4,314
Views
55
CrossRef citations to date
0
Altmetric
Review

Cell culture-based influenza vaccines: A necessary and indispensable investment for the future

Pages 1223-1234 | Received 21 Oct 2014, Accepted 16 Jan 2015, Published online: 27 May 2015

Abstract

The traditional platform of using embryonated chicken eggs for the production of influenza vaccines has several drawbacks including the inability to meet the volume of required doses in the case of widespread epidemics and pandemics. Cell culture platforms have therefore been explored in the last 2 decades, and have attracted further attention following the H1N1 pandemic outbreak. This platform, while not the most economical for large-scale production, has several advantages, and can supplement the vaccine requirement when needed. Recent developments in production technologies have contributed greatly to fine-tuning this platform. In combination with other technologies such as live attenuated and recombinant protein or virus-like particle vaccines, and different adjuvants and delivery systems, cell culture-based influenza vaccine platform can be used both for production of seasonal vaccine, and to mitigate vaccine shortages in pandemic situations.

Introduction

The importance of influenza and its prevention

Vaccines have saved billions of lives over the last century, to the extent that non-communicable diseases have more impact on human health in many countries nowadays.Citation1 However, influenza still occupies an important place in our preventive healthcare programs. This is mainly because (a) influenza is associated with high attack rates and explosive spread, (b) it is most debilitating to children and the elderly, thus incurring high healthcare costs, (c) can afflict a large proportion of the working class, or affect them indirectly, thus reducing productivity, (d) the virus strains change every year, making it difficult to prepare and stockpile the vaccine in advance, (e) cross-protection engendered by infection or vaccination is incomplete, and (f) it is nearly impossible to predict a widespread outbreak. Influenza, along with secondary complications, is possibly the most important infectious cause of human morbidity and mortality.

Based on host tropism and severity of disease, influenza viruses are classified into types A, B, and C. Of these, type A, and to a lesser extent type B, are clinically important to humans. They are further classified based on the subtype of one of the 2 viral surface glycoproteins, the hemagglutinin (H) and the neuraminidase (N). Eighteen H and 9 N subtypes have been described so far. Although the H and N types can theoretically combine randomly, only a limited combination of H1, H2, H3, H5, H7 or H9, and N1 or N2 have been typically observed in humans. Influenza viruses are named by the virus type, geographic location from where the virus was first isolated, the isolate or strain number, the year of isolation, and the H and N subtype. For example, A/California/04/2009(H1N1) refers to the fourth strain isolated in California in 2009, and belonging to type A, subtypes H1 and N1.

At least 15 influenza pandemics are known to have occurred in recorded history, with an estimated mortality of 50 million people in 1918 alone.Citation2 The major outbreaks of human influenza have so far involved the A/H1N1, A/H1N2, and A/H3N2 viruses, whereas a highly pathogenic avian influenza A/H5N1 caused considerable mortality in humans directly exposed to afflicted domestic and wild birds in the mid-1990s. Besides these major outbreaks, influenza viruses continually circulate among humans, and the subtypes and strains may vary annually or appear in a cyclical pattern with regular intervals of a few years. Global attack rates range from 5 to 10% in adults, 20 to 30% in children and the elderly, and up to 50% in specific populations and settings, with varying severity of disease. The annual global burden of severe influenza is estimated to be 3,000,000 to 5,000,000, causing 250,000 to 500,000 deaths, with 95% of these predicted to occur in developing countries.Citation3,4 However, these estimates are misleading since many cases in developing countries are likely to remain unconfirmed, and a proportion of the so called influenza-like illness (ILI) cases could be due to other respiratory viral infections. Given the difficulty of accurately assessing the burden of influenza, it is difficult to estimate the economic benefit of influenza vaccines.Citation5 There have been a number of recent studies that have delved into the burden of influenza and ILI as well as the impact of vaccination in various countries. Describing these studies in detail goes beyond the scope of this review, however, studies modeling the impact of vaccination have unequivocally shown that vaccines are critical to the reduction of influenza-related illness and healthcare costs, and that the benefits of vaccination outweigh the costs.Citation4-7 This article reviews the current status of cell culture-based influenza vaccine platform, its advantages and drawbacks, and the future of influenza vaccine research.

Immunity to influenza and correlates of protection

Several excellent studies and reviews have described and discussed immunity to influenza infection and vaccination, and only points pertaining to the evaluation of vaccines are presented here. Although all arms of the immune system are triggered by influenza virus, primary protective immunity in humans is thought to be mediated predominantly by B cell responses and the resultant antibodies. These comprise mainly the airway mucosal immunoglobulin A (IgA) but also the immunoglobulin G (IgG) transduced from the serum to the respiratory tract.Citation8 It has been observed that specific B cells appear in blood as early as 7 to 10 d after the onset of symptoms, and neutralizing antibodies directed to the hemagglutinin (HA), and to a lesser extent to the neuraminidase (NA) or the matrix (M) protein, mediate protection against infection and disease.Citation8 Indeed, the level of hemagglutination inhibition (HI) antibodies has been used as a surrogate measure of protection against influenza when assessing vaccine immunogenicity and efficacy. However, there is substantial evidence suggesting that broadly neutralizing non-HI antibodies may also play an important role.Citation8,9

As far as T lymphocyte responses are concerned, several studies have shown that human CD4+ and CD8+ T cell responses typically cross-react to multiple influenza strains and subtypes, and mainly target internal components of the virus,Citation10-12 and that both CD4+ and CD8+ T cells may independently protect humans from infection or disease.Citation13,14 Evidence from elderly subjects, in whom the antibody response is quantitatively poor compared to adults,Citation15 suggests that CD4+ and CD8+ T cell responses may correlate better with protection.Citation16-18 Studies focusing on identifying early predictors of correlates of protection following infection or vaccination have identified an array of other molecular signatures.Citation19 While these markers correlate with later induction of functional antibodies, standardized regulatory criteria are yet to be defined for any of the markers or assays for evaluating cell-mediated immunity.Citation20,21 In any case, while it is intuitive that T cell responses against conserved regions of the genome would be expected to be boosted every year, it is known that prior immunity is generally insufficient to protect against future infections, and that annual seasonal vaccines are required against even the same subtype. These findings argue against extensive correlation between T cell responses and protection. On the other hand, the relative contribution of antibody- versus cell-mediated immunity to protection following vaccination may differ significantly from infection-induced immunity.

For any infectious disease, the correlates of vaccine-mediated protection are likely to be similar to those in subjects who suffer from mild to moderate illness and recover, although the quality, quantity and the breadth of immune response may be much wider in the case of infection due to the widespread replication of the virus in the host. Indeed, adaptive immune memory to influenza, in terms of humoral and poly-functional effector or memory T cell responses, has been found to be similar between those who were vaccinated and those who experienced a mild form of the disease.Citation22 However, the type and extent of immune response depends on the type of vaccine being administered, whether it has been adjuvanted or not, the route of administration, as well as the age of the vaccinee.Citation23 The inactivated vaccines administered by the intramuscular route induce robust secondary recall IgG, good CD4+ T cell responses with the ability to help B cells, and weak CD8+ T cell as well as mucosal IgA responses, whereas the intranasally administered live attenuated vaccines (LAV) produce good mucosal IgA, relatively lower IgG, and pronounced and diverse CD4+ and CD8+ T cell as well as type I interferon (IFN) responses.Citation24,25 Specific B cell and polyclonal antibody responses are significantly higher with inactivated compared to LAV vaccines, whereas IgA, nucleoprotein-specific and cross-reactive B cell responses are better with LAV.Citation26 Efficacy of inactivated vaccines correlates well with HI antibody titres, which in turn may be predicted by CD4+ T cell responses,Citation27-29 whereas using HI titres as a yardstick may underestimate the efficacy of LAV because of weaker antibody responses.Citation30-32 On the other hand, LAV efficacy may be better correlated with neutralizing antibody titres, the frequency of IgG+ B cells, virus-specific airway IgA, or a combination of these immune parameters.Citation31-34

Current regulatory guidelines on influenza vaccine immunogenicity and efficacy assessments in humans are based solely on antibody responses. Although it might be intuitive to assess respiratory mucosal IgA, serum IgG levels have been shown to correlate with protection, and form the basis for vaccine evaluation. The most commonly employed serological assay evaluates the ability of antibodies to inhibit hemagglutination of chicken or human type ‘O’ erythrocytes. Another acceptable assay is the single radial hemolysis (SRH) assay, while the virus neutralization assay has recently been put forward as criteria for regulatory acceptance. A hemagglutination inhibition (HI) titer of ≥40 or hemolytic zone area of ≥25 mm2 in SRH assay are considered as protective titres. Titres of antibodies are converted to seroconversion (SRC), seroprotection (SRP), and geometric mean titres (GMT). SRC is defined as a pre-vaccination (baseline) HI titer of ≤10 and a post-vaccination titer of ≥40 or at least a four-fold increase if baseline titer is ≥10. SRP is defined by the HI titer of at least 40, and GMT is defined as the arithmetic mean of the logarithm of the last positive dilution of serum. Geometric mean ratio (GMR) is calculated as the fold increase in GMT from baseline to post-vaccination time-point. A vaccine is deemed to meet regulatory criteria when SRC, SRP and GMR are >70%, >40 %, and 2.5 for subjects of 18–60 y of age, and >60%, >30% and 2.0 for the elderly, respectively. The proportion of subjects showing protective titres constitutes the efficacy of the vaccine, whereas effectiveness is an estimate of protection against the occurrence of disease.

Vaccine production using chicken embryos: Eggs rule the roost

Based on a method developed a century ago, influenza vaccines have been produced since the 1940s in embryonated chicken eggs (ECE). High titers can be obtained by growing influenza virus in ECE, and extensive experience in large-scale production has led to the streamlining and automation of a highly standardized process. There is also extensive safety data available as billions of doses of ECE-produced influenza vaccine have been administered to humans. However, besides being labor intensive, ECE-derived influenza vaccines have several drawbacks. First and foremost, the dependency on eggs is a matter of concern. Because 1 to 2 ECE are required for the production of each human dose of influenza vaccine, the method requires the availability of a large number of eggs simultaneously or within a short window of time, necessitating considerable planning (up to a year in advance) to produce sufficient numbers of ECE. In addition, the eggs need to be set synchronously since inoculations need to be carried out 10–12 d after initiation of incubation. More importantly, the eggs need to be from specific pathogen-free flocks or at least certified as ‘clean’ in order to avoid adventitious agents. This could be an issue especially in developing countries since the vaccine manufacturer will have to rely on the quality control of the supplier of the ECE. Secondly, some virus strains, especially the recent H3N2 strains, do not grow well in ECE, and others such as the highly pathogenic avian influenza strains, viz., H5N1, could be lethal to embryos, resulting in low titres.Citation35-37 Indeed, this is one of the reasons why seasonal influenza vaccine strains are generated through reassortment with a high yielding strain such as A/PR/8/34. Third, occasional breakdown in sterility during downstream processing could lead to rejection of large volume of vaccine bulk, leading to a need to revisit the long process of planning and execution, or to the lack of availability of vaccine when needed. Fourth, influenza viruses appear to mutate more frequently around the receptor binding site and be selected when passaged in ECE, compared to passage in cells cultured in vitro.Citation38-43 This could potentially affect vaccine efficacy, as has been seen in animal models.Citation44,45 Moreover, egg-based antigens may elicit weaker immunogenicity compared to cell culture-derived antigens.Citation46,47 Further, there have been reports of narrower reactivity when ECE-derived antigens are used in immunogenicity assays as compared cell-derived antigens.Citation46,48 Fifth, despite an extensive purification process, residual allergenicity of egg proteins is a serious concern. Finally, ECE-based vaccine production simply cannot meet global demand in case of widespread epidemics and pandemics, as current production capacities can only deliver about half a billion doses every year. A large increase in ECE production is probably not possible, in part because of eggs being a major human proteinaceous food source.

Vaccine production using cultured cells: ‘tis time to roust

In the last 2 decades, cell-culture based systems have been advocated because of the drawbacks with ECE-derived vaccine technology. Vaccine production in cultured cells has several advantages. First, cell lines can be extensively characterized and stored for future use without the need for repeated full range testing, and cell culture avoids dependency on supply and quality control of a raw material such as ECE. Second, certain viruses grow better in cells, avoiding the down-time required for the generation of high growth reassortants. Alternatively, high growth reassortants can be directly generated in cells.Citation49,50 Third, a much more standardized and controlled process can be set up to track vaccine manufacturing. Fourth, scalability is better with cell culture than with egg-based production platforms. Fifth, allergies to egg proteins can be avoided. Sixth, virus propagated in mammalian cell systems has been shown to be structurally or antigenically more similar or identical to the field virus as compared to that grown in ECE.Citation37,41,42,45,47,51-54 Furthermore, immune responses elicited by mammalian cell-derived vaccines have been shown to be more cross-reactive than responses produced by ECE-derived vaccines,Citation46,55 although protective efficacy may not be affected.Citation46 Finally, the same facilities can be used for the production of other vaccines when not being used for the production of influenza vaccine for extended periods.

Vero cell-derived influenza vaccines: Here we come to boost

Vero (African monkey kidney epithelial) cells have been used for human vaccine production for nearly 40 years, and were the only World Health Organization approved continuous cell line substrates for some time. Beginning with polio, several vaccines have been produced or developed using Vero cells.Citation56 One of the advantages of Vero cells is that they do not produce IFN, thus allowing unhindered replication of viruses.Citation57 However, Vero cells were only recently shown to be suitable for influenza virus isolation and growth.Citation54,58 This observation was immediately followed by the adaptation of Vero cells to produce high titres of influenza virus, for vaccine production at a commercial scale,Citation57,58 and for the generation of reassortant vaccine strains using the reverse genetics technology.Citation49 The resultant vaccines were found in animal studies to be safe and immunogenic.Citation59-65 An A/H5N1 vaccine produced following generation of the reassortant strain in Vero cells could elicit antibodies which could neutralize a broad range of A/H5N1 strains in mice and guinea pigs.Citation62 The whole virion killed vaccine also induced subtype-specific T-helper type 1 (Th1) and subtype cross-reactive T-helper type 2 (Th2) responses in mice.Citation62 Mice were also protected against at least 2 heterologous A/H5N1 strains belonging to 2 different clades.Citation62 Indeed, Vero cell-derived vaccines were better inducers of T cell proliferation and Th1 responses in mice than were ECE-derived vaccines.Citation66

Human clinical trials for Vero cell-derived A/H5N1 influenza vaccines showed that doses ranging from 3.75 to 45 μg of hemagglutinin (HA) protein equivalent were well tolerated, induced robust clade- and cross-clade-reactive antibodies in pediatric and adult populations, and that these responses could be boosted 6–24 months later.Citation67-72 Seroconversion rates ranged from 6% to 86%; the responses were typically higher with a higher dose or following boosting, but lower when adjuvanted with aluminum salts, in older individuals, and against viruses belonging to heterologous clades. Significantly, responses persisted for 12–24 months in a considerable proportion (26–86%) of the subjects.Citation67,69,71,72 Seroprotective levels of antibodies were achieved in 10–100% of the individuals, generally mirroring the variation observed in seroconversion.Citation67-72 In general, SRP levels were observed in >70% of the subjects after 2 conventional doses, and the proportion of subjects rose to >90% of the adults and >70% of the elderly following another dose at 6, 12–15 or 24 months following a single or a double dose administered earlier.Citation70 GMRs ranging from 1.6 to 32.8 have been observed, again following a pattern similar to SRC and SRP.Citation67,69,71,72 The vaccine could also induce (a) strong and effective, albeit not boostable, anti-neuraminidase antibodies,Citation73 and (b) clade and cross-clade-reactive CD4+ T cell responses in both adults and the elderly, the responses lasting at least 6 months at significant levels in adults.Citation74 Importantly, the CD4+ T-cell responses cross-reacted with A/H1N1, but not to A/H1N2 or B seasonal influenza strains.Citation74 In addition, human serum antibodies protected mice against virus challenge.Citation75

A large-scale multi-center trial of Vero cell-derived seasonal influenza vaccine in adults found that SRC rates were 70.4% against A/H1N1, 79.1% against A/H3N2 and 65.7% against influenza B, SRP rates were 88% against A/H1N1, 93.3% against A/H3N2 and 97.1% against influenza B, in a population which showed baseline SRP values of 29.5%, 39.4% and 56.4% against A/H1N1, A/H3N2 and influenza B, respectively.Citation76 In the same study, GMR of 11.1, 13.5 and 7.6 were achieved against A/H1N1, A/H3N2 and influenza B, respectively. The protective efficacy was 79%, 50% and 100% by culture positivity assay, and 75.2%, 50% and 60.1% by nucleic acid positivity assay, against A/H1N1, A/H3N2 and influenza B, respectively.Citation76 Overall efficacy of Vero cell-derived seasonal influenza vaccines has been reported to be 73–82% against antigenically matched and 68–83% against all strains of influenza viruses.Citation77,78 With the 2009 pandemic A/H1N1 (H1N1pdm09) strain, higher and comparatively longer lasting protective efficacy was reached, with a single dose in adults and older adolescents, and with 2 doses in young adolescents and children, and these responses were boosted strongly by the A/H1N1 component of the seasonal trivalent vaccine.Citation76,79

Safety profiles of Vero cell-derived influenza vaccines have shown that they are generally as well tolerated as the ECE-derived vaccines. Total local adverse events (AEs) have ranged from 11% to 29%, typically higher after the first dose than the second dose, as well as when adjuvanted with aluminum salts, and in younger individuals.Citation67,70,71,79 The most frequently reported local AEs have been pain at injection site (10–43% of the subjects), followed by redness, swelling, induration and ecchymosis.Citation67,70,71,79-81 Total systemic AEs have been reported in 18–51% of the subjects,Citation65,68,69 the most common being headache, fatigue, myalgia and malaise (3–20% of the subjects) in adults as well as the elderly.Citation70,71,81 A wider variety of events have been reported in children.Citation79,80 However, the frequency of occurrence of the AEs is not significantly different from subjects administered a placebo or an ECE-derived vaccine.

MDCK cell-derived influenza vaccines: The final joust?

Compared to other cell lines, the MDCK cells present several advantages for influenza vaccine production. First, MDCK cells are the most suitable substrates among cultured cells to obtain primary isolates of influenza viruses.Citation37,54,82-85 This might be partly because (a) of the inability of canine IFN-induced myxovirus resistance protein 1 (Mx1) to inhibit influenza virus replication, and (b) trypsin used to facilitate virus entry during infection of cells exerts negative effects on IFN-induced antiviral proteins.Citation86-88 Together, the ineffectiveness of the IFN system may allow the virus to replicate to higher titres. A comparison of various cell lines for supporting the replication of live attenuated influenza viruses showed that MDCK cells are better than Vero, Medical Research Council-5 (MRC-5) human fetal lung fibroblast, Wistar Institute-38 (WI-38) human fetal diploid lung, fetal rhesus lung (FRhL), A549 human lung carcinoma and National Cancer Institute (NCI) H292 human mucoepidermoid bronchiolar carcinoma cells.Citation89-91 Whereas A549, NCI H292, MRC-5, WI-38 and FRhL cells produced low to moderate titres of virus, both Vero and MDCK cells could support the replication of influenza virus to higher titres.Citation89,90 However, Vero cells could only produce high virus titres with certain strains and when grown in the presence of serum.Citation89 MDCK cells may also be better suited for neutralization assays.Citation92 Second, MDCK cells have been shown to be the most suitable for large-scale production of influenza virus.Citation89,93,94 Head-to-head comparison in laboratory scale bioreactors showed that MDCK cells yielded more virus than did Vero cells.Citation94 Third, influenza virus replicates more rapidly in MDCK cells compared to other cell lines,Citation95 and can be adapted to produce high titres in MDCK cells in as few as 3 to 10 passages, i.e., in 10–30 days, depending on the strain. This may reduce the lead time for vaccine production.Citation96 Fewer passages during adaptation would also reduce the chances of accumulation of mutations around the receptor binding site of the HA protein. In addition, trypsin does not need to be added frequently for virus propagation in MDCK cells, avoiding potential chances of contamination, although trypsin inhibitors have also been reported to be secreted by MDCK cells.Citation97 Fourth, the use of MDCK cells may be significantly more advantageous for the production of some influenza B virus vaccines.Citation93 Finally, MDCK cells are refractory to human and mouse prions,Citation98 and in vitro data suggests that MDCK cell derived components are not allergenic.Citation99,100

Extensive literature exists on the adaptation of MDCK cells for scaling up and influenza vaccine production. The cells can be easily adapted to and be grown in serum-free media, and in suspension, as well as on various microcarriers maintained under various bioreactor conditions.Citation93,101-108 Subclones of MDCK cells adopted to grow in suspension and support robust virus production have also been described,Citation91,108,109 although adherent MDCK cells appear to support more robust virus production than suspension MDCK cells.Citation110

Influenza vaccines derived from MDCK cells are also safe and immunogenic. Initial studies which compared ECE- and MDCK cell-derived vaccines in Phase I clinical trials demonstrated the comparable safety and immunogenicity of the 2 vaccines in children, healthy adults and the elderly.Citation111-114 Other studies found that MDCK cell-derived vaccines were at least equivalent, and sometimes better and more efficacious as compared to ECE-derived antigens.Citation111,112,114-120 In one instance, it was reported that at risk adult and elderly subjects who did not respond serologically to a previous ECE-derived vaccine responded better when boosted with MDCK cell-derived vaccine as compared to an ECE-based vaccine.Citation121

Since the early 1990s, reports of more than 20 clinical studies involving greater than 20,000 subjects in over a dozen countries, as well as large-scale immunization programs have further confirmed the safety and immunogenicity of MDCK cell-derived influenza vaccines. As far as safety is concerned, overall AEs have been reported in up to 84% of the subjects,Citation112,114,116,117,122-125 with a higher incidence in adults (60–84%) as compared to children (50–60%) and lowest (typically 15–25%, but sometimes up to 50%) in the elderly.Citation114,122,123,125 Total local AEs have ranged from 10% to 84%,Citation112,114–117,122,124,126 again, typically higher in adults than in children, and lowest in the elderly.Citation114,116,118,122 Local AEs are also higher in the case of adjuvanted vaccine formulations as compared to unadjuvanted vaccine.Citation122,123,126 The most common local AE has been pain at injection site (12–75%), followed by erythema (2–20%), induration (6–15%), swelling (2–15%), and ecchymosis (0–18%).Citation112,115,116-120,122,123,125–129 Some investigators have also reported limitation in movement, tenderness and bruising.Citation114,127 In general, the local reactions are mild, and are not significantly different from subjects administered ECE-derived vaccine or a placebo. Mild to severe reactions requiring medical attention are observed at the most in 25% of the total local AEs, and are usually more frequent in children.Citation112,114,124

Systemic AEs to MDCK cell-derived influenza vaccines have been found to be lower as compared to local AEs. Total systemic AEs have ranged from 20% to 55%.Citation112,114-117,122,126 Similar to local AEs, systemic AEs are also lowest in the elderly.Citation114,116,118,122 However, in contrast to the local AEs, systemic AEs are only slightly lower in children as compared to that in adults.Citation114,122 Adjuvanted preparations typically produce higher local AEs but systemic AEs are either similar or only slightly more as compared to unadjuvanted vaccines.Citation122,123,126 The commonest systemic AE is headache, being reported in 6.7–32% of the subjects, followed by myalgia (2–30%), fatigue (4–24%), malaise (3–25%), sweating (0–16%), chills (0–14%), and arthralgia (0–15%).Citation112,114-120,122,123,125,126,128,129 Other systemic AEs, which are typically observed in less than 10% of the subjects, include nausea, loss of appetite, diarrhea, vomiting, fever and rash. A wider variety of systemic reactions, including sleepiness, inappetence, irritability, and unusual crying have been reported in young children.Citation129 None of the systemic AEs are significantly different from those due to ECE-derived vaccine. In addition, the systemic AEs typically disappear following a short symptomatic treatment.

Immunogenicity studies with MDCK cell-derived influenza vaccines in humans have revealed that (a) the SRC rates range from 25% to 100%, (b) the SRP rates are achieved in 70–100% of the subjects, and (c) the GMRs vary widely from as low as 1.89 to as high as 478, depending on the quantity of antigen injected, the frequency and time duration between the doses, the age of the vaccinee and whether the vaccine was formulated with an adjuvant.Citation111,112,114-120,122,123,125,127,128 The SRC rates and GMR are typically the best in adults, lower in children and the lowest in the elderly, whereas the SRP rates are similar in children and adults while being slightly lower in the elderly.Citation114,116,118,120,122,123,125,128 When comparing between subtypes of the viruses used as part of a trivalent vaccine, overall responses in terms of SRC, SRP and GMR are equivalent against the different influenza A subtypes H3N2 and H1N1, whereas responses to influenza B virus are comparatively weaker.Citation117,125,128 Some studies have shown that substantial proportion of the subjects retain seroprotective titres for at least 6 months, and in some cases, more than a year,Citation115,123 and that the dose can be reduced at least by half by using MF59 as an adjuvant.Citation122,127,130-132

Whereas all the above described studies involve killed vaccines, a recent study has shown that an MDCK cell-derived LAV vaccine containing 3 reassortant virus strains does not cause AEs that are significantly different from placebo, but immunogenicity, as determined by serum antibody titres, could only be demonstrated for H3N2, but not for H1N1 or B, influenza viruses.Citation133 However, since no ILI was observed, it is possible that either airway IgA or cell-mediated responses may have been responsible for protection against infection and/or disease.

Other cell lines tested for influenza vaccine production

Several other cell lines including the human embryonic retinal cell line PER.C6, the chick embryo cell line PBS-1, the duck embryo retinal cell line AGE.CR, and the duck embryonic stem cell line EB66 have been explored for producing influenza vaccine. The only cell line from humans is PER.C6, which was derived by transformation of embryonic retinal epithelial cells with human adenovirus type 5 E1 region genes.Citation134 The cell line has been characterized extensively to meet regulatory requirements.Citation135 These cells can be propagated in suspension and be adopted to serum-free and bioreactor conditions.Citation136 Influenza viruses can be propagated in PER.C6 cells, and reassortant vaccine strains can be generated in these cells.Citation63,136-138 Avian influenza H7N1 vaccine produced in PER.C6 cells has been shown to be well tolerated by humans.Citation139 However, immunogenicity studies indicated slightly lower antibody titres, which were enhanced somewhat by aluminum adjuvant.Citation139 There was also a correlation with antibody response and antibody secreting cells and interleukin-2 production.Citation139 However, poor immunogenicity in humans is a concern with influenza vaccine produced from PER.C6 cells, although more extensive studies are required. The related human embryonic kidney 293 cells, which are also derived through transformation using sheared adenovirus genome containing the E1 region genes,Citation140 have also been shown to support production of scalable quantities of influenza virus.Citation141

Three avian cell lines have also been developed for the production of influenza vaccines. The best characterized of these are the AGE1.CR and the EB66 cell lines. The AGE1.CR cell line was derived by transforming Muscovy duck embryo retinal cells through stable transfection with human adenovirus type 5 E1A/E1B genes. The cells have been extensively characterized to meet regulatory requirements, and can grow in suspension, although they require the addition of trypsin for influenza virus propagation,Citation105,142-144 The EB66 cell line was derived as a stable line through a non-chemical non-genetic selection process, and can be adopted to various culture conditions, including suspension culture, bioreactor conditions and to serum-free or chemically defined media, and can grow to high density.Citation145 Another avian cell line potentially useful for influenza virus production is the immortalized chick embryo cell line PBS-1. This is a non-tumorigenic cell line which has been reported to support the propagation of influenza virus to titres higher than that obtained with Vero, MDCK and primary chick embryo kidney cells.Citation146 The cells have also been shown to be free of adventitious agents, and are readily adaptable to a variety of culture conditions, including growth in serum-free media and on microcarrier beads.Citation146,147 Importantly, propagation of influenza viruses in PBS-1 cells does not require the addition of exogenous trypsin, reducing chances of contamination, and simplifying downstream processing.Citation146 However, pre-clinical development and clinical trial studies are necessary for influenza vaccines produced in any of the avian cell lines.

Another cell line demonstrated to support growth of influenza viruses to titres comparable to those with MDCK cells is the mink lung epithelial cell line Mv1Lu.Citation148 Cytopathic effects can be observed earlier in these cells, and titres of influenza A viruses are higher in these cells than in MDCK cells.Citation92,148-150 However, further characterization and clinical studies would be required before they could be adopted as cell substrates for influenza vaccine production.

Issues related to cell culture-derived influenza vaccines

No vaccine production system is without shortcomings and cell-based influenza vaccine platforms are not an exception. Besides being vulnerable to occasional contamination, the cells must be free from additional or specific extraneous agents relevant to the species of origin of the cell line. This is typically not an issue since tests carried out for characterization of cell banks include extensive adventitious agent testing. Another issue is the fact that the reference reagents used for quality control and potency assays were largely produced using ECE-grown virus, and there is evidence that reactivity could be broader when MDCK-grown virus is used.Citation46,55,92 However, this could be overcome by shifting to producing reagents using cell culture-derived virus. It is also possible that some virus strains may not grow well in certain cell types, and several cell lines or clones may need to be tested for initial expansion of the vaccine virus.Citation37 A particular concern with the use of cell lines is that intellectual property issues may hinder wide utilization of an available system or technology. And finally, manufacturing influenza vaccines in cell culture is typically more expensive than vaccine production using ECEs.

Initial problems with the use of Vero cells in influenza vaccine production stemmed from the fact that virus replication was found to be low.Citation151,152 It was demonstrated that one of the reasons for this was that Vero cells contain an anti-tryptic activity, and that repeated addition of TPCK-treated trypsin could aid in multicycle replication and generation of high virus yields.Citation153 Even so, comparative studies have shown that whereas peak titres could be higher,Citation94 the growth of influenza A and B viruses may be slower in Vero cells compared to that in MDCK cells,Citation48,93-95,101 and that extended passaging may be required to obtain high titer virus suitable for industrial scale production.Citation154,155 This may be disadvantageous in case of serious epidemics or pandemics, when vaccines need to be available in a short amount of time. In addition, as the whole process could take 1 to 3 months, the virus may accumulate mutations during the passaging process. The trypsin concentration may also need to be optimized for each culture condition as the enzyme may influence adherence of cells which are already under bioreactor shear forces.Citation93 On the other hand, the ratio of genome copies to infectious unit or infectious unit to total virions have been found to be better with influenza A propagated in Vero cells than in MDCK cells.Citation94,95 The virus produced in Vero cells has also been shown to be more stable at least with A/H1N1, compared to MDCK-derived virus, especially when propagated in serum-free medium.Citation94 In addition, reverse genetics systems have also been developed so that high-yielding reassortant vaccine strains can be rapidly generated directly in Vero cells.Citation49,156,157

A major issue with the use of MDCK cells for the production of influenza vaccines is tumorigenicity. MDCK cells were derived in 1958 from the kidney of an apparently healthy female cocker spaniel dog, and are reported to be reverse transcriptase negative and non-tumorigenic.Citation158 Since then, multiple independent lines showing varying degrees of tumorigenicity, including apparently non-tumorigenic clones, have been derived by various groups.Citation113,159–162 However, several lines of argument have been put forward to dispel concerns about tumorigenicity. First of all, the probability of live cell remaining in the final product is extremely low (10−25 to −35/dose) due to the multiple redundant manufacturing steps which remove live cells.Citation163-166 Moreover, any live MDCK cell is likely to be rejected by the immune system as it is xenogenic to humans.Citation158 Indeed, except in completely immunocompromised nude mice, MDCK cells have not been shown to be tumorigenic, and either cell extracts or DNA extracted from cells are not oncogenic even in nude mice.Citation163,164 In addition, models for mitigation of risks, including those due to adventitious agents, have found that MDCK cells are safe cell substrates, and equivalent or better than ECE or Vero cells for producing vaccines.Citation165,167-173 In addition to MDCK cells, PER.C6 cells are also potentially tumorigenic, although these risks can be mitigated through the production process.Citation174 However, concerns about tumorigenicity might be more relevant, even if infinitesimally minor, for LAV vaccines produced in these cells.

Other concerns relating to the use of MDCK cells should also be noted here. One issue is that the use of MDCK cells to isolate viruses from respiratory samples can also yield viruses other than influenza virus, but this may be only a minor problem as these other adventitious viruses could be rapidly eliminated upon passaging.Citation173 A couple of studies have also reported that passage of influenza virus in MDCK cells may select antigenically and structurally distinct variants, e.g., those that mirror ECE-derived viruses,Citation175 or those that show altered fusion and entry characteristics,Citation176 and these altered viruses may outgrow other, desired phenotypes. Another study has also reported quasispecies consisting of HA variants in Vero and MDCK cells.Citation154 It is not clear how these would affect vaccine production or overall immunogenicity of the vaccine. On the other hand, certain other mutations have been shown to affect serological assays, and may influence the serological evaluation of the vaccine.Citation177,178 Other issues with cell culture-derived vaccines include lower levels of neuraminidase in the virion compared to virus grown in ECE,Citation46 and differential glycosylation patterns.Citation92,152,176,177 Although these may influence immunogenicity in animal models,Citation179 overwhelming evidence from clinical studies in humans suggests that there is no significant difference in immunogenicity. However, this has not been supported with a detailed characterization of how these differences affect the various components of the immune response.

Despite advancements, the use of the cell culture platform for influenza vaccine production faces commercial challenges. First, each manufacturer will almost certainly have to optimize their cells for growth conditions and to support propagation of influenza virus. In addition, growth of cells in serum- and animal component-free media, and in suspension or in bioreactor conditions may need to be standardized. Second, the virus yield and titer per unit volume are lower and variable as compared to ECE-derived vaccine production, necessitating the use of steps to concentrate the virus. However, this could be circumvented by generating a high yielding reassortment recipient virus. Third, there is a need for extensive testing for adventitious agents, not only for human pathogens but also for other mammalian pathogens, especially those originating from the host from which the cells were derived. In addition, since the virus is highly lytic, host cell-derived proteins and DNA need to be carefully removed to meet regulatory requirements. Fourth, the cost of production per unit dose is high. Fifth, the technology for the production of influenza vaccines using ECEs is unrestricted whereas widespread application of the cell culture platform for influenza vaccine production is hindered by restrictions due to intellectual property rights. Finally, influenza is still not considered to be a serious disease in most of the developing and underdeveloped countries, where diseases such as malaria, tuberculosis, acquired immunodeficiency syndrome, dengue etc. are more important. However, it has to be noted that experience with the production of cell culture-based influenza vaccines is barely 2 decades old compared to 6 decades with the production using ECEs, and it is anticipated that recent investments in new facilities, research and development, and production and downstream processes will increase the practical utility of this vaccine platform.

The road ahead

Keeping in mind the enormity of the seasonal influenza burden and the constant potential for a pandemic, multiple approaches will be required to reach the goal of controlling influenza. Here are 10 mutually non-exclusive principles on which to base our efforts toward conquering influenza virus. Most of these areas are already the focus of investigations by vaccinologists worldwide.

  1. Employing one particular technology or platform of vaccine production may not be sufficient, especially for meeting demands in the case of pandemics. Besides ECE and cell culture-based classical vaccines, partial or periodical dependence on other technological platforms may be inevitable.

  2. Vaccines that elicit neutralizing antibodies targeting conserved or least variable regions of HA, possibly in combination with other antibodies which target other viral targets, would be highly desirable.

  3. In addition to focusing only on humoral immunity, mechanisms that induce strong effector and memory cell-mediated immune responses, which probably contribute to extended and long-term protection, will need to be investigated.

  4. In addition, standardized assays measuring cell-mediated immune correlates of protection following vaccination, which can be used to formulate regulatory guidelines will need to be identified. However, one needs to keep in mind that different populations may react differently, and the requirements may vary from population to population.

  5. Directed stimulation of innate immune signals may be required to further fine-tune and strengthen adaptive immune responses to enhance protective efficacy.

  6. Reducing the dose of vaccine antigen either by calibrating potency, by using adjuvants or by targeted stimulation of innate immune responses could extend the number of doses available, and help mitigate shortages especially during times of pandemics.

  7. Better assays which assess more than just HI antibodies are needed to estimate potency and immunogenicity of the vaccines.

  8. Different vaccine doses and vaccination regimens will need to be drawn up for immunizing children, adults, and the elderly.

  9. Other routes of administration, such as transdermal delivery, need to be further investigated.

  10. The use of immune enhancers or immunotherapeutics may have to be investigated either alone or in combination with vaccination, especially in the case of severe epidemics or pandemics. It may also be worthwhile exploring the use of passive immunity.

Disclosure of Potential Conflicts of Interest

The author's organization, Ella Foundation, was involved in the development and pre-clinical testing as well as consultation during clinical evaluation, data analysis and publication relating to HNVAC, an H1N1pdm09 vaccine of Bharat Biotech International Limited (BBIL), Hyderabad, India. Ella Foundation also provides other advice and consultancy on production and assay development for other products of BBIL.

Acknowledgments

I thank Dr. Kimberley Goldsmith, Institute of Psychology, Psychiatry and Neuroscience, King's College London, for critical review of the manuscript.

Funding

All of the activities of Ella Foundation were funded by BBIL.

References

  • Rappuoli R, Pizza M, del Guidice G, de Gregorio E. Vaccines, new opportunities for a new society. Proc Natl Acad Sci U SA 2014; 111:12288-93; PMID:25136130; http://dx.doi.org/10.1073/pnas.1402981111
  • Morens DM, Taubenberger JK, Folkers GK, Fauci AS. Pandemic influenza's 500th anniversary. Clin Infect Dis 2010; 51:1442-4; PMID:21067353; http://dx.doi.org/10.1086/657429
  • Nair H, Simões EA, Rudan I, Gessner BD, Azziz-Baumgartner E, Zhang JS, Feikin DR, Mackenzie GA, Moïsi JC, Roca A, Baggett HC, et al. Global and regional burden of hospital admissions for severe acute lower respiratory infections in young children in 2010: a systematic analysis. Lancet 2013; 381:1380-90; PMID:23369797; http://dx.doi.org/10.1016/S0140-6736(12)61901-1
  • Carrillo-Santisteve P, Ciancio BC, Nicoll A, Lopalco PL. The importance of influenza prevention for public health. Hum Vaccin Immunother 2012; 8:89-95; PMID:22251992; http://dx.doi.org/10.4161/hv.8.1.19066
  • Jit M, Newall AT, Beutels P. Key issues for estimating the impact and cost-effectiveness of seasonal influenza vaccination strategies. Hum Vaccin Immunother 2013; 9:834-40; PMID:23357859; http://dx.doi.org/10.4161/hv.23637
  • de Waure C. Veneziano MA, Cadeddu C, Capizzi S, Specchia ML, Capri S, Ricciardi W. Economic value of influenza vaccination. Hum Vaccin Immunother 2012; 8:119-29; PMID:22251999; http://dx.doi.org/10.4161/hv.8.1.18420
  • Preaud E, Durand L, Macabeo B, Farkas N, Sloesen B, Palache A, Shupo F, Samson SI. Annual public health and economic benefits of seasonal influenza vaccination: a European estimate. BMC Public Health 2014; 14:813; PMID:25103091; http://dx.doi.org/10.1186/1471-2458-14-813
  • Chiu C, Ellebedy AH, Wrammert J, Ahmed R. B cell responses to influenza infection and vaccination. Curr Top Microbiol Immunol 2014; 386:381-98; Epub ahead of print, Sep 6; PMID:25193634; http://dx.doi.org/10.1007/82_2014_425
  • Fox A, Mai LQ, Thanh LT, Wolbers M, Le Khanh Hang N, Thai PQ, Thu Yen NT, Minh Hoa LN, Bryant JE, Duong TN, et al. Hemagglutination inhibiting antibodies and protection against seasonal and pandemic influenza infection. J Infect 2015; 70:187-96. http://dx.doi.org/10.1016/j.jinf.2014.09.003
  • Babon JA, Cruz J, Orphin L, Pazoles P, Co MD, Ennis FA, Terajima M. Genome-wide screening of human T-cell epitopes in influenza A virus reveals a broad spectrum of CD4(+) T-cell responses to internal proteins, hemagglutinins, and neuraminidases. Hum Immunol 2009; 70:711-21; PMID:19524006; http://dx.doi.org/10.1016/j.humimm.2009.06.004
  • Rimmelzwaan GF, Kreijtz JH, Bodewes R, Fouchier RA, Osterhaus AD. Influenza virus CTL epitopes, remarkably conserved and remarkably variable. Vaccine 2009; 27:6363-5; PMID:19840674; http://dx.doi.org/10.1016/j.vaccine.2009.01.016
  • Grant E, Wu C, Chan KF, Eckle S, Bharadwaj M, Zou QM, Kedzierska K, Chen W. Nucleoprotein of influenza A virus is a major target of immunodominant CD8+ T-cell responses. Immunol Cell Biol 2013; 91:184-94; PMID:23399741; http://dx.doi.org/10.1038/icb.2012.78
  • Wilkinson TM, Li CK, Chui CS, Huang AK, Perkins M, Liebner JC, Lambkin-Williams R, Gilbert A, Oxford J, Nicholas B, et al. Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat Med 2012; 18:274-80; PMID:22286307; http://dx.doi.org/10.1038/nm.2612
  • Sridhar S, Begom S, Bermingham A, Hoschler K, Adamson W, Carman W, Bean T, Barclay W, Deeks JJ, Lalvani A. Cellular immune correlates of protection against symptomatic pandemic influenza. Nat Med 2013; 19:1305-12; PMID:24056771; http://dx.doi.org/10.1038/nm.3350
  • Sasaki S, Sullivan M, Narvaez CF, Holmes TH, Furman D, Zheng N-Y, Nishtala M, Wrammert J, Smith K, James JA, et al. Limited efficacy of inactivated influenza vaccine in the elderly individuals is associated with decreased production of vaccine-specific antibodies. J Clin Invest 2014; 121:3109-19; http://dx.doi.org/10.1172/JCI57834
  • McElhaney JE, Ewen C, Zhou X, Kane KP, Xie D, Hager WD, Barry MB, Kleppinger A, Wang Y, Bleackley RC. Granzyme B: Correlates with protection and enhanced CTL response to influenza vaccination in older adults. Vaccine 2009; 27:2418-25; PMID:19368783; http://dx.doi.org/10.1016/j.vaccine.2009.01.136
  • Hsu JP, Phoon MC, Koh GC, Chen MI, Lee VJ, Wu Y, Xie ML, Cheong A, Leo YS, Chow VT. Comparison of neutralizing antibody and cell-mediated immune responses to pandemic H1N1 2009 influenza virus before and after H1N1 2009 influenza vaccination of elderly subjects and healthcare workers. Int J Infect Dis 2012; 16:e621-7; PMID:22704721; http://dx.doi.org/10.1016/j.ijid.2012.04.010
  • Dolfi DV, Mansfield DK, Kurupati RK, Kanan S, Doyle SA, Ertl HC, Schmader KE, Wherry EJ. Vaccine-induced boosting of influenza virus-specific CD4 T cells in younger and aged humans. PLoS One 2013; 8:e77164; PMID:24155927; http://dx.doi.org/10.1371/journal.pone.0077164
  • Miller MS, Palese P. Peering into the crystal ball: influenza pandemics and vaccine efficacy. Cell 2014; 157:294-9; PMID:24725400; http://dx.doi.org/10.1016/j.cell.2014.03.023
  • Cox RJ. Correlates of protection to influenza virus, where do we go from here? Hum Vaccin Immunother 2013; 9:405-8; PMID:23291930; http://dx.doi.org/10.4161/hv.22908
  • Reber A, Katz J. Immunological assessment of influenza vaccines and immune correlates of protection. Exp Rev Vaccines 2013; 12:519-36; PMID:23659300; http://dx.doi.org/10.1586/erv.13.35
  • Bonduelle O, Carrat F, Luyt CE, Leport C, Mosnier A, Benhabiles N, Krivine A, Rozenberg F, Yahia N, Samri A, et al. Characterization of pandemic influenza immune memory signature after vaccination or infection. J Clin Invest 2014; 124:3129-36; PMID:24911149; http://dx.doi.org/10.1172/JCI74565
  • He XS, Holmes TH, Mahmood K, Kemble GW, Dekker CL, Arvin AM, Greenberg HB. Phenotypic changes in influenza-specific CD8+ T cells after immunization of children and adults with influenza vaccines. J Infect Dis 2008; 197:803-11; PMID:18279048; http://dx.doi.org/10.1086/528804
  • Hoft DF, Babusis E, Worku S, Spencer CT, Lottenbach K, Truscott SM, Abate G, Sakala IG, Edwards KM, Creech CB, et al. Live and inactivated influenza vaccines induce similar humoral responses, but only live vaccines induce diverse T-cell responses in young children. J Infect Dis 2011; 204:845-53; PMID:21846636; http://dx.doi.org/10.1093/infdis/jir436
  • Rudenko L, Kiseleva I, Naykhin AN, Erofeeva M, Stukova M, Donina S, Petukhova G, Pisareva M, Krivitskaya V, Grudinin M, et al. Assessment of human immune responses to H7 avian influenza virus of pandemic potential: results from a placebo-controlled, randomized double-blind phase I study of live attenuated H7N3 influenza vaccine. PLoS One 2014; 9:e87962; PMID:24533064; http://dx.doi.org/10.1371/journal.pone.0087962
  • Sasaki S, Holmes TH, Albrecht RA, Garcia-Sastre A, Dekker CL, He XS, Greenberg HB. Distinct cross-reactive B-cell responses to live attenuated and inactivated influenza vaccines. J Infect Dis 2014; 210:865-74; PMID:24676204; http://dx.doi.org/10.1093/infdis/jiu190
  • Pedersen GK, Madhun AS, Breakwell L, Hoschler K, Sjursen H, Pathirana RD, Goudsmit J, Cox RJ. T-helper 1 cells elicited by H5N1 vaccination predict seroprotection. J Infect Dis 2012; 206:158-66; PMID:22551811; http://dx.doi.org/10.1093/infdis/jis330
  • Nayak JL, Fitzgerald TF, Richards KA, Yang H, Treanor JJ, Sant AJ. CD4+ T-cell expansion predicts neutralizing antibody responses to monovalent, inactivated 2009 pandemic influenza A(H1N1) virus subtype H1N1 vaccine. J Infect Dis 2013; 207:297-305; PMID:23148285; http://dx.doi.org/10.1093/infdis/jis684
  • Pedersen GK, Madhun AS, Beakwell L, Hoschler K, Sjursen H, Pathirana RD, Goudsmit J, Cox RJ. T-helper 1 cells elicited by H5N1 vaccination predict seroprotection. J Infect Dis 2012; 206:158-66; PMID:22551811; http://dx.doi.org/10.1093/infdis/jis330
  • Edwards EM, Dupont WD, Westrich MK, Plummer WD Jr, Palmer PS, Wright PF. A randomized controlled trial of cold adapted and inactivated vaccines for the prevention of influenza A disease. J Infect Dis 1994; 169:68-76; PMID:8277200; http://dx.doi.org/10.1093/infdis/169.1.68
  • Belshe RB, Gruber WC, Mendelman PM, Mehta HB, Mahmood K, Reisinger K, Treanor J, Zangwill K, Hayden FG, Bernstein DI, et al. Correlates of immune protection induced by live, attenuated, cold-adapted, trivalent, intranasal influenza vaccine. J Infect Dis 2000; 181(3):1133-1137; PMID:10720541; http://dx.doi.org/10.1086/315323
  • Beyer WE, Palache AM, de Jong JC, Osterhaus AD. Cold-adapted live influenza vaccine versus inactivated vaccine: systemic vaccine reactions, local and systemic antibody response, and vaccine efficacy. A meta-analysis. Vaccine 2002; 20(9–10):134-153; PMID:11818152; http://dx.doi.org/10.1016/S0264-410X(01)00471-6
  • Lee MS, Mahmood K, Adhikary L, August JM, Cordova J, Cho I, Kemble G, Reisinger K, Walker RE, Mendelman PM. Measuring antibody responses to a live attenuated influenza vaccine in children. Pediatr Infect Dis J 2004; 23:852-6; PMID:15361726; http://dx.doi.org/10.1097/01.inf.0000137566.87691.3b
  • Sasaki S, Jaimes MC, Holmes TH, Dekker CL, Mahmood K, Kemble GW, Arvin AM, Greenberg HB. Comparison of the influenza virus-specific effector and memory B-cell responses to immunization of children and adults with live attenuated or inactivated influenza vaccines. J Virol 2007; 81:215-28; PMID:17050593; http://dx.doi.org/10.1128/JVI.01957-06
  • Lu B, Zhou H, Ye D, Kemble G, Jin H. Improvement of influenza A/Fujian/411/02 (H3N2) virus growth in embryonated chicken eggs by balancing the hemagglutinin and neuraminidase activities, using reverse genetics. J Virol 2005; 79:6763-71; PMID:15890915; http://dx.doi.org/10.1128/JVI.79.11.6763-6771.2005
  • Horimoto T, Takada A, Fujii K, Goto H, Hatta M, Watanabe S, Iwatsuki-Horimoto K, Ito M, Tagawa-Sakai Y, Yamada S, et al. The development and characterization of H5 influenza virus vaccines derived from a 2003 human isolate. Vaccine 2006; 24:3669-76; PMID:16378663; http://dx.doi.org/10.1016/j.vaccine.2005.07.005
  • Donis RO; The Influenza Cell Culture Working Group. Performance characteristics of qualified cell lines for isolation and propagation of influenza viruses for vaccine manufacturing. Vaccine 2014; 32(48):6583-90; Epub ahead of print, Jun 24; http://dx.doi.org/10.1016/j.vaccine.2014.06.045
  • Robertson JS, Naeve CW, Webster RG, Bootman JS, Newman R, Schild GC. Alternations in the hemagglutinin associated with adaptation of influenza B virus to growth in eggs. Virology 1985; 143:166-74; PMID:4060580; http://dx.doi.org/10.1016/0042-6822(85)90105-9
  • Katz JM, Naeve CW, Webster RG. Host cell-mediated variation in H3N2 influenza viruses. Virology 1987; 156:386-95; PMID:3811239; http://dx.doi.org/10.1016/0042-6822(87)90418-1
  • Robertson JS, Bootman JS, Newman R, Oxford JS, Daniels RS, Webster RG, Schild GC. Structural changes in the hemagglutinin which accompany egg adaptation of an influenza A (H1N1) virus. Virology 1987; 160:31-7; PMID:3629978; http://dx.doi.org/10.1016/0042-6822(87)90040-7
  • Katz JM, Webster RG. Antigenic and structural characterization of multiple subpopulations of H3N2 influenza virus from an individual. Virology 1988; 165:446-56; PMID:3407150; http://dx.doi.org/10.1016/0042-6822(88)90588-0
  • Wang ML, Katz JM, Webster RG. Extensive heterogeneity in the hemagglutinin of egg-grown influenza viruses from different patients. Virology 1989; 171:275-9; PMID:2741346; http://dx.doi.org/10.1016/0042-6822(89)90538-2
  • Katz JM, Webster RG. Amino acid sequence identity between the HA1 of influenza A (H3N2) viruses grown in mammalian and primary chick kidney cells. J Gen Virol 1992; 73:1159-65; PMID:1588320; http://dx.doi.org/10.1099/0022-1317-73-5-1159
  • Wood JM, Oxford JS, Dunleavy U, Newman RW, Major D, Robertson JS. Influenza A (H1N1) vaccine efficacy in animal models is influenced by two amino acid substitutions in the hemagglutinin molecule. Virology 1989; 171:214-21; PMID:2741341; http://dx.doi.org/10.1016/0042-6822(89)90528-X
  • Kodihalli S, Justewicz DM, Gubareva LV, Webster RG. Selection of a single amino acid substitution in the hemagglutinin molecule by chicken eggs can render influenza A virus (H3) candidate vaccine ineffective. J Virol 1995; 69:4888-97; PMID:7609057
  • Alymova IV, Kodihalli S, Govorkova EA, Fanget B, Gerdil C, Webster RG. Immunogenicity and protective efficacy in mice of influenza B virus vaccines growth in mammalian cells. J Virol 1998; 72:4472-7; PMID:9557744
  • Katz JM, Webster RG. Efficacy of inactivated influenza A virus (H3N2) vaccines grown in mammalian cells or embryonated eggs. J Infect Dis 1989; 160:191-8; PMID:2760480; http://dx.doi.org/10.1093/infdis/160.2.191
  • Govorkova EA, Kodihalli S, Alymova IV, Fanget B, Webster RG. Growth and immunogenicity of influenza viruses cultivated in Vero or MDCK cells and in embryonated chicken eggs. Dev Biol Stand 1999; 98:39-51; PMID:10494958
  • Nicolson C, Major D, Wood JM, Robertson JS. Generation of influenza vaccine viruses on Vero cells by reverse genetics: an H5N1 candidate vaccine strain produced under a quality system. Vaccine 2005; 23:2943-52; PMID:15780743; http://dx.doi.org/10.1016/j.vaccine.2004.08.054
  • Jung EJ, Lee KH, Seong BL. Reverse genetic platform for inactivated and live-attenuated influenza vaccine. Exp Mol Med 2010; 42:116-21; PMID:20054235; http://dx.doi.org/10.3858/emm.2010.42.2.013
  • Katz JM, Wang M, Webster RG. Direct sequencing of the HA gene of influenza (H3N2) virus in original clinical samples reveals identity with mammalian cell-grown virus. J Virol 1990; 64:1808-11; PMID:2319652
  • Robertson JS, Bootman JS, Nicolson C, Major D, Robertson EW, Wood JM. The hemagglutinin of influenza B virus present in clinical material is a single species identical to that of mammalian cell-grown virus. Virology 1990; 179:35-40; PMID:2219728; http://dx.doi.org/10.1016/0042-6822(90)90270-2
  • Robertson JS, Nicolson C, Bootman JS, Major D, Robertson EW, Wood JM. Sequence analysis of the haemagglutinin (HA) of influenza A (H1N1) viruses present in clinical material and comparison with the HA of laboratory-derived virus. J Gen Virol 1991; 72:2671-77; PMID:1940864; http://dx.doi.org/10.1099/0022-1317-72-11-2671
  • Govorkova EA, Murti G, Meigneier B, deTaisne C, Webster RG. African green monkey (Vero) cells provide an alternative host cell system for influenza A and B viruses. J Virol 1996; 70:5519-24; PMID:8764064
  • Kishida N, Fujisaki S, Yokoyama M, Sato H, Saito R, Ikematsu H, Xu H, Takashita E, Tashiro M, Takao S, et al. Evaluation of influenza virus A/H3N2 and B vaccines on the basis of cross-reactivity of postvaccination human serum antibodies against influenza viruses A/H3N2 and B isolated in MDCK cells and embryonated hen eggs. Clin Vac Immunol 2012; 19:897-908; PMID:22492743; http://dx.doi.org/10.1128/CVI.05726-11
  • Barrett PN, Mundt W, Kistner O, Howard MK. Vero cell platform in vaccine production: moving towards cell culture-based viral vaccines. Expert Rev Vaccines 2009; 8:607-18; PMID:19397417; http://dx.doi.org/10.1586/erv.09.19
  • Desmyter J, Melnick JL, Rawls WE. Defectiveness of interferon production and of rubella virus interference in a line of African green monkey kidney cells (Vero). J Virol 1968; 2:955-61; PMID:4302013
  • Govorkova EA, Kaverin NV, Gubareva LV, Meignier B, Webster RG. Replication of influenza A viruses in a green monkey kidney continuous cell line (Vero). J Infect Dis 1995; 172:250-3; PMID:7797924; http://dx.doi.org/10.1093/infdis/172.1.250
  • Kistner O, Barrett PN, Mundt W, Reiter M, Schober-Bandixen S, Dorner F. Development of a mammalian cell (Vero) derived candidate influenza virus vaccine. Vaccine 1998; 16:960-8; PMID:9682344; http://dx.doi.org/10.1016/S0264-410X(97)00301-0
  • Zhou F, Zhou J, Ma L, Song S, Zhang X, Li W, Jiang S, Wang Y, Liao G. High-yield production of a stable Vero cell-based vaccine candidate against the highly pathogenic avian influenza virus H5N1. Biochem Biophys Res Commun 2012; 421:850-4; PMID:22554519; http://dx.doi.org/10.1016/j.bbrc.2012.04.101
  • Kistner O, Barrett PN, Mundt W, Reiter M, Schober-Bendixen S, Eder G, Dorner F. Development of a Vero cell-derived influenza whole virus vaccine. Dev Biol Stand 1999; 98:101-10; discussion 111; PMID:10494963
  • Kistner O, Howard MK, Spruth M, Wodal W, Brühl P, Gerencer M, Crowe BA, Savidis-Dacho H, Livey I, Reiter M, et al. Cell culture (Vero) derived whole virus (H5N1) vaccine based on wild-type virus strain induces cross-protective immune responses. Vaccine 2007; 25:6028-36; PMID:17614165; http://dx.doi.org/10.1016/j.vaccine.2007.05.013
  • Whiteley A, Major D, Legastelois I, Campitelli L, Donatelli I, Thompson CI, Zambon MC, Wood JM, Barclay WS. Generation of candidate human influenza vaccine strains in cell culture – rehearsing the European response to an H7N1 pandemic threat. Infl Other Resp Viruses 2007; 1:157-66; PMID:19432631; http://dx.doi.org/10.1111/j.1750-2659.2007.00022.x
  • Howard MK, Kistner O, Barrett PN. Pre-clinical development of cell culture (Vero)-derived H5N1 pandemic vaccines. Biol Chem 2008; 389:569-77; PMID:18953724; http://dx.doi.org/10.1515/BC.2008.060
  • Kistner O, Crowe BA, Wodal W, Kerschbaum A, Savidis-Dacho H, Sabarth N, Falkner FG, Mayerhofer I, Mundt W, Reiter M, et al. A whole virus pandemic influenza H1N1 vaccine is highly immunogenic and protective in active immunization and passive protection mouse models. PLoS One 2010; 5:e9349; PMID:20186321; http://dx.doi.org/10.1371/journal.pone.0009349
  • Bruhl P, Kerschbaum A, Kistner O, Barrett N, Dorner F, Gerencer M. Humoral and cell-mediated immunity to Vero cell-derived influenza vaccine. Vaccine 2000; 19:1149-58; PMID:11137251; http://dx.doi.org/10.1016/S0264-410X(00)00316-9
  • Ehrlich HJ, Muller M, Oh HM, Tambyah PA, Joukhadar C, Montomoli E, Fisher D, Berezuk G, Fritsch S, Löw-Baselli A, et al. A clinical trial of a whole-virus H5N1 vaccine derived from cell culture. N Engl J Med 2008; 358:2573-84; PMID:18550874; http://dx.doi.org/10.1056/NEJMoa073121
  • Ehrlich HJ, Muller M, Fritsch S, Zeitlinger M, Berezuk G, Loew-Baselli A, van der Velden MV, Pollabauer EM, Maritsch F, Pavlova BG, et al. A cell culture (Vero)-derived H5N1 whole-virus vaccine induces cross-reactive memory responses. J Infect Dis 2009; 200:1113-8; PMID:19712040; http://dx.doi.org/10.1086/605608
  • Keitel WA, Keitel WA, Dekker CL, Mink C, Campbell JD, Edwards KM, Patel SM, Ho DY, Talbot HK, Guo K, et al. Safety and immunogenicity of inactivated, Vero cell culture-derived whole virus influenza A/H5N1 vaccine given alone or with aluminum hydroxide adjuvant in healthy adults. Vaccine 2009; 27:6642-8; PMID:19773098; http://dx.doi.org/10.1016/j.vaccine.2009.03.015
  • Tambyah PA, Wilder-Smith A, Pavlova BG, Barrett PN, Oh HM, Hui DS, Yuen KY, Fritsch S, Aichinger G, Loew-Baselli A, et al. Safety and immunogenicity of two different doses of a Vero cell-derived, whole virus clade H5N1 (A/Indonesia/05/2005) influenza vaccine. Vaccine 2012; 30:329-35; PMID:22080174; http://dx.doi.org/10.1016/j.vaccine.2011.10.088
  • van der Velden MV, Aichinger G, Pollabauer EM, Loew-Baselli A, Fritsch S, Benamara K, Kistner O, Muller M, Zeitlinger M, Kollaritsch H, et al. Cell culture (Vero cell) derived whole-virus non-adjuvanted H5N1 influenza vaccine induces long-lasting cross-reactive memory immune response: homologous or heterologous booster response following two dose or single dose priming. Vaccine 2012; 30:6127-35; PMID:22884662; http://dx.doi.org/10.1016/j.vaccine.2012.07.077
  • van der Velden MV, Fritz R, Pöllabauer EM, Portsmouth D, Howard MK, Kreil TR, Dvorak T, Fritsch S, Vesikari T, Diez-Domingo J, et al. Safety and immunogenicity of a Vero cell culture-derived whole-virus influenza A(H5N1) vaccine in a pediatric population. J Infect Dis 2014; 209:12-23; PMID:24041789; http://dx.doi.org/10.1093/infdis/jit498
  • Fritz R, Sabarth N, Kiermayr S, Hohenadl C, Howard MK, Ilk R, Kistner O, Ehrlich HJ, Barrett PN, Kreil TR. A Vero cell-derived whole-virus H5N1 vaccine effectively induces neuraminidase-inhibiting antibodies. J Infect Dis 2012; 205:28-34; PMID:22090447; http://dx.doi.org/10.1093/infdis/jir711
  • Crowe BA, Bruhl P, Gerencer M, Schwendinger MG, Pilz A, Kistner O, Koelling-Schlebusch K, Aichinger G, Singer J, Zeitlinger M, et al. Evaluation of the cellular immune responses induced by a non-adjuvanted inactivated whole virus A/H5N1/VN/1203 pandemic influenza vaccine in humans. Vaccine 2010; 29:166-73; PMID:21055500; http://dx.doi.org/10.1016/j.vaccine.2010.10.065
  • Howard MK, Sabarth N, Savidis-Dacho H, Portsmouth D, Kistner O, Kreil TR, Ehrlich HJ, Barrett PN. H5N1 whole-virus vaccine induces neutralizing antibodies in humans which are protective in a mouse passive transfer model. PLoS One 2011; 6:e23791; PMID:21876771; http://dx.doi.org/10.1371/journal.pone.0023791
  • Ehrlich HJ, Berezuk G, Fritsch S, Aichinger G, Singer J, Portsmouth D, Hart MK, El-Amin W, Kistner O, Barrett PN. Clinical development of a Vero cell culture-derived seasonal influenza vaccine. Vaccine 2012; 30:4377-86; PMID:22172502; http://dx.doi.org/10.1016/j.vaccine.2011.11.114
  • Ehrlich HJ, Muller M, Kollaritsh H, Pinl F, Schmitt B, Zeitlinger M, Loew-Baselli A, Kreil TR, Kistner O, Portsmouth D, Fritsch S, et al. Pre-vaccination immunity an immune responses to a cell culture-derived whole-virus H1N1 vaccine and similar to a seasonal influenza vaccine. Vaccine 2012; 30:4543-51; PMID:22475864; http://dx.doi.org/10.1016/j.vaccine.2012.03.061
  • Ehrlich HJ, Singer J, Berezuk G, Fritsch S, Aichinger G, Hart MK, El-Amin W, Portsmouth D, Kistner O, Barrett PN. A cell culture-derived influenza vaccine provides consistent protection against infection and reduces the duration and severity of disease in infected individuals. Clin Infect Dis 2012; 54:946-54; PMID:22267715; http://dx.doi.org/10.1093/cid/cir959
  • Loew-Baselli A, Pavlova BG, Fritsch S, Poellabauer EM, Draxler W, Kistner O, Behre U, Angemayr R, Neugebauer J, Kirsten K, et al. A non-adjuvanted whole-virus H1N1 pandemic vaccine is well tolerated and highly immunogenic in children and adolescents and induces substantial immunological memory. Vaccine 2012; 30:5956-66; PMID:22846396; http://dx.doi.org/10.1016/j.vaccine.2012.07.039
  • Waddington CS, Walker WT, Oeser C, Reiner A, John T, Wilkins S, Casey M, Eccleston PE, Allen RJ, Okike I, et al. Safety and immunogenicity of AS03B adjuvanted split virion versus non-adjuvanted whole virion H1N1 influenza vaccine in UK children aged 6 months-12 years: open label, randomised, parallel group, multicentre study. BMJ 2010; 340:c2649; PMID:20508026; http://dx.doi.org/10.1136/bmj.c2649
  • Barrett PN, Berezuk G, Fritsch S, Aichinger G, Hart MK, El-Amin W, Kistner O, Ehrlich HJ. Efficacy, safety, and immunogenicity of a Vero-cell-culture-derived trivalent influenza vaccine: a multicentre, double-blind, randomised, placebo-controlled trial. Lancet 2011; 377:751-9; PMID:21329971; http://dx.doi.org/10.1016/S0140-6736(10)62228-3
  • Frank AL, Couch RB, Griffis CA, Baxter BD. Comparison of different tissue cultures for isolation and quantitation of influenza and parainfluenza viruses. J Clin Microbiol 1979; 10:32-6; PMID:227920
  • Schepetuik SK, Kok T. The use of MDCK, MEK, and LLCMK2 cell lines with enzyme immunoassay for the isolation of influenza and parainfluenza viruses from clinical specimens. J Virol Methods 1993; 42:241-50; PMID:8390473; http://dx.doi.org/10.1016/0166-0934(93)90036-Q
  • Reina J, Fernandez-Baca V, Blanco I, Munar M. Comparison of Madin-Darby canine kidney cells (MDCK) with a green monkey continuous cell line (Vero) and human lung embryonated cells (MRC-5) in the isolation of influenza A virus from nasopharyngeal aspirates by shell vial culture. J Clin Microbiol 1997; 35:1900-1; PMID:9196221
  • Romanova J, Katinger D, Ferko B, Voglauer R, Mochalova L, Bovin N, Lim W, Katinger H, Egorov A. Distinct host range of influenza H3N2 virus isolates in Vero and MDCK cells is determined by cell specific glycosylation pattern. Virology 2003; 307:90-7; PMID:12667817; http://dx.doi.org/10.1016/S0042-6822(02)00064-8
  • Seitz C, Frensing T, Höper D, Kochs G, Reichl U. High yields of influenza A virus in Madin-Darby canine kidney cells are promoted by an insufficient interferon-induced antiviral state. J Gen Virol 2010; 91:1754-63; PMID:20357039; http://dx.doi.org/10.1099/vir.0.020370-0
  • Frensing T, Seitz C, Heynisch B, Patzina C, Kochs G, Reichl U. Efficient influenza B virus propagation due to deficient interferon-induced antiviral activity in MDCK cells. Vaccine 2011; 29:7125-9; PMID:21651940; http://dx.doi.org/10.1016/j.vaccine.2011.05.069
  • Seitz C, Isken B, Heynisch B, Rettkowski M, Frensing T, Reichl U. Trypsin promotes efficient influenza vaccine production in MDCK cells by interfering with the antiviral host response. Appl Microbiol Biotechnol 2012; 93:601-11; PMID:21915610; http://dx.doi.org/10.1007/s00253-011-3569-8
  • Liu J, Shi X, Schwartz R, Kemble G. Use of MDCK cells for production of live attenuated influenza vaccine. Vaccine 2009; 27:6460-3; PMID:19559113; http://dx.doi.org/10.1016/j.vaccine.2009.06.024
  • Hussain AI, Cordeiro M, Sevilla E, Liu J. Comparison of egg and high yielding MDCK cell-derived live attenuated influenza virus for commercial production of trivalent influenza vaccine: in vitro cell susceptibility and influenza virus replication kinetics in permissive and semi-permissive cells. Vaccine 2010; 28:3848-55; PMID:20307595; http://dx.doi.org/10.1016/j.vaccine.2010.03.005
  • van Wielink R, Kant-Eenbergen HC, Harmsen MM, Martens DE, Wijffels RH, Coco-Martin JM. Adaptation of a Madin-Darby canine kidney cell line to suspension growth in serum-free media and comparison of its ability to produce avian influenza virus to Vero and BHK21 cell lines. J Virol Methods 2011; 171:53-60; PMID:20933017; http://dx.doi.org/10.1016/j.jviromet.2010.09.029
  • Zhai W, Zhang DN, Mai C, Choy J, Jian G, Sra K, Galinski MS. Comparison of different cell substrates on the measurement of human influenza virus neutralizing antibodies. PLoS One 2012; 7:e52327; PMID:23284988; http://dx.doi.org/10.1371/journal.pone.0052327
  • Merten OW, Hannoun C, Manuguerra JC, Ventre F, Petres S. Production of influenza virus in cell cultures for vaccine preparation. Adv Exp Med Biol 1996; 397:141-51; PMID:8718593; http://dx.doi.org/10.1007/978-1-4899-1382-1_19
  • Genzel Y, Dietzsch C, Rapp E, Schwarzer J, Reichl U. MDCK and Vero cells for influenza virus vaccine production: a one-to-one comparison up to lab-scale bioreactor cultivation. Appl Microbiol Biotechnol 2010; 88:461-75; PMID:20617311; http://dx.doi.org/10.1007/s00253-010-2742-9
  • Youil R, Su Q, Toner TJ, Szymkowiak C, Kwan WS, Rubin B, Petrukhin L, Kiseleva I. Shaw AR, DiStefano D. Comparative study of influenza virus replication in Vero and MDCK cell lines. J Virol Methods 2004; 120:23-31; PMID:15234806; http://dx.doi.org/10.1016/j.jviromet.2004.03.011
  • Makizumi K, Kimachi K, Fukada K, Nishimura T, Kudo Y, Goto S, Odagiri T, Tahiro M, Kino Y. Timely production of A/Fujian-like influenza vaccine matching the 2003-2004 epidemic strain may have been possible using Madin-Darby canine kidney cells. Vaccine 2008; 26:6852-58; PMID:19149013; http://dx.doi.org/10.1016/j.vaccine.2008.09.075
  • Nishiyama K, Sgawara K, Nouchi T, Kawano N, Soejima K, Abe S, Mizokami H. Purification and cDNA cloning of a novel protease inhibitor secreted into culture supernatant by MDCK cells. Biologics 2008; 36:122-33; PMID:17892946; http://dx.doi.org/10.1016/j.biologicals.2007.07.004
  • Polymenidou M, Trusheim H, Stallmach L, Moos R, Julius C, Miele G, Lenz-Bauer C, Aguzzi A. Canine MDCK cell lines are refractory to infection with human and mouse prions. Vaccine 26:2601-14; PMID:18423803; http://dx.doi.org/10.1016/j.vaccine.2008.03.035
  • Wanich N, Bencharitiwong R, Tsai T, Nowak-Wegrzyn A. In vitro assessment of the allergenicity of a novel influenza vaccine produced in dog kidney cells in individuals with dog allergy. Ann Allergy Asthma Immunol 2010; 104:426-33; PMID:20486334; http://dx.doi.org/10.1016/j.anai.2010.03.012
  • Bencharitiwong R, Leonard S, Tsai T, Nowak-Węgrzyn A. In vitro assessment of the allergenicity of novel MF59-adjuvanted pandemic H1N1 influenza vaccine produced in dog kidney cells. Hum Vaccin Immunother 2012; 8:863-5; PMID:22777093; http://dx.doi.org/10.4161/hv.20324
  • Merten OW, Manuguerra JC, Hannoun C, van der Werf S. Production of influenza virus in serum-free mammalian cell cultures. Dev Biol Stand 1999; 98:23-37; PMID:10494957
  • Voeten JT, Claas EC, Brands R, Palache AM, van Scharrenburg GJ, Rimmelzwaan GF, Osterhaus AD. Generation and characterization of reassortant influenza A viruses propagated in serum-free cultured MDCK-SF1 cells. Dev Biol Stand 1999; 98:77-87; PMID:10494961
  • Tree JA, Richardson C, Fooks AR, Clegg JC, Looby D. Comparison of large-scale mammalian cell culture systems with egg culture for the production of influenza virus A vaccine strains. Vaccine 2001; 19:3444-50; PMID:11348709; http://dx.doi.org/10.1016/S0264-410X(01)00053-6
  • Genzel Y, Reichl U. Continuous cell lines as a production system for influenza vaccines. Expert Rev Vaccines 2009; 8:1681-92; PMID:19943763; http://dx.doi.org/10.1586/erv.09.128
  • Genzel Y, Vogel T, Buck J, Behrendt I, Ramirez DV, Schiedner G, Jordan I, Reichl U. High cell density cultivations by alternating tangential flow (ATF) perfusion for influenza A virus production using suspension cells. Vaccine 2014; 32:2770-81; PMID:24583003; http://dx.doi.org/10.1016/j.vaccine.2014.02.016
  • George M, Farooq M, Dang T, Cortes B, Liu J, Maranga L. Production of cell culture (MDCK) derived live attenuated influenza vaccine (LAIV) in a fully disposable platform process. Biotechnol Bioeng 2010; 106:906-17; PMID:20589670; http://dx.doi.org/10.1002/bit.22753
  • Sun B, Yu X, Kong W, Sun S, Yang P, Zhu C, Zhang H, Wu Y, Chen Y, Shi Y, et al. Production of influenza H1N1 vaccine from MDCK cells using a novel disposable packed-bed bioreactor. Appl Microbiol Biotechnol 2013; 97:1063-70; PMID:22945265; http://dx.doi.org/10.1007/s00253-012-4375-7
  • Tapia F, Vogel T, Genzel Y, Behrendt I, Hirschel M, Gangemi JD, Reichl U. Production of high-titer human influenza A virus with adherent and suspension MDCK cells cultured in a single-use hollow fiber bioreactor. Vaccine 2014; 32:1003-11; PMID:24269322; http://dx.doi.org/10.1016/j.vaccine.2013.11.044
  • Lohr V, Genzel Y, Behrendt I, Scharfenberg K, Reichl U. A new MDCK suspension line cultivated in a fully defined medium in stirred-tank and wave bioreactor. Vaccine 2010; 28:6256-64; PMID:20638458; http://dx.doi.org/10.1016/j.vaccine.2010.07.004
  • Vlecken DH, Plegrim RP, Ruminski S, Bakker WA, van der Pol LA. Comparison of initial feasibility of host cell lines for viral vaccine production. J Virol Methods 2013; 193:28-41; PMID:23684847; http://dx.doi.org/10.1016/j.jviromet.2013.04.020
  • Palache AM, Brands R, van Scharrenburg GJ. Immunogenicity and reactogenicity of influenza subunit vaccines produced in MDCK cells or fertilized chicken eggs. J Infect Dis 1997; 176:S20-S23; PMID:9240689; http://dx.doi.org/10.1086/514169
  • Halperin SA, Nestruck AC, Eastwood BJ. Safety and immunogenicity of a new influenza vaccine growth in mammalian cell culture. Vaccine 1998; 16:1331-5; PMID:9682398; http://dx.doi.org/10.1016/S0264-410X(98)00011-5
  • Percheson PB, Trepanier P, Dugre R, Mabrouk T. A Phase I, randomized controlled clinical trial to study the reactogenicity and immunogenicity of a new split influenza vaccine derived from a non-tumorigenic cell line. Dev Biol Stand 1999; 98:127-132; PMID:10494965
  • Halperin SA, Smith B, Mabrouk T, Germain M, Trepanier P, Hassell T, Treanor J, Gauthier R, Mills EL. Safety and immunogenicity of a trivalent, inactivated, mammalian cell culture-derived influenza vaccine in healthy adults, seniors, and children. Vaccine 2002; 20:1240-7; PMID:11803087; http://dx.doi.org/10.1016/S0264-410X(01)00428-5
  • Ambrozaitis A, Groth N, Bugarini R, Sparacio V, Podda A, Lattanzi M. A novel mammalian cell-culture technique for consistent production of a well-tolerated and immunogenic trivalent subunit influenza vaccine. Vaccine 2009; 27:6022-9; PMID:19666152; http://dx.doi.org/10.1016/j.vaccine.2009.07.083
  • Groth N, Montomoli E, Gentile C, Manini I, Bugarini R, Podda A. Safety, tolerability and immunogenicity of a mammalian cell-culture-derived influenza vaccine: a sequential Phase I and Phase II clinical trial. Vaccine 2009; 27:786-91; PMID:19027046; http://dx.doi.org/10.1016/j.vaccine.2008.11.003
  • Reisinger KS, Block SL, Izu A, Groth N, Holmes SJ. Subunit influenza vaccines produced from cell culture or in embryonated chicken eggs: comparison of safety, reactogenicity, and immunogenicity. J Infect Dis 2009; 200:849-57; PMID:19673652; http://dx.doi.org/10.1086/605506
  • Szymczakiewicz-Multanowska A, Groth N, Bugarini R, Lattanzi M, Casula D, Hilbert A, Tsai T, Podda A. Safety and immunogenicity of a novel influenza subunit vaccine produced in mammalian cell culture. J Infect Dis 2009; 200:841-8; PMID:19673651; http://dx.doi.org/10.1086/605505
  • Frey S, Vesikari T, Szymczakiewicz-Multanowska A, Lattanzi M, Izu A, Groth N, Holmes S. Clinical efficacy of cell culture–derived and egg‐derived inactivated subunit influenza vaccines in healthy adults. Clin Infect Dis 2010; 51:997-1004; PMID:20868284; http://dx.doi.org/10.1086/656578
  • Vesikari T, Block SL, Guerra F, Lattanzi M, Holmes S, Izu A, Gaitatzis N, Hilbert AK, Groth N. Immunogenicity, safety and reactogenicity of a mammalian cell-culture-derived influenza vaccine in healthy children and adolescents three to seventeen years of age. Pediatr Infect Dis J 2012; 31:494-500; PMID:22301476; http://dx.doi.org/10.1097/INF.0b013e31824bb179
  • Oxford JS, Al-Jabri AA, Lambkin R, Palache AM, Fleming DM. Non-responders to egg grown influenza vaccine seroconvert after booster immunization with MDCK cell grown vaccine. Vaccine 2003; 21:2743-6; PMID:12798612; http://dx.doi.org/10.1016/S0264-410X(03)00214-7
  • Fukase H, Furuie H, Yasuda Y, Komatsu R, Matsushita K, Minami T, Suehiro Y, Yotsuvanagi H, Kusadokoro H, Sawata H, et al. Assessment of the immunogenicity and safety of varying doses of an MF59®-adjuvanted cell culture-derived A/H1N1 pandemic influenza vaccine in Japanese paediatric, adult and elderly subjects. Vaccine 2012; 30:5030-7; PMID:22472791; http://dx.doi.org/10.1016/j.vaccine.2012.03.053
  • Hatz C, Cramer JP, Vertruyen A, Schwarz TF, von Sonnenburg F, Borkowski A, Lattanzi M, Hilbert AK, Cioppa GD, Leroux-Roels G. A randomised, single-blind, dose-range study to assess the immunogenicity and safety of a cell-culture-derived A/H1N1 influenza vaccine in adult and elderly populations. Vaccine 2012; 30:4820-7; PMID:22626675; http://dx.doi.org/10.1016/j.vaccine.2012.05.013
  • Reynales H, Astudillo P, de Valliere S, Hatz C, Schlagenhauf P, Rath B, Velentgas P, Farina A, Sales-Carmona V, Groth N. A prospective observational safety study on MF59® adjuvanted cell culture-derived vaccine, Celtura® during the A/H1N1 (2009) influenza pandemic. Vaccine 2012; 30:6436-43; PMID:22902681; http://dx.doi.org/10.1016/j.vaccine.2012.08.005
  • Vinnemeier CD, Fischer-Herr J, Meyer S, Liebig K, Thee⇓ W, Burchard GD, Cramer JP. Immunogenicity and safety of an inactivated 2012/2013 trivalent influenza vaccine produced in mammalian cell culture (Optaflu®): an open label, uncontrolled study. Hum Vaccin Immunother 2014; 10:441-8; PMID:24240428; http://dx.doi.org/10.4161/hv.27140
  • Pan SC, Kung HC, Kao TM, Wu H, Dong SX, Hu MH, Chou AH, Chong P, Hsieh SM, Chang SC. The Madin-Darby canine kidney cell culture derived influenza A/H5N1 vaccine: a phase I trial in Taiwan. J Microbiol Immunol Infect 2013; 46:448-55; PMID:23022464; http://dx.doi.org/10.1016/j.jmii.2012.08.002
  • Clark TW, Pareek M, Hoschler K, Dillon H, Nicholson KG, Groth N, Stephenson I. 2009. Trial of influenza A (H1N1) 2009 monovalent MF59-adjuvanted vaccine – preliminary report. New Engl J Med 2009; 361:2424-35; PMID:19745215; http://dx.doi.org/10.1056/NEJMoa0907650
  • Loebermann M, Voss U, Meyer S, Bosse D, Fritzche C, Klammt S, Frimmel S, Riebold D, Reisinger EC. Clinical trial to evaluate the safety and immunogenicity of a trivalent surface antigen seasonal influenza vaccine produced in mammalian cell culture and administered to young and elderly adults with and without A(H1N1) pre-vaccination. PLoS One 2013; 8:e70866; PMID:23976960; http://dx.doi.org/10.1371/journal.pone.0070866
  • Basavaraj VH, Sampath G, Hegde NR, Mohan VK, Ella KM. Evaluation of safety and immunogenicity of HNVAC, an MDCK-based H1N1 pandemic influenza vaccine, in Phase I single centre and Phase II/III multi-centre, double-blind, randomized, placebo-controlled, parallel assignment studies. Vaccine 2014; 32:4592-7; PMID:24856782; http://dx.doi.org/10.1016/j.vaccine.2014.05.039
  • Yasuda Y, Komatsu R, Matsushita K, Minami T, Suehiro Y, Sawata H, Nakura N, Jaeger RK, Lattanzi M. Comparison of half and full doses of an MF59-adjuvanted cell culture-derived A/H1N1v vaccine in Japanese children. Adv Ther 2010; 27:444-57; PMID:20586002; http://dx.doi.org/10.1007/s12325-010-0043-4
  • Hatz C, von Sonnenburg F, Casula D, Lattanzi M, Leroux-Roels G. A randomized clinical trial to identify the optimal antigen and MF59® adjuvant dose of a monovalent A/H1N1 pandemic influenza vaccine in healthy adult and elderly subjects. Vaccine 2012; 30:3470-7; PMID:22446638; http://dx.doi.org/10.1016/j.vaccine.2012.03.017
  • Reisinger KS, Holmes SJ, Pedotti P, Arora AK, Lattanzi M. A dose-ranging study of MF59®-adjuvanted and non-adjuvanted A/H1N1 pandemic influenza vaccine in young to middle-aged and older adult populations to assess safety, immunogenicity, and antibody persistence one year after vaccination. Hum Vaccin Immunother 2014; 10:(8):2395-407; [Epub ahead of print]; PMID:25424947; http://dx.doi.org/10.4161/hv.29393
  • Heldens J, Hulskotte E, Voeten T, Breedveld B, Verwey P, van Duijnhoven W, Rudenko L, van Damme P, van den Bosch H. Safety and immunogenicity in man of a cell culture derived trivalent live attenuated seasonal influenza vaccine: a Phase I dose escalating study in healthy volunteers. Vaccine 2014; 32:5118-24; PMID:24858566; http://dx.doi.org/10.1016/j.vaccine.2014.05.030
  • Fallaux FJ, Bout A, van der Velde I, van den Wollenberg DJ, Hehir KM, Keegan J, Auger C, Cramer SJ, van Ormondt H, van der Eb AJ, et al. New helper cells and matched early region 1-deleted adenovirus vectors prevent generation of replication-competent adenoviruses. Hum Gene Ther 1998; 9:1909-17; PMID:9741429; http://dx.doi.org/10.1089/hum.1998.9.13-1909
  • Lewis JA, Brown EL, Duncan PA. Approaches to the release of a master cell bank of PER.C6 cells; a novel cell substrate for the manufacture of human vaccines. Dev Biol (Basel) 2006; 123:165-76; discussion 183-97; PMID:16566444
  • Koudstaal W, Hartgroves L, Havenga M, Legastelois I, Ophorst C, Sieuwerts M, Zuijdgeest D, Vogels R, Custers J, de Boer-Luijtze E, et al. Suitability of PER.C6 cells to generate epidemic and pandemic influenza vaccine strains by reverse genetics. Vaccine 2009; 27:2588-93; PMID:19428865; http://dx.doi.org/10.1016/j.vaccine.2009.02.033
  • Pau MG, Ophorst C, Koldijk MH, Schouten G, Mehtali M, Uytdehaag F. The human cell line PER.C6 provides a new manufacturing system for the production of influenza vaccines. Vaccine 2001; 19:2716-21; PMID:11257414; http://dx.doi.org/10.1016/S0264-410X(00)00508-9
  • Hartgroves LC, Koudstaal W, McLeod C, Moncorgé O, Thompson CI, Ellis J, Bull C, Havenga MJ, Goudsmit J, Barclay WS. Rapid generation of a well-matched vaccine seed from a modern influenza A virus primary isolate without recourse to eggs. Vaccine 2010; 28:2973-9; PMID:20188682; http://dx.doi.org/10.1016/j.vaccine.2010.02.012
  • Cox RJ, Madhun AS, Hauge S, Sjursen H, Major D, Kuhne M, Höschler K, Saville M, Vogel FR, Barclay W, et al. A phase I clinical trial of a PER.C6 cell grown influenza H7 virus vaccine. Vaccine 2009; 27:1889-97; PMID:19368768; http://dx.doi.org/10.1016/j.vaccine.2009.01.116
  • Graham FL, Smiley J, Russell WC, Nairn R. Characteristics of a human cell line transformed by DNA from human adenovirus type 5. J Gen Virol 1977; 36:59-74; PMID:886304; http://dx.doi.org/10.1099/0022-1317-36-1-59
  • Le Ru A, Jacob D, Transfiguracion J, Ansorge S, Henry O, Kamen AA. Scalable production of influenza virus in HEK-293 cells for efficient vaccine manufacturing. Vaccine 2010; 28: 3661-71; PMID:20347632; http://dx.doi.org/10.1016/j.vaccine.2010.03.029
  • Lohr V, Genzel Y, Jordan I, Katinger D, Mahr S, Sandig V, Reichl U. Live attenuated influenza viruses produced in suspension process with avail AGE1.CR.pIX cells. BMC Biotechnol 2012; 12:79; PMID:23110398; http://dx.doi.org/10.1186/1472-6750-12-79
  • Jordan I, Vos A, Beilfuss S, Neubert A, Breul S, Sandig V. An avian cell line designed for production of highly attenuated viruses. Vaccine 2009; 27:748-56; PMID:19071186; http://dx.doi.org/10.1016/j.vaccine.2008.11.066
  • Lohr V, Hadicke O, Genzel Y, Jordan I, Buntemeyer H, Klamt S, Reichl U. The avian cell line AGE1.CR.pIX characterized by metabolic flux analysis. BMC Biotechnol 2014; 14:72; PMID:25077436; http://dx.doi.org/10.1186/1472-6750-14-72
  • Brown SW, Mehtali M. The avian EB66R cell line, application to vaccines, and therapeutic protein production. PDA J Pharm Sci Technol 2010; 64:419-25; PMID:21502045
  • Smith KA, Colvin CJ, Weber PS, Spatz SJ, Coussens PM. High titer growth of human and avian influenza viruses in an immortalized chick embryo cell line without the need for exogenous proteases. Vaccine 2008; 26:3778-82; PMID:18524432; http://dx.doi.org/10.1016/j.vaccine.2008.04.048
  • Coussens PM, Smith KA, Weber PS, Colvin CJ. Immortalized chick embryo cell line adapted to serum-free growth conditions and capable of replicating human and reassortant H5N1 influenza strains for vaccine production. Vaccine 2011; 29:8661-8; PMID:21911025; http://dx.doi.org/10.1016/j.vaccine.2011.08.122
  • Schultz-Cherry S, Dybdahl-Sissoko N, McGregor M, Hinshaw VS. Mink lung epithelial cells: unique cell line that supports influenza A and B virus replication. J Clin Microbiol 1998; 36:3718-20; PMID:9817906
  • Huang YT, Turchek BM. Mink lung cells and mixed mink lung and A549 cells for rapid detection of influenza virus and other respiratory viruses. J Clin Microbiol 2000; 38:422-3; PMID:10618131
  • Hamilton SB, Wyatt DE, Wahlgren BT, O'Dowd MK, Morrissey JM, Daniels DE, Lednicky JA. Higher titers of some H5N1 and recent human H1N1 and H3N2 influenza viruses in Mv1 Lu vs. MDCK cells. Virol J 2011; 8:66; PMID:21314955; http://dx.doi.org/10.1186/1743-422X-8-66
  • Nakamura K, Homma M. Protein synthesis in Vero cells abortively infected with influenza B virus. J Gen Virol 1981; 56:199-202; PMID:7299373; http://dx.doi.org/10.1099/0022-1317-56-1-199
  • Lau SC, Scholtissek C. Abortive infection of Vero cells by an influenza A virus (FPV). Virology 1995; 212:225-31; PMID:7676635; http://dx.doi.org/10.1006/viro.1995.1473
  • Kaverin NV, Webster RG. Impairment of multicycle influenza virus growth in Vero (WHO) cells by loss of trypsin activity. J Virol 1995; 69:2700-3; PMID:7884927
  • Roedig JV, Rapp E, Hoper D, Genzel Y, Reichl U. Impact of host cell line adaptation on quasispecies composition and glycosylation of influenza A virus hemagglutinin. PLoS One 2011; 6:e27989; PMID:22163276; http://dx.doi.org/10.1371/journal.pone.0027989
  • Tseng YF, Hu AY, Huang ML, Yeh WZ, Weng TC, Chen YS, Chong P, Lee MS. Adaptation of high-growth influenza H5N1 vaccine virus in Vero cells: implications for pandemic preparedness. PLoS One 2011; 6:e24057; PMID:22022351; http://dx.doi.org/10.1371/journal.pone.0024057
  • Fodor E, Devenish L, Engelhardt OG, Palese P, Brownlee GG, García-Sastre A. Rescue of influenza A virus from recombinant DNA. J Virol 1999; 73:9679-82; PMID:10516084
  • Ozaki H, Govorkova EA, Li C, Xiong X, Webster RG, Webby RJ. Generation of high-yielding influenza A viruses in African green monkey kidney (Vero) cells by reverse genetics. J Virol 2004; 78:1851-7; PMID:14747549; http://dx.doi.org/10.1128/JVI.78.4.1851-1857.2004
  • Krause PR. Use of MDCK Cells for Manufacture of Inactivated Influenza Vaccines: Introduction. Vaccines and Related Biological Products Advisory Committee, Center for Biologics Evaluation and Research, Taylor & Francis. 2005; www.fda.gov/ohrms/dockets/ac/05/slides/5-4188S1-1draft.PPT
  • Groner A, Vorlop J (inventors); Novartis Vaccines and Diagnostics, Inc., (assignee). Animal cells and processes for the replication of influenza viruses. 1997. Patent No. WO/1997/037000.
  • Liu J, Mani S, Schwartz R, Richman L, Tabor DE. Cloning and assessment of tumorigenicity and oncogenicity of a Madin-Darby canine kidney (MDCK) cell line for influenza vaccine production. Vaccine 2010; 28:1285-93; PMID:19944150; http://dx.doi.org/10.1016/j.vaccine.2009.11.023
  • Omeir RL, Teferedegne B, Foseh GS, Beren JJ, Snoy PJ, Brinster LR, Cook JL, Peden K, Lewis AM Jr. Heterogeneity of the tumorigenic phenotype expressed by Madin-Darby canine kidney cells. Comp Med 2011; 61:243-50; PMID:21819694
  • Vepachedu RS, Menon A, Hussain AI, Liu J. Evaluation of tumorigenic potential of high yielding cloned MDCK cells for live-attenuated influenza vaccine using in vitro growth characteristics, metastatic gene expression and in vivo nude mice model. Biologicals 2012; 40:482-94; PMID:22902973; http://dx.doi.org/10.1016/j.biologicals.2012.06.005
  • Madin-Darby canine kidney continuous cell line. Briefing document, Solvay Pharmaceuticals, Vaccines and Related Biologicals Advisory Committee (VRBPAC), Center for Biologics Evaluation and Research, Taylor & Francis. 2005; http://www.fda.gov/ohrms/dockets/ac/05/briefing/5-4188b1_19a.pdf
  • Use of Madin Darby canine kidney cells for the manufacture of live, attenuated influenza vaccines. Briefing document, MedImmune, Vaccines and Related Biologicals Advisory Committee (VRBPAC), Center for Biologics Evaluation and Research, Taylor & Francis. 2008; http://www.fda.gov/ohrms/dockets/ac/08/briefing/2008-4384B1_4.pdf
  • Onions D, Egan W, Jarrett R, Novicki D, Gregersen JP. Validation of the safety of MDCK cells as a substrate for the production of a cell-derived influenza vaccine. Biologicals 2010; 38:544-51; PMID:20537553; http://dx.doi.org/10.1016/j.biologicals.2010.04.003
  • Hegde NR, Kumar D, Rao PP, Kumari PK, Kaushik Y, Ravikrishnan R, Prasad SD, Ella KM. Development and preclinical testing of HNVAC, a cell culture-based H1N1 pandemic influenza vaccine from India. Vaccine 2014; 32:3636-43; PMID:24801052; http://dx.doi.org/10.1016/j.vaccine.2014.04.072
  • Lewis AM Jr, Krause P, Peden K. A defined-risk approach to the regulatory assessment of the use of neoplastic cells as substrates for viral vaccine manufacture. Dev Biol 2001; 106:513-35; PMID:11761266
  • Medema JK, Meijer J, Kersten AJ, Horton R. Safety assessment of Madin Darby canine kidney cells as vaccine substrate. Dev Biol 2006; 123:243-50; PMID:16566450
  • Gregersen JP. A risk-assessment model to rate the occurrence and relevance of adventitious agents in the production of influenza vaccines. Vaccine 2008; 26:3297-304; PMID:18468737; http://dx.doi.org/10.1016/j.vaccine.2008.03.076
  • Gregersen JP. A quantitative risk assessment of exposure to adventitious agents in a cell culture-derived subunit influenza vaccine. Vaccine 2008; 26:3332-40;PMID:18485545; http://dx.doi.org/10.1016/j.vaccine.2008.03.075
  • Murata H, Macauley J, Lewis AM Jr, Peden K. Plaque purification as a method to mitigate the risk of adventitious-agent contamination in influenza vaccine virus seeds. Vaccine 2011; 29:3155-61; PMID:21354480; http://dx.doi.org/10.1016/j.vaccine.2011.02.041
  • Hess RD, Weber F, Watson K, Schmitt S. Regulatory, biosafety and safety challenges for novel cells as substrates for human vaccines. Vaccine 2012; 30:2715-27; PMID:22342707; http://dx.doi.org/10.1016/j.vaccine.2012.02.015
  • Roth B, Mohr H, Enders M, Garten W, Gregersen JP. Isolation of influenza viruses in MDCK 33016PF cells and clearance of contaminating respiratory viruses. Vaccine 2012; 30:517-22; PMID:22119922; http://dx.doi.org/10.1016/j.vaccine.2011.11.063
  • Ledwith BJ, Lanning CL, Gumprecht LA, Anderson CA, Coleman JB, Gatto NT, Balasubramanian G, Farris GM, Kemp RK, Harper LB, et al. Tumorigenicity assessments of Per.ER C6 cells and of an Ad5-vectored HIV-1 vaccine produced on this continuous cell line. Dev Biol 2006; 123:251-63; discussion 265; PMID:16566451
  • Robertson JS, Cook P, Attwell AM, Williams SP. Replicative advantage in tissue culture of egg-adapted influenza virus over tissue culture-derived virus: implications for vaccine manufacture. Vaccine 1995; 13:1583-8; PMID:8578846; http://dx.doi.org/10.1016/0264-410X(95)00085-F
  • Lin YP, Wharton SA, Martin J, Skehel JJ, Wiley DC, Steinhauer DA. Adaptation of egg-grown and transfectant influenza viruses for growth in mammalian cells: selection of hemagglutinin mutants with elevated pH of membrane fusion. Virology 1997; 233:402-10; PMID:9217063; http://dx.doi.org/10.1006/viro.1997.8626
  • Lee HK, Tang JW, Kong DH, Loh TP, Chiang DK, Lam TT, Koay ES. Comparison of mutation patterns in full-genome A/H3N2 influenza sequences obtained directly from clinical samples and the same samples after a single MDCK passage. PLoS One 2013; 8:e79252; PMID:24223916; http://dx.doi.org/10.1371/journal.pone.0079252
  • Opitz L, Zimmermann A, Lehmann S, Genzel Y, Lübben H, Reichl U, Wolff MW. Capture of cell culture-derived influenza virus by lectins: strain independent, but host cell dependent. J Virol Methods 2008; 154:61-8; PMID:18840469; http://dx.doi.org/10.1016/j.jviromet.2008.09.004
  • Hutter J, Rodig JV, Hoper D, Seeberger PH, Reichl U, Rapp E, Lepenies B. Toward animal cell culture-based influenza vaccine design: viral hemagglutinin N-glycosylation markedly impacts immunogenicity. J Immunol 2013; 190:220-30; PMID:23225881; http://dx.doi.org/10.4049/jimmunol.1201060

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.