key: cord-0872884-k3cme2s2 authors: Ayalew, Lisanework E.; Kumar, Pankaj; Gaba, Amit; Makadiya, Niraj; Tikoo, Suresh K. title: Bovine adenovirus-3 as a vaccine delivery vehicle date: 2015-01-15 journal: Vaccine DOI: 10.1016/j.vaccine.2014.11.055 sha: 7f60c9d00f903457244719182bd27821116dcfa6 doc_id: 872884 cord_uid: k3cme2s2 The use of vaccines is an effective and relatively inexpensive means of controlling infectious diseases, which cause heavy economic losses to the livestock industry through animal loss, decreased productivity, treatment expenses and decreased carcass quality. However, some vaccines produced by conventional means are imperfect in many respects including virulence, safety and efficacy. Moreover, there are no vaccines for some animal diseases. Although genetic engineering has provided new ways of producing effective vaccines, the cost of production for veterinary use is a critical criterion for selecting the method of production and delivery of vaccines. The cost effective production and intrinsic ability to enter cells has made adenovirus vectors a highly efficient tool for delivery of vaccine antigens. Moreover, adenoviruses induce both humoral and cellular immune responses to expressed vaccine antigens. Since nonhuman adenoviruses are species specific, the development of animal specific adenoviruses as vaccine delivery vectors is being evaluated. This review summarizes the work related to the development of bovine adenovirus-3 as a vaccine delivery vehicle in animals, particularly cattle. Adenoviruses (AdVs) are non-enveloped double stranded DNA viruses with icosahedral capsid symmetry composed of 252 capsomers. The genome size ranges between 24 and 45 kb [1, 2] . Although adenoviruses were first identified in the early 1950s from the adenoids of humans with acute respiratory infection [3, 4] , today, there are over 120 serotypes, isolated from different species, including, mammals, birds, reptiles and fish. While adenoviruses can infect a wide variety of animals, including humans, birds and livestock, they are usually species-specific [2, 5, 6] . AdVs have been proposed to be clustered in five phylogenitically distinct groups namely; Mastadenovirus, Aviadenovirus, Atadenovirus, Siadenovirus and Ichtadenovirus, with each virus group having a distinct and characteristic genome structure [7, 8] . The overall genetic organization of members of the Mastadenovirus including human adenovirus (HAdV)-5 [9] , canine adenovirus (CAdV)-2 [10] , Simian adenovirus (SAdV) [11] , bovine adenovirus (BAdV)-3 [12] , porcine adenovirus (PAdV)-5 [13] and PAdV-3 [14] appears to be conserved. The Mastadenovirus genome is divided into five discrete transcriptional units. The early regions E1 and E4 are located at either end of the genome, while E2 and E3 are separate and internal. The central core region constitutes the late region. In addition, there are two delayed early transcriptional units named IVa2 and pIX. Despite this genetic conservation, some regions are highly divergent (based on structure of transcripts and proteins) among different members of Mastadenoviruses. In particular, the deduced amino acid sequence of ORFs in the E1, E3 and E4 regions of BAdV-3 show little or no homology to the corresponding proteins encoded by genomes of other members of Mastadenoviruses [12] . Since these proteins are involved in virus cell interaction and viral gene expression, variations in these proteins may reflect the diverse host range and pathogenic potential of different members of Mastadenoviruses. In recent years, much attention has been focused on evaluating adenoviruses as viral vectors due to their ability to infect both dividing and non-dividing cells, capacity to package large foreign genes, relative ease to produce high titer recombinants in cell culture [15] , elicit strong antigen specific T cell responses and lack of virulence [16, 17] . Although recombinant HAdVs have been proven to deliver vaccine antigens to domestic animals [18] [19] [20] [21] and birds [22, 23] , regulatory concerns regarding safety has limited their use in domestic animals. Moreover, species specificity limiting host range, restricted replication in non host species and http://dx.doi.org/10.1016/j.vaccine.2014.11.055 0264-410X/© 2014 Elsevier Ltd. All rights reserved. stability of nonhuman adenoviruses has led to the evaluation of animal [CAdV-2 [24] , PAdV-3 [25] , PAdV-5 [26] , BAdV-3 [27, 28] and poultry (Fowl adenovirus (FAdV) [29] specific adenoviruses as vaccine delivery vectors. Since BAdV-3 is a natural nonpathogenic virus with restricted host-range, grows to high titers and can be delivered intranasally without affecting the meat quality of food producing animals, BAdV-3 is being evaluated as vaccine delivery vector in animals including cattle. BAdV-3 is 75 nm in diameter, non enveloped icosahedral particle containing a double stranded DNA genome. The genome of BAdV-3 is 34, 446 base pairs [12] flanked on either end by 195 bp inverted terminal repeats (ITRs) [30] , which play a key role in DNA replication. Unlike other adenoviruses, the ITRs of BAdV-3 are longer and contain a high GC content [12] . Like other adenoviruses, the packaging domain is located in the left end of the viral genome overlapping the transcriptional control region of E1A, but E1A region expression distinctively appears to be essential for packaging in BAdV-3 [31, 32] . Though the TATA or CAAT boxes are absent in the regions between the left ITR and upstream of the E1A start codon, the expression of the E1A open reading frame is driven by a promoter located within the ITR [33] . Based on the transcriptional analysis [12, [34] [35] [36] [37] , BAdV-3 genome appears to be organized ( Fig. 1) into early, intermediate and late regions [12] . Although genome organization appears similar to other members of Mastadenoviruses, the structure and function of some proteins encoded by different regions of BAdV-3 appears to be different [38] [39] [40] [41] [42] [43] [44] . Initial attempts to generate recombinant BAdV-3 using homologous recombination in Madin Darby bovine kidney (MDBK) cells co-transfected with purified BAdV-3 genome and a transfer vector containing a deletion in E3 region were inefficient and mostly unsuccessful [45] [46] [47] due to low transfection efficiency and inefficient recombination in MDBK cells. However, two improvements namely availability of homologous recombination machinery of Escherichia coli [48] and use of bovine retina cells for transfection of restriction enzyme excised modified BAdV-3 genome from plasmids has facilitated the efficient generation of recombinant BAdV-3 [47, 49] . Recently, a more efficient and time saving method of generating recombinant BAdV-3 has been reported by transfection of endonuclease I-SceI expressing non bovine (cotton rat lung) cells with circular BAdV-3 genomic DNA flanked by I-SceI recognition site [50] . Isolation of replication competent BAdV-3 containing partial deletion of E1B small suggested that this region is not essential for replication of BAdV-3 [51] in fetal bovine retina cells and may be used for foreign gene insertion. However, viable recombinant BAdV-3 with E1A [52] [53] [54] or E1B large deletion [33, 52] could not be rescued suggesting that these regions are essential for replication of BAdV-3. However, packaging cell lines expressing E1 region of BAdV-3 (FBK-34; MDBK-221; [53] ) or HAdV-5 (VIDO R2, FBRT-HE1; BHH3 and BHH8 [53, 54] supported the replication of only E1A (541 bp) deleted BAdV-3 ( Fig. 2) and not complete E1 region deleted BAdV-3 [52] . Replication-defective BAdV-3s containing insertion of 2.3 kb of foreign DNA in E1A region of E1-E3 deleted BAdV-3 could be isolated only in E1A complementing cell lines [52] . Since replication-defective BAdV-3 vector cannot undergo multiple replication cycle and therefore are less likely to spread to the environment, they are considered safer. One of the major concerns with regard to the use of live viral vectors for vaccination in animals is the release of potential recombinants into the environment. However, since the replication-incompetent recombinants undergo an abortive infection, the level of foreign gene expression is lower than that obtained using replication-competent BAdV-3. The E3 region of BAdV-3 is 1.591 kb. Since E3 region is non essential for replication of adenoviruses [46, 55] , initial attempts resulted in isolating a viable recombinant BAdV-3 containing deletion of 1.245 kb of E3 region [46] . Although potential insertion capacity of E3 deleted vector is 3 kb [46] , the insertion of 2.8 kb foreign DNA [56] has been successful in generating viable replication-competent recombinant BAdV-3. The foreign open reading frames (ORFs) inserted in E3 are efficiently expressed using upstream endogenous (E3/MLP) promoters [28, 46, 56, 57] or exogenous promoters [56, 57] . However, insertion of foreign ORFs antiparallel to E3 transcription does not lead to the generation of viable recombinant BAdV-3. Moreover, addition of exogenous consensus sequence for polyadenylation of foreign ORF affects the replication efficiency of recombinant BAdV-3 (Lobanov and Tikoo, unpublished data). Interestingly, efficient expression of a RNA virus required addition of an intron and consensus sequence for polyadenylation upstream and downstream, respectively, of the gene [55] . Since E3 deleted recombinant BAdV-3 are replication competent (Fig. 2) , less amount of virus may be required to induce a protective immune response. The first site for insertion of foreign ORF has been identified in a transcriptionally inactive region between start of E4 and right ITR of BAdV-3 [12, 34] . Isolation of replication competent BAdV-3 containing insertion of 1.9 kb foreign DNA at nucleotide (34059) suggested that this region is non-essential for BAdV-3 replication (Fig. 2) . However, exogenous promoter and consensus sequence for polyadenylation are required for expression of the foreign ORF. The other sites for potential insertion of foreign ORF have been identified in transcriptionally active E4 region of BAdV-3 [58] . Isolation of replication competent BAdV-3 containing deletion of N-terminus 1.501 kb or C-terminus 1.342 kb (Fig. 2 ) in E4 region suggested that these regions are not essential for replication of BAdV-3 [58] , thus increasing the potential insertion capacity of E3-E4 deleted BAdV-3 to 4.5 kb [58] . Much of the initial efforts of developing replication-competent BAdV-3 based vectors have focused on non-essential E3 region. Earlier, successful expression of a reporter ORF inserted in partially deleted E3 region of BAdV-3 [45] suggested the feasibility of utilizing E3 region for expression of foreign genes. Thus, subsequent expression of different forms of (BHV)-1 vaccine antigen gD ORF [46] , (BVDV) E2 ORF [57] or (BCV) virus HE ORF [55] inserted individually in fully deleted E3 region of BAdV-3 demonstrated the potential of developing BAdV-3 based recombinant vaccines for cattle. These ORFs were selected as earlier reports have suggested the potential of bovine herpesvirus (BHV)-1 glycoprotein gD [59] , bovine viral diarrhea virus (BVDV) E2 glycoprotein [60] 40 78 87 Late (L) Region corona virus (BCV) hemagglutinin ORF [61] to induce protective immune responses in animals and thus act as vaccine antigen. Reports suggest that cotton rats can serve as small-animal model for investigation of immune responses to BAdV-3 vectored vaccines [62] . The feasibility of using replication-competent recombinant BAdV-3 in inducing antigen specific immune responses in animals was demonstrated by immunizing cotton rats twice intranasally three weeks apart, with recombinant BAdV-3 expressing BHV-1 gD/gDt [46, 56] or BVDV E2 protein [57] . One of the concerns with the use of BAdV-3 as a vector has been the presence of varying levels of BAdV-3 vector specific pre-existing antibodies in calves in the field [27, [63] [64] [65] [66] . To address the concern, the induction of BAdV-3 specific immune response was analyzed in calves containing significant level of pre-existing BAdV-3 specific neutralizing antibodies. The results suggested that intranasal immunization of calves induced significant BAdV-3 specific antibody responses in the presence of BAdV-3 specific neutralizing antibodies [67] . Next, the usefulness of replication-competent BAdV-3 as an effective vaccine delivery vehicle was tested in natural host (calves) containing significant level of pre-existing BAdV-3 specific neutralizing antibodies [27] . We demonstrated the induction of protective immune responses against BHV-1 challenge in calves immunized with recombinant replication-competent BAdV-3 expressing BHV-1 gD/gDt [27] . In this study, calves were immunized twice intranasally, four weeks apart and challenged two weeks later with BHV-1. The immunized calves were protected from clinical disease and showed BHV-1 shedding for reduced time [27] . Thus, species-specific vectors can be developed and used to induce mucosal immunity and protection against a disease in a natural host. Moreover, mucosal (intranasal) immunization helps to bypass the effect of vector (BAdV-3) specific pre-existing antibodies on the induction of protective immune responses. In contrast, intratracheal/subcutaneous immunization with replication-competent recombinant BAdV-3 expressing gDt was not as efficient as intranasal immunization in inducing protective immune responses in calves [65] . Despite the requirement of high doses, replication-defective human adenovirus based vectors have been successfully used in inducing protective immune responses in animals [20, 68, 69, 19] and poultry [22, 70, 71] . In contrast, replication-defective BAdV-3 expressing gDt does not appear efficient in inducing a protective immune response in calves [65] . This could be due to the low amount of replication-defective recombinant used for immunization, which may not be adequate for producing sufficient amount of vaccine antigen [65] . Although the use of high amount of replication-defective BAdV-3 vectors may induce protective immune responses in claves, the cost-effective production of such vaccines may be problematic. Since veterinary vaccines have to be safe and cost effective, the development of a recombinant BAdV-3 vectored vaccine providing immunity to more than one pathogen is desirable. Recently, we demonstrated that single recombinant BAdV-3 expressing vaccine antigens from two respiratory pathogens (gDt of BHV-1 and gG of bovine respiratory syncytial virus [BRSV]) of cattle could be isolated [72] . Moreover, two intranasal immunizations, four weeks apart of calves with single recombinant BAdV-3 expressing BHV-1 gDt and BRSV gG induced significant neutralizing antibodies against BHV-1 and BRSV in cotton rats [72] . Thus, the construction of single recombinant BAdV-3 with multiple vaccine antigens is feasible to induce virus specific immunity for protection from multiple pathogens in calves [72] . Several recent studies in experimental models have demonstrated that it is possible to bias the immune response as required by administration of recombinant cytokines along with vaccine antigens, which has helped to modulate disease progression in animal models [73] [74] [75] [76] [77] . Moreover, inoculation of replication-defective HAdV-5-expressing porcine IFN-␣, protected pigs from foot-andmouth disease (FMD) [78] , and delayed and reduced disease signs in cattle after challenge with FMD virus [79] . However, despite the availability of different bovine cytokine genes, very few studies have been reported investigating the immunomodulation potential of known bovine (Bo) cytokines in cattle [80] [81] [82] . We have demonstrated the feasibility of constructing replication-competent (E3 deleted) recombinant BAdV-3 viruses, expressing bovine BoIFN-␥ [80, 83] (Fig. 3, panels A-C) , BoIL-2 [84] (Fig. 3, panels, A, B and D) , or BoIL-6 [28] genes, inserted into E3 region of BAdV-3 (Fig. 3) . Required effect of cytokine in modulating immune response is best achieved, when both vaccine antigen and cytokine are synthesized in the same cells. To this end, we demonstrated the feasibility of constructing replication competent (E3 deleted) recombinant BAdV-3 expressing antigen-cytokine chimera (gDt-BoIL-6) or expressed antigen-and cytokine as individual proteins (gDt, BoIL-6) from a single mRNA [28] . In addition, the co-expressed gDt retained antigenicity and BoIL-6 retained biological activity [28] . Hence, constructing and evaluating single BAdV-3 expressing two antigens (vaccine antigen and cytokine) is possible. Since gD/gDt expressed by recombinant BAdV-3 did not induce efficient mucosal IgA response required for eliminating BHV-1 shedding in immunized calves, we evaluated the effectiveness of recombinant BAdV-3 co-expressing BHV-1 gDt and BoIL-6 [85] in the immunomodulatory effects of BoIL-6 on induction of BHV-1 gDt specific immune responses in calves [28] . Although co-expression of BoIL-6 did not modulate the gDt specific immune response, the study provided the feasibility of delivering vaccine antigens with other cytokines with potent adjuvant effects to modulate immune responses in cattle. Although BAV-3 vector can efficiently deliver vaccine antigens to respiratory mucosal surfaces of cattle, the prospects of oral delivery of BAdV-3 based vaccines should help in reducing the indirect cost (man power and animal handling) associated with vaccination, thus providing additional economic benefits. Though, BAdV-3s potential use in delivering vaccine antigens to enteric mucosal surfaces of cattle has not been successful, it should be possible to develop BAdV-3 vectors which can deliver vaccine antigens to different mucosal surfaces by genetic modification of capsid proteins. This technique can also be applied to deliver vaccine antigens to other animals (e.g. cats, dogs, poultry). We have tested the feasibility of changing tropism of BAdV-3 by genetic manipulation of major capsid protein fiber and minor capsid protein pIX of BAdV-3. First, the construction of a recombinant BAdV-3 with a chimeric fiber by replacing the knob region of BAdV-3 fiber with the knob region of HAdV-5 fiber alters the tropism of the recombinant BAdV-3 [44] . Second, we have provided proof of principle that BAdV-3 minor capsid protein pIX can be used for stable insertion of longer polypeptides exposed on the surface of BAdV-3 virions [86] . Moreover, incorporation of targeting ligand into the C-terminus of pIX enhanced fiber-knob independent tropism of BAdV-3 [86] . BAdV-3 pIX based targeting is being evaluated in developing improved BAdV-3 vectors for bovine vaccination. Thus, it should be possible to exchange the knob region of BAdV-3 fiber with knob region of an adenovirus fiber with tropism for enteric mucosa of cattle to generate BAdV-3 vectored vaccines that can be delivered orally. Alternatively, it should be possible to develop recombinant BAdV-3 expressing chimeric pIX (pIX containing targeting ligand) recognizing M cells [87] [88] [89] in ileal Peyer's patches of enteric mucosa. Although advances in recombinant DNA technology have led to the identification of potent vaccine antigens, one of the impediments in the development of effective vaccines has been the appropriate delivery of vaccine. Moreover, the development of new generation of veterinary vaccine need to take into account a number of factors including the cost of production, ease of delivery, safety and efficacy. The development and use of vaccines based on BAdV-3 vector system will be cost effect and safe with no risk of producing disease. In addition, these vaccines can also be used for eradication programs since infected and vaccinated animals can be differentiated. Interestingly, intranasal inoculation of calves with wild-type [67] or replication-competent BAdV-3 [27] causes in apparent infection with negligible (number of animals and duration of virus secretion) excreting of virus. Since BAdV-3 has restrictive host range, [35, 44, 90] , can be isolated from a healthy cattle [64] and causes in apparent infection in calves [27, 67] , the use of replication-competent BAdV-3 vector based vaccines in cattle may not be a major regulatory concern for use in the field. Earlier reports have suggested the oncogenicity of BAdV-3 in Hamster [91] and rat embryo cells [92] . However, like other adenoviruses, BAdV-3 is not known to induce tumor formation in cattle (its natural host). In fact, certain replication competent human adenoviruses (containing transforming E1 region) are already in different phases of clinical trial for certain cancer vaccination and have been used as vaccines in humans [93] . Based on the very long substantial safety record of replicationcompetent human adenovirus-4 and 7 vaccines in U.S. military, replication competent adenovirus based vaccines under development are also expected to be safe [93] . The replication-competent adenoviral vectors have a "dose sparing" effect providing an advantage for manufacturing. They also mimic a natural wild-type infection inducing all arms of the immune system including; innate, cellular, humoral and mucosal immunity. Theoretically, replication competent vaccine vectors have the safety of inactivated vaccines and the improved efficacy of live attenuated vaccines [93] . Gomez-Roman et al. demonstrated the detection of vector in the stools of rhesus macaques within a week after vaccination with oral enteric coated replication competent adenovirus-5 based vectors; however, no spread of the virus to the upper respiratory tract was detected and no clinical symptoms of adenovirus induced disease were observed during the 10 weeks of follow up period, proving the safety of the vector and very good tolerance by vaccinated animals [94] . A phase I clinical trial of an oral, replicating adenovirus-4 vector vaccine for H1N1 influenza in humans was also well tolerated and demonstrated to have a good safety profile with low transmissibility [95] . Generally, experimental inoculation of humans or chimpanzees with replication-competent adenovirus vectors has proven to be safe [95, 96] . Similarly, replication competent BAdV-3 vector is expected to be safe and tolerable in cattle [27, 64, 67] . The authors have no conflict of interest to declare. Genetic content and evolution of adenoviruses The viruses and their replication fields virology Isolation of a cytopathogenic agent from human adenoids undergoing spontaneous degeneration in tissue culture Recovery of new agent from patients with acute respiratory illness Adenoviruses: group name proposed for new respiratory tract viruses The viruses and their replication Family adenoviridae Family adenoviridae The sequence of the genome of adenovirus type 5 and its comparison with the genome of adenovirus type 2 Functional organization of the major late transcriptional unit of canine adenovirus type 2 Complete genome sequence of simian adenovirus 1: an old world monkey adenovirus with two fiber genes Nucleotide sequence, genome organization, and transcription map of bovine adenovirus type 3 The complete nucleotide sequence of porcine adenovirus sero-type 5 Nucleotide sequence and transcription map of porcine adenovirus type 3 Update on adenovirus and its vectors Detailed analysis of the CD8+ T-cell response following adenovirus vaccination T-cell immunity generated by recombinant adenovirus vaccines A recombinant human adenovirus vaccine against rabies Tropism of human adenovirus type 5-based vectors in swine and their ability to protect against transmissible gastroenteritis coronavirus Induction of protective immunity to bovine herpesvirus type 1 in cattle by intranasal administration of replication-defective human adenovirus type 5 expressing glycoprotein gC or gD Time-dependent biodistribution and transgene expression of a recombinant human adenovirus serotype 5-luciferase vector as a surrogate for rAd5-FMDV vaccines in cattle Avian influenza vaccination in chickens and pigs with replicationcompetent adenovirus-free human recombinant adenovirus 5 Protection of mice and poultry from lethal H5N1 avian influenza virus through adenovirusbased immunization Canine adenovirus based rabies vaccines Vaccination of pigs with a recombinant porcine adenovirus expressing the gD gene from pseudorabies virus Construction and characterization of recombinant porcine adenovirus serotype 5 expressing the transmissible gastroenteritis virus spike gene Mucosal immunization of calves with recombinant bovine adenovirus-3: induction of protective immunity to bovine herpesvirus-1 Mucosal immunization of calves with recombinant bovine adenovirus-3 co-expressing truncated form of bovine herpesvirus-1 gD and bovine IL-6 Oral inoculation of chickens with a candidate fowl adenovirus 9 vector Phylogenetic relationships between adenoviruses as inferred from nucleotide sequences of inverted terminal repeats Identification of cis-acting sequences required for selective packaging of bovine adenovirus type 3 DNA Bovine adenovirus-3 E1A coding region contain cis-acting DNA packaging motifs E1A promoter of bovine adenovirus type 3 Transcription map and expression of bovine herpesvirus-1 glycoprotein D in early region 4 of bovine adenovirus-3 Characterization of early region 1 and pIX of bovine adenovirus-3 Transcription mapping and characterization of 284R and 121R proteins produced from early region 3 of bovine adenovirus type 3 Transcription units of E1a, E1b and pIX regions of bovine adenovirus type 3 Role of bovine adenovirus-3 33 K protein in viral replication Interaction of bovine adenovirus-3 33 K protein with other viral proteins Conserved regions of bovine adenovirus-3 pVIII contain functional domains involved in nuclear localization and packaging in mature infectious virion Functional characterization of 100 K protein of bovine adenovirus type 3. Saskatoon: University of Saskatchewan Mapping of nuclear import signal and importin␣3 binding regions of 52 K protein of bovine adenovirus-3 The fibre of bovine adenovirus type 3 is very long but bent Altered tropism of recombinant bovine adenovirus type-3 expressing chimeric fiber Development of a bovine adenovirus type 3-based expression vector Construction and characterization of E3-deleted bovine adenovirus type 3 expressing full-length and truncated form of bovine herpesvirus type 1 glycoprotein gD Generation of infectious genome of bovine adenovirus type 3 by homologous recombination in bacteria Efficient generation of recombinant adenovirus vectors by homologous recombination in Escherichia coli Novel human gene transfer vectors: evaluation of wild-type and recombinant animal adenoviruses in human-derived cells Efficient replication and generation of recombinant bovine adenovirus-3 in nonbovine cotton rat lung cells expressing I-SceI endonuclease Bovine adenovirus type 3 E1B (small) protein is essential for growth in bovine fibroblast cells Replication-defective bovine adenovirus type 3 as an expression vector Characterization of bovine adenovirus type 3 E1 proteins and isolation of E1-expressing cell lines Development and characterization of bovine X human hybid cell lines that efficiently support the replication of both wildtype bovine and human adenoviruses and those with E1 deleted Optimization of bovine coronavirus hemagglutinin-esterase glycoprotein expression in E3 deleted bovine adenovirus-3 Evaluation of promoters for foreign gene expression in E3 region of bovine adenovirus type 3 Recombinant bovine adenovirus type 3 expressing bovine viral diarrhea virus glycoprotein E2 induces an immune response in cotton rats Mutational analysis of early region 4 of bovine adenovirus type 3 Protection of cattle from BHV-1 infection by immunization with recombinant glycoprotein gIV Immunology of BVDV vaccines Monoclonal antibodies to bovine coronavirus glycoproteins E2 and E3: demonstration of in vivo virus-neutralizing activity Pathogenesis and immunogenicity of bovine adenovirus type 3 in cotton rats (Sigmodon hispidus) Sero-prevalences of selected cattle diseases in the Kafue flats of Zambia Neutralizing antibodies to bovine adenovirus serotype 3 in healthy cattle and cattle with respiratory tract disease The immunogenicity and efficacy of replication-defective and replicationcompetent bovine adenovirus-3 expressing bovine herpesvirus-1 glycoprotein gD in cattle Serological evaluation of relationship between viral pathogens (BHV-1, BVDV, BRSV, PI-3V, and Adeno3) and dairy calf pneumonia by indirect ELISA Experimental inoculation of heifers with bovine adenovirus type 3 Early protection against homologous challenge after a single dose of replication-defective human adenovirus type 5 expressing capsid proteins of foot-and-mouth disease virus (FMDV) strain A24 Replicationdefective adenovirus serotype 5 vectors elicit durable cellular and humoral immune responses in nonhuman primates Protection of mice and poultry from lethal H5N1 avian influenza virus through adenovirusbasede immunization Protective avian influenza in-ovo vaccination with non-replicating human adenovirus vector Recombinant bovine adenovirus -3 co-expressing bovine respiratory syncytial virus glycoprotein G and truncated glycoprotein gD of bovine herpesvirus-1 induce immune responses in cotton rats Interleukin-5 expressed by a recombinant virus vector enhances specific mucosal IgA responses in vivo The role of interleukin-6 in mucosal IgA antibody responses in vivo Recombinant adenovirus vectors expressing interleukin-5 and -6 specifically enhance mucosal immunoglobulin A responses in the lung Codelivery of the chemokine CCL3 by an adenovirus-based vaccine improves protection from retrovirus infection Improved vaccine protection against retrovirus infection after co-administration of adenoviral vectors encoding viral antigens and type I interferon subtypes Novel viral disease control strategy: adenovirus expressing alpha interferon rapidly protects swine from foot-and-mouth disease Adenovirus-mediated type I interferon expression delays and reduces disease signs in cattle challenged with foot-and-mouth disease virus The in vivo effects of recombinant bovine herpesvirus-1b expressing bovine interferon-gamma GM-CSF and IL-2 as adjuvant enhance the immune effect of protein vaccine against foot-and-mouth disease The effects of IL-6 and TNFalpha as molecular adjuvants on immune responses to FMDV and maturation of dendritic cells by DNA vaccination Cloning, sequence, and expression of bovine interferon-gamma Bovine interleukin 2: cloning, high level expression, and purification Nucleotide sequence of bovine interleukin-6 cDNA Bovine adenovirus type 3 containing heterologous protein in the C-terminus of minor capsid protein IX Enhancing oral vaccine potency by targeting intestinal M cells Exploiting M cells for drug and vaccine delivery In-vivo phage display to identify M cell-targeting ligands Comparative transduction efficiencies of human and nonhuman adenoviral vectors in human, murine, bovine, and porcine cells in culture Oncogenicity of bovine adenovirus-3 in hamsters Replicating and non-replicating viral vectors for vaccine development Oral delivery of replication-competent adenovirus vectors is well tolerated by SIV-and SHIV-infected rhesus macaques Safety and immunogenicity of an oral, replicating adenovirus serotype 4 vector vaccine for H5N1 influenza: a randomized, double blind, placebo-controlled, phase 1 study Experimental infections of humans with wild-type adenoviruses and with replication-competent adenovirus vectors: replication, safety and transmission Authors are thankful to all past and present members of Tikoo's Laboratory who have contributed to the work. The work was supported by grants from NSERC Canada, Saskatchewan Agriculture Development Fund, Ontario Cattlemen Association, Funding Consortium of Alberta, Cattle Industry Development Council, BC, Canada, and Saskatchewan Health Research Foundation, Canada. Published as VIDO-InterVac article no. 734.