Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/257777621

The Application of Science- and Risk-Based Concepts to Drug Substance


Stability Strategies

Article  in  Journal of Pharmaceutical Innovation · December 2012


DOI: 10.1007/s12247-012-9135-9

CITATIONS READS

10 933

6 authors, including:

Stephen Colgan Jon Beaman


Pfizer Pfizer
38 PUBLICATIONS   437 CITATIONS    9 PUBLICATIONS   46 CITATIONS   

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Jon Beaman on 01 August 2016.

The user has requested enhancement of the downloaded file.


J Pharm Innov
DOI 10.1007/s12247-012-9135-9

CASE REPORT

The Application of Science- and Risk-Based Concepts


to Drug Substance Stability Strategies
Stephen T. Colgan & Timothy J. Watson & Robert D. Whipple &
Roger Nosal & Jon V. Beaman & David M. De Antonis

# Springer Science+Business Media New York 2012

Abstract The International Conference on Harmonization Introduction


(ICH) has provided practical guidance on the amount and type
of drug substance stability data needed to support marketing The International Conference on Harmonization (ICH) Q8
applications (International Conference on Harmonization states that pharmaceutical development should include, at a
2001, 2002, 2003a, b). Additional guidance has been issued minimum, a definition of the quality target product profile as
by the World Health Organization (WHO 2009). Recent sci- it relates to quality, safety, and efficacy, considering stability
entific advances and practices have resulted in improved and several other factors. Q8 also states that a greater prod-
scientific understanding of the chemical and physical attrib- uct understanding can create a basis for more flexible regu-
utes that contribute directly or indirectly to drug substance latory approaches, the degree of which is predicated on the
stability. Combining this improved understanding with the level of relevant scientific knowledge. Although Q8 covers
science- and risk-based approaches detailed in ICH Q8, Q9, drug products, its principles can be applied to drug sub-
and Q10 allows for alternative and more scientifically stance as well. ICH Q11 recently issued at stage 4 states that
driven approaches to meet the scientific and regulatory objec- drug substance critical quality attributes typically include
tives for drug substance stability (International Conference on those properties or characteristics that affect identity, purity,
Harmonization 2005, 2008, 2009). In this paper, proposals are biological activity, and stability. In addition, changes to the
presented to more fully leverage enhanced product knowledge manufacturing process should be evaluated for impact on
to design improved stability strategies. The chemical and the quality of the drug substance. The evaluation should be
physical attributes that potentially impact drug substance sta- based on scientific understanding of the manufacturing pro-
bility are discussed, and strategies that leverage accelerated cess and should determine appropriate process point(s) and
stability studies are presented. testing to analyze the impact of the proposed change.
In regulatory filings, formal stability studies are designed
Keywords Stability . Drug substance . Stability-related to evaluate quality over time under the influence of temper-
quality attributes . Control strategy ature, humidity, and light and to facilitate the establishment
of appropriate controls, retest periods, and underwrites pack-
aging and storage recommendations. Drug substance stabil-
Some of these concepts were presented at the AAPS Current Trends in ity is related to inherent properties of the specific compound,
Stability Workshop (Washington, D.C., September 2009): Colgan S. as well as its stability-related quality attributes. A stability-
“The Application of Quality by Design’s Science and Risk Based
Concepts to API Stability Strategies” related quality attribute (typically a subset of quality attrib-
utes) directly or indirectly impacts stability but may or may
S. T. Colgan (*) : T. J. Watson : R. D. Whipple : R. Nosal :
not impact other considerations (such as manufacture of the
D. M. De Antonis
Pfizer Global Research and Development, drug product). Identification, quantification, and control of
Groton, CT, USA the stability-related quality attributes ensure drug substance
e-mail: stephen.t.colgan@pfizer.com quality over time.
There are potential benefits for the pharmaceutical industry,
J. V. Beaman
Pfizer Global Research and Development, global regulators, and patients that could be realized by adapt-
Sandwich, UK ing scientific stability strategies that leverage understanding of
J Pharm Innov

these chemical and physical attributes which can impact drug most suitable drug substance form and understanding and
substance stability. Consistent with the Q8 and Q11 principles, controlling the stability-related quality attributes of the drug
a fundamental understanding of stability, coupled with risk- substance.
based- and prospective control strategies, could reduce the
overall number and/or type of stability studies (regulatory
flexibility), while increasing confidence in the proposed drug Identification of Stability-Related Quality Attributes
substance storage retest periods.
Understanding which attributes or variables that do not Regulatory stability guidances are based on general princi-
directly or indirectly affect drug substance stability is as ples of stability and chemical degradation, and were written
important as well because one can justify that these can be primarily to harmonize regulatory standards for marketing
controlled through internal (non-regulatory) quality systems. applications. These guidances describe what is expected in
This approach will allow industry and regulators to focus on terms of the number of batches, time on stability, stability
the drug substance variables and attributes that have the high- conditions, and indicate that “Stability studies should in-
est probability of affecting stability and potentially safety and/ clude testing of those attributes of the drug substance that
or efficacy as well. Examples of variables that generally do not are susceptible to change during storage and are likely to
directly or indirectly affect drug substance stability include the influence quality, safety, and/or efficacy.” Interestingly, cur-
synthetic route, processes, site of manufacture, and batch size. rent guidance does not necessarily require an understanding
Here, the stability-related quality attributes are unaffected as of the attributes most critical to the stability. Identification of
these variables change. In summary, the stability-related qual- those attributes that directly or indirectly contribute to drug
ity attributes need to be identified, understood, and controlled. substance stability is critical to establishing the fundamental
The other factors mentioned above (synthetic route, etc.) can understanding and design of a robust control strategy for
then be assessed to determine whether they impact the stability. Stability-related quality attributes can be catego-
stability-related quality attributes. This concept can be applied rized into those with the potential to directly or indirectly
during development, for marketing applications, and for post- impact drug substance stability and those that are unlikely to
approval changes. impact stability.
Attributes with the potential to impact drug substance
stability include ratio of polymorphs, degree of crystal-
Control Strategies for Stability linity, surface area, moisture content, and identity of
residual solvent and/or level. Certain impurities may
A control strategy as defined in ICH Q10 is “A planned set cause degradation (i.e., acids/bases, metals) and in rare
of controls, derived from current product and process un- cases may improve stability (e.g., antioxidants). In many
derstanding that assures process performance and product cases however, these attributes will not alter drug substance
quality. The controls can include parameters and attributes stability.
related to drug substance and drug product materials and Attributes unlikely to impact drug substance stability
components, facility and equipment operating conditions, include appearance, physical attributes (i.e., flowability,
in-process controls, finished product specifications, and the bulk density), or minor changes in the impurity profile. Note
associated methods and frequency of monitoring and con- that while an attribute like appearance is unlikely to affect
trol.” Although not explicitly stated in Q10, the concept of a stability (batches of different color will demonstrate similar
control strategy can also be applied to drug substance and its behavior on stability), appearance is an attribute that may
related stability. change on stability. Instability usually is initially detected by
The foundation of a comprehensive control strategy for changes in degradant levels but also can be manifested by
stability is the proper selection of a stable drug substance. changes in other attributes such as assay or solid form.
Ideally (from a stability viewpoint), the polymorph of the Process changes that result in new or increased levels of
drug substance should be the lowest energy (most stable) process-related impurities usually have no effect on the
form. The drug substance form chosen (e.g., counter ion) overall stability of the drug substance because these impu-
may need to balance stability and other considerations such rities are chemically inert. As such, changes in the impurity
as bioavailability and ease of manufacture of the drug prod- profiles should be carefully assessed relative to the specific
uct. Project teams should then work to understand and impurity and its reactivity with the drug substance. Al-
control the stability-related quality attributes that affect the though each drug candidate should be evaluated on a case-
inherent stability of the drug substance. Although drug by-case basis, additional long-term stability studies to sup-
substance instability can be managed downstream (in the port changes to the attributes detailed above are likely to be
drug product or though protective packaging), this may not of low value and unlikely to provide any benefit related to
be as effective as controlling instability by selecting the patient safety.
J Pharm Innov

As detailed earlier, changes to the synthetic route, site of Table 1 Summary of regulatory expectations for registration stability
manufacture, or batch size generally do not affect drug Regulatory Comments/experiences
substance stability unless the changes result in a change in governance
a stability-related quality attribute. As the synthetic process
is developed and drug substance attributes change from FDA/ICH Three pilot-scale batches by the same synthetic
route, method of manufacture and procedure
batch-to-batch, project teams should consider short acceler- and representative of commercial route. Although
ated studies (vide infra) to evaluate if the change has altered 12 months is the expectation, 6 months may be
drug substance stability as a result of a change in a stability- acceptable in some cases.
related quality attribute. By conducting side-by-side scien- WHO Three pilot-scale batches by the same synthetic
tifically based stability challenges as the drug substance route, method of manufacture and procedure and
representative of commercial route.
process evolves, a constantly expanding stability knowledge
Japan Although not an actual requirement, there is a JP
base will be established. If the stability of the new batch is
expectation that “pilot-scale” is 10 % commercial
different, a traditional stability study should be considered. scale.
The practice of evaluating process changes with short ac- China Expectation that “pilot scale” is equal to amount of
celerated studies also can minimize the number of “low API needed to manufacture 10 % of the
value” traditional long-term stability programs either commercial Drug Product scale.
for batches with equivalent stability or unacceptable Brazil At least one batch should be made at the proposed
stability performance. A low-value stability program is charac- commercial site
terized as one that does not provide an improved understanding
of stability.
approaches can be used when scientifically justified. Similarly,
ICH Q8, Q9, and Q10 encourage science- and risk-based
Current Regulatory Guidance and Expectations approaches which can lead to innovation and continual im-
provement. Unfortunately, these scientifically sound alterna-
The ICH guidances for stability (Q1A through E), which are tive approaches are often not fully embraced globally in lieu of
intended for commercial product registration, outline gener- the more conservative prescriptive interpretation of the gui-
al expectations for batch selection, storage conditions, and dances. The flexibility to use alternative approaches as de-
testing frequency for accelerated and long-term drug sub- tailed in Q1A can be partially offset by different regional
stance stability programs. Stability programs should include interpretations of the guidelines, and supply chain logistics
at least three batches of drug substance manufactured to a can be challenging when flexibility is not globally accepted.
minimum of pilot scale by the same synthetic route, method
of manufacture, and procedure that simulates the final pro- Traditional Industry Practices
cess used for production batches. The overall quality of the
batches of drug substance placed on formal stability studies Clinical Stability
should be representative of the quality of the material to be
made on a production scale. Unfortunately, interpretation of A typical industry practice has been to place each clinical
ICH guidances varies within industry, from country-to- drug substance batch manufactured using a new synthetic
country and between regulatory agencies. To avoid risk, route or process on both accelerated and long-term stability.
both industry and regulators have often adopted a conserva- These batches are then monitored for various quality attrib-
tive interpretation of these guidelines which does not focus utes (e.g., assay, purity, polymorphic form, water content,
on the fundamentals of stability relative to the manufactur- etc.) regardless of whether or not these attributes are
ing design space. Table 1 below outlines some of the differ- stability-related or are impacted by the change in the route
ent regional interpretations of registration stability or process. In many instances, this conservative approach to
expectations for new chemical entities (NCEs). selection of drug substance stability lots has led to an
In addition to the expectations for stability data detailed unwieldy number of clinical stability programs, with little
above, regulatory authorities on occasion have requested regard to the actual scientific justification or value of the
supplementary confirmatory stability studies when there data (Li et al., in preparation).
have been concerns that these assessments are insufficient
to ensure quality through expiry/retest period. Registration Stability
While the ICH Q1A details reasonable expectations for
the type and quantity of stability data required to establish a Based on what may be considered a conservative interpre-
drug substance retest period, flexibility is allowed to encom- tation of the ICH guidance, some companies have perceived
pass a variety of practical considerations, and alternative a regulatory expectation that the drug product stability
J Pharm Innov

batches be manufactured with the same drug substance When the traditional stability strategies detailed above
batches used for the registration drug substance stability are considered, several questions emerge:
programs. As such, to achieve a minimum of 12 months of
drug product and drug substance stability data for three & Are these practices actually providing useful information?
pilot-scale batches using the proposed commercial route & Can we leverage our enhanced understanding when we
and processes, companies often manufactured three consec- approach stability studies?
utive batches in their commercial facility 18+ months prior & Are there alternative (perhaps better) ways to meet our
to product registration. Even though the material may be stability commitments?
used for ongoing clinical trials and commercial product
launch, there is typically a significant expense associated Stability Strategies that Leverage Enhanced Understanding
with use of the commercial facility, as well as the risk of
unanticipated project termination. Additionally, the use of Clinical Stability
three consecutive batches manufactured using the same
batches of starting materials, manufacturing equipment, As an alternative risk-based approach to supporting clinical
and manufacturing process does little to demonstrate stabil- programs, only those changes which affect potential
ity robustness over several years of commercial production stability-related quality attributes (e.g., polymorph, surface
(Fig. 1, top). The batches produced with the same drug sub- area) would trigger the need for a subsequent batch to be
stance starting materials, manufactured in the same equipment, placed on stability. Early in development, the stability-
by the same staff will be essentially identical. This approach related quality attributes may not be fully understood, and
provides limited statistical value, limits understanding of the in those instances, accelerated stability studies may be un-
stability-related quality attributes, and limits understanding of dertaken to determine whether stability performance has
the manufacturing design space. been affected before committing to long-term stability stud-
ies. These studies are particularly valuable to efficiently
Post-approval Stability demonstrate equivalent or superior stability performance
during the development process in lieu of or in addition to
Stability studies are traditionally carried out on the first three long-term stability programs.
commercial production batches (if not part of the original Although the harsh conditions used in forced degradation
registration) and at least one batch annually thereafter to conditions may generate degradants that are never seen when
confirm the commercial retest period. Drug substance batches the drug substance is stored under normal conditions, stress
were also traditionally set up and evaluated for stability to testing and forced degradation studies can help identify po-
support site changes. tential degradation products, pathways, and the intrinsic
Impurity Levels

z
nt
e
lv

Water Content
So

Initial 3 months 6 months 9 months 12 months


Impurity Levels

nt
e
lv

Water Content
So

Retest Limit 3 Batches w/ Similar Material Attributes

Internal Limit 3 Batches w/ Different Material Attributes

Fig. 1 Manufacturing three consecutive batches produced using the with different processes or ingoing materials helps to identify stability-
same manufacturing parameters and ingoing materials will result in drug related quality attributes which require control. The batch represented by
substance with similar stability-related quality attributes. On stability, the yellow ball is less stable than the other two batches and is illustrative
these batches demonstrate essentially equivalent behavior over time of a batch that has a stability-related quality attribute that requires control
(top row). Alternatively, using developmental materials manufactured
J Pharm Innov

stability of the moiety. These studies also facilitate develop- stability-related quality attribute was not controlled (Fig. 2)
ment of the analytical procedures capable of detecting these and (2) batch-to-batch variability at extreme conditions
degradants if they actually do occur. Similar predictive stabil- (Fig. 3), but equivalent retest periods under normal storage
ity studies may also be used to identify and assess stability- conditions (Fig. 4). A third example leveraging accelerated
related quality attributes. Identification of these attributes dur- stability conditions to evaluate equivalence between batches is
ing the development process provides the opportunity to con- shown below. For this drug substance, stability is demonstrat-
trol these attributes as part of the overall manufacturing design ed for both batches even under extreme conditions (Tables 2
space strategy and minimize lot-to-lot variability. and 3). These data are consistent with a very stable drug
Accelerated stability assessment program (ASAP) [9] is substance with all of the stability-related quality attributes
an example of a predictive stability tool that has been used consistent between batches.
to identify stability-related quality attributes. As an exam- Using ASAP or other accelerated stability challenges,
ple, for a specific drug substance process in development, stability issues that traditionally would have only been
sodium thiosulfate was generated at differing levels as a detected after extended time can be rapidly detected. Chem-
function of the crystallization process. Figure 2 illustrates ists can quickly determine what are the stability-related
that higher levels of this impurity significantly improved the quality attributes and the impact of changes to stability-
overall stability of the drug substance and minimized for- related quality attributes with process changes. This type
mation of the primary degradation product on stability. This of information can be used during development to address
study revealed that the levels of sodium thiosulfate (a questions such as:
stability-related quality attribute) must be controlled to min-
imize lot-to-lot variability on stability. & Can this batch of drug substance be used to manufacture
Although Fig. 2 illustrates how an accelerated study was drug product (or is a rework/reprocess of the drug sub-
used to demonstrate apparent batch-to-batch differences in stance advised)?
drug substance stability performance, this represents the & Should this batch of drug substance be set up on a
exception to the usual case wherein ASAP demonstrates formal stability program?
similar stability behavior between batches. & With enhanced product knowledge, if subsequent drug
Figure 3 illustrates the stability data from another ASAP substance batches are placed on stability, would a leaner
experiment for a different drug substance. In this case, differ- stability strategy be appropriate?
ences in degradation rates from batch-to-batch are observed With these questions in mind, the following stability
under extreme conditions of high temperature and humidity. strategies are advocated for stable drug substance stability
Interestingly, using the ASAP moisture-corrected Arrhenius programs to support clinical trials (if adequately justified):
equation to model the long-term stability of these same data
(Fig. 4), there is little batch-to-batch variability in the pre- & Initial retest period assignment of 18 months based on
dicted review periods, regardless of changes in the manufac- purposeful degradation studies, in silico modeling, or
turing process. stressed conditions (70 °C/75 % RH)
The examples detailed above illustrated (1) batch-to-batch & Stability on first good manufacturing practice (GMP)
variability of a relatively unstable drug substance because a batch set up prior to release, at accelerated, long term,

Fig. 2 Results of accelerated Lot-to-Lot Variability in


stability assessment program Primary Degratation at (30°C/60% RH)
used to reveal lot-to-lot variabil- 14
ity based on experimentally
designed process changes. Lot 4 12
had the highest levels of sodium Lot 1 Lot 2
% Degradation Product

thiosulfate and was the most sta- Lot 3 Lot 4


10
ble. As the levels of sodium Lot 5 Lot 6
thiosulfate decreased, instability
8
increased. Lot 2 had the lowest
levels of sodium thiosulfate;
6
30 °C/60 % RH is considered an
accelerated condition for this
4
drug substance because it is a
relatively unstable compound
2

0
Initial Day 20 Day 30 Day 44
Time at 30°C/60%RH
J Pharm Innov

Fig. 3 Stability data from an Use of Accelerated Stability Assessment Programs to


ASAP experiment showing lot-
to-lot variability under extreme Evaluated Impact of Process on Stability Performance
3.5
conditions of high temperature
High Shear Wet Mill
and humidity. Air classified 3 Sonocrystallisation

Impurity Levels (%)


milling (blue) shows signifi- Un-milled
cantly higher impurity levels 2.5 Air Classified Milling
after storage under several
2
different conditions
1.5

0.5

70C/5%RH

80C/5%RH

80C/5%RH
50C/75%RH

60C/40%RH

70C/75%RH

70C/75%RH

80C/40%RH

80C/40%RH

80C/40%RH
60 days 60 days 40 days 30 days 60 days 30 days 60 days 20 days 40 days 60 days

Storage Conditions

and 1 condition below long term for trending across selected as the drug substance synthetic process is devel-
temperatures. Reduced testing employed at initial, 3, oped and ideally should vary in the levels of the stability-
12, 24, and 36 months. But no testing at 6 weeks, 6, 9, related quality attributes, as long as the attributes remain
or 18 months. within their control limits. In addition, these batches should
& No external standard assay testing if the lot on stability also vary in the levels of attributes that are not stability-
is the same as that used for the working standard. related as these studies can validate that these attributes do
& For route changes, if scientific justification has deter- not have an effect on stability. An example of this would be
mined that the original batch placed on stability remains to select batches for stability that vary in the level of residual
representative of the new route, then the new batch is not solvent. Changes in residual solvents levels generally will
placed on stability. Otherwise, a reduced testing strategy not have an effect on the stability of the drug substance.
is prosecuted to justify the drug substance retest period Batches should not be specifically made for stability studies
assignment. but should be scientifically selected as batches are made
during development to support clinical trials, safety studies,
Registration Stability site qualification, or for other uses. The registration stability
package can be further strengthened by including additional
The stability studies conducted to support registration supportive stability data on batches made with processes
should be information rich. Batches should be scientifically that differ from the commercial process but can help support

Fig. 4 Results of ASAP Retest Period Estimates Based on Modeling of Degradation Product
statistical model showing in Batches Manufactured using Different Milling Processes
estimated retest periods based on
14
the same data presented in Fig. 3.
This clearly demonstrates that
despite significant changes in 12
Air Classified Milling
impurity profiles under extreme
High Shear Wet Mill
stability conditions, batches from 10
Retest Period (years)

all four processes are expected Sonocrystallisation


to have a similar retest period 8 Un-milled
under milder storage conditions.
If the drug substance storage 6
conditions were specified as
25 °C/60 % RH, all batches could
4
easily achieve a conservative
retest period of 5 years
2

0
25C/60% RH 30C/65% RH 40C/75% RH
Storage Condition
J Pharm Innov

Table 2 Stability results for batch # 1 that matrixing for drug substance stability will be of limited
Test 70 °C/75 %RH 80 °C/40 %RH utility and traditionally would not be considered by most
Initial 14 Days 14 Days pharmaceutical companies for registration stability. Certain-
ly, a matrixed approach would not be suitable for a com-
Appearance Off-white Off-white Off-white pound that has moved quickly through development with
powder powder powder
limited product understanding. For cases where the appli-
Assay 99.5 99.0 99.5
cant will propose supplying less than 12 months data at the
Impurities
time of the filing, a matrixed approach may not be suitable
(% area)
either. That said, for drug candidates that have been in
(a) NMT 0.05 NMT 0.05 NMT 0.05
development for long periods, for those with extensive
(b) NMT 0.05 NMT 0.05 NMT 0.05
product knowledge, or for those cases with a large number
Water content 0.24 0.18 0.21
(%) of drug substance batches that have been monitored during
PXRD Form A Form A Form A development, a matrixed strategy for the registration stabil-
ity batches should be considered.
When considering risk and risk management for registra-
proposed retest dates and the proposed drug substance tion stability studies, industry should recognize that stability is
specifications. an important part of the much larger CTD Module 3, which is
Several options for the selection of batches to be included part of the complete registration dossier. The dossier will be
in the Common Technical Document (CTD) are illustrated deemed acceptable (or not) based on the quality of the dossier
in Fig. 5. As described previously, the traditional and most and other considerations such as the safety and efficacy of the
conservative approach is located on the lower left corner of product and whether the product represents an unmet medical
the figure. Although this approach will not likely be chal- need. In general, regulatory flexibility is more likely if the
lenged by the regulators, it can be the most costly and will product falls within a few special categories such as orphan
provide little opportunity to increase stability understanding. drugs or those that fill an unmet and/or critical medical need.
The option in the lower right corner represents a very Here, the applicant may be able to negotiate options such as
aggressive regulatory strategy and may be unacceptable if providing additional stability data during the review or flexi-
the drug substance batches selected for stability are not bility on what is considered a pilot-scale batch. Worst case
representative of the commercial process. Other cases are scenarios should also be reviewed as part of the risk analysis.
presented that moderate both risk and scientific value. It is The worst case may only require that the applicant initiate
evident that there are many options that should be consid- additional stability studies or provide data as a post-approval
ered when selecting the stability batches that will be used to commitment. The applicant should determine if the worst case
support registration. The risk (of not meeting regulatory scenarios are acceptable or require mitigation and/or negotia-
standards) associated with each of the Fig. 5 options can tion before filing.
be mitigated with increased product understanding. As de-
tailed earlier in this paper, accelerated studies can be used Post-approval Stability
during development to mitigate risk by identifying the
attributes that will have an effect on stability. Post-approval, there is a global GMP expectation that at
A stability strategy that may be appropriate for some drug least one batch of drug substance is placed on stability per
candidates is matrixing. Interestingly, ICH Q1D indicates year (assuming adequate production). But if the stability-
related quality attributes are understood and if post-approval
Table 3 Stability results for batch # 2 batches meet all acceptance criteria, a traditional post-
approval stability program will not be value-added. To meet
Test 70 °C/75 %RH 80 °C/40 %RH
Initial 14 Days 14 Days a regulatory commitment for annual batches or the GMP
expectation for annual stability batches, a short ASAP study
Appearance Off-white powder Off-white powder Off-white powder may be an appropriate alternative to a traditional annual
Assay 99.5 99.0 99.5 stability program and can be proposed to the regulators.
Impurities For post-approval changes to the synthetic process, site
(% area) change, or batch size, if the stability-related quality attrib-
(a) NMT 0.05 NMT 0.05 NMT 0.05 utes are within specification, a traditional stability program
(b) 0.09 0.09 0.09 is not scientifically needed.
Water 0.18 0.20 0.20 Recognizing the current regulatory environment, it would
content
PXRD Form A Form A Form A
be overly optimistic to plan for global acceptance of all the
post-approval strategies detailed above. However, leaner or
J Pharm Innov

Fig. 5 Several options for High


registration stability that vary Batches scientifically Batches scientifically
selected as the DS selected as the DS
scientific value and regulatory process is developed. process is developed
risk Batches are but may have minor
representative of the differences from the
commercial process. commercial process.

Scientific
Batches selected as the DS
Value process is developed. One
batch made at the
commercial site.

Clinical stability for each


new process and 3 Phase 1, 2 DS used for
commercial campaigns Registration Stability
Low for Registration stability

Low Regulatory Risk High

more nimble stability programs may be acceptable. With a resistance is directly proportional to impact and inversely pro-
stable and well understood drug substance, the number of portional to trust (resistance α impact/trust), industry should
stability pull points can be greatly reduced, especially dur- request a reasonable amount of flexibility (lowering the impact)
ing the first year of storage. A matrixed testing protocol also and work hard to clearly articulate their proposals with sound
may be acceptable where all tests are not monitored at each scientific data (and hence maximizing trust). While formal risk
time point. Traditionally, the testing done at release is large- assessments of manufacturing processes are well established
ly duplicated on stability even though many of these tests and leveraged in quality by design (QbD) submissions, risk
are not stability indicating (identification, water content), are assessments of stability are less well established and have been
not stability-related quality attributes, and/or are not shelf- underutilized by industry. A process for formal stability risk
life limiting attributes (assay). If the purpose of stability assessments has been presented [11] and these were leveraged
studies is to confirm the drug substance retest date, then by Pfizer for several NCE CTDs that were submitted for global
the only attributes that needs to be evaluated on stability are approval in 2011. While regulatory feedback varied across
those that may limit the retest period. Use of a quality by regions and even within regions (for different products), a trend
design approach to justify the removal of the assay from towards acceptance of leaner and “alternative” stability strate-
active pharmaceutical ingredients (API) stability testing pro- gies is clearly evident. Interestingly, these concepts are finding
tocol has recently been proposed [10]. acceptance not only in the major markets but in the Emerging
The lean strategies advocated above were recently Markets region as well. Day-80 feedback for a NCE under
employed in several marketing applications for several review in the EU is an example of current thinking: [12]
new chemical entities. Leveraging significant product un-
“The proposed post-approval testing frequencies, stor-
derstanding that was detailed in the marketing application,
age conditions, and testing methods are based on a QbD
regulatory relief was secured on post-approval stability
approach and take into consideration the findings of the
commitments in several markets:
above stability data. Testing done annually at 12, 24,
& Six months of API stability was provided in the initial and 36 months will include appearance and purity. The
filing and a 24-month retest period was approved. development formal registration stability studies con-
& Appearance and purity were the only tests on both post- firm that more frequent testing and storage at the accel-
approval and annual stability commitment protocols erated condition do not provide critical information
& The first stability pull point was at 12 months and annually needed to establish the retest period or monitor stability
thereafter. and quality of the drug substance. Assay was considered
& Only one storage condition (25 °C and 60 % RH) as non-stability indicating as the level of the total impu-
& Markets that approved this included: USA, European rities remained consistent for the pilot long-term stabil-
Union, Japan, Switzerland, and Canada. Approvals in ity batches while the assay values varied. In addition,
addition regions are pending. chiral purity, microbial quality, and water content were
also investigated and deemed non-stability indicating.”
Stability Risk Assessments and Regulatory Acceptance
of New Stability Strategies A Proposed Path Forward

New concepts that require regulatory approval will understand- A continued partnership and scientific-based collaboration
ably generate resistance from the authorities. Recognizing that between industry and the regulators is needed. Industry needs
J Pharm Innov

to identify stability-related quality attributes and the process 3. International Conference on Harmonization. International Confer-
ence on Harmonization Guideline Q1E: evaluation of stability
parameters that may have an effect on stability. With this
data. Rockville: FDA; 2003b.
scientific understanding, stability studies can be better engi- 4. International Conference on Harmonization. International Conference
neered and low-value programs can be avoided. Alternative on Harmonization Guideline Q7A: good manufacturing practice guid-
stability approaches should be leveraged and proposed to ance for active pharmaceutical ingredients. Rockville: FDA; 2001.
5. WHO. Stability testing of active pharmaceutical ingredients and
regulators as alternatives to traditional stability commitments.
finished pharmaceutical products. In: WHO Expert Committee on
Achieving global acceptance of the strategies for stability Specifications for Pharmaceutical Preparations. 43rd report. Gene-
studies detailed in this paper will take some time but will be va, World Health Organization, 2009, Annex 2 (WHO Technical
worth the effort because the patients will ultimately benefit. Report Series, No. 953).
6. International Conference on Harmonization. International Confer-
ence on Harmonization Guideline Q8(R2): pharmaceutical devel-
Acknowledgments The authors would like to thank Bob Baum, opment. Rockville: FDA; 2009.
Stephane Caron, Chi-wan Chen, Dave Damon, Allan Edwards, John 7. International Conference on Harmonization. International Confer-
Groskoph, Chuck Hoiberg, Vince McCurdy, Megan McMahon, Julian ence on Harmonization Guideline Q9: quality risk management.
Meekings, Greg Sluggett, Robert Timpano, Ken Waterman, Kevin Rockville: FDA; 2005.
Ryan, Roger Weaver, and PhRMA Stability Expert Team. 8. International Conference on Harmonization. International Confer-
ence on Harmonization Guideline Q10: pharmaceutical quality
system. Rockville: FDA; 2008.
9. Waterman KC, Carella AJ, Gumkowski MJ, Lukulay P, Macdonald
BC, Roy MC, Shamblin SL. Improved protocol and data analysis for
References accelerated shelf-life estimation of solid dosage forms. Pharm Res.
2007;24(4):780–90.
10. Skrdla PJ, Wang T, Antonucci V, Dowling T, Ge Z, Ellison D, Curran
1. International Conference on Harmonization. International Confer- J, Mohan G, Wyvratt J. Use of a quality-by-design approach to justify
ence on Harmonization Guideline Q1A(R2): stability testing of removal of the HPLC weight % assay from routine stability testing
new drug substances and drug products. Rockville: FDA; 2003a. protocols. J Pharm Biomed Anal. 2009;50:794–6.
2. International Conference on Harmonization. International Confer- 11. Weaver R. AAPS Annual Meeting presentation stability risk
ence on Harmonization Guideline Q1D: bracketing and matrixing assessments with a focus on physical stability issues. 2011.
designs for stability testing of new drug substances and drug 12. Co-Rapporteur Day 80 Critical Assessment Report for a New
products. Rockville: FDA; 2002. Chemical Entity. 2012.

View publication stats

You might also like