Prime-boost vaccination with Bacillus Calmette Guerin and a recombinant adenovirus co-expressing CFP10, ESAT6, Ag85A and Ag85B of Mycobacterium tuberculosis induces robust antigen-specific immune responses in mice

  • Authors:
    • Wu Li
    • Min Li
    • Guangcun Deng
    • Liping Zhao
    • Xiaoming Liu
    • Yujiong Wang
  • View Affiliations

  • Published online on: May 12, 2015     https://doi.org/10.3892/mmr.2015.3770
  • Pages: 3073-3080
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tuberculosis (TB) remains to be a prevalent health issue worldwide. At present, Mycobacterium bovis Bacillus Calmette Guerin (BCG) is the singular anti‑TB vaccine available for the prevention of disease in humans; however, this vaccine only provides limited protection against Mycobacterium tuberculosis (Mtb) infection. Therefore, the development of alternative vaccines and strategies for increasing the efficacy of vaccination against TB are urgently required. The present study aimed to evaluate the ability of a recombinant adenoviral vector (Ad5‑CEAB) co‑expressing 10‑kDa culture filtrate protein, 6‑kDa early‑secreted antigenic target, antigen 85 (Ag85)A and Ag85B of Mtb to boost immune responses following primary vaccination with BCG in mice. The mice were first subcutaneously primed with BCG and boosted with two doses of Ad5-CEAB via an intranasal route. The immunological effects of Ad5-CEAB boosted mice primed with BCG were then evaluated using a series of immunological indexes. The results demonstrated that the prime‑boost strategy induced a potent antigen‑specific immune response, which was primarily characterized by an enhanced T cell response and increased production of cytokines, including interferon‑γ, tumor necrosis factor‑α and interleukin‑2, in mice. In addition, this vaccination strategy was demonstrated to have an elevated humoral response with increased concentrations of antigen‑specific bronchoalveolar lavage secretory immunoglobulin (Ig)A and serum IgG in mice compared with those primed with BCG alone. These data suggested that the regimen of subcutaneous BCG prime and mucosal Ad5‑CEAB boost was a novel strategy for inducing a broad range of antigen‑specific immune responses to Mtb antigens in vivo, which may provide a promising strategy for further development of adenoviral-based vaccine against Mtb infection.

Introduction

Tuberculosis (TB) is one of the most prevalent infectious diseases worldwide, accounting for ~1.4 million mortalities and 8.7 million novel cases annually, which occurs as a results of Mycobacterium tuberculosis (Mtb) infection (1). Due to Mtb reactivation at a latent state in immunocompromised individuals, slow progress in dealing with drug-resistant Mtb infection and the co-infection of Mtb with human immunodeficiency virus, the global burden of TB remains high, particularly in developing countries (2,3).

Effective vaccines are of key importance in ending the global TB epidemic (1). However, a consistently effective vaccine is not currently available. The only available TB vaccine, attenuated Mycobacterium bovis Bacillus Calmette Guerin (BCG) has made a marked contribution to Mtb infection control, especially in juvenile population and newborns (4). However, BCG does not provide effective protection for all age groups, particularly in adults; its protective efficacy is highly varied from different trials, with certain studies observing negative effects associated with BCG revaccination (0-80%) (5,6). Therefore, the development of more effective vaccines or feasible vaccination strategies that provide better protection from Mtb infection are urgently required.

It is widely accepted that homologous boosting with the same vaccine is not sufficient for protecting against Mtb (7); therefore, heterologous prime-boost immunization strategies using BCG and a novel anti-TB vaccine have been investigated. Such prime-boost vaccination strategies have demonstrated the potential to elicit protective immune responses, including cellular immune responses against Mtb infection in animal models and in humans (812).

Mtb is an intracellular pathogen transmitted via a mucosal route; mucosal and cellular immunity have thus been suggested to have pivotal roles in protection against Mtb infection. Therefore, a vector that is able to be delivered via a mucosal route and elicit potent antigen-specific immune responses may be an ideal candidate for anti-TB vaccines. Recombinant adenoviral vectors have gained increasing attention in anti-TB vaccine development due to their properties of type 1 immune adjuvant activity, excellent safety record in humans and high levels of antigen release as well as their suitability for parenteral and intranasal mucosal delivery (13,14). In addition, recombinant adenoviral vectors are highly effective at eliciting robust cellular immunity in experimental animal models (15), implicating them as promising antigen delivery vectors for the development of an anti-TB vaccine. In addition to a delivery vector, proper Mtb antigens used for vaccine development are also key factor for effectiveness of a vaccine candidate (16,17). Previously, a number of microbial antigens of Mtb were tested as TB vaccine candidates, including 10-kDa culture filtrate protein (CFP10), 6-kDa early-secreted antigenic target (ESAT6), the 30–32 kDa family of three proteins [antigen 85 (Ag85)A, Ag85B and Ag85C], the Mtb protein 64 (MPT64) and TB10.4 (a protein of 96 amino acids with a theoretical molecular mass of 10.4 kDa) (1823). Among them, CFP10 and ESAT6 are immunodominant antigens encoded by region of difference-1 (RD1) that are present in virulent strains of Mtb and Mycobacterium bovis; however, these antigens are absent in BCG (2426). Loss of RD1 was hypothesized to be the contributing factor for the attenuation of BCG (27,28); therefore, RD1-encoded CFP10 and ESAT6 have often been selected as potential antigen candidates in the development of novel anti-TB vaccines (19,2932). In addition to CFP10 and ESAT6, Ag85A and Ag85B have also been widely employed in anti-TB vaccine development (3236).

In the present study, BCG and a recombinant adenoviral vector (Ad5-CEAB) co-expressing CFP10, ESAT6, Ag85A and Ag85B of Mtb were used in combination to investigate the effects of this prime-boost strategy in mice.

Materials and methods

Animals

Female ICR mice (n=72, 6-8 weeks old) were purchased from the Animal facility of Ningxia Medical University (Yinchuan, China) and housed in a special pathogen-free environment with free access to food and water and a constant temperature of 18°C. All animal experiments were performed in accordance with the guidelines of the Chinese Council on Animal Care and were approved by the Committee for Animal Care and Use of Ningxia University.

Bacterial strains and Mtb antigens

The BCG vaccine, which was produced by Chengdu Institute of Biological Products (Chengdu, China), was a gift from the Centers for Disease Control and Prevention in Ningxia Province of China (Ningxia, China) while colony-forming units (CFU) were determined on 7H11 agar plates. For preparation of Mtb antigens of CFP10, ESAT6, Ag85 and Ag85B, the target gene fragments were amplified from Mtb H37Rv genomic DNA, which was extracted using Myco DNAout Kit (Beijing Tiandz Gene Technology Company, Beijing, China), by polymerase chain reaction (PCR), as previously described (18). PCR fragments were codon optimized prior to being subcloned in frame into a prokaryotic expression plasmid pET-28a (Novagen, Madison, WI, USA) for E. coli expression of His-tagged proteins (Novagen) (26). The His-tagged CFP10, ESAT6, Ag85A and Ag85B proteins were purified using ÄKTA protein purification system (GE Healthcare, Pittsburgh, PA, USA) according to the manufacturer’s instruction. Endotoxins were removed from the purified proteins using ToxinEraser™ Endotoxin Removal kit (GenScript, Piscataway, NJ, USA) prior to use. The antigenic proteins used in the present study had a purity of >85%, which was determined as previously described (18).

Recombinant adenovirus Ad5-CEAB preparation and immunization

The recombinant adenovirus Ad5-CEAB in which the four genes of CFP10, ESAT6, Ag85A and Ag85B were expressed as a mixture of proteins, rather than a fusion protein, was prepared as described previously, and the titer of virus stock was determined by a plaque assay (18). For immunization, mice were randomly divided into three groups (n=8 per group) as follows: Phosphate-buffered saline (PBS; Beijing Solarbio Science & Technology Co., Ltd., Beijing, China) group, mice were treated intranasally with 100 μl PBS three times separated by two-week intervals; BCG group, mice were injected subcutaneously with 1×106 CFU of BCG vaccine three times separated by 2 week intervals; BCG/Ad5 group, mice received a subcutaneous injection with 1×106 CFU of BCG and following a 2 week interval, mice were intranasally boosted with 100 μl 1×109 plaque-forming units (PFU) of Ad5-CEAB twice with 2 week intervals. At 2 weeks following the final immunization, animals were euthanized under ether anesthesia (Tianjin Kemiou Chemical Reagent Co., Ltd., Tianjin, China) by exsanguination for analysis of immune responses.

Flow cytometric analysis of splenocytes

Lymphocytes were isolated from the spleens of mice 2 weeks following the final immunization. Briefly, following sacrifice, spleens were aseptically harvested and the mouse spleen cells were obtained by carefully mashing the spleens with a syringe plunger, passing the product through a cell strainer (BD Biosciences, San Jose, CA, USA) and suspending it in preheated (37°C) RPMI-1640 medium (Gibco-BRL, Carlsbad, CA, USA). Splenocytes from each mouse were then isolated through density gradient centrifugation (1.092±0.001 g/ml; 500 × g, 20 min) with Mouse Lymphocyte Separation Medium (Solarbio Science & Technology Co., Ltd, Beijing, China). Splenocytes at a concentration of 5×106/ml were cultured in RPMI-1640 medium with 5% fetal calf serum (FCS) (Gibco-BRL, Eggenstein, Germany) supplemented with a mixture of purified Mtb proteins (containing 5 μg/ml of each purified Mtb: CFP10, ESAT6, Ag85A and Ag85B; Mtb CEAB antigen mixture) in an atmosphere of 5% CO2 at 37°C for 48 h. The frequencies of CD4+ and CD8+ T cells were characterized through flow cytometric analysis on a FACSCalibur instrument (BD Biosciences). Briefly, splenocytes from each animal were stained with a combination of allophycocyanin (APC) rat anti-mouse CD4 (1 μg/mouse; 553051), phycoerythrin (PE) rat anti-mouse CD8a (1 μg/mouse; 553033) and peridinin chlorophyll (PerCP) hamster anti-mouse CD3e (1 μg/mouse; 553067) antibodies (diluted 1:500; BD Biosciences) for 30 min at 4°C. The APC rat immunoglobulin (Ig)G2aκ, PE rat IgG2aκ and PerCP hamster IgG1κ isotype controls were included for isotype control staining (diluted 1:500; BD Biosciences).

Antigen-specific lymphocyte proliferation test

A total of 5×105 isolated splenocytes from individual mice were seeded into 96-well plates and stimulated in triplicate, with or without Mtb CEAB antigen mixture, in 5% FCS RPMI-1640 medium at 37°C in an atmosphere of 5% CO2 for 72 h. T cell proliferation was evaluated using the CellTiter 96® Aqueous One Solution Cell Proliferation Assay (Promega Corp., Madison, WI, USA), which is a sensitive fluorescence based microplate assay, according to the manufacturer’s protocol. The proliferative responses were expressed as stimulation index (SI) that was calculated using the following formula: SI = mean optical density (OD) value of antigen-stimulated cells/mean OD value of control cells.

Enzyme-linked immunospot (ELISPOT) assays for interferon (IFN)-γ

The frequency of splenic antigen-specific IFN-γ-secreting spot forming cells (SFC) was determined by ELISPOT using a Mouse IFN-γ ELISPOT Ready-SET-Go! Reagent set (eBioscience, San Diego, CA, USA) according to the manufacturer’s instructions with minor modifications. Briefly, isolated splenocytes were seeded at a density of 1×105cells/well in a 96-well filter plate (MSIPS4510; Millipore, Billerica, MA, USA) pre-coated with mouse IFN-γ antibodies overnight. Cells were then incubated with or without the stimulator (Mtb CEAB antigen mixture) for 40 h at 37°C. Visible spots were counted under a light microscope (SZX16; Olympus, Tokyo, Japan).

Cytokine induction and quantification

Splenocytes at a concentration of 5×105/well were seeded into 24-well plates and cultured in 5% FCS RPMI-1640 medium containing Mtb CEAB antigen mixture for 72 h at 37°C in an atmosphere of 5% CO2. Culture supernatants were then harvested by centrifugation at 500 x g for 10 min and the concentrations of IFN-γ, tumor necrosis factor (TNF)-α and interleukin (IL)-2 were determined using an enzyme-linked immunosorbent assay (ELISA) cytokine detection system (RayBiotech, Inc., Norcross, GA, USA) according to the manufacture’s instructions. All experiments were performed in triplicate.

ELISA assay for antigen-specific secretory (s)IgA and IgG

For sIgA measurement, bronchoalveolar lavage (BAL) samples were collected according to a method previously described (37). SIgA in the supernatant of BAL fluid was determined using an ELISA kit (eBioscience) according to the manufacturer’s protocol. ELISA plates were pre-coated with Mtb CEAB antigen mixture (5 mg/ml) at 4°C overnight.

For IgG measurement, mouse peripheral blood (~600 ml) was collected 2 weeks following the final immunization and the concentration of serum antigen-specific IgG was ascertained using a mouse ELISA Ready-SET-Go! kit (eBio-science) according to the manufacturer’s instructions with minor modifications. ELISA plates were customized by pre-coating with Mtb CEAB antigen mixture at 4°C overnight, rather than directly coated with the capturing antibodies provided in the kits.

Statistical analysis

Experimental data were expressed as the mean ± standard deviation. Differences between groups were analyzed using a one-way analysis of variance followed by Tukey’s post-hoc test with SPSS 13.0 software (SPSS, Inc., Chicago, IL, USA).

Results

Antigen-specific T cell responses

It is widely accepted that T cell responses have important roles in the host defense against Mtb infection (16). In the present study, the Mtb antigen-specific T cell response was analyzed in vitro by assessing the ability of the prime-boosted strategy to induce T cell responses. The results of Mtb CEAB antigen-specific splenic T cell responses revealed a significantly elevated splenic T cell proliferation in immunized mice in the BCG and BCG/Ad5 groups compared with the PBS-treated group (Fig. 1A). In addition, the IFN-γ ELISPOT assay revealed an increased frequency of Mtb antigen-specific IFN-γ-secreting splenic T cells in the mice immunized with BCG (P<0.05) and BCG/Ad5 (P<0.001) compared with the PBS-treated group (Fig. 1B). Furthermore, higher frequencies of CD4+ (Fig. 2A) and CD8+ (Fig. 2B) T cell populations were observed in mice immunized with BCG (P<0.001) and BCG/Ad5 (P<0.001) compared with the PBS-treated group. Of note, all of the above examined indexes of immune response in mice immunized with BCG/Ad5 were significantly increased compared with those of the BCG group. These results clearly demonstrated that the subcutaneous BCG prime mucosal Ad5-CEAB-boosted strategy was capable of stimulating a more potent antigen-specific T cell response in mice compared with that of the BCG group.

Antigen-specific cytokines responses

Cytokines were previously suggested to have important roles in the host defense against Mtb (16). In the present study, concentrations of cyto-kines INF-γ, TNF-α and IL-2 were detected using ELISA analysis of the culture supernatant of lymphocytes stimulated with Mtb antigens in vitro (Fig. 3). The results showed that the levels of all the tested cytokines were significantly higher in the BCG prime-Ad5-CEAB-boosted group compared with the PBS-treated group. Of note, significantly elevated levels of antigen-induced cytokines INF-γ (P<0.001) (Fig. 3A), TNF-α (P<0.05) (Fig. 3B) and IL-2 (P<0.001) (Fig. 3C) were reported in the BCG prime-Ad5-CEAB-boosted group compared with the BCG group. This therefore indicated that the BCG prime-Ad5-CEAB boost strategy had a greater potency to enhance antigen-specific immunity in mice compared with BCG alone.

Antigen-specific antibody responses

Mucosal immunity is known to have important roles against Mtb infection and sIgA is the most abundant antibody isotype produced in mucosal tissues (38); therefore, sIgA production was examined in BAL fluid of mice. As shown in Fig. 4A, sIgA levels in BAL were markedly elevated in mice immunized with the BCG/Ad5 compared with the BCG group (P<0.01). However, no statistically significant difference was observed between the BCG group and PBS-treated group (P>0.05) (Fig. 4A). These data suggested that the prime-boost strategy was able to potently augment mucosal immune responses in vivo.

Humoral immunity has also been demonstrated to have a protective role in mycobacterial infections (39). In order to evaluate the IgG antibody response in immunized mice in the BCG or BCG prime Ad5-CEAB-boosted groups, the titers of IgG in mice were examined at 2 weeks following the final immunization. As shown in Fig. 4B, mice immunized with BCG or BCG/Ad5 elicited significantly higher titer of antigen-specific IgG compared with the group treated with PBS. Furthermore, IgG levels in mice immunized with BCG/Ad5 were significantly higher compared with those of the BCG group (P<0.001), which indicated that the prime-boosted strategy may elicit more efficient antibody responses.

Discussion

It has been reported that repeated vaccination with the same vaccine induces increased levels of antibody production compared with a single vaccination. However, such homologous boosts with the same vaccine may not be sufficient for protection against intracellular pathogens, such as Mtb (7). Studies in humans have demonstrated that revaccination with BCG does not confer additional protection against TB (40,41) and certain studies in humans and animals even reported negative effects associated with BCG revaccination (40-43). However, heterologous prime-boost strategies using BCG and a novel anti-TB vaccine may elicit robust immune responses, which are more efficacious than BCG alone. Since BCG is the only commonly used anti-TB vaccine in most developing countries, employment of a second vaccine to boost BCG-primed immunity may be the most practical novel strategy. In accordance with the heterologous prime-boost strategy, the present study investigated the safety and effi-cacy of a novel recombinant vaccine candidate, Ad5-CEAB, using the BCG-prime-boost strategy in mice. To the best of our knowledge, the recombinant adenovirus Ad5-CEAB used as booster in the present study was the first attempt for the co-expression of four Mtb antigens as a mixture of individual proteins. The results demonstrated that the adenovirus vector may be a promising novel vaccine platform capable of boosting BCG-induced immunity.

Vaccines against intracellular infections are dependent on the induction of cell-mediated immunity (44). As an intracellular pathogen, Mtb localizes to the vacuole of the host’s macrophages and cellular immunity has a crucial role in the immune response against Mtb infection. The cellular immune response is primarily composed of CD4+, CD8+ and other subsets of T cells. CD4+ T cells were reported to contribute to the initial resistance to Mtb via the production of IFN-γ and other cytokines in order to induce macrophage activation (45). However, CD8+ T cells produce IFN-γ and cytokines in addition to producing perforin and granulysin, which act to directly kill Mtb-infected cells and attack the invaded Mtb bacilli (46). In the present study, significantly increased frequencies of antigen-specific CD4+, CD8+ and INF-γ-secreting T cells were detected in the splenocytes of mice boosted with Ad5-CEAB compared with those primed with BCG alone. In addition to its ability to induce antigen-specific T cell responses, the prime-boost strategy also displayed a capacity to augment antigen-specific T helper type-1 cytokine production, including the secretion of INF-γ, TNF-α and IL-2. Cytokines have also been demonstrated to have important roles in host defense against Mtb infection. For example, IFN-γ was reported to activate infected macrophages and directly inhibit intracellular replication and growth of Mtb (47,48). By contrast, TNF-α was demonstrated to be essential for the initiation of the immune response against Mtb infection (49).

In addition to T cell responses, the prime-boost strategy exhibited a capacity to evoke antibody responses in the present study. Antibody responses have a protective role in preventing mycobacterial infections, particularly the mucosal antibodies. For example, sIgA, the most abundant naturally-produced antibody isotype in mucosal tissue, has an indispensable role in preventing primary Mtb infection at the mucosal surfaces; in addition, sIgA was reported to prevent the adsorption of pathogens at the mucosal epithelium (50,51). A murine study demonstrated that sIgA may act to prevent the entrance of mycobacterial bacilli into the lungs (52). The results of the present study revealed that the BCG prime and mucosal Ad5-CEAB boost strategy was able to significantly augment mucosal immune responses in vivo.

Antigen-specific IgG antibodies are commonly used as biomarkers to confirm the expression of Mtb antigens in animal models; however, the role of serum antibodies in the pathogenesis and control of TB has been controversial for a long time. Previous studies have demonstrated that serum antibodies may have protective effect in animal models of tuberculosis (39,53). In addition, analysis of the isotypic distribution of immuno-globulin may offer an insight into the possible immunological mechanisms involved in cellular immunity (54). Together with the observation of increased CD4+ and CD8+ T cell populations in BCG/Ad5-CEAB-immunized mice, the results of the present study clearly demonstrated that the BCG prime mucosal Ad5-CEAB boost vaccination strategy effectively evoked the immune system for T cell- and antibody-mediated antigen-specific immune responses in mice.

The mucosal immune response is the first line of defense against infectious agents and is crucial for the immune response against Mtb infection. Increasing evidence has indicated the effectiveness of vaccination at the mucosal site compared with vaccination via other routes for inducing protection from mucosal infectious diseases (15,55). Numerous studies have verified that mucosal immunity may provide unique advantages for protection against Mtb infection (34,5658). Therefore, any vaccines or vaccination strategies that are able to elicit the mucosal immune response may enhance the efficacy of protection against Mtb infection. Great efforts have been made to improve the protective efficacy of TB vaccines and various types of vaccine candidates or vectors have been developed, including the recombinant BCG (rBCG), DNA vaccines, nanoparticle vaccines, recombinant modified vaccinia virus Ankara and recombinant adenoviral-based vaccines (12,15,32,59,60). Among them, the adenoviral-based TB vaccines have gained increased attention, as they were first evaluated as mucosal TB vaccine candidates (15). An increasing number of studies have thus focused on mucosal immunity; these studies have suggested that intranasal/intrapulmonary vaccination with recombinant adenoviral vaccines may induce an antigen-specific mucosal immune response (15,6166). In line with these findings, the results of the present study demonstrated that intranasal boost with the recombinant adenovirus Ad5-CEAB was able to enhance the BCG-primed immune response. In addition, the present study reported that levels of antigen-specific sIgA in BAL fluid were significantly increased in the Ad5-CEAB-boosted group, indicating that intranasal boost with Ad5-CEAB may induce a potent antigen-specific mucosal immune response in mice.

In conclusion, the results of the present study demonstrated that the heterologous prime-boost strategy of subcutaneously primed BCG-intranasal boost with recombinant adenovirus Ad5-CEAB was able to elicit an enhanced antigen-specific immune response in mice compared with that conferred by homologous prime-boost immunization with BCG. These results provided evidence for the effectiveness of TB vaccines from recombinant adenoviral vectors and novel anti-TB vaccination strategies. The BCG prime Ad5-CEAB boost vaccination strategy appears promising as an anti-TB vaccination strategy, thus we aim to evaluate this in mice infected with Mtb in future studies.

Acknowledgments

The present study was supported by grants from the National Natural Science Foundation of China (no. 31160515), the National Key Basic Research Program of China (973 Program) (nos. 2012CB126301 and 2012CB518801) and the Key Technologies Research and Development Program of China (no. 2012BAD12B07-4).

References

1 

World Health Organization (WHO): Global Tuberculosis Report 2012. WHO; Geneva, Switzerland: pp. 3062012

2 

Churchyard GJ, Chaisson RE, Maartens G and Getahun H: Tuberculosis preventive therapy: An underutilised strategy to reduce individual risk of TB and contribute to TB control. S Afr Med J. 104:339–343. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Muwonge A, Malama S, Johansen TB, et al: Molecular epidemiology, drug susceptibility and economic aspects of tuberculosis in Mubende district, Uganda. PLoS One. 8:e647452013. View Article : Google Scholar : PubMed/NCBI

4 

Awasthi S and Moin S: Effectiveness of BCG vaccination against tuberculous meningitis. Indian Pediatr. 36:455–460. 1999.

5 

Nuttall JJ, Davies MA, Hussey GD and Eley BS: Bacillus Calmette-Guérin (BCG) vaccine-induced complications in children treated with highly active antiretroviral therapy. Int J Infect Dis. 12:e99–e105. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Bolger T, O’Connell M, Menon A and Butler K: Complications associated with the bacille Calmette-Guérin vaccination in Ireland. Arch Dis Child. 91:594–597. 2006. View Article : Google Scholar : PubMed/NCBI

7 

McShane H and Hill A: Prime-boost immunisation strategies for tuberculosis. Microbes Infect. 7:962–967. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Dean G, Whelan A, Clifford D, et al: Comparison of the immunogenicity and protection against bovine tuberculosis following immunization by BCG-priming and boosting with adenovirus or protein based vaccines. Vaccine. 32:1304–1310. 2014. View Article : Google Scholar

9 

Hoft DF, Blazevic A, Stanley J, et al: A recombinant adenovirus expressing immunodominant TB antigens can significantly enhance BCG-induced human immunity. Vaccine. 30:2098–2108. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Perez de Val B, Villarreal-Ramos B, Nofrarías M, et al: Goats primed with Mycobacterium bovis BCG and boosted with a recombinant adenovirus expressing Ag85A show enhanced protection against tuberculosis. Clin Vaccine Immunol. 19:1339–1347. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Dou J, Wang Y, Yu F, et al: Protection against Mycobacterium tuberculosis challenge in mice by DNA vaccine Ag85A-ESAT-6-IL-21 priming and BCG boosting. Int J Immunogenet. 39:183–190. 2012. View Article : Google Scholar

12 

Cervantes-Villagrana AR, Hernández-Pando R, Biragyn A, et al: Prime-boost BCG vaccination with DNA vaccines based in β-defensin-2 and mycobacterial antigens ESAT6 or Ag85B improve protection in a tuberculosis experimental model. Vaccine. 31:676–684. 2013. View Article : Google Scholar

13 

Xing Z and Lichty BD: Use of recombinant virus-vectored tuberculosis vaccines for respiratory mucosal immunization. Tuberculosis. 86:211–217. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Lasaro MO and Ertl HC: New insights on adenovirus as vaccine vectors. Mol Ther. 17:1333–1339. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Wang J, Thorson L, Stokes RW, et al: Single mucosal, but not parenteral, immunization with recombinant adenoviral-based vaccine provides potent protection from pulmonary tuberculosis. J Immunol. 173:6357–6365. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Li W, Deng G, Li M, Liu X and Wang Y: Roles of mucosal immunity against Mycobacterium tuberculosis infection. Tuberc Res Treat. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Cripps AW, Kyd JM and Foxwell AR: Vaccines and mucosal immunisation. Vaccine. 19:2513–2515. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Li W, Deng G, Li M, et al: A recombinant adenovirus expressing CFP10, ESAT6, Ag85A and Ag85B of Mycobacterium tuberculosis elicits strong antigen-specific immune responses in mice. Mol Immunol. 62:86–95. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Zhang H, Peng P, Miao S, et al: Recombinant Mycobacterium smegmatis expressing an ESAT6-CFP10 fusion protein induces anti-mycobacterial immune responses and protects against Mycobacterium tuberculosis challenge in mice. Scand J Immunol. 72:349–357. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Liang Y, Wu X, Zhang J, et al: Immunogenicity and therapeutic effects of Ag85A/B chimeric DNA vaccine in mice infected with Mycobacterium tuberculosis. FEMS Immunol Med Microbiol. 66:419–426. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Pydi SS, Bandaru AR, Venkatasubramanian S, et al: Vaccine for tuberculosis: up-regulation of IL-15 by Ag85A and not by ESAT-6. Tuberculosis (Edinb). 91:136–139. 2011. View Article : Google Scholar

22 

Sibley L, Reljic R, Radford DS, et al: Recombinant Bacillus subtilis spores expressing MPT64 evaluated as a vaccine against tuberculosis in the murine model. FEMS Microbiol Lett. 358:170–179. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Shi S, Yu L, Sun D, Liu J and Hickey AJ: Rational design of multiple TB antigens TB10.4 and TB10.4-Ag85B as subunit vaccine candidates against Mycobacterium tuberculosis. Pharm Res. 27:224–234. 2010. View Article : Google Scholar

24 

Berthet FX, Rasmussen PB, Rosenkrands I, Andersen P and Gicquel B: A Mycobacterium tuberculosis operon encoding ESAT-6 and a novel low-molecular-mass culture filtrate protein (CFP-10). Microbiology. 144(Pt 11): 3195–3203. 1998. View Article : Google Scholar : PubMed/NCBI

25 

Gordon SV, Brosch R, Billault A, Garnier T, Eiglmeier K and Cole ST: Identification of variable regions in the genomes of tubercle bacilli using bacterial artificial chromosome arrays. Mol Microbiol. 32:643–655. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Behr MA, Wilson MA, Gill WP, et al: Comparative genomics of BCG vaccines by whole-genome DNA microarray. Science. 284:1520–1523. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Lewis KN, Liao R, Guinn KM, et al: Deletion of RD1 from Mycobacterium tuberculosis mimics Bacille Calmette-Guerin attenuation. J Infect Dis. 187:117–123. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Pym AS, Brodin P, Brosch R, Huerre M and Cole ST: Loss of RD1 contributed to the attenuation of the live tuberculosis vaccines Mycobacterium bovis BCG and Mycobacterium microti. Mol Microbiol. 46:709–717. 2002. View Article : Google Scholar : PubMed/NCBI

29 

You Q, Wu Y, Jiang D, et al: Immune responses induced by heterologous boosting of recombinant bacillus Calmette-Guerin with Ag85B-ESAT6 fusion protein in levamisole-based adjuvant. Immunol Invest. 41:412–428. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Yuan W, Dong N, Zhang L, et al: Immunogenicity and protective efficacy of a tuberculosis DNA vaccine expressing a fusion protein of Ag85B-Esat6-HspX in mice. Vaccine. 30:2490–2497. 2012. View Article : Google Scholar

31 

Esparza-González SC, Troy A, Troudt J, et al: Recombinant adenovirus delivery of calreticulin-ESAT-6 produces an antigen-specific immune response but no protection against a Mycobacterium tuberculosis challenge. Scand J Immunol. 75:259–265. 2012. View Article : Google Scholar

32 

Lin CW, Su IJ, Chang JR, Chen YY, Lu JJ and Dou HY: Recombinant BCG coexpressing Ag85B, CFP10 and interleukin-12 induces multifunctional Th1 and memory T cells in mice. APMIS. 120:72–82. 2012. View Article : Google Scholar

33 

Betts G, Poyntz H, Stylianou E, et al: Optimising immunoge-nicity with viral vectors: mixing MVA and HAdV-5 expressing the mycobacterial antigen Ag85A in a single injection. PLoS One. 7:e504472012. View Article : Google Scholar

34 

Dietrich J, Andersen C, Rappuoli R, Doherty TM, Jensen CG and Andersen P: Mucosal administration of Ag85B-ESAT-6 protects against infection with Mycobacterium tuberculosis and boosts prior bacillus Calmette-Guerin immunity. J Immunol. 177:6353–6360. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Dou J, Tang Q, Yu F, et al: Investigation of immunogenic effect of the BCG priming and Ag85A-GM-CSF boosting in Balb/c mice model. Immunobiology. 215:133–142. 2010. View Article : Google Scholar

36 

Lu D, Garcia-Contreras L, Muttil P, et al: Pulmonary immunization using antigen 85-B polymeric microparticles to boost tuberculosis immunity. AAPS J. 12:338–347. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Wakeham J, Wang J, Magram J, et al: Lack of both types 1 and 2 cytokines, tissue inflammatory responses and immune protection during pulmonary infection by Mycobacterium bovis bacille Calmette-Guerin in IL-12-deficient mice. J Immunol. 160:6101–6111. 1998.PubMed/NCBI

38 

Williams A, Reljic R, Naylor I, et al: Passive protection with immunoglobulin A antibodies against tuberculous early infection of the lungs. Immunology. 111:328–333. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Borrero R, García Mde L, Canet L, et al: Evaluation of the humoral immune response and cross reactivity against Mycobacterium tuberculosis of mice immunized with liposomes containing glycolipids of Mycobacterium smegmatis. BMC Immunol. 14(Suppl 1): 132013. View Article : Google Scholar

40 

Rodrigues LC, Pereira SM, Cunha SS, et al: Effect of BCG revaccination on incidence of tuberculosis in school-aged children in Brazil: the BCG-REVAC cluster-randomised trial. Lancet. 366:1290–1295. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Dantas OM, Ximenes RA, de Albuquerque Mde F, et al: A case-control study of protection against tuberculosis by BCG revaccination in Recife, Brazil. Int J Tuberc Lung Dis. 10:536–541. 2006.PubMed/NCBI

42 

Basaraba RJ, Izzo AA, Brandt L and Orme IM: Decreased survival of guinea pigs infected with Mycobacterium tuberculosis after multiple BCG vaccinations. Vaccine. 24:280–286. 2006. View Article : Google Scholar

43 

Buddle B, Wedlock D, Parlane N, et al: Revaccination of neonatal calves with Mycobacterium bovis BCG reduces the level of protection against bovine tuberculosis induced by a single vaccination. Infect Immun. 71:6411–6419. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Seder RA and Hill AV: Vaccines against intracellular infections requiring cellular immunity. Nature. 406:793–798. 2000. View Article : Google Scholar : PubMed/NCBI

45 

Flynn JL and Chan J: Immunology of tuberculosis. Annu Rev Immunol. 19:93–129. 2001. View Article : Google Scholar : PubMed/NCBI

46 

Woodworth JS, Wu Y and Behar SM: Mycobacterium tuberculosis-specific CD8+T cells require perforin to kill target cells and provide protection in vivo. J Immunol. 181:8595–8603. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Szabo SJ, Sullivan BM, Stemmann C, Satoskar AR, Sleckman BP and Glimcher LH: Distinct effects of T-bet in TH1 lineage commitment and IFN-gamma production in CD4 and CD8 T cells. Science. 295:338–342. 2002. View Article : Google Scholar : PubMed/NCBI

48 

Sharma M, Sharma S, Roy S, Varma S and Bose M: Pulmonary epithelial cells are a source of interferon-γ in response to Mycobacterium tuberculosis infection. Immunol Cell Biol. 85:229–237. 2007.PubMed/NCBI

49 

Bean AG, Roach DR, Briscoe H, et al: Structural deficiencies in granuloma formation in TNF gene-targeted mice underlie the heightened susceptibility to aerosol Mycobacterium tuber- culosis infection, which is not compensated for by lymphotoxin. J Immunol. 162:3504–3511. 1999.PubMed/NCBI

50 

Mazanec MB, Nedrud JG, Kaetzel CS and Lamm ME: A three-tiered view of the role of IgA in mucosal defense. Immunol Today. 14:430–435. 1993. View Article : Google Scholar : PubMed/NCBI

51 

Williams R and Gibbons R: Inhibition of bacterial adherence by secretory immunoglobulin A: a mechanism of antigen disposal. Science. 177:697–699. 1972. View Article : Google Scholar : PubMed/NCBI

52 

Tjärnlund A, Rodríguez A, Cardona PJ, et al: Polymeric IgR knockout mice are more susceptible to mycobacterial infections in the respiratory tract than wild-type mice. Int Immunol. 18:807–816. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Olivares N, Marquina B, Mata-Espinoza D, et al: The protective effect of immunoglobulin in murine tuberculosis is dependent on IgG glycosylation. Pathog Dis. 69:176–183. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Mosmann T and Coffman R: TH1 and TH2 cells: Different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol. 7:145–173. 1989. View Article : Google Scholar : PubMed/NCBI

55 

Lamichhane A, Azegamia T and Kiyonoa H: The mucosal immune system for vaccine development. Vaccine. 32:6711–6723. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Goonetilleke NP, McShane H, Hannan CM, Anderson RJ, Brookes RH and Hill AV: Enhanced immunogenicity and protective efficacy against Mycobacterium tuberculosis of bacille Calmette-Guérin vaccine using mucosal administration and boosting with a recombinant modified vaccinia virus Ankara. J Immunol. 171:1602–1609. 2003. View Article : Google Scholar : PubMed/NCBI

57 

Chen L, Wang J, Zganiacz A and Xing Z: Single intranasal mucosal Mycobacterium bovis BCG vaccination confers improved protection compared to subcutaneous vaccination against pulmonary tuberculosis. Infect Immun. 72:238–246. 2004. View Article : Google Scholar :

58 

Santosuosso M, McCormick S, Zhang X, Zganiacz A and Xing Z: Intranasal boosting with an adenovirus-vectored vaccine markedly enhances protection by parenteral Mycobacterium bovis BCG immunization against pulmonary tuberculosis. Infect Immun. 74:4634–4643. 2006. View Article : Google Scholar : PubMed/NCBI

59 

Yu F, Wang J, Dou J, et al: Nanoparticle-based adjuvant for enhanced protective efficacy of DNA vaccine Ag85A-ESAT-6-IL-21 against Mycobacterium tuberculosis infection. Nanomedicine. 8:1337–1344. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Ndiaye BP, Thienemann F, Ota M, et al: Safety, immunogenicity, and efficacy of the candidate tuberculosis vaccine MVA85A in healthy adults infected with HIV-1: A randomised, placebo-controlled, phase 2 trial. Lancet Respir Med. 3:190–200. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Croyle MA, Patel A, Tran KN, et al: Nasal delivery of an adenovirus-based vaccine bypasses pre-existing immunity to the vaccine carrier and improves the immune response in mice. PLoS One. 3:e35482008. View Article : Google Scholar : PubMed/NCBI

62 

Lemiale F, Kong WP, Akyurek LM, et al: Enhanced mucosal immunoglobulin A response of intranasal adenoviral vector human immunodeficiency virus vaccine and localization in the central nervous system. J Virol. 77:10078–10087. 2003. View Article : Google Scholar : PubMed/NCBI

63 

Richardson JS, Abou MC, Tran KN, Kumar A, Sahai BM and Kobinger GP: Impact of systemic or mucosal immunity to adenovirus on Ad-based Ebola virus vaccine efficacy in guinea pigs. J Infect Dis. 204(Suppl 3): 1032–1042. 2011. View Article : Google Scholar

64 

Santosuosso M, Zhang X, McCormick S, Wang J, Hitt M and Xing Z: Mechanisms of mucosal and parenteral tuberculosis vaccinations: adenoviral-based mucosal immunization preferentially elicits sustained accumulation of immune protective CD4 and CD8 T cells within the airway lumen. J Immunol. 174:7986–7994. 2005. View Article : Google Scholar : PubMed/NCBI

65 

Shim BS, Stadler K, Nguyen HH, et al: Sublingual immunization with recombinant adenovirus encoding SARS-CoV spike protein induces systemic and mucosal immunity without redirection of the virus to the brain. Virol J. 9:2152012. View Article : Google Scholar : PubMed/NCBI

66 

Kaufman DR, Bivas-Benita M, Simmons NL, Miller D and Barouch DH: Route of adenovirus-based HIV-1 vaccine delivery impacts the phenotype and trafficking of vaccine-elicited CD8+ T lymphocytes. J Virol. 84:5986–5996. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2015
Volume 12 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li W, Li M, Deng G, Zhao L, Liu X and Wang Y: Prime-boost vaccination with Bacillus Calmette Guerin and a recombinant adenovirus co-expressing CFP10, ESAT6, Ag85A and Ag85B of Mycobacterium tuberculosis induces robust antigen-specific immune responses in mice. Mol Med Rep 12: 3073-3080, 2015
APA
Li, W., Li, M., Deng, G., Zhao, L., Liu, X., & Wang, Y. (2015). Prime-boost vaccination with Bacillus Calmette Guerin and a recombinant adenovirus co-expressing CFP10, ESAT6, Ag85A and Ag85B of Mycobacterium tuberculosis induces robust antigen-specific immune responses in mice. Molecular Medicine Reports, 12, 3073-3080. https://doi.org/10.3892/mmr.2015.3770
MLA
Li, W., Li, M., Deng, G., Zhao, L., Liu, X., Wang, Y."Prime-boost vaccination with Bacillus Calmette Guerin and a recombinant adenovirus co-expressing CFP10, ESAT6, Ag85A and Ag85B of Mycobacterium tuberculosis induces robust antigen-specific immune responses in mice". Molecular Medicine Reports 12.2 (2015): 3073-3080.
Chicago
Li, W., Li, M., Deng, G., Zhao, L., Liu, X., Wang, Y."Prime-boost vaccination with Bacillus Calmette Guerin and a recombinant adenovirus co-expressing CFP10, ESAT6, Ag85A and Ag85B of Mycobacterium tuberculosis induces robust antigen-specific immune responses in mice". Molecular Medicine Reports 12, no. 2 (2015): 3073-3080. https://doi.org/10.3892/mmr.2015.3770