Next Article in Journal
Activity of a Carbohydrate-Binding Module Therapy, Neumifil, against SARS-CoV-2 Disease in a Hamster Model of Infection
Next Article in Special Issue
Outreach Efforts to Prevent Newcastle Disease Outbreaks in Southern California
Previous Article in Journal
A Unique Robust Dual-Promoter-Driven and Dual-Reporter-Expressing SARS-CoV-2 Replicon: Construction and Characterization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Development and Scalable Production of Newcastle Disease Virus-Vectored Vaccines for Human and Veterinary Use

Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
*
Author to whom correspondence should be addressed.
Viruses 2022, 14(5), 975; https://doi.org/10.3390/v14050975
Submission received: 22 March 2022 / Revised: 29 April 2022 / Accepted: 2 May 2022 / Published: 6 May 2022
(This article belongs to the Special Issue Newcastle Disease Virus and Other Avian Paramyxoviruses)

Abstract

:
The COVID-19 pandemic has highlighted the need for efficient vaccine platforms that can rapidly be developed and manufactured on a large scale to immunize the population against emerging viruses. Viral-vectored vaccines are prominent vaccine platforms that have been approved for use against the Ebola virus and SARS-CoV-2. The Newcastle Disease Virus is a promising viral vector, as an avian paramyxovirus that infects poultry but is safe for use in humans and other animals. NDV has been extensively studied not only as an oncolytic virus but also a vector for human and veterinary vaccines, with currently ongoing clinical trials for use against SARS-CoV-2. However, there is a gap in NDV research when it comes to process development and scalable manufacturing, which are critical for future approved vaccines. In this review, we summarize the advantages of NDV as a viral vector, describe the steps and limitations to generating recombinant NDV constructs, review the advances in human and veterinary vaccine candidates in pre-clinical and clinical tests, and elaborate on production in embryonated chicken eggs and cell culture. Mainly, we discuss the existing data on NDV propagation from a process development perspective and provide prospects for the next steps necessary to potentially achieve large-scale NDV-vectored vaccine manufacturing.

1. Introduction

Infectious diseases have been an important issue throughout human history, with epidemics affecting entire populations, as well as causing losses in livestock [1]. Recently, due to the COVID-19 pandemic, the world turned its attention to viral diseases and vaccine manufacturing. The pipeline to develop and manufacture new vaccines became a global concern, leading to significantly more resources being leveraged into this field [2]. With this increased demand comes a need for the development and optimization of vaccine platforms—technologies that can rapidly be adapted to target an emerging disease, with minimal changes to the manufacturing process [3].
In this context, vaccines based on RNA and viral vectors have shown to be promising options for vaccine platforms. Both avoid the costly biosafety level 3 manipulation of the target pandemic pathogen and are quickly adaptable by modifying the RNA sequence used or the antigen expressed on the viral vector backbone. RNA vaccines stand out for their fast development and ease of production, comprising a straightforward chemical synthesis, with the approved Moderna and Pfizer-BioNTech vaccines playing an essential role in the race for immunization against SARS-CoV-2 worldwide [4]. However, their lack of stability leads to the challenging requirement of frozen storage (lower than −20 °C), which poses issues with transportation, and hinders access in low-income countries and remote regions which lack the cold chain infrastructure [5]. Live virus vaccines, on the other hand, are commonly freeze-dried to be stored between 2 and 8 °C [6].
Viral-vectored vaccines represent a promising alternative with several approved vaccines, including the adenovirus-vectored vaccines used against SARS-CoV-2 (Oxford-Astrazeneca, Johnson & Johnson, Gamaleya and CanSino) [7], and the vectors used against Ebola virus [8] (adenovirus [9], modified Vaccinia Ankara [6,10] and vesicular stomatitis virus [11]). This type of vaccine is highly versatile, with a wide range of human and non-human viruses being studied as vector candidates [12] and the possibility to genetically engineer each vector to modify the surface proteins, generate chimeric strains and select for the desired characteristics, including thermostability [13,14]. This allows researchers to optimize the balance between immunogenicity and safety by modulating virulence and evading pre-existing immunity. It is also possible to design unique vaccination strategies, such as co-expressing different antigens, for a more robust response or even generating a bivalent vector that provides immunization against multiple pathogens [15,16].
Newcastle Disease Virus (NDV) is an avian virus that has been extensively researched as an oncolytic virus, with a long history of clinical trials for this application [17]. Due to host range restriction, it is not pathogenic in humans, which avoids the issue of pre-existing immunity in the population [1]. As such, it is an ideal candidate for a vaccine vector in terms of safety and immunogenicity, and has been implemented in several studies targeting human and veterinary diseases [16]. These vaccine candidates currently rely on the well-established production process in embryonated chicken eggs (ECEs), which can be a cost-effective way of using existing facilities that produce influenza vaccines to produce large quantities of doses [18]. However, there are very few studies exploring the production of NDV in cell culture, which could be scaled to bioreactor production facilities. Cell culture-based processes provide several advantages over production in eggs: they avoid issues with allergens, eliminate dependence on chicken egg supply, and allow greater control over each operation parameter, which leads to more reproducibility, scalability and optimization of the process [3,19].
This review elaborates on the potential of NDV as a viral vector based on its fundamental biology, outlines the recommendations and limitations for designing recombinant NDV constructs, and summarizes the history of developed vaccine candidates for human and veterinary use, including the strains and routes of administration used, as well as highlighting innovative strategies. Lastly, we discuss what has been done in terms of viral propagation and process development, and how this could potentially be leveraged for scalable manufacturing in bioreactors.

2. Characteristics of NDV as a Viral Vector

The Newcastle Disease Virus is an avian paramyxovirus that stands out as a promising viral vector based on many advantageous characteristics. Although NDV poses a concern in the poultry industry for the neurological and respiratory disease it can cause among chicken [1], this virus typically does not lead to disease in humans [20]. Only a few cases of conjunctivitis have been reported among those who work closely with poultry or virus samples [21,22,23,24]. This avoids the issue of pre-existing immunity in the population that can arise when using widely spread human viruses as viral vectors [12], such as highly seroprevalent adenoviruses [25]. In addition, NDV replicates efficiently in the respiratory tract, enabling it to be a vector for intranasal vaccines [20,26,27,28,29,30,31,32,33]. This type of vaccine generates both mucosal and systemic immunity against the target disease, which is especially useful for vaccines against highly contagious respiratory diseases, including SARS-CoV-2 [2,33].
Another key aspect of this viral vector is safety. NDV strains are classified into three different pathotypes based on their level of virulence in chicken: lentogenic, mesogenic and velogenic. Lentogenic strains, such as LaSota or B1, show the lowest virulence in chicken [1] and have an extensive documented history of being safe in humans, as has been shown in clinical trials using NDV as an oncolytic agent [34,35]. More detailed information on such trials can be found in the most recent reviews on the oncolytic application of NDV [17,36,37]. Mesogenic and velogenic strains, on the other hand, are typically not used as vaccine vectors due to their virulence in chicken [1] and their status as a “Select Agent” in the United States [13]. As an RNA virus with cytoplasmic replication, NDV also poses very low risk when it comes to the chance of recombination with the host’s DNA, and it has been suggested to lack gene exchange with other viruses [35].
Aside from safety and efficacy, NDV also offers versatility when it comes to production processes. This virus has been produced extensively in embryonated chicken eggs [31,38] and has also shown the capacity of infecting continuous cell lines such as HEK293 [3], Vero [3,20,39], DF-1 [20,38,40], MDCK [41] and HeLa [42]. Although there are very few studies focusing on process development for NDV, it shows the potential to be produced in either egg-based or cell culture-based processes depending on which production facilities are available and which strategies are adopted.

3. Designing and Generating Recombinant NDV

NDV contains an RNA genome which is single-stranded, negative-sense and non-segmented, ranging from 15,186 to 15,198 nucleotides in length [43]. The genome comprises six transcriptional units, encoding a nucleocapsid protein (N), a phosphoprotein (P), a matrix protein (M), a fusion protein (F), a hemagglutinin-neuraminidase protein (HN) and a large polymerase protein (L) [1]. In addition, the RNA of the P gene can be edited to generate the V or W proteins, which are non-structural and generally associated with modulating the avian host’s immune response [44]. Techniques for generating recombinant NDV constructs are already well established, with protocols following the general steps of: antigenomic plasmid construction, transfection, rescue and amplification [45].
It is important to consider the limitations and recommendations for recombinant NDV when designing the antigenome plasmid. As a non-segmented genome, large increases in genome length can impair virus replication, and it has been suggested that the maximum transgene size tolerated by NDV is around 3 kb [46,47] or 5 kb [27,36]. NDV constructs bearing genes encoding the SARS-CoV S protein [28] and the SARS-CoV-2 S protein [27], both around 3.8 kb in size, have been successfully generated, demonstrating this vector’s capacity for inserts in this size range. NDV has also been shown to tolerate multiple transgenes [48,49,50,51,52,53,54,55,56,57,58] using different co-expression strategies [59]. Each foreign gene must be flanked by untranslated regions (UTRs) of NDV genes called gene start (GS) and gene end (GE) sequences (Figure 1). The level of transgene expression varies according to the GS and GE, with the highest expression achieved using UTRs from the M and F genes [60]. Any GS or GE-like sequences within the transgene should be removed through silent mutagenesis [61].
Transgenes are typically inserted in the optimal site between the P and M genes (Figure 1), although other sites can also be used [62], and must follow the “rule of six,” which determines that the genome length should be an even multiple of six. This is important for efficient virus replication, as each nucleocapsid protein monomer covers around six nucleotides, so this rule ensures that the nucleotide sequence can be completely encapsidated [43].
For rescue, the antigenome plasmid containing T7 promoter and terminator sequences must be co-transfected with plasmids expressing the N, P and L genes [45]. The T7 DNA-dependent RNA polymerase is commonly introduced by: (i) infecting the cell with a recombinant virus, such as Modified Vaccinia Ankara; (ii) using a stable cell line expressing this polymerase; or (iii) introducing an additional plasmid expressing the T7 polymerase [63]. After rescue, the virus is typically amplified in embryonated chicken eggs [45] or permissive cell lines [38]. Once amplified, the infectious stock of recombinant virus is ready to be used for further production by infecting embryonated chicken eggs or cell culture.

4. NDV-Vectored Vaccines

4.1. Vaccines for Human Use

NDV has been explored as a vector for human vaccines over the past two decades, with nearly 30 published studies assessing these vaccine candidates in animals (Table 1). Other reviews have listed these studies previously [1,15,64], and an updated table is provided below.
NDV-vectored vaccine candidates have been developed for a range of pathogens, including HIV, EBOV and, predominantly, respiratory viruses such as influenza, SARS-CoV and SARS-CoV-2 (Table 1). Most of the vaccine candidates employed recombinant lentogenic NDV strains as an intranasal live vectored vaccine. The LaSota strain is predominantly used, although other lentogenic strains such as Hitchner B1 and VG/GA, as well as the mesogenic strain Beaudette C (BC), are also present. Interestingly, a few vaccine candidates use chimeric NDV strains, such as the LaSota/VF. This strain is based on a LaSota backbone with the BC strain F protein cleavage sequence, which slightly increases the virulence in birds but allows the virus to replicate in cell culture without the need for added trypsin [28]. In other studies, the mesogenic BC strain was modified by exchanging the ectodomains in the surface glycoproteins F and HN by their equivalents from the LaSota strain [72] or avian paramyxovirus 3 (APMV-3) [13] to reduce virulence and increase safety.
Due to host range restriction, NDV was shown to have attenuated replication in primates, while still generating sufficient mucosal immunity as a respiratory virus [20], demonstrating both safety and immunogenicity. As such, many studies targeting respiratory diseases have implemented the intranasal route of inoculation, including most of the recently developed vaccines targeting SARS-CoV-2 [18,26,27,30,31,76]. Combinations of intranasal and intramuscular doses have also been assessed for several vaccine candidates [30,31,70,76]. Inactivated NDV-vectored vaccines, on the other hand, have been administered exclusively by the intramuscular route, as the inactivated virus can no longer replicate in the mucosal passages [31,75,76,78].
A few studies have used multiple antigens to target the pathogen of concern. Notably, an NDV vector co-expressing the poliovirus P1 and 3CD proteins resulted in the formation of poliovirus viral-like particles (VLPs) in the host cells upon vaccination, which means antigens are presented in a form most similar to the native pathogen while still being safer than live poliovirus vaccines [49,71]. The replication of NDV also serves as a natural adjuvant, increasing the immunogenicity of the vaccine. In another study, NDV was used to co-express the HIV Env and Gag proteins, testing several different orders and positions in the genome [50]. Most constructs in this study also generated VLPs, enhancing the immune response and providing a promising vaccination platform for HIV. Other studies have evaluated the effect of multiple antigens by co-infecting animals with different NDV constructs expressing each antigen separately [69,74], although this approach was less efficient than inoculating a single construct in a study for JEV vaccines [74]. This strategy could avoid issues with slow NDV propagation due to co-expression of multiple transgenes but would require the production of different NDV vectors for the same vaccine, similar to influenza vaccines containing multiple strains [19].
Certain studies have combined different vaccination approaches for a heterologous immunization strategy. One study used an NDV vector expressing the HIV Env (gp160) protein for a priming dose and boosted with purified recombinant proteins (gp120 or gp140), which was the most efficient regimen of vaccination tested [73]. This mixed regimen induced higher magnitude of immune response than the regimen with only NDV-vectored doses, while also providing a longer lasting immune memory in comparison to the purified protein-only regimen. As such, the NDV-vectored prime was important for long-term immunity, while the protein boost enhanced immunogenicity. Another study explored combining different vectors by priming with an adenovirus-vectored vaccine and boosting with an NDV-vectored vaccine, or vice versa [77]. This heterologous regimen induced a more potent and robust response than the homologous alternatives, potentially due to avoiding pre-existing immunity to each vector.
NDV-vectored vaccines have great potential to be used against pandemic diseases, having shown efficient protection against EBOV [13,29,77], SARS-CoV [28] and SARS-CoV-2 [18,26,27,30,31,75,76,78]. Intranasal vaccines against EBOV have been shown to induce neutralizing antibody responses in monkeys [29], guinea pigs [13] and mice [77], although the latter study found more robust responses when mixing adenovirus and NDV-vectored doses as compared to a homologous NDV-vectored regimen. An intranasal vaccine against SARS-CoV [28] also generated a protective antibody response in monkeys and highlighted the need for two doses, as one dose provided insufficient immunogenicity. A similar result was found in a study against SARS-CoV-2 in hamsters [27], in which two doses induced a protective neutralizing response while the single-dose regimen did not significantly reduce viral loads upon infection. Inactivated NDV-vectored SARS-CoV-2 vaccines have also been successful, inducing higher neutralizing responses in mice than a purified protein vaccine [78] and significantly reducing viral loads in a hamster model [75].
Notably, some NDV-vectored SARS-CoV-2 vaccines provided potent protective immunity and reduced viral loads to undetectable amounts on day 4 or 5 post infection, but they did not induce sterilizing immunity, as there were still detectable amounts of virus on day 2 post infection that could potentially lead to shedding [26,27,75]. However, it is considered that widely available vaccines capable of reducing disease severity are highly beneficial in a pandemic, even if sterilizing immunity is not achieved [75].
Although there have been numerous NDV-vectored vaccine candidates tested in animal models (Table 1), only a select few have proceeded to clinical trials. The increased demand for vaccines throughout the COVID-19 pandemic advanced the field, leading to a series of clinical trials for two vaccine candidates (Table 2). A research group based in the Icahn School of Medicine at Mount Sinai (USA) engineered the HexaPro-S (HXP-S) version of the SARS-CoV-2 S protein, which is stabilized in its pre-fusion conformation and anchored in the NDV membrane by containing domains from the NDV F protein [31]. Using the LaSota strain backbone expressing the HXP-S antigen, two vaccine candidates were generated: a live version [18] (“Patria”), in Phase II clinical trials in Mexico, and an inactivated version [75], in Phase I/II clinical trials in Thailand (“HXP-GPOVac”), Vietnam (“COVIVAC”) and Brazil (“ButanVac”) [76]. These vaccines are produced in embryonated chicken eggs (ECEs) in GMP-certified facilities in each country, taking advantage of the existing infrastructure for production of influenza vaccines. The inactivated version uses beta-propiolactone (BPL) for inactivation and CpG 1018 as an adjuvant, while the live version has no adjuvant addition, as the replicative virus is considered self-adjuvanted [76].
Interim results for Phase I clinical trials of the NDV-vectored vaccine against SARS-CoV-2 have been reported using the live version in Mexico [79] and the inactivated version in Thailand [80]. The results showed both versions of the vaccine to be safe in humans. The live version was sufficiently immunogenic at the highest dose tested (108 EID50), and all formulations were safe. Interestingly, adequate immunogenicity was only achieved when doses were administered twice intramuscularly or intramuscularly followed by intranasally. The exclusively intranasal regime was found to generate cellular immunity but lacked a robust systemic antibody response. As such, the high-dose formulations with IM-IM and IN-IM routes were chosen for the Phase II trials [79]. As for the inactivated vaccine candidate, the immunogenicity was proportional to the dosage and was not considerably affected by the use of the adjuvant CpG 1018. The mid-dose (3 μg) was considered sufficiently immunogenic and was chosen for the Phase II trials, with and without CpG 1018 [80].

4.2. Vaccines for Veterinary Use

NDV has been extensively explored as a vector for veterinary vaccines, with over 60 published studies (Table A1) on vaccine candidates over the past two decades mainly targeting use in poultry or cattle. Other review papers have listed and summarized some of these studies [1,15,16,64,81], and an up-to-date table is provided in the Appendix A (Table A1).
Importantly, as virulent NDV strains cause severe disease in chicken and economic loss, the lentogenic strains are often used to vaccinate chicken for protection against virulent NDV. As such, several studies have implemented a bivalent vaccine approach for poultry in which NDV expresses antigens of another avian virus to immunize chicken against both diseases, including highly pathogenic influenza virus (HPAIV) [41,53,55,82,83,84], avian reovirus (ARV) [85], infectious laryngotracheitis virus (ILTV) [86,87] and fowl adenovirus serotype 2 (FAdV-4) [88]. Notably, certain bivalent vaccines against NDV and HPAIV have been licensed for use in poultry [15,81]. Bivalent NDV vaccines have also been developed for ducks against duck Tembusu virus (DTMUV) [89] and HPAIV H5N1 [90]; for geese against goose parvovirus (GPV) [91] and goose astrovirus (GoAstV) [92]; and for turkeys against avian metapneumovirus (AMPV) [52].
Similar to vaccine candidates for human use, several NDV vaccines expressing multiple antigens have been developed for veterinary use either by co-expressing the antigens within the same vector [51,52,53,54,55,56,57,58] or by inoculating different vectors, each expressing a different antigen, in the same formulation [71,86,93,94]. For the latter, two studies have found that a formulation with only one vector/antigen yielded better results and sufficient protection [86,93], while another study found adequate immune responses regardless of inoculating one or multiple vectors [71].
NDV strains have also been engineered to generate chimeric or modified strains with novel characteristics. Thermostability is a key characteristic for poultry immunization through spraying or drinking water, while also relieving difficulties associated with cold chain requirements [14]. To address this, thermostable NDV strains have been isolated and modified to be used as vectors in avian influenza vaccines [14,95]. Another concern in poultry immunization is pre-existing immunity to NDV [64], which led to the development of chimeric NDV strains in which the native surface glycoproteins (F and HN) were substituted by the corresponding genes from another virus, namely APMV-8 [96] or APMV-2 [97,98]. Chimeric strains have also been developed to modulate virulence by substituting the F and HN proteins of the mesogenic BC strain (partly or completely) with the corresponding proteins from the lentogenic LaSota strain, or by modifying basic residues in the F protein cleavage site [32,99].

5. Manufacturing of NDV-Vectored Vaccines

5.1. Workflow for Viral Vector Production

Viral vectors are typically produced in ECEs or cell culture, from which they are harvested and purified for vaccine formulation (Figure 2). NDV has been extensively produced in ECEs for poultry vaccination and for pre-clinical studies, with only a few studies propagating this virus in cell lines (Table 1 and Table A1) and, to our knowledge, only four studies producing it in lab-scale bioreactors [3,100,101,102].
For production in ECEs (Figure 2A), eggs must be acquired and incubated at 37 °C prior to inoculation. They are infected between 9 and 11 days old, and are incubated for another 24 h for viral production. ECEs containing dead embryos before 24 h of incubation are discarded, while embryos that die after that timepoint are stored at 4 to 8 °C for several hours before collecting the allantoic fluid to harvest NDV [103].
For production in cell culture (Figure 2B), bioreactors are inoculated with defined medium and the chosen cell line for the cell growth phase. Critical operation parameters such as pH, oxygen concentration, agitation and temperature are kept constant throughout the entire run. Once the required cell density is reached, cells are infected with NDV to initiate the viral production phase. For batch productions, the bioreactor is interrupted for harvest when the known peak production timepoint is reached, typically between 24 and 48 h post infection (hpi) [3,100].
The processes also differ in terms of waste treatment: infected ECEs result in solid waste that is typically incinerated [104], while reusable bioreactors use methods such as cleaning/sterilization in place and chemical treatment of liquid waste, and single-use bioreactors are disposed of through chemical or physical treatments [105].

5.2. Parameters for NDV Production in Cell Culture

When using lentogenic NDV strains to infect cell lines for propagation, an exogenous protease must be provided to cleave the F protein and activate infection [28]. This can be achieved by providing allantoic fluid at a concentration from 5 to 10% [20,72,106], or by adding TPCK-treated trypsin [3,28], as is done for influenza [107]. Trypsin is more suitable than allantoic fluid due to the variability regarding undefined animal products in cell culture. This is the same reason why serum-free media is preferred over the use of fetal bovine serum (FBS) in industry [108]. Alternatively, there are strains which can replicate without trypsin addition, including mesogenic strains such as BC and R2B, or strains with a modified F cleavage site, such as LaSota/VF [28]. The optimal trypsin concentration to produce recombinant NDV LaSota in suspension Vero cells was found to be 1 μg/mL [3], although NDV constructs expressing certain antigens can become self-sufficient for viral entry and no longer require trypsin [109].
Another key parameter is the multiplicity of infection (MOI), which has been optimized in suspension Vero cells using serum-free media. Our past research showed that MOIs of 0.1, 0.01 and 0.001 resulted in similar peak production titers (around 108 TCID50/mL), while production at an MOI of 0.0001 was approximately 100-fold lower [3]. Replication of NDV in DF-1 and adherent Vero cells is usually achieved with an MOI of 0.01 [58,69,70,72,85,106], which is also within this range. A study using adherent Vero cells with microcarriers in media containing FBS showed similar titers of around 4 × 107 TCID50/mL for MOI 2 and 0.2 [100]. When infecting BHK-21 cells, an MOI of 5 resulted in a peak of around 107 EID50/mL at 24 hpi, while an MOI of 0.01 caused a delay in that peak to 96 hpi [109]. Thus, NDV seems to have wide range of MOIs that achieve adequate production and the optimal MOI can depend on other process and culture parameters, although 0.01 seems to be a suitable MOI for most conditions.
NDV can be adapted to a cell line by serial passaging, which selects for more efficient replication. This facilitates the next infection and can lead to a higher yield, with an observed increase of around 500-fold in lentogenic strains after four passages in suspension Vero cells and 13-fold in suspension HEK293 [3]. Another study found an increase from 5- to 25-fold in lentogenic strains and from 6- to 10-fold in mesogenic strains after eight passages in adherent Vero cells [39].
Recombinant lentogenic NDV production in several cell lines resulted in titers around 107 and 108 infectious units per mL between 30 and 48 hpi, including suspension Vero cells [3] and adherent cells: Vero [100,106], DF-1 [13,72], BHK-21 [109] and MDCK [41]. The highest titers reported were around 109 PFU/mL in DF-1 [70], 5 × 108 PFU/mL in MDCK [41] and 2.37 × 108 TCID50/mL in suspension Vero cells [3]. Production of the mesogenic strain R2B in adherent Vero cells were between 6 × 108 and 6 × 107 TCID50/mL [58,85,110]. The production in ECEs is similar but still overall higher, with titers around 108 and 109 infectious units per mL [13,41,54,67,82,109].

5.3. NDV Production in Lab-Scale Bioreactors

Although there is still a small number of studies available, a few successful productions of NDV in bioreactors have been published. It is important to note that serum-free media is preferred for industrial bioprocesses, as the use of FBS implies an undefined composition and lot-to-lot variability [108]. In addition, suspension cell lines are preferred over adherent cells, as they are not limited by the surface area available, resulting in a more straightforward scale-up and homogenization of cultures. It is still possible to use adherent cells in a stirred-tank bioreactor using microcarriers, which are beads that the cell can attach to, whereas the microcarriers themselves remain suspended and stirred in the media [111]. Adherent cells can also be used in other bioreactor models, such as fixed-bed and wave, but all methods are ultimately limited by surface area [108].
In 2010, a pioneer work reported the production of the lentogenic F strain in adherent Vero cells using microcarriers in a 2 L bioreactor scale, reaching a peak production of 4.79 × 107 TCID50/mL [100]. Although the production was sufficient, the process developed has limited scalability due to the use of serum and adherent cell culture. The same authors tested DF-1 cells in the same microcarrier and stirred-tank bioreactor system, but the resulting titer was low (1.03 × 103 TCID50/mL) [102].
In 2021, our group published 1 L bioreactor productions of NDV constructs based on the LaSota strain [3] using a recently developed suspension Vero cell line [112] in commercial serum-free media. Adequate production comparable to ECEs was achieved for all constructs, with a peak titer of 2.37 × 108 TCID50/mL for NDV-GFP and 3.16 × 107 TCID50/mL for the COVID-19 vaccine candidate NDV-FLS [3]. This work established the basis for the upstream and analytics of a cell culture-based process for NDV production, showing the potential for a scalable process.

5.4. Downstream Processing and Formulation

Aside from the upstream production in cell culture, it is also important to establish downstream protocols and formulation. Although it is possible to inject the harvested allantoic fluid from eggs directly into animals for experiments [74], a few studies have implemented sucrose gradient centrifugation to purify NDV [18,27,70,75]. However, this is not the ideal method for industrialization, as it can be impractical and lack reproducibility. Chromatography-based purification methods would be more appropriate for industrial applications due to high scalability and reproducibility [113]. Chromatography purification protocols have been developed for other enveloped viruses, such as lentivirus [114], and could potentially be developed for NDV as well.
Formulation is also an essential aspect that must be studied for NDV. Vaccines with low stability might require storage and transportation in temperatures below −20 °C, which poses a major bottleneck in related to cold chain infrastructure [5]. A few studies have implemented lyophilized versions of their vaccine candidates [27,115], which represents a promising way of simplifying transportation. The lyophilization of a COVID-19 vaccine candidate did not significantly reduce the virus infectivity and allowed for storage at 4 °C [27], which greatly decreased the burden on storage. Further optimization of formulation strategies for NDV and stability studies for the existing vaccine candidates are extremely important to prepare for large-scale manufacturing and distribution.

6. Conclusions

In a time where vaccines are in high demand, it is important to establish vaccine platforms with rapid development and scalable manufacturing. NDV is a promising viral vector for vaccines with well-established recombinant technology, a long history of safety in humans and animals, and extensive pre-clinical research. With the progression of clinical trials for NDV-vectored vaccines in humans, it is important more than ever to fill the gap of process development for this virus by testing and optimizing scalable production methods for NDV using cell culture in bioreactors. The basis for a cell culture-based NDV production process has been established, but improvements in the upstream and downstream processing are critical, especially when it comes to purification, formulation and process intensification.
The results achieved in batch bioreactor productions of NDV were comparable to those in ECEs but could potentially be further optimized through process intensification. Different modes of operation, such as fed-batch and perfusion, could reduce by-products and replenish nutrients in the media, potentially allowing cells to reach a higher cell density and higher titers of viral production due to a more favorable metabolic state [19]. Perfusion could be particularly beneficial for NDV, seeing as this virus has been shown to lose infectivity over time in bioreactors [3]. This indicates viral degradation in the bioreactor due to unfavorable temperatures and shear stress from agitation, which could be avoided using perfusion to continuously harvest the virus and reduce the retention time in the vessel, as has been done for the VSV [112]. This can also potentially be integrated with purification for a continuous or semicontinuous manufacturing process [19].
Further development in these aspects is necessary so that NDV-vectored vaccines approved in the future can be produced sufficiently at a large scale without major transportation and cold chain issues.

Author Contributions

J.P.C.F.: writing—original draft preparation, writing—review and editing. A.A.K.: supervision, funding acquisition, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Research Council of Canada (NRC), grant number PR-013.1 for Priority Research on COVID-19. A.A.K. is funded through a Canada Research Chair (CRC-240394). J.P.C.F. received funding from a Mitacs Globalink Graduate Fellowship.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. NDV-vectored vaccine candidates for veterinary use in chronological order (publication date).
Table A1. NDV-vectored vaccine candidates for veterinary use in chronological order (publication date).
Vaccine TypePathogenDiseaseAntigen
Expressed
Animal ModelProduction Platform *Route * Dose *Reference
Live B1 StrainHPAIV H7N2Respiratory infectionHAChickenECEsioone dose of 1 × 106 EID50[116]
Live B1 strainSIVSimian AIDSGagMouseECEsiv, ip, inone or two doses of 5 × 107 PFU[117]
Live LaSota strainIBDVInfectious bursal diseaseVP2ChickenECEsioone or two doses of 1 × 104 EID50[118]
Live Clone 30 strainHPAIV H5N1Respiratory infectionHAChickenECEsonone dose of 106 EID50[82]
Live B1 StrainHPAIV H7N2Respiratory infectionHAChickenECEs, MDCK cell lineioone or two doses of 5 × 105 to 1.25 × 106 mean EID50[41]
Live LaSota strainHPAIV H5N1Flu (respiratory infection)HAChicken, mouseECEson (chicken), ip (mice)one dose of 106 EID50 (chicken), two doses of 108 EID50 (mice)[83]
Live LaSota strainHPAIV H7N1Respiratory infectionHAChickenECEsinone dose of 1 × 106 EID50[119]
Live LaSota strainHPAIV H5N1Respiratory infectionHAChickenECEsonone dose of 1 × 106 EID50[84]
Live LaSota strainHPAIV H5N2Respiratory infectionHAChickenECEsioone dose of 6 × 104, 6 × 105, 6 × 106 or 6 × 107 mean EID50[120]
Live LaSota strainRVFVRift Valley feverGnCalfECEsin, imone dose of 2 × 106 TCID50 (in), one dose of 3 × 107 TCID50 (im)[121]
Live LaSota strainBHV-1Respiratory infectiongDCalfECEsin + itone dose of 1.5 × 107 PFU[122]
Live LaSota strainRVFVRift Valley feverGn/GcMouse, lambECEsimtwo doses of 2 × 105 TCID50 (mice), two doses of 2 × 107 TCID50 (lambs)[51]
Live Clone 30 strainHPAIV H6N2Respiratory infectionHAChicken, turkeyECEsonone dose of 106 EID50 (chicken) or 107 EID50 (turkeys)[123]
Live LaSota/VF strainBorrelia burgdorferiLymeBmpA, OspCHamsterECEsin, im, ipone or two doses of 1 × 106 PFU[69]
Live LaSota strainRVRabiesGMouse, cat, dogECEsimone dose of 2 × 108 EID50, 2 × 107 EID50 or 2 × 106 EID50 (mice); three doses of 6 × 109 EID50 (cats); three doses of 6 × 109 EID50, 2 × 109 EID50 or 2 × 108 EID50 (dogs)[109]
Live LaSota strainAMPVRespiratory infectionGTurkeyECEsin, ioone or two doses of 106 TCID50[124]
Live LaSota strainNiVEncephalitisF, GMouse, pigECEsimtwo doses of 1 × 108 EID50 (mice), two doses of 2 × 109 EID50 (pigs)[71]
Live LaSota strainHPAIV H7N9Respiratory infectionHAMouseECEsintwo doses of 104 or 106 FFU[125]
Live chimeric NDVHPAIV H5N1Respiratory infectionHAChickenECEsonone dose of 106 TCID50[96]
Live LaSota strainIBVRespiratory infectionS2ChickenECEsioone dose of 106 EID50 followed by one dose of an attenuated IBV vaccine[126]
Live LaSota strainHPAIV H5N1Respiratory infectionHADuckECEsiotwo doses of 106 EID50[90]
Live LaC30L strainIBDVInfectious bursal diseaseVP2Chicken embryoECEsin ovoone dose of 3 × 102, 3 × 103, 3 × 104 or 3 × 105 EID50[127]
Live LaSota strainILTVRespiratory infectiongB, gC, gDChickenECEsontwo doses of 2 × 105 TCID50[86]
Live LaSota strainILTVRespiratory infectiongB, gDChickenECEsin + ioone dose of 1 × 106 TCID50[87]
Live NA strainGPVDerzsy’s disease (goose hepatitis)VP3GoslingECEssctwo doses of 106 EID50[91]
Live LaSota strainCDVCanine distemperF, HNMinkECEsimtwo doses of 109 EID50[93]
Live LaSota strainH7N9, H5N1Respiratory infectionHAChickenECEsim, ontwo doses of 5 × 106 PFU[128]
Live LaSota strainVSVVesicular stomatitisGMouseECEsimtwo doses of 1 × 107 TCID50[129]
Live LaSota strainHPAIV H9N2Respiratory infectionHAChickenECEson, imtwo doses of 107 FFU[130]
Live LaSota strainWNVWest Nile feverPrM/EMouse, horse, chicken, duck, gooseECEsim, in, oraltwo doses of 1 × 108 EID50 (im) (mice); two doses of 2 × 109 EID50 (im) (horses); two doses of 1 × 108 EID50 (im) or 1 × 1010 EID50 (oral) (chicken); two doses of 5 × 108 EID50 (im, in, or oral) (geese)[131]
Live LaSota strainBEFVBovine ephemeral feverGMouse, calfECEsimone dose of 1 × 106 TCID50 (mice), two doses 8 × 107 TCID50 (calves)[132]
Live Clone 30 strainPaBV-4, CnBV-2Proventricular dilatation diseaseN/PCockatiel, canaryECEsimone dose of 8 × 105 FFU to 1 × 106 FFU (cockatiel) or 4 × 106 FFU (canary)[94]
Live chimeric NDVHPAIV H5N1Respiratory infectionHAChickenECEsontwo doses of 106 PFU/mL[97]
Live LaSota strainIBVRespiratory infectionS1ChickenECEsonone or two doses of 106 PFU[133]
Live LaSota strainAMPVRespiratory infectionF/GTurkeyECEsin, ioone dose of 106 TCID50[52]
Live LaSota strain or chimeric NDVHPAIV H5N1Respiratory infectionHA, HA/NA, HA/M1, HA/NS1ChickenECEsonone dose of 106 PFU/mL or two doses: prime with chimeric NDV construct, boost with LaSota construct (106 PFU/mL)[55]
Live F strainIBDVInfectious bursal diseaseVP2ChickenECEs, Vero cell lineintwo doses of 105, 106 or 107 EID50[106]
Live LaSota strainMERS-CoVRespiratory infectionSMouse, camelECEsimtwo doses of 108 EID50 (mouse) or 1 × 109 EID50 (camel)[134]
Live or inactivated LaSota strainHPAIV H5N2Respiratory infectionHAChickenECEsim, sprayingtwo doses of 5 × 106 TCID50 (im live) or 107 TCID50 (im inactivated); around 106 TCID50 (spraying)[135]
Live LX strainHPAIV H7N9Respiratory infectionHAChickenECEsintwo doses of 5 × 106 EID50[136]
Live attenuated GM strainDTMUVDuck Tembusu virus diseasePrM/EDuckECEssctwo doses of 106 EID50[89]
Live LaSota strain or chimeric NDVSIVSimian AIDSEnv (gp160)Guinea pigECEsintwo doses of 105 TCID50[32]
Live chimeric NDVHPAIV H9N2Respiratory infectionHAChickenECEsonone dose of 106 EID50[98]
Live LaSota strain or chimeric NDVHPAIV H5N2Respiratory infectionHA/NAChickenECEsintwo doses: prime with chimeric NDV construct (1 × 105 PFU), boost with LaSota construct (2 × 105 PFU)[53]
Live rAI4 strainHPAIV H7N9Respiratory infectionHAChickenECEsin, ioone dose of 106 EID50[137]
Live NA StrainHPAIV H9N2Respiratory infectionHAChickenECEsonone or two doses of 106 EID50[138]
Live LaSota strainIBVRespiratory infectionSChickenECEsonone or two doses of 1 × 106 PFU[139]
Live LaSota strain or chimeric NDVHPAIV H7N8Respiratory infectionHA, HA/NAChicken, turkeyECEsintwo doses: prime with chimeric NDV construct (1 × 105 PFU), boost with LaSota construct (2 × 105 PFU)[56]
Live R2B strainARVViral arthritis/tenosynovitisσCChickenECEsoral + in, imtwo doses of 1 × 105 EID50[85]
Live SH12 strainGoAstVVisceral goutCapGoslingECEsonone dose of 1 × 107 TCID50[92]
Live LaSota strainCSFVClassical swine feverE2, ErnsPigECEsintwo doses of 103 TCID50[140]
Live LaSota strainIBVRespiratory infectionS1 (multi-epitope)ChickenECEsonone dose of 1 × 106 EID50[141]
Live LaSota strainPRRSVPorcine reproductive and respiratory syndromeGP5, GP3/GP5PigletECEsimtwo doses of 4 × 108 EID50[57]
Live TS09-C (thermostable) strainHPAIV H5N1Respiratory infectionHA, HA1ChickenECEsin, iotwo doses of 106 TCID50[95]
Live LaSota strainFAdV-4Hepatitis-hydropericardium syndromeFiber 2ChickenECEsimone dose of 107 EID50[88]
Live Clone 30 strainPPRVPeste des petits ruminants (PPR)HGoatCEFsscone or two doses of 6 × 106 TCID50/mL[142]
Live R2B strain or chimeric NDVRVRabiesGMouseECEsimtwo doses of 2 × 106 TCID50[110]
Live HR09 (thermostable) strainHPAIV H9N2Respiratory infectionHA, chimeric HAChickenECEsonone dose of 106 EID50[14]
Live LaSota strainIBVRespiratory infectionN (multi-epitope)ChickenECEsonone dose of 106 EID50[143]
Live LaSota strainPPRVPeste des petits ruminants (PPR)HSheep, goatECEsimtwo doses of 1 × 108, 5 × 108 or 3 × 109 EID50[115]
Live R2B strainCIAVChicken infectious anaemiaVP1/VP2ChickenVero cell lineonthree doses of 1 × 106 TCID50/mL[58]
Live K148/08 strainHPAIV H5N6Respiratory infectionHAChicken, duckECEson, spraytwo doses of 107 EID50[144]
* ECEs: embryonated chicken eggs, CEFs: chicken embryo fibroblasts, io: intraocular, iv: intravenous, ip: intraperitoneal, in: intranasal, on: oculonasal, im: intramuscular, it: intratracheal, sc: subcutaneous, EID50: embryo infectious dose 50%, PFU: plaque-forming units, TCID50: tissue culture infectious dose 50%, FFU: focus-forming units.

References

  1. Kim, S.-H.; Samal, S.K. Newcastle Disease Virus as a Vaccine Vector for Development of Human and Veterinary Vaccines. Viruses 2016, 8, 183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Annas, S.; Zamri-Saad, M. Intranasal Vaccination Strategy to Control the COVID-19 Pandemic from a Veterinary Medicine Perspective. Animals 2021, 11, 1876. [Google Scholar] [CrossRef] [PubMed]
  3. Fulber, J.P.C.; Farnós, O.; Kiesslich, S.; Yang, Z.; Dash, S.; Susta, L.; Wootton, S.K.; Kamen, A.A. Process Development for Newcastle Disease Virus-Vectored Vaccines in Serum-Free Vero Cell Suspension Cultures. Vaccines 2021, 9, 1335. [Google Scholar] [CrossRef] [PubMed]
  4. Kon, E.; Elia, U.; Peer, D. Principles for designing an optimal mRNA lipid nanoparticle vaccine. Curr. Opin. Biotechnol. 2022, 73, 329–336. [Google Scholar] [CrossRef] [PubMed]
  5. Crommelin, D.J.A.; Anchordoquy, T.J.; Volkin, D.B.; Jiskoot, W.; Mastrobattista, E. Addressing the Cold Reality of mRNA Vaccine Stability. J. Pharm. Sci. 2021, 110, 997–1001. [Google Scholar] [CrossRef]
  6. Crommelin, D.J.A.; Volkin, D.B.; Hoogendoorn, K.H.; Lubiniecki, A.S.; Jiskoot, W. The Science is There: Key Considerations for Stabilizing Viral Vector-Based COVID-19 Vaccines. J. Pharm. Sci. 2021, 110, 627–634. [Google Scholar] [CrossRef]
  7. Samaranayake, L.P.; Seneviratne, C.J.; Fakhruddin, K.S. Coronavirus disease 2019 (COVID-19) vaccines: A concise review. Oral Dis. 2021, 1–11. [Google Scholar] [CrossRef]
  8. Tomori, O.; Kolawole, M.O. Ebola virus disease: Current vaccine solutions. Curr. Opin. Immunol. 2021, 71, 27–33. [Google Scholar] [CrossRef]
  9. European Medicines Agency (EMA). Zabdeno Summary of Product Characteristics. Available online: https://www.ema.europa.eu/en/documents/product-information/zabdeno-epar-product-information_en.pdf (accessed on 8 October 2021).
  10. European Medicines Agency (EMA). Mvabea Summary of Product Characteristics. Available online: https://www.ema.europa.eu/en/documents/product-information/mvabea-epar-product-information_en.pdf (accessed on 8 October 2021).
  11. Henao-Restrepo, A.M.; Camacho, A.; Longini, I.M.; Watson, C.H.; Edmunds, W.J.; Egger, M.; Carroll, M.W.; Dean, N.E.; Diatta, I.; Doumbia, M.; et al. Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: Final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ça Suffit!). Lancet 2017, 389, 505–518. [Google Scholar] [CrossRef] [Green Version]
  12. Pinschewer, D.D. Virally vectored vaccine delivery: Medical needs, mechanisms, advantages and challenges. Swiss Med. Wkly. 2017, 147, w14465. [Google Scholar] [CrossRef] [Green Version]
  13. Yoshida, A.; Kim, S.-H.; Manoharan, V.K.; Varghese, B.P.; Paldurai, A.; Samal, S.K. Novel avian paramyxovirus-based vaccine vectors expressing the Ebola virus glycoprotein elicit mucosal and humoral immune responses in guinea pigs. Sci. Rep. 2019, 9, 5520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Zhang, X.; Bo, Z.; Meng, C.; Chen, Y.; Zhang, C.; Cao, Y.; Wu, Y. Generation and Evaluation of Recombinant Thermostable Newcastle Disease Virus Expressing the HA of H9N2 Avian Influenza Virus. Viruses 2021, 13, 1606. [Google Scholar] [CrossRef] [PubMed]
  15. Choi, K.-S. Newcastle disease virus vectored vaccines as bivalent or antigen delivery vaccines. Clin. Exp. Vaccine Res. 2017, 6, 72–82. [Google Scholar] [CrossRef] [PubMed]
  16. Bello, M.B.; Yusoff, K.; Ideris, A.; Hair-Bejo, M.; Jibril, A.H.; Peeters, B.P.H.; Omar, A.R. Exploring the Prospects of Engineered Newcastle Disease Virus in Modern Vaccinology. Viruses 2020, 12, 451. [Google Scholar] [CrossRef]
  17. Burman, B.; Pesci, G.; Zamarin, D. Newcastle Disease Virus at the Forefront of Cancer Immunotherapy. Cancers 2020, 12, 3552. [Google Scholar] [CrossRef]
  18. Sun, W.; Leist, S.R.; McCroskery, S.; Liu, Y.; Slamanig, S.; Oliva, J.; Amanat, F.; Schäfer, A.; Dinnon, K.H.; García-Sastre, A.; et al. Newcastle disease virus (NDV) expressing the spike protein of SARS-CoV-2 as a live virus vaccine candidate. EBioMedicine 2020, 62, 103132. [Google Scholar] [CrossRef]
  19. Silva, C.A.T.; Kamen, A.A.; Henry, O. Recent advances and current challenges in process intensification of cell culture-based influenza virus vaccine manufacturing. Can. J. Chem. Eng. 2021, 99, 2525–2535. [Google Scholar] [CrossRef]
  20. Bukreyev, A.; Huang, Z.; Yang, L.; Elankumaran, S.; Claire, M.S.; Murphy, B.R.; Samal, S.K.; Collins, P.L. Recombinant Newcastle Disease Virus Expressing a Foreign Viral Antigen Is Attenuated and Highly Immunogenic in Primates. J. Virol. 2005, 79, 13275–13284. [Google Scholar] [CrossRef] [Green Version]
  21. Shimkin, N. Conjunctival haemorrhage due to an infection of Newcastle virus of fowls in man (laboratory and contact infection). Br. J. Ophthalmol. 1946, 30, 260. [Google Scholar] [CrossRef] [Green Version]
  22. Nelson, C.; Pomeroy, B.; Schrall, K.; Park, W.; Lindeman, R. An outbreak of conjunctivitis due to Newcastle disease virus (NDV) occurring in poultry workers. Am. J. Public Health Nations Health 1952, 42, 672–678. [Google Scholar] [CrossRef]
  23. Mustaffa-Babjee, A.; Ibrahim, A.L.; Khim, T.S. A case of human infection with Newcastle disease virus. Southeast Asian J. Trop. Med. Public Health 1976, 7, 622–624. [Google Scholar] [PubMed]
  24. Prajna, N.V.; Lalitha, P.; Chen, C.; Zhong, L.; Lietman, T.M.; Doan, T.; Seitzman, G.D. Acute keratoconjunctivitis resulting from coinfection with avian Newcastle virus and human adenovirus. Cornea 2022, 41, 630–631. [Google Scholar] [CrossRef] [PubMed]
  25. Fausther-Bovendo, H.; Kobinger, G.P. Pre-existing immunity against Ad vectors. Hum. Vaccines Immunother. 2014, 10, 2875–2884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Park, J.-G.; Oladunni, F.S.; Rohaim, M.A.; Whittingham-Dowd, J.; Tollitt, J.; Hodges, M.D.J.; Fathallah, N.; Assas, M.B.; Alhazmi, W.; Almilaibary, A.; et al. Immunogenicity and protective efficacy of an intranasal live-attenuated vaccine against SARS-CoV-2. iScience 2021, 24, 102941. [Google Scholar] [CrossRef] [PubMed]
  27. Warner, B.M.; Santry, L.A.; Leacy, A.; Chan, M.; Pham, P.H.; Vendramelli, R.; Pei, Y.; Tailor, N.; Valcourt, E.; Leung, A.; et al. Intranasal vaccination with a Newcastle disease virus-vectored vaccine protects hamsters from SARS-CoV-2 infection and disease. iScience 2021, 24, 103219. [Google Scholar] [CrossRef] [PubMed]
  28. DiNapoli, J.M.; Kotelkin, A.; Yang, L.; Elankumaran, S.; Murphy, B.R.; Samal, S.K.; Collins, P.L.; Bukreyev, A. Newcastle disease virus, a host range-restricted virus, as a vaccine vector for intranasal immunization against emerging pathogens. Proc. Natl. Acad. Sci. USA 2007, 104, 9788. [Google Scholar] [CrossRef] [Green Version]
  29. DiNapoli, J.M.; Yang, L.; Samal, S.K.; Murphy, B.R.; Collins, P.L.; Bukreyev, A. Respiratory tract immunization of non-human primates with a Newcastle disease virus-vectored vaccine candidate against Ebola virus elicits a neutralizing antibody response. Vaccine 2010, 29, 17–25. [Google Scholar] [CrossRef] [PubMed]
  30. Tcheou, J.; Raskin, A.; Singh, G.; Kawabata, H.; Bielak, D.; Sun, W.; González-Domínguez, I.; Sather, D.N.; García-Sastre, A.; Palese, P.; et al. Safety and Immunogenicity Analysis of a Newcastle Disease Virus (NDV-HXP-S) Expressing the Spike Protein of SARS-CoV-2 in Sprague Dawley Rats. Front. Immunol. 2021, 12, 791764. [Google Scholar] [CrossRef]
  31. Lara-Puente, J.H.; Carreño, J.M.; Sun, W.; Suárez-Martínez, A.; Ramírez-Martínez, L.; Quezada-Monroy, F.; de la Rosa, G.P.; Vigueras-Moreno, R.; Singh, G.; Rojas-Martínez, O.; et al. Safety and Immunogenicity of a Newcastle Disease Virus Vector-Based SARS-CoV-2 Vaccine Candidate, AVX/COVID-12-HEXAPRO (Patria), in Pigs. mBio 2021, 12, e01908-21. [Google Scholar] [CrossRef]
  32. Manoharan, V.K.; Khattar, S.K.; Labranche, C.C.; Montefiori, D.C.; Samal, S.K. Modified Newcastle Disease virus as an improved vaccine vector against Simian Immunodeficiency virus. Sci. Rep. 2018, 8, 8952. [Google Scholar] [CrossRef] [Green Version]
  33. Gallo, O.; Locatello, L.G.; Mazzoni, A.; Novelli, L.; Annunziato, F. The central role of the nasal microenvironment in the transmission, modulation, and clinical progression of SARS-CoV-2 infection. Mucosal. Immunol. 2021, 14, 305–316. [Google Scholar] [CrossRef] [PubMed]
  34. Freeman, A.I.; Zakay-Rones, Z.; Gomori, J.M.; Linetsky, E.; Rasooly, L.; Greenbaum, E.; Rozenman-Yair, S.; Panet, A.; Libson, E.; Irving, C.S.; et al. Phase I/II Trial of Intravenous NDV-HUJ Oncolytic Virus in Recurrent Glioblastoma Multiforme. Mol. Ther. 2006, 13, 221–228. [Google Scholar] [CrossRef] [PubMed]
  35. Schirrmacher, V. Fifty Years of Clinical Application of Newcastle Disease Virus: Time to Celebrate! Biomedicines 2016, 4, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Schirrmacher, V.; van Gool, S.; Stuecker, W. Breaking Therapy Resistance: An Update on Oncolytic Newcastle Disease Virus for Improvements of Cancer Therapy. Biomedicines 2019, 7, 66. [Google Scholar] [CrossRef] [Green Version]
  37. Meng, Q.; He, J.; Zhong, L.; Zhao, Y. Advances in the Study of Antitumour Immunotherapy for Newcastle Disease Virus. Int. J. Med. Sci. 2021, 18, 2294–2302. [Google Scholar] [CrossRef] [PubMed]
  38. Santry, L.A.; McAusland, T.M.; Susta, L.; Wood, G.A.; Major, P.P.; Petrik, J.J.; Bridle, B.W.; Wootton, S.K. Production and purification of high-titer Newcastle disease virus for use in preclinical mouse models of cancer. Mol. Ther.-Methods Clin. Dev. 2018, 9, 181–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Yurchenko, K.; Yi, J.; Shestopalov, A. Adaptation of the Newcastle disease virus to cell cultures for enhancing its oncolytic properties. Acta Nat. 2019, 11, 66–73. [Google Scholar] [CrossRef]
  40. DiNapoli, J.M.; Yang, L.; Suguitan, A., Jr.; Elankumaran, S.; Dorward, D.W.; Murphy, B.R.; Samal, S.K.; Collins, P.L.; Bukreyev, A. Immunization of primates with a Newcastle disease virus-vectored vaccine via the respiratory tract induces a high titer of serum neutralizing antibodies against highly pathogenic avian influenza virus. J. Virol. 2007, 81, 11560–11568. [Google Scholar] [CrossRef] [Green Version]
  41. Park, M.-S.; Steel, J.; García-Sastre, A.; Swayne, D.; Palese, P. Engineered viral vaccine constructs with dual specificity: Avian influenza and Newcastle disease. Proc. Natl. Acad. Sci. USA 2006, 103, 8203–8208. [Google Scholar] [CrossRef] [Green Version]
  42. Zhan, Y.; Yu, S.; Yang, S.; Qiu, X.; Meng, C.; Tan, L.; Song, C.; Liao, Y.; Liu, W.; Sun, Y.; et al. Newcastle Disease virus infection activates PI3K/Akt/mTOR and p38 MAPK/Mnk1 pathways to benefit viral mRNA translation via interaction of the viral NP protein and host eIF4E. PLOS Pathog. 2020, 16, e1008610. [Google Scholar] [CrossRef]
  43. Schirrmacher, V. Immunobiology of Newcastle disease virus and its use for prophylactic vaccination in poultry and as adjuvant for therapeutic vaccination in cancer patients. Int. J. Mol. Sci. 2017, 18, 1103. [Google Scholar] [CrossRef] [PubMed]
  44. Yang, Y.; Xue, J.; Teng, Q.; Li, X.; Bu, Y.; Zhang, G. Mechanisms and consequences of Newcastle disease virus W protein subcellular localization in the nucleus or mitochondria. J. Virol. 2021, 95, e02087-20. [Google Scholar] [CrossRef] [PubMed]
  45. Vijayakumar, G.; Zamarin, D. Design and Production of Newcastle Disease Virus for Intratumoral Immunomodulation. In Oncolytic Viruses; Engeland, C.E., Ed.; Springer New York: New York, NY, USA, 2020; pp. 133–154. [Google Scholar]
  46. Gao, Q.; Park, M.-S.; Palese, P. Expression of Transgenes from Newcastle Disease Virus with a Segmented Genome. J. Virol. 2008, 82, 2692–2698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Cardenas-Garcia, S.; Afonso, C.L. Reverse genetics of Newcastle disease virus. In Reverse Genetics of RNA Viruses; Springer: Berlin/Heidelberg, Germany, 2017; pp. 141–158. [Google Scholar]
  48. Pühler, F.; Willuda, J.; Puhlmann, J.; Mumberg, D.; Römer-Oberdörfer, A.; Beier, R. Generation of a recombinant oncolytic Newcastle disease virus and expression of a full IgG antibody from two transgenes. Gene Ther. 2008, 15, 371–383. [Google Scholar] [CrossRef]
  49. Viktorova, E.G.; Khattar, S.K.; Kouiavskaia, D.; Laassri, M.; Zagorodnyaya, T.; Dragunsky, E.; Samal, S.; Chumakov, K.; Belov, G.A.; Pfeiffer, J.K. Newcastle Disease Virus-Based Vectored Vaccine against Poliomyelitis. J. Virol. 2018, 92, e00976-18. [Google Scholar] [CrossRef] [Green Version]
  50. Khattar, S.K.; Manoharan, V.; Bhattarai, B.; LaBranche, C.C.; Montefiori, D.C.; Samal, S.K.; Meng, X.-J. Mucosal Immunization with Newcastle Disease Virus Vector Coexpressing HIV-1 Env and Gag Proteins Elicits Potent Serum, Mucosal, and Cellular Immune Responses That Protect against Vaccinia Virus Env and Gag Challenges. mBio 2015, 6, e01005. [Google Scholar] [CrossRef] [Green Version]
  51. Kortekaas, J.; de Boer, S.M.; Kant, J.; Vloet, R.P.M.; Antonis, A.F.G.; Moormann, R.J.M. Rift Valley fever virus immunity provided by a paramyxovirus vaccine vector. Vaccine 2010, 28, 4394–4401. [Google Scholar] [CrossRef]
  52. Hu, H.; Roth, J.P.; Zsak, L.; Yu, Q. Engineered Newcastle disease virus expressing the F and G proteins of AMPV-C confers protection against challenges in turkeys. Sci. Rep. 2017, 7, 4025. [Google Scholar] [CrossRef] [Green Version]
  53. Cho, Y.; Lamichhane, B.; Nagy, A.; Chowdhury, I.R.; Samal, S.K.; Kim, S.-H. Co-expression of the Hemagglutinin and Neuraminidase by Heterologous Newcastle Disease Virus Vectors Protected Chickens against H5 Clade 2.3.4.4 HPAI Viruses. Sci. Rep. 2018, 8, 16854. [Google Scholar] [CrossRef]
  54. Murr, M.; Karger, A.; Steglich, C.; Mettenleiter, T.C.; Römer-Oberdörfer, A. Coexpression of soluble and membrane-bound avian influenza virus H5 by recombinant Newcastle disease virus leads to an increase in antigen levels. J. Gen. Virol. 2020, 101, 473–483. [Google Scholar] [CrossRef]
  55. Kim, S.-H.; Samal, S.K. Heterologous prime-boost immunization of Newcastle disease virus vectored vaccines protected broiler chickens against highly pathogenic avian influenza and Newcastle disease viruses. Vaccine 2017, 35, 4133–4139. [Google Scholar] [CrossRef] [PubMed]
  56. Roy Chowdhury, I.; Yeddula, S.G.R.; Pierce, B.G.; Samal, S.K.; Kim, S.-H. Newcastle disease virus vectors expressing consensus sequence of the H7 HA protein protect broiler chickens and turkeys against highly pathogenic H7N8 virus. Vaccine 2019, 37, 4956–4962. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, H.; Nan, F.; Li, Z.; Zhao, G.; Xie, C.; Ha, Z.; Zhang, J.; Han, J.; Xiao, P.; Zhuang, X.; et al. Construction and immunological evaluation of recombinant Newcastle disease virus vaccines expressing highly pathogenic porcine reproductive and respiratory syndrome virus GP3/GP5 proteins in pigs. Vet. Microbiol. 2019, 239, 108490. [Google Scholar] [CrossRef] [PubMed]
  58. Chellappa, M.M.; Dey, S.; Pathak, D.C.; Singh, A.; Ramamurthy, N.; Ramakrishnan, S.; Mariappan, A.K.; Dhama, K.; Vakharia, V.N. Newcastle Disease Virus Vectored Chicken Infectious Anaemia Vaccine Induces Robust Immune Response in Chickens. Viruses 2021, 13, 1985. [Google Scholar] [CrossRef]
  59. He, L.; Zhang, Z.; Yu, Q. Expression of Two Foreign Genes by a Newcastle Disease Virus Vector From the Optimal Insertion Sites through a Combination of the ITU and IRES-Dependent Expression Approaches. Front. Microbiol. 2020, 11, 769. [Google Scholar] [CrossRef] [PubMed]
  60. Kim, S.-H.; Samal, S.K. Role of Untranslated Regions in Regulation of Gene Expression, Replication, and Pathogenicity of Newcastle Disease Virus Expressing Green Fluorescent Protein. J. Virol. 2010, 84, 2629–2634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Ayllon, J.; García-Sastre, A.; Martínez-Sobrido, L. Rescue of recombinant Newcastle disease virus from cDNA. J. Vis. Exp. JoVE 2013, 80, 50830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Zhao, W.; Zhang, Z.; Zsak, L.; Yu, Q. P and M gene junction is the optimal insertion site in Newcastle disease virus vaccine vector for foreign gene expression. J. Gen. Virol. 2015, 96, 40–45. [Google Scholar] [CrossRef]
  63. Molouki, A.; Peeters, B. Rescue of recombinant Newcastle disease virus: Current cloning strategies and RNA polymerase provision systems. Arch. Virol. 2017, 162, 1–12. [Google Scholar] [CrossRef]
  64. Hu, Z.; Ni, J.; Cao, Y.; Liu, X. Newcastle Disease Virus as a Vaccine Vector for 20 Years: A Focus on Maternally Derived Antibody Interference. Vaccines 2020, 8, 222. [Google Scholar] [CrossRef]
  65. Nakaya, T.; Cros, J.; Park, M.-S.; Nakaya, Y.; Zheng, H.; Sagrera, A.; Villar, E.; García-Sastre, A.; Palese, P. Recombinant Newcastle disease virus as a vaccine vector. J. Virol. 2001, 75, 11868–11873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Martinez-Sobrido, L.; Gitiban, N.; Fernandez-Sesma, A.; Cros, J.; Mertz, S.E.; Jewell, N.A.; Hammond, S.; Flano, E.; Durbin, R.K.; García-Sastre, A.; et al. Protection against Respiratory Syncytial Virus by a Recombinant Newcastle Disease Virus Vector. J. Virol. 2006, 80, 1130–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Carnero, E.; Li, W.; Borderia, A.V.; Moltedo, B.; Moran, T.; García-Sastre, A. Optimization of Human Immunodeficiency Virus Gag Expression by Newcastle Disease Virus Vectors for the Induction of Potent Immune Responses. J. Virol. 2009, 83, 584–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Maamary, J.; Array, F.; Gao, Q.; García-Sastre, A.; Steinman, R.M.; Palese, P.; Nchinda, G. Newcastle Disease Virus Expressing a Dendritic Cell-Targeted HIV Gag Protein Induces a Potent Gag-Specific Immune Response in Mice. J. Virol. 2011, 85, 2235–2246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Xiao, S.; Kumar, M.; Yang, X.; Akkoyunlu, M.; Collins, P.L.; Samal, S.K.; Pal, U. A host-restricted viral vector for antigen-specific immunization against Lyme disease pathogen. Vaccine 2011, 29, 5294–5303. [Google Scholar] [CrossRef] [PubMed]
  70. Khattar, S.K.; Samal, S.; DeVico, A.L.; Collins, P.L.; Samal, S.K. Newcastle Disease Virus Expressing Human Immunodeficiency Virus Type 1 Envelope Glycoprotein Induces Strong Mucosal and Serum Antibody Responses in Guinea Pigs. J. Virol. 2011, 85, 10529–10541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Kong, D.; Wen, Z.; Su, H.; Ge, J.; Chen, W.; Wang, X.; Wu, C.; Yang, C.; Chen, H.; Bu, Z. Newcastle disease virus-vectored Nipah encephalitis vaccines induce B and T cell responses in mice and long-lasting neutralizing antibodies in pigs. Virology 2012, 432, 327–335. [Google Scholar] [CrossRef] [Green Version]
  72. Kim, S.-H.; Chen, S.; Jiang, X.; Green, K.Y.; Samal, S.K.; López, S. Newcastle Disease Virus Vector Producing Human Norovirus-Like Particles Induces Serum, Cellular, and Mucosal Immune Responses in Mice. J. Virol. 2014, 88, 9718–9727. [Google Scholar] [CrossRef] [Green Version]
  73. Khattar, S.K.; DeVico, A.L.; LaBranche, C.C.; Panda, A.; Montefiori, D.C.; Samal, S.K.; Silvestri, G. Enhanced Immune Responses to HIV-1 Envelope Elicited by a Vaccine Regimen Consisting of Priming with Newcastle Disease Virus Expressing HIV gp160 and Boosting with gp120 and SOSIP gp140 Proteins. J. Virol. 2016, 90, 1682–1686. [Google Scholar] [CrossRef] [Green Version]
  74. Nath, B.; Vandna; Saini, H.M.; Prasad, M.; Kumar, S. Evaluation of Japanese encephalitis virus E and NS1 proteins immunogenicity using a recombinant Newcastle disease virus in mice. Vaccine 2020, 38, 1860–1868. [Google Scholar] [CrossRef]
  75. Sun, W.; McCroskery, S.; Liu, W.-C.; Leist, S.R.; Liu, Y.; Albrecht, R.A.; Slamanig, S.; Oliva, J.; Amanat, F.; Schäfer, A.; et al. A Newcastle Disease Virus (NDV) Expressing a Membrane-Anchored Spike as a Cost-Effective Inactivated SARS-CoV-2 Vaccine. Vaccines 2020, 8, 771. [Google Scholar] [CrossRef] [PubMed]
  76. Sun, W.; Liu, Y.; Amanat, F.; González-Domínguez, I.; McCroskery, S.; Slamanig, S.; Coughlan, L.; Rosado, V.; Lemus, N.; Jangra, S.; et al. A Newcastle disease virus expressing a stabilized spike protein of SARS-CoV-2 induces protective immune responses. Nat. Commun. 2021, 12, 6197. [Google Scholar] [CrossRef] [PubMed]
  77. Zhao, W.; Zhang, P.; Bai, S.; Lv, M.; Wang, J.; Chen, W.; Yu, Q.; Wu, J. Heterologous prime-boost regimens with HAdV-5 and NDV vectors elicit stronger immune responses to Ebola virus than homologous regimens in mice. Arch. Virol. 2021, 166, 3333–3341. [Google Scholar] [CrossRef] [PubMed]
  78. Jung, B.-K.; An, Y.H.; Jang, J.-J.; Jeon, J.H.; Jang, S.H.; Jang, H. The human ACE-2 receptor binding domain of SARS-CoV-2 express on the viral surface of the Newcastle disease virus as a non-replicating viral vector vaccine candidate. PLoS ONE 2022, 17, e0263684. [Google Scholar] [CrossRef]
  79. Ponce-de-León, S.; Torres, M.; Soto-Ramírez, L.E.; José Calva, J.; Santillán-Doherty, P.; Carranza-Salazar, D.E.; Carreño, J.M.; Carranza, C.; Juárez, E.; Carreto-Binaghi, L.E.; et al. Safety and immunogenicity of a live recombinant Newcastle disease virus-based COVID-19 vaccine (Patria) administered via the intramuscular or intranasal route: Interim results of a non-randomized open label phase I trial in Mexico. medRxiv 2022, preprint. [Google Scholar] [CrossRef]
  80. Pitisuttithum, P.; Luvira, V.; Lawpoolsri, S.; Muangnoicharoen, S.; Kamolratanakul, S.; Sivakorn, C.; Narakorn, P.; Surichan, S.; Prangpratanporn, S.; Puksuriwong, S.; et al. Safety and immunogenicity of an inactivated recombinant Newcastle disease virus vaccine expressing SARS-CoV-2 spike: Interim results of a randomised, placebo-controlled, phase 1 trial. eClinicalMedicine 2022, 45, 101323. [Google Scholar] [CrossRef]
  81. Kim, S.-H.; Samal, S.K. Innovation in Newcastle Disease Virus Vectored Avian Influenza Vaccines. Viruses 2019, 11, 300. [Google Scholar] [CrossRef] [Green Version]
  82. Veits, J.; Wiesner, D.; Fuchs, W.; Hoffmann, B.; Granzow, H.; Starick, E.; Mundt, E.; Schirrmeier, H.; Mebatsion, T.; Mettenleiter, T.C.; et al. Newcastle disease virus expressing H5 hemagglutinin gene protects chickens against Newcastle disease and avian influenza. Proc. Natl. Acad. Sci. USA 2006, 103, 8197–8202. [Google Scholar] [CrossRef] [Green Version]
  83. Ge, J.; Deng, G.; Wen, Z.; Tian, G.; Wang, Y.; Shi, J.; Wang, X.; Li, Y.; Hu, S.; Jiang, Y.; et al. Newcastle Disease Virus-Based Live Attenuated Vaccine Completely Protects Chickens and Mice from Lethal Challenge of Homologous and Heterologous H5N1 Avian Influenza Viruses. J. Virol. 2007, 81, 150–158. [Google Scholar] [CrossRef] [Green Version]
  84. Nayak, B.; Rout, S.N.; Kumar, S.; Khalil, M.S.; Fouda, M.M.; Ahmed, L.E.; Earhart, K.C.; Perez, D.R.; Collins, P.L.; Samal, S.K. Immunization of chickens with Newcastle disease virus expressing H5 hemagglutinin protects against highly pathogenic H5N1 avian influenza viruses. PLoS ONE 2009, 4, e6509. [Google Scholar] [CrossRef]
  85. Saikia, D.P.; Yadav, K.; Pathak, D.C.; Ramamurthy, N.; D’Silva, A.L.; Marriappan, A.K.; Ramakrishnan, S.; Vakharia, V.N.; Chellappa, M.M.; Dey, S. Recombinant Newcastle Disease Virus (NDV) Expressing Sigma C Protein of Avian Reovirus (ARV) Protects against Both ARV and NDV in Chickens. Pathogens 2019, 8, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Kanabagatte Basavarajappa, M.; Kumar, S.; Khattar, S.K.; Gebreluul, G.T.; Paldurai, A.; Samal, S.K. A recombinant Newcastle disease virus (NDV) expressing infectious laryngotracheitis virus (ILTV) surface glycoprotein D protects against highly virulent ILTV and NDV challenges in chickens. Vaccine 2014, 32, 3555–3563. [Google Scholar] [CrossRef] [PubMed]
  87. Zhao, W.; Spatz, S.; Zhang, Z.; Wen, G.; Garcia, M.; Zsak, L.; Yu, Q.; Sandri-Goldin, R.M. Newcastle Disease Virus (NDV) Recombinants Expressing Infectious Laryngotracheitis Virus (ILTV) Glycoproteins gB and gD Protect Chickens against ILTV and NDV Challenges. J. Virol. 2014, 88, 8397–8406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Tian, K.-Y.; Guo, H.-F.; Li, N.; Zhang, Y.-H.; Wang, Z.; Wang, B.; Yang, X.; Li, Y.-T.; Zhao, J. Protection of chickens against hepatitis-hydropericardium syndrome and Newcastle disease with a recombinant Newcastle disease virus vaccine expressing the fowl adenovirus serotype 4 fiber-2 protein. Vaccine 2020, 38, 1989–1997. [Google Scholar] [CrossRef]
  89. Sun, M.; Dong, J.; Li, L.; Lin, Q.; Sun, J.; Liu, Z.; Shen, H.; Zhang, J.; Ren, T.; Zhang, C. Recombinant Newcastle disease virus (NDV) expressing Duck Tembusu virus (DTMUV) pre-membrane and envelope proteins protects ducks against DTMUV and NDV challenge. Vet. Microbiol. 2018, 218, 60–69. [Google Scholar] [CrossRef]
  90. Ferreira, H.L.; Pirlot, J.F.; Reynard, F.; van den Berg, T.; Bublot, M.; Lambrecht, B. Immune Responses and Protection Against H5N1 Highly Pathogenic Avian Influenza Virus Induced by the Newcastle Disease Virus H5 Vaccine in Ducks. Avian Dis. 2012, 56, 940–948. [Google Scholar] [CrossRef]
  91. Wang, J.; Cong, Y.; Yin, R.; Feng, N.; Yang, S.; Xia, X.; Xiao, Y.; Wang, W.; Liu, X.; Hu, S.; et al. Generation and evaluation of a recombinant genotype VII Newcastle disease virus expressing VP3 protein of Goose parvovirus as a bivalent vaccine in goslings. Virus Res. 2015, 203, 77–83. [Google Scholar] [CrossRef]
  92. Xu, D.; Li, C.; Liu, G.; Chen, Z.; Jia, R. Generation and evaluation of a recombinant goose origin Newcastle disease virus expressing Cap protein of goose origin avastrovirus as a bivalent vaccine in goslings. Poult. Sci. 2019, 98, 4426–4432. [Google Scholar] [CrossRef]
  93. Ge, J.; Wang, X.; Tian, M.; Gao, Y.; Wen, Z.; Yu, G.; Zhou, W.; Zu, S.; Bu, Z. Recombinant Newcastle disease viral vector expressing hemagglutinin or fusion of canine distemper virus is safe and immunogenic in minks. Vaccine 2015, 33, 2457–2462. [Google Scholar] [CrossRef]
  94. Olbert, M.; Römer-Oberdörfer, A.; Herden, C.; Malberg, S.; Runge, S.; Staeheli, P.; Rubbenstroth, D. Viral vector vaccines expressing nucleoprotein and phosphoprotein genes of avian bornaviruses ameliorate homologous challenge infections in cockatiels and common canaries. Sci. Rep. 2016, 6, 36840. [Google Scholar] [CrossRef]
  95. Xu, L.; Qin, Z.; Qiao, L.; Wen, J.; Shao, H.; Wen, G.; Pan, Z. Characterization of thermostable Newcastle disease virus recombinants expressing the hemagglutinin of H5N1 avian influenza virus as bivalent vaccine candidates. Vaccine 2020, 38, 1690–1699. [Google Scholar] [CrossRef] [PubMed]
  96. Steglich, C.; Grund, C.; Ramp, K.; Breithaupt, A.; Höper, D.; Keil, G.; Veits, J.; Ziller, M.; Granzow, H.; Mettenleiter, T.C. Chimeric newcastle disease virus protects chickens against avian influenza in the presence of maternally derived NDV immunity. PLoS ONE 2013, 8, e72530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Kim, S.-H.; Paldurai, A.; Samal, S.K. A novel chimeric Newcastle disease virus vectored vaccine against highly pathogenic avian influenza virus. Virology 2017, 503, 31–36. [Google Scholar] [CrossRef] [PubMed]
  98. Liu, J.; Xue, L.; Hu, S.; Cheng, H.; Deng, Y.; Hu, Z.; Wang, X.; Liu, X. Chimeric Newcastle disease virus-vectored vaccine protects chickens against H9N2 avian influenza virus in the presence of pre-existing NDV immunity. Arch. Virol. 2018, 163, 3365–3371. [Google Scholar] [CrossRef] [PubMed]
  99. Yadav, K.; Pathak, D.C.; Saikia, D.P.; Debnath, A.; Ramakrishnan, S.; Dey, S.; Chellappa, M.M. Generation and evaluation of a recombinant Newcastle disease virus strain R2B with an altered fusion protein cleavage site as a vaccine candidate. Microb. Pathog. 2018, 118, 230–237. [Google Scholar] [CrossRef]
  100. Arifin, M.A.; Mel, M.; Abdul Karim, M.I.; Ideris, A. Production of Newcastle Disease Virus by Vero Cells Grown on Cytodex 1 Microcarriers in a 2-Litre Stirred Tank Bioreactor. J. Biomed. Biotechnol. 2010, 2010, 586363. [Google Scholar] [CrossRef] [Green Version]
  101. Jaafar, J.N. Investigation of the Effectiveness of Newcastle Disease Virus Production in Different Bioreactors; International Islamic University Malaysia: Gombak, Malaysia, 2009. [Google Scholar]
  102. Arifin, M.A. Optimization of Cell Culture Conditions for the Production of Newcastle Disease Virus; International Islamic University Malaysia: Kuala Lumpur, Malaysia, 2011. [Google Scholar]
  103. Al-Ziaydi, A.G.; Al-Shammari, A.M.; Hamzah, M.I. Propagation of oncolytic Newcastle Disease Virus in Embryonated Chicken Eggs and its Research Applications in Cell lines. In Proceedings of the 1st International Virtual Conference on Pure Science, Qadisiyah, Iraq, 10–11 June 2020; College of Science, University of Al-Qadisiyah: Al-Qadisiyah, Iraq, 2020; Volume 1664, p. 012129. [Google Scholar]
  104. Chen, L.-M.; Donis, R.O.; Suarez, D.L.; Wentworth, D.E.; Webby, R.; Engelhardt, O.G.; Swayne, D.E. Biosafety risk assessment for production of candidate vaccine viruses to protect humans from zoonotic highly pathogenic avian influenza viruses. Influenza Other Respir. Viruses 2020, 14, 215–225. [Google Scholar] [CrossRef] [PubMed]
  105. Sharma, R.; Harrison, S.T.L.; Tai, S.L. Advances in Bioreactor Systems for the Production of Biologicals in Mammalian Cells. ChemBioEng Rev. 2022, 9, 42–62. [Google Scholar] [CrossRef]
  106. Dey, S.; Chellappa, M.M.; Pathak, D.C.; Gaikwad, S.; Yadav, K.; Ramakrishnan, S.; Vakharia, V.N. Newcastle Disease Virus Vectored Bivalent Vaccine against Virulent Infectious Bursal Disease and Newcastle Disease of Chickens. Vaccines 2017, 5, 31. [Google Scholar] [CrossRef] [Green Version]
  107. Le Ru, A.; Jacob, D.; Transfiguracion, J.; Ansorge, S.; Henry, O.; Kamen, A.A. Scalable production of influenza virus in HEK-293 cells for efficient vaccine manufacturing. Vaccine 2010, 28, 3661–3671. [Google Scholar] [CrossRef]
  108. Kiesslich, S.; Kamen, A.A. Vero cell upstream bioprocess development for the production of viral vectors and vaccines. Biotechnol. Adv. 2020, 44, 107608. [Google Scholar] [CrossRef] [PubMed]
  109. Ge, J.; Wang, X.; Tao, L.; Wen, Z.; Feng, N.; Yang, S.; Xia, X.; Yang, C.; Chen, H.; Bu, Z. Newcastle Disease Virus-Vectored Rabies Vaccine Is Safe, Highly Immunogenic, and Provides Long-Lasting Protection in Dogs and Cats. J. Virol. 2011, 85, 8241–8252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Debnath, A.; Pathak, D.C.; D’silva, A.L.; Batheja, R.; Ramamurthy, N.; Vakharia, V.N.; Chellappa, M.M.; Dey, S. Newcastle disease virus vectored rabies vaccine induces strong humoral and cell mediated immune responses in mice. Vet. Microbiol. 2020, 251, 108890. [Google Scholar] [CrossRef] [PubMed]
  111. Kiesslich, S.; Vila-Chã Losa, J.P.; Gélinas, J.-F.; Kamen, A.A. Serum-free production of rVSV-ZEBOV in Vero cells: Microcarrier bioreactor versus scale-X™ hydro fixed-bed. J. Biotechnol. 2020, 310, 32–39. [Google Scholar] [CrossRef] [PubMed]
  112. Shen, C.F.; Guilbault, C.; Li, X.; Elahi, S.M.; Ansorge, S.; Kamen, A.; Gilbert, R. Development of suspension adapted Vero cell culture process technology for production of viral vaccines. Vaccine 2019, 37, 6996–7002. [Google Scholar] [CrossRef]
  113. Segura, M.M.; Kamen, A.A.; Garnier, A. Overview of current scalable methods for purification of viral vectors. Viral Vectors Gene Ther. 2011, 737, 89–116. [Google Scholar]
  114. Moreira, A.S.; Cavaco, D.G.; Faria, T.Q.; Alves, P.M.; Carrondo, M.J.T.; Peixoto, C. Advances in Lentivirus Purification. Biotechnol. J. 2021, 16, 2000019. [Google Scholar] [CrossRef]
  115. Fakri, F.Z.; Bamouh, Z.; Elmejdoub, S.; Elkarhat, Z.; Tadlaoui, K.; Chen, W.; Bu, Z.; Elharrak, M. Long term immunity against Peste Des Petits Ruminants mediated by a recombinant Newcastle disease virus vaccine. Vet. Microbiol. 2021, 261, 109201. [Google Scholar] [CrossRef]
  116. Swayne, D.E.; Suarez, D.L.; Schultz-Cherry, S.; Tumpey, T.M.; King, D.J.; Nakaya, T.; Palese, P.; Garcia-Sastre, A. Recombinant Paramyxovirus Type 1-Avian Influenza-H7 Virus as a Vaccine for Protection of Chickens Against Influenza and Newcastle Disease. Avian Dis. 2003, 47, 1047–1050. [Google Scholar] [CrossRef]
  117. Nakaya, Y.; Nakaya, T.; Park, M.-S.; Cros, J.; Imanishi, J.; Palese, P.; García-Sastre, A. Induction of Cellular Immune Responses to Simian Immunodeficiency Virus Gag by Two Recombinant Negative-Strand RNA Virus Vectors. J. Virol. 2004, 78, 9366–9375. [Google Scholar] [CrossRef] [Green Version]
  118. Huang, Z.; Elankumaran, S.; Yunus, A.S.; Samal, S.K. A Recombinant Newcastle Disease Virus (NDV) Expressing VP2 Protein of Infectious Bursal Disease Virus (IBDV) Protects against NDV and IBDV. J. Virol. 2004, 78, 10054–10063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Schröer, D.; Veits, J.; Grund, C.; Dauber, M.; Keil, G.; Granzow, H.; Mettenleiter, T.C.; Römer-Oberdörfer, A. Vaccination with Newcastle Disease Virus Vectored Vaccine Protects Chickens Against Highly Pathogenic H7 Avian Influenza Virus. Avian Dis. 2009, 53, 190–197. [Google Scholar] [CrossRef] [PubMed]
  120. Sarfati-Mizrahi, D.; Lozano-Dubernard, B.; Soto-Priante, E.; Castro-Peralta, F.; Flores-Castro, R.; Loza-Rubio, E.; Gay-Gutiérrez, M. Protective Dose of a Recombinant Newcastle Disease LaSota–Avian Influenza Virus H5 Vaccine Against H5N2 Highly Pathogenic Avian Influenza Virus and Velogenic Viscerotropic Newcastle Disease Virus in Broilers with High Maternal Antibody Levels. Avian Dis. 2010, 54, 239–241. [Google Scholar] [CrossRef] [PubMed]
  121. Kortekaas, J.; Dekker, A.; de Boer, S.M.; Weerdmeester, K.; Vloet, R.P.M.; Wit, A.A.C.d.; Peeters, B.P.H.; Moormann, R.J.M. Intramuscular inoculation of calves with an experimental Newcastle disease virus-based vector vaccine elicits neutralizing antibodies against Rift Valley fever virus. Vaccine 2010, 28, 2271–2276. [Google Scholar] [CrossRef] [PubMed]
  122. Khattar, S.K.; Collins, P.L.; Samal, S.K. Immunization of cattle with recombinant Newcastle disease virus expressing bovine herpesvirus-1 (BHV-1) glycoprotein D induces mucosal and serum antibody responses and provides partial protection against BHV-1. Vaccine 2010, 28, 3159–3170. [Google Scholar] [CrossRef] [PubMed]
  123. Schröer, D.; Veits, J.; Keil, G.; Römer-Oberdörfer, A.; Weber, S.; Mettenleiter, T.C. Efficacy of Newcastle Disease Virus Recombinant Expressing Avian Influenza Virus H6 Hemagglutinin Against Newcastle Disease and Low Pathogenic Avian Influenza in Chickens and Turkeys. Avian Dis. 2011, 55, 201–211. [Google Scholar] [CrossRef]
  124. Hu, H.; Roth, J.P.; Estevez, C.N.; Zsak, L.; Liu, B.; Yu, Q. Generation and evaluation of a recombinant Newcastle disease virus expressing the glycoprotein (G) of avian metapneumovirus subgroup C as a bivalent vaccine in turkeys. Vaccine 2011, 29, 8624–8633. [Google Scholar] [CrossRef]
  125. Goff, P.H.; Krammer, F.; Hai, R.; Seibert, C.W.; Margine, I.; García-Sastre, A.; Palese, P. Induction of Cross-Reactive Antibodies to Novel H7N9 Influenza Virus by Recombinant Newcastle Disease Virus Expressing a North American Lineage H7 Subtype Hemagglutinin. J. Virol. 2013, 87, 8235–8240. [Google Scholar] [CrossRef] [Green Version]
  126. Toro, H.; Zhao, W.; Breedlove, C.; Zhang, Z.; van Santen, V.; Yu, Q. Infectious Bronchitis Virus S2 Expressed from Recombinant Virus Confers Broad Protection Against Challenge. Avian Dis. 2013, 58, 83–89. [Google Scholar] [CrossRef]
  127. Ge, J.; Wang, X.; Tian, M.; Wen, Z.; Feng, Q.; Qi, X.; Gao, H.; Wang, X.; Bu, Z. Novel in-ovo chimeric recombinant Newcastle disease vaccine protects against both Newcastle disease and infectious bursal disease. Vaccine 2014, 32, 1514–1521. [Google Scholar] [CrossRef]
  128. Liu, Q.; Mena, I.; Ma, J.; Bawa, B.; Krammer, F.; Lyoo, Y.S.; Lang, Y.; Morozov, I.; Mahardika, G.N.; Ma, W.; et al. Newcastle Disease Virus-Vectored H7 and H5 Live Vaccines Protect Chickens from Challenge with H7N9 or H5N1 Avian Influenza Viruses. J. Virol. 2015, 89, 7401–7408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Zhang, M.; Ge, J.; Li, X.; Chen, W.; Wang, X.; Wen, Z.; Bu, Z. Protective efficacy of a recombinant Newcastle disease virus expressing glycoprotein of vesicular stomatitis virus in mice. Virol. J. 2016, 13, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Nagy, A.; Lee, J.; Mena, I.; Henningson, J.; Li, Y.; Ma, J.; Duff, M.; Li, Y.; Lang, Y.; Yang, J.; et al. Recombinant Newcastle disease virus expressing H9 HA protects chickens against heterologous avian influenza H9N2 virus challenge. Vaccine 2016, 34, 2537–2545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Wang, J.; Yang, J.; Ge, J.; Hua, R.; Liu, R.; Li, X.; Wang, X.; Shao, Y.; Sun, E.; Wu, D.; et al. Newcastle disease virus-vectored West Nile fever vaccine is immunogenic in mammals and poultry. Virol. J. 2016, 13, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Zhang, M.; Ge, J.; Wen, Z.; Chen, W.; Wang, X.; Liu, R.; Bu, Z. Characterization of a recombinant Newcastle disease virus expressing the glycoprotein of bovine ephemeral fever virus. Arch. Virol. 2017, 162, 359–367. [Google Scholar] [CrossRef] [Green Version]
  133. Zhao, R.; Sun, J.; Qi, T.; Zhao, W.; Han, Z.; Yang, X.; Liu, S. Recombinant Newcastle disease virus expressing the infectious bronchitis virus S1 gene protects chickens against Newcastle disease virus and infectious bronchitis virus challenge. Vaccine 2017, 35, 2435–2442. [Google Scholar] [CrossRef]
  134. Liu, R.-q.; Ge, J.-y.; Wang, J.-l.; Shao, Y.; Zhang, H.-l.; Wang, J.-l.; Wen, Z.-y.; Bu, Z.-g. Newcastle disease virus-based MERS-CoV candidate vaccine elicits high-level and lasting neutralizing antibodies in Bactrian camels. J. Integr. Agric. 2017, 16, 2264–2273. [Google Scholar] [CrossRef]
  135. Ma, J.; Lee, J.; Liu, H.; Mena, I.; Davis, A.S.; Sunwoo, S.Y.; Lang, Y.; Duff, M.; Morozov, I.; Li, Y.; et al. Newcastle disease virus-based H5 influenza vaccine protects chickens from lethal challenge with a highly pathogenic H5N2 avian influenza virus. npj Vaccines 2017, 2, 33. [Google Scholar] [CrossRef] [Green Version]
  136. Hu, Z.; Liu, X.; Jiao, X.; Liu, X. Newcastle disease virus (NDV) recombinant expressing the hemagglutinin of H7N9 avian influenza virus protects chickens against NDV and highly pathogenic avian influenza A (H7N9) virus challenges. Vaccine 2017, 35, 6585–6590. [Google Scholar] [CrossRef]
  137. Shi, L.; Hu, Z.; Hu, J.; Liu, D.; He, L.; Liu, J.; Gu, H.; Gan, J.; Wang, X.; Liu, X. Single Immunization with Newcastle Disease Virus-Vectored H7N9 Vaccine Confers a Complete Protection against Challenge with Highly Pathogenic Avian Influenza H7N9 Virus. Avian Dis. 2018, 63, 61–67. [Google Scholar] [CrossRef]
  138. Xu, X.; Xue, C.; Liu, X.; Li, J.; Fei, Y.; Liu, Z.; Mu, J.; Bi, Y.; Qian, J.; Yin, R.; et al. A novel recombinant attenuated Newcastle disease virus expressing H9 subtype hemagglutinin protected chickens from challenge by genotype VII virulent Newcastle disease virus and H9N2 avian influenza virus. Vet. Microbiol. 2019, 228, 173–180. [Google Scholar] [CrossRef] [PubMed]
  139. Abozeid, H.H.; Paldurai, A.; Varghese, B.P.; Khattar, S.K.; Afifi, M.A.; Zouelfakkar, S.; El-Deeb, A.H.; El-Kady, M.F.; Samal, S.K. Development of a recombinant Newcastle disease virus-vectored vaccine for infectious bronchitis virus variant strains circulating in Egypt. Vet. Res. 2019, 50, 12. [Google Scholar] [CrossRef] [PubMed]
  140. Kumar, R.; Kumar, V.; Kekungu, P.; Barman, N.N.; Kumar, S. Evaluation of surface glycoproteins of classical swine fever virus as immunogens and reagents for serological diagnosis of infections in pigs: A recombinant Newcastle disease virus approach. Arch. Virol. 2019, 164, 3007–3017. [Google Scholar] [CrossRef] [PubMed]
  141. Tan, L.; Wen, G.; Qiu, X.; Yuan, Y.; Meng, C.; Sun, Y.; Liao, Y.; Song, C.; Liu, W.; Shi, Y.; et al. A Recombinant La Sota Vaccine Strain Expressing Multiple Epitopes of Infectious Bronchitis Virus (IBV) Protects Specific Pathogen-Free (SPF) Chickens against IBV and NDV Challenges. Vaccines 2019, 7, 170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Murr, M.; Hoffmann, B.; Grund, C.; Römer-Oberdörfer, A.; Mettenleiter, T.C. A Novel Recombinant Newcastle Disease Virus Vectored DIVA Vaccine against Peste des Petits Ruminants in Goats. Vaccines 2020, 8, 205. [Google Scholar] [CrossRef] [PubMed]
  143. Qin, Y.; Tu, K.; Teng, Q.; Feng, D.; Zhao, Y.; Zhang, G.; Gallagher, T. Identification of Novel T-Cell Epitopes on Infectious Bronchitis Virus N Protein and Development of a Multi-epitope Vaccine. J. Virol. 2021, 95, e00667-21. [Google Scholar] [CrossRef]
  144. Lee, J.; Kim, D.-H.; Noh, J.; Youk, S.; Jeong, J.-H.; Lee, J.-B.; Park, S.-Y.; Choi, I.-S.; Lee, S.-W.; Song, C.-S. Live Recombinant NDV-Vectored H5 Vaccine Protects Chickens and Domestic Ducks from Lethal Infection of the Highly Pathogenic H5N6 Avian Influenza Virus. Front. Vet. Sci. 2022, 8, 773715. [Google Scholar] [CrossRef]
Figure 1. A representation of the NDV single-stranded negative-sense RNA genome including the six native transcriptional units. Transgenes are typically inserted between the P and M genes and flanked by NDV gene start (GS) and gene end (GE) sequences. The total genome length must be an even multiple of six (“rule of six”) to ensure complete encapsidation.
Figure 1. A representation of the NDV single-stranded negative-sense RNA genome including the six native transcriptional units. Transgenes are typically inserted between the P and M genes and flanked by NDV gene start (GS) and gene end (GE) sequences. The total genome length must be an even multiple of six (“rule of six”) to ensure complete encapsidation.
Viruses 14 00975 g001
Figure 2. Overview of production processes for viral-vectored vaccines in (A) embryonated chicken eggs (ECEs) and (B) suspension cell cultures in stirred-tank bioreactors.
Figure 2. Overview of production processes for viral-vectored vaccines in (A) embryonated chicken eggs (ECEs) and (B) suspension cell cultures in stirred-tank bioreactors.
Viruses 14 00975 g002
Table 1. NDV-vectored vaccine candidates for human use in chronological order (publication date).
Table 1. NDV-vectored vaccine candidates for human use in chronological order (publication date).
Vaccine TypePathogenDiseaseAntigenAnimal ModelProduction Platform *Route *Dose *Reference
Live B1 strainInfluenza A H1N1Respiratory infectionHAMouseECEsiv, iptwo doses; 3 × 107 PFU[65]
Live LaSota or BC strainHPIV3Respiratory infectionHNAfrican green monkey; rhesus monkeyECEs, DF-1 cell linein + ittwo doses; 3 × 106 PFU[20]
Live B1 strainHRSVRespiratory infectionFMouseECEsinone dose; 5 × 105 PFU[66]
Live BC or LaSota/VF strainSARS-CoVRespiratory infectionS, S1African green monkeyECEs, DF-1 cell linein + itone or two doses; 2 × 107 PFU[28]
Live LaSota strainHPAIV H5N1Respiratory infectionHAAfrican green monkeyDF-1 cell lineintwo doses; 2 × 107 PFU[40]
Live B1 strainHIVAIDSGagMouseECEs, Vero cell lineintwo doses; 5 × 105 PFU prime and 106 PFU boost[67]
Live LaSota or BC strainEBOVEbola virus disease (hemorrhagic fever)GPRhesus monkeyDF-1 cell linein + ittwo doses; 107 PFU[29]
Live LaSota strainHIVAIDSGagMouseECEsintwo doses; 5 × 105 PFU prime and 106 PFU boost[68]
Live LaSota/VF strainBorrelia burgdorferiLymeBmpA, OspCHamsterECEsin, im, ipone or two doses; 106 PFU[69]
Live LaSota strainHIVAIDSEnv (gp160)Guinea pigECEsin, im, in + imtwo or three doses; 3 × 105 PFU (in) or 5 × 105 PFU (im)[70]
Live LaSota strainNiVEncephalitisF, GMouse, pigECEsimtwo doses; 108 EID50 (mice) or 2 × 109 EID50 (pigs)[71]
Live LaSota or Lasota/BC strainNoVGastroenteritisVP1MouseECEsinone dose; 106 EID50[72]
Live LaSota strainHIVAIDSEnv (gp160)/Gag (p55)Guinea pig, mouseECEsintwo doses; 2 × 105 PFU (guinea pigs) or 4 × 104 PFU (mice)[50]
Live LaSota strainHIVAIDSEnv (gp160)Guinea pigECEsinfour doses of 2 × 105 PFU or two doses of 2 × 105 PFU followed by two doses of recombinant protein (gp120 or gp140)[73]
Live LaSota strainPoliovirusPoliomyelitisP1/3CDGuinea pigECEsintwo doses; 105 PFU[49]
Live chimeric NDV strainEBOVEbola virus disease (hemorrhagic fever)GPGuinea pigECEsintwo doses; 2 × 106 TCID50[13]
Live LaSota strainJEVEncephalitisE, NS1MouseECEsinone dose; 106 EID50[74]
Live LaSota strainSARS-CoV-2COVID-19S, S-F chimeraMouseECEsintwo doses; 10 or 50 μg[18]
Inactivated LaSota strainSARS-CoV-2COVID-19S, S-F chimeraMouse, hamsterECEsimtwo doses; 5 or 10 μg[75]
Live B1 strainSARS-CoV-2COVID-19SMouse, hamsterECEsinone or two doses; 104 PFU (mice) or 106 PFU (hamster)[26]
Live or inactivated LaSota strainSARS-CoV-2COVID-19HXP-SPigECEsin, im, in + imtwo doses of 107, 3 × 107, 108 or 3 × 108 EID50 (live); two doses of 108 EID50 (inactivated)[31]
Live or inactivated LaSota strainSARS-CoV-2COVID-19HXP-SHamster, mouseECEsin, im, in + imtwo doses of 1.0, 0.3, 0.1, 0.03 or 0.03 μg (im, inactivated, hamsters); two doses of 106 EID50 (in, live, hamsters); two doses of 104, 105 or 106 EID50 (in prime and im boost, live, mice); two doses of 1 μg (im, inactivated, mice)[76]
Live LaSota strainSARS-CoV-2COVID-19HXP-SRatECEsin, im, in + imtwo doses; 7.4 × 108 EID50[30]
Live LaSota strainSARS-CoV-2COVID-19S, truncated SHamsterECEsinone or two doses; 107 PFU[27]
Live LaSota strainEBOVEbola virus disease (hemorrhagic fever)GPMouseECEsintwo doses of 106 PFU; one dose of 106 PFU and one dose of adenovirus-vectored vaccine[77]
Inactivated VG/GA strainSARS-CoV-2COVID-19RBDMouseECEsimtwo doses of 1, 5 or 10 μg[78]
* ECEs: embryonated chicken eggs, iv: intravenous, ip: intraperitoneal, in: intranasal, it: intratracheal, im: intramuscular, PFU: plaque-forming units, TCID50: tissue culture infectious dose 50%, EID50: embryo infectious dose 50%.
Table 2. Recombinant NDV-vectored vaccine candidate in clinical trials for human use.
Table 2. Recombinant NDV-vectored vaccine candidate in clinical trials for human use.
Responsible GroupVaccine TypePathogenDiseaseAntigenPhaseRoute *Dose *Reference
Icahn School of Medicine at Mount Sinai, USALive LaSota strainSARS-CoV-2COVID-19HXP-SIin, im,
in + im
3.3 × 108 EID50
1 × 109 EID50
NCT05181709
Laboratorio Avi-Mex, MexicoLive LaSota strainSARS-CoV-2COVID-19HXP-SI/IIim, in + im108 EID50NCT04871737 [79]
NCT05205746
Institute of Vaccines and Medical Biologicals,
Vietnam
Inactivated LaSota strainSARS-CoV-2COVID-19HXP-SI/IIim1, 3 or 10 μgNCT04830800
Butantan Institute, BrazilInactivated LaSota strainSARS-CoV-2COVID-19HXP-SI/IIim1, 3 or 10 μgNCT04993209
Mahidol University, ThailandInactivated LaSota strainSARS-CoV-2COVID-19HXP-SI/IIim1, 3 or 10 μgNCT04764422 [80]
* in: intranasal, im: intramuscular, EID50: embryo infectious dose 50%.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fulber, J.P.C.; Kamen, A.A. Development and Scalable Production of Newcastle Disease Virus-Vectored Vaccines for Human and Veterinary Use. Viruses 2022, 14, 975. https://doi.org/10.3390/v14050975

AMA Style

Fulber JPC, Kamen AA. Development and Scalable Production of Newcastle Disease Virus-Vectored Vaccines for Human and Veterinary Use. Viruses. 2022; 14(5):975. https://doi.org/10.3390/v14050975

Chicago/Turabian Style

Fulber, Julia P. C., and Amine A. Kamen. 2022. "Development and Scalable Production of Newcastle Disease Virus-Vectored Vaccines for Human and Veterinary Use" Viruses 14, no. 5: 975. https://doi.org/10.3390/v14050975

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop