Next Article in Journal
Dual Role of p53 in Innate Antiviral Immunity
Next Article in Special Issue
Heterologous Prime-Boost HIV-1 Vaccination Regimens in Pre-Clinical and Clinical Trials
Previous Article in Journal
Evasion of the Interferon-Mediated Antiviral Response by Filoviruses
Previous Article in Special Issue
Preliminary Report on HIV-1 Vaccine Preparedness in Nigeria: Advantages of Recruiting University Students
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Development of an AIDS Mucosal Vaccine

1
AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
2
Department of Microbiology and Research Center of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
*
Author to whom correspondence should be addressed.
Viruses 2010, 2(1), 283-297; https://doi.org/10.3390/v2010283
Submission received: 12 October 2009 / Revised: 20 January 2010 / Accepted: 20 January 2010 / Published: 22 January 2010
(This article belongs to the Special Issue AIDS Vaccine)

Abstract

:
It is well known that mucosal tissues contain the largest surface area of the human body and are the front line of natural host defense against various pathogens. In fact, more than 80% of infectious disease pathogens probably gain entry into the susceptible human hosts through open mucosal surfaces. Human immunodeficiency virus type one (HIV-1), a mainly sexually transmitted virus, also primarily targets the vaginal and gastrointestinal mucosa as entry sites for viral transmission, seeding, replication and amplification. Since HIV-1 establishes its early replication in vaginal or rectal mucosal tissues, the induction of sufficient mucosal immunity at the initial site of HIV-1 transmission becomes essential for a protective vaccine. However, despite the fact that current conventional vaccine strategies have remained unsuccessful in preventing HIV-1 infection, sufficient financial support and resources have yet to be given to develop a vaccine able to elicit protective mucosal immunity against sexual transmissions. Interestingly, Chinese ancestors invented variolation through intranasal administration about one thousand years ago, which led to the discovery of a successful smallpox vaccine and the final eradication of the disease. It is the hope for all mankind that the development of a mucosal AIDS vaccine will ultimately help control the AIDS pandemic. In order to discover an effective mucosal AIDS vaccine, it is necessary to have a deep understanding of mucosal immunology and to test various mucosal vaccination strategies.

1. Introduction

Mucosal membranes contain the largest surface area of the human body, providing protection for various body cavities exposed to the external environment and internal organs. Mucosal membranes comprise the linings of gastrointestinal, urogenital and respiratory tracts [1]. They are actually the portal of entry for more than 80% of infectious disease pathogens, and the first line of defense for host protection from infection through various mechanisms including physical barriers, mechanical discharge, chemical degradation, innate immunity, and specialized mucosal immune responses [2,3].
Mucosal tissues represent a critical immunological repertoire, which is compartmentalized and independent from systemic immune organs [4]. Mucosal tissues in a healthy human body comprise as much as 80% of all lymphocytes, which may transfer between various mucosal-associated lymphoid tissues (MALTs) [4]. B-lymphocytes in MALTs mainly produce secretory IgA or sIgA, which is released as a covalently linked dimer. This unique structure makes sIgA more stable and resistant to many kinds of digestive enzymes. sIgA is considered to be the major protective immunoglobulin on mucosal surfaces [4]. T lymphocytes on the other hand either act as cytotoxic T lymphocytes (CTL) (e.g., CD8+ T cells) or help the induction of both cellular mediated immunity and antibody response (e.g., CD4+ T cells) [4].
Upon encountering foreign antigens, the activated B and T cells may migrate from inductive sites (e.g., Peyer’s patches in the gut, and nasopharynx-associated lymphoid tissue (NALT) in the oropharyngeal cavity), travel through lymph nodes and the peripheral circulation and back home to effector sites (e.g., such as the lamina propria of the intestinal and respiratory tracts, and glandular tissues). This results in the generation of antigen-specific T helper 2 (Th2) cell-dependent IgA responses, and T helper 1 (Th1) cell- and CTL-dependent immune responses, which function as the front line of defense at mucosal surfaces. The homing behavior of these cells is mainly attributed to the up-regulated expression of site-specific adhesion molecules and cytokine receptors that guide the cells back to mucosa by recognition of mucosal tissue-specific receptors on vascular endothelial cells [5,6]. Critically, mucosal inductive sites stand as sentinels to the intestinal and respiratory systems and represent the major sites where mucosal immune responses are initiated.
Human immunodeficiency virus type one (HIV-1), the causative agent of acquired immunodeficiency syndrome (AIDS), is a mainly sexually transmitted virus. HIV-1 primarily targets the gastrointestinal and vaginal mucosa as entry sites for viral transmission, seeding, replication and amplification. Since HIV-1 establishes its early replication in vaginal or rectal mucosal tissues, the induction of sufficient mucosal immunity at the initial site of HIV-1 transmission becomes essential for a protective vaccine. Here, we provide an overview of the development of an AIDS mucosal vaccine and perspectives of mucosal vaccine candidates against HIV-1.

2. A Brief History of Mucosal Vaccines

A vaccine is a biological preparation that induces immunity against a particular disease. The term vaccine derives from Edward Jenner's use of the term cowpox (Latin variolæ vaccinæ, adapted from the Latin vaccīn-us, from vacca cow) in 1796. Edward Jenner demonstrated that cowpox, when administered to humans, provided them protection against smallpox. The history of using a biological preparation to prevent smallpox, however, was traced back to Northern Song Dynasty (960–1127 A.D.) when Chinese Ancestors collected scabs from skin lesions of smallpox patients and used the ground down particles to prevent new infections, a procedure called variolation [7]. Interestingly, the procedure involved the delivery of the biological preparation through intranasal inoculation, a common route of modern mucosal vaccination. This technique was subsequently introduced to European countries by Lady Mary Montagu prior to 1762, which later led to the discovery of vaccines and modern immunology [8].
During recent decades, with the rapid development of modern immunology, significant progress has been made to understand the structure and function of the mucosal immune system. It is now clear that mucosal vaccines educate immune cells in mucosal inductive sites and can thus induce protective immunity in both the mucosal and systemic compartments. One of the classical mucosal vaccines is the oral polio vaccine (OPV), a live-attenuated strain with spontaneous mutations in the viral genome. OPV was generated by a series of passages of a polio virus through non-human cells at sub-physiological temperature [9]. In comparison to the inactive polio vaccine (IPV) given by injection, OPV not only elicits neutralizing antibodies to prevent the spread of poliovirus to the nervous system, but also induces excellent sIgA response in the intestinal mucosa, the primary site of poliovirus entry and replication, which contributes greatly to the worldwide eradication of polio. However, because of concerns over the reversion of the live-attenuated vaccine into a virulent form, OPV has been largely replaced by IPV in developed countries.
Another well known mucosal vaccine in recent years is FluMist®, which is a live attenuated, trivalent cold-adapted influenza vaccine (CAIV-T) manufactured by MedImmune, Inc [10-12]. FluMist is given as a gentle nasal mist. This nasal vaccine has been proved to be safe, effective and well tolerated in adults including those infected with HIV-1. It was approved by the United States Food and Drug Administration (FDA) in 2003 and became available on the market in the same year [10-12]. The current version of CAIV-T is a refrigerator-stable formulation of FluMist®, containing components of live attenuated influenza viruses of each of the three strains for the 2009-2010 season: A/South Dakota/6/2007(H1N1) (an A/Brisbane/59/2007-like), A/Uruguay/716/2007 (H3N2) (an A/Brisbane/10/2007-like), and B/Brisbane/60/2008. These findings have demonstrated the successful application of mucosal vaccines in protecting humans from viral infections.

3. HIV-1 infection in mucosal associated lymph tissues

Since conventional vaccines have not proven successful against HIV-1 infections, it is necessary to explore other strategies including mucosal vaccination. Genetically divergent HIV-1 and its cousin, simian immunodeficiency virus (SIV), use host proteins CD4 and the chemokine receptor CCR5 as primary and secondary receptors [13,14]. Moreover, HIV-1 and SIV both replicate optimally in activated memory CD4+CCR5+ T cells, a cell type that is abundant in mucosal lymph tissues [15]. It was first demonstrated in the SIV/macaque model system that the mucosal lymph tissues are the initial and predominant sites of SIV infection [15,16]. At the acute stage of infection, not only was the high viral load detected in the intestinal mucosa, but SIV infection resulted in pathogenic effects as indicated by the depletion of a significant proportion of CD4+ T cells in the guts but not in peripheral blood [17].
Since at this stage, similar effects have not been found in peripheral lymphoid tissues, the intestine appears to be a major target for SIV replication and the major site of CD4+ T cell loss in early SIV infection. In fact, the gut-associated lymph tissues contain about 40% of the total lymphocytes [18]. Similar findings were subsequently made in macaques infected with human/simian immunodeficiency virus (SHIV) harboring CCR5-tropic HIV-1 envelope, implicating the mucosal lymph tissues as being the initial and predominant sites of HIV-1 infection [19,20]. CD4+ T cell loss predominates in the effecter sub-compartment of the gastrointestinal (GI) mucosa at all stages of HIV-1 disease [21]. This loss did not appear to be easily reversible as there was a significantly greater CD4+ T cell loss in the GI mucosa despite over five years of fully suppressive therapy [21,22].
Since sexually transmitted HIV-1 establishes its initial replication in mucosal tissues, the induction of sufficient mucosal immunity at the initial site of HIV-1 transmission becomes essential for the development of a protective vaccine [16,18]. Some studies have tried to understand the early events following the transmission of SIV or HIV-1 via the mucosal surfaces. Both viruses were found to replicate predominantly in CD4+ T cells at the portal of entry and in lymphoid tissues [23]. The viruses were found propagating not only in activated and proliferating T cells but also in resting T cells. Because most of the HIV-1-infected resting T cells persisted after antiretroviral therapy, it poses great difficulties when attempting to eradicate the virus in vivo [23]. However, SIV did not readily disseminate to the systemic circulation but rather localized to the mucosa of viral inoculation at the very early stage of infection [23,24]. Moreover, the mucosal barrier greatly limits the infection of cervicovaginal tissues, and thus the initial founder populations of infected cells are smaller [25,26]. Recent findings have further indicated that a dramatic evolutionary bottleneck occurs, with 80% of heterosexual infections apparently initiated by a single variant when HIV-1 is transmitted into a new recipient [27,28]. The initially small founder populations and dependence on continuous seeding, to establish a productive infection in systemic lymph tissues, define a small window of maximum vulnerability for the virus in which there is an opportunity for the host, vaccines, or other interventions to prevent or control infection [26]. The depletion of memory CD4+ T cells, especially in the gut during the acute phase of infection, provide further evidence that the induction of mucosal immunity should be considered a high priority in the development of vaccines against mucosally transmitted HIV-1 [18,29,30]. These findings have provided supporting evidence for strategies by focusing on mucosal vaccines for inducing protective HIV-1 immunity at the site of viral transmission.

4. Protective mucosal immune responses at the site of viral transmission

One of the critical bottlenecks in the development of an effective HIV-1 vaccine is the current inability to induce a local mucosal immunity with a durable, high level of antibody response and high frequency cellular immune response in humans. Mucosal sIgA is considered to have an important role in the prevention of HIV-1 transmission through sexual intercourse [31]. Several studies have evaluated the neutralizing activity of sIgA purified from plasma and mucosal samples from HIV-1 highly exposed persistently seronegative individuals (HEPSIs) or infected people [32-34]. Against HIV-1 primary isolates of different viral clades and phenotypes, specific neutralizing activity of the purified IgA from cervicovaginal fluid was found in the majority of samples [32-34]. In contrast, the cervicovaginal fluid of low-risk, uninfected HIV-seronegative individuals lacked neutralizing IgA [32,33]. Moreover, mucosal HIV-1 gp41-specific IgA derived from highly exposed seronegative individuals was able to block HIV-1 epithelial transcytosis and neutralized CD4+ cell infection [35]. However, by analyzing a group of exposed uninfected female commercial sex workers from Gambia, no significant vaginal sIgA or IgG responses against HIV-1 or HIV-2 were detected, and none of the vaginal secretions tested displayed any HIV-1 neutralizing activity [36,37]. Since these studies seem controversial, it becomes necessary to study neutralizing IgA or IgG induced by a mucosal vaccine [31]. Interestingly, a recent study indicated that anti-CCR5 IgG and IgA might have contributed to protection against HIV-1 sexual transmission among exposed seronegative people [38]. It is known that mucosal polymeric IgA plays a predominant role in the protection of influenza virus-induced pathology in the upper respiratory tract [39]. Whether or not similar effects can be achieved against HIV-1 remain unknown. It was evident that functional polymeric IgA-like 2F5, by switching 2F5IgG to the IgA isotype, had a stronger impact on the protective potential of these two antibodies and could interfere with HIV-1 entry across a mucosal epithelial layer in vitro [40]. These findings indicate that although sIgA is the major humoral defense mechanism at mucosal surfaces, it is wise to induce locally produced IgM and IgG as well as serum-derived IgG via mucosal vaccination. Mucosal neutralizing IgG is also important because it protects macaques from repeated intravaginal exposure to low doses of a SHIV that uses CCR5 as its co-receptor [41,42].
It has been demonstrated in SIV/macaque models that CD8+ cytotoxic T lymphocytes (CTL) play a crucial role in suppressing SIV replication in vivo during acute and chronic infections [43,44]. In fact, based on a mathematical model, it has been suggested that the first T cell response to transmitted/founder viruses in humans is early and contributes to the initial decline of plasma virus in acute infection [45]. Besides mucosal antibody response, high frequency mucosal cellular immune responses detected are essential for preventing the local productive infection and eliminating the initial founder populations of infected cells [45-49]. HIV-1-specific CD8+ T cell responses were found in the genital mucosa of HIV-1-resistant sex workers in the absence of detectable HIV-1 infection, suggesting the protective role of cell-mediated immunity [50]. Moreover, cellular immune responses, especially those mediated by CTL and CD4+ helper T lymphocytes, are needed to control HIV-1 or SIV in the mucosal tissues [46-48,51]. In particular, the magnitude and timing of the establishment of an excess of effector cells versus targets were found to correlate with the extent of control and SIV infection outcome [16]. Therefore, vaccines capable of inducing high frequency broadly reactive cell-mediated responses are considered critical for controlling the spread of the virus [45,52]. Although multiple approaches have been developed for inducing mucosal CTL responses, these CTLs were weak or at low frequency and could not completely prevent the initial viral replication and dissemination [46-48,53].

5. Vector-based AIDS mucosal vaccine strategies

To develop an effective HIV-1 mucosal vaccine, besides immunogen design, there are three factors to be considered: the vector for antigen delivery, the adjuvant to enhance mucosal immune response and the route of immunization to induce both mucosal and systemic immune responses as early as possible [54-57]. The selection of a proper antigen delivery vector is necessary to the induction of an efficient mucosal immune response. Studies in macaques found that immunization with a live attenuated SIV vaccine can induce specific local CTLs in mucosa and dramatically reduce viremia after mucosal challenge with SIV [58-60]. Since a live attenuated HIV-1 is too risky to be used as a human vaccine, other viral vectors are under investigation for stimulating mucosal immunity, such as live attenuated vesicular stomatitis virus, non-replicating adenovirus, vaccinia virus including modified vaccinia Ankara (MVA) vectors, and Venezuelan equine encephalitis virus (VEE). To this end, the vaccinia viral vector is probably one of the most intensively studied live recombinant vectors.
Several studies have demonstrated the ability of vaccinia-based vaccines in inducing mucosal immune responses against infectious pathogens [61-63]. For example, either mucosal or systemic routes of immunization with the live, attenuated vaccinia NYVAC/SIVgpe recombinant vaccine resulted in gag-specific CD8+ T-cell responses in mucosal tissues of macaques [49]. Furthermore, vaccines based on the MVA vector were effective in inducing protective responses against different respiratory viruses such as SARS-CoV, influenza and respiratory syncytial virus following immunization via mucosal routes [64-70]. Intranasal inoculation of the MVA-based HIV-1 vaccine (named MVA-HIV) was immunogenic, whereas the intravaginal route was disappointing [62]. Since the mucosal immunogenicity of MVA-HIV is low, new approaches are needed to improve the poxvirus vector system [62]. Several approaches have been evaluated in animals. First, intranasal co-delivery of MVA-HIV plus adjuvant cholera toxin (CT) significantly enhanced the cellular and humoral immune response against HIV-1 antigens at the mucosal surfaces of vaccinated mice. Due to the limitation of CT for human use, cytokines IL-1alpha, IL-12, IL-18 and GM-CSF have been evaluated as substitutes for the induction of systemic and mucosal CTL after nasal immunization [71]. Thus, the functional activity of these candidate adjuvants needs to be tested in the context of MVA-HIV. Second, a heterologous DNA-HIV prime-MVA-HIV boost by intranasal immunization, together with CT, produced a cellular immune response in the mouse spleen 10-fold superior to that in the absence of CT [62]. Moreover, in a study using rhesus macaques, intranasal heterologous vaccination with SHIV-DNA plus IL-2/Ig DNA and rMVA induced both systematic and mucosal immunity and protected animals from disease progression against the intrarectal challenge with pathogenic SHIV89.6P [72]. In a separate study, rhesus macaques vaccinated intrarectally with a DNA construct producing replication-defective SHIV particles and boosted with rMVA-SHIV produced virus-specific mucosal and systemic humoral IgA and cell-mediated immune responses [63]. This mucosal vaccination regimen was also sufficient to delay progression to AIDS [63,73]. Although these findings are promising, there seem to be limited advantages for vaccination via the mucosal routes in comparison to the non-mucosal route of vaccination. For example, intramuscular inoculation of a heterologous DNA/MVA vaccine regimen showed a similar level of protection in monkeys [74]. It remains unknown whether or not a much better protection could be achieved after vaccinia, or other vector systems, are further optimized for stimulating host mucosal immune responses.

6. New strategies for the development of an HIV-1 mucosal vaccine

A variety of novel biological vehicles have been studied for the delivery of HIV-1 antigens including inert system and live recombinant vector system [3]. The inert system involves the direct incorporation of HIV-1 antigens into a vehicle in order to increase their immunogenicity, to protect them from degradation, and to enhance their uptake by mucosal surfaces. Commonly used inert vectors include liposomes, immunostimulating complexs (ISCOMs), lactic acid and lectins. Most of these vectors, however, are still at the early stage of preclinical studies. Since these vectors can be easily manipulated in vitro, the targeted delivery of HIV-1 antigens into antigen presentation cells (APC) might help improve the immunogenicity of mucosal vaccines of these types. For the live recombinant vector system, a number of studies have been carried out using bacterial vectors encoding HIV-1 antigens. In a phase I study, the safety and immunogenicity of an oral attenuated Salmonella enterica serovar Typhimurium delivering an HIV-1 Gag antigen via the Salmonella Type III secretion system was investigated among healthy volunteers [75]. Although more than 80% of the subjects had mucosal immune responses to vector antigens, responses specific to HIV-1 Gag were low and disappointing. Since some vaccines in the high dose group developed severe adverse events, such as diarrhea, fever, and abnormal transaminases, the vaccine probably requires further attenuation and laboratory research [76].
We have recently reported a novel replication-competent modified vaccina Tian-Tan (MVTT) as a promising mucosal vaccination vector [77]. Using SARS-CoV spike glycoprotein as a test antigen, we conducted a head-to-head comparison of MVA-S and MVTT-S. After these two vaccines were inoculated into mice via seven different routes of immunization, we found that the neutralizing antibody (Nab) response induced was significantly different between MVTT-S and MVA-S. First, intranasal (i.n.) and intraoral (i.o.) routes clearly induced the highest level of Nabs for MVTT-S but not for MVA-S. Second, if we compare the levels of Nabs elicited via the i.n. and i.o. immunizations, MVTT-S induced more than 100-fold higher Nab response than MVA-S did in this experiment. Moreover, we increased the inoculation dose of MVA-S by 10-fold (106 pfu/mouse) and did not observe significant improvement in the Nab response in mice via either i.o. or i.n. routes. In contrast, 104 pfu of MVTT-S was still able to induce significant levels of Nab response in mice via the same i.n. and i.o. routes. Therefore, our data indicate that MVTT-S may preferentially target the mucosa immune system, signifying its superiority to the MVA vector system. Further studies will evaluate MVTT as a mucosal AIDS vaccine using an SIV/macaque model. Although some animal investigations have led to the hypothesis that systemic vaccines can induce virus-specific CTLs at mucosal sites [48,78,79], it remains plausible that mucosal vaccination remains the most effective way for inducing high and long-term protective immunity at the mucosal sites of viral transmission including HIV-1 [80].

7. Summary and perspectives

With the increasing efforts in understanding the mucosal transmission of HIV-1, a greater attention has been drawn to study AIDS mucosal vaccines. Since conventional vaccine strategies have not been successful for use against AIDS, it is necessary to provide sufficient financial support and resources to study protective mucosal immunity and new vaccine methods against HIV-1 sexual transmissions. Up till now, there are still many unanswered questions for the development of a protective HIV-1 mucosal vaccine. These unanswered questions are related to HIV-1 diversity in mucosal tissues, the role of innate mucosal immunity, the lack of understanding of immune correlates of mucosal protection, the rapid formation of a latently infected CD4+ T-cell pool in mucosal reservoirs, the depletion of CD4+ T cells especially in guts through all stages of infection, the absence of effective mucosal adjuvants for human use and the poor immunogenicity of current envelope antigens to induce broadly neutralizing anti-HIV antibodies at mucosal surfaces.
The failure of Merck’s rAd5-based HIV-1 vaccine has called for efforts to further study HIV-1 immunology and vaccinology. It has been evident that it is possible to identify cell-mediated correlates of protection when the vaccine regimen is strong enough. An encouraging finding came from a recent study where the heterologous rAD26SIVgag/rAD5SIVgag regimen elicited strong cell-mediated immune responses, which correlated with significant immune control of a pathogenic SIV challenge [81]. To this end, the evaluation of various heterologous prime and boost vaccine regimens should be studied especially through mucosal routes of vaccination. This is a critical area of investigation because the magnitude and timing of the establishment of an excess of effector cells versus targets were found to be essential to the mucosal control of SIV infection [16].
Recent news from the U.S. Military HIV Research Program and the Thai Ministry of Public Health indicated that a prime-boost combination of two AIDS vaccine candidates has shown partial efficacy. Those administered with the combination vaccine has a 31% increased chance of preventing infection with HIV-1, as shown in a phase III efficacy trial in Thailand when the ‘intent-to-treat’ strategy of data analysis was modified [82]. When data were analyzed based on ‘per-protocol’ or ‘intent-to-treat’ analyses, differences dropped to 26% and results did not reach statistical significance [82]. Although it remains unknown whether or not vaccine-induced immunity has contributed to the partial protection at this stage, this finding is the first demonstration that a candidate AIDS vaccine provides benefit in humans. The AIDS vaccine research and development remain a long march. In order to discover an effective mucosal AIDS vaccine, it is necessary to have a deep understanding of mucosal immunology and to test various vaccine strategies including mucosal vaccination, which follows the fundamental rules of host natural defense.

Acknowledgments

The authors thank financial supports from Hong Kong research grant council (HK-RGC762208 and HK-RGC762209 to ZC), the National Science and Technology Major Programs (2008ZX10001-011 and 2008ZX10001-015) and the University Development Fund of the University of Hong Kong to its AIDS Institute. The authors also thank D.D. Ho and K.Y. Yuen for scientific advice and discussions.

References

  1. Holmgren, J.; Czerkinsky, C.; Lycke, N.; Svennerholm, A.M. Mucosal immunity: implications for vaccine development. Immunobiology 1992, 184, 157–179. [Google Scholar] [PubMed]
  2. Schneider, T.; Ullrich, R.; Zeitz, M. Gastrointestinal manifestations of HIV infection. Zeitschrift fuer Gastroenterologie 1994, 32, 174–181. [Google Scholar]
  3. Holmgren, J.; Czerkinsky, C. Mucosal immunity and vaccines. Nat. Med. 2005, 11, S45–S53. [Google Scholar] [CrossRef] [PubMed]
  4. Mestecky, J. Mucosal Immunology, 3rd edElsevier Academic Press: Burlington, MA, USA, 2005; p. 372. [Google Scholar]
  5. Brandtzaeg, P. Review article: Homing of mucosal immune cells--a possible connection between intestinal and articular inflammation. Aliment. Pharmacol. Ther. 1997, 11, 24–39. [Google Scholar] [PubMed]
  6. Kunkel, E.J.; Butcher, E.C. Plasma-cell homing. Nat. Rev. Immunol. 2003, 3, 822–829. [Google Scholar] [CrossRef]
  7. Paul, W.E. Fundamental Immunology, 5th edLippincott Williams & Wilkins: Philadelphia, PA, USA; London, UK, 2003; pp. 23–46. [Google Scholar]
  8. Paul, W.E. Fundamental Immunology, 6th edWolters Kluwer/Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2008; p. 1236. [Google Scholar]
  9. Sabin, A.B.; Ramos-Alvarez, M.; Alvarez-Amezquita, J.; Pelon, W.; Michaels, R.H.; Spigland, I.; Koch, M.A.; Barnes, J.M.; Rhim, J.S. Live, orally given poliovirus vaccine. Effects of rapid mass immunization on population under conditions of massive enteric infection with other viruses. JAMA 1960, 173, 1521–1526. [Google Scholar] [PubMed]
  10. Jin, H.; Lu, B.; Zhou, H.; Ma, C.; Zhao, J.; Yang, C.F.; Kemble, G.; Greenberg, H. Multiple amino acid residues confer temperature sensitivity to human influenza virus vaccine strains (FluMist) derived from cold-adapted A/Ann Arbor/6/60. Virology 2003, 306, 18–24. [Google Scholar] [CrossRef] [PubMed]
  11. Buonagurio, D.A.; Bechert, T.M.; Yang, C.F.; Shutyak, L.; D'Arco, G.A.; Kazachkov, Y.; Wang, H.P.; Rojas, E.A.; O'Neill, R.E.; Spaete, R.R.; Coelingh, K.L.; Zamb, T.J.; Sidhu, M.S.; Udem, S.A. Genetic stability of live, cold-adapted influenza virus components of the FluMist/CAIV-T vaccine throughout the manufacturing process. Vaccine 2006, 24, 2151–2160. [Google Scholar] [CrossRef] [PubMed]
  12. Harper, S.A.; Fukuda, K.; Cox, N.J.; Bridges, C.B. Using live, attenuated influenza vaccine for prevention and control of influenza: supplemental recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm. Rep. 2003, 52, 1–8. [Google Scholar] [PubMed]
  13. Zhang, L.; He, T.; Huang, Y.; Chen, Z.; Guo, Y.; Wu, S.; Kunstman, K.J.; Brown, R.C.; Phair, J.P.; Neumann, A.U.; Ho, D.D.; Wolinsky, S.M. Chemokine coreceptor usage by diverse primary isolates of human immunodeficiency virus type 1. J. Virol. 1998, 72, 9307–9312. [Google Scholar] [PubMed]
  14. Chen, Z.; Zhou, P.; Ho, D.D.; Landau, N.R.; Marx, P.A. Genetically divergent strains of simian immunodeficiency virus use CCR5 as a coreceptor for entry. J. Virol. 1997, 71, 2705–2714. [Google Scholar] [PubMed]
  15. Veazey, R.; Lackner, A. The mucosal immune system and HIV-1 infection. AIDS Rev 2003, 5, 245–252. [Google Scholar] [PubMed]
  16. Li, Q.; Skinner, P.J.; Ha, S.J.; Duan, L.; Mattila, T.L.; Hage, A.; White, C.; Barber, D.L.; O'Mara, L.; Southern, P.J.; Reilly, C.S.; Carlis, J.V.; Miller, C.J.; Ahmed, R.; Haase, A.T. Visualizing antigen-specific and infected cells in situ predicts outcomes in early viral infection. Science 2009, 323, 1726–1729. [Google Scholar] [CrossRef] [PubMed]
  17. Veazey, R.S.; DeMaria, M.; Chalifoux, L.V.; Shvetz, D.E.; Pauley, D.R.; Knight, H.L.; Rosenzweig, M.; Johnson, R.P.; Desrosiers, R.C.; Lackner, A.A. Gastrointestinal tract as a major site of CD4+ T cell depletion and viral replication in SIV infection. Science 1998, 280, 427–431. [Google Scholar] [CrossRef] [PubMed]
  18. Lackner, A.A.; Mohan, M.; Veazey, R.S. The gastrointestinal tract and AIDS pathogenesis. Gastroenterology 2009, 136, 1965–1978. [Google Scholar] [CrossRef] [PubMed]
  19. Harouse, J.M.; Gettie, A.; Tan, R.C.; Blanchard, J.; Cheng-Mayer, C. Distinct pathogenic sequela in rhesus macaques infected with CCR5 or CXCR4 utilizing SHIVs. Science 1999, 284, 816–819. [Google Scholar] [CrossRef] [PubMed]
  20. Chen, Z.; Zhao, X.; Huang, Y.; Gettie, A.; Ba, L.; Blanchard, J.; Ho, D.D. CD4+ lymphocytopenia in acute infection of Asian macaques by a vaginally transmissible subtype-C, CCR5-tropic Simian/Human Immunodeficiency Virus (SHIV). J. Acquir. Immune Defic. Syndr. 2002, 30, 133–145. [Google Scholar] [PubMed]
  21. Brenchley, J.M.; Schacker, T.W.; Ruff, L.E.; Price, D.A.; Taylor, J.H.; Beilman, G.J.; Nguyen, P.L.; Khoruts, A.; Larson, M.; Haase, A.T.; Douek, D.C. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J. Exp. Med. 2004, 200, 749–759. [Google Scholar] [CrossRef] [PubMed]
  22. Mehandru, S.; Poles, M.A.; Tenner-Racz, K.; Horowitz, A.; Hurley, A.; Hogan, C.; Boden, D.; Racz, P.; Markowitz, M. Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J. Exp. Med. 2004, 200, 761–770. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, Z.; Schuler, T.; Zupancic, M.; Wietgrefe, S.; Staskus, K.A.; Reimann, K.A.; Reinhart, T.A.; Rogan, M.; Cavert, W.; Miller, C.J.; Veazey, R.S.; Notermans, D.; Little, S.; Danner, S.A.; Richman, D.D.; Havlir, D.; Wong, J.; Jordan, H.L.; Schacker, T.W.; Racz, P.; Tenner-Racz, K.; Letvin, N.L.; Wolinsky, S.; Haase, A.T. Sexual transmission and propagation of SIV and HIV in resting and activated CD4+ T cells. Science 1999, 286, 1353–1357. [Google Scholar] [CrossRef] [PubMed]
  24. Spira, A.I.; Marx, P.A.; Patterson, B.K.; Mahoney, J.; Koup, R.A.; Wolinsky, S.M.; Ho, D.D. Cellular targets of infection and route of viral dissemination after an intravaginal inoculation of simian immunodeficiency virus into rhesus macaques. J. Exp. Med. 1996, 183, 215–225. [Google Scholar] [CrossRef] [PubMed]
  25. Smith, S.M.; Baskin, G.B.; Marx, P.A. Estrogen protects against vaginal transmission of simian immunodeficiency virus. J. Infect. Dis. 2000, 182, 708–715. [Google Scholar] [CrossRef] [PubMed]
  26. Miller, C.J.; Li, Q.; Abel, K.; Kim, E.Y.; Ma, Z.M.; Wietgrefe, S.; La Franco-Scheuch, L.; Compton, L.; Duan, L.; Shore, M.D.; Zupancic, M.; Busch, M.; Carlis, J.; Wolinsky, S.; Haase, A.T. Propagation and dissemination of infection after vaginal transmission of simian immunodeficiency virus. J. Virol. 2005, 79, 9217–9127. [Google Scholar] [CrossRef] [PubMed]
  27. Salazar-Gonzalez, J.F.; Salazar, M.G.; Keele, B.F.; Learn, G.H.; Giorgi, E.E.; Li, H.; Decker, J.M.; Wang, S.; Baalwa, J.; Kraus, M.H.; Parrish, N.F.; Shaw, K.S.; Guffey, M.B.; Bar, K.J.; Davis, K.L.; Ochsenbauer-Jambor, C.; Kappes, J.C.; Saag, M.S.; Cohen, M.S.; Mulenga, J.; Derdeyn, C.A.; Allen, S.; Hunter, E.; Markowitz, M.; Hraber, P.; Perelson, A.S.; Bhattacharya, T.; Haynes, B.F.; Korber, B.T.; Hahn, B.H.; Shaw, G.M. Genetic identity, biological phenotype, and evolutionary pathways of transmitted/founder viruses in acute and early HIV-1 infection. J. Exp. Med. 2009, 206, 1273–1289. [Google Scholar] [CrossRef] [PubMed]
  28. Keele, B.F.; Giorgi, E.E.; Salazar-Gonzalez, J.F.; Decker, J.M.; Pham, K.T.; Salazar, M.G.; Sun, C.; Grayson, T.; Wang, S.; Li, H.; Wei, X.; Jiang, C.; Kirchherr, J.L.; Gao, F.; Anderson, J.A.; Ping, L.H.; Swanstrom, R.; Tomaras, G.D.; Blattner, W.A.; Goepfert, P.A.; Kilby, J.M.; Saag, M.S.; Delwart, E.L.; Busch, M.P.; Cohen, M.S.; Montefiori, D.C.; Haynes, B.F.; Gaschen, B.; Athreya, G.S.; Lee, H.Y.; Wood, N.; Seoighe, C.; Perelson, A.S.; Bhattacharya, T.; Korber, B.T.; Hahn, B.H.; Shaw, G.M. Identification and characterization of transmitted and early founder virus envelopes in primary HIV-1 infection. Proc. Natl. Acad. Sci. USA 2008, 105, 7552–7557. [Google Scholar] [CrossRef]
  29. Mattapallil, J.J.; Douek, D.C.; Hill, B.; Nishimura, Y.; Martin, M.; Roederer, M. Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection. Nature 2005, 434, 1093–1097. [Google Scholar] [CrossRef] [PubMed]
  30. Li, Q.; Duan, L.; Estes, J.D.; Ma, Z.M.; Rourke, T.; Wang, Y.; Reilly, C.; Carlis, J.; Miller, C.J.; Haase, A.T. Peak SIV replication in resting memory CD4+ T cells depletes gut lamina propria CD4+ T cells. Nature 2005, 434, 1148–1152. [Google Scholar] [PubMed]
  31. Alexander, R.; Mestecky, J. Neutralizing antibodies in mucosal secretions: IgG or IgA? Curr. HIV Res. 2007, 5, 588–593. [Google Scholar] [CrossRef] [PubMed]
  32. Devito, C.; Hinkula, J.; Kaul, R.; Lopalco, L.; Bwayo, J.J.; Plummer, F.; Clerici, M.; Broliden, K. Mucosal and plasma IgA from HIV-exposed seronegative individuals neutralize a primary HIV-1 isolate. Aids 2000, 14, 1917–1920. [Google Scholar] [CrossRef] [PubMed]
  33. Devito, C.; Hinkula, J.; Kaul, R.; Kimani, J.; Kiama, P.; Lopalco, L.; Barass, C.; Piconi, S.; Trabattoni, D.; Bwayo, J.J.; Plummer, F.; Clerici, M.; Broliden, K. Cross-clade HIV-1-specific neutralizing IgA in mucosal and systemic compartments of HIV-1-exposed, persistently seronegative subjects. J. Acquir. Immune Defic. Syndr. 2002, 30, 413–420. [Google Scholar] [PubMed]
  34. Broliden, K.; Hinkula, J.; Devito, C.; Kiama, P.; Kimani, J.; Trabbatoni, D.; Bwayo, J.J.; Clerici, M.; Plummer, F.; Kaul, R. Functional HIV-1 specific IgA antibodies in HIV-1 exposed, persistently IgG seronegative female sex workers. Immunol. Lett. 2001, 79, 29–36. [Google Scholar] [CrossRef] [PubMed]
  35. Tudor, D.; Derrien, M.; Diomede, L.; Drillet, A.S.; Houimel, M.; Moog, C.; Reynes, J.M.; Lopalco, L.; Bomsel, M. HIV-1 gp41-specific monoclonal mucosal IgAs derived from highly exposed but IgG-seronegative individuals block HIV-1 epithelial transcytosis and neutralize CD4(+) cell infection: an IgA gene and functional analysis. Mucosal Immunol. 2009, 2, 412–426. [Google Scholar] [CrossRef]
  36. Dorrell, L.; Hessell, A.J.; Wang, M.; Whittle, H.; Sabally, S.; Rowland-Jones, S.; Burton, D.R.; Parren, P.W. Absence of specific mucosal antibody responses in HIV-exposed uninfected sex workers from the Gambia. Aids 2000, 14, 1117–1122. [Google Scholar] [CrossRef] [PubMed]
  37. Schneider, J.A.; Alam, S.A.; Ackers, M.; Parekh, B.; Chen, H.Y.; Graham, P.; Gurwith, M.; Mayer, K.; Novak, R.M. Mucosal HIV-binding antibody and neutralizing activity in high-risk HIV-uninfected female participants in a trial of HIV-vaccine efficacy. J. Infect. Dis. 2007, 196, 1637–1644. [Google Scholar] [CrossRef] [PubMed]
  38. Pastori, C.; Weiser, B.; Barassi, C.; Uberti-Foppa, C.; Ghezzi, S.; Longhi, R.; Calori, G.; Burger, H.; Kemal, K.; Poli, G.; Lazzarin, A.; Lopalco, L. Long-lasting CCR5 internalization by antibodies in a subset of long-term nonprogressors: a possible protective effect against disease progression. Blood 2006, 107, 4825–4833. [Google Scholar] [CrossRef] [PubMed]
  39. Renegar, K.B.; Small Jr., P.A.; Boykins, L.G.; Wright, P.F. Role of IgA versus IgG in the control of influenza viral infection in the murine respiratory tract. J. Immunol. 2004, 173, 1978–1986. [Google Scholar] [PubMed]
  40. Wolbank, S.; Kunert, R.; Stiegler, G.; Katinger, H. Characterization of human class-switched polymeric (immunoglobulin M [IgM] and IgA) anti-human immunodeficiency virus type 1 antibodies 2F5 and 2G12. J. Virol. 2003, 77, 4095–40103. [Google Scholar] [CrossRef] [PubMed]
  41. Hessell, A.J.; Poignard, P.; Hunter, M.; Hangartner, L.; Tehrani, D.M.; Bleeker, W.K.; Parren, P.W.; Marx, P.A.; Burton, D.R. Effective, low-titer antibody protection against low-dose repeated mucosal SHIV challenge in macaques. Nat. Med. 2009, 15, 951–954. [Google Scholar] [CrossRef] [PubMed]
  42. Hessell, A.J.; Rakasz, E.G.; Poignard, P.; Hangartner, L.; Landucci, G.; Forthal, D.N.; Koff, W.C.; Watkins, D.I.; Burton, D.R. Broadly neutralizing human anti-HIV antibody 2G12 is effective in protection against mucosal SHIV challenge even at low serum neutralizing titers. PLoS Pathog. 2009, 5, e1000433:1–e1000433:9. [Google Scholar] [CrossRef] [Green Version]
  43. Jin, X.; Bauer, D.E.; Tuttleton, S.E.; Lewin, S.; Gettie, A.; Blanchard, J.; Irwin, C.E.; Safrit, J.T.; Mittler, J.; Weinberger, L.; Kostrikis, L.G.; Zhang, L.; Perelson, A.S.; Ho, D.D. Dramatic rise in plasma viremia after CD8(+) T cell depletion in simian immunodeficiency virus-infected macaques. J. Exp. Med. 1999, 189, 991–998. [Google Scholar] [CrossRef] [PubMed]
  44. Schmitz, J.E.; Kuroda, M.J.; Santra, S.; Sasseville, V.G.; Simon, M.A.; Lifton, M.A.; Racz, P.; Tenner-Racz, K.; Dalesandro, M.; Scallon, B.J.; Ghrayeb, J.; Forman, M.A.; Montefiori, D.C.; Rieber, E.P.; Letvin, N.L.; Reimann, K.A. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science 1999, 283, 857–860. [Google Scholar] [CrossRef] [PubMed]
  45. Goonetilleke, N.; Liu, M.K.; Salazar-Gonzalez, J.F.; Ferrari, G.; Giorgi, E.; Ganusov, V.V.; Keele, B.F.; Learn, G.H.; Turnbull, E.L.; Salazar, M.G.; Weinhold, K.J.; Moore, S.; Letvin, N.; Haynes, B.F.; Cohen, M.S.; Hraber, P.; Bhattacharya, T.; Borrow, P.; Perelson, A.S.; Hahn, B.H.; Shaw, G.M.; Korber, B.T.; McMichael, A.J. The first T cell response to transmitted/founder virus contributes to the control of acute viremia in HIV-1 infection. J. Exp. Med. 2009, 206, 1253–1272. [Google Scholar] [CrossRef] [PubMed]
  46. Vogel, T.U.; Reynolds, M.R.; Fuller, D.H.; Vielhuber, K.; Shipley, T.; Fuller, J.T.; Kunstman, K.J.; Sutter, G.; Marthas, M.L.; Erfle, V.; Wolinsky, S.M.; Wang, C.; Allison, D.B.; Rud, E.W.; Wilson, N.; Montefiori, D.; Altman, J.D.; Watkins, D.I. Multispecific vaccine-induced mucosal cytotoxic T lymphocytes reduce acute-phase viral replication but fail in long-term control of simian immunodeficiency virus SIVmac239. J. Virol. 2003, 77, 13348–13360. [Google Scholar] [CrossRef] [PubMed]
  47. Evans, D.T.; Chen, L.M.; Gillis, J.; Lin, K.C.; Harty, B.; Mazzara, G.P.; Donis, R.O.; Mansfield, K.G.; Lifson, J.D.; Desrosiers, R.C.; Galan, J.E.; Johnson, R.P. Mucosal priming of simian immunodeficiency virus-specific cytotoxic T-lymphocyte responses in rhesus macaques by the Salmonella type III secretion antigen delivery system. J. Virol. 2003, 77, 2400–2409. [Google Scholar] [CrossRef] [PubMed]
  48. Baig, J.; Levy, D.B.; McKay, P.F.; Schmitz, J.E.; Santra, S.; Subbramanian, R.A.; Kuroda, M.J.; Lifton, M.A.; Gorgone, D.A.; Wyatt, L.S.; Moss, B.; Huang, Y.; Chakrabarti, B.K.; Xu, L.; Kong, W.P.; Yang, Z.Y.; Mascola, J.R.; Nabel, G.J.; Carville, A.; Lackner, A.A.; Veazey, R.S.; Letvin, N.L. Elicitation of simian immunodeficiency virus-specific cytotoxic T lymphocytes in mucosal compartments of rhesus monkeys by systemic vaccination. J. Virol. 2002, 76, 11484–11490. [Google Scholar] [CrossRef] [PubMed]
  49. Stevceva, L.; Alvarez, X.; Lackner, A.A.; Tryniszewska, E.; Kelsall, B.; Nacsa, J.; Tartaglia, J.; Strober, W.; Franchini, G. Both mucosal and systemic routes of immunization with the live, attenuated NYVAC/simian immunodeficiency virus SIV(gpe) recombinant vaccine result in gag-specific CD8(+) T-cell responses in mucosal tissues of macaques. J. Virol. 2002, 76, 11659–11676. [Google Scholar] [CrossRef] [PubMed]
  50. Kaul, R.; Plummer, F.A.; Kimani, J.; Dong, T.; Kiama, P.; Rostron, T.; Njagi, E.; MacDonald, K.S.; Bwayo, J.J.; McMichael, A.J.; Rowland-Jones, S.L. HIV-1-specific mucosal CD8+ lymphocyte responses in the cervix of HIV-1-resistant prostitutes in Nairobi. J. Immunol. 2000, 164, 1602–1611. [Google Scholar] [PubMed]
  51. Ibarrondo, F.J.; Anton, P.A.; Fuerst, M.; Ng, H.L.; Wong, J.T.; Matud, J.; Elliott, J.; Shih, R.; Hausner, M.A.; Price, C.; Hultin, L.E.; Hultin, P.M.; Jamieson, B.D.; Yang, O.O. Parallel human immunodeficiency virus type 1-specific CD8+ T-lymphocyte responses in blood and mucosa during chronic infection. J. Virol. 2005, 79, 4289–4297. [Google Scholar] [CrossRef] [PubMed]
  52. Bourinbaiar, A.S.; Metadilogkul, O.; Jirathitikal, V. Mucosal AIDS vaccines. Viral Immunol. 2003, 16, 427–445. [Google Scholar] [PubMed]
  53. Jiang, J.Q.; Patrick, A.; Moss, R.B.; Rosenthal, K.L. CD8+ T-cell-mediated cross-clade protection in the genital tract following intranasal immunization with inactivated human immunodeficiency virus antigen plus CpG oligodeoxynucleotides. J. Virol. 2005, 79, 393–400. [Google Scholar] [CrossRef] [PubMed]
  54. Walker, R.I. New strategies for using mucosal vaccination to achieve more effective immunization. Vaccine 1994, 12, 387–400. [Google Scholar] [CrossRef] [PubMed]
  55. Kyd, J.M.; Cripps, A.W. Killed whole bacterial cells, a mucosal delivery system for the induction of immunity in the respiratory tract and middle ear: an overview. Vaccine 1999, 17, 1775–1781. [Google Scholar] [CrossRef] [PubMed]
  56. Kyd, J.; Cripps, A. Identifying vaccine antigens and assessing delivery systems for the prevention of bacterial infections. J. Biotechnol. 2000, 83, 85–90. [Google Scholar] [CrossRef] [PubMed]
  57. Cripps, A.W.; Kyd, J.M.; Foxwell, A.R. Vaccines and mucosal immunisation. Vaccine 2001, 19, 2513–2515. [Google Scholar] [CrossRef] [PubMed]
  58. Nilsson, C.; Makitalo, B.; Thorstensson, R.; Norley, S.; Binninger-Schinzel, D.; Cranage, M.; Rud, E.; Biberfeld, G.; Putkonen, P. Live attenuated simian immunodeficiency virus (SIV)mac in macaques can induce protection against mucosal infection with SIVsm. AIDS (London, England) 1998, 12, 2261–2270. [Google Scholar] [PubMed]
  59. Cromwell, M.A.; Veazey, R.S.; Altman, J.D.; Mansfield, K.G.; Glickman, R.; Allen, T.M.; Watkins, D.I.; Lackner, A.A.; Johnson, R.P. Induction of mucosal homing virus-specific CD8(+) T lymphocytes by attenuated simian immunodeficiency virus. J. Virol. 2000, 74, 8762–8766. [Google Scholar] [CrossRef] [PubMed]
  60. Tenner-Racz, K.; Stahl Hennig, C.; Uberla, K.; Stoiber, H.; Ignatius, R.; Heeney, J.; Steinman, R.M.; Racz, P. Early protection against pathogenic virus infection at a mucosal challenge site after vaccination with attenuated simian immunodeficiency virus. Proc. Natl. Acad. Sci. USA 2004, 101, 3017–3022. [Google Scholar] [CrossRef]
  61. Kent, S.J.; Dale, C.J.; Ranasinghe, C.; Stratov, I.; De Rose, R.; Chea, S.; Montefiori, D.C.; Thomson, S.; Ramshaw, I.A.; Coupar, B.E.; Boyle, D.B.; Law, M.; Wilson, K.M.; Ramsay, A.J. Mucosally-administered human-simian immunodeficiency virus DNA and fowlpoxvirus-based recombinant vaccines reduce acute phase viral replication in macaques following vaginal challenge with CCR5-tropic SHIVSF162P3. Vaccine 2005, 23, 5009–5021. [Google Scholar] [CrossRef] [PubMed]
  62. Gherardi, M.M.; Perez-Jimenez, E.; Najera, J.L.; Esteban, M. Induction of HIV immunity in the genital tract after intranasal delivery of a MVA vector: enhanced immunogenicity after DNA prime-modified vaccinia virus Ankara boost immunization schedule. J. Immunol. 2004, 172, 6209–6220. [Google Scholar] [PubMed]
  63. Wang, S.W.; Bertley, F.M.; Kozlowski, P.A.; Herrmann, L.; Manson, K.; Mazzara, G.; Piatak, M.; Johnson, R.P.; Carville, A.; Mansfield, K.; Aldovini, A. An SHIV DNA/MVA rectal vaccination in macaques provides systemic and mucosal virus-specific responses and protection against AIDS. AIDS Res. Hum. Retroviruses 2004, 20, 846–859. [Google Scholar] [CrossRef] [PubMed]
  64. Chen, Z.; Zhang, L.; Qin, C.; Ba, L.; Yi, C.E.; Zhang, F.; Wei, Q.; He, T.; Yu, W.; Yu, J.; Gao, H.; Tu, X.; Gettie, A.; Farzan, M.; Yuen, K.Y.; Ho, D.D. Recombinant modified vaccinia virus Ankara expressing the spike glycoprotein of severe acute respiratory syndrome coronavirus induces protective neutralizing antibodies primarily targeting the receptor binding region. J. Virol. 2005, 79, 2678–2688. [Google Scholar] [CrossRef] [PubMed]
  65. Bisht, H.; Roberts, A.; Vogel, L.; Bukreyev, A.; Collins, P.L.; Murphy, B.R.; Subbarao, K.; Moss, B. Severe acute respiratory syndrome coronavirus spike protein expressed by attenuated vaccinia virus protectively immunizes mice. Proc. Natl. Acad. Sci. USA 2004, 101, 6641–6646. [Google Scholar] [CrossRef]
  66. Degano, P.; Schneider, J.; Hannan, C.M.; Gilbert, S.C.; Hill, A.V. Gene gun intradermal DNA immunization followed by boosting with modified vaccinia virus Ankara: enhanced CD8+ T cell immunogenicity and protective efficacy in the influenza and malaria models. Vaccine 1999, 18, 623–632. [Google Scholar] [CrossRef] [PubMed]
  67. Sutter, G.; Wyatt, L.S.; Foley, P.L.; Bennink, J.R.; Moss, B. A recombinant vector derived from the host range-restricted and highly attenuated MVA strain of vaccinia virus stimulates protective immunity in mice to influenza virus. Vaccine 1994, 12, 1032–1040. [Google Scholar] [CrossRef] [PubMed]
  68. Olszewska, W.; Suezer, Y.; Sutter, G.; Openshaw, P.J. Protective and disease-enhancing immune responses induced by recombinant modified vaccinia Ankara (MVA) expressing respiratory syncytial virus proteins. Vaccine 2004, 23, 215–221. [Google Scholar] [CrossRef] [PubMed]
  69. de Waal, L.; Wyatt, L.S.; Yuksel, S.; van Amerongen, G.; Moss, B.; Niesters, H.G.; Osterhaus, A.D.; de Swart, R.L. Vaccination of infant macaques with a recombinant modified vaccinia virus Ankara expressing the respiratory syncytial virus F and G genes does not predispose for immunopathology. Vaccine 2004, 22, 923–926. [Google Scholar] [CrossRef] [PubMed]
  70. Wyatt, L.S.; Whitehead, S.S.; Venanzi, K.A.; Murphy, B.R.; Moss, B. Priming and boosting immunity to respiratory syncytial virus by recombinant replication-defective vaccinia virus MVA. Vaccine 1999, 18, 392–397. [Google Scholar] [CrossRef] [PubMed]
  71. Staats, H.F.; Bradney, C.P.; Gwinn, W.M.; Jackson, S.S.; Sempowski, G.D.; Liao, H.X.; Letvin, N.L.; Haynes, B.F. Cytokine requirements for induction of systemic and mucosal CTL after nasal immunization. J. Immunol. 2001, 167, 5386–5394. [Google Scholar] [PubMed]
  72. Bertley, F.M.; Kozlowski, P.A.; Wang, S.W.; Chappelle, J.; Patel, J.; Sonuyi, O.; Mazzara, G.; Montefiori, D.; Carville, A.; Mansfield, K.G.; Aldovini, A. Control of simian/human immunodeficiency virus viremia and disease progression after IL-2-augmented DNA-modified vaccinia virus Ankara nasal vaccination in nonhuman primates. J. Immunol. 2004, 172, 3745–3757. [Google Scholar] [PubMed]
  73. Makitalo, B.; Lundholm, P.; Hinkula, J.; Nilsson, C.; Karlen, K.; Morner, A.; Sutter, G.; Erfle, V.; Heeney, J.L.; Wahren, B.; Biberfeld, G.; Thorstensson, R. Enhanced cellular immunity and systemic control of SHIV infection by combined parenteral and mucosal administration of a DNA prime MVA boost vaccine regimen. J. Gen. Virol. 2004, 85, 2407–2419. [Google Scholar] [CrossRef] [PubMed]
  74. Amara, R.R.; Villinger, F.; Altman, J.D.; Lydy, S.L.; O'Neil, S.P.; Staprans, S.I.; Montefiori, D.C.; Xu, Y.; Herndon, J.G.; Wyatt, L.S.; Candido, M.A.; Kozyr, N.L.; Earl, P.L.; Smith, J.M.; Ma, H.L.; Grimm, B.D.; Hulsey, M.L.; Miller, J.; McClure, H.M.; McNicholl, J.M.; Moss, B.; Robinson, H.L. Control of a mucosal challenge and prevention of AIDS by a multiprotein DNA/MVA vaccine. Science 2001, 292, 69–74. [Google Scholar] [CrossRef] [PubMed]
  75. Kotton, C.N.; Lankowski, A.J.; Scott, N.; Sisul, D.; Chen, L.M.; Raschke, K.; Borders, G.; Boaz, M.; Spentzou, A.; Galan, J.E.; Hohmann, E.L. Safety and immunogenicity of attenuated Salmonella enterica serovar Typhimurium delivering an HIV-1 Gag antigen via the Salmonella Type III secretion system. Vaccine 2006, 24, 6216–6224. [Google Scholar] [CrossRef] [PubMed]
  76. Lewis, G.K. Live-attenuated Salmonella as a prototype vaccine vector for passenger immunogens in humans: are we there yet? Expert Rev. Vaccines 2007, 6, 431–440. [Google Scholar] [CrossRef] [PubMed]
  77. Huang, X.; Lu, B.; Yu, W.; Fang, Q.; Liu, L.; Zhuang, K.; Shen, T.; Wang, H.; Tian, P.; Zhang, L.; Chen, Z. A novel replication-competent vaccinia vector MVTT is superior to MVA for inducing high levels of neutralizing antibody via mucosal vaccination. PLoS One 2009, 4, e4180:1–e4180:9. [Google Scholar]
  78. Mitchell, E.A.; Bergmeier, L.A.; Doyle, C.; Brookes, R.; Hussain, L.A.; Wang, Y.; Lehner, T. Homing of mononuclear cells from iliac lymph nodes to the genital and rectal mucosa in non-human primates. Eur. J. Immunol. 1998, 28, 3066–3074. [Google Scholar] [CrossRef] [PubMed]
  79. Belyakov, I.M.; Hammond, S.A.; Ahlers, J.D.; Glenn, G.M.; Berzofsky, J.A. Transcutaneous immunization induces mucosal CTLs and protective immunity by migration of primed skin dendritic cells. J. Clin. Invest. 2004, 113, 998–1007. [Google Scholar] [PubMed]
  80. Yu, W.; Fang, Q.; Zhu, W.; Wang, H.; Tien, P.; Zhang, L.; Chen, Z. One time intranasal vaccination with a modified vaccinia Tiantan strain MVTTZCI protects animals against pathogenic viral challenge. Vaccine 2009. [Google Scholar]
  81. Liu, J.; O'Brien, K.L.; Lynch, D.M.; Simmons, N.L.; La Porte, A.; Riggs, A.M.; Abbink, P.; Coffey, R.T.; Grandpre, L.E.; Seaman, M.S.; Landucci, G.; Forthal, D.N.; Montefiori, D.C.; Carville, A.; Mansfield, K.G.; Havenga, M.J.; Pau, M.G.; Goudsmit, J.; Barouch, D.H. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature 2009, 457, 87–91. [Google Scholar] [CrossRef] [PubMed]
  82. Rerks-Ngarm, S.; Pitisuttithum, P.; Nitayaphan, S.; Kaewkungwal, J.; Chiu, J.; Paris, R.; Premsri, N.; Namwat, C.; de Souza, M.; Adams, E.; Benenson, M.; Gurunathan, S.; Tartaglia, J.; McNeil, J.G.; Francis, D.P.; Stablein, D.; Birx, D.L.; Chunsuttiwat, S.; Khamboonruang, C.; Thongcharoen, P.; Robb, M.L.; Michael, N.L.; Kunasol, P.; Kim, J.H. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N. Engl. J. Med. 2009, 361, 2209–2220. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Tang, X.; Chen, Z. The Development of an AIDS Mucosal Vaccine. Viruses 2010, 2, 283-297. https://doi.org/10.3390/v2010283

AMA Style

Tang X, Chen Z. The Development of an AIDS Mucosal Vaccine. Viruses. 2010; 2(1):283-297. https://doi.org/10.3390/v2010283

Chicago/Turabian Style

Tang, Xian, and Zhiwei Chen. 2010. "The Development of an AIDS Mucosal Vaccine" Viruses 2, no. 1: 283-297. https://doi.org/10.3390/v2010283

Article Metrics

Back to TopTop