6
Enhancing enterovirus A71 vaccine production yield by microcarrier profusion bioreactor culture Chia-Chyi Liu a,, Suh-Chin Wu b , Shang-Rung Wu c , Hsiao-Yu Lin a , Meng-Shin Guo a , Alan Yung-Chih Hu a , Yen-Hung Chow a,d , Jen-Ron Chiang e , Dar-Bin Shieh c , Pele Chong a,d a National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan b Institute of Biotechnology, Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan c Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan d Graduate Institute of Immunology, China Medical University, Taichung, Taiwan e Vaccine Center, Centers for Disease Control, Taipei, Taiwan article info Article history: Available online 6 March 2017 Keywords: Enterovirus A71 Inactivated whole virion vaccine Serum-free bioreactor Microcarrier culture Perfusion technology abstract Hand, foot and mouth diseases (HFMD) are mainly caused by Enterovirus A71 (EV-A71) infections. Clinical trials in Asia conducted with formalin-inactivated EV-A71 vaccine candidates produced from serum-free Vero cell culture using either roller bottle or cell factory technology, are found to be safe and highly efficacious. To increase vaccine yields and reduce the production costs, the bioprocess improvement for EV-A71 vaccine manufacturing is currently being investigated. The parameters that could affect and enhance the production yields of EV-A71 virus growth in the microcarrier bioreactor were investigated. The medium replacement culture strategy included a multi-harvested semi-batch pro- cess and perfusion technology and was found to increase the production yields more than 7–14 folds. Based on the western blot and cryo-EM analyses of the EV-A71 virus particles produced from either the multi-harvested semi-batch (MHSBC) or perfusion cultures were found to be similar to those virus particles obtained from the single batch culture. Mouse immunogenicity studies indicate that the EV-A71 vaccine candidates produced from the perfusion culture have similar potency to those obtained from single batch bioprocess. The physical structures of the EV-A71 particles revealed by the cryo-EM analysis were found to be spherical capsid particles. These results provide feasible technical bioprocesses for increasing virus yields and the scale up of EV-A71 vaccine manufacturing using the bioreactor cell culture methods. Ó 2017 Elsevier Ltd. All rights reserved. 1. Introduction Enterovirus A71 (EV-A71) is a non-enveloped single (+) strand RNA virus of the family Picornaviridae, genus Enterovirus. EV-A71 viral infections have caused several epidemics of hand, foot and mouth disease (HFMD) that were associated with severe neurolog- ical diseases in young children. It has become a serious public health problem in the Asia-Pacific region [1–6]. To prevent EV-A71 infections and periodic EV-A71 outbreaks, an effective vaccine against EV-A71 infection is urgently needed. Several EV-A71 vaccine candidates have been investigated including the live-attenuated virus, inactivated whole virus, recombinant viral protein, virus-like particle, recombinant plant, VP1 peptide and DNA vaccines [4]. Most of these EV-A71 vaccine candidates could induce various level of virus neutralizing antibody responses in the mouse immunogenicity studies, but due to the success of polio vaccine and easy in regulatory issues, the chemical inactivated whole-virion EV-A71 vaccine candidates were first selected for clinical development [3,4,7]. The chemical inactivated EV-A71 vaccine candidates based on EV-A71 grown in the Vero cell culture using either roller bottle technology or cell factory technology have been tested in human clinical trials in Taiwan, mainland China and Singapore [8–14]. Serum-free media were used in the cell-culture to overcome the high serum-protein content that could complicate the downstream purification process and the risk of potential contamination with prions due to the bovine resource [15]. The whole-virion EV-A71 vaccine candidate either in empty virus particle (E-particle) or fully infectious virus particle (F-particle) could be either co-purified http://dx.doi.org/10.1016/j.vaccine.2017.02.042 0264-410X/Ó 2017 Elsevier Ltd. All rights reserved. Corresponding author at: Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan. E-mail address: [email protected] (C.-C. Liu). Vaccine 36 (2018) 3134–3139 Contents lists available at ScienceDirect Vaccine journal homepage: www.elsevier.com/locate/vaccine

Enhancing enterovirus A71 vaccine production yield by ...can.ncku.edu.tw/upload/2019/01/20190116144041aCl.pdfEnhancing enterovirus A71 vaccine production yield by microcarrier profusion

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Enhancing enterovirus A71 vaccine production yield by ...can.ncku.edu.tw/upload/2019/01/20190116144041aCl.pdfEnhancing enterovirus A71 vaccine production yield by microcarrier profusion

Vaccine 36 (2018) 3134–3139

Contents lists available at ScienceDirect

Vaccine

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

Enhancing enterovirus A71 vaccine production yield by microcarrierprofusion bioreactor culture

http://dx.doi.org/10.1016/j.vaccine.2017.02.0420264-410X/� 2017 Elsevier Ltd. All rights reserved.

⇑ Corresponding author at: Vaccine R&D Center, National Institute of InfectiousDiseases and Vaccinology, National Health Research Institutes, Zhunan Town,Miaoli County 350, Taiwan.

E-mail address: [email protected] (C.-C. Liu).

Chia-Chyi Liu a,⇑, Suh-Chin Wu b, Shang-Rung Wu c, Hsiao-Yu Lin a, Meng-Shin Guo a, Alan Yung-Chih Hu a,Yen-Hung Chow a,d, Jen-Ron Chiang e, Dar-Bin Shieh c, Pele Chong a,d

aNational Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwanb Institute of Biotechnology, Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwanc Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, TaiwandGraduate Institute of Immunology, China Medical University, Taichung, TaiwaneVaccine Center, Centers for Disease Control, Taipei, Taiwan

a r t i c l e i n f o a b s t r a c t

Article history:Available online 6 March 2017

Keywords:Enterovirus A71Inactivated whole virion vaccineSerum-free bioreactorMicrocarrier culturePerfusion technology

Hand, foot and mouth diseases (HFMD) are mainly caused by Enterovirus A71 (EV-A71) infections.Clinical trials in Asia conducted with formalin-inactivated EV-A71 vaccine candidates produced fromserum-free Vero cell culture using either roller bottle or cell factory technology, are found to be safeand highly efficacious. To increase vaccine yields and reduce the production costs, the bioprocessimprovement for EV-A71 vaccine manufacturing is currently being investigated. The parameters thatcould affect and enhance the production yields of EV-A71 virus growth in the microcarrier bioreactorwere investigated. The medium replacement culture strategy included a multi-harvested semi-batch pro-cess and perfusion technology and was found to increase the production yields more than 7–14 folds.Based on the western blot and cryo-EM analyses of the EV-A71 virus particles produced from eitherthe multi-harvested semi-batch (MHSBC) or perfusion cultures were found to be similar to those virusparticles obtained from the single batch culture. Mouse immunogenicity studies indicate that theEV-A71 vaccine candidates produced from the perfusion culture have similar potency to those obtainedfrom single batch bioprocess. The physical structures of the EV-A71 particles revealed by the cryo-EManalysis were found to be spherical capsid particles. These results provide feasible technical bioprocessesfor increasing virus yields and the scale up of EV-A71 vaccine manufacturing using the bioreactor cellculture methods.

� 2017 Elsevier Ltd. All rights reserved.

1. Introduction

Enterovirus A71 (EV-A71) is a non-enveloped single (+) strandRNA virus of the family Picornaviridae, genus Enterovirus. EV-A71viral infections have caused several epidemics of hand, foot andmouth disease (HFMD) that were associated with severe neurolog-ical diseases in young children. It has become a serious publichealth problem in the Asia-Pacific region [1–6]. To preventEV-A71 infections and periodic EV-A71 outbreaks, an effectivevaccine against EV-A71 infection is urgently needed. SeveralEV-A71 vaccine candidates have been investigated including thelive-attenuated virus, inactivated whole virus, recombinant viral

protein, virus-like particle, recombinant plant, VP1 peptide andDNA vaccines [4]. Most of these EV-A71 vaccine candidates couldinduce various level of virus neutralizing antibody responses inthe mouse immunogenicity studies, but due to the success of poliovaccine and easy in regulatory issues, the chemical inactivatedwhole-virion EV-A71 vaccine candidates were first selected forclinical development [3,4,7].

The chemical inactivated EV-A71 vaccine candidates based onEV-A71 grown in the Vero cell culture using either roller bottletechnology or cell factory technology have been tested in humanclinical trials in Taiwan, mainland China and Singapore [8–14].Serum-free media were used in the cell-culture to overcome thehigh serum-protein content that could complicate the downstreampurification process and the risk of potential contamination withprions due to the bovine resource [15]. The whole-virion EV-A71vaccine candidate either in empty virus particle (E-particle) or fullyinfectious virus particle (F-particle) could be either co-purified

Page 2: Enhancing enterovirus A71 vaccine production yield by ...can.ncku.edu.tw/upload/2019/01/20190116144041aCl.pdfEnhancing enterovirus A71 vaccine production yield by microcarrier profusion

C.-C. Liu et al. / Vaccine 36 (2018) 3134–3139 3135

using the gel-filtration chromatography [8,9,13] or individuallypurified by sucrose gradient ultracentrifugation [14]. The ratio ofthe E-particle to the F-particle was 7:3 in the single batch culture[14]. To develop the large-scale manufacturing process forEV-A71 vaccine production, cells grown on the surface of a 5 g/LCytodex 1 microcarrier and cultured in a serum-free bioreactortechnology have been investigated and established [14,16,17].Recently, a 200L-scale serum-free microcarrier bioreactor was suc-cessfully established to produce an EV-A71 vaccine candidateusing a single-use bioreactor [18]. Their results show that thetwo types of EV-A71 viral particles were purified from the micro-carrier bioreactor culture that could induce good immunogenicityagainst the EV-A71 virus.

Several parameters for viral vaccine production have beeninvestigated that include cell lines, virus strains, culture media,bioreactor technologies and cultivation strategies [19]. In our pre-vious studies, we used the following two-temperature culturemethod to increase the EV-A71 virus titer: the cell growth phasewas at 37 �C and 32 �C for the infection phase [14,16,17]. TheEV-A71 virus was released at higher titers in the supernatant thanintra-cellular using microcarrier culture [16]. The glucose/glutamine feedings strategy could improve EV-A71 productionyields during infection in batch culture [16], and this method hasalso been applied in a 200 L-scale bioreactor [18]. Either rollerbottle technology or microcarrier bioreactor technology for200 L-scale cGMP-grade EV-A71 vaccine production requires over2 months to amplify Vero cells. To increase vaccine yields, the con-ventional batch production system is modified with the fed-batchand perfusion methods for cell culture based manufacturing bio-processes [19]. The perfusion method utilize feeding the freshmedium, while withdrawing the old culture medium to achievehigher cell concentrations and extended the bioreactor run times.The development of technically feasible large-scale manufacturingmethods to enhance EV-A71 virus production yields could alsoreduce the production costs.

In this study, we present a medium replacement culture strat-egy as a good approach to improve EV-A71 virus production yieldsin the serum-free microcarrier bioreactor. Both the multi-harvested semi-bath culture (MHSBC) and perfusion culture tech-nology in microcarrier culture were tested, and it was found thatthese technologies could enhance the production of the EV-A71virus. This slow cell-death indicates that the semi-bath culturestrategy and perfusion culture technology could be developed toenhance production yields in the scaled-up manufacturingprocesses. We also examined the immunogenicity of the EV-A71antigens in mouse model. The results from the current studieswould provide valuable information for developing large-scaleserum-free microcarrier cell culture for manufacturing thechemical inactivated EV-A71 vaccine.

2. Materials and methods

2.1. Cells, culture medium, and virus

Vero cells (green monkey kidney cells) were kindly providedby the Taiwan Centers of Disease Control (Taiwan CDC), whichwere obtained from the original Vero cell line from the AmericanType Culture Collection (ATCC). Vero cells were grown in aVP-SFM medium (Gibco) and were passaged twice weekly inT-flasks. The EV-A71 E59 strain (genotype B4), the clinical isolateof the EV-A71 virus, was obtained from the Taiwan CDC [13].EV-A71 viral stocks were harvested from the supernatants ofinfected Vero cells three days post infection (DPI). The titers ofthe virus stocks were determined by a TCID50 assay, and thestocks were stored at �80 �C.

2.2. EV-A71 virus cultivation using a bioreactor system

The EV-A71 virus was cultivated using serum-free VP-SFMmedium in a BIOFLO 310 bioreactor (Eppendorf AG) based on thetwo-temperature microcarrier cell culture bioprocess as previouslyreported [14,16,17]. The bioreactor cultures (5 L of working vol-umes) were stirred at a rate of 60 rpm at pH 7, and the dissolvedoxygen (DO) content was controlled at 50% by a gas mixer appara-tus. The Vero cells used in the bioreactor were first harvested fromthe roller bottle and were detached using TrypLE select. Bioreactorculture medium containing 5 g of Cytodex 1 per liter was inocu-lated with 2 � 105 cells per mL, and the cell density would reachto 2–2.5 � 106 cells per mL after six days of cultivation. After a70% culture medium replacement, the Vero cells were infectedwith the EV-A71 virus at a multiplicity of infection (MOI) = 10�5

[13,14]. EV-A71 virus stocks were harvested from the culturesupernatants at 6–13 days post infection. The multi-harvestedsemi-batch culture method was defined as EV-A71 virus and washarvested by replacing 70% of the culture medium every two daysand collected from the 6th, 8th, 10th and 12th DPI. A perfusion cul-ture method was used for harvesting and collecting EV-A71 virusby replacing 50% of the medium every day from 7th to 13th DPI.

2.3. EV-A71 virus purification by continuous sucrose gradientultracentrifugation

The upstream harvesting of the EV-A71 viral stocks was per-formed as previously described [14], the debris of the Vero cellswas removed by a 0.65 mm filter (Sartorius), and viral stocks wereconcentrated using a 100 K TFF capsule (Pall). The concentratedviral stocks were loaded to a 10–50% continuous sucrose gradientfor ultracentrifugation at 32,000 rpm for 3 h using a zonal rotorin a Hitachi CP80 ultracentrifuge. These resultant fractions weresubjected to SDS-PAGE and western analyses to detect purifiedEV-A71 viruses. The purified EV-A71 viral stocks were diafiltratedusing an Amicon 100 K tube (Millipore) by centrifuging at 3000g.The concentrated stock was re-suspended in PBS. The total proteinconcentration of the re-suspended viral stock was determinedusing a BCA protein assay (Pierce).

2.4. Determination of virus titers and specific titers

Virus titers were determined from the median end point of thetissue culture’s infectious dose (TCID50) according to the protocoldescribed previously by Chang et al. [13]. The TCID50 values werecalculated using the Reed-Muench methods. The specific titerswere measured and defined as the total harvested virus yields afterpost-infection/total initially infected cell numbers produced undervarious experimental conditions.

2.5. SDS-PAGE analysis and western blotting

SDS-PAGE and western blot analyses of the purified EV-A71virus fractions were performed in a NuPAGE 4–12% Bis-Tris Gel(Invitrogen) according to a previously described protocol [14]. AnMAB979 monoclonal antibody (Millipore) was used to detectEV-A71 antigens. Novex� Sharp Pre-stained Protein Standards(ThermoFisher, LC5800) and PiNK Plus Prestained Protein Ladder(GeneDireX, PM005) protein markers were used in westernblotting.

2.6. Characterization of viral particles by cryo-electron microscopy(cryo-EM)

The purified viral stocks were inactivated by 0.025% formalde-hyde solution [14] and were adsorbed to grids coated with a holey

Page 3: Enhancing enterovirus A71 vaccine production yield by ...can.ncku.edu.tw/upload/2019/01/20190116144041aCl.pdfEnhancing enterovirus A71 vaccine production yield by microcarrier profusion

3136 C.-C. Liu et al. / Vaccine 36 (2018) 3134–3139

carbon film, blotted, and plunge-frozen in ethane using a GatanCp3 Cryo Plunger. The frozen grids were observed and recordedwith a JEM-2100F transmission electron microscope with an accel-erating voltage of 200 kV. The micrographs were recorded on a4 k � 4 k CCD camera (Gatan Ultrascan 4000 4 k � 4 k CCD CameraSystemModel 895). Viral particles were found to have diameters ofapproximately 30–35 nm.

2.7. Animal immunogenicity study

The animal protocol (NHRI-IACUC-101042-A) was reviewed andapproved by the Institutional Animal Care and Use Committee,NHRI. Mouse immunogenicity studies were performed accordingto the protocols described previously [9,14]. Virus stocks weremixed with a 37% formaldehyde solution (Merck) at a of 4,000:1volume ratio and stored for 3 days at 37 �C for virus inactivation.The aluminumphosphate (alum)was produced in NHRI cGMP facil-ity [11]. Three groups of six female BALB/c mice (6–8 weeks old)were intraperitoneally (i.p.) immunized with 0.5 mL of PBS/alum(300 mg) as the control, the viral empty particle (alum-absorbed0.25 mg antigen per dose), or the viral full particle (alum-absorbed0.25 mg antigen per dose). Mice were boosted twice with the samedose at two week intervals after priming. The immunized micewere bled one week after the final boost, and the serum was col-lected and used to analyze virus neutralization.

2.8. Virus neutralizing assay

The sera collected from immunized mice were inactivated at56 �C for 30 min. A virus neutralization assay was performedaccording to Liu et al. [14]. The TCID50 values were obtained bycounting the cytopathic effects (CPE) in the infected Vero cells.The 50% neutralization inhibition dose that is the geometric recip-rocal of the serum dilution yielding a 50% reduction in the viraltiter was obtained using the Reed-Muench methods.

3. Results

3.1. EV-A71 virus culture in a bioreactor by perfusion culture methods

The preliminary MHSBC experiments were performed in100-mL spinner culture system (Supplementary S1). To develop a

Fig. 1. EV-A71 E59 virus production in a microcarrier bioreactor culture. Vero cells cultustrain at MOI = 10�5. (j) In batch culture, there was no replacement. (h) In the MHSBC, 70DPI. (N) In perfusion culture, 50% of the culture medium was replaced every day on 7th toaccumulated in different culture conditions.

feasible technical bioprocess for EV-A71 virus production, a 5-Lworking volume bioreactor was investigated. As the control, thesingle-batch method was performed in Vero cells grown on 5 g/LCytodex 1 microcarriers reaching 2–2.5 � 106 cells/mL, 70% ofthe culture medium was replaced with a fresh VP-SFM mediumand the cells were infected with the EV-A71 virus at an MOI = 10�5

in the bioreactor at 32 �C. Similar to the previous report [14], theviral titer was consistently found to be 0.7–1 � 106 TCID50/mL ateither the 6th or 8th DPI harvested virus culture (Fig. 1A). In theMHSBC the medium was replaced at the 6th, 8th and 10th DPI,and the average viral titers from 3 different runs were found tobe 13, 7, 4 and 0.7 � 106 TCID50/mL in the 6th, 8th, 10th and12th DPI harvested virus culture media, respectively (Fig. 1A).From 3 different perfusion studies in which 50% of the culturemedium was replaced every day after infection, the average viraltiters found in the harvested virus culture were ranging from 1to 2 � 107 TCID50/mL for the 7th to 13th DPI (Fig. 1A). The averageharvested virus titers were calculated to be 2 � 106 TCID50/mL inthe 4.5 L of a single batch culture, 5.8 � 106 TCID50/mL in a total15 L of a semi-batch culture and 1.0 � 107 TCID50/mL in a total18 L of perfusion culture. Both the MHSBC and the perfusion cul-ture methods could significantly increase the virus productionyields by 7–14-fold in comparison to those obtained from thesingle-batch culture method. The current perfusion technologycertainly has more capability than the MHSBC culture method asmeasured by the specific titer (Fig. 1B). In addition, the currentresults indicate that the medium replacement strategy couldprovide savings in time and the cost of raw materials, such asmicrocarrier beads and a Vero cell bank seed.

To understand how the fresh culture media support virus pro-duction, the cell morphology and the CPE of the virus-infected Verocells were examined after EV-A71 infection. The CPE was consis-tently observed starting at the 5th to 6th DPI and essentially allVero cells were dissociated from microcarriers on the 8th DPI inthe single-batch studies. In MHSBC studies, the CPE was alsoobserved on the 6th DPI, but the Vero cells were slowly and grad-ually dissociated from microcarriers until the 12th DPI. In perfu-sion studies, CPE was also observed and the Vero cells weregradually dissociated from microcarriers during the 7th to 13thDPI. Taking the observation of CPE and the results of virus titersin the harvested virus culture together, it seems that EV-A71 virusreplications maintain a stable rate as long as the cells still attach to

red on 5 g/L microcarrier in 5 L working volumes and infected with the EV-A71 E59% of the culture mediumwas replaced every two days on the 6th, 8th, 10th and 12th13th DPI. (B) Specific titers of the EV-A71 E59 virus production. Specific titers were

Page 4: Enhancing enterovirus A71 vaccine production yield by ...can.ncku.edu.tw/upload/2019/01/20190116144041aCl.pdfEnhancing enterovirus A71 vaccine production yield by microcarrier profusion

C.-C. Liu et al. / Vaccine 36 (2018) 3134–3139 3137

the microcarrier beads. Additionally, the fresh culture medium cer-tainly helps the cells continue to grow as the toxic materialsderived from the dead cells are removed from the media. TheEV-A71 virus is stable at 32 �C for a few days that provide enoughtimes to operate MHSBC and perfusion culture methods.

3.2. Biochemical characterizations of virus collected from the MHSBCand perfusion culture media

We investigated whether the MHSBC and/or perfusion cell cul-ture methods could produce EV-A71 virus particles that are similarto previous studies. The results described above indicate that con-sistent EV-A71 virus titers were produced from both the MHSBCand perfusion methods during the 6th to 13th DPI. Harvested virusculture from four time points (6th, 8th, 10th and 12th DPI) wasindividually concentrated and separated using a 10–50% continu-ous sucrose gradient ultracentrifugation into two types of EV-A71 virus particles as detected by the western blotting analysis(Fig. 2A). Using MAB979 VP2-specific monoclonal antibody, at all4 time points, the EV-A71 E-particle characterized with a VP0 viralprotein (36 kDa) was observed in Fractions #9 to #11, and the EV-A71 F-particle characterized with a VP2 protein (28 kDa) wasrevealed in Fractions #15 to #17. These results are identical tothose observed from the single-batch bioreactor study reportedpreviously [14]. A similar separation pattern of the virus particleswas also detected by western blotting analysis of the 13th DPI har-vested virus culture of the perfusion bioreactor run (Fig. 2B). Thecurrent results certainly confirm that the MHSBC and/or perfusionmethods could produce two EV-A71 virus particles that are similarto the virus particles obtained from the single-batch culture or

Fig. 2. Purification of the EV-A71 virus from the bioreactor culture by sucrose gradientsupernatants (6th, 8th, 10th and 12th DPI) were concentrated and separated into 25 fracfinal perfusion culture harvesting supernatant (13th DPI) was concentrated and separatEV-A71 antigens were detected by western blot using MAB979 VP2-specific monoclonal a(N), and the VP2 protein was labeled at the expected molecular weight (4).

roller-bottle technology. We collected viral particles and evaluatedthe protein concentration from Fractions #9 to #11 (E-particle) andFractions #15 to #17 (F-particle). The ratio of the two particles(E-particle/F-particle) was found to be 2.5 in the single-batch cul-ture method, 1.7 in the MHSBC method and 1.4 in the perfusionmethod. In the roller-bottle technology, the ratio was found to be2.3 in the single-batch production run. These results indicated boththe MHSBC and perfusion methods could enhance more infectiousvirus yields from Vero cell.

3.3. Characterization of viral particles by cryo-EM

To have good preservation of structural features, the purifiedEV-A71 viral particles were individually inactivated with formalin(4000:1), suspended in vitreous ice and analyzed by cryo-EM(Fig. 3). The intact and spherical particles of the EV-A71 virusesare shown with a diameter of 30–35 nm in the cryo-EM micro-graphs, and their morphology was found to closely resemble thoseof the enteroviruses of the Picornaviridae family.

3.4. Immunogenicity study of formalin-inactivated EV-A71 virionderived from perfusion bioreactor

To determine the immunogenicity of the EV-A71 virionobtained from the perfusion technology using the 5-L bioreactor,the sucrose gradient ultracentrifugation purified virus stocks weretreated with formaldehyde solution for virus inactivation asdescribed by Liu et al. [14]. Mice were immunized with alum-absorbed formalin-inactivated EV-A71 virions, and the virus neu-tralization titers obtained a titer of 1280 using the E-particle

zonal ultracentrifugation. (A) Four multi-harvested semi-batch culture harvestingtions. Marker: Novex� Sharp Pre-stained Protein Standards (ThermoFisher). (B) Theed into 25 fractions. Marker: PiNK Plus Prestained Protein Ladder (GeneDireX). Thentibody. The VP0 protein (VP4 + VP2) was labeled at the expected molecular weight

Page 5: Enhancing enterovirus A71 vaccine production yield by ...can.ncku.edu.tw/upload/2019/01/20190116144041aCl.pdfEnhancing enterovirus A71 vaccine production yield by microcarrier profusion

Fig. 3. Cryo-EM micrographs of the formalin-inactivated viral particles from the perfusion culture. (A) EV-A71 empty-particle at fraction 10. (B) EV-A71 full-particle atfraction 16.

3138 C.-C. Liu et al. / Vaccine 36 (2018) 3134–3139

immunization and 2560 using the F-particle immunization(Table 1). The PBS/alum control group had <8. The current resultsindicate that the inactivated EV-A71 virions prepared from the per-fusion bioreactor culture technology can induce virus neutralizingtiters that are comparable to those obtained from the batch biore-actor culture [9,14].

4. Discussion

The current EV-A71 vaccine candidates are produced from avirus grown in Vero cell using either the roller bottle or cell factorytechnologies as they are easy to implement and operate, and com-panies can establish the cGMP infrastructure and have a flexiblefacility for multiple applications [8]. However, these technologiesare labor intensive and need a bigger facility to implement moreroller bottles or cell factories in large-scale vaccine manufacturing.The microcarrier bioreactor technology can provide large-scalemanufacturing in fixed or disposable bioreactors. The purpose ofthe current work is to develop a medium replacement culturestrategy to increase EV-A71 virus production yields. This concepthas been used in large-scale cell culture for viral vaccines, biophar-maceuticals and recombinant protein production [19–23].

The stirred tank bioreactors can be run in the following differ-ent modes: batch, fed-batch and perfusion culture [19,24]. Theadvantage of perfusion cultures is that cells are retained in thebioreactor, and the product is continuously harvested. Therefore,a smaller bioreactor can achieve higher productivities comparedto those obtained in batch or fed-batch cultures. In addition, theseculture methods are very easy to perform and do not need more

Table 1The EV-A71 virus neutralization titers of mouse antisera generated against varioustypes of EV-A71 E59 viral particle fractions using TCID50 neutralization assay.

Process Sample Totalprotein(mg)

The virusneutralizationtiter(GMT ± SE)

Perfusion bioreactorPBS/alum control 0 <8E-particle 2.5 1280 ± 388F-particle 2.5 2560 ± 675

Batch bioreactora E-particle 2.5 906 ± 351

F-particle 2.5 2020 ± 799

a This data is derived from our previous article [14]. To be consistent, the samestarting materials (cell bank, virus seed, media and bioreactor) to culture andproduce the EV-A71 virus particles were used.

expensive investments for the development of manufacturing pro-cesses. Perfusion culture has been successfully applied to improverabies virus production [25,26] and flu virus production [23,27].Cell culture based flu vaccine products are currently manufacturedusing perfusion culture system has recently been reviewed [23].Microcarrier beads are designed for anchor-dependent cell linesin a stirred tank in which the culture parameters are easily moni-tored and controlled for cell growth. The microcarrier beads have adensity of 1.04 g/mL and a mean diameter of 200 mm. These char-acteristics of the microcarrier are easy to keep in the bioreactorwhen the medium is replaced by gravity or a filter. In the currentMHSBC, as the stirrer shuts down for 5–15 min, most microcarrierbeads were settled in the bottom of the bioreactor. It was easy toharvest the virus supernatant and replace 70% of the fresh medium.It is worthwhile to note in the perfusion culture that a 70 mm meshfilter is set in the harvesting channel to enhance the virus produc-tion yield as the filter could prevent the cell-attached microcarriersfrom flowing through and being lost. However, there are alsopotential disadvantages regarding additional media costs, productstability and operational risks during the extended cultivationperiod.

In this work, we discovered that the infectious EV-A71 viruswas consistently replicating when the fresh culture medium wasreplaced. Therefore, the medium replacement culture strategyworks well for increasing EV-A71 virus production yields. After6–7 DPI at the low MOI (10�5), the rate of CPE was slowed whena fresh medium replacement was performed. The MHSBC methodcould significantly increase the harvested virus production yieldsfor EV-A71 in comparison with the single-batch culture method.We observed a 7–14 folds increase in the EV-A71 virus productionyield in the perfusion culture technology. Two types of EV-A71viral particles are produced in the cell culture production biopro-cess. The F-particle ratio was increased in the medium replacementculture strategy. This method provides a feasible technical biopro-cess for producing more F-particles.

The biochemical and biophysical analyses demonstrated thatthe MHSBC and perfusion culture methods produced a comparableEV-A71 particle pattern to the batch culture method(Fig. 2A and B). In cryo-EM observation, the inactivated EV-A71virions prepared from the bioreactor culture technology were stillmaintaining a complete smooth particle structure (Fig. 3). EV-A71structure proteins are assembled with viral RNAs to formF-particle, and E-particle is packed without viral RNAs [14]. Inour previous studies, we detected a large amount of viral RNAs atF-particle fractions using real-time PCR [14]. Furthermore, a mouseimmunogenicity study of the EV-A71 particles produced from a

Page 6: Enhancing enterovirus A71 vaccine production yield by ...can.ncku.edu.tw/upload/2019/01/20190116144041aCl.pdfEnhancing enterovirus A71 vaccine production yield by microcarrier profusion

C.-C. Liu et al. / Vaccine 36 (2018) 3134–3139 3139

perfusion bioreactor elicited the virus neutralizing titers that arecomparable to those obtained from the batch bioreactor culture.These results showed that the medium replacement culture strat-egy did not change the basic biochemical properties, physicalstructures or immunogenicity of the EV-A71 particles.

5. Conclusion

The current results certainly confirm the success of theperfusion strategy and indicate that there are savings in time, thecost of the microcarriers and the usage of Vero cells. Therefore,the present study provides valuable information for the large-scale bioreactor manufacturing of the FI-EV-A71 vaccine.

Acknowledgements

This work was supported by the Ministry of Health andWelfare,the Ministry of Science and Technology (NSC 99A1-VCSP01-014),the Taiwan CDC and the National Health Research Institutes(02A1-IVPP12-014), Taiwan.

Appendix A. Supplementary material

Supplementary data associated with this article can be found, inthe online version, at http://dx.doi.org/10.1016/j.vaccine.2017.02.042.

References

[1] Ho M, Chen ER, Hsu KH, Twu SJ, Chen KT, Tsai SF, et al. An epidemic ofenterovirus 71 infection in Taiwan. N Engl J Med 1999;341:929–35.

[2] McMinn PC. An overview of the evolution of enterovirus 71 and its clinical andpublic health significance. FEMS Microbiol Rev 2002;26:91–107.

[3] Xu J, Qian Y, Wang S, Serrano JMG, Li W, Huang Z, et al. EV71: an emerginginfectious disease vaccine target in the Far East? Vaccine 2010;28:3516–21.

[4] Lee MS, Chang LY. Development of enterovirus 71 vaccines. Expert Rev Vacc2010;9:149–56.

[5] Ooi MH, Wong SC, Lewthwaite P, Cardosa MJ, Solomon T. Clinical features,diagnosis, and management of enterovirus 71. Lancet Neurol2010;9:1097–105.

[6] McMinn PC. Recent advances in the molecular epidemiology and control ofhuman enterovirus 71 infection. Curr Opin Virol 2012;2:199–205.

[7] Chou AH, Liu CC, Chang JY, Lien SP, Guo MS, Tasi HP, et al. Immunologicalevaluation and comparison of different EV71 vaccine candidates. Clin DevImmunol 2012;2012:831282.

[8] Chong P, Liu CC, Chow YH, Chou AH, Klein M. Review of enterovirus 71vaccines. Clin Infect Dis 2015;60:797–803.

[9] Chou AH, Liu CC, Chang CP, Guo MS, Hsieh SY, Yang WH, et al. Pilot scaleproduction of highly efficacious and stable enterovirus 71 vaccine candidates.PLoS ONE 2012;7:e34834.

[10] Li YP, Liang ZL, Gao Q, Huang LR, Mao QY, Wen SQ, et al. Safety andimmunogenicity of a novel human Enterovirus 71 (EV71) vaccine: arandomized, placebo-controlled, double-blind, Phase I clinical trial. Vaccine2012;30:3295–303.

[11] Cheng A, Fung CP, Liu CC, Lin YT, Tsai HY, Chang SC, et al. A Phase I,randomized, open-label study to evaluate the safety and immunogenicity of anenterovirus 71 vaccine. Vaccine 2013;31:2471–6.

[12] Zhu FC, Meng FY, Li JX, Li XL, Mao QY, Tao H, et al. Efficacy, safety, andimmunology of an inactivated alum-adjuvant enterovirus 71 vaccine inchildren in China: a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2013;381:2024–32.

[13] Chang JY, Chang CP, Tsai HH, Lee CD, Lian WC, Su IJ, et al. Selection andcharacterization of vaccine strain for Enterovirus 71 vaccine development.Vaccine 2012;30:703–11.

[14] Liu CC, Guo MS, Lin FH, Hsiao KN, Chang KH, Chou AH, et al. Purification andcharacterization of enterovirus 71 viral particles produced from Vero cellsgrown in a serum-free microcarrier bioreactor system. PLoS ONE 2011;6:e20005.

[15] Merten OW. Safety issues of animal products used in serum-free media. DevBiol Stand 1999;99:167–80.

[16] Wu SC, Liu CC, Lian WC. Optimization of microcarrier cell culture process forthe inactivated enterovirus type 71 vaccine development. Vaccine2004;22:3858–64.

[17] Liu CC, Lian WC, Butler M, Wu SC. High immunogenic enterovirus 71 strainand its production using serum-free microcarrier Vero cell culture. Vaccine2007;25:19–24.

[18] Wu CY, Lin YW, Kuo CH, Liu WH, Tai HF, Pan CH, et al. Inactivated Enterovirus71 vaccine produced by 200-L scale serum-free microcarrier bioreactor systemprovides cross-protective efficacy in human SCARB2 transgenic mouse. PLoSONE 2015;10:e0136420.

[19] Gallo-Ramírez LE, Nikolay A, Genzel Y, Reichl U. Bioreactor concepts for cellculture-based viral vaccine production. Expert Rev Vacc 2015;14:1181–95.

[20] Voisard D, Meuwly F, Ruffieux PA, Baer G, Kadouri A. Potential of cell retentiontechniques for large-scale high-density perfusion culture of suspendedmammalian cells. Biotechnol Bioeng 2003;82:751–65.

[21] Bleckwenn NA, Golding H, Bentley WE, Shiloach J. Production of recombinantproteins by vaccinia virus in a microcarrier based mammalian cell perfusionbioreactor. Biotechnol Bioeng 2005;90:663–74.

[22] Warnock JN, Al-Rubeai M. Bioreactor systems for the production ofbiopharmaceuticals from animal cells. Biotechnol Appl Biochem2006;45:1–12.

[23] Genzel Y, Reichl U. Continuous cell lines as a production system for influenzavaccines. Expert Rev Vacc 2009;8:1681–92.

[24] Shuler ML, Kargi F. Bioprocess engineering: basic concepts. 2th ed. PrenticeHall; 2001.

[25] Trabelsi K, Rourou S, Loukil H, Majoul S, Kallel H. Optimization of virus yield asa strategy to improve rabies vaccine production by Vero cells in a bioreactor. JBiotechnol 2006;121:261–71.

[26] Rourou S, van der Ark A, van der Velden T, Kallel H. A microcarrier cell cultureprocess for propagating rabies virus in Vero cells grown in a stirred bioreactorunder fully animal component free conditions. Vaccine 2007;25:3879–89.

[27] Petiot E, Jacob D, Lanthier S, Lohr V, Ansorge S, Kamen AA. Metabolic andkinetic analyses of influenza production in perfusion HEK293 cell culture. BMCBiotechnol 2011;11:84.