Вы находитесь на странице: 1из 10

J Pharm Innov (2013) 8:5665

DOI 10.1007/s12247-013-9147-0

CASE REPORT

Best Practices for Drug Substance Stress and Stability Studies


During Early-Stage Development Part IIConducting
Abbreviated Long-Term and Accelerated Stability Testing
on the First Clinical Drug Substance Batch to Confirm
and Adjust the Drug Substance Retest Period/Powder for Oral
Solution Shelf Life
Q. Chan Li & K. Cohen & T. Tougas & F. Qiu & J. Li &
J. McCaffrey & T. Purdue & Jinhua J. Song & F. Swanek &
S. Abelaira

Published online: 24 January 2013


# Springer Science+Business Media New York 2013

Abstract In pursuit of continuous process improvement,


the authors streamlined the drug substance (DS) stress and
stability testing process from preclinical to the first clinical
batch. High temperature/high humidity stress and ICH Q1B
confirmatory photostability testing on an early DS batch
provide stability data that help quickly assess the new DS
stability behavior, extrapolate the initial DS retest period
and the initial powder for oral solution (PFOS) shelf life,
and evaluate packaging requirements. Then, concurrent stability testing on the first clinical batch (or alternatively a
representative nonclinical batch) is conducted under longterm (LT) and accelerated (AC) storage conditions. The
LT/AC stability testing is used to verify the solid stress
testing results and to adjust the DS retest period and the
shelf life of (PFOS), if necessary. This paper describes how
to perform abbreviated LT/AC stability testing, compares
LT/AC stability results with the solid stress results, shows
how to adjust the DS retest period and PFOS shelf life if

Q. C. Li (*) : K. Cohen : T. Tougas : F. Qiu : J. Li :


J. McCaffrey : T. Purdue : J. J. Song : F. Swanek
Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT
06877, USA
e-mail: chan.li@boehringer-ingelheim.com
S. Abelaira
Boehringer Ingelheim, Argentina, Juana Azurduy 1534,
1429, Buenos Aires, Argentina

necessary, and shares our regulatory experiences with this


new approach. Our science- and risk-based DS stress and
stability process has offered a quick turnaround in obtaining
adequate stability information for new DS development,
eliminated redundancies, improved efficiency and consistency, achieved an optimal balance between risk and cost for
early drug development, and received regulatory acceptances.
Keywords Drug substance . Powder for oral solution
(PFOS) . Early-stage development . Solid stress . High
temperature/high humidity . Photostability . Retest period .
Shelf life
Abbreviations
AC
Accelerated storage condition (e.g., 40 C/75 %
RH)
HT/HH
High temperature and high humidity (e.g., 70 C/
75 % RH)
ICH
International Conference on Harmonisation
1 ICH
Light exposure for confirmatory studies, not
Q1B
less than 1.2 million lux hours of overall illumination of visible light and an integrated near
ultraviolet energy of not less than 200 Wh/m2
LT
Long-term storage condition, e.g., 25 C/60 %
RH
LT/AC
Long-term storage condition and accelerated
storage condition (25 C/60 % RH and 40 C/
75 % RH)

J Pharm Innov (2013) 8:5665

57

As stated in our part I paper [1], establishing the stability of


drug substance (DS) and drug product is a very important
drug development task, and is a regulatory expectation to
ensure product quality, which may affect product safety and
efficacy. For the stability testing of drug substance and drug
product to support registration, regulatory guidances [26]
harmonize and delineate expectations (e.g., protocol design,
the amount of data). For drug stability to support phase I and
phase II clinical trials (early development), regulatory guidances [7, 8] are usually worded generally. For phase I drug
substance stability, the US FDA guidance [7] states that A
brief description of the stability study and the test methods
used to monitor the stability of the drug substance should be
submitted. Preliminary tabular data based on representative
material may be submitted. Neither detailed stability data
nor the stability protocol should be submitted. The EMA
guidance [8] has similar requirements.

The generality of the regulatory guidances for the


early development stage is intended to allow flexibility
when changes and improvements are constantly made.
This flexibility leads pharmaceutical companies and regulatory agencies to make their own interpretations of the
guidances. Consequently, companies either conduct more
stability studies than necessary just to avoid regulatory
questions but at the expense of valuable resources, or
perform insufficient stability work, resulting in regulatory questions that must be answered and/or that may
demand more stability work and thus cause delays in
drug development. Also, regulatory interpretations and
expectations are not entirely consistent from agency to
agency and even from reviewer to reviewer. Hence, there
exist a pressing need and a great opportunity for pharmaceutical companies to share drug stability testing
practices, rationales, and regulatory experiences for the
early stages of development. Li [1], Waterman [911],
Colgan [12, 13], and Mazzo [14] have published scientific, practical, and efficient stability testing proposals
and practices.
As described in the part I paper [1], several years ago, we
improved our DS stability testing process to support phase I
clinical trials. The improved (recommended) process proceeds in two steps:

A suitably selected early lab scale batch of a new drug


substance in both open and closed containers is stressed
at high temperature/high humidity (HT/HH), preferably
70 C/75 % relative humidity (RH) for 3 weeks, and at
confirmatory photostability testing condition per International Conference on Harmonisation (ICH) Q1B [3]. This
design is called solid stress testing or simply solid stress.
The open container subjects the DS to a continuous
exposure to moisture (a worst case for bulk packaging)
and thus helps quickly determine whether moisture
affects DS stability. The closed container, which represents the packaging of powder for oral solution (PFOS),
protects the DS from exposure to moisture. This solid
stress is designed to get a quick readout of the new DS
stability behavior. Testing results for the open container
are used to extrapolate a retest period for the DS, whereas testing results for the closed containers are used to
extrapolate a shelf life for PFOS. All the solid stress
results are used to define packaging requirements.
The extrapolation principles and criteria have been described in part I [1]. Briefly, the extrapolation is based on the

classic Arrhenius relationship between temperature and the


reaction (degradation) rate, by assuming that the degradation reaction proceeds in a pseudo-zero order and that the
degradation rate constant doubles for every 10 C increase
in temperature, i.e., the 2 for 10 rule (k(T)=2T/10), which
roughly corresponds to an activation energy (Ea) of 12
14 Kcal/mol (5059 kJ/mol). This assumed Ea is very
conservative when compared with the Ea value of
83.144 kJ/mol (20 Kcal/mol) used for the calculation of
mean kinetic temperature for various climatic zones in the
US Pharmacopoeia [15]. The extrapolation criteria and
Arrhenius extrapolations from 70 and 60 to 25 C are
presented in Table 1 and Table 2, respectively.
The part I paper [1] has also described the solid stress
designs, DS batch selection, the stress testing results of
11 new drug substances, the extrapolations of initial DS
retest periods, and the PFOS shelf lives. As stated in [1],
the stress-extrapolated retest periods and shelf lives
should be verified with stability studies of the first clinical batch (or alternatively a representative nonclinical
batch) under long-term (LT) and accelerated (AC)

PFOS
RH

Powder for oral solution (DS in bottle, without


excipients)
Relative humidity

Introduction

58
Table 1 Solid stress extrapolation criteria

J Pharm Innov (2013) 8:5665

Tests

Stress extrapolation criteria

Description
Assay (compared with time 0a)
Individual degradation products
Total degradation products
Peak homogeneity
Chiral impurity (if applicable)

Report result and compare with initial result


Within 97.0102.0 % of the time 0 assay value
Each NMT 2 ICH 3A qualification thresholdb
NMT 2.0 %
The DS peak is spectrally homogeneous
NMT 2 ICH 3A qualification threshold

storage conditions. Part II (this paper) describes how to


perform abbreviated LT/AC stability testing, compares
LT/AC stability results with the solid stress results,
shows how to adjust the DS retest periods and PFOS
shelf lives if necessary, and shares our regulatory experiences with the improved process.

and/or extend a shelf life for the clinical PFOS whose


primary packaging is more protective (typically glass and
tight screw cap). A DS stability design example is given
in Table 3, which uses the ICH Q1A construct [2] but
with fewer time points at LT conditions, i.e., the 9- and
18-month time points are not included (thus called abbreviated LT/AC stability testing). The Table 3 design is
efficient and justified because stability data from 3-, 6-,
12-, and 24-month time points can adequately verify the
DS stability behavior from the solid stress, and 9- and
18-month time points do not add much new information
based on our many years of experience and that statistical analyses requiring a sufficient number of data points
are hardly ever done or needed in early development. On
the other hand, the 9- and 18-month time points can be
tested if retest periods/shelf lives of 21 and 30 months,
respectively, are desired. Typical tests (A) are: description, assay and impurities by HPLC, water content, and
X-ray powder diffraction. Chiral impurity by HPLC is
generally not performed unless the solid stress testing
shows chiral conversion. Although literature [16] discourages the assay test for routine stability testing, our
HPLC methods typically combine assay and impurity
determination in the same procedure, which does not
demand much extra time, but can yield useful information when the DS is not stable.

NMT not more than


a

Initial (time zero) assay result is


determined by area percent (in the
absence of a reference standard)

2 ICH 3A qualification threshold = two times the qualification


threshold by ICH Q3A [17]

Abbreviated LT/AC Stability Testing of the First


Clinical Batch
DS Batch Selection
Ideally, the first clinical batch of the DS is selected for the
LT/AC stability studies, which are usually conducted concurrently with the Phase 1a clinical trial. Alternatively, a
nonclinical batch can be selected for the LT/AC stability
study, provided that the quality and particularly stabilityrelated quality attributes of the batch, packaging configurations, and analytical methods are all representative. In the
alternate case, the stability testing is not required for the first
clinical batch. However, it is recommended that sufficient
samples from the clinical batch be stored at the LT/AC
storage conditions in the event testing is requested.
Bulk DS Stability Design
Drug substance material is packaged in polyethylene
bags closed with twist ties and placed in fiber shipping
containers. This packaging simulates the worst case protection among various packaging configurations that can
be used for bulk drug substance. Because this packaging
is less protective, the stability results can be used to set
Table 2 Arrhenius extrapolations of retest period and shelf
life in month at 25 C

PFOS Stability Design


One representative fill weight should be placed on stability.
This fill weight typically contains the least amount of drug
substance to allow full testing. Samples from clinical supplies
are preferred for this study. A PFOS LT/AC stability design

Temperature (C)

Storage duration (weeks)

Extrapolated retest period/shelf life in month at 25 C

70

1
2
3
4
5
6

5
11
15.6 reduced to 15
10
13
15.6 reduced to 15

60

J Pharm Innov (2013) 8:5665

59

Table 3 Bulk drug substance LT/AC stability design example


Storage conditions

Time 0

3 months

6 months

12 months

24 months

25 C/60 % RHa
30 C/75 % RHb
40 C/75 % RHa

Typically release testing results

If the stress data suggest that the bulk DS should be refrigerated, then the long-term storage condition 25 C/60 % RH is changed to 53 C, and
the accelerated storage condition 40 C/75 % RH is changed to 252 C/605 % RH

Samples may be stored but tested only if significant degradation is observed at 40 C/75 % RH

example is shown in Table 4. Typical tests (B) are description


and assessment of packaging material. The assay and impurities by HPLC test is performed only if needed, for example, if
there is a stability question relating to the clinical PFOS. The
PFOS stability design has fewer tests and is shorter in duration
than the bulk DS stability design in Table 3. This is because, as
mentioned above, the bulk DS stability results are used to set
and extend the clinical PFOS shelf life.
Data Evaluation and Retest Period/Shelf Life Assignment
The results from the LT/AC stability study are evaluated to
confirm and/or adjust the DS retest period and PFOS shelf
life. While formal statistical evaluations are typically not
performed at this stage of development, data trending should
be examined to evaluate changes that may occur during a
stability study and to assess whether stability results are
approaching or have exceeded the acceptance criteria for
specified impurities/degradation products. Two times (2)
the ICH Q3A [17] qualification threshold is used as the
criterion for evaluating each unspecified impurity/degradation product in this early stage of drug development. It is
understood that other pharmaceutical companies, depending
on their risk tolerance, may choose more lenient criteria.
Table 5 shows our recommendations for setting DS retest
period and PFOS shelf life on the basis of both the solid
stress testing results and the LT/AC stability testing results,
when both data sets show no degradation trends towards an
unacceptable level of degradant. It should be pointed out
that longer or shorter shelf lives may be proposed,
Table 4 PFOS stability design example
Storage conditions

Time 0

3 months

6 months

End
of study

25 C/60 % RHa
40 C/75 % RHa

B
B

Bb

If the stress data suggest that the PFOS should be refrigerated, then
the long-term storage condition 25 C/60 % RH is changed to 5 C3
C, and the accelerated storage condition 40 C/75 % RH is changed to
252 C/605 % RH

Performed at the end of study to cover the duration of clinical trials

depending on the rigor of scientific justifications, the risk


tolerance of the pharmaceutical company, and the regulatory
expectations of authorities that approve the clinical trials
[14]. It is also noted that Beaman [18] proposed the duration
of primary stability testing and a decision tree to assign
product shelf lives for registration. Also, if a significant
degradation trend is observed in samples stored at accelerated conditions, but the long-term data show no degradation
trend, the DS retest period/PFOS shelf life are set shorter
than those proposed in Table 5, and this has to be handled on
a case-by-case basis. In such a case, testing DS stored at the
intermediate condition 30 C/75 % RH would provide useful data.

Comparisons Between Solid Stress Testing Results


and LT/AC Stability Results
The improved approach has been implemented on 11 new
drug substances, all of which underwent solid stress testing
and 10 of which underwent the abbreviated LT/AC stability
testing. The chemical stability results are summarized in
Table 6 for DS 16, Table 7 for DS 79, and Table 8 for
DS 11. DS 10 was not subjected to LT/AC stability testing
because the first clinical batch was not manufactured. For all
the drug substances, the physical stress/stability data (typically for XRPD and DSC and water content), which were
used for understanding DS physical behavior and for formulation development purposes, were gathered and analyzed but are not presented in this paper, as all 10
compounds exhibited no physical stability-related changes.
As shown in Table 6, DS 1 showed a single, minor
degradation product (Imp 3) when stressed at 70 C/75 %
RH, but no degradation under the ICH Q1B confirmatory
photostability testing. Since the maximum daily dose for
phase I was below 25 mg, the solid stress extrapolation
criterion for degradation was set to not more than (NMT)
0.30 %, which is two times the ICH Q3A qualification
threshold (20.15 %=0.30 %). Because the degradation
product (Imp 3) was NMT 0.14 % in both open and closed
containers at 3 weeks at 70 C/75 % RH, both the initial DS
retest period and the initial PFOS shelf life were

60
Table 5 DS retest period and
PFOS shelf life extrapolated
from long-term and accelerated
stability data, along with solid
stress testing data
a

Both data sets show no degradation trends towards an unacceptable level of degradant

In case these time points are


analyzed

J Pharm Innov (2013) 8:5665

Available stability dataa

Proposed retest period and shelf life

Time 0
3-month accelerated and long-term data
6-month accelerated and long-term data
9-month long-term datab
12-month long-term data
18-month long-term datab
24-month long-term data

15
15
18
21
24
30
36

extrapolated to 15 months per Table 2. The stress findings


and the conservatively extrapolated 15 months shelf life
were confirmed by the LT/AC stability results. After
12 months at 25 C/60 % RH and 5 months at 40 C/75 %
RH, the degradant Imp 3 did not exceed 0.08 and 0.14 %,
respectively (Table 6). According to Table 5, the DS retest
period was extended to 24 months. The 15-month PFOS
shelf life could have been extended to 24 months. However,
15 months was sufficient to cover packaging, labeling,
shipping, storage, and the clinical trial duration. In our
experience, a 15-month shelf life for a PFOS can cover
phase Ia clinical trials in most cases.
As shown in Tables 6 and 7, drug substances 2, 3, and
9 showed little or no degradation in open or closed
containers after 3 weeks stress at 70 C/75 % RH but
showed degradation under the Q1B confirmatory photostability testing. Thus, according to Table 2, both the
initial DS retest period and the PFOS shelf life were
set to 15 months with the storage requirement that each
DS be protected from light. The results of the 3-week
stress at 70 C/75 % RH and the 15-month shelf life
were confirmed by the LT/AC stability results. DS 2 and
DS 3 in Table 6 did not show degradation after storage
of 12 months at 25 C/60 % RH and 6 months at 40
C/75 % RH. According to Table 5, the retest period for
DS 2 and DS 3 was extended to 24 months. Since DS 9
showed no degradation after 6 months at 25 C/60 % RH
and 6 months at 40 C/75 % RH (Table 7), the retest
period for DS 9 was extended to 18 months (Table 5).
For these three drug substances, there was no business
need to extend the initial PFOS shelf life beyond
15 months, although longer shelf lives were justified
(Table 5) based on the LT/AC stability results.
As shown in Tables 6 and 7, drug substances 48 showed
no degradation after 3 weeks stress at 70 C/75 % RH in
open or closed containers and by the confirmatory photostability testing. In our experience, this stable behavior is
typical of most of the drug substances. For each of these
drug substances, the initial DS retest period and the initial
PFOS shelf life were set to 15 months according to Table 2.
The results of the 3-week stress at 70 C/75 % RH and the
15-month shelf life were confirmed by the LT/AC stability

months
months (support stress extrapolation)
months
months
months
months
months

results, since no degradation was observed at 25 C/60 %


RH after 6 or 12 months and at 40 C/75 % RH after
6 months (Tables 6 and 7). For DS 46, the DS retest period
was extended to 24 months (Table 5) on the basis of the
LT/AC stability results obtained after 6 months at 40
C/75 % RH and after 12 months at 25 C/60 % RH (Table 6).
For DS 78, the DS retest period was extended to 18 months
based on the LT/AC stability results obtained after 6 months
at 40 C/75 % RH and at 25 C/60 % RH (Table 7).
DS 11 is an interest example as shown in Table 8 and
described in detail in part I [1]. The first lab batch was
subjected to 70 C/75 % RH and the Q1B confirmatory
photostability testing, and one significant, predominant
degradation product Imp 2 was observed. Imp 2 was
0.12 % at time 0. Imp 2 increased to 0.43, 0.66, and
0.80 % in the open container, and to 0.23, 0.29, and
0.37 % in the closed container, at 70 C/75 % RH after
1 week (8 days), 2 weeks, and 3 weeks, respectively. It
should be noted that the open container showed greater
degradation than the closed container, pointing to water
as a stability-related quality attribute. Imp 2 increased to
1.8 % under the confirmatory photostability testing. The
extrapolated DS retest period was only 6 months based
on Table 2. Efforts were immediately made to identify
Imp 2 and the degradation pathway. LC-MS work identified Imp 2 as a rotamer (a conformational isomer produced by restricted rotation). It was then hypothesized
that high contents of volatiles (residual solvent and water) could promote the formation of the rotamer (Imp 2),
particularly when the volatiles possess high kinetic energy at high temperature (e.g., 70 C). Note that the first
lab batch contained the relatively high levels of isopropanol (1.6 %) and water (0.26 %). Thus, a second lab
batch was produced intentionally with low levels of
water (0.08 %) and residual solvent ethanol (0.10 %),
which replaced isopropanol for recrystallization. When
this second lab batch was subjected to 70 C/75 % RH
stress for 2 and 3 weeks, Imp 2 virtually did not change,
i.e., from 0.09 % at time 0 to 0.11 % at 3 weeks, in both
open and closed containers (Table 8). From the second
lab batch stress data, the initial DS retest period and the
initial PFOS shelf life were extrapolated to 15 months,

J Pharm Innov (2013) 8:5665

61

Table 6 Comparisons between solid stress and LT/AC stability results for DS 16
Test

Lab batch solid stress

Time 0

DS 1
Description
Assay (%)
Imp 1 (%)
Imp 2 (%)
Imp 3 (%)
DS 2
Description
Assay (%)
Imp 1 (%)
Imp 2 (%)
Imp 3 (%)
Imp 4 (%)
DS 3
Description
Assay (%)
Imp 1 (%)
Imp 2 (%)
DS 4
Description
Assay (%)
Imp 1 (%)
Imp 2 (%)
Imp 3 (%)
Chiral Imp (%)
DS 5
Description
Assay (%)
Imp 1 (%)
Imp 2 (%)
DS 6
Description
Assay (%)
Imp 1 (%)

1st clinical batch (or representative non-GMP batch) (DS in polyethylene


bag placed in fiber tube)

70 C/75 % RH

Photo

Time 0

25 C/60 % RH (LT)

40 C/75 % RH (AC)

6 months

12 months

3 months

6 months

3 weeks, open

3 weeks,
closed

1 ICH
closed

White
100.0
0.07
0.07
<0.05

White
100.0
0.08
0.08
0.14

White
100.0
0.07
0.07
0.14

White
99.0
0.08
0.07
<0.05

White
100.0
0.07
0.07
<0.05

Whitea
100.3a
0.08a
0.08a
0.07a

White
99.0
0.08
0.07
0.08

White
99.8
0.08
0.07
<0.05

Whiteb
100.2b
0.08b
0.07b
0.14b

Off-white
100.0
<0.05
<0.05

Off-white
99.2
0.10
0.11

Off-white
99.9
<0.05
0.10

Off-white
99.3
<0.05
<0.05

Off-white
100.1
<0.05
0.08

Off-white
99.2
0.10
0.08

Off-white
99.3
<0.05
0.08

Off-white
100.3
<0.05
0.08

Off-white
99.9
<0.05
0.08

<0.05
0.55

<0.05
0.59

<0.05
0.56

0.66
0.56

<0.05
0.52

0.10
0.51

<0.05
0.51

<0.05
0.52

<0.05
0.51

Off-white
99.9
0.09
<0.05

Off-white
99.2
0.09
<0.05

Off-white
99.2
0.12
<0.05

Off-whitec
98.9c
0.09c
1.7c

White
101.3
<0.05
<0.05

White
99.4
<0.05
<0.05

White
98.9
<0.05
<0.05

White
99.2
<0.05
<0.05

White
100.4
<0.05
<0.05

White
99.8
<0.05
<0.05
<0.05
0.21

White
98.9
<0.05
<0.05
<0.05
0.22

White
100.0
<0.05
<0.05
<0.05
0.21

White
100.6
<0.05
<0.05
<0.05
0.21

White
99.4
0.06
0.10
0.08
0.17

White
99.8
0.06
0.09
0.08
0.18

White
100.3
0.05
0.09
0.09
0.18

White
99.6
0.05
0.09
0.08
0.18

White
99.6
0.06
0.09
0.09
0.16

Off-white
99.9
0.05
<0.05

Off-white
99.8
0.06
<0.05

Off-white
99.8
0.06
<0.05

Off-white
99.7
0.05
<0.05

White
99.9
0.07
<0.05

White
100.4
0.08
<0.05

White
99.9
0.11
<0.05

White
100.1
0.06
<0.05

White
100.4
0.08
<0.05

Off-white
100.0
0.09

Off-white
101.0
0.09

Off-white
100.9
0.09

Off-white
NA
0.09

Off-white
99.3
0.12

Off-white
99.3
0.16

Off-white
99.9
0.12

Off-white
98.8
0.16

Off-white
99.4
0.17

General designations Imp 1 and Imp 2 are specific to each DS. All impurity levels <0.05 % including not detected are simply reported as <0.05 %.
Photo 1 ICH closed is the confirmatory photostability of ICH Q1B
Imp impurity, White white powder, Off-white off-white powder
a

Five months at 25 C/60 % RH; Normal practice is 6 months

Five months at 40 C/75 % RH; Normal practice is 6 months

2 ICH photo stress in this case, although normal practice is 1 ICH photo stress

with protection from light, according to Table 2. The


stress results of the second lab batch were confirmed
by the LT/AC stability results of the first clinical batch:

no degradation was observed after 12 months at 25


C/60 % RH and after 6 months at 40 C/75 % RH
(Table 8). The DS retest period was then extended to

62

J Pharm Innov (2013) 8:5665

Table 7 Comparisons between solid stress and LT/AC stability results for DS 79
Test

Lab batch solid stress


Time 0

DS 7
Description
Assay (%)
Imp 1 (%)
Imp 2 (%)
Imp 3 (%)
DS 8
Description
Assay (%)
Imp 1 (%)
Imp 2 (%)
DS 9
Description
Assay (%)
Imp 1 (%)
Imp 2 (%)
Imp 3 (%)
Imp 4 (%)
Imp 5 (%)
Imp 6 (%)
Imp 7 (%)
Imp 8 (%)
Imp 9 (%)
Imp 10 (%)
Imp 11 (%)

1st clinical batch (DS in polyethylene bag placed in fiber tube)

70 C/75 % RH

Light

3 weeks,
open

3 weeks,
closed

1 ICH
closed

White
100.1
0.06
0.10
0.20

White
99.0
0.07
0.10
0.17

White
99.7
0.06
0.10
0.17

White
101.1
0.06
0.10
0.20

Light yellow
100.0
<0.05
<0.05

Light yellow
98.1
<0.05
<0.05

Light yellow
98.9
<0.05
<0.05

Off-white
100.0
0.06
0.08
<0.05
0.07
<0.05
<0.05
0.17
0.11
0.13
0.52
0.13

Off-white
100.4
0.21
0.10
<0.05
0.06
0.06
0.08
0.19
0.10
0.13
0.50
0.12

Off-white
102.2
0.12
0.10
<0.05
0.07
0.07
<0.05
0.19
0.11
0.13
0.50
0.12

Time 0

25 C/60 % RH (LT)

40 C/75 % RH (AC)

3 months

6 months

3 months

6 months

White
100.4
<0.05
<0.05
<0.05

White
100.2
<0.05
<0.05
<0.05

White
99.8
<0.05
<0.05
<0.05

White
99.9
<0.05
<0.05
<0.05

White
99.8
<0.05
<0.05
<0.05

Light yellow
99.5
<0.05
0.08

Light yellow
99.7
<0.05
<0.05

Light yellow
100.3
<0.05
<0.05

Light yellow
99.4
<0.05
<0.05

Light yellow
100.2
<0.05
<0.05

Light yellow
99.3
<0.05
<0.05

Off-white
97.0
<0.05
0.10
0.45
0.06
0.54
0.54
0.20
0.11
0.10
0.46
0.12

White
100.2
<0.05
<0.05
<0.05
0.06
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05

White
100.0
<0.05
<0.05
<0.05
0.06
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05

White
100.1
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05

White
99.7
<0.05
<0.05
<0.05
0.06
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05

White
100.2
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05
<0.05

General designations Imp 1 and Imp 2 are specific to each DS. All impurity levels <0.05 % including not detected are simply reported as <0.05 %.
Photo 1 ICH closed is the confirmatory photostability of ICH Q1B
Imp impurity, White white powder, Off-white off-white powder, Light yellow light yellow powder

24 months per Table 5. Note that the clinical batch


contained low levels of ethanol (0.18 %) and water
(0.08 %) (Table 8), consistent with the stability-related
quality attributes identified by the stress stability data
from lab batch 1 and lab batch 2. Thus, it is obvious
that the HT/HH and photo stress can quickly reveal the
instability behavior and stability-related quality attributes
of the new DS. After the identification of degradation
products and degradation mechanisms, stability-related
quality attributes were confirmed and controlled, the
chemical manufacturing process improved, and the packaging properly selected.
An approach alternative to our simple HT/HH stress is the
Accelerated Stability Assessment Program (ASAP) [911].
ASAP uses several combinations of temperature and

humidity conditions, requires multiple stress chambers and


the amounts of degradation be kept consistent (isoconversion,
tied to specification limits) at different conditions, involves
statistical analysis, and uses a humidity-corrected Arrhenius
equation to extrapolate a DS retest period. If isoconversion is
not achieved, more iterations of the experiments are conducted. However, in early development, specification limits
are either not set or changeable, therefore isoconversion is
hard to achieve. For the requirements and amount of work,
ASAP is best used for drug substances that have shown
chemical stability challenges. Since most drug substances in
our experience are stable or moderately stable, our one
HT/HH stress condition that requires only one stability chamber (or two conditions on occasion) is reliable, efficient, and
effective for assessing all new DS stability behaviors.

J Pharm Innov (2013) 8:5665

63

Table 8 Comparisons between solid stress and LT/AC stability results for DS 11
1st lab batch solid stress
Conditions

Open container

Time 0
70 C/75 % RH 8 daysa
2 weeks
3 weeks
1 ICH photo

Description

% Assay % Imp 1 % Imp 2 % IPA

% Water Description

% Assay % Imp 1 % Imp 2

Brown
Brown

100.0
100.2

100.0
100.5

Brown
100.4
Brown
99.0
Dark brown 98.6

2nd lab batch solid stress


Conditions

Open container

Time 0
70 C/75 % RH 1 week
2 weeks
3 weeks

Description
Brown
NP
Brown
Brown

1st clinical batch LT/AC (DS


Conditions
Time 0
25 C/60 % RH 6 months
(LT)
12 months
40 C/75 % RH 3 months
(AC)
6 months

Closed container

% Assay
100.5
NP
99.6
100.1

<0.05
<0.05

0.12
0.43

1.6
NP

0.26
NP

Brown
Brown

0.06
0.08
0.08

0.66
0.80
1.8

NP
NP
NP

NP
NP
NP

Brown
99.8
Brown
99.8
Dark brown 98.6

% Water
0.08
NP
NP
NP

Description
Brown
NP
Brown
Brown

% Assay
100.5
NP
100.4
99.9

% Imp 2
0.14
0.14
0.13
0.14
0.14

% Ethanol
0.18
NP
NP
NP
NP

<0.05
<0.05

0.12
0.23

0.06
0.08
0.08

0.29
0.37
2.1

Closed container
% Imp 1 % Imp 2 % Ethanol
<0.05
0.09
0.10
NP
NP
NP
<0.05
0.10
NP
<0.05
0.11
NP

in polyethylene bag placed in fiber tube)


Description
% Assay
Brown
99.1
Brown
100.8
Brown
99.3
Brown
100.5
Brown
101.8

% Imp 1
<0.05
<0.05
<0.05
<0.05
<0.05

% Imp 1 % Imp 2
<0.05
0.09
NP
NP
<0.05
0.10
<0.05
0.11

% Water
0.08
0.15
0.12
0.17
0.16

General designations Imp 1 and Imp 2 are specific to each DS. All impurity levels <0.05 % including not detected are simply reported as <0.05 %.
Photo 1 ICH closed is the confirmatory photostability of ICH Q1B
Imp Impurity, Brown brown powder, IPA isopropanol, NP not performed
a

Eight days in this case, although 1 week (7 days) is the normal practice

From the studies of these 10 drug substances, it is evident


that the solid stress testing and extrapolations are reliable,
useful to quickly evaluate DS stability behaviors, whether
the substances are stable or not (DS 1 and DS 11, in particular, being not stable), and the abbreviated LT/AC stability
study is confirmatory and concurrent with the clinical trial,
satisfying both scientific rigor and regulatory expectations.

Regulatory Experiences
Of the 10 drug substances cited in Tables 6, 7, and 8, six
have been filed with a regulatory agency for phase Ia clinical trials conducted in Europe. Table 9 summarizes relevant
stability data and shelf lives presented in these six IMPDs
(Investigational Medicinal Product Dossier).

Table 9 DS stability information and PFOS shelf lives presented in six IMPDs
DS #

5, 7, 8, 9

Solid stress results for 3 weeks No degradation


No degradation except at ICH Little or no degradation except
at 70 C/75 % RH (open and
Q1B photo
that DS 9 showed degradation
closed glass vials) and at ICH
at ICH Q1B photo
Q1B photo
LT/AC stability test results
Data not available at filing. Committed to
No degradation after 6 months No degradation after 3 months
perform concurrent LT/AC stability testing at LT/AC
at LT/AC
PFOS shelf life proposed
12 months
18 months (DS and its PFOS 15 months (DS 9 and its PFOS
must be protected from light) must be protected from light)

64

In the DS 6 IMPD, we elected to be conservative by


proposing a PFOS shelf life of 12 months instead of 15 months
because it was the first time we used the solid stress testing
results to derive a PFOS shelf life. Initially, the regulatory
agency questioned the basis for this proposal. After we
responded by presenting the Arrhenius equation, explaining
thoroughly our extrapolations and rationales for the solid
stress testing, and reaffirming our commitment to the LT/AC
stability testing concurrent with the phase 1a clinical trial, the
regulatory agency accepted our proposed shelf life of
12 months.
In the DS 3 IMPD, we submitted solid stress data, presented the Arrhenius equation, explained our extrapolations
and rationales for the solid stress testing, presented up to
6 months of LT/AC stability results for the clinical batch,
and committed to continue the abbreviated LT/AC stability
testing on the clinical batch. We proposed a PFOS shelf life
of 18 months and received regulatory acceptance of this
proposal without question.
In the DS 8 IMPD, we submitted solid stress data, presented the Arrhenius equation, provided a summary of our
extrapolations and rationales for the solid state stress testing,
and committed to perform the LT/AC stability testing on the
clinical batch. There were no LT/AC stability results of the
clinical batch available at the time of initial filing. We
proposed a PFOS shelf life of 15 months. Initially, the
regulatory agency questioned the basis for proposing a shelf
life of 15 months. We responded by (1) explaining more
thoroughly the Arrhenius equation, extrapolations, and
rationales, (2) providing references for the solid state stress
testing, (3) providing 3 months of LT/AC stability results for
the clinical batch, and (4) reaffirming our commitment to
continue to perform the LT/AC stability testing concurrent
with the phase 1a clinical trial. The regulatory agency accepted our proposed shelf life of 15 months.
In the DS 5, DS 7, and DS 9 IMPDs, we submitted solid
stress data, presented the Arrhenius equation, explained our
extrapolations and rationales for the solid stress testing,
presented 3 months LT/AC stability results for the clinical
batches, and committed to continue the LT/AC stability
testing on the clinical batch. We proposed a PFOS shelf life
of 15 months and the regulatory agency accepted our proposals without question.
For future initial IMPD filings, we recommend following the paradigm of the DS 5, 7, 9, or 3. Our experiences
indicate that the solid stress results and 3-month (or 6month) LT/AC stability results have received regulatory
acceptance of a 15-month (or 18-month) shelf life without
question. Again, longer or shorter shelf lives may be
proposed, depending on the rigor of scientific justifications, the risk tolerance of the pharmaceutical company,
and the expectations of authorities that approve the clinical
trials [14].

J Pharm Innov (2013) 8:5665

Conclusions
In pursuit of continuous process improvement, we improved
the drug substance stress and stability testing process from
preclinical to the first clinical batch. A high temperature/high humidity stress (typically 3 weeks in open and closed
containers at 70 C/75 % RH) and ICH Q1B confirmatory
photostability testing on an early DS batch provide stability
data that help quickly assess the new DS stability behavior,
extrapolate the initial DS retest period and the initial PFOS
shelf life, and evaluate packaging requirements. Then, concurrent with the phase 1a clinical trial, an abbreviated longterm and accelerated stability testing on the first clinical
batch (or representative nonclinical batch) provides stability
data that are used to verify the solid stress testing conclusions and to adjust the DS retest period and the PFOS shelf
life, if necessary. This improved process was based on
established scientific principles, experimentally tested, and
implemented on 10 new drug substances. Since implementation, the improved DS stability process has offered a quick
turnaround in obtaining adequate stability information for
new DS development, eliminated redundancies, improved
efficiency and consistency, achieved an optimal balance
between risk and cost for early drug development when
attrition rate is very high, and received regulatory acceptances. Parts III (currently in preparation) of this series will
discuss how to make risk-based and data-driven evaluation
of the need for further stability studies on DS batches
subsequent to the first clinical batch.
Acknowledgments Special thanks go to Mr. Gordon Hansen, Drs.
Chris Senanayake and Keith Horspool, and Ms. Patricia Watson for
management support and to Ms. Cornelia Field, Drs. Christian Kulinna
and Reggie Saraceno, and many BI colleagues for constructive discussions and contributions.

References
1. Li QC, Qiu F, Cohen K, Tougas T, Li J, McCaffrey J, Purdue
T, Song J, Swanek F, Abelaira S. Recommended best practices
for drug substance stress and stability studies to support drug
development from preclinical to phase II clinical trials, part I
conducting drug substance solid stress to support phase Ia
clinical trials. J Pharm Innov (JPI). 2012;7(Issue 3):21424.
2. International Conference on Harmonisation Guidance (ICH) Q1A
(R2): Stability testing of new drug substances and products,
February 2003.
3. International Conference on Harmonisation Guidance (ICH) Q1B:
Stability testing: photostability testing of new drug substances and
products, November 1996.
4. International Conference on Harmonisation Guidance (ICH) Q1D:
Bracketing and matrixing designs for stability testing of new drug
substances and products, February 2002.
5. International Conference on Harmonisation Guidance (ICH) Q1E:
Stability data, February 2003.

J Pharm Innov (2013) 8:5665


6. WHO Expert Committee on Specifications for Pharmaceutical
Preparations. Forty-third report, WHO technical report series,
vol. No. 953. Geneva: World Health Organization; 2009.
Annex 2.
7. US FDA Guidance for Industry Content and Format of
Investigational New Drug Applications (INDs) for Phase 1 Studies
of Drugs, Including Well-Characterized, Therapeutic
Biotechnology-derived Products, November 1995.
8. European Medicines Agency (EMA): Guideline on the
requirements to the chemical and pharmaceutical quality documentation concerning investigational medicinal products in
clinical trialsCHMP/QWP/185401/2004 Final, 31 March
2006.
9. Waterman KC, Carella AJ, Gumkowski MJ, Lukulay P,
Macdonald BC, Roy MC, Shamblin SL. Improved protocol and
data analysis for accelerated shelf-life estimation of solid dosage
forms. Pharm Res. 2007;24(4):78090.
10. Waterman KC, Colgan ST, Waterman KC. A science-based approach to setting expiry dating for solid drug products. Regulatory
Rapporteur. 2008;5(7):914.
11. Waterman KC. Accelerated Stability Assessment Program
(ASAP): using science to set shelf life, pharmaceutical outsourcing. March/April 2012.

65
12. Colgan ST, Whipple RD, Watson TJ, Nosal R, Beaman JV,
DeAntonis DM. The Application of Quality by Design's Science
and Risk Based Concepts to API Stability Strategies. AAPS
Stability Workshop, September 2009.
13. Colgan ST, Watson TJ, Whipple RD, Nosal R, Beaman JV, De
Antonis DM. The application of science- and risk-based concepts
to drug substance stability strategies. J. Pharm. Innov. 2012;7(3
4):20513. doi:10.1007/s12247-012-9135-9.
14. Acken B, Alasandro M, Colgan S, Curry P, Diana F, Li QC, Li ZJ,
Mazzeo T, Rignall A, Tan ZJ, Timpano R. Early development GMPs
for stabilityan industry perspective (part IV). Pharmaceutical
Technology, 2012;8694.
15. US Pharmacopoeia (USP) 34-National Formulary (NF) 29 S1,
general chapter <1150> pharmaceutical stability.
16. Skrdla PJ, Wang T, Antonucci V, Dowling T, Ge Z, Ellison D,
Curran J, Mohan G, Wyvratt J. Use of a quality-by-design approach to justify removal of the HPLC weight % assay from
routine API stability testing protocols. J PharmBiomed Anal.
2012;50:7946.
17. International Conference on Harmonisation Guidance (ICH) Q3A
(R2): Impurities in New Drug Substances, October 2006.
18. Beaman JV. Stability testing: doing everything or doing the right
thing? Pharm Rev. 2010;736.

Вам также может понравиться