Вы находитесь на странице: 1из 4

FOCUS ON...

MANUFACTURING

Essentials in Quality By Design


by Thomas A. Little

Q
uality by design (QbD) is a
systematic approach to drug
development. It begins with
predefined objectives and
emphasizes product and process
understanding and process control, all
based on sound science and quality
risk management (QRM). As
introduced by the US Food and Drug
Administration (FDA), QbD brings
modern drug development
methodologies to chemistry,
manufacturing, anc control (CMC)
teams working on biologics,
pharmaceuticals, and vaccines. The GRAPHIC STOCK WWW.GRAPHICSTOCK.COM

innovations associated with QbD are


not so much the development concepts • Use of modern control strategies A CLEAR LINE OF SIGHT
(which have been proven in multiple and process analytical technologies There must be a clear definition of all
industries for well over 20 years). (PAT) (1) product requirements and
Rather, such innovations are the • Continuous improvement and performance characteristics of a drug.
application of those principles in the validation throughout a product’s Information for CMC teams and
development, submission, and lifecycle. others clinical indication, stability,
manufacturing of drug products and As stated in the International product form, dose, method of
drug substances. Conference on Harmonisation of administration, and other product
QbD was introduced over 10 years Technical Requirements for requirements as stated in a quality
ago; however, regulatory agencies are Registration of Pharmaceuticals for target product profile (QTPP).
now in the process of moving it from Human Use (ICH) Q8 Pharmaceutical Critical quality attributes (CQAs) are
recommended (optional) to mandatory Development guideline, Section 2 (2): defined early in a development
in drug submissions and filings. QbD process, and they should be managed
The aim of pharmaceutical
has 10 guiding principles: and updated throughout a product’s
development is to design a quality
• A clear line of sight from clinical life cycle. A complete list of analytical
product and its manufacturing
to product release and stability methods associated with CQAs help
process to consistently deliver the
• QRM in every aspect of to clarify the line of site from CQA to
intended performance of the
development assay (Figure 1).
product. The information and
• Enhanced product understanding Systematic drug development and
knowledge gained from
• Assay understanding QRM ensure that a line of site is
pharmaceutical development studies
• Process understanding and maintained from a target product
and manufacturing experience
characterization profile to the QTPP. It continues
provide scientific understanding to
• Generation of transfer functions through CQA generation and
support the establishment of the
• Improved product specification analytical method selection, unit
design space, specifications, and
limits and justification operation characterization,
manufacturing controls.
• Robust design space and edge of specification and acceptance criteria
failure limit generation, critical process
2 BioProcess International 12(3) M ARCH 2014
parameter (CPP) and control managed to ensure patient safety and data minimize and help control risk.
selection, then finally method and drug efficacy. An effective QRM The major benefits of QRM and risk
process validation. ICH Q6, Q8, Q9, process can help manufacturers assessments are to improve
Q10, and Q11 documents (2–6) detail produce high-quality of drug products manufacturers’ ability to develop drug
the pharmaceutical development by providing a proactive means to products and drug substances and to
process and how to set limits. The identify and control potential risks to answer the questions of when, what,
QRM process is linked to product and quality during development and how much, and where additional
process CQAs because they are the manufacturing. Customer and market development is needed. Development
drug quality attributes we wish to understanding, product definition and activities reduce potential risks to
maintain. understanding, and process safety and efficacy and help
understanding are all the main manufacturers achieve the benefits of
QRM IN EVERY ASPECT objectives of QRM throughout all a drug for a targeted indication.
OF PRODUCT DEVELOPMENT development activities (Figure 2).
QRM is an essential element of drug Two key QRM principles are as ENHANCED PRODUCT
development and manufacturing follows: UNDERSTANDING
throughout a product’s life cycle. • Risk assessment should be based You should understand all critical and
QRM as stated in ICH Q9 is on scientific knowledge associated key multiple factors influencing your
designed to ensure that drug CQAs with product and process product and all primary sources of
are defined and maintained from understanding variation. The three parts of product
phase to phase during drug • The level of effort and detail understanding are system design,
development and manufacturing. Its associated with risk assessment and parameter design, and tolerance
meant to secure that changes in drug management should be commensurate design.
product formulation, definition, with the level of risk being identified System design is the selection of a
analytical methods, and associated and evaluated. technology, chemistry, active
process changes are understood and Science guides us, and facts and pharmaceutical ingredient (API), and

Figure 1: Quality target product profile and critical quality attribute (CQA) identification and line of sight

Drug Substance Critical Quality Attributes

Product Development Team Leader: Approval/Review Date: Product Development Team Members:

Test or Attribute Attribute Attribute CQA Development Process Prior


DS Component Attribute CQA CQA Measurement Capability CQA Risk
Target Upper Limit Lower Limit Knowledge
Name Purpose Method Strategy (Optional)
Active 1 10
Pharmaceutical 7
2
Ingredient (API)
3 5

4 3

5 1

Appearance 1
and 2
Description
3
4
5

Figure 2: QRM and quality by design

Quality Risk Management

Market Analysis/Opportunity Prior and Clinical Risk Assessments


Clinical Benefits — Indication Knowledge Before Development Process Control: Critical process
Voice of the Customer (VOC) Analytical Method parameter selection, process
Safety and Efficacy Data Development capability, method and
Use Environment
Modes of Administration Precision, Accuracy, Linearity, process validation, input and
Product Requirements Selectivity, Standards, LOQ, material control, feed forward,
Stability and Supply Chain
LOD, and Controls feed back or in-situ control
CQA
Generation tracking and trending
Process Development
Release Testing: In-process
Target Product Profile DoE for process characterization,
test, OOS, and limits, technology
Formulation, set points, NOR/PAR, and
design space transfer, and equivalence
Stability, and Dose

Customer Understanding Product Understanding Process Understanding

M ARCH 2014 12(3) BioProcess International 3


materials as well as an understanding controlled. Experimental design and (Figure 3).
of impurities found in a drug product partition of variation can be used to
and drug substance. understand and minimize assay PROCESS UNDERSTANDING
Parameter design is the selection of variation. AND CHARACTERIZATION
key targets, set points, and Precision, accuracy, linear range, Factors influencing a production
concentrations. limit of quantitation (LOQ ), limit of process and associated variations
Tolerance design is the allowable detection (LOD), and percent of should be understood. Process
range or limits for each parameter. tolerance must be defined and understanding has the same three
Capability assessment indicates how validated during method development. components as product understanding:
well you can meet defined tolerances. Reference standards and assay controls system, parameter, and tolerance
The enhanced approach is systematic, must be determined to ensure that data design. Process understanding is a
with a mechanistic understanding of generated from the assay are prerequisite to stage 1 process
the multivariate factors influencing dependable and do not confuse product validation.
products. and process variation with variation With a QbD approach, some
from the analytical method. Relative process parameters may be adjustable
ASSAY UNDERSTANDING standard deviation or coefficients of within a defined design space. Many
Care must be taken to properly select variation alone are not sufficient processes have both dynamic and
analytical methods for measuring all measures of assay accuracy. They static properties. Understanding and
CQAs. Each selected assay must be fit should be used with other measures of controlling process dynamics is at the
for use and valid for the condition to assay validity and systems suitability. heart of an enhanced approach and
be measured. During method Accuracy to precision profilers should will reduce variation at the product
development, key factors and process be used to explore assay design space and process target. CPPs and their
steps that influence method variation and visualize whether a method is associated effect size must be
must be identified, optimized, and acceptable relative to product limits identified and controlled during
process development.
Figure 3: Accuracy to precision modeling
GENERATION OF TRANSFER FUNCTIONS
Accuracy to Precision, Prediciton Profiler Manufacturers should understand how
% Measurements within Tolerance

98.95%
X factors influence Y responses, where
100
each is in the form of an equation and
90 either reflect scientific knowledge or
be empirically derived from structured
80
multiple factor experimentation.
70 Ultimately, knowledge should be

60 Equation: Transfer function from a drying


process
50 0.704 0.994
15.1468749681755 +
–3 –2 –1 0 1 2 3 0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 –0.001311354985*Solution Conc from QC[OD/mL] +
0.00562627469256*Primary Drying Shelf Temp[°C] +
Bias Precision –0.0263141752003*Primary Drying Pressure [mTorr} +
[Primary Drying Shelf Temp [°C] – 30]*
Accuracy to Precision [[Primary Drying Shelf Temp [°C] – 30]* 0.00654278237764]
Horiz. Vert. Factor Current X + –0.0757524304844 * Total Drying Time [hrs]
Bias 0
Precision 1
Response Contour Current Y Lo Limit Hi Limit Figure 4: Design space
% Measurements within tolerance 0.95 0.9973002 0.95
Coupling Reagent Contact Time (min)

6.2-
1.5 6.0-
5.8-
Precision

1.0 5.6-
ion
r e cis 5.4-
0.5 0 P
Bias 5.2-

% Measurements within tolerance 5.0-


Yield, OD/mmol
–3 –2 1 0 1 2 3
-

Bias 55 60 65
Activator Volume (mL)

4 BioProcess International 12(3) M ARCH 2014


expressed in the form of an equation CONTROL STRATEGIES AND PAT teams. Until manufacturers have
to be fully useful (Equation box). Controls are defined to ensure successfully used QbD, achieved
Many software packages such as potency, purity, safety, and efficacy. positive regulatory submissions, and
SAS/JMP will generate the transfer The three parts of a control strategy produced good outcomes in their
function during process are in-process test and release, GMP operations, they will have some
characterization and experimentation. postprocess test and release, and degree of fear or uncertainty in QbD’s
closed-loop process measurement and use. Consulting and training are
IMPROVED PRODUCT SPECIFICATION adjustment during processing. The typically used to fill the gap until the
LIMITS AND JUSTIFICATION first two are traditional, but closed- teams have sufficient experience to do
Specification limits must be a part of loop in-process controls are relatively the development on their own.
an overall product control strategy. new to good manufacturing practice The QbD framework for drug
They should be linked to CQAs and (GMP) operations. Adjustment of development became mandatory in
the QTPP (line of sight). These limits temperature, pH, time, and amount of 2013 from the FDA and EU. For now,
should be based on scientific a key ingredient are all examples of completing a design space is optional.
knowledge, transfer functions, and/or enhanced closed-loop controls.
statistical distributions. Ideally, a Closed-loop controls need four REFERENCES
transfer-function approach to setting parts: sensors to detect deviation from 1 PAT: A Framework for Innovative
limits is best. Using statistical target in a product or process, alarms Pharmaceutical Development, Manufacturing,
and Quality Assurance. US Food and Drug
distributions to set limits is a to determine when to adjust and when Administration: Rockville, MD, September
reasonable approach when all unit to leave alone, control logic and 2004.
samples indicate good safety and adjustment rules (how much 2 ICH Q8: Pharmaceutical Development.
efficacy and no harm relative to adjustment to make), and verification International Conference of Harmonisation of
clinical outcomes. If edges of failure that adjustments are correct and Technical Requirements for the Registration of
are detected, then statistical effective. The PAT framework is a Pharmaceuticals for Human Use: Geneva,
Switzerland, November 2009.
distributions should not be used. good guide for how closed-loop
3 ICH Q6: Specifications: Test Procedures
controls should be defined and and Acceptance Criteria for New Drug Substances
ROBUST DESIGN SPACE validated. and New Drug Products: Chemical Substances.
AND EDGE OF FAILURE International Conference of Harmonisation of
ICH Q8 defines design space as “The CONTINUOUS IMPROVEMENT Technical Requirements for the Registration of
multidimensional combination and AND VALIDATION Pharmaceuticals for Human Use, May 2000.
4 ICH Q9: Quality Risk Management.
interaction of input variables (e.g., Tracking and trending have generally
International Conference of Harmonisation of
material attributes) and process been the method for monitoring drug Technical Requirements for the Registration of
parameters that have been products and drug substances to assess Pharmaceuticals for Human Use: Geneva,
demonstrated to provide assurance of lot variation and process trends. Switzerland, June 2006.
quality” (2). That definition, however, Properly designed, such systems and 5 ICH Q10: Pharmaceutical Quality
has a logical flaw: Design space is a controls will lead to preventative System. International Conference of
Harmonisation of Technical Requirements for
visualization of the average action rather than corrective action
the Registration of Pharmaceuticals for Human
performance at fixed set points. In before OOS events and lot failures. Use: Geneva, Switzerland, April 2009.
truth, it often may misrepresent the Measures of process capability, 6 ICH Q11: Development and Manufacture
edges of failure and the actual process variation, and controllability will of Drug Substances. International Conference of
dynamics at play during processing. continuously validate a process and Harmonisation of Technical Requirements for
Simulation and edge-of-failure demonstrate it is fit for use. They also the Registration of Pharmaceuticals for Human
Use: Geneva, Switzerland, Draft. 
analysis augment design-space provide acceptable process outcomes.
visualization to ensure that the
allowable range of variation will cause CLOSING THE GAPS Thomas A. Little, PhD, is president of
no harm and will not result in out-of- QbD is a modern, updated, and Thomas A. Little Consulting, 12401 North
specification (OOS) conditions. enhanced method of drug Wildflower Lane, Highland, UT 84003;
Design space is proposed by an development, manufacturing, and 1-925-285-1847; drlittle@dr-tom.com.
applicant and approved by a regulatory control. It closes many gaps associated
agency. A design space allows a with traditional drug development and
manufacturing operation to adjust provides higher levels of quality For electronic or printed reprints, contact
Rhonda Brown of Foster Printing Service,
parameters within the space to improve assurance, reduced variation, and rhondab@fosterprinting.com, 1-866-879-9144
drug substance consistency and control. improved control. x194. Download low-resolution PDFs online at
Proven acceptable ranges and normal The industry is currently www.bioprocessintl.com.
operating ranges must be set and experiencing a “knowledge and
visualized relative to the particular experience deficit” regarding the use
design space. of QbD principles for many CMC

6 BioProcess International 12(3) M ARCH 2014

Вам также может понравиться