You are on page 1of 6

Journal of Pharmaceutical Sciences 111 (2022) 2639−2644

Contents lists available at ScienceDirect

Journal of Pharmaceutical Sciences


jo urn a l h om ep ag e : w ww.jp harms ci.o rg

Note

Impact of Annealing and Controlled Ice Nucleation on Properties


of A Lyophilized 50 mg/ml MAB Formulation
Jijun Wanga,*, James A. Searlesa, Ekaterina Torresa, Serguei A. Tchessalovb,
Anthony L. Younga
a
Pharmaceutical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO 63017, USA
b
Pharmaceutical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Andover, MA 01810, USA

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

Article history: We compared “ice-fog” controlled ice nucleation at -6 °C to annealing at the same temperature for a
Received 7 January 2022 50 mg/mL monoclonal antibody formulation, using shelf-ramp freezing as a control. Cake structure, drying
Revised 18 May 2022 time, reconstitution time, specific surface area, calculated cake resistance and size exclusion chromatography
Accepted 18 May 2022
were all compared. Controlled nucleation resulted in the fastest reconstitution, shortest primary drying, low-
Available online 22 May 2022
est calculated cake resistance, lowest specific surface area and highest moisture content. There was no effect
upon the results for size exclusion chromatography. Results for annealing were between those for controlled
Keywords:
nucleation and shelf-ramp freezing. All results were consistent with “ice-fog” controlled nucleation at -6 °C
Freeze-drying
Lyophilization
having greater impact upon the ice crystal morphology than annealing at the same temperature for 3 h.
Protein © 2022 The Authors. Published by Elsevier Inc. on behalf of American Pharmacists Association. This is an open
Monoclonal antibody access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/)
Morphology
Nucleation
Protein formulation(s)
Drying
Solid-state
Amorphism

Introduction There are several reports that show annealing can reduce recon-
stitution time when annealing results in crystallization of formula-
Lyophilization is a well-established drying process for stabilization tion components such as mannitol.11,12 Annealing as also been
of biologics.1-4 There are three major steps in the lyophilization pro- shown to affect reconstitution time for amorphous protein formula-
cess: freezing (solidification) (may or may not include an annealing tions similar to what we used.13-16 Blue et al. showed that incorporat-
step), primary drying (ice sublimation), and secondary drying (mois- ing an annealing step after freezing can reduce the reconstitution
ture desorption).1,5 The structure of the dry layer above the ice inter- time for two separate mAbs by up to 60%.13
face determines the cake resistance to water vapor flow during Controlled ice nucleation (CN) is receiving consistent attention
primary drying, and hence the required primary drying time. This due to its benefits in terms of improving lyophilization process per-
structure, while it can be affected during drying by temperature formance and enhancing product quality attributes of lyophilized
excursions above the collapse temperature, is initially set by the freez- drug products. Several controlled ice nucleation systems have been
ing and annealing steps.6 The final cake structure has a strong influ- developed and are commercially available, such as ice-fog, pressuri-
ence upon reconstitution time and other product quality attributes. zation/depressurization, vacuum-induced, or partial vacuum.17,18
Annealing is frequently used after shelf-ramp freezing to reduce Several papers have reported that controlled ice nucleation can
product resistance during primary drying, resulting in a higher pri- also reduce the reconstitution time of high concentration protein
mary drying rate.7-9 However, annealing also causes reduction of the formulations.12,16,19,20 Geidobler et al. found that CN at -5 °C led to a
product specific surface area, which can decrease the water desorp- 66% reduction in reconstitution time of a lyophilized mAb at
tion rate in secondary drying and may lead to increased residual 161 mg/mL.19 About 50% reduction was reported by Singh et al. for a
moisture content in the final product.4,10 mAb at 108 mg/mL.20
In contrast to others, Luoma et al. compared different CN techni-
ques with 10 mg/mL and 100 mg/mL mAb formulations and found
* Correspondence author. that the partial vacuum and depressurization techniques actually

https://doi.org/10.1016/j.xphs.2022.05.016
0022-3549/© 2022 The Authors. Published by Elsevier Inc. on behalf of American Pharmacists Association. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)
2640 J. Wang et al. / Journal of Pharmaceutical Sciences 111 (2022) 2639−2644

increased reconstitution time, and that ice-fog CN produced results “Controlled nucleation -6 8C” cycle used a VERISEQÒ ice-fog nucleation
similar to shelf-ramp freezing.18 system (IMA Life/Linde, Tonawanda, NY). For this process, the sample
There is a lack of published data comparing annealing and con- vials were loaded and held on the -6 8C shelf for 2.5 h, after which the
trolled nucleation in which both processes are carried out at the same chamber pressure was reduced to 2.8 psia and nitrogen gas carrying
temperature. We compared controlled ice nucleation at a shelf tem- ice crystals was introduced into the chamber to induce nucleation. The
perature of -6 °C with annealing at same temperature (-6 °C), evaluat- shelf temperature was then ramped to -45 8C at 0.58C/min.
ing cake structure, primary drying time, reconstitution time, specific Primary drying conditions were set to maintain the product tem-
surface area, and size exclusion chromatography. We saw improve- perature near or below its collapse temperature while in the presence
ments in lyophilization cycle time and in product reconstitution time of ice, which was determined to be approximately -28 8C using
with controlled nucleation and found that similar benefit obtained by freeze-drying microscopy. At the end of freeze drying, the product
using controlled nucleation could also be achieved using annealing. chamber was backfilled with nitrogen to approximately 600 Torr and
the vials were stoppered before unloading. Samples were crimped to
Materials and Methods seal with flip-off seals and kept refrigerated until analysis.

Materials Moisture Determination

The protein used in this study was a IgG2 monoclonal antibody Karl Fischer (Mettler Toledo, Columbus, OH) was used to determine
(mAb) manufactured by Pfizer. The mAb formulation was 50 mg/mL residual water content in the lyophilized cake. The titration sample was
IgG2 in 10 mM histidine buffer pH 5.5 containing 8% sucrose and prepared by dissolving/suspending the cake in a diluent of 50% formam-
0.2 mg/mL polysorbate-20. The excipients chosen for the studies were ide/ 50% methanol mixture. The samples were then vortexed to extract
based on those typically used in protein formulations.21 The mAb bulk residual water before the coulometric titration for water content.
formulation was filtered through a 0.22 mm membrane filter and was
filled into 10 mL Type I clear glass vials with 4 mL fill volume in a gen- Scanning Electron Microscopy (SEM)
eral laboratory environment. The vials were partially stoppered with
20 mm FlurotecÒ coated butyl rubber stoppers for lyophilization. Lyophilized cake samples were mounted on Scotch tape-covered
Histidine was purchased from JT Baker (Center Valley, PA). Poly- stubs using silver paste. Samples were sputter-coated with 10 mm gold
sorbate-20 (containing low carbonyl and peroxide) was purchased using Gressington 108 instrument (Gressington Scientific Instruments,
from Thermo Scientific (Rockford, IL). Sucrose was obtained from Watford, UK) prior to imaging. Gold coated samples were analyzed using
Pfanstiehl Laboratories (Waukegan, IL). EDTA was from JT Baker (Cen- a JOEL JCM-600-Plus scanning electron microscope (JOEL, Peabody, MA)
ter Valley, PA). Distilled, deionized water was used throughout. using secondary electron detector at 5 keV and 200-300 mBar.

Lyophilization and Controlled Nucleation (CN) Specific Surface Area (SSA)

All samples were freeze-dried using a SP LyoStar-3 lab scale Gemini 2380 instrument (Micromeritics, Norcross, GA) was used
freeze-dryer (SP Scientific, Stone Ridge, NY). One shelf was loaded to measure the SSA of the freeze-dried samples. Samples were placed
with 30-50 samples surrounded by placebo vials to completely fill in the bulb glass tubes in a dry box and passively purged with dry
the shelf. Product temperature was measured using thermocouples nitrogen for at least 48 h. The instrument provides an option to use
inserted into sample vials. Table 1 shows the freezing and drying con- free space balancing with the glass beads and this approach makes
ditions used for the three different cycles tested. nitrogen gas acceptable for measurement of low SSA. Sample and ref-
The controlled nucleation and annealing temperature of -6 °C (shelf erence tubes were evacuated at 200 mmHg/min. SSA of each sample
temperature) was selected to be far enough below zero to give confi- was measured using Nitrogen with multi-point BET analysis method.
dence that even the edge vials on the shelf would not melt completely. Free space differential was measured using helium. To facilitate
For shelf ramp freezing without annealing, the shelves were at 20 °C measurements with the low SSA tube was partially filled with glass
for loading, the chamber was ramped at 1 °C/min to 5 8C and then beads to minimize differences in the free space volume. The weight
ramped at 0.5 °C/min to -45 8C and holding for 90 minutes. The cycle of samples was used to calculate the specific surface area
named “Annealed -6 8C” samples were loaded on the shelf pre-cooled
to 5 8C, held for 10 minutes, then the shelf temperature was ramped Reconstitution Time
down to -45 °C at 0.5 8C/min then held for 90 minutes. For annealing
the shelf was ramped up to -6 8C at 0.5 8C/min, held for 3 h, re-cooled The lyophilized cakes were reconstituted by addition of 3.6 mL
to -45 8C at 0.5 8C/min, and held for 90 minutes to re-freeze. The water for injection (WFI). Reconstitution was performed by applying

Table 1
Lyophilization cycle parameters. All shelf temperature ramp rates up and down were 0.5 °C/min except where noted. All temperatures are shelf fluid inlet temperature.

Shelf Ramp Freezing Annealed -6 °C Controlled Nucleation -6 °C

Sample loading 20 °C 5 °C -6 °C
Freezing Ramp 20 °C to 5 °C at 1 °C/min then Ramp 5 °C to -45 °C, hold 90 min Hold -6 °C for 150 min, ice fog controlled
to -45 °C at 0.5 °C/min, hold 90 min nucleation, ramp to -45 C and hold 90 min
Annealing n/a Ramp -45 °C to -6°C, hold 180 min, n/a
ramp -6 to -45 °C, hold 90 min

Drying pressure 50 mTorr


Primary drying Ramp − 45 °C to -25 °C, hold 90 h
Secondary drying Ramp -25 °C to +35 °C at 0.2 °C/min, hold 10 h
Total cycle time (includes 110 h 116 h 110 h
annealing and CN equilibration time)
J. Wang et al. / Journal of Pharmaceutical Sciences 111 (2022) 2639−2644 2641

gentle swirling after the addition of water. Reconstitution time was monomeric species is identified by its characteristic retention time
determined by visual observation of complete dissolution. The recon- relative to a reference standard. Typical method precision was
stitution time was determined with three cake samples and reported approximately 0.1% RSD for IgG monomer.
with an average of the three measurements.

Results and Discussion


Size Exclusion Chromatography (SEC)
The lyophilization cycle run profiles for the three cycles are shown
Separation was performed with YMC-Pack Diol-200, 300 x 8 col- in Fig. 1a-c, and plots of freezing are in Fig. 1d-f. Primary drying
umns, neutral phosphate buffer/NaCl mobile phase, and detection at times, shown in Table 2, were calculated as the time for the Pirani
280 nm sing Agilent HPLC system (Agilent, Santa Clara, CA). The pressure sensor to equilibrate close to the pressure measured by the

Figure 1. Lyophilization parameter profiles: (a-c) entire cycle (green: pirani pressure, red: CM pressure, blue: CM temp, all other color: probe temp); (d-f) freezing; (g) cake resis-
tance. Runs: (a and d) shelf-ramp frozen, no annealing; (b and e) annealed; (c and f) controlled nucleation.
2642 J. Wang et al. / Journal of Pharmaceutical Sciences 111 (2022) 2639−2644

Table 2
Lyophilization and product quality results.

Sample Primary Drying Time (hr) Moisture (%) Recon. Time (sec) Surface Area Multi-point BET (m2/g) HMMS (%) Monomer (%)

Shelf-Ramp Freezing 87 0.2 129 § 11 1.4 1.9 98.1


Annealed -6 °C 62 0.5 71 § 6 0.8 2.0 98.0
Controlled Nucleation -6 °C 61 0.9 49 § 4 0.6 1.9 98.1

capacitance manometer. Primary drying time was significantly The lyophilized cakes from the three cycles each appear slightly
reduced (30% reduction) by CN compared to shelf-ramp freezing different. As seen in Fig. 2, the CN cake appears less elegant due to
without annealing. This result agrees with other reported CN larger pores and some cracks. Microscopic appearance in Fig. 3 con-
studies. For example, Awotwe-Otoo et al compared conventional firms the larger pores of the CN cake, especially at the bottom of the
shelf-ramp freezing to pressurization/ depressurization CN, and cake. The SEM images in Fig. 3 also show that pore sizes at the bottom
primary drying time was reduced by 19%.22 A similar study of the cakes are progressively larger in the order shelf ramp frozen <
reported by Konstantinidis et al achieved a 41% reduction in pri- annealed < CN. Our findings in this regard are not unique. Gieseler
mary drying using CN.23 In comparison to the 30% decrease in reported the cake with CN had a more porous and fragile cracked
primary drying that we observed with CN at -6 °C, we found a structure.25
nearly identical reduction of 29% with simple annealing at -6 °C. SSA of the samples were 1.4 m2/g for shelf-ramp frozen without
Note that residence time of product between -6°C and 0°C annealing, 0.8 m2/g for annealed, and 0.6 m2/g for controlled nucle-
(Fig. 1b), after nucleation was induced, was about 1 h as opposed ation. These results align with the other observations given that the
to 3 h during annealing step. SSA of the dried cake is initially established by the freezing / anneal-
Cake resistance to the flow of escaping water vapor is an impor- ing process (as long as the lyophilization cycle is sufficiently conser-
tant factor used to characterize how ice crystal size impacts product vative to avoid collapse). The larger SSA of the product prepared with
temperature and drying rate during lyophilization.22 Fig. 1g shows CN method agrees with other reports.20, 22 Awotwe-Otoo et al
calculated cake resistance curves for the three cycles used in this reported that CN resulted in cake of a mAB formulation with SSA of
study. The resistance of cake as a function of dry layer was calculated 0.46 m2/g compared to shelf-ramp frozen SSA of 0.90 m2/g.22
from the product temperature profile, using the approach used by As discussed above, others have found that CN could reduce
Tchessalov et al.24 The cake resistance for the controlled nucleation reconstitution time of lyophilized products.19,22 We observed a simi-
and annealed cycles were much lower than the shelf-ramp frozen lar result in which the reconstitution time of the CN cake was
cycle. For parts of the cycle the annealed product had slightly higher reduced to 49 seconds from 129 seconds (Table 2). In contrast, the
resistance than the CN product. Our findings are similar as reported reconstitution time of annealed samples was 71 seconds, slightly lon-
by Gieseler who found that cake resistance after controlled nucle- ger than CN but still faster than the controls. This result agreed with
ation freezing at -3 °C was reduced by about 25% compared to shelf the results reported by others.13,15,16 The shorter reconstitution times
ramp freezing. Annealing at -15 °C for 6 h resulted in cake resistance for the CN and annealed cakes were likely due to the more open net-
similar to CN.25 work of larger pores.15

Figure 2. Cake Appearance: self ramp frozen (A), annealed -6 °C (B), and controlled nucleation (C) cycles.
J. Wang et al. / Journal of Pharmaceutical Sciences 111 (2022) 2639−2644 2643

Figure 3. SEM of lyo cake: Shelf ramp frozen (A), annealed -6C (B), and controlled nucleation (C) cycles. Top images are from the top of the cakes and bottom images are from the
bottom of the cakes.

The residual moisture contents (Table 2) were in order: 0.2% for moisture content trended inversely with these factors, meaning that
shelf-ramp frozen without annealing, 0.5% for annealed, and 0.9% for shorter primary drying would be at least partially offset by additional
CN. Higher moisture for CN has been reported in many secondary drying time and/or higher secondary drying temperature.
publications5,19,22,26 and is consistent with the SSA differences.
Higher SSA for shelf-ramp freezing product is responsible for its
Declaration of Competing Interests
lower moisture content. The higher moisture level in the annealed
cake observed in this study indicated that the -6 °C annealing also
The authors declare that they have no known competing financial
increased the crystal size and decreased the water desorption rate in
interests or personal relationships that could have appeared to influ-
secondary drying, which was similar to what was observed for the
ence the work reported in this paper.
CN cycle.
Protein aggregates can occur during formulation, lyophilization
processes and storage.27 To assess the quality of the lyophilized cake Acknowledgements
produced from shelf-ramp frozen, annealed, and CN cycles, soluble
aggregates of the protein in reconstituted solutions were determined The authors thank Bakul Bhatnagar for his valuable conversations
using the SEC method (Table 2). There was no significant change in in the experiment design and tests. This research did not receive any
high molecular weight species or % monomer for all samples regard- specific grant from funding agencies in the public, commercial, or
less of lyophilization conditions. Fang et al reported that lyophilized not-for-profit sectors.
IgG and rHSA proteins with lower SSA can reduce aggregation28 due
to the reduction of adsorption of protein at the ice/freeze-concentrate
References
interface.29 We did not see a difference in aggregation among the
controlled and non-controlled nucleation methods. This could be due 1. Gangurde JS, Erande KB, Shevale LM. Freeze drying: a review. World J Pharm Res.
to the presence of polysorbate-20 in the formulation, the purpose of 2019;8(3):592–603.
which is to reduce aggregation, as well as the relatively high protein 2. Awotwe-Otoo D, Khan M. Regulatory aspects of freeze-drying. eds Ward KR,
Matejtschuk P, eds. Regulatory aspects of freeze-drying. Lyophilization of Pharma-
concentration of 50 mg/mL. ceuticals and Biologicals. 2019:173–192.
3. Carpenter JF, Chang BS, Garzon-Rodriguez W, Randolph TW. Rational design of sta-
Conclusions ble lyophilized protein formulations: theory and practice. In: Carpenter JF,
Manning MC, eds. Rationale Design of stable protein formulations-theory and prac-
tice. New York: Kluwer Academic/Plenum publishers; 2002:109–133.
This study compared controlled ice nucleation and traditional 4. Tang X (Charlie), Pikal MJ. Design of freeze-drying processes for pharmaceuticals:
annealing processes, in which both were carried out at -6 °C. Primary practical advice. Pharm Res. 2004;21(2):191–200.
5. Siew A. Controlling ice nucleation during the freezing step of lyophilization. Pharm
drying time, cake reconstitution time, specific surface area, crystal
Technol. 2013;37(5):36–40. (Review).
and pore size, SEC and calculated dry layer cake resistance were eval- 6. Hottot A, Vessot S, Andrieu J. A direct characterization method of the ice morphol-
uated. The results show that while “ice-fog” CN at -6 8C was most ogy. Relationship between mean crystals size and primary drying times of freeze-
effective at reducing primary drying time, cake resistance, specific drying processes. Drying Technol. 2004;22(8):2009–2021.
7. Searles JA, Carpenter JF, Randolph TW. Annealing to optimize the primary drying
surface area, and reconstitution time; annealing at -6 8C after shelf- rate, reduce freezing-induced drying rate heterogeneity, and determine T'g in
ramp freezing yielded results that were nearly as good. As expected, pharmaceutical lyophilization. J Pharm Sci. 2001;90(7):872–887.
2644 J. Wang et al. / Journal of Pharmaceutical Sciences 111 (2022) 2639−2644

8. Pikal M J, Shah S, Senior D, Lang JE. Physical chemistry of freeze-drying: measure- 19. Geidobler R, Konrad I, Winter G. Can controlled ice nucleation improve freeze dry-
ment of sublimation rates for frozen aqueous solutions by a microbalance tech- ing of highly-concentrated protein formulations? J Pharm Sci. 2013;102(11):3915–
nique. J Pharm Sci. 1983;72:635–650. 3919.
9. Fonte P, Lino PR, Seabra V, et al. Annealing as a tool for the optimization of lyophili- 20. Singh S, Kumar S, Bondar V, et al. Unexplored benefits of controlled ice nucleation:
zation and ensuring of the stability of protein-loaded PLGA nanoparticles. Int J Lyophilization of a highly concentrated monoclonal antibody solution. Int J Pharm.
Pharm. 2016;503(1−2):163–173. 2018;552:171–179.
10. Pikal MJ, Shah S, Roy ML, Putman R. The secondary drying stage of freeze drying: 21. Carpenter JF, Pikal MJ, et al. Rational design of stable lyophilized protein formula-
drying kinetics as a function of temperature and chamber pressure. Int J Pharm. tions: some practical advice. Pharm Res. 1997;14(8):969–975.
1990;60:203–217. 22. Awotwe-Otoo D, Agarabi C, Read EK, et al. Impact of controlled ice nucleation on
11. Kulkarni SS, Patel SM, Bogner RH. Reconstitution time for highly concentrated process performance and quality attributes of a lyophilized monoclonal antibody.
lyophilized proteins: role of formulation and protein. J Pharm Sci. 2020;109 Int J Pharm. 2013;450(1-2):70–78.
(10):2975–2985. 23. Konstantinidis AK, Kuu WEI, Otten L, Nail SL, Sever RR. Controlled nucleation in
12. Kulkarni SS, Patel SM, Suryanarayanan R, Rinella JV, Bogner RH. Key factors gov- freeze-drying: effects on pore size in the dried product layer, mass transfer resis-
erning the reconstitution time of high concentration lyophilized protein formula- tance, and primary drying rate. J Pharm Sci. 2011;100(8):3453–3470.
tions. Eur J Pharm Biopharm. 2021;165:361–373. 24. Tchessalov S, Latshaw D, Nulu S, Sharp T, Ewan S, Chen X. Application of first prin-
13. Blue J, Yoder H. Successful lyophilization development of protein therapeutics. ciples primary drying model to lyophilization process design and transfer: case
Amer Pharm Rev. 2009;12:90–96. studies from the industry. J Pharm Sci. 2021 Feb;110:968–981.
14. Lin L-F H, Bunnell R. Overcoming challenges in the reconstitution of a high-concen- 25. Gieseler H, Startzel P. Controlled nucleation in freeze-drying. Eur Pharm Rev. 2012.
tration protein drug product. BioPharm Int. 2013;26(3):28–39. Available at: https://www.europeanpharmaceuticalreview.com/article/15427/con
15. Beech KE, Biddlecombe JG, Van Der Walle CF, et al. Insights into the influence of the trolled-nucleation-in-freeze-drying/. Accessed October 22, 2012 .
cooling profile on the reconstitution times of amorphous lyophilized protein for- 26. Khana MA, Shaha RB. Impact of controlled ice nucleation on process performance
mulations. Eur J Pharm Biopharm. 2015;96:247–254. and quality attributes of a lyophilized monoclonal antibody. Int J Pharm.
16. Kulkarni SS, Suryanarayanan R, Rinella JV, Bogner RH. Mechanisms by which crys- 2013;450:70–78.
talline mannitol improves the reconstitution time of high concentration lyophi- 27. Rathore N, Rajan RS. Current perspectives on stability of protein drug formulation
lized protein formulations. Eur J Pharm Biopharm. 2018;131:70–81. fill and finish operations. Biotechnol Prog. 2008;24(3):504–514.
17. Gitter JH, Geidobler R, Presser I, Winter G. A comparison of controlled ice nucle- 28. Fang R, Bogner RH, Nail SL, Pikal MJ. Stability of freeze-dried protein formulations:
ation techniques for freeze-drying of a therapeutic antibody. J Pharm Sci. 2018;107 contributions of ice nucleation temperature and residence time in the freeze-con-
(11):1–7. centrate. J Pharm Sci. 2020;109(6):1896–1904.
18. Luoma J, Ingham E, Martinez CL, Allmendinger A. Comparison of techniques to con- 29. Xu Y, Grobelny P, Von Allmen A, et al. Protein quantity on the air-solid interface
trol ice nucleation during lyophilization. Processes. 2020;8(11):1439. https://doi. determines degradation rates of human growth hormone in lyophilized samples. J
org/10.3390/pr8111439. Pharm Sci. 2014;103(5):1356–1366.

You might also like