader than, the scope included in APR, the
APR includes additional sections not required for CPV.
The APR will provide comprehensive assessments of
product performance that are only made once a year.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 80
12.2 Description of tools to trend and
evaluate data
Tools to trend and assist in the evaluation of CPV data rules by which decisions will be made, and the expected
include types of charts and mathematical treatments. outcomes, are recorded in the Plan. A CPV plan should be
The rationale and approaches to evaluation should created and issued following the risk assessment and before
be documented in a standardized way. The three key the start of Stage 3. As stated previously, the definition of an
documents are a Risk Assessment justification, CPV initial phase and long-term phase of CPV Plan development
Plan and CPV Report. Process Risk Assessments that may be helpful. This approach is designed to ensure that
support CPV plan development and implementation variation in process performance is understood before long-
have the important purpose of justifying the scope and term control limits are established in the control strategy.
frequency with which CPV reports are required. They Regular assessments of process performance should be
are performed at the start of CPV plan preparation. For documented in CPV Reports. The frequency with which
new processes they should draw on the outcomes of these are created will depend on the assessment of risk
PPQ and it is recommended they also take into account a as described previously. CPV Reports should detail
Process Failure Mode Effects Analysis (FMEA), which may any control alerts, as defined by the CPV Plan, whether
have been conducted during Stage 1 process design and these have led to a formal Quality investigation or not.
updated after the Stage 2 PPQ experience. Justification supporting the response made to these alerts
For legacy products, risk assessments should include should be given, including any outcomes for the control
the following sets of data: process capability, analysis of strategy. Typically, CPV Reports will contain calculations of
campaign trends, historical causes of discards, customer process capability, statistical process control (SPC) charts,
complaints and failure investigations. any other chosen charts, control limits and alerts arising
from the data.
A CPV Plan should be written with the purpose of
specifying what must be monitored to provide for CPV and In combination, these tools help make any changes in
how data should be interpreted. Interpretation should the performance of the process obvious, revealing any
involve: how data will be collected, transformed, and non-random patterns. The following sections expand on
evaluated. It is strongly recommended that data driven these tools.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 81
12.3 Process capability index
Process capability assessment evaluates the risk that an In this paper, we recommend an initial period in which
attribute will fail to meet specifications; in other words, control limits are estimated from Stage 1 and Stage 2
it quantifies the likelihood that an attribute will routinely and experience with similar processes. Given the nature
meet specifications. of Ppk, it becomes most useful as actual manufacturing
Any assessment of process capability requires the experience develops and robust control limits are
assumption that the same sources of variation that affected established for the long term.
previous results will continue to affect future results, and In cases where longer-term variation exceeds short-term
the expected range of variability does not change. variation, and the longer-term shifts cannot be tolerated,
There are many indices that measure process capability, Cpk can be used. However, Cpk should not be calculated
but two are especially popular: Ppk and Cpk . Both indices until the SPC charts provide evidence that the process is
compare the width of the specification range to the width in a state of statistical control, such that no signals of non-
of the typical variation range. The key difference is that Cpk random variation are present. This means there should be
uses a short-term estimate of variation, whereas Ppk uses no evidence of shifts, trends, or results outside of control
a long-term estimate of variation [13, Chapter 7]. These limits. This is a prerequisite to calculate a meaningful
indices take into account the centering of the process Cpk. Ppk can be calculated even when some non-random
within the specification range, and higher values of either signals are present, as long as those signals are considered
index indicates higher process capability (or lower risk of to be part of the routine, expected, longer-term variation
missing specifications). inherent to a process.

The short-term estimate of sigma is a best-case value that The index also assumes the data follow a normal
represents the minimum variation that could be achieved if distribution. Transformations are sometimes needed
all longer-term sources of variation were eliminated from to enable non-normal data to be analyzed in a rigorous
the process. The long-term estimate of sigma includes both statistical manner. A common source of non-normal
the short-term and the longer-term sources of variation. data is where negative values cannot arise, and the most
For many manufacturing processes, these longer-term frequent values are close to zero. Such data sets may be
sources of variation are an expected part of the total, termed ‘log-normal’ and taking the logarithm of the data
routine process variation. can transform it into a normal distribution. There is more
discussion on this topic in Section 12.4 that follows, but in
Cpk provides a more optimistic estimate of the potential the context of this paper, we refer the reader to Reference
process capability if all longer-term sources of variation 13, recognize there are many other texts on this topic and
are removed; Ppk provides a more realistic estimate of recommend consulting a trained statistician.
the process capability that has been achieved in routine
production. For this reason, Ppk is preferred here over Cpk When reporting process capability, a control chart of the
as an indicator of expected process capability; although same data should always be constructed to provide a visual
either measure could be used, depending on process check that the index is reasonable.
circumstances. Table 12.3 contains guidelines rating the For discussion of frequency and scope of CPV analysis see
level of control over the process based on Ppk values. Section 8.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 82
Table 12.3. Rating of PpK Index

Ppk Index Rating

>1.33 Limited Opportunity:


Attribute meets specifications with a very high
level of consistency. It may be more valuable
to look for opportunities in other areas of the
process or other processes.

1 - 1.33 Some Opportunity:


Attribute is routinely meeting specifications but
there are indications that it might not always do
so consistently.

0.68 – 1.00 Considerable Opportunity:


Attribute typically meets specifications, but does
not do so to the extent that might be expected.

≤ 0.67 Significant Opportunity:


Attribute/process cannot be expected to
routinely meet specifications.

Note: Ppk values are recognized indicators of process


capability. Although quantitative ranges have been
selected and matched with the potential for improvement
opportunities, these ranges should not replace reasonable
investigation efforts to determine the factors influencing
the Ppk value obtained. The levels presented in Table 12.3
reflect one example of a commonly accepted set of ranges for a
monoclonal antibody manufacturing process.
Other approaches to both assessing process capability as well
as matching this assessment to the potential improvement
opportunity may be more suitable for specific processes.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 83
12.4 Control charts
Control charts consist of a few simple elements: and Cumulative Sum (CUSUM) control charts are
• 
Results plotted in time order; recommended. These are easy to set up using software,
and have the advantage of using previous observations
• 
A centerline, usually at the average of the results; to filter noise and detect small shifts more quickly.
• 
Statistical control limits. However, they are slower than Shewhart charts to detect
large shifts, and are more difficult to explain to shop floor
There are many varieties of control charts. The type of
personnel and business leaders.
chart should be selected based on the type of data to be
plotted [13, Chapter 11]. Most attributes plotted for CPV Statistical control charts are based on some assumptions
are individual continuous measurements. about the process results. When these assumptions
are not met, the probability of false signals can rise
One important consideration for control charts is the
dramatically. The most important assumptions are that the
choice of time order. Results may be plotted in date of
results are independent over time and that the underlying
manufacturing order (upstream or downstream) or in
results are approximately normally distributed.
date of test order. Any of these orders can be informative,
since each order highlights sources of variation that occur In real manufacturing processes, results are rarely
in the related process steps. When processes are carried independent over time. Instead, there is some correlation
out in strict first-in, first-out sequence from upstream to across sequential results (autocorrelation). This can be a
downstream to assay, the time order will be the same and natural result of operating conditions, such as raw material
the choice of time variable has no impact on the charts. lots that are used for several upstream lots in sequence.
For CPV, generally one time order will be selected and The presence of autocorrelation can produce many small
used; other time orders may be plotted as needed for shifts and trends within the control limits; the science
investigations and process understanding. and technology process support and statistical personnel
should document in the CPV Plan whether these types of
Another important consideration is the method for
shifts and trends will be addressed as signals of unusual
calculating control limits. Control limits should always be
variation, or treated as expected routine variation.
set at plus and minus three sigma (standard deviations),
but sigma may be estimated using either a short-term The assumption of normally distributed data is also
formula or a long-term formula, the same as for Cpk and important, and should be checked using a histogram,
Ppk. For most charts, the long-term estimate of sigma is box plot, and normal quartile plot. Statistical tests for
recommended; this corresponds to the use of Ppk rather normality are not generally recommended, since they will
than Cpk. Control limits based on the long-term estimate be triggered by other issues in the data, such as occasional
of sigma will encompass the longer-term sources of outliers, or the very shifts and trends that the control chart
variation that are an expected part of total routine process is intended to identify.
variation; short-term estimates will generally be narrower, One common violation of the normality assumption is
and may produce false statistical signals when longer-term proportional variance. Proportional variance is variability
variation impacts process results. that is proportional to the level of results. For example,
Standard Shewhart control charts are simple to set up, measurements of concentration often have higher
easy to understand and explain, and good at detecting variability at higher concentrations, and lower variability
large shifts quickly. However, the Shewhart charts at lower concentrations. When variation is expressed as a
are not as good at detecting small shifts (relative to percentage or Relative Standard Deviation, RSD) instead
variation), and do not build any memory of previous of a simple standard deviation, this is an indication that the
observations. For quick detection of small shifts, EWMA variance may be proportional, and should be checked.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 84
Most common SPC tools were designed for results with These signals would require a response from the process
constant variance, not proportional variance. When owners in CPV. In the chart on the right, the same results
proportional variance is present, a simple solution is to are transformed to the log scale. The log transformation
transform results from the original scale to a log scale. expands the scale at the lower end of the range, and
Proportional variance on the original scale is stabilized to compresses the scale at the upper end of the range. Now
constant variance by transforming to the log scale. The the results are more symmetric within the limits, and
choice of natural or base 10 log scale does not matter, no results exceed either upper or lower control limits.
but one or the other should be used consistently. It is Capability indices are best calculated on the transformed
worth noting that significant data are required to make data, using specifications, mean, and sigma in the
transformation a justifiable approach. If there is any doubt transformed scale.
about there being sufficient data, transformation is better
In many situations, the average of a data set is expected
avoided and a statistician should be consulted.
to remain fairly constant over time. However, there are
The control charts below illustrate the importance of circumstances when the mean is expected to change
finding the appropriate scale for results before identifying over time, based on process knowledge and experience.
special causes of variation or estimating capability. In When this is the case, additional tools may be used to
the chart on the left, log-normally distributed data are monitor for departures from the expected behavior. These
treated assuming they are actually normally distributed. include “tool wear charts” and other similar charts based
The results are not symmetrically distributed within the on monitoring the residual result (actual – predicted by a
control limits, and there are several results outside the model). These tools will not be expanded upon further in
upper control limit. this document.

Figure 12.4.1. Control charts, showing the effect of different scales To summarize the recommendations for trending and
evaluating data:
• 
Start simply – always plot the data;
• 
Use long-term estimates of sigma for most situations,
both to set the control limits, and to evaluate process
capability (Ppk);
• 
For some special cases when standard control charts
are not providing satisfactory assessments, consult a
statistician and other process experts. These special
cases include:
– Results with low resolution (few distinct values are
possible within the specification range);
– The original scale does not provide normally
distributed data, and a scale transformation may
be needed;
– EWMA charts or CUSUM charts may be preferred
when it is important to detect relatively small shifts
in data with high variation [14, Chapter 9];
– Statistical methods may not produce meaningful
estimates, for example when a very small number
of results are available, so technically justified
limits must be used either initially or permanently.
An algorithm can be used to support the selection of
control charts (Figure 12.4.2).

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 85
Figure 12.4.2. Guide to selecting control charts

Obtain data
for an attribute

yes no yes no
At least 30 Many At least Detecting
results related results five distinct small shifts in
available? available? values? noisy data?

no yes no yes

Plot on a run chart Plot on a run chart CUSUM


MVDA
(no limits) (no limits) EWM

Shewart
chart

12.5 Multivariate data analysis


Multivariate Data Analysis (MVDA)25 combines multiple
parameters to provide greater power for detecting
changes in results, and to develop deeper understanding
of the sources of variation in a process. MVDA requires
access to larger bodies of data than univariate approaches.
When MVDA is feasible, it should be considered as a
powerful extension of CPV; it is most effective when it
can be applied to process data in real-time, while there is
still an opportunity to adjust and improve batch results.
MVDA is often used to evaluate and improve within-
batch performance, while CPV is most commonly focused
inter-batch monitoring. CPV using univariate approaches
represents the most common approach within the industry
at the moment; MVDA represents where the industry is
heading in the future.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 86
12.6 Responses to shifts and trends
CPV is only one part of an overall CS. It is envisaged to A typical path for root cause analysis in response to a
function at a supervisory level and is not typically tied signal includes:
directly to lot release. There are other basal level control • 
Establish that the results are valid;
elements such as alarms and alerts, in-process testing and
release analytics that are designed to be more immediate • 
Check for any indications of inconsistency,
controls of product quality. Any result that is Out Of e.g. within a laboratory, during the timeframe
Specification (OOS) should be investigated under existing the result was obtained;
Quality procedures for handling deviations. • 
Evaluate any other attributes that typically
CPV is intended to serve as an ‘early warning system’ correlate with the result, to determine if all
where process drift can be detected before it can cause an attributes trended together as expected, or if
OOS or failure that could otherwise have product quality the particular result was exceptional;
impact. Thus, responses to shifts and trends discovered • 
Walk the process upstream from the sample
during CPV typically include those that remain within point, and collect process performance
specifications. Investigations or other activities may be data to understand any unusual patterns in
triggered to identify the root cause of the process and/ process operations during the timeframe the
or quality shift or perturbation; however, closure of the result was obtained.
investigation triggered in this way, is not typically tied to
The explanation of within-specification shifts and trends
lot release; unless an ‘out-of-specification’ (OOS) situation
should be documented in the routine CPV Report. If the
has also occurred.
reason for a shift or trend cannot be identified during
Shifts and trends that remain within specifications should CPV, it may be escalated to the status of an official quality
be evaluated by trained personnel who are most familiar deviation, for further investigation. It may be advantageous
with the process and assay; typically floor support to define a ‘tiered’, risk-based approach linked to anticipated
engineers and laboratory scientists. The response to the actions when a shift or trend is observed. This approach
shift or trend may be determined by the local engineering could be developed over time.
or technology function, with consultation from the
quality, operations, and statistical functions. These
investigations form part of the CPV Plan and in most
cases, a formal quality investigation/deviation will not
be required, as an OOS situation will not normally have
occurred alongside a CPV trend.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 87
12.7 Establishing initial limits 12.8 Establishing long-term limits
Note: Establishing initial control limits and the choice of Once sufficient process history is established, long-term
analysis tools is also discussed in Sections 9 and 10. This section control limits should be established against which the
provides further thoughts on their application to CPV, given the performance of the process can be assessed. Long-term
importance of establishing them as part of a CPV Plan. control limits are sometimes said to be ‘fixed’ meaning
In general, statistical control limits should be set at the they should not be changed without a sound, recorded
centerline plus and minus 3 sigma (standard deviation). justification. Fixed limits should be based on a minimum
Sigma may be estimated from short-term variation [13, of 30 batches, and should reflect all expected sources
Chapter 11] or long-term variation, using the formula for of variation. If there are potential sources of significant
standard deviation. longer term variation, such as a change of raw material lot,
it is important to gather data over a sufficient time period
Long-term variation is preferred for two reasons. The to account for that variation.
most important is that long-term sigma includes all the
sources of variation that are expected to be inherent to Initial OOS results which are determined to be invalid
the process. This provides more realistic control limits, may be excluded when setting limits. Examples of invalid
which will be better able to distinguish between expected results may include laboratory errors. Such data points
and unusual instances of variation in results. The second should be excluded from sigma calculations and charts.
reason is that during initial production, when fewer than However, if the root cause is unknown or representative
30 results are available for calculating sigma, the long- of the process or testing method, the data points should be
term estimate stabilizes more quickly than the short-term retained in the sigma calculation and chart.
estimate. For independent, normally distributed results, When a process change or improvement shifts the mean
the long-term and short-term formulas for sigma will or changes the variability, control limits should be re-set
provide very similar values. based on a minimum of 30 results following the change.
During initial production or after a process change, when
fewer than 30 results are available to estimate sigma, it
is recommended that limits are set, based on technical
knowledge of the process. If statistical approaches must be
used to set initial limits, use the long-term sigma formula.
Set temporary limits to be updated once 30 or more results
are available. If longer-term sources of variation occur
over extended timeframes, more than 30 results may be
needed to capture typical long-term variation within sigma
and the control limit values.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 88
12.9 Finding signals of special
cause variation
Additional rules may be applied to a control chart to
increase the sensitivity of detecting shifts and trends
that may remain within the control limits. The Western
Electric15 and Nelson16 rules are implemented in
commercially available SPC software. Note that these
tests were designed on the assumptions of independent
successive results (no autocorrelation) and a normal
distribution. The possibility of false alarms may rise
dramatically when either of these assumptions is violated.
Low-resolution data in particular will generate many false
alarms, and the rules should not be applied for data not
meeting these assumptions. Also, the number of rules
applied should be limited, at least in the initial phase of
CPV, since the probability of false alarms increases with
each additional test applied.
Low resolution results occur frequently in regulated
processes, often because results have been rounded
before they are charted. Whenever possible, use the
unrounded result for SPC charting and estimation of
sigma. It is recommended that the validation procedure
and history of measuring systems is checked for
repeatability and reproducibility. Such data helps
decide whether rounding is appropriate and to what
extent. If in doubt, a statistician should be consulted.
When low resolution data is to be evaluated, the
recommended approach is to plot the data on a run
chart in time order, but avoid setting statistical control
limits. Limits may be set using technical judgment and
process experience17, 18.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 89
13.0
Change management
Good change management is critical to getting the greatest possible benefits from a CPV system. Some important
general principles of change management are presented here, along with some more specific scenarios related to
CPV observations.

Change management is complementary to effective setting) future long-term control limits may be necessary
CPV. Product and process-related changes may impact to manage control of changes to reduce variability or
the CPV plan (including any CPV limits document that achieve an improved outcome. A deviation which leads
might exist separately from the plan), and the RA and CS to a corrective or preventative action to begin monitoring
documents that form the basis for the CPV plan. Changes a new variable trend not previously monitored, or to re-
to these controlled documents should proceed per the activate monitoring a trend discontinued for CPV may also
company’s normal change control procedures, which should need to be added to the plan.
include the description, rationale and justification for all
Changes to the control strategy need to consider the
significant changes. Changes that do not impact monitored
potential impact to the current status of the CPV plan.
parameters, sampling points or control limits do not require
Facility, process, equipment, field measurements, or
revision of the CPV plan; however the change control
analytical laboratory method changes (examples of normal
should consider whether evaluation of normal process
change controls) require a review of the CS, any related
variability for a particular parameter should start again at
risk assessments, and the current monitoring plan for
the initial CPV phase for analysis. After noting in the change
the steps being changed as well as the downstream steps
control documentation, this may be noted in the next CPV
that have linked parameters or attributes. If a process
analysis / report.
variable is re-classified in the CS based on new process
Process changes or experience with special or common understanding, changes to the CPV monitoring plan
cause variation may require investigations and a revision may also be needed and any impact to registered details
of the CPV execution plan during the lifetime of a process. of regulatory licenses need to be addressed. Below, a
Adjustments to the frequency of periodic monitoring, or decision flow outlines a process for managing changes
a return to collecting baseline data before setting (or re- needed due to ongoing CPV monitoring (Figure 10.13).

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 90
Figure 13. Decision Flow for changes to CPV Execution Plan for Drug Substance Lifetime. OOT stands for Out-of-Trend, NOR is Normal Operating Range and
PAR is Proven Acceptable Range.

Unexpected incident Confirm OOT, investigate to


reported/impending change determine if a special cause and
notification communicated excluding data from future natural
CPV data OOT variation sigma calculation

Review control strategies and risk Is it completely No, but it is not in


assessment for actual potentially new? Not addressed CPV monitoring,
impacted control parameters and in existing risk justify adding or not
performance indicators and, if assessments or adding to program in
classification rationale is still justified, control strategies investigation or change
or if decision needs to be adjusted yet impact assessment

Does Incident/Change indicate Yes. Complete new or


No. Current status setpoints or ranges for any supplement existing
can continue (routine fixed or response variables risk assessment, revise
control or CPV need additional monitoring, control strategy
enhanced monitoring verification, requalification,
as currently in place) resetting, redefinition (setpoints,
alert/action limits or NOR/PAR
controls)

Yes, determine sample Generate Suspend any Commercial data collection/assessment


test/data capture information existing CPV to establish long term confidence in new
requirements needed to establish criteria/limits understanding, and assurance of control
initial process and replace with
understanding lower confidence
product knowledge control ranges via
(R&D, SSM, supplier QMS mechanism
data, historical data of protocal change
analysis) management

Assessments of CPV trends and CPV plan deviations In this A-Mab example, for cases where a change would
are to be documented in CPV reports. This should cancel out capability indices or remove confidence in
indicate actions taken and capture rationale to justify continued use of existing control limits, a plan deviation
any recommended CPV plan revisions. These reports would be used to document suspending or making these
are then used as inputs into a periodic APR / Product control limits obsolete. The process performance trend can
Quality Review package. Assessments may be as simple then be monitored against a new provisional control range
as chart status, or include an evaluation into whether (justified via the deviation and based on PARs, recent
a capability index value demonstrates it is possible to history, equipment capability, or qualified small scale
reduce or stop monitoring a particular parameter or model studies). Data collected prior to the change may be
attribute. If sufficient data has been obtained to calculate used for comparison to assess the impact of the change.
and implement long-term limits with high confidence,
The following table presents several cases where CPV-
this should be documented in the CPV Report, before
related changes may occur, the impacted documents and
the CPV Plan is changed. An assessment may conclude
required actions. The change management process is very
that monitoring will continue to a new planned milestone
similar to the initial setup of the CPV plan, as presented
without change setting new limits. If the capability index
in preceding sections of this paper. An assessment of the
is low, the monitoring plan may need to be changed (e.g. to
RA document and its impact on the CS document is always
obtain data more frequently, or a Corrective Action and
required, even if not explicitly stated in table 13-1 below.
Preventive Action (CAPA), might be considered to improve
control. In any case the decision should be documented in
a CPV Report with its rationale.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 91
Table 13-1. Change Management Examples

Impacted
Reason for Change Actions
Document(s)

Changes within CPV Plan

Transition from initial control limits to long-term limitsEnough CPV Plan / Limits Document (e.g. in a CPV limits document) how statistically based
data available (e.g. n = 30) to establish statistically based Document control limits (CL) were determined. Start routine monitoring with
control limits CL’s and run rules

OOT results (e.g. control limit or run rule violations) due to CPV Limits Document Justify removal of current CPV limits (if applicable), and continue to
newly experienced, but normal, variability (e.g. due to a new collect data until sufficient to recalculate new control limits. During
raw material lot) the extended data collection period, continue to monitor for trends
(e.g. average ± 3 SD), but without run rules.

Shift in mean or a change in variability (e.g. due to a process CPV Limits Document Justify removal of current CPV limits, and reset counter for the
improvement) number of runs required to set new control limits for the impacted
attribute / parameter. Continue to monitor for trends (e.g. average
± 3 SD).

Add / Remove control elements based on process knowledge RA / CS Update to reflect the new knowledge
gained through CPV, e.g. after periodic monitoring. This data
could point to ways to improve the product or optimize the
process. Elements may be deleted (or have reduced sampling CPV Plan / Limits Update. New elements may be monitored (e.g. using average ± 3 SD)
frequency) if their process capability index is high and Document until statistically based control limits can be established (n ≥ 30).
variability is well controlled. Elements may be added if a new
source of process variability is discovered.

Changes external to CPV Plan

• Add or Remove control elements based on new process PPQ Perform PPQ run(s) if deemed necessary (consult with QA and
knowledge, e.g. from lab scale studies, CAPA’s, complaints, etc. Regulatory Affairs).
• Process changes (e.g. new raw material) RA/ CS See “Add/Remove control elements” above
• Change or add manufacturing site
CPV Plan / Limits See “Add/Remove control elements” above
• New equipment (excluding like-for-like changes)
Document

New or changed analytical capabilities RA/ CS Only if impacted

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 92
14.0
Data verification
This section describes in general the types of data sources in a CPV reporting
system. It goes on to make recommendations as to how data in these systems
should be verified and hence how the systems should be validated.
Three major sources of data are used for CPV: 2. Single data entry and independent verification: The
1. Process data e.g. viable cell concentration in the data are entered from the original data source by
bioreactor26, offline pH / conductivity measurements one operator and independently verified by a second
or process volumes which are recorded in either operator using the source documents. This process
paper or electronic BRcs. is used in many companies, but cannot completely
eliminate errors because the data verifier may not
2. Analytical data generated in QC laboratories which always catch all data entry errors.
are typically recorded in a LIMS or Data Historian.
3. SMART Data entry / Error proofing: for a parameter
3. Data recorded by inline instruments e.g. pH of or attribute being transcribed, the data archival
bioreactor, or buffer flow rates for chromatography system can limit the allowable values that can be
operations. These data are archived and managed entered, thus any errors can be readily pointed out
by the data historian component of the plant control and corrections can be made, for example:
system. Examples of data archival systems include
Plant Information (PI_System) by OSISoft and • Offline pH measurements can be restricted to
values from 0.0 to 14.0;
InfoPlus 21 by Aspentech.
Data recorded on paper-based systems require
• Viable cell concentration data can be restricted
to the expected range of values26.
transcribing into a secure electronic archiving system (e.g.
a database) so that it can be retrieved in the future for Alternatively, the data archival system can generate a
data analysis, control charting and CPV. The manual data report of observed minimum and maximum values for
entry process is prone to human error. Several options a process parameter or attribute. Any discrepant result
are available to ensure data integrity during the data outside of the normally observed range can be readily
transcription process. These are described below. detected.
1. Blinded data entry: Data are independently entered While a SMART Data entry system can detect some errors,
into the data archival system by two different it is also not foolproof. Suppose the observed values for
operators. The data archival system prompts the chromatography step yield vary from 50.2 to 80.4 %. For
operators if there is a discrepancy noted between a particular batch, if the yield recorded in the source BRc
the two separate entries for a process parameter or is 62.6%; this could be wrongly entered as 66.2% which
attribute so corrections can be made. However there would still fall within the expected observed values and
is a remote possibility that both data entry operators may remain undetected.
can make the same mistake.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 93
In summary, there are hidden sources of error in Whether the source data are from paper systems
transcribing data from paper to electronic data archival or electronic systems, a record should be kept (and
systems. It is important to recognize that verification continually updated) of the BRc step number or the Tag
is critical for some parameters (e.g. CPPs) but may not ID for electronic sources. It is recommended (although
be necessary for other process parameters. Therefore not an absolute requirement) for the data archival system
verification depends on purpose and criticality of the to have the ability to record data entry operator ID /
parameter being measured. The intention should to reduce time stamp, any corrections made; i.e. an audit trail of all
data entry errors to zero. This may require continuous entries and changes.
improvement and it is important to know the extent to
Finally it would be good practice for any process
which any error impacts the process parameter.
parameter or quality attribute observed to be out of
Transcribing data from electronic sources e.g. electronic controllable range (trend) or for observed shifts and
BRcs or plant information into data archival systems, is trends to be independently verified by re-examining the
not likely to introduce errors provided the data transfer source data.
system is designed robustly and the ability to transfer
Thus in conclusion, it is recommended that robust systems
data is validated. For either paper or electronic sources,
and procedures are designed and developed in order to
the ability to retrieve data from the data archival system
archive data and validate the accuracy of data retrieval in
should undergo an initial validation. Similarly if the
order to minimize errors during CPV. It is of course, the
data from the data archival system is used routinely
responsibility of each individual organization to apply the
for generating control charts or data tables for process
recommendations in this section as they see fit.
monitoring and CPV, the procedure for generating
control charts or data tables should be validated or as
recommended in section 12 of this document, established
software should be used. A number of software suppliers
will provide a quality statements regarding the validation
of their statistical software [e.g. 19, 20, 21].

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 94
15.0
Discretionary elements of a CPV program
The Table below shows some elements that may be included within a CPV program based upon the needs and
decisions of the individual operating company. The following elements are not considered mandatory for inclusion in
CPV, but may provide the operating company with information valuable to the manufacturing of drug substances.

Table 15.1. Discretionary Elements of CPV

Element Description

Operational or Performance Elements Process performance attributes (i.e., bioreactor titer, column elution volumes) or input parameters linked
to process performance attributes.

Multivariate Data Analysis MVDA, particularly Partial least squares (PLS) regression or Principal component analysis (PCA) may be
useful to identify latent variables within the CPV data set and increase an understanding of the design
space of the drug substance manufacturing process.

Column/Resin/UF Membrane Cleaning and The full scale verification of column and/or ultra-filtration membrane cleaning and performance, out to
Performance Lifetime Verification established lifetime limits, may be included within the scope of the operating company’s CPV program.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 95
References
1 Technical Report No. 60. Process Validation: A Lifecycle Approach. PDA, Inc. 2013
2 ISPE PQLI Guidance Series Part 4
3 A-Mab: A case study in Bioprocess Development. CMC Biotech Working Group. 2009
4 FDA Code of Federal Regulations 21 Part 211, Jan. 2013
5 Process Validation: General Principles and Practices, guidance for industry, FDA (CDER, CBER, and CVM), January 2011
6 A-Mab study; Pharmaceutical Quality by Design: Product and Process Development, Understanding, and Control; By Lawrence X. Yu,
Ph. D.; Director for Science; Office of Generic Drugs; Food and Drug Administration
7 ICH Q8 Pharmaceutical Development: http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/
Q8_R1/Step4/Q8_R2_Guideline.pdf
8 ICH Q9 Quality Risk Management: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/
WC500002873.pdf
9 ICH Q10 Pharmaceutical Quality System: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_
guideline/2009/09/WC500002871.pdf
10 Technical Report No. 15. Validation of Tangential Flow Filtration in Biopharmaceutical applications. PDA, Inc. 2009
11 Technical Report No. 14. Validation of column-based chromatography processes for purification of proteins. PDA, Inc. 2008
12 Biopharmaceutical Manufacturing Facilities- Volume 6. ISPE Baseline Pharmaceutical Engineering Guide. June 2004
13 Pena-Rodriguez, ME. Statistical Process Control for the FDA-Regulated Industry, 2013. ASQ Press: Milwaukee, WI
14 Montgomery DC. Introduction to Statistical Quality Control, 2013. 7th edition, Wiley: Hoboken, NJ
15 Western Electric. Statistical Quality Control Handbook 1956, Western Electric Corporation, Indianapolis, IN
16 Nelson, LS. The Shewhart Control Chart – Tests for Special Causes, Journal of Quality Technology 1984; 16: 237-239
17 Woodall, WH. Controversies and Contradictions in Statistical Process Control, Journal of Quality Technology 2000; 32: 341-350
18 Limpert, E, Stahel, WA, Abbt, M, Log-normal Distributions across the Sciences: Keys and Clues. BioScience 2001; Vol. 51 No. 5
19 www.jmp.com/software/qualitystatement.shtml
20 www.minitab.com/en-US/support/documentation/software-validation.aspx?langType=1033
21 www.statsoft.com/services/validation-services/
22 ICH Q11 Development and Manufacture of drug substances (Chemical Entities and Biotechnological/Biological Entities)
23 http://www.ema.europa.eu/docs/en_GB/document_library/Other/2013/08/WC500148215.pdf
24 ISPE Discussion Paper: Applying Continued Process Verification Expectations to New and Existing Products”, D. Bika, P.
Butterell, J. Walsh, K. Epp and J. Barrick, 2012. https://www.google.com/url?sa=t&rct=j&q=&esrc=s&frm=1&source=w
eb&cd=1&cad=rja&ved=0CCkQFjAA&url=https%3A%2F%2Fwww.ispe.org%2Fdiscussion-papers%2Fstage-3-process-
validation.pdf&ei=FIEXU8z1Gurx0wGftoGIBg&usg=AFQjCNFvOl1WjtGX-vPHZjFsPq8n44cI3Q&sig2=QzSLW0X3-
cQvbUnwYDGYDA&bvm=bv.62286460,d.dmQ
25  uidance for Industry Analytical Procedures and Methods Validation for Drugs and Biologics, Draft, February 2014 http://www.fda.
G
gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM386366.pdf
26 European Pharmacopoeia, 2.7.29. Nucleated cell count and viability, p233

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 96
Glossary

Term Expanation Source (s)

Acceptance criteria Numerical limits, ranges, or other suitable measures for acceptance which the drug substance Q6b
or drug product or materials at other stages of their manufacture should meet to conform to the
specification for analytical procedures.

Action limits An action limit is an internal (in-house) value used to assess the consistency of the process at less Q6b
critical steps. These limits are the responsibility of the manufacturer.

Batch Records (BRc) A record of specific identifiers for the batch of material being produced, that includes all activities [4]
required to prepare for production, produce the material and close down the process. It provides
traceability of who did what, when, and the outcomes of those actions, including any observations
on or deviations from the process or anticipated results.

Batch Release (BR) The process by which the product is tested and results reviewed to ensure product quality under Q8(R2)
cGMP regulations and guidelines.

BPOG BioPhorum Operations Group, a collaboration of biopharmaceutical companies, seeking to


accelerate the rate at which the industry achieves a lean state.

Bulk Drug Substance (BDS) According to 21CFR207.3(a)(4) this means any substance that is represented for use in a drug 21CFR207.3(a)[4]
and that, when used in the manufacturing, processing, or packaging of a drug, becomes an active
ingredient or a finished dosage form of the drug, but the term does not include intermediates used
in the synthesis of such substances.

Capability of a Process (Ppk) Ability of a process to realise a product that will fulfil the requirements of that product. The Q10
concept of process capability can also be defined in statistical terms via the process performance
index Ppk or the process capability index Cpk (ISO 9000:2005).

CMC BWG Chemistry, manufacturing and control, biotech working group of the International Society for
Pharmaceutical Engineers (ISPE).

Continued Process A formal process that enables the detection of variation in the manufacturing process that might [5]
Verification have an impact on the product. It provides opportunities to proactively control variation and
assure that, during routine production the process remains in a state of control.

Control Strategy A planned set of controls, derived from current product and process understanding that assures Q10
process performance and product quality. The controls can include parameters and attributes
related to drug substance and drug product materials and components, facility and equipment
operating conditions, in-process controls, finished product specifications, and the associated
methods and frequency of monitoring and control.

Critical Describes a process step, process condition, test requirement, or other relevant parameter or Q7
item that must be controlled within predetermined criteria to ensure that the API meets its
specification.

Critical Material Attribute A material attribute, whose variability has an impact on a critical quality attribute and therefore Q8(R2)
(CMA) should be monitored or controlled to ensure the process produces the desired quality.

Critical Process Parameter A process parameter whose variability has an impact on a critical quality attribute and therefore Q8(R2)
(CPP) should be monitored or controlled to ensure the process produces the desired quality.

Critical Quality Attribute A physical, chemical, biological or microbiological property or characteristic that should be within Q8(R2)
(CQA) an appropriate limit, range, or distribution to ensure the desired product quality.

Design Space The multidimensional combination and interaction of input variables (eg, material attributes) and Q8(R2)
process parameters that have been demonstrated to provide assurance of quality. Working within
the design space is not considered as a change. Movement out of the design space is considered to
be a change and would normally initiate a regulatory post approval change process. Design space is
proposed by the applicant and is subject to regulatory assessment and approval.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 97
Term Expanation Source (s)

Detect-ability The ability to discover or determine the existence, presence, or fact of a hazard. Detect-ability is a Q9
component of a Failure Modes Effects Analysis (FMEA).

Drug product (Dosage form; A pharmaceutical product type that contains a drug substance, generally in association Q6b
Finished product) with excipients. Drug substance (Bulk material): The drug substance is the material which is
subsequently formulated with excipients to produce the drug product. It can be composed of the
desired product, product-related substances, and product- and process-related impurities. It may
also contain excipients and other components, such as buffers.

Failure Modes Effects One of the first systematic techniques for failure analysis. It was developed by reliability engineers Q8, IEC 60812 Analysis
Analysis (FMEA) in the 1950s to study problems that might arise from malfunctions of military systems. A FMEA techniques for system
is often the first step of a system reliability study. It involves reviewing as many components, reliability—Procedure
assemblies, and subsystems as possible to identify failure modes, and their causes and effects. for failure mode and
For each component, the failure modes and their resulting effects on the rest of the system are effects analysis (FMEA).
recorded in a specific FMEA worksheet. There are numerous variations of such worksheets.

General process parameter An adjustable parameter (variable) of the process that does not have a critical effect on product CMC-BWG
(GPP) quality or process performance. Ranges for GPPs are established during process development, and
changes to operating ranges will be managed within the quality system.

Impurity Any component present in the drug substance or drug product that is not the desired product, a Q6b
product-related substance, or an excipient (including added buffer components). It may be either
process- or product-related.

In-process quality attributes Parameters used in the A-Mab Case Study model of a control strategy, to provide a link between [3]
(IPQA) KPPs and KPAs

In-Process Control also called Checks performed during production in order to monitor and if necessary to adjust the process Q7
Process Control and/or to ensure that the intermediate or API conforms to its specifications.

In-process test In-process inspection and testing should be performed by monitoring the process or by actual WHO Portal
sample analysis at defined locations and times. The results should conform to established process http://apps.who.
parameters or acceptable tolerances. Work instructions should delineate the procedure to follow int/medicinedocs
and how to use the inspection and test data to control the process. /en/d/Jh1792e/
20.7.3.3.html

In-process tests Tests which may be performed during the manufacture of either the drug substance or drug Q6a
product, rather than as part of the formal battery of tests which are conducted prior to release.

Intermediate For biotechnological/ biological products, a material produced during a manufacturing process Q5c
that is not the drug substance or the drug product but for which manufacture is critical to the
successful production of the drug substance or the drug product. Generally, an intermediate will
be quantifiable and specifications will be established to determine the successful completion of
the manufacturing step before continuation of the manufacturing process. This includes material
that may undergo further molecular modification or be held for an extended period before further
processing.

Key Process Attribute (KPA) An important attribute or output measure of the process used in this paper to maintain consistency A-MaB Case Study [3]
with the language used in the A-MaB case study. N.B. It is important not to confuse a KPA, which is
a measure of process consistency with measures of quality such as CQAs.
N.B. at the time of writing, the European Medicines Agency (EMA) draft guidance on Process
Validation is out for consultation, referring to KPAs as 'performance indicators'.

Key Process Parameter (KPP) An adjustable parameter (variable) of the process that, when maintained within a narrow range, CMC BWG
ensures optimum process performance. A key process parameter does not meaningfully affect
critical product quality attributes. Ranges for KPPs are established during process development,
and changes to operating ranges will be managed within the quality system. N.B. this category of
parameter is not recognised by the FDA or the EMA for use in formal submissions and reports,
though they do not oppose its use internally. The agencies see all parameters that may have an
impact on CQAs as Critical and hence CPPs [23].

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 98
Term Expanation Source (s)

Knowledge Management Systematic approach to acquiring, analyzing, storing, and disseminating information related to Q10
products, manufacturing processes and components.

Multivariate Analysis MDVA, particularly Partial least squares (PLS) regression or Principal component analysis (PCA) may [25]
be useful to identify latent variables within the CPV data set and increase an understanding of the
design space of the drug substance manufacturing process.

Normal Operating Range A defined range, within the Proven Acceptable Range, specified in the manufacturing instructions PQRI
(NOR) as the target and range at which a process parameter is controlled, while producing unit operation
material or final product meeting release criteria and Critical Quality Attributes.

Performance Indicators Measurable values used to quantify quality objectives to reflect the performance of an Q10
organisation, process or system, also known as ―performance metrics in some regions.

Pharmaceutical Quality Management system to direct and control a pharmaceutical company with regard to quality. ICH Q10
System (PQS)

Plan A detailed description of how something is going to be done. Oxford English


Dictionary online

Potency Potency is the measure of the biological activity using a suitably quantitative biological assay (also Q6b
called potency assay or bioassay), based on the attribute of the product which is linked to the
relevant biological properties.

Prior product knowledge The accumulated laboratory, nonclinical, and clinical experience for a specific product quality CMC BWG
attribute. This knowledge may also include relevant data from other similar molecules or from the
scientific literature.

Procedure A written, established way of doing something in the operating environment. Oxford English
Dictionary online

Process Analytical A system for designing, analyzing, and controlling manufacturing through timely measurements (ie, Q8(R2)
Technology (PAT) during processing) of critical quality and performance attributes of raw and in-process materials and
processes with the goal of ensuring final product quality.

Process Control See In-Process Control. Q7

Process Robustness Ability of a process to tolerate variability of materials and changes of the process and equipment Q8(R2)
without negative impact on quality.

Process-related impurities Impurities that are derived from the manufacturing process. They may be derived from cell Q6b
substrates, culture (eg, inducers, antibiotics, or media components), or from downstream
processing (eg, processing reagents or column leachables).

Process-related impurities These are impurities that develop from, or are introduced by, the biological or chemical processes Q3B(R2),
by which the product is made.

Product lifecycle All phases in the life of a product from the initial development through marketing until the Q8(R2)
product's discontinuation.

All phases in the life of the product from the initial development through marketing until the Q9
product‘s discontinuation.

Product-related impurities Product-related impurities are molecular variants of the desired product arising from processing Q6b
or during storage (eg, certain degradation products) which do not have properties comparable to
those of the desired product with respect to activity, efficacy, and safety.

Product-related substances Product-related substances are molecular variants of the desired product which are active and Q6b
have no deleterious effect on the safety and efficacy of the drug product. These variants possess
properties comparable to the desired product and are not considered impurities.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 99
Term Expanation Source (s)

Protocol Method for carrying out an experiment and/or creating an official record of scientific or Oxford English
experimental observations. Written GMP Protocols define prospectively the conditions which will Dictionary online
be tested, sample testing plan, and acceptance criteria for results.

Proven Acceptable Range A characterized range of a process parameter for which operation within this range, while keeping Q8(R2)
other parameters constant, will result in producing a material meeting relevant quality criteria.

Quality The degree to which a set of inherent properties of a product, system or process fulfils Q9
requirements.

Quality Attribute (QA) A molecular or product characteristic that is selected for its ability to help indicate the quality Q5e
of the product. Collectively, the quality attributes define the adventitious agent safety, purity,
potency, identity, and stability of the product. Specifications measure a selected subset of the
quality attributes.

Quality by Design A systematic approach to development that begins with predefined objectives and emphasizes Q8(R2)
product and process understanding and process control, based on sound science and quality risk
management.

Quality Control (QC) Checking or testing, that specifications are met. Q7

Quality Target Product Profile A prospective summary of the quality characteristics of a drug product that ideally will be achieved Q8 (R2)
(QTPP) to ensure the desired quality, taking into account safety and efficacy of the drug product.

Quality risk management A systematic process for the assessment, control, communication, and review of risks to the quality Q9
of the drug product across the product lifecycle.

Raw material Raw material is a collective name for substances or components used in the manufacture of the Q6b
drug substance or drug product.

Reference standards/ In addition to the existing international/national standards, it is usually necessary to create in- Q6b
materials house reference materials.

In-house primary reference material: a primary reference material is an appropriately


characterized material prepared by the manufacturer from a representative lot(s) for the purpose
of biological assay and physicochemical testing of subsequent lots, and against which in-house
working reference material is calibrated.

Risk The combination of the probability of occurrence of harm and the severity of that harm (ISO/IEC Q9
Guide 51).

Risk analysis The estimation of the risk associated with the identified hazards. Q9

Risk assessment A systematic process of organizing information to support a risk decision to be made within a risk Q9
management process. It consists of the identification of hazards and the analysis and evaluation of
risks associated with exposure to those hazards.

Risk evaluation The comparison of the estimated risk to given risk criteria using a quantitative or qualitative scale Q9
to determine the significance of the risk.

Severity A measure of the possible consequences of a hazard, which is a component of a Failure Modes Q9
Effects Analysis (FMEA).

Specification A specification is a list of tests, references to analytical procedures, and appropriate acceptance Q6b
criteria with numerical limits, ranges, or other criteria for the tests described, which establishes
the set of criteria to which a drug substance or drug product or materials at other stages of their
manufacture should conform to be considered acceptable for its intended use.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 100
Term Expanation Source (s)

Specification - Release The combination of physical, chemical, biological and microbiological tests and acceptance criteria Q1a(R2)
that determine the suitability of a drug product at the time of its release.

Statistical Process Control Statistical process control (SPC) is a method of quality control which uses statistical methods. Q8 (R2), [13],
(SPC) SPC is applied in order to monitor and control a process. Monitoring and controlling the process
ensures that it operates at its full potential. At its full potential, the process can make as much
conforming product as possible with a minimum (if not an elimination) of waste (rework or
Scrap). SPC can be applied to any process where the "conforming product" (product meeting
specifications) output can be measured. Key tools used in SPC include control charts; a focus on
continuous improvement; and the design of experiments. An example of a process where SPC is
applied is manufacturing lines.

Testing plan A determination as to whether routine monitoring, characterization testing, in process monitoring, CMC-BWG
stability testing, or no testing is conducted as a part of the overall control strategy. Extended
testing plans may be put in place to demonstrate that valuable resources can be used more than
once. It may also be necessary to establish additional tests to understand sources of variation
and to demonstrate that changes to the process have addressed sources of variation that are
considered to present appreciable risk to product quality.

TPP See Quality Target Product Profile. Q8 (R2)

Viable Cell Concentration A measure of the number of viable cells per unit volume. [25]
(VCC)

Well Controlled Critical A process parameter which is controlled by process design and standardized procedures or CMC-BWG
Process Parameter (WC-CPP) automated control systems that ensure it remains within the design space of the process. It is only
likely to vary beyond the design space if there is a failure in the control system and failure modes
for this situation are likely to be mitigated.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 101
Permission to use
The contents of this report may be used unaltered as
long as the copyright is acknowledged appropriately
with correct source citation, as follows “Entity,
Author(s), Editor, Title, Location: Year”

Disclaimer
This document represents a consensus view, and as
such it does not represent fully the internal policies of
the contributing companies.

Neither BioPhorum nor any of the contributing


companies accept any liability to any person arising
from their use of this document.

The views and opinions contained herein are that of


the individual authors and should not be attributed to
the authors’ employers.

©BioPhorum Operations Group Ltd | April 2020 Continued Process Verification : An industry position paper with example plan 102

You might also like