Sei sulla pagina 1di 9

European Journal of Pharmaceutics and Biopharmaceutics 125 (2018) 76–84

Contents lists available at ScienceDirect

European Journal of Pharmaceutics and Biopharmaceutics


journal homepage: www.elsevier.com/locate/ejpb

Research paper

Accurate prediction of vaccine stability under real storage conditions and T


during temperature excursions
Didier Clénet
Formulation & Stability Platform, Bioprocess R&D Department, Sanofi Pasteur, 1541 Avenue Marcel Mérieux, 69280 Marcy L'Étoile, France

A R T I C L E I N F O A B S T R A C T

Keywords: Due to their thermosensitivity, most vaccines must be kept refrigerated from production to use. To successfully
Vaccine stability carry out global immunization programs, ensuring the stability of vaccines is crucial. In this context, two im-
Shelf life predictions portant issues are critical, namely: (i) predicting vaccine stability and (ii) preventing product damage due to
Stability modeling excessive temperature excursions outside of the recommended storage conditions (cold chain break). We applied
Cold chain break
a combination of advanced kinetics and statistical analyses on vaccine forced degradation data to accurately
Storage temperature
describe the loss of antigenicity for a multivalent freeze-dried inactivated virus vaccine containing three var-
iants. The screening of large amounts of kinetic models combined with a statistical model selection approach
resulted in the identification of two-step kinetic models. Predictions based on kinetic analysis and experimental
stability data were in agreement, with approximately five percentage points difference from real values for long-
term stability storage conditions, after excursions of temperature and during experimental shipments of freeze-
dried products. Results showed that modeling a few months of forced degradation can be used to predict various
time and temperature profiles endured by vaccines, i.e. long-term stability, short time excursions outside the
labeled storage conditions or shipments at ambient temperature, with high accuracy. Pharmaceutical applica-
tions of the presented kinetics-based approach are discussed.

1. Introduction the time period during which a drug product remains capable of ac-
ceptable performance. Several shelf life estimation methods exist, those
The thermal sensitivity of formulated biologics, including vaccines, described in International Conference on Harmonisation (ICH) guide-
is a critical factor impacting product quality and potency, hence in- lines [8] are generally based on linear or nonlinear regression and
fluencing their worldwide distribution [1,2]. The thermal stability of statistical modeling through poolability tests. Focusing on the fact that
vaccines is of concern to the vaccine industry, health authorities, gov- an agreement does not currently exist, an investigation of current sta-
ernment institutions, and philanthropic organizations attempting to tistical methods for estimating shelf life, based on stability data, was
increase the distribution of vaccines to people living in countries with conducted by a Product Quality Research Institute (PQRI) working
poor infrastructure, unreliable transportation and substandard storage group [9]. Additionally, to improve ICH procedure, the application of
facilities for the preservation of vaccines requiring refrigeration [3]. quantile regression or mixed model tolerance interval methods and
World Health Organization (WHO) sets guidelines for the stability batch effects, was proposed [10,11].
evaluation of vaccines [4] and recommends conducting regular and Kinetics is typically used to estimate vaccine degradation rates
accelerated stability studies. These guidelines aim to provide a frame- through accelerated stability programs which expose products to tem-
work for shelf life and storage conditions, monitor vaccine stability in peratures greater than those recommended for vaccine storage (typi-
the post licensure period and support manufacturing changes by de- cally 5 °C, 25 °C, 37 °C). During kinetic analysis of the data, very often
monstrating consistency of vaccine batches. Such guidelines can be the simplified kinetics models, such as zero- or first order mechanisms,
used for the selection of optimal stabilizing conditions, shelf life esti- are considered. Such models fail to correctly describe the complicated
mation, temperature excursion modeling, and investigation of changes course of decomposition of biological materials, which frequently show
to manufacturing process that may potentially alter vaccine stability complex and multi-step degradation behavior [12–15]. The rate con-
[5]. Many different stability indicating assays can be used, including stant derived from simplified models are often of little value during
viral titer, immunochemical assays, liquid chromatography and gel kinetic workflow; for example the determination of the kinetic values of
electrophoresis [6,7]. An often used parameter is shelf life, this refers to pre-exponential factor and activation energy from the Arrhenius plot

E-mail address: didier.clenet@sanofi.com.

https://doi.org/10.1016/j.ejpb.2018.01.005
Received 28 July 2017; Received in revised form 29 November 2017; Accepted 8 January 2018
Available online 16 January 2018
0939-6411/ © 2018 The Author. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).
D. Clénet European Journal of Pharmaceutics and Biopharmaceutics 125 (2018) 76–84

[7,16]. Superior kinetic description of the experimental decomposition specific sandwich ELISA. Freeze-dried products were stored at 5 °C
data of biologicals [17,18] was obtained with kinetic parameters de- before analysis and reconstituted extemporaneously prior to testing.
rived from two-step models, these better mimic the complicated de- Three vials were analyzed for each kinetic time point and a mean of
composition of the investigated samples. Also of great use is the Šesták- these independent dosage values was displayed. Results are system-
Berggren approach [19], being a phenomenological kinetic model atically expressed as a percentage of the mean value at T = 0 and re-
which allows kinetic analysis, even of complex reactions [14,20]. ferred to as antigen recovery. To estimate prediction accuracy, per-
In this report, we applied a general procedure combining advanced centage point was used as the arithmetic difference between
kinetic and statistical analyses of vaccine forced degradation data to antigenicity recoveries, experimentally determined and predicted.
identify kinetic models which best describe loss of antigenicity for three
variants of inactivated viruses in freeze-dried form. Kinetic analysis was 2.3. Advanced kinetic analyses
applied to predict the long-term vaccine degradation of the freeze-dried
products, either under isothermal or specific temperature excursions. A general stability modeling procedure using AKTS-Thermokinetics
Additionally, the antigenicity recoveries predicted by kinetic models software [21] (version 4.1, AKTS AG, Advanced Kinetics and Tech-
were compared with the experimental data of tested samples. nology Solutions, Siders, Switzerland) was already described in detail
Pharmaceutical applications of the presented kinetics-based approach [20] and applied to predict stability for various products constituting
are discussed. vaccines [14]. Briefly, according to a least-squares regression analysis,
the software uses ‘one-step’ and ‘two-step’ kinetic models based on the
2. Material and methods truncated Šesták-Berggren equation (‘two-step’ in Eq. (1) to determine
the function(s) which best fits the experimental data. Hence, reaction
2.1. Materials rate was described as follows [20]:
dα E E
A multivalent freeze-dried inactivated vaccine by Sanofi Pasteur = A1 exp ⎛− 1 ⎞ (1−α )n1α m1 + A2 exp ⎛− 2 ⎞ (1−α )n2α m2
dt ⎝ RT ⎠ ⎝ RT ⎠ (1)
was used for this study; it is composed of three different viral variants
referred as A, B, C. To allow the detection of the antigenicity recovery With α: the reaction progress, A: pre-exponential factor, E: activation
losses during temperature excursions outside of the refrigerated storage energy, n: reaction order, m: a parameter introduced to take into ac-
condition (cold chain break), a thermosensitive formulated product was count the possible autocatalytic behavior of reaction (1) and (2), for the
selected. Final bulk products were prepared using an appropriate sta- first and second step, respectively. During evaluation one, two or more
bilizer containing a mixture of excipients (amino acids, sugar, surfac- reaction stages may be considered, depending on the complexity of the
tant and chelator) in a HEPES buffer to maintain pH at 7. All reagents reaction. For a two-step reaction, consecutive and competitive type
were purchased from Sigma-Aldrich (St. Louis, MO, USA). 0.5 mL of models were screened. Competitive two-step models use one more ad-
final bulk products were dispensed in 3 mL type I glass vials partially justable parameter than consecutive two-step model, with the con-
stoppered with septum closure and subsequently freeze-dried, then tribution of the reaction rate of the first and second stage to the overall
fully stoppered and crimped with aluminum cap. Dried products were reaction rate. The quality of regression fit is quantified according to RSS
investigated with low residual moisture content (0.7%) measured by (Residual Sum of Squares) value. The combination of different kinetic
coulometric titration (Karl Fisher). models best describing the reaction course is identified with software
Antigenicity was tested by enzyme-linked immunosorbent assay according to the higher Akaikie Information Criteria and Baysian In-
(ELISA) at different time intervals. formation Criteria weighted scores, referred to as wAIC and wBIC, re-
spectively. In kinetic analysis, when applying the different models de-
2.2. Stability monitoring and antigen content by ELISA scribing the reaction course, an important issue is to find the best
method for their discrimination [22]. The criteria for comparing models
Product stability was studied by storing freeze-dried formulations at applied in this study were based on the information theory introduced
5 °C, 25 °C, 37 °C and 45 °C into temperature-controlled incubators for by Akaike [23] and its Bayesian counterpart [24,25]. Both these criteria
up to 6 months. This forced degradation plan was carefully designed to indicate not only which model is more likely to better fit the analyzed
obtain experimental datasets able to be fitted by the software, with a data, but also allow quantification of likelihood. An example of criteria
minimum of 20 data points in total obtained at three or more storage application, using limited experimental points for the prediction of
temperatures, with significant degree of reaction reached for the more thermal stability of biological materials, is described in detail elsewhere
drastic condition. Hence, time intervals for the forced degradation plan [20]. Finally, by using the selected model, the software was employed
were chosen, as follows: 0, 30, 90 and 180 days at 5 °C; 7, 14, 30, 60, 90 to run bootstrap analysis (resampled 1000 times with replacement) and
and 180 days at 25 °C; 1, 3, 7, 14, 30, 60 and 180 days at 37 °C, and 1, 3, determine the confidence intervals (CI), in the form of the upper and
7, 14, 30 and 90 days at 45 °C. lower 95 percentiles (predictive band at 95%), which were evaluated
During studies of the influence of temperature excursions, the two for each of the fitted parameters [20]. After determination of the best
following temperature programs were applied to freeze-dried products kinetic models the software was able to simulate the reaction progress
before analysis by ELISA: (i) 1 month at 5 °C, followed by 9 months at during arbitrarily chosen aging time and under any temperature pro-
25 °C and then returned to 5 °C for 2 months; (ii) 1 month at 5 °C, fol- files. We have used this approach to continuously predict degradation
lowed by 6 months at 37 °C and then returned to 5 °C for 4.5 months. To progresses of variants when vaccines were kept under isothermal con-
evaluate the impact of experimental shipments under uncontrolled cold ditions (long-term stability), during temperature excursions and during
chain conditions, product samples kept for 534 days (∼1.5 year) at 5 °C exposures under real atmospheric temperature profiles (e.g. shipments
were shipped from Lyon (France) to Toronto (Canada) at ambient at ambient temperature).
temperature. Another set of samples was also sent from Lyon (France)
to Barcelona (Spain). Real temperature profiles were recorded with 3. Results
temperature monitoring devices (TempTale®4 USB Monitors, Sensitech
Inc., Beverly, USA) placed into the box containing the vaccines. The 3.1. Thermal degradation of virus-based vaccine described by two-step
temperature profiles recorded continuously during shipment were used kinetic models
for predictions. All samples came back to France and were kept at 5 °C
before ELISA analyses. Lots of models, starting from the simplest ones (zero and first-order)
The antigen titer of vaccine samples was determined by a variant- and ending with more complex were screened to fit the six months

77
D. Clénet European Journal of Pharmaceutics and Biopharmaceutics 125 (2018) 76–84

Fig. 1. Kinetic modeling of vaccine antigenicity. Antigenicity predictions are displayed as lines for variants A, B and C at 5 °C (blue), 25 °C (red), 37 °C (green) and 45 °C (pink) based on
one-step models (first-order and zero-order models displayed as solid lines and dashed lines, respectively) in left column and based on two-step models (competitive and consecutive
models are displayed as solid and dashed lines, respectively) in right column. ELISA data used for kinetic modeling are displayed as filled circles. Additional experimental data obtained
later in time, for another vaccine batch, are displayed as open triangles (blue: 5 °C, red: 25 °C after 2.5 months).

experimental data. The kinetic parameters (A, E, n, m) were system- models were preferred to consecutive models for two-step type kinetics.
atically adjusted during the fitting procedure, comprising ‘one-step’ or Consequently, the most appropriate kinetic models for variants A, B,
‘two-step’ kinetic models. The software compared and ranked the and C were competitive two-step models, mathematically described as
models from the worst to the best, for each variant. One-step models follows:
such as zero- and first-order kinetics did not correctly fit experimental Variant A:
data, especially at temperatures higher than 5 °C for variant A and C
dα 83.2E3 119.3E3
⎞ (1−α1) 4 + 0.591∗1.68E 31 exp ⎛− ⎞ (1−α2)2
(Fig. 1, left). Better fits were obtained by using two-step models for all dt
= 0.409∗1.08E 8 exp ⎛−
⎝ RT ⎠ ⎝ RT ⎠ (2)
variants (Fig. 1, right). Based on ranking statistical scores wAIC and
wBIC, competitive two-step model containing six adjustable kinetic Variant B:
parameters were selected, since they provided the best quality of fit dα 72.0E3 197.0E3
= 0.115∗8.89E 6 exp ⎛− ⎞ (1−α1) + 0.885∗5.13E 25 exp ⎛− ⎞ (1−α2)3
(low RSS value) and best wAIC and wBIC scores. It was confirmed for all dt ⎝ RT ⎠ ⎝ RT ⎠ (3)
three serotypes, with wAIC and wBIC scores at least equal to 15%
Variant C:
(Tables S1–S3), in contrast to the other scores, close to zero, obtained
with one-step and consecutive two-step models (Tables S1–S3). Firstly, dα
= 0.327∗4.79E13 exp ⎛−
111.3E3
⎞ (1−α1) + 0.673∗3.48E 20 exp ⎛−
164.1E3
⎞ (1−α2)
this means that the adjustable kinetic parameters used with one-step dt ⎝ RT ⎠ ⎝ RT ⎠ (4)
models were not enough to fit datasets, and secondly, that competitive where for example, in Eq. (2), the values 0.409 and 0.591 represent the

78
D. Clénet European Journal of Pharmaceutics and Biopharmaceutics 125 (2018) 76–84

contribution of the reaction rates of the first and second stages of the In that particular case, no significant loss of antigenicity was predicted
overall reaction rate, 1.08E8 and 1.68E31 depict the values of the pre- when compared to sample kept at 5 °C (Fig. 3, left, middle), indicating a
exponential factors A1 and A2 for first- and second steps, 83.2E3 and lower thermal sensitivity for variant B, compared to variants A and C. In
119.3E3 represent the values of the activation energy E1 and E2, ex- the same condition, at 25 °C, antigenicity evolution were predicted in
ponents 2 and 4 show the value of the reaction order in the first- and two stages for variant A and C, with a rapid initial decrease followed by
second step, respectively. For all three variants, such contribution was a more gradual loss of antigenicity (Fig. 3, left, top and bottom). It
higher for the second stage of the reaction rate. Activation energies should be noted that variant C exhibited a faster initial degradation
obtained for the second stage were also higher than the activation en- profile when exposed at elevated temperature (Fig. 3, bottom), in-
ergies of the first stages (e. g. in Eq. (2), 119.3 kJ/mol for the second dicating a particular thermosensitivity for this variant. Finally, ex-
stage vs. 83.2 kJ/mol for the first stage). Finally, a similar observation posure at 37 °C induced pronounced loss of antigenicity for all variants,
could be made regarding pre-exponential factors with higher values for with two-step profiles (Fig. 3, right). Quantitatively, antigenicity re-
the second stage as compared to the first stage (e. g. in Eq. (2), 1.68E31 coveries were predicted at 72%, 86% and 58% for variants A, B and C,
for the second stage vs. 1.08E8 for the first stage). All three variants respectively when vaccine was exposed for 9 months at 25 °C (Table 2).
were described with competitive two-step models containing fixed Those predictions were later confirmed experimentally, with less than
order, from n = 1 to n = 4 (Eqs. (2)–(4)). five percentage points difference from experimental values (Table 2). In
Based on experimental data generated for up to 6 months, the re- addition, predictions based on two-step kinetic models indicated that
spective models were used for the simulation of stability for 3 years at exposure of vaccine for 6 months at 37 °C induced a loss of antigenicity
5 °C, 25 °C, 37 °C and 45 °C (Fig. 2, left). For the three variants, loss of recoveries up to 44%, 60% and 24% for variants A, B and C, respec-
antigenicity exhibited first a rapid initial decay followed by a gradual tively. Experimental data obtained by ELISA were included in the
decrease (Fig. 2, left). The initial loss of antigenicity occurred with a predictive band (CI 95%), confirming agreement between antigenicity
magnitude depending on incubation temperatures and variants. At 5 °C, recovery predictions and experimental data with not more than five
the first step of the degradation process reached an antigenicity re- percentage points difference (Table 2).
covery of around 85%, 90% and 72% after approximately 1 year,
0.5 year and 1.5 years for variants A, B and C, respectively (Fig. 2, left). 3.2.2. Confronting vaccine stability predictions with real data obtained after
Then, the second step of the degradation process was predicted to be shipment of products at ambient temperatures
more gradual and slowly reached an antigenicity recovery around 80%, Recorded oscillations around ambient temperature indicated
88% and 67% for variants A, B and C, respectively after 3 years at 5 °C minimum/maximum temperatures at 21 °C/27 °C during shipment to
(Fig. 2, left). This two-step degradation behavior looked more pro- Toronto and at 4 °C/48 °C during shipment to Barcelona (Fig. 4). Gra-
nounced with higher storage temperatures and depending on variants. dual decreases of antigenicity for all the three variants were predicted
As an example, 50% of antigenicity recovery was predicted to be by the software (Eqs. (2)–(4)) depending on the recorded temperature
reached after 2 years at 25 °C for variant A, 1.5 years for variant C and profiles (i.e. for shipment in Barcelona – Fig. 4). Predictions identified
for variant B 3 years and beyond (data not shown), making this ob- variant B as the most stable and variant C as the least stable. Quanti-
viously the more thermostable variant (Fig. 2, left). Incubation at high tatively, antigenicity recoveries after the Lyon – Barcelona round trip
temperatures (37 °C, 45 °C) rapidly induced loss of antigenicity of sev- were predicted at 81%, 88% and 66% for variants A, B and C, respec-
eral tens of percents (Fig. 2, left). This phenomenon was especially tively (Table 3). Those predictions were later confirmed experimen-
pronounced for variant C, obviously the more thermosensitive variant. tally, with approximate five percentage points difference from experi-
Focusing on 5 °C, the experimental data generated for 6 months are mental values (Table 3). Interestingly, similar antigenicity recoveries
inside the predictive band determined by bootstrap analysis (CI 95% – were obtained after the return flight between Lyon and Toronto (82%,
Fig. 2, right). The same was observed for 2.5 months data originating 89% and 67% for variants A, B and C, respectively – Table 3).
from another batch (Fig. 2, right, open triangles). Similarly, further To assess the impact of maintaining freeze-dried products at am-
experimental data (obtained later) not included in kinetic modeling, bient temperature for an extended period of time, we designed a case
collected after 1.8 years at 5 °C for all variants and after 2.8 years for study using simulations of storage at ambient temperature in Kenya
variant A, were mostly contained within the predictive bands (Fig. 2, (Mombassa) and Argentina (Buenos Aires). The software mimicked the
right). Only one of the three data points was outside the predictive band real atmospheric temperature profiles in both locations, with oscilla-
for variant B. Quantitatively, predictions were performed for each tions between 23 °C and 32 °C for Mombassa, and between 4 °C and
variant based on their respective two-step models (Eqs. (2)–(4)), as- 30 °C for Buenos Aires (Fig. S1). The impact of such storage outside the
suming an aging of 1.8 years at 5 °C. Antigenicity recoveries, which standard refrigerated conditions on vaccine antigenicity was predicted
were predicted at 82%, 89% and 68% for variants A, B and C, respec- for 2 years for variant A, B and C as a rapid initial loss of antigenicity,
tively, were later confirmed by real stability data, with approximately followed by a gradual decrease (Fig. S1). The first degradation step
five percentage points difference from experimental values (Table 1). predicted a loss of antigenicity of variant A to approximately 79% and
72% in Buenos Aires and Mombassa, respectively (Fig. S1), and a gra-
3.2. Vaccine stability predicted during excursions of temperature dual decrease to approximately 72% and 64% after 2 years, in Buenos
Aires and Mombassa, respectively (Fig. S1). For comparison, anti-
3.2.1. Confronting vaccine stability predictions with real data obtained after genicity recovery of variant A kept under refrigerated conditions
experimental temperature excursions for months at 25 °C and 37 °C (2–8 °C) for 2 years was predicted at 82%. For variant B, the first pre-
Based on kinetic modeling performed with 6 months forced de- dicted degradation step amounted to approximately 90% in both
gradation data, selected ‘two-step’ models (Eqs. (2)–(4)) were applied countries, followed by a gradual decrease to approximately 87% and
to temperature excursion to determine if antigenicity of variants could 79% after 2 years in Buenos Aires and Mombassa, respectively (Fig. S1).
be adequately predicted. Since the recommended vaccine storage con- Finally, the predictions for variant C indicated a loss of antigenicity up
dition is 5 °C ± 3 °C, excursions to higher temperatures (9 months at to approximately 65% in both countries for the first rapid step, and to
25 °C and 6 months at 37 °C) were applied to assess the degree of de- approximately 60% and 40% after 2 years in Buenos Aires and
gradation obtained at other arbitrarily selected storage conditions and Mombassa, respectively (Fig. S1).
verify the simulated predictions. Antigenicity changes under these
temperature excursions were simulated on the basis of the selected 4. Discussion
model. Gradual decreases were predicted for all variants during ex-
posure at elevated temperatures, except for variant B at 25 °C (Fig. 3). In this study, an experimental vaccine used as complex biomaterial

79
D. Clénet European Journal of Pharmaceutics and Biopharmaceutics 125 (2018) 76–84

Fig. 2. Long-term prediction of vaccine antigenicity based on two-step kinetic models. Left column: Antigenicity predictions are displayed as lines for variants A, B and C for 3 years at
5 °C (blue), 25 °C (red), 37 °C (green) and 45 °C (pink). ELISA data used for kinetic modeling are displayed as filled circles. Right column: At 5 °C, antigenicity predictions (lines) are
shown with predictive band representing 95% CI (dotted lines). Experimental data used for kinetic modeling are displayed as filled circles. Additional experimental data obtained later in
time and not used for kinetic modeling are displayed as open circles, after 1.8 and 2.8 years for variant A and after 1.8 years for variants B and C. Experimental data obtained for another
vaccine batch are also displayed as open triangles.

Table 1
Comparison of antigenicity predicted by kinetic models (one-step and two-step) with experimental data (average of three independent ELISA results ± standard deviation) for vaccine
containing variants A, B and C kept under refrigerated conditions (2–8 °C). Differences between prediction and experimental antigenicity recovery are displayed (percentage point).

Time under refrigerated conditions Antigenicity recovery (%)

Two-step model One-step model (first-order)

Variant Experimental data ± SD (%) Prediction (%) Percentage point (%) Prediction (%) Percentage point (%)

1.8 years A 87 ± 2 82 −5 97 10
B 94 ± 8 89 −5 99 4
C 74 ± 8 68 −6 99 25

2.8 years A 79 ± 8 79 0 94 15

80
D. Clénet European Journal of Pharmaceutics and Biopharmaceutics 125 (2018) 76–84

Fig. 3. Vaccine antigenicity predicted from kinetic models during temperature excursions outside the cold-chain. Left column: Antigenicity predictions are displayed as lines for variant
A, B and C for customized temperature excursions for 9 months in incubator set at 25 °C (red, solid line) and for 12 months under refrigerated conditions (2–8 °C, blue and dotted line).
Experimental data determined at the end of temperature excursions and not used for kinetic modeling are displayed as open red circles. Right column: Antigenicity predictions for
variants A, B and C for customized temperature excursions for 6 months in incubator set at 37 °C (red, solid line) and for 12 months under refrigerated conditions (2–8 °C, blue and dotted
line). Experimental data determined at the end of temperature excursions and not used for kinetic modeling are displayed as open red circles.

and containing three variants (variant A, B and C) was thermally kinetics were applied, leading to long-term antigenicity predictions
stressed in a forced degradation program comprising of four incubation overestimated by up to 25%. Software such as ASAPprime® [26,27] and
temperatures ranging between 5 °C and 45 °C. Vaccine antigenicity was ScienTek® [28] are not able to correctly describe the course of the
periodically evaluated by ELISA up to 6 months for each variant. No or complex degradations of viruses studied here. AKTS-Thermokinetics
partial fit of experimental data was shown when zero- and first-order software [21] however, applies simultaneous combination of two-step

Table 2
Comparison of antigenicity predicted by two-step models with experimental data (average of three independent ELISA results ± standard deviation) for vaccine containing variants A, B
and C, subjected to customized temperature excursions at 25 °C and 37 °C. Differences between prediction and experimental antigenicity recovery are displayed (percentage point).

Shipments and storage conditions Antigenicity recovery (%)

Two-step model

Variant Experimental data ± SD (%) Prediction (%) Percentage point (%)

1 month in a cold chamber (2–8 °C), followed by 9 months in an incubator set at 25 °C, then A 68 ± 6 72 4
returned to the cold chamber for 2 months B 83 ± 14 86 3
C 56 ± 4 58 2

1 month in a cold chamber (2–8 °C), followed by 6 months in an incubator set at 37 °C, then A 44 ± 2 47 3
returned to the cold chamber for 4.5 months B 60 ± 4 62 2
C 24 ± 5 19 5

81
D. Clénet European Journal of Pharmaceutics and Biopharmaceutics 125 (2018) 76–84

degradation. The selection of two-step reactions model is purely based


on the statistical analysis and goodness of fit with two major pathways
of phenomenological kinetic model of the reaction rate, which are most
likely built up from the sum of multiple reactions. At that point, it is
necessary to clearly underline that this result does not have a real
mechanistic basis, but it rather only depicts a phenomenological
mathematical model applied for fitting the reaction course. Kinetic
models mathematically described in Eqs. (2)–(4) for variant A, B and C
respectively, showed a higher contribution of the second step (gradual
decrease of antigenicity) as compared to the first step (rapid loss of
antigenicity). This is particularly marked for variant B, indicating that
stability is mainly governed by the second step with a contribution of
Fig. 4. Vaccine antigenicity predicted from kinetic models during experimental shipment
88.5%, as compared with the first step at 11.5% (Eq. (3). This very slow
of vaccine containing variants A, B and C performed at ambient temperature (red, solid
rate of antigenicity decrease predicted by the kinetic model was found
lines, top). Temperature oscillation (4–48 °C) suffered by vaccine during shipment is
displayed as red, solid line (bottom). For comparison, antigenicity predictions for samples to be in agreement with real stability data presented in this study
kept under refrigerated conditions (real temperature oscillation as blue line, bottom) are (Figs. 2 and 3, Tables 1 and 2). Variant B was shown to be the most
displayed for variants A, B and C as blue and dotted line (top). thermostable vaccine variant.
Conversely, results obtained demonstrated that kinetic models de-
models enabling the consideration of an unlimited amount of kinetic scribing degradation changes of variants A, B and C are able to accu-
models, for the best description of the reaction course. When complex rately predict vaccine stability under any kind of storage conditions,
degradation progresses were observed experimentally, it was demon- with just a few percentage points difference from experimental values
strated that the application of wAIC and wBIC scores allowed determi- (Tables 1–3). First of all, long-term stability at usual recommended
nation of whether one or two-steps model was more likely to be correct storage temperature for freeze-dried product (5°C ± 3 °C) was pre-
[20]. These criteria take into account not only the quality of fit, such as dicted with success for all variants, with less than 6% as differences
the sum of residual squares (RSS), but also the number of experimental between predicted and experimental values (Table 1). The use of the
points available and model parameters used. Additionally, they de- Monte Carlo method, such as bootstrap analysis, provides accurate
termine which model is more likely to be correct, and quantify how predictions. It was already shown that the bootstrap CI gives a tight and
much more likely. Hence, possible errors of over-fitting were avoided, realistic estimate of the true CI [33]. One of the main interests of
and it was statistically confirmed that the two-step models better de- bootstrap CI determination is that this approach takes into account the
scribe the loss of antigenicity of variants A, B and C. Thus, exhibiting scattering of experimental data without imposing a Gaussian distribu-
highest statistical scores (wAIC and wBIC), competitive two-step models tion around predicted values, and that it does not need to know the
were selected for each variant (see Eqs. (2)–(4) and Tables S1–S3), standard deviation of the analytical technique used. For the prediction
describing a rapid initial loss of antigenicity followed by a gradual of degradation processes, the lower 95 percentile (predictive band at
decrease. 95%), obtained with bootstrap analysis, can advantageously be used to
Two-phase degradation profiles have already been described during determine product shelf life, i.e. time to reach the lowest acceptable
accelerated stability studies of several viruses (measles vaccine [29], specification value. This approach could improve applicable standards
attenuated RBOK vaccine [30], canine distemper vaccine [31], ALVAC used to determine shelf life, such as compliance with the acceptance
poxvirus [14], and a freeze-dried live, attenuated virus vaccine [15]). criterion as the time associated with the last measurement within the
For these enveloped viruses, from different viral families, two steps in specification [4].
the degradation process is also described (initial rapid drop followed by Our results confirm that the impact of experimental shipments with
a long gradual decrease phase), regardless of the readout used (in- rupture in the cold chain and short time excursions above 40 °C can be
fectious tiers, antigenic titers etc.) [15,29,32]. One hypothesis put predicted with high accuracy. During shipments to Lyon-Barcelona and
forward in such forced virus degradation studies is that this biphasic Lyon-Toronto, predictions in term of antigenicity losses were confirmed
behavior entails the presence of at least two components with different by experimental ELISA data, with no more than 6% expressed as dif-
inactivation kinetics which are affected by the thermal treatment ferences between predicted and experimental values (Table 3). Inter-
[29,32] (e.g. epitope unfolding, capsid integrity loss, major virus con- estingly, even if temperature excursions endured by freeze-dried pro-
formational reorganization, key protein functionality loss or any ducts during our experimental shipments were different, including
adaptive viral strategy). It is important to mention that many chemical different gaps between the minimum and the maximum exposure
degradations and physical transitions occur during virus thermal temperatures, similar predictions were obtained regarding the level of

Table 3
Comparison of antigenicity predicted by two-step models with experimental data (average of 3 independent ELISA results ± standard deviation) for vaccine containing variants A, B and
C, subject to experimental shipments performed at ambient temperature. Differences between prediction and experimental antigenicity recovery are displayed (percentage point).

Storage conditions Antigenicity recovery (%)

Two-step model

Min. – Max. temp. exposure Variant Experimental data ± SD (%) Prediction (%) Percentage point
during shipment (°C) (%)

534 days in a cold chamber (2–8 °C) then 10 days travel at 21–27 A 86 ± 3 82 −4
ambient temperature during shipment from Lyon to Toronto B 90 ± 7 89 −1
C 69 ± 18 67 −2

534 days in a cold chamber (2–8 °C) then 10 days travel at 4–48 A 80 ± 6 81 1
ambient temperature during shipment from Lyon to B 94 ± 4 88 −6
Barcelona C 69 ± 9 66 −3

82
D. Clénet European Journal of Pharmaceutics and Biopharmaceutics 125 (2018) 76–84

degradation of variants. This observation could be explained by the fact and adjuvants (e.g. emulsions, immunomodulators) could also be suc-
that shipments were performed after 1.5 years of storage at 5 °C. In- cessfully predicted. Such forecasts may significantly support the for-
deed, after this period of time, the initial rapid loss of antigenicity was mulation, process development and help in optimization of supply
passed and samples were in the second step of the degradation process, chain procedures.
engaging thermo-resistant behavior with a gradual decrease of anti-
genicity. As shipments were executed during the stable phase, it could 5. Conclusion
be assumed that variants were less impacted by temperature oscillation
profiles, which could explain why antigenicity losses were similar de- A kinetic-based modeling approach was studied to evaluate in-
spite such different shipment conditions (by car to Barcelona and by activated virus-based vaccine stability, estimate impact of temperature
plane to Toronto). In addition, the simulation indicated that these excursions outside recommended storage conditions and predict the
shipments, performed at ambient temperature, did not dramatically long-term stability of a vaccine containing three variants (variant A, B
impact antigenicity level when compared to shipments performed and C), formulated as a freeze-dried product. Based on two-step kinetic
under controlled refrigerated conditions at 5 °C. Thus, predicted anti- models, losses of virus antigenicity were accurately predicted for freeze-
genicity values only differed by 2.5% between experimental shipments dried product in isothermal long-term storage conditions, as well as in
Lyon-Barcelona performed under controlled cold chain versus ambient real temperature profiles (i.e. temperature excursions during ship-
temperature. This result illustrated how kinetic-based modeling could ments, maintaining vaccine products at ambient temperature in dif-
help to recommend storage conditions for vaccine shipments and to ferent climatic zones). Based on experimental data collected during
evaluate the quality of vaccine under any temperature exposure. several months, these results uncover the potential of a kinetic-based
Kinetic-based modeling approach presented in this work can ad- modeling approach to (i) predict shelf life of vaccines, (ii) recommend
vantageously be used to evaluate impact of storage temperature on appropriate storage conditions, and (iii) evaluate the impact of tem-
vaccine quality. It was illustrated by simulating antigenicity losses for perature excursions (cold chain breaks), with an error amounting to
variant A, B and C during 2 years under different storage conditions (i.e. approximately five percentage points. From these results, it seems
ambient temperature in Mombassa, in Buenos Aires and under re- reasonable to assume that development of recommendations, based on
frigerated conditions). Depending on variants, antigenicity losses advanced kinetics and stability modeling approaches can be a very
reached from 10% to 60% for a vaccine kept at ambient temperature, useful tool. It can also support the conclusions gained from procedures
whereas for vaccines kept in refrigerated conditions the losses were described in international guidelines [8,38,39]. The application of ad-
limited to approximately 10% to 30%. One can take advantage of such vanced kinetic software may have a significant impact on shortening
long-term predictions to justify, document and recommend the storage time-consuming shelf life determinations, evaluating impact of tem-
temperature of products. Improvements in knowledge of vaccine sta- perature excursions on vaccine efficacy, and evaluating the pharma-
bility behavior, and their thermosensitivity level could certainly have ceutical product stability of vaccines, monoclonal antibodies, antibody-
economic impacts and financial gain. drug conjugates, peptides and viral vectors.
It was stated that the inherent temperature sensitivity of vaccines is
particularly significant for live virus-based vaccines that require careful Acknowledgments
control of the storage cold chain [34]. As the cold chain in the last mile
is particularly demanding, a controlled temperature chain (CTC) label The author would like to express his cordial thanks to Marie-Julie
[35] was recently implemented to ensure that vaccines can withstand a Diana, Elisabeth Vialon and Romain Pizatto for ELISA analyses, Rayane
temperature excursion up to 40 °C for up to four days. The kinetic-based Seba, Laure Cigolotti and Manvi Hasija for their technical support
approach described here could easily, and advantageously, be used to during shipments of samples and Aure Saulnier for fruitful discussions
predict degradation profiles of vaccines during storage at 40 °C for four and critical scientific review of the manuscript. Editorial assistance with
days. In addition, kinetic modeling is able to predict the impact of any the preparation of the manuscript was provided by Rebecca Hornby,
kind of temperature excursions, independent of their complexity pro- inScience Communications, Springer Healthcare. Funding for this as-
vided that temperature was accurately recorded and used for predic- sistance was provided by Sanofi Pasteur.
tion. Hence, new ways of supply chain management could emerge. The Didier Clenet is an employee of Sanofi Pasteur.
accurate prediction of changes in vaccine properties during shipment
would require appropriate controlled temperature monitoring which, in Appendix A. Supplementary material
turn, allows the use of real temperature profiles in the kinetic analysis.
Hence, electronic time-temperature devices would be able to inform, in Supplementary data associated with this article can be found, in the
real time, the level of vaccine degradation provided that specific and online version, at https://doi.org/10.1016/j.ejpb.2018.01.005.
convenient kinetic models such as the ones described in this study could
be integrated into compact and connected devices, suggesting real-time References
tracking of temperature excursions could be achieved. This could be of
major interest, knowing that maintaining the cold chain for storing and [1] D. Chen, D. Kristensen, Opportunities and challenges of developing thermostable
transporting vaccines is often difficult for health workers in poor vaccines, Expert Rev. Vaccines 8 (2009) 547–557.
[2] D.T. Brandau, L.S. Jones, C.M. Wiethoff, J. Rexroad, C.R. Middaugh, Thermal sta-
countries, especially in remote areas. The WHO has stated that as many bility of vaccines, J. Pharm. Sci. 92 (2003) 218–231.
as 25% of all vaccine products reach their destination in a degraded [3] A. Black, E-Logistics in Cold Chain Management, <http://www.samedanltd.com/
state [3] and it was demonstrated that major providers expose vaccine magazine/11/issue/17/article/899> (accessed on 05/22/2017).
[4] World Health Organization, WHO/BS/06.2049, Guidelines on Stability Evaluation
products to inappropriate temperatures [36]. Considering that the cold of Vaccines, 2006.
chain, from production to use of vaccines, amounts to about 80% of the [5] T.L. Schofield, Vaccine stability study design and analysis to support product li-
total cost of vaccination programs [37], supply chain product man- censure, Biologicals 37 (2009) 387–396.
[6] M. Hasija, L. Li, N. Rahman, S.F. Ausar, Forced degradation studies: an essential tool
agement integrating kinetic modeling could significantly reduce fi- for the formulation development of vaccines, Vaccine Dev. Ther. 3 (2013) 11–33.
nancial losses due to cold chain breaks. [7] J. Zhang, E. Pritchard, X. Hu, T. Valentin, B. Panilaitis, F.G. Omenetto, D.L. Kaplan,
Finally, the experimental and thermosensitive virus-based vaccine Stabilization of vaccines and antibiotics in silk and eliminating the cold chain, Proc.
Natl. Acad. Sci. U.S.A. 109 (2012) 11981–11986.
used for this study, can be considered as one of the most complex
[8] International Conference on Harmonisation (ICH) Q1E, CPMP/ICH/420/02,
biomaterials with multi-step degradation processes. Therefore, it is ra- Evaluation of stability data, current Step 4 version dated 6 February 2003 by the
tional to assume that stability behavior of other biologicals (e.g. pep- ICH Steering Committee, Evaluation of stability data, 2003.
tides, viral vectors, monoclonal antibodies, antibody-drug conjugates) [9] R. Capen, D. Christopher, P. Forenzo, C. Ireland, O. Liu, S. Lyapustina, J. O’Neill,

83
D. Clénet European Journal of Pharmaceutics and Biopharmaceutics 125 (2018) 76–84

N. Patterson, M. Quinlan, D. Sandell, J. Schwenke, W. Stroup, T. Tougas, On the parameters of gegradation of anthocyanins in grape pomase, J. Food Process Eng.
shelf life of pharmaceutical products, AAPS PharmSciTech 13 (3) (2012) 911–918. 34 (2011) 1220–1233.
[10] W. Stroup, M. Quinlan, Alternative shelf life estimation methodologies, JSM [26] K.C. Waterman, The application of the accelerated stability assessment program
Proceedings, Biopharmaceutical Section, American Statistical Association, (ASAP) to quality by design (QbD) for drug product stability, AAPS PharmSciTech.
Alexandria, VA, 2010, pp. 2056–2066. 12 (2011) 932–937.
[11] M. Quinlan, W. Stroup, J. Schwenke, D. Christopher, Evaluating the performance of [27] R. Waterman, J. Lewis, K.C. Waterman, Accelerated stability modeling for peptides:
the ICH guidelines for shelf-life estimation, J. Biopharm. Stat. 23 (2013) 881–896. a case study with bacitracin, AAPS PharmSciTech 18 (5) (2017) 1692–1698.
[12] V. Kayser, N. Chennamsetty, V. Voynov, B. Helk, K. Forrer, B.L. Trout, Evaluation of [28] ScienTek®, <http://www.stabilitysystem.com/statistical-data-analysis.htm>.
a non-Arrhenius model for therapeutic monoclonal antibody aggregation, J. Pharm. [29] L.M.C. Allison, G.F. Mann, F.T. Perkins, A.J. Zuckerman, An accelerated stability
Sci. 100 (2011) 2526–2542. test procedure for measles vaccines, J. Biol. Stand. 9 (1981) 185–194.
[13] A. Oliva, A. Santovena, M. Llabres, J.B. Farina, Stability study of human serum [30] J.C. Mariner, J.A. House, A.E. Sollod, C. Stem, M. Van Den End, C.A. Mebus,
albumin pharmaceutical preparations, J. Pharm. Pharmacol. 51 (1999) 385–392. Comparison of the effect of various chemical stabilizers and lyophilization cycles on
[14] D. Clenet, F. Imbert, P. Probeck, N. Rahman, S.F. Ausar, Advanced kinetic analysis the thermostability of a vero cell-adapted rinderpest vaccine, Vet. Microbiol. 21 (3)
as a tool for formulation development and prediction of vaccine stability, J. Pharm. (1990) 195–209.
Sci. 103 (2014) 3055–3064. [31] P.M. Precausta, D. Simatos, M. Le Pemp, B. Devaux, F. Kato, Influence of residual
[15] L. Hansen, J.V. Renterghem, R. Daoussi, C. Vervaet, J.P. Remon, T.D. Beer, moisture and sealing atmosphere on viability of two freeze-dried viral vaccines, J.
Spectroscopic evaluation of a freeze-dried vaccine during an accelerated stability Clin. Microbiol. 12 (4) (1980) 483–489.
study, Eur. J. Pharm. Biopharm. 104 (2016) 89–100. [32] F. Higashikawa, L.J. Chang, Kinetic analyses of stability of simple and complex
[16] R.K. Evans, D.K. Nawrocki, L.A. Isopi, D.M. Williams, D.R. Casimiro, S. Chin, retroviral vectors, Virology 280 (1) (2001) 124–131.
M. Chen, D.M. Zhu, J.W. Shiver, D.B. Volkin, Development of stable liquid for- [33] D.K. Mishra, K.D. Dolan, L. Yang, Bootstrap confidence intervals for the kinetics
mulations for adenovirus-based vaccines, J. Pharm. Sci. 93 (2004) 2458–2475. parameters of degradation of anthocyanins in grape pomace, J. Food Process Eng.
[17] M.A. Watzky, A.M. Morris, E.D. Ross, R.G. Finke, Fitting yeast and mammalianprion 34 (2011) 1220–1233.
aggregation kinetic data with the Finke-Watzky two-step model of nucleation and [34] U. Kartoglu, J. Milstien, Tools and approaches to ensure quality of vaccines
autocatalytic growth, Biochemistry 47 (2008) 10790–10800. throughout the cold chain, Expert Rev. Vaccines 13 (7) (2014) 843–854.
[18] A.M. Morris, M.A. Watzky, R.G. Finke, Protein aggregation kinetics, mechanism and [35] S. Zipurskya, M. Harouna Djingarey, J.C. Lodjo, L. Olodo, S. Tiendrebeogo,
curve-fitting: a review of the literature, Biochim. Biophys. Acta 1794 (2009) O. Ronveaux, Benefits of using vaccines out of the cold chain: Delivering Meningitis
375–397. A vaccine in a controlled temperature chain during the mass immunization cam-
[19] J. Šesták, G. Berggren, Study of the kinetics of the mechanism of solid-state reac- paign in Benin, Vaccine 32 (2014) 1431–1435.
tions at increasing temperature, Thermochm. Acta 3 (1971) 1–13. [36] D.R. Levinson, Vaccines for children program: vulnerabilities in vaccine manage-
[20] B. Roduit, M. Hartmann, P. Folly, A. Sarbach, R. Baltensperger, Prediction of ment. Department of Health and Human Services, Office of Inspector General OEI-
thermal stability of materials by modified kinetic and model selection approaches 04-10-00430 (2012), available from: http://www.oig.hhs.gov/oei/reports/oei-04-
based on limited amount of experimental points, Thermochim. Acta 579 (2014) 10-00430.pdf.
31–39. [37] Making vaccines not need refrigeration, http://blog.pnas.org/2013/04/making-
[21] AKTS-Thermokinetics Software, <http://www.akts.com>. vaccines-not-need-refrigeration/, posted on April 16 (2013) by PNAS (accessed on
[22] J.B. Johnson, K.S. Omland, Model selection in ecology and evolution, Trends Ecol. 05/22/2017).
Evol. 19 (2004) 101–109. [38] International Conference on Harmonisation (ICH) Q5C, CPMP/ICH/138/95,
[23] H. Akaike, A new look at the statistical model identification, IEEE Trans. Automatic Quality of Biotechnological Products: Stability Testing of Biotechnological/
Control 19 (1974) 716–723. Biological Products, 1996.
[24] K.P. Burnham, D.R. Anderson, Model Selection and Multimodel Inference: A [39] European Medicines Agency, EMA/CHMP/BWP/354898/2008, Guideline on the
Practical Information – Theoretic Approach, second ed., Springer, New York, 2002. requirements for quality documentation concerning biological investigational
[25] D.K. Mishra, K.D. Dolan, L. Yang, Bootsrap confidence intervals for the kinetic medicinal products in clinical trials, 2012.

84

Potrebbero piacerti anche