1
|
Needle-free, spirulina-produced Plasmodium falciparum circumsporozoite vaccination provides sterile protection against pre-erythrocytic malaria in mice. NPJ Vaccines 2022; 7:113. [PMID: 36195607 PMCID: PMC9532447 DOI: 10.1038/s41541-022-00534-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 09/05/2022] [Indexed: 02/02/2023] Open
Abstract
Antibodies against the Plasmodium falciparum circumsporozoite protein (PfCSP) can block hepatocyte infection by sporozoites and protect against malaria. Needle-free vaccination strategies are desirable, yet most PfCSP-targeted vaccines like RTS,S require needle-based administration. Here, we evaluated the edible algae, Arthrospira platensis (commonly called 'spirulina') as a malaria vaccine platform. Spirulina were genetically engineered to express virus-like particles (VLPs) consisting of the woodchuck hepatitis B core capsid protein (WHcAg) displaying a (NANP)15 PfCSP antigen on its surface. PfCSP-spirulina administered to mice intranasally followed by oral PfCSP-spirulina boosters resulted in a strong, systemic anti-PfCSP immune response that was protective against subcutaneous challenge with PfCSP-expressing P. yoelii. Unlike male mice, female mice did not require Montanide adjuvant to reach high antibody titers or protection. The successful use of spirulina as a vaccine delivery system warrants further development of spirulina-based vaccines as a useful tool in addressing malaria and other diseases of global health importance.
Collapse
|
2
|
Molina-Franky J, Cuy-Chaparro L, Camargo A, Reyes C, Gómez M, Salamanca DR, Patarroyo MA, Patarroyo ME. Plasmodium falciparum pre-erythrocytic stage vaccine development. Malar J 2020; 19:56. [PMID: 32013956 PMCID: PMC6998842 DOI: 10.1186/s12936-020-3141-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 01/25/2020] [Indexed: 12/13/2022] Open
Abstract
Worldwide strategies between 2010 and 2017 aimed at controlling malarial parasites (mainly Plasmodium falciparum) led to a reduction of just 18% regarding disease incidence rates. Many biologically-derived anti-malarial vaccine candidates have been developed to date; this has involved using many experimental animals, an immense amount of work and the investment of millions of dollars. This review provides an overview of the current state and the main results of clinical trials for sporozoite-targeting vaccines (i.e. the parasite stage infecting the liver) carried out by research groups in areas having variable malaria transmission rates. However, none has led to promising results regarding the effective control of the disease, thereby making it necessary to complement such efforts at finding/introducing new vaccine candidates by adopting a multi-epitope, multi-stage approach, based on minimal subunits of the main sporozoite proteins involved in the invasion of the liver.
Collapse
Affiliation(s)
- Jessica Molina-Franky
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Laura Cuy-Chaparro
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Anny Camargo
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - César Reyes
- PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia.,Animal Science Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Bogotá, Colombia.,3D Structures Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Marcela Gómez
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - David Ricardo Salamanca
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia. .,Basic Sciences Department, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.
| | - Manuel Elkin Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia. .,Medical School, Universidad Nacional de Colombia, Bogotá, Colombia.
| |
Collapse
|
3
|
Goh YS, McGuire D, Rénia L. Vaccination With Sporozoites: Models and Correlates of Protection. Front Immunol 2019; 10:1227. [PMID: 31231377 PMCID: PMC6560154 DOI: 10.3389/fimmu.2019.01227] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Despite continuous efforts, the century-old goal of eradicating malaria still remains. Multiple control interventions need to be in place simultaneously to achieve this goal. In addition to effective control measures, drug therapies and insecticides, vaccines are critical to reduce mortality and morbidity. Hence, there are numerous studies investigating various malaria vaccine candidates. Most of the malaria vaccine candidates are subunit vaccines. However, they have shown limited efficacy in Phase II and III studies. To date, only whole parasite formulations have been shown to induce sterile immunity in human. In this article, we review and discuss the recent developments in vaccination with sporozoites and the mechanisms of protection involved.
Collapse
Affiliation(s)
- Yun Shan Goh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore
| | - Daniel McGuire
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
4
|
Improved lysis efficiency and immunogenicity of Salmonella ghosts mediated by co-expression of λ phage holin-endolysin and ɸX174 gene E. Sci Rep 2017; 7:45139. [PMID: 28332591 PMCID: PMC5362813 DOI: 10.1038/srep45139] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/15/2017] [Indexed: 12/19/2022] Open
Abstract
Bacterial ghosts (BGs) are empty cell envelopes derived from Gram-negative bacteria by bacteriophage ɸX174 gene E mediated lysis. They represent a novel inactivated vaccine platform; however, the practical application of BGs for human vaccines seems to be limited due to the safety concerns on the presence of viable cells in BGs. Therefore, to improve the lysis efficiency of the gene E, we exploited the peptidoglycan hydrolyzing ability of the λ phage holin-endolysins to expedite the process of current BG production system. In this report, we constructed a novel ghost plasmid encoding protein E and holin-endolysins in tandem. We observed that sequential expressions of the gene E and the holin-endolysins elicited rapid and highly efficient Salmonella lysis compared to the lysis mediated by gene E only. These lysed BGs displayed improved immunogenicity in mice compared to the gene E mediated BGs. Consequently, seventy percent of the mice immunized with these novel ghosts survived against a lethal challenge while all the mice vaccinated with gene E mediated ghosts died by day 9 post-infection. We conclude that this novel strategy has the potential to generate highly efficient inactivated candidate vaccines that could replace the currently available bacterial vaccines.
Collapse
|
5
|
Lewis GK. Live-attenuatedSalmonellaas a prototype vaccine vector for passenger immunogens in humans: are we there yet? Expert Rev Vaccines 2014; 6:431-40. [PMID: 17542757 DOI: 10.1586/14760584.6.3.431] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
It has been nearly 20 years since the first Phase I clinical trial of a live-attenuated bacterial vaccine was created by recombinant DNA methods, opening the door to the use of these organisms as mucosal delivery vehicles for passenger antigens. Over this time, a number of animal studies have indicated the feasibility of this approach. These include studies showing that bacteria can deliver antigens expressed by the bacterium itself and that bacteria can deliver DNA vaccines to be expressed in target eukaryotic cells. Concomitant studies have identified a number of attenuating mutations that render the bacterial vectors both safe and immunogenic in humans. Both avenues of research indicate the significant promise of this approach to mucosal vaccine development; however, this promise remains largely unrealized at the level of human clinical trials. This review sketches the history of this problem and points toward possible solutions using Salmonella vaccine vectors as the prototypes.
Collapse
Affiliation(s)
- George K Lewis
- Division of Basic Science and Vaccine Research, Institute of Human Virology, University of Maryland Biotechnology Institute and University of Maryland Baltimore, 725 W. Lombard Street, Baltimore, MD 21218, USA.
| |
Collapse
|
6
|
Chin'ombe N, Ruhanya V. Recombinant Salmonella Bacteria Vectoring HIV/AIDS Vaccines. Open Virol J 2013; 7:121-6. [PMID: 24478808 PMCID: PMC3905348 DOI: 10.2174/1874357901307010121] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/13/2013] [Accepted: 11/18/2013] [Indexed: 11/29/2022] Open
Abstract
HIV/AIDS is an important public health problem globally. An affordable, easy-to-deliver and protective HIV
vaccine is therefore required to curb the pandemic from spreading further. Recombinant Salmonella bacteria can be
harnessed to vector HIV antigens or DNA vaccines to the immune system for induction of specific protective immunity.
These are capable of activating the innate, humoral and cellular immune responses at both mucosal and systemic
compartments. Several studies have already demonstrated the utility of live recombinant Salmonella in delivering
expressed foreign antigens as well as DNA vaccines to the host immune system. This review gives an overview of the
studies in which recombinant Salmonella bacteria were used to vector HIV/AIDS antigens and DNA vaccines. Most of
the recombinant Salmonella-based HIV/AIDS vaccines developed so far have only been tested in animals (mainly mice)
and are yet to reach human trials.
Collapse
Affiliation(s)
- Nyasha Chin'ombe
- Department of Medical Microbiology, University of Zimbabwe, Harare, Zimbabwe ; Division of Medical Virology, University of Cape Town, Cape Town, South Africa
| | - Vurayai Ruhanya
- Department of Medical Microbiology, University of Zimbabwe, Harare, Zimbabwe
| |
Collapse
|
7
|
Ouédraogo A, Tiono AB, Kargougou D, Yaro JB, Ouédraogo E, Kaboré Y, Kangoye D, Bougouma EC, Gansane A, Henri N, Diarra A, Sanon S, Soulama I, Konate AT, Watson NL, Brown V, Hendriks J, Pau MG, Versteege I, Wiesken E, Sadoff J, Nebie I, Sirima SB. A phase 1b randomized, controlled, double-blinded dosage-escalation trial to evaluate the safety, reactogenicity and immunogenicity of an adenovirus type 35 based circumsporozoite malaria vaccine in Burkinabe healthy adults 18 to 45 years of age. PLoS One 2013; 8:e78679. [PMID: 24244339 PMCID: PMC3823848 DOI: 10.1371/journal.pone.0078679] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/11/2013] [Indexed: 11/28/2022] Open
Abstract
Background Ad35.CS.01 is a pre-erythrocytic malaria candidate vaccine. It is a codon optimized nucleotide sequence representing the P. falciparum circumsporozoite (CS) surface antigen inserted in a replication deficient Adenovirus 35 backbone. A Phase 1a trial has been conducted in the USA in naïve adults and showed that the vaccine was safe. The aim of this study is to assess the safety and immunogenicity of ascending dosages in sub Saharan Africa. Methods A double blind, randomized, controlled, dose escalation, phase Ib trial was conducted in a rural area of Balonghin, the Saponé health district (Burkina Faso). Forty-eight healthy adults aged 18-45 years were randomized into 4 cohorts of 12 to receive three vaccine doses (day 0, 28 and 84) of 109, 1010, 5X1010, 1011 vp of Ad35.CS.01 or normal saline by intra muscular injection. Subjects were monitored carefully during the 14 days following each vaccination for non serious adverse events. Severe and serious adverse events were collected throughout the participant study duration (12 months from the first vaccination). Humoral and cellular immune responses were measured on study days 0, 28, 56, 84, 112 and 140. Results Of the forty-eight subjects enrolled, forty-four (91.7%) received all three scheduled vaccine doses. Local reactions, all of mild severity, occurred in thirteen (27.1%) subjects. Severe (grade 3) laboratory abnormalities occurred in five (10.4%) subjects. One serious adverse event was reported and attributed to infection judged unrelated to vaccine. The vaccine induced both antibody titers and CD8 T cells producing IFNγ and TNFα with specificity to CS while eliciting modest neutralizing antibody responses against Ad35. Conclusion Study vaccine Ad35.CS.01 at four different dose levels was well-tolerated and modestly immunogenic in this population. These results suggest that Ad35.CS.01 should be further investigated for preliminary efficacy in human challenge models and as part of heterologous prime-boost vaccination strategies. Trial Registration ClinicalTrials.gov NCT01018459 http://clinicaltrials.gov/ct2/show/NCT01018459
Collapse
Affiliation(s)
- Alphonse Ouédraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Alfred B. Tiono
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Désiré Kargougou
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Jean Baptiste Yaro
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Esperance Ouédraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Youssouf Kaboré
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - David Kangoye
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Edith C. Bougouma
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Adama Gansane
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Noelie Henri
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Amidou Diarra
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Souleymane Sanon
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Issiaka Soulama
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Amadou T. Konate
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Nora L. Watson
- The EMMES Corporation, Rockville, Maryland, United States of America
| | - Valerie Brown
- The EMMES Corporation, Rockville, Maryland, United States of America
| | | | | | | | | | | | - Issa Nebie
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Sodiomon B. Sirima
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
- Groupe d’action et de Recherche en Santé, Ouagadougou, Burkina Faso
- * E-mail:
| |
Collapse
|
8
|
Remakus S, Sigal LJ. Memory CD8+ T Cell Protection. CROSSROADS BETWEEN INNATE AND ADAPTIVE IMMUNITY IV 2013; 785:77-86. [DOI: 10.1007/978-1-4614-6217-0_9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
9
|
Balasubramaniam D, Arockiasamy A, Kumar PD, Sharma A, Krishnaswamy S. Asymmetric pore occupancy in crystal structure of OmpF porin from Salmonella typhi. J Struct Biol 2012; 178:233-44. [PMID: 22525817 DOI: 10.1016/j.jsb.2012.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 04/03/2012] [Accepted: 04/07/2012] [Indexed: 11/25/2022]
Abstract
OmpF is a major general diffusion porin of Salmonella typhi, a Gram-negative bacterium, which is an obligatory human pathogen causing typhoid. The structure of S. typhi Ty21a OmpF (PDB Id: 3NSG) determined at 2.8 Å resolution by X-ray crystallography shows a 16-stranded β-barrel with three β-barrel monomers associated to form a trimer. The packing observed in S. typhi Ty21a rfOmpF crystals has not been observed earlier in other porin structures. The variations seen in the loop regions provide a starting point for using the S. typhi OmpF for structure-based multi-valent vaccine design. Along one side of the S. typhi Ty21a OmpF pore there exists a staircase arrangement of basic residues (20R, 60R, 62K, 65R, 77R, 130R and 16K), which also contribute, to the electrostatic potential in the pore. This structure suggests the presence of asymmetric electrostatics in the porin oligomer. Moreover, antibiotic translocation, permeability and reduced uptake in the case of mutants can be understood based on the structure paving the way for designing new antibiotics.
Collapse
Affiliation(s)
- D Balasubramaniam
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625 021, India
| | | | | | | | | |
Collapse
|
10
|
Mafamane H, Szabó I, Schmidt MFG, Filter M, Walk N, Tedin K, Scharek-Tedin L. Studies on the effect of an Enterococcus faecium probiotic on T cell populations in peripheral blood and intestinal epithelium and on the susceptibility to Salmonella during a challenge infection with Salmonella Typhimurium in piglets. Arch Anim Nutr 2012; 65:415-30. [PMID: 22256673 DOI: 10.1080/1745039x.2011.623351] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Although Enterococcus faecium is used as a probiotic feed supplement in animal production, feeding of the bacterium to piglets resulted in a more severe infection with Salmonella Typhimurium DT104 during a challenge experiment. To enlighten the mode of action by which E. faecium affected the piglets' health, we investigated the influence of the probiotic bacterium on the development of intestinal and circulating immune cells during a challenge experiment with S. Typhimurium DT104. To minimise varying impacts of the maternal immunity on the course of infection, only piglets were implemented that descended from Salmonella-free sows. In addition, the potency of purified blood and intraepithelial immune cells to control the growth of Salmonella was tested in vitro. In animals treated with E. faecium, a reduction of intraepithelial CD8alphabeta T cells, reduced circulating CD8alphabeta T cells and a less efficient control of intracellular Salmonella growth, mediated by peripheral blood mononuclear cells, were observed.
Collapse
Affiliation(s)
- Hassan Mafamane
- Institute of Immunology and Molecular Biology, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
11
|
Weiss WR, Jiang CG. Protective CD8+ T lymphocytes in primates immunized with malaria sporozoites. PLoS One 2012; 7:e31247. [PMID: 22355349 PMCID: PMC3280278 DOI: 10.1371/journal.pone.0031247] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 01/05/2012] [Indexed: 11/26/2022] Open
Abstract
Live attenuated malaria vaccines are more potent than the recombinant protein, bacterial or viral platform vaccines that have been tested, and an attenuated sporozoite vaccine against falciparum malaria is being developed for humans. In mice, attenuated malaria sporozoite vaccines induce CD8+ T cells that kill parasites developing in the liver. We were curious to know if CD8+ T cells were also important in protecting primates against malaria. We immunized 9 rhesus monkeys with radiation attenuated Plasmodium knowlesi sporozoites, and found that 5 did not develop blood stage infections after challenge with live sporozoites. We then injected 4 of these protected monkeys with cM-T807, a monoclonal antibody to the CD8 molecule which depletes T cells. The fifth monkey received equivalent doses of normal IgG. In 3 of the 4 monkeys receiving cM-T807 circulating CD8+ T cells were profoundly depleted. When re-challenged with live sporozoites all 3 of these depleted animals developed blood stage malaria. The fourth monkey receiving cM-T807 retained many circulating CD8+ T cells. This monkey, and the vaccinated monkey receiving normal IgG, did not develop blood stage malaria at re-challenge with live sporozoites. Animals were treated with antimalarial drugs and rested for 4 months. During this interval CD8+ T cells re-appeared in the circulation of the depleted monkeys. When all vaccinated animals received a third challenge with live sporozoites, all 5 monkeys were once again protected and did not develop blood stage malaria infections. These data indicate that CD8+ T cells are important effector cells protecting monkeys against malaria sporozoite infection. We believe that malaria vaccines which induce effector CD8+ T cells in humans will have the best chance of protecting against malaria.
Collapse
Affiliation(s)
- Walter R Weiss
- Infectious Disease Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America.
| | | |
Collapse
|
12
|
Baban CK, Cronin M, O'Hanlon D, O'Sullivan GC, Tangney M. Bacteria as vectors for gene therapy of cancer. Bioeng Bugs 2011; 1:385-94. [PMID: 21468205 DOI: 10.4161/bbug.1.6.13146] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 07/23/2010] [Accepted: 07/26/2010] [Indexed: 12/13/2022] Open
Abstract
Anti-cancer therapy faces major challenges, particularly in terms of specificity of treatment. The ideal therapy would eradicate tumor cells selectively with minimum side effects on normal tissue. Gene or cell therapies have emerged as realistic prospects for the treatment of cancer, and involve the delivery of genetic information to a tumor to facilitate the production of therapeutic proteins. However, there is still much to be done before an efficient and safe gene medicine is achieved, primarily developing the means of targeting genes to tumors safely and efficiently. An emerging family of vectors involves bacteria of various genera. It has been shown that bacteria are naturally capable of homing to tumors when systemically administered resulting in high levels of replication locally. Furthermore, invasive species can deliver heterologous genes intra-cellularly for tumor cell expression. Here, we review the use of bacteria as vehicles for gene therapy of cancer, detailing the mechanisms of action and successes at preclinical and clinical levels.
Collapse
Affiliation(s)
- Chwanrow K Baban
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jr. Laboratory, University College Cork, Cork, Ireland
| | | | | | | | | |
Collapse
|
13
|
Ahmad S, Casey G, Cronin M, Rajendran S, Sweeney P, Tangney M, O'Sullivan GC. Induction of effective antitumor response after mucosal bacterial vector mediated DNA vaccination with endogenous prostate cancer specific antigen. J Urol 2011; 186:687-93. [PMID: 21683415 DOI: 10.1016/j.juro.2011.03.139] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Indexed: 11/15/2022]
Abstract
PURPOSE The induction of systemic immune responses against antigenic targets that are over expressed by cancer cells represents a powerful therapeutic strategy to target metastatic cancer. We generated specific antitumor immune responses in a murine model of prostate cancer by oral administration of an attenuated strain of Salmonella typhimurium containing a plasmid coding for murine prostate stem cell antigen. MATERIALS AND METHODS Trafficking of S. typhimurium SL7207 in the initial 10 hours after gavage feeding was determined using a bacterial lux expressing strain and live bioluminescence imaging. For vaccination trials male C57 BL/6 mice were gavage fed SL7207/murine prostate stem cell antigen expressing plasmid or controls twice at 2-week intervals. One week after the last feeding the mice were challenged subcutaneously with TRAMPC1 murine prostate carcinoma cells. Tumor dynamics and animal survival were recorded. RESULTS Clearance of bacterial vector from animals was complete 9 hours after feeding. Delivery of vector transformed with a firefly luciferase reporter plasmid resulted in maximal eukaryotic reporter gene expression in splenocytes 48 hours after feeding. Induction of tumor protective immunity was achieved by feeding the mice murine prostate stem cell antigen expressing plasmid bearing bacteria and greater than 50% of immunized mice remained tumor free. No significant toxicity was observed. Induction of T-helper type 1 immune responses was determined by measuring interferon-γ produced by splenocytes from vaccinated mice. When adoptively transferred to naive animals, splenocytes from vaccinated mice prevented tumor growth in 66% of challenged animals. CONCLUSIONS Endogenous prostate cancer antigen gene delivery using a bacterial vector resulted in breaking immune tolerance to murine prostate stem cell antigen and significant retardation of tumor growth.
Collapse
Affiliation(s)
- Sarfraz Ahmad
- Leslie C. Quick Jr. Laboratory, Cork Cancer Research Centre, and Department of Surgery, Mercy University Hospital, University College Cork, Cork, Ireland
| | | | | | | | | | | | | |
Collapse
|
14
|
Ashraf S, Kong W, Wang S, Yang J, Curtiss R. Protective cellular responses elicited by vaccination with influenza nucleoprotein delivered by a live recombinant attenuated Salmonella vaccine. Vaccine 2011; 29:3990-4002. [PMID: 21466806 DOI: 10.1016/j.vaccine.2011.03.066] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 03/11/2011] [Accepted: 03/20/2011] [Indexed: 12/27/2022]
Abstract
Orally administered recombinant attenuated Salmonella vaccines (RASVs) elicit humoral and mucosal immune responses against the immunizing antigen. The challenge in developing an effective vaccine against a virus or an intracellular bacterium delivered by RASVs is to introduce the protective antigen inside the host cell cytoplasm for presentation to MHC-I molecules for an efficient cell mediated immune response. To target the influenza nucleoprotein (NP) into the host cell cytosol, we constructed a regulated delayed lysis in vivo RASV strain χ11246(pYA4858) encoding influenza NP with a chromosomal deletion of the sifA gene to enable it to escape from the endosome prior to lysis. Oral immunization of mice with χ11246(pYA4858) (SifA⁻) with 3 booster immunizations resulted in complete protection (100%) against a lethal influenza virus (rWSN) challenge (100 LD₅₀) compared to 25% survival of mice immunized with the isogenic χ11017(pYA4858) (SifA⁺) strain. Reducing the number of booster immunizations with χ11246(pYA4858) from 3 to 2 resulted in 66% survival of mice challenged with rWSN (100 LD₅₀). Immunization with χ11246(pYA4858) via different routes provided protection in 80% orally, 100% intranasally and 100% intraperitoneally immunized mice against rWSN (100 LD₅₀). A Th1 type immune response was elicited against influenza NP in all experiments. IFN-γ secreting NP₁₄₇₋₁₅₅ specific T cells were not found to be correlated with protection. The role of antigen-specific CD8⁺ T cells remains to be determined. To conclude, we showed that Salmonella can be designed to deliver antigen(s) to the host cell cytosol for presumably class I presentation for the induction of protective immune responses.
Collapse
Affiliation(s)
- Shamaila Ashraf
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, AZ 85287-5401, USA
| | | | | | | | | |
Collapse
|
15
|
Braga CJM, Massis LM, Sbrogio-Almeida ME, Alencar BCG, Bargieri DY, Boscardin SB, Rodrigues MM, Ferreira LCS. CD8+ T cell adjuvant effects of Salmonella FliCd flagellin in live vaccine vectors or as purified protein. Vaccine 2009; 28:1373-82. [PMID: 19932669 DOI: 10.1016/j.vaccine.2009.11.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Revised: 11/01/2009] [Accepted: 11/03/2009] [Indexed: 12/21/2022]
Abstract
Salmonella flagellin, the flagellum structural subunit, has received particular interest as a vaccine adjuvant conferring enhanced immunogenity to soluble proteins or peptides, both for activation of antibody and cellular immune responses. In the present study, we evaluated the Salmonella enterica FliCd flagellin as a T cell vaccine adjuvant using as model the 9-mer (SYVPSAEQI) synthetic H2(d)-restricted CD8(+) T cell-specific epitope (CS(280-288)) derived from the Plasmodium yoelii circumsporozoite (CS) protein. The FliCd adjuvant effects were determined under two different conditions: (i) as recombinant flagella, expressed by orally delivered live S. Dublin vaccine strains expressing the target CS(280-288) peptide fused at the central hypervariable domain, and (ii) as purified protein in acellular vaccines in which flagellin was administered to mice either as a recombinant protein fused or admixed with the target CS(280-288) peptide. The results showed that CS(280-288)-specific cytotoxic CD8(+) T cells were primed when BALB/c mice were orally inoculated with the expressing the CS(280-288) epitope S. Dublin vaccine strain. In contrast, mice immunized with purified FliCd admixed with the CS(280-288) peptide and, to a lesser extent, fused with the target peptide developed specific cytotoxic CD8(+) T cell responses without the need of a heterologous booster immunization. The CD8(+) T cell adjuvant effects of flagellin, either fused or not with the target peptide, correlated with the in vivo activation of CD11c(+) dendritic cells. Taken together, the present results demonstrate that Salmonella flagellins are flexible adjuvant and induce adaptative immune responses when administered by different routes or vaccine formulations.
Collapse
Affiliation(s)
- Catarina J M Braga
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP 05008-000, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
16
|
An enteric pathogen Salmonella enterica serovar Typhimurium suppresses tumor growth by downregulating CD44high and CD4T regulatory (Treg) cell expression in mice: the critical role of lipopolysaccharide and Braun lipoprotein in modulating tumor growth. Cancer Gene Ther 2009; 17:97-108. [PMID: 19713997 PMCID: PMC2808459 DOI: 10.1038/cgt.2009.58] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
An antitumor activity associated with several bacterial pathogens, including Salmonella enterica serovar Typhimurium, has been reported; however, the underlying immunological mechanism(s) that lead to an antitumor effect are currently unclear. Furthermore, such pathogens cannot be used to suppress tumor growth because of their potential for causing sepsis. Recently, we reported the characterization of S. Typhimurium isogenic mutants from which Braun lipoprotein genes (lppA and B) and the multicopy repressor of high temperature requirement (msbB) gene were deleted. In a mouse infection model, two mutants, namely, lppB/msbB and lppAB/msbB, minimally induced proinflammatory cytokine production at high doses and were nonlethal to animals. We showed that immunization of mice with these mutants, followed by challenge with the wild-type S. Typhimurium, could significantly suppress tumor growth, as evidenced by an 88% regression in tumor size in lppB/msbB mutant-immunized animals over a 24-day period. However, the lppAB/msbB mutant alone was not effective in modulating tumor growth in mice, although the lppB/msbB mutant alone caused marginal regression in tumor size. Importantly, we showed that CD44(+) cells grew much faster than CD44(-) cells from human liver tumors in mice, leading us to examine the possibility that S. Typhimurium might downregulate CD44 in tumors and splenocytes of mice. Consequently, we found in S. Typhimurium-infected mice that tumor size regression could indeed be related to the downregulation of CD44(high) and CD4(+)CD25(+) T(reg) cells. Importantly, the role of lipopolysaccharide and Braun lipoprotein was critical in S. Typhimurium-induced antitumor immune responses. Taken together, we have defined new immune mechanisms leading to tumor suppression in mice by S. Typhimurium.
Collapse
|
17
|
Overstreet MG, Cockburn IA, Chen YC, Zavala F. Protective CD8 T cells against Plasmodium liver stages: immunobiology of an 'unnatural' immune response. Immunol Rev 2009; 225:272-83. [PMID: 18837788 DOI: 10.1111/j.1600-065x.2008.00671.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
SUMMARY Immunization with high doses of irradiated sporozoites delivered by the bites of infected mosquitoes has been shown to induce protective responses against malaria, mediated in part by CD8(+) T cells. In contrast, natural transmission involving low exposure to live sporozoite antigen fails to elicit strong immunity. In this review, we examine how irradiated sporozoite immunization breaks the natural host-parasite interaction and induces protective CD8(+) T cells. Upon biting, the malaria-infected mosquitoes deposit parasites in the skin, many of which eventually exit to the bloodstream and infect hepatocytes. However, certain antigens, including the circumsporozoite (CS) protein, remain in the skin and are presented in the draining lymph node. These antigens prime specific CD8(+) T cells, which migrate to the liver where they eliminate parasitized hepatocytes. We discuss the relevance of the different tissue compartments involved in the induction and effector phases of this response, as well as the cellular requirements for priming and memory development of CD8(+) T cells, which include a complete dependence on dendritic cells and a near absolute need for CD4(+) T-cell help. Finally, we discuss the impact of the immunodominant CS protein on this protection and the implications of these findings for vaccine design.
Collapse
Affiliation(s)
- Michael Glen Overstreet
- Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
18
|
Abstract
Over the last decade, significant advances have been made in the methodology for studying immune responses in vivo. It is now possible to follow almost every aspect of pathogen-specific immunity using in vivo models that incorporate physiological infectious doses and natural routes of infection. This new ability to study immunity in a relevant physiological context will greatly expand our understanding of the dynamic interplay between host and pathogen. Visualizing the resolution of primary infection and the development of long-term immunological memory should also aid the development of new vaccines and therapeutics for infectious diseases. In this review, we will describe the application of in vivo visualization technology to Salmonella infection, describe our current understanding of Salmonella-specific immunity, and discuss some unanswered questions that remain in this model.
Collapse
Affiliation(s)
- James J. Moon
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, MN, 55455
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455
| | - Stephen J. McSorley
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, 55455
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455
- Center for Infectious Diseases & Microbiology Translational Research, University of Minnesota Medical School, Minneapolis, MN, 55455
| |
Collapse
|
19
|
Tartz S, Rüssmann H, Kamanova J, Sebo P, Sturm A, Heussler V, Fleischer B, Jacobs T. Complete protection against P. berghei malaria upon heterologous prime/boost immunization against circumsporozoite protein employing Salmonella type III secretion system and Bordetella adenylate cyclase toxoid. Vaccine 2008; 26:5935-43. [PMID: 18804138 DOI: 10.1016/j.vaccine.2008.08.057] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2008] [Revised: 08/12/2008] [Accepted: 08/31/2008] [Indexed: 01/13/2023]
Abstract
Sterile immunity against malaria can be achieved by the induction of IFNgamma-producing CD8(+) T cells that target infected hepatocytes presenting epitopes of the circumsporozoite protein (CSP). In the present study we evaluate the protective efficacy of a heterologous prime/boost immunization protocol based on the delivery of the CD8(+) epitope of Plasmodium berghei CSP into the MHC class I presentation pathway, by either a type III secretion system of live recombinant Salmonella and/or by direct translocation of a recombinant Bordetella adenylate cyclase toxoid fusion (ACT-CSP) into the cytosol of professional antigen-presenting cells (APCs). A single intraperitoneal application of the recombinant ACT-CSP toxoid, as well as a single oral immunization with the Salmonella vaccine, induced a specific CD8(+) T cell response, which however conferred only a partial protection on mice against a subsequent sporozoite challenge. In contrast, a heterologous prime/boost vaccination with the live Salmonella followed by ACT-CSP led to a significant enhancement of the CSP-specific T cell response and induced complete protection in all vaccinated mice.
Collapse
Affiliation(s)
- Susanne Tartz
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Wang L, Coppel RL. Oral vaccine delivery: can it protect against non-mucosal pathogens? Expert Rev Vaccines 2008; 7:729-38. [PMID: 18665772 DOI: 10.1586/14760584.7.6.729] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Vaccination is an efficient and cost-effective form of preventing infectious diseases. However, most currently available vaccines are delivered by injection, which makes mass immunization more costly and less safe, particularly in resource-poor developing countries. Oral vaccines have several attractive features compared with parenteral vaccines, but studies on their use have been limited almost exclusively to protection against mucosally transmitted pathogens. Their potential for controlling non-mucosally transmitted diseases has not yet been appreciated in general. In this article, we provide evidence that oral immunization is a feasible alternative for preventing infections transmitted through non-mucosal routes, including infections such as malaria, Japanese encephalitis and hepatitis B. Although there are still hurdles to overcome before such approaches can be deployed widely, recent progress in the oral vaccination field and the availability of a range of delivery systems offers hope for the development of a larger number of oral vaccines.
Collapse
Affiliation(s)
- Lina Wang
- Department of Microbiology, Monash University, Clayton, Victoira 3800, Australia.
| | | |
Collapse
|
21
|
Donnelly JJ, Ulmer JB, Liu MA. Overview: Biologicals & Immunologicals: Recombinant vaccines: technology and applications. Expert Opin Ther Pat 2008. [DOI: 10.1517/13543776.5.3.211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
22
|
Sedegah M, Weiss WW, Hoffman SL. Cross-protection between attenuated Plasmodium berghei and P. yoelii sporozoites. Parasite Immunol 2008; 29:559-65. [PMID: 17944745 PMCID: PMC2955969 DOI: 10.1111/j.1365-3024.2007.00976.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An attenuatedPlasmodium falciparum sporozoite (PfSPZ) vaccine is under development, in part, based on studies in mice withP. berghei. We usedP. berghei andP. yoelii to study vaccine-induced protection against challenge with a species of parasite different from the immunizing parasite in BALB/c mice. One-hundred percent of mice were protected against homologous challenge. Seventy-nine percent immunized with attenuatedP. berghei sporozoite (PbSPZ)(six experiments) were protected against challenge withP. yoelii sporozoite (PySPZ), and 63% immunized with attenuatedPySPZ(three experiments) were protected against challenge withPbSPZ. Antibodies in sera of immunized mice only recognized homologous sporozoites and could not have mediated protection against heterologous challenge. Immunization with attenuatedPySPZ orPbSPZ induced CD8+ T cell-dependent protection against heterologous challenge. Immunization with attenuatedPySPZ induced CD8+ T cell-dependent protection against homologous challenge. However, homologous protection induced by attenuatedPbSPZ was not dependent on CD8+ or CD4+ T cells, and depletion of both populations only reduced protection by 36%. Immunization of C57BL/10 mice withPbSPZ induced CD8+ T cell-dependent protection againstP. berghei, but no protection againstP. yoelii. The cross-protection data in BALB/c mice support testing a human vaccine based on attenuatedPfSPZ for its efficacy againstP. vivax.
Collapse
Affiliation(s)
- M Sedegah
- Malaria Program, Naval Medical Research Center, Silver Spring, MD, USA
| | | | | |
Collapse
|
23
|
Chinchilla M, Pasetti MF, Medina-Moreno S, Wang JY, Gomez-Duarte OG, Stout R, Levine MM, Galen JE. Enhanced immunity to Plasmodium falciparum circumsporozoite protein (PfCSP) by using Salmonella enterica serovar Typhi expressing PfCSP and a PfCSP-encoding DNA vaccine in a heterologous prime-boost strategy. Infect Immun 2007; 75:3769-79. [PMID: 17502396 PMCID: PMC1951980 DOI: 10.1128/iai.00356-07] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Two Salmonella enterica serovar Typhi strains that express and export a truncated version of Plasmodium falciparum circumsporozoite surface protein (tCSP) fused to Salmonella serovar Typhi cytolysin A (ClyA) were constructed as a first step in the development of a preerythrocytic malaria vaccine. Synthetic codon-optimized genes (t-csp1 and t-csp2), containing immunodominant B- and T-cell epitopes present in native P. falciparum circumsporozoite surface protein (PfCSP), were fused in frame to the carboxyl terminus of the ClyA gene (clyA::t-csp) in genetically stabilized expression plasmids. Expression and export of ClyA-tCSP1 and ClyA-tCSP2 by Salmonella serovar Typhi vaccine strain CVD 908-htrA were demonstrated by immunoblotting of whole-cell lysates and culture supernatants. The immunogenicity of these constructs was evaluated using a "heterologous prime-boost" approach consisting of mucosal priming with Salmonella serovar Typhi expressing ClyA-tCSP1 and ClyA-tCSP2, followed by parenteral boosting with PfCSP DNA vaccines pVR2510 and pVR2571. Mice primed intranasally on days 0 and 28 with CVD 908-htrA(pSEC10tcsp2) and boosted intradermally on day 56 with PfCSP DNA vaccine pVR2571 induced high titers of serum NANP immunoglobulin G (IgG) (predominantly IgG2a); no serological responses to DNA vaccination were observed in the absence of Salmonella serovar Typhi-PfCSP priming. Mice primed with Salmonella serovar Typhi expressing tCSP2 and boosted with PfCSP DNA also developed high frequencies of gamma interferon-secreting cells, which surpassed those produced by PfCSP DNA in the absence of priming. A prime-boost regimen consisting of mucosal delivery of PfCSP exported from a Salmonella-based live-vector vaccine followed by a parenteral PfCSP DNA boosting is a promising strategy for the development of a live-vector-based malaria vaccine.
Collapse
Affiliation(s)
- Magaly Chinchilla
- Center for Vaccine Development, University of Maryland, 685 W. Baltimore Street, HSF I, Room 480, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Moorthy G, Ramasamy R. Mucosal immunisation of mice with malaria protein on lactic acid bacterial cell walls. Vaccine 2007; 25:3636-45. [PMID: 17280749 DOI: 10.1016/j.vaccine.2007.01.070] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Revised: 11/02/2006] [Accepted: 01/11/2007] [Indexed: 12/28/2022]
Abstract
Mice were orally and nasally immunised with a malaria parasite protein, MSA2, expressed in Lactococcus lactis, covalently attached to the peptidoglycan (MSA2cP), or non-covalently rebound to L. lactis cell walls, live Lactobacillus reuteri and Lb. salivarius (MSA2cA). Although there was marked variation within mice of the same inbred strain, the data suggest that the characteristics of serum IgG antibodies formed against MSA2 are influenced by the mouse strain and mode of MSA2 presentation. MSA2-specific IgA antibodies in sera and faecal pellets, and IFNgamma-secreting spleen cells were detectable in some immunised animals. MSA2cA on lactobacilli, although not effective in eliciting serum IgG antibodies, showed a potential for eliciting IgA antibodies in the gut. The presence of MSA2 in L. lactis tended to divert the antibody response from lactococcal antigens. The data suggest that anti-MSA2 and anti-lactococcal antibody responses are not necessarily associated in the mice. The findings are related to the use of lactic acid bacteria expressing heterologous proteins for therapy and vaccination.
Collapse
Affiliation(s)
- G Moorthy
- National Science Foundation, Maitland Place, Colombo, Sri Lanka; Institute of Fundamental Studies, Kandy, Sri Lanka
| | | |
Collapse
|
25
|
Hafalla JCR, Cockburn IA, Zavala F. Protective and pathogenic roles of CD8+ T cells during malaria infection. Parasite Immunol 2006; 28:15-24. [PMID: 16438672 DOI: 10.1111/j.1365-3024.2006.00777.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
CD8+ T cells play a key role in protection against pre-erythrocytic stages of malaria infection. Many vaccine strategies are based on the idea of inducing a strong infection-blocking CD8+ T cell response. Here, we summarize what is known about the development, specificity and protective effect of malaria-specific CD8+ T cells and report on recent developments in the field. Although work in mouse models continues to make progress in our understanding of the basic biology of these cells, many questions remain to be answered - particularly on the roles of these cells in human infections. Increasing evidence is also emerging of a harmful role for CD8+ T cells in the pathology of cerebral malaria in rodent systems. Once again, the relevance of these results to human disease is one of the primary questions facing workers in this field.
Collapse
Affiliation(s)
- J C R Hafalla
- Department of Medical Parasitology, New York University School of Medicine, New York, NY, USA
| | | | | |
Collapse
|
26
|
Wang L, Webster DE, Wesselingh SL, Coppel RL. Orally delivered malaria vaccines: not too hard to swallow. Expert Opin Biol Ther 2005; 4:1585-94. [PMID: 15461570 DOI: 10.1517/14712598.4.10.1585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Vaccines offer efficient and cost-effective protection against a wide range of infectious diseases. Unfortunately, no effective vaccine is yet available against malaria, and this infection remains one of the most important causes of human morbidity and mortality in the developing world. Over the past two decades a number of candidate proteins for inclusion in a subunit vaccine have been identified. Malariologists believe that an effective malaria vaccine will need to include multiple proteins that induce protective immune responses against different stages of the Plasmodium life cycle. The construction of such multivalent vaccines is beset by considerable logistical difficulties, not least of which is how to deliver them to a population living in endemic areas. Compared with other routes of vaccine administration, oral delivery has several advantages that make it an attractive strategy for vaccine development. This review summarises the progress towards an oral vaccine delivery system for malaria and discusses the feasibility of this approach.
Collapse
Affiliation(s)
- Lina Wang
- Monash University, Department of Microbiology, Clayton, Victoria 3800, Australia.
| | | | | | | |
Collapse
|
27
|
Abstract
Transgenic (Tg) mice carrying a T-cell receptor (TCR) specific for a CD8(+) T-cell epitope expressed in pre-erythrocytic stages of Plasmodium yoelii has proven to be a valuable tool to advance our understanding of this anti-parasite T-cell response, as it occurs in vivo. The visualization of CD8(+) T cells in vivo and ex vivo greatly facilitated research aimed at characterizing basic features of this T-cell response such as the kinetics of differentiation and proliferation and the in vivo antigen presentation. Importantly, this research unveiled the existence of early self-regulatory mechanisms controlling the magnitude of the CD8(+) T-cell response and also identified CD4(+) T cells as critical elements in the development of memory populations. This review discusses our recent research using Tg mice and highlights our progress in understanding the CD8(+) T-cell-mediated immunity against malaria liver stages.
Collapse
Affiliation(s)
- Alexandre Morrot
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
28
|
Ringrose JH, Meiring HD, Speijer D, Feltkamp TEW, van Els CACM, de Jong APJM, Dankert J. Major histocompatibility complex class I peptide presentation after Salmonella enterica serovar typhimurium infection assessed via stable isotope tagging of the B27-presented peptide repertoire. Infect Immun 2004; 72:5097-105. [PMID: 15322003 PMCID: PMC517416 DOI: 10.1128/iai.72.9.5097-5105.2004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Reactive arthritis (ReA) induced by infection with several gram-negative bacteria is strongly associated with expression of the major histocompatibility complex class I molecule HLA-B27. It is thought that due to the intracellular lifestyle of ReA-inducing bacteria, bacterial fragments can be presented by HLA-B27. Cytotoxic T cells recognizing such bacterial peptides or other induced host peptides could cross-react with self peptides presented in the joints, giving rise to disease. Studies to analyze the B27 peptide repertoire in relation to infection were severely hampered, as complex peptide profiles obtained from separate infected and noninfected cell preparations had to be compared. For this study, we applied a new approach to examine the effect of Salmonella enterica serovar Typhimurium infection on the B27 peptide repertoire presented by the HLA-B*2704 subtype associated with disease. Firstly, we showed that both host cell and S. enterica serovar Typhimurium proteins can be tagged metabolically with stable-isotope-labeled arginine. We then designed experiments so that either the tagged endogenous or tagged bacterial B*2704-presented peptide repertoires from infected cells could be analyzed by mass spectrometry from single peptide preparations that included uninfected controls. Using this new approach, we found no evidence for significant changes in endogenous B*2704 peptide presentation after infection or for any S. enterica serovar Typhimurium-derived B27-bound peptide. In conclusion, the hypothesis that S. enterica serovar Typhimurium induces changes in B27 peptide presentation could not be supported.
Collapse
Affiliation(s)
- Jeffrey H Ringrose
- Department of Medical Microbiology, Academic Medical Centre, University of Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
29
|
Affiliation(s)
- A K Sahni
- Associate Professor, Department of Microbiology, Armed Forces Medical College, Pune - 411 040
| | - A Nagendra
- Professor and Head, Department of Microbiology, Armed Forces Medical College, Pune - 411 040
| |
Collapse
|
30
|
Chauhan VS, Bhardwaj D. Current status of malaria vaccine development. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2003; 84:143-82. [PMID: 12934936 DOI: 10.1007/3-540-36488-9_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
There is an urgent need to develop an effective vaccine against malaria--a disease that has approximately 10% of the world population at risk of infection at any given time. The economic burden this disease puts on the medico-social set-up of countries in Sub-Saharan Africa and South East Asia is phenomenal. Increasing drug resistance and failure of vector control strategies have necessitated the search for a suitable vaccine that could be integrated into the extended program of immunization for countries in the endemic regions. Malaria vaccine development has seen a surge of activity in the last decade or so owing largely to the advances made in the fields of genetic engineering and biotechnology. This revolution has brought sweeping changes in the understanding of the biology of the parasite and has helped formulate newer more effective strategies to combat the disease. Latest developments in the field of malaria vaccine development will be discussed in this chapter.
Collapse
Affiliation(s)
- Virander Singh Chauhan
- Malaria Research Group, International Center for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi-110067, India.
| | | |
Collapse
|
31
|
Abstract
CD8 T cells respond to viral infections but also participate in defense against bacterial and protozoal infections. In the last few years, as new methods to accurately quantify and characterize pathogen-specific CD8 T cells have become available, our understanding of in vivo T cell responses has increased dramatically. Pathogen-specific T cells, once thought to be quite rare following infection, are now known to be present at very high frequencies, particularly in peripheral, nonlymphoid tissues. With the ability to visualize in vivo CD8 T cell responses has come the recognition that T cell expansion is programmed and, to a great extent, independent of antigen concentrations. Comparison of CD8 T cell responses to different pathogens also highlights the intricate relationship between microbially induced innate inflammatory responses and the kinetics, magnitude, and character of long-term T cell responses. This review describes recent progress in some of the major murine models of CD8 T cell-mediated immunity to viral, bacterial, and protozoal infection.
Collapse
Affiliation(s)
- Phillip Wong
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| | | |
Collapse
|
32
|
Bagley KC, Shata MT, Onyabe DY, DeVico AL, Fouts TR, Lewis GK, Hone DM. Immunogenicity of DNA vaccines that direct the coincident expression of the 120 kDa glycoprotein of human immunodeficiency virus and the catalytic domain of cholera toxin. Vaccine 2003; 21:3335-41. [PMID: 12804865 DOI: 10.1016/s0264-410x(03)00038-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Passive antibody studies unequivocally demonstrate that sterilizing immunity against lentiviruses is obtainable through humoral mechanisms. In this regard, DNA vaccines represent an inexpensive alternative to subunit vaccine for mass vaccination programs designed to induce such responses to human immunodeficiency virus type I (HIV-1). At present, however, this vaccine modality has proven relatively ineffective at inducing humoral responses. In this report, we describe the immunogenicity of DNA vaccines that direct the coincident expression of the cholera toxin catalytic domain (CTA1) with that of the human immunodeficiency virus type I gp120 through genes either encoded in individual plasmids or in a single dicistronic plasmid. In BALB/cJ mice, coincident expression of CTA1 in either a separate plasmid or in the dicistronic plasmid in the DNA vaccines induced serum IgG responses to gp120 that were at least 1000-fold greater, and remained elevated longer than, the analogous responses in mice vaccinated with a DNA vaccine that expressed gp120 alone. In addition, mice vaccinated with CTA1 and gp120 produced significantly more gp120-specific IFN-gamma ELISPOTs than mice vaccinated with the gp120 DNA vaccine. Combined, these data show that the adjuvant properties of cholera toxin can be harnessed in DNA vaccine modalities.
Collapse
Affiliation(s)
- K C Bagley
- Division of Vaccine Research and Basic Science, Institute of Human Virology, University of Maryland Biotechnology Institute, 725 W. Lombard Street, Baltimore, MD, 212001, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Pasetti MF, Levine MM, Sztein MB. Animal models paving the way for clinical trials of attenuated Salmonella enterica serovar Typhi live oral vaccines and live vectors. Vaccine 2003; 21:401-18. [PMID: 12531639 DOI: 10.1016/s0264-410x(02)00472-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Attenuated Salmonella enterica serovar Typhi (S. Typhi) strains can serve as safe and effective oral vaccines to prevent typhoid fever and as live vectors to deliver foreign antigens to the immune system, either by the bacteria expressing antigens through prokaryotic expression plasmids or by delivering foreign genes carried on eukaryotic expression systems (DNA vaccines). The practical utility of such live vector vaccines relies on achieving a proper balance between minimizing the vaccine's reactogenicity and maximizing its immunogenicity. To advance to clinical trials, vaccine candidates need to be pre-clinically evaluated in relevant animal models that attempt to predict what their safety and immunogenicity profile will be when administered to humans. Since S. Typhi is a human-restricted pathogen, a major obstacle that has impeded the progress of vaccine development has been the shortcomings of the animal models available to assess vaccine candidates. In this review, we summarize the usefulness of animal models in the assessment of the degree of attenuation and immunogenicity of novel attenuated S. Typhi strains as vaccine candidates for the prevention of typhoid fever and as live vectors in humans.
Collapse
Affiliation(s)
- Marcela F Pasetti
- Center for Vaccine Development, University of Maryland School of Medicine, Room 480, 685 West Baltimore Street, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|
34
|
Pasetti MF, Salerno-Gonçalves R, Sztein MB. Salmonella enterica serovar Typhi live vector vaccines delivered intranasally elicit regional and systemic specific CD8+ major histocompatibility class I-restricted cytotoxic T lymphocytes. Infect Immun 2002; 70:4009-18. [PMID: 12117906 PMCID: PMC128131 DOI: 10.1128/iai.70.8.4009-4018.2002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated the ability of live attenuated Salmonella enterica serovar Typhi strains delivered to mice intranasally to induce specific cytotoxic T-lymphocyte (CTL) responses at regional and systemic levels. Mice immunized with two doses (28 days apart) of Salmonella serovar Typhi strain Ty21a, the licensed oral typhoid vaccine, and genetically attenuated mutants CVD 908 (DeltaaroC DeltaaroD), CVD 915 (DeltaguaBA), and CVD 908-htrA (DeltaaroC DeltaaroD DeltahtrA) induced CTL specific for Salmonella serovar Typhi-infected cells in spleens and cervical lymph nodes. CTL were detected in effector T cells that had been expanded in vitro for 7 days in the presence of Salmonella-infected syngeneic splenocytes. A second round of stimulation further enhanced the levels of specific cytotoxicity. CTL activity was observed in sorted alphabeta+ CD8+ T cells, which were remarkably increased after expansion, but not in CD4+ T cells. CTL from both cervical lymph nodes and spleens failed to recognize Salmonella-infected major histocompatibility complex (MHC)-mismatched cells, indicating that the responses were MHC restricted. Studies in which MHC blocking antibodies were used showed that H-2L(d) was the restriction element. This is the first demonstration that Salmonella serovar Typhi vaccines delivered intranasally elicit CD8+ MHC class I-restricted CTL. The results further support the usefulness of the murine intranasal model for evaluating the immunogenicity of typhoid vaccine candidates at the preclinical level.
Collapse
Affiliation(s)
- Marcela F Pasetti
- Center for Vaccine Development, Department of Pediatrics, University of Maryland School of Medicine, Baltimore 21201, USA.
| | | | | |
Collapse
|
35
|
Qian F, Pan W. Construction of a tetR-integrated Salmonella enterica serovar Typhi CVD908 strain that tightly controls expression of the major merozoite surface protein of Plasmodium falciparum for applications in human Vaccine production. Infect Immun 2002; 70:2029-38. [PMID: 11895968 PMCID: PMC127878 DOI: 10.1128/iai.70.4.2029-2038.2002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Attenuated Salmonella strains are an attractive live vector for delivery of a foreign antigen to the human immune system. However, the problem with this vector lies with plasmid segregation and the low level of expression of the foreign gene in vivo when constitutive expression is employed, leading to a diminished immune response. We have established inducible expressions of foreign genes in the Salmonella enterica serovar Typhi CVD908 vaccine strain using the tetracycline response regulatory promoter. To set up this system, a tetracycline repressor (tetR) was integrated into a defined Delta aroC locus of the chromosome via suicide plasmid pJG12/tetR-neo. To remove the neo gene conferring kanamycin resistance from the locus, a cre expression vector under the control of the tetracycline response promoter was transformed into the clone; expression of the Cre recombinase excised the neo gene and generated the end strain CVD908-tetR. Expression of the luciferase reporter gene in this strain is dependent on the presence of tetracycline in the medium and can be regulated up to 4,773-fold. Moreover, the tightly controlled expression of major merozoite surface protein 1 (MSP1) and parts of Plasmodium falciparum was achieved, and the product yield was increased when the inducible expression system was employed. Inoculation of bacteria harboring plasmid pZE11/MSP1(42) in mice produced the protein in liver and spleen controlled by the inducer. The persistence of the plasmid-carrying bacteria in mice was determined. Peak colonization of both liver and spleen was detected on the third day postinoculation and was followed by a decline in growth curves. After 14 days postinfection, the majority of the bacteria (>90%) recovered from the liver and spleen of the mice retained the plasmid when expression was induced; this clearly indicated that stability of the expression vector in vivo was improved by inducible expression. Establishment of the regulatory system in the vaccine strain may broaden the range of its use by enhancing plasmid stability and expression levels in vivo. Moreover, the availability of the vaccine strain inducibly expressing the entire MSP1 provides possibilities for examining its immunogenicity, particularly the cellular response in animal models.
Collapse
Affiliation(s)
- Feng Qian
- Department of Etiologic Biology, Second Military Medical University, Shanghai, China
| | | |
Collapse
|
36
|
Zheng C, Xie P, Chen Y. Recombinant Mycobacterium bovis BCG producing the circumsporozoite protein of Plasmodium falciparum FCC-1/HN strain induces strong immune responses in BALB/c mice. Parasitol Int 2002; 51:1-7. [PMID: 11880222 DOI: 10.1016/s1383-5769(01)00094-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The current vaccine against tuberculosis, Mycobacterium bovis strain bacillus Calmette-Guerin (BCG), offers potential advantages as a live, innately immunogenic vaccine vehicle for expression and delivery of protective recombinant antigens. Malaria is one of the severest parasitic diseases in humans especially in the developing world. No efficacious vaccine is currently available. However, circumsporozoite protein (CSP) is a malaria vaccine candidate currently undergoing clinical trials. We analyzed the immune response to recombinant BCG (rBCG) vaccine expressing Plasmodium falciparum CSP (BCG-CSP) under the control of heat shock protein 70 promoter in BALB/c mice. The lymphocytes proliferative response to P. falciparum soluble antigen was significantly higher than those in the groups of BCG and normal saline, and the production of cytokines (IFN-gamma and IL-2) in response to malaria antigen was significantly higher in rBCG and BCG groups than control group of normal saline. A specific IgG antibody response against P. falciparum antigen of CSP was also characterized. The booster injection could enhance the production of cytokine, proliferation responses of spleen lymphocytes and the antibodies titer of BCG-CSP. The results in the study demonstrated that rBCG vaccine producing CSP is an appropriate vaccine for further evaluation in non-human primates.
Collapse
Affiliation(s)
- Chunfu Zheng
- Institute of Infectious and Parasitic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China.
| | | | | |
Collapse
|
37
|
Müller-Schollenberger V, Beyer W, Schnitzler P, Merckelbach A, Roth S, Kalinna BH, Lucius R. Immunisation with Salmonella typhimurium-delivered glyceraldehyde-3-phosphate dehydrogenase protects mice against challenge infection with Echinococcus multilocularis eggs. Int J Parasitol 2001; 31:1441-9. [PMID: 11595231 DOI: 10.1016/s0020-7519(01)00244-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recombinant glyceraldehyde-3-phosphate dehydrogenase of the cestode parasite Echinococcus multilocularis was expressed in Escherichia coli and in Salmonella typhimurium. The potential of different forms of the recombinant antigen to protect BALB/c mice against oral challenge infections with E. multilocularis eggs was evaluated. Oral or intraperitoneal immunisation with live attenuated S. typhimurium as a carrier for recombinant glyceraldehyde-3-phosphate dehydrogenase of the E. multilocularis resulted in significant protection, reducing the number of developing metacestodes up to 79.8%. The sera of protected animals did not contain detectable amounts of antibody against glyceraldehyde-3-phosphate dehydrogenase of E. multilocularis. By contrast, although anti-glyceraldehyde-3-phosphate dehydrogenase of E. multilocularis antibodies were detectable in the sera, immunisation with E. coli-expressed recombinant glutathione-S-transferase-fusion protein or with glyceraldehyde-3-phosphate dehydrogenase of E. multilocularis fused to a 6HIS-tag failed to protect the animals against oral challenge infections. These data emphasise that antigen delivery systems play a critical role in vaccination and the induction of protective immunity against helminth parasites.
Collapse
Affiliation(s)
- V Müller-Schollenberger
- Division of Parasitology, Department of Zoology, University of Hohenheim, 70599 Stuttgart, Germany
| | | | | | | | | | | | | |
Collapse
|
38
|
Soloski MJ, Metcalf ES. The involvement of class Ib molecules in the host response to infection with Salmonella and its relevance to autoimmunity. Microbes Infect 2001; 3:1249-59. [PMID: 11755413 DOI: 10.1016/s1286-4579(01)01485-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Class I molecules with limited polymorphism have been implicated in the host response to infectious agents. Following infection with Salmonella typhimurium, mice develop a CD8+ CTL response that specifically recognizes bacteria infected cells. An immunodominant component of the CTL response recognizes a peptide epitope derived from the Salmonella GroEL molecule that is presented by the non-polymorphic MHC class Ib molecule Qa-1. T cells recognizing the bacterial peptide also cross-recognize a homologous peptide from the mammalian hsp60 molecule. Since Qa-1 has a functional equivalent in humans, this observation may be relevant not only to the host response involved in clearing infection but also in understanding the link between infection with Gram-negative pathogens and autoimmune disease.
Collapse
Affiliation(s)
- M J Soloski
- Division of Rheumatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA.
| | | |
Collapse
|
39
|
Abstract
Mucosal administration of vaccines is an important approach to the induction of appropriate immune responses to microbial and other environmental antigens in systemic sites and peripheral blood as well as in most external mucosal surfaces. The development of specific antibody- or T-cell-mediated immunologic responses and the induction of mucosally induced systemic immunologic hyporesponsiveness (oral or mucosal tolerance) depend on complex sets of immunologic events, including the nature of the antigenic stimulation of specialized lymphoid structures in the host, antigen-induced activation of different populations of regulatory T cells (Th1 versus Th2), and the expression of proinflammatory and immunoregulatory cytokines. Availability of mucosal vaccines will provide a painless approach to deliver large numbers of vaccine antigens for human immunization. Currently, an average infant will receive 20 to 25 percutaneous injections for vaccination against different childhood infections by 18 months of age. It should be possible to develop for human use effective, nonliving, recombinant, replicating, transgenic, and microbial vector- or plant-based mucosal vaccines to prevent infections. Based on the experience with many dietary antigens, it is also possible to manipulate the mucosal immune system to induce systemic tolerance against environmental, dietary, and possibly other autoantigens associated with allergic and autoimmune disorders. Mucosal immunity offers new strategies to induce protective immune responses against a variety of infectious agents. Such immunization may also provide new prophylactic or therapeutic avenues in the control of autoimmune diseases in humans.
Collapse
|
40
|
Abstract
Mucosal administration of vaccines is an important approach to the induction of appropriate immune responses to microbial and other environmental antigens in systemic sites and peripheral blood as well as in most external mucosal surfaces. The development of specific antibody- or T-cell-mediated immunologic responses and the induction of mucosally induced systemic immunologic hyporesponsiveness (oral or mucosal tolerance) depend on complex sets of immunologic events, including the nature of the antigenic stimulation of specialized lymphoid structures in the host, antigen-induced activation of different populations of regulatory T cells (Th1 versus Th2), and the expression of proinflammatory and immunoregulatory cytokines. Availability of mucosal vaccines will provide a painless approach to deliver large numbers of vaccine antigens for human immunization. Currently, an average infant will receive 20 to 25 percutaneous injections for vaccination against different childhood infections by 18 months of age. It should be possible to develop for human use effective, nonliving, recombinant, replicating, transgenic, and microbial vector- or plant-based mucosal vaccines to prevent infections. Based on the experience with many dietary antigens, it is also possible to manipulate the mucosal immune system to induce systemic tolerance against environmental, dietary, and possibly other autoantigens associated with allergic and autoimmune disorders. Mucosal immunity offers new strategies to induce protective immune responses against a variety of infectious agents. Such immunization may also provide new prophylactic or therapeutic avenues in the control of autoimmune diseases in humans.
Collapse
Affiliation(s)
- P L Ogra
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, and Children's Hospital of Buffalo, Buffalo, New York 14222, USA.
| | | | | |
Collapse
|
41
|
Eo SK, Pack C, Kumaraguru U, Rouse BT. Optimisation of DNA vaccines for the prophylaxis and modulation of herpes simplex virus infections. Expert Opin Biol Ther 2001; 1:213-25. [PMID: 11727531 DOI: 10.1517/14712598.1.2.213] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Herpes simplex virus (HSV) lacks an effective vaccine. Despite its prevalence and importance HSV infection is not controlled with an acceptable vaccine. Perhaps the best candidate and so far untested approach is the use of plasmid DNA encoding viral proteins. Immunomodulators are also holding some hope as a potential therapeutic. In this review various DNA vaccine approaches used in animal model systems to prevent HSV infections are discussed. Judgements are made as to which of these may prove effective for prophylactic or therapeutic vaccines in humans.
Collapse
Affiliation(s)
- S K Eo
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | | | | | | |
Collapse
|
42
|
Mastroeni P, Chabalgoity JA, Dunstan SJ, Maskell DJ, Dougan G. Salmonella: immune responses and vaccines. Vet J 2001; 161:132-64. [PMID: 11243685 DOI: 10.1053/tvjl.2000.0502] [Citation(s) in RCA: 153] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Salmonella infections are a serious medical and veterinary problem world-wide and cause concern in the food industry. Vaccination is an effective tool for the prevention of Salmonella infections. Host resistance to Salmonella relies initially on the production of inflammatory cytokines leading to the infiltration of activated inflammatory cells in the tissues. Thereafter T- and B-cell dependent specific immunity develops allowing the clearance of Salmonella microorganisms from the tissues and the establishment of long-lasting acquired immunity to re-infection. The increased resistance that develops after primary infection/ vaccination requires T-cells cytokines such as IFNgamma TNFalpha and IL12 in addition to opsonising antibody. However for reasons that are not fully understood seroconversion and/or the presence of detectable T-cell memory do not always correlate with the development of acquired resistance to infection.Whole-cell killed vaccines and subunit vaccines are used in the prevention of Salmonella infection in animals and in humans with variable results. A number of early live Salmonella vaccines derived empirically by chemical or u.v. mutagenesis proved to be immunogenic and protective and are still in use despite the need for repeated parenteral administration. Recent progress in the knowledge of the genetics of Salmonella virulence and modern recombinant DNA technology offers the possibility to introduce multiple defined attenuating and irreversible mutations into the bacterial genome. This has recently allowed the development of Salmonella strains devoid of significant side effects but still capable of inducing solid immunity after single oral administration. Live attenuated Salmonella vaccines have been used for the expression of heterologous antigens/proteins that can be successfully delivered to the immune system. Furthermore Salmonella can transfer plasmids encoding foreign antigens under the control of eukaryotic promoters (DNA vaccines) to antigen-presenting cells resulting in targeted delivery of DNA vaccines to these cells. Despite the great recent advances in the development of Salmonella vaccines a large proportion of the work has been conducted in laboratory rodents and more research in other animal species is required.
Collapse
Affiliation(s)
- P Mastroeni
- Centre for Veterinary Science, University of Cambridge, Madingley Road, Cambridge, CB3 OES, UK.
| | | | | | | | | |
Collapse
|
43
|
Abstract
Epidemiological studies have revealed that HIV-1 infections occur through contact with contaminated blood or during unprotected vaginal or anal intercourse. Hence, to protect against HIV infection, vaccines should ideally induce both mucosal and systemic immune responses. We present a brief review of the different delivery systems and adjuvants which can be used to elicit mucosal immune responses. Oral or nasal administration of recombinant attenuated bacteria or viruses can induce both mucosal and systemic immune responses against the carried antigen. The oral delivery of mucosal adjuvants (such as cholera toxin) in association with antigens has been shown to enhance mucosal and systemic immune responses against them. Recently developed vaccination strategies using naked DNA or other antigen delivery systems are also discussed.
Collapse
Affiliation(s)
- D Velin
- Centre d'Immunologie Pierre Fabre (CIPF), St. Julien en Genevois, France
| | | |
Collapse
|
44
|
Hess J, Schaible U, Raupach B, Kaufmann SH. Exploiting the immune system: toward new vaccines against intracellular bacteria. Adv Immunol 2001; 75:1-88. [PMID: 10879281 DOI: 10.1016/s0065-2776(00)75001-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- J Hess
- Department of Immunology, Max-Planck-Institute for Infection Biology, Berlin, Germany
| | | | | | | |
Collapse
|
45
|
Oliveira-Ferreira J, Daniel-Ribeiro C. Protective CD8+ T cell responses against the pre-erythrocytic stages of malaria parasites: an overview. Mem Inst Oswaldo Cruz 2001; 96:221-7. [PMID: 11285500 DOI: 10.1590/s0074-02762001000200014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
CD8+ T cells have been implicated as critical effector cells in protection against the pre-erythrocytic stage of malaria in mice and humans following irradiated sporozoite immunization. Immunization experiments in animal models by several investigators have suggested different strategies for vaccination against malaria and many of the targets from liver stage malaria antigens have been shown to be immunogenic and to protect mice from the sporozoite challenge. Several prime/boost protocols with replicating vectors, such as vaccinia/influenza, with non-replicating vectors, such as recombinant particles derived from yeast transposon (Ty-particles) and modified vaccinia virus Ankara, and DNA, significantly enhanced CD8+ T cell immunogenicity and also the protective efficacy against the circumsporosoite protein of Plasmodium berghei and P. yeti. Based on these experimental results the development of a CD8+ T cell inducing vaccine has moved forward from epitope identification to planning stages of safety and immunogenicity trials of candidate vaccines.
Collapse
Affiliation(s)
- J Oliveira-Ferreira
- Laboratório de Pesquisas em Malária, Instituto Oswaldo Cruz, Rio de Janeiro, RJ, 21045-900, Brasil
| | | |
Collapse
|
46
|
Revaz V, Benyacoub J, Kast WM, Schiller JT, De Grandi P, Nardelli-Haefliger D. Mucosal vaccination with a recombinant Salmonella typhimurium expressing human papillomavirus type 16 (HPV16) L1 virus-like particles (VLPs) or HPV16 VLPs purified from insect cells inhibits the growth of HPV16-expressing tumor cells in mice. Virology 2001; 279:354-60. [PMID: 11145916 DOI: 10.1006/viro.2000.0717] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Human papillomaviruses, mainly type 16 (HPV16), are responsible for cervical intraepithelial neoplasia, which can lead, in association with other factors, to cervical cancer. Both Salmonella recombinant vaccine strains assembling HPV16 virus-like particles (VLPs) and HPV16 VLPs purified from insect cells are able to induce HPV16 neutralizing antibodies in genital secretions of mice after nasal immunization. Anti-HPV16-specific antibodies in cervical secretions of women may prevent genital infection with HPV16, although this cannot be critically evaluated in the absence of an experimental model for genital papillomavirus infection. Induction of HPV16-specific cell-mediated immunity in the genital mucosa could improve the efficacy of a vaccine and a mucosal route of immunization might be necessary to do so. It has been shown that systemic immunization of mice with purified HPV16 VLPs confers protection against an HPV16-expressing tumor cell challenge through the induction of cytotoxic T-lymphocytes. Using the same C3 tumor model, we show that intranasal immunization of mice with purified HPV16 VLPs in a prophylactic setting also induces anti-tumor immunity. More interestingly, mucosal vaccination of mice with a Salmonella recombinant strain stably expressing HPV16 L1 VLPs also induces anti-tumor immunity in prophylactic as well as in therapeutic settings. Our data suggest that attenuated Salmonella strains expressing chimeric VLPs containing nonstructural viral proteins might be a promising candidate vaccine against cervical cancer by inducing both neutralizing antibodies and cell-mediated immunity.
Collapse
Affiliation(s)
- V Revaz
- Department of Gynecology, Centre Hospitalier Universitaire Vaudois, Lausanne, CH-1011, Switzerland
| | | | | | | | | | | |
Collapse
|
47
|
Lo-Man R, Langeveld JP, Dériaud E, Jehanno M, Rojas M, Clément JM, Meloen RH, Hofnung M, Leclerc C. Extending the CD4(+) T-cell epitope specificity of the Th1 immune response to an antigen using a Salmonella enterica serovar typhimurium delivery vehicle. Infect Immun 2000; 68:3079-89. [PMID: 10816447 PMCID: PMC97535 DOI: 10.1128/iai.68.6.3079-3089.2000] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We analyzed the CD4 T-cell immunodominance of the response to a model antigen (Ag), MalE, when delivered by an attenuated strain of Salmonella enterica serovar Typhimurium (SL3261*pMalE). Compared to purified MalE Ag administered with adjuvant, the mapping of the peptide-specific proliferative responses showed qualitative differences when we used the Salmonella vehicle. We observed the disappearance of one out of eight MalE peptides' T-cell reactivity upon SL3261*pMalE immunization, but this phenomenon was probably due to a low level of T-cell priming, since it could be overcome by further immunization. The most striking effect of SL3261*pMalE administration was the activation and stimulation of new MalE peptide-specific T-cell responses that were silent after administration of purified Ag with adjuvant. Ag presentation assays performed with MalE-specific T-cell hybridomas showed that infection of Ag-presenting cells by this intracellular attenuated bacterium did not affect the processing and presentation of the different MalE peptides by major histocompatibility complex (MHC) class II molecules and therefore did not account for immunodominance modulation. Thus, immunodominance of the T-cell response to microorganisms is governed not only by the frequency of the available T-cell repertoire or the processing steps in Ag-presenting cells that lead to MHC presentation but also by other parameters probably related to the infectious process and to the bacterial products. Our results indicate that, upon infection by a microorganism, the specificity of the T-cell response induced against its Ags can be much more effective than with purified Ags and that it cannot completely be mimicked by purified Ags administered with adjuvant.
Collapse
Affiliation(s)
- R Lo-Man
- Unité de Biologie des Régulations Immunitaires, Institut Pasteur, Paris, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lee JJ, Sinha KA, Harrison JA, de Hormaeche RD, Riveau G, Pierce RJ, Capron A, Wilson RA, Khan CM. Tetanus toxin fragment C expressed in live Salmonella vaccines enhances antibody responses to its fusion partner Schistosoma haematobium glutathione S-transferase. Infect Immun 2000; 68:2503-12. [PMID: 10768937 PMCID: PMC97452 DOI: 10.1128/iai.68.5.2503-2512.2000] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tetanus toxoid has been used widely as an adjuvant. The atoxic fragment C from tetanus toxin (TetC) is potently immunogenic when expressed in Salmonella vaccine strains and has been used as a fusion partner for antigens (Ag). However, there has been no formal comparison of the immunomodulatory impact of TetC on its fusion partners. In this study, we have addressed this important issue. The protective 28-kDa glutathione S-transferase (GST) from Schistosoma haematobium (Sh28GST) was expressed either as a fusion to TetC or as the full-length Sh28GST alone in a nonvirulent aroA-attenuated strain of Salmonella enterica serovar Typhimurium. The Sh28GST proteins were soluble and stably expressed in Salmonella, as evaluated by Western blotting with TetC and/or Sh28GST antisera. Mice were immunized orally with a single dose of the live recombinant Salmonella. The constructs were stable in mice but, dramatically, only the strain expressing the TetC-Sh28GST fusion elicited significant antibody (Ab) responses directed against Sh28GST as determined by enzyme-linked immunosorbent assay. An analysis of the isotype profiles showed that these mice also produced anti-Sh28GST immunoglobulin A and GST-neutralizing assays revealed high levels of neutralizing Abs in sera. These are important correlates of protection in schistosomiasis. In addition, stimulation of spleen cells from immunized mice with Sh28GST Ag showed that both strains, expressing Sh28GST alone or the TetC-Sh28GST fusion, were able to stimulate the secretion of Th1-related cytokines (gamma interferon and interleukin 2) to comparable levels. Thus, TetC has modulated the immune responses generated against its fusion partner, Sh28GST, by markedly enhancing the Ab responses elicited. These results have important implications in the rational development of live vaccines.
Collapse
Affiliation(s)
- J J Lee
- Department of Microbiology and Immunology, The Medical School, University of Newcastle, Newcastle upon Tyne NE2 4HH, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
An LL, Rodriguez F, Harkins S, Zhang J, Whitton JL. Quantitative and qualitative analyses of the immune responses induced by a multivalent minigene DNA vaccine. Vaccine 2000; 18:2132-41. [PMID: 10715528 DOI: 10.1016/s0264-410x(99)00546-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Vaccines containing minigenes - isolated antigenic epitopes encoded by short open reading frames - can, under certain circumstances, confer protective immunity upon the vaccinee. Here we evaluate the efficacy of the minigene vaccine approach using DNA immunization and find that, to be immunogenic, a minigene-encoded epitope requires a perfect "Kozak" translational initiation region. In addition, using intracellular cytokine staining, we show that immunization with a plasmid encoding a full-length protein induces epitope-specific CD8(+) T cells which are detectable directly ex vivo, and constitute approximately 2% of the vaccinee's splenic CD8(+) T cells. In contrast, such cells are undetectable directly ex vivo in recipients of a minigene vaccine. Nevertheless, the minigene plasmid does induce a low number of epitope-specific CD8(+) T cells, which can be amplified to detectable levels by in vivo stimulation. Indeed, 4 days after in vivo stimulation (by virus infection), all vaccinated mice - regardless of whether they had been vaccinated with the minigene or with the full-length gene - had similar numbers of epitope-specific CD8(+) T cells. However, despite these strong responses at 4 days post-infection, recipients of the minigene vaccine showed no enhanced ability to limit virus replication and dissemination. We therefore observe a dichotomy; minigene vaccinees are not protected, despite the presence of strong virus-specific immune responses at 4 days post-challenge. We suggest that the protective benefits of vaccination exert themselves very soon - perhaps within minutes or hours - after virus challenge. If the vaccine-induced immune response is too low to achieve this early protective effect, virus-specific T cells will expand rapidly, but ineffectually, leading to the strong but non-protective response measured at 4 days post-infection. Thus, vaccine-induced immunity should be monitored very early in infection, since the extent to which these responses may later be amplified is largely irrelevant to the protection observed.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Base Sequence
- CD8-Positive T-Lymphocytes/immunology
- Codon/genetics
- Cytokines/biosynthesis
- Epitopes/genetics
- Epitopes/immunology
- Genes, Synthetic
- Immunity, Cellular
- Lymphocyte Count
- Lymphocytic choriomeningitis virus/genetics
- Lymphocytic choriomeningitis virus/immunology
- Lymphocytic choriomeningitis virus/physiology
- Mengovirus/genetics
- Mengovirus/immunology
- Mengovirus/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Molecular Sequence Data
- Open Reading Frames
- Plasmids/genetics
- Plasmids/immunology
- Regulatory Sequences, Nucleic Acid
- Respiratory Syncytial Viruses/genetics
- Respiratory Syncytial Viruses/immunology
- Respirovirus/genetics
- Respirovirus/immunology
- Spleen/immunology
- Time Factors
- Vaccination
- Vaccines, DNA/immunology
- Vesicular stomatitis Indiana virus/genetics
- Vesicular stomatitis Indiana virus/immunology
- Viral Vaccines/immunology
- Virus Replication
Collapse
Affiliation(s)
- L L An
- Department of Neuropharmacology, CVN-9, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
50
|
Oliveira-Ferreira J, Miyahira Y, Layton GT, Savage N, Esteban M, Rodriguez D, Rodriguez JR, Nussenzweig RS, Zavala F, Myahira Y. Immunogenicity of Ty-VLP bearing a CD8(+) T cell epitope of the CS protein of P. yoelii: enhanced memory response by boosting with recombinant vaccinia virus. Vaccine 2000; 18:1863-9. [PMID: 10699335 DOI: 10.1016/s0264-410x(99)00344-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We characterized the immunogenicity of the hybrid Ty-virus-like carrying the CD8(+) T cell epitope (SYVPSAEQI) of the circumsporozoite (CS) protein of Plasmodium yoelii (TyCS-VLP), a rodent malaria parasite. Balb/c mice were immunized with hybrid TyCS-VLP, and their CS-specific CD8(+) T cell response was quantitatively evaluated with the ELISPOT assay, based on the enumeration of epitope specific gamma-interferon secreting CD8(+) T cell. A single immunization with the TyCS-VLP by a variety of routes and doses indicated that the maximal response occurred in mice, which were immunized with 50 micrograms of these particles, administered via intramuscular. Combined immunization of mice with this TyCS-VLP followed by recombinant vaccinia virus expressing the entire P. yoelii CS protein (VacPyCS) or irradiated sporozoites, induced high levels of IFN-gamma-producing cells. The immunization regime, priming with TyCS-VLP and boosting with VacPyCS generated a potent protective immune response, which strongly inhibited P. yoelii liver stages development and protected 62% of the mice against a subsequent live P. yoelii sporozoite challenge.
Collapse
Affiliation(s)
- J Oliveira-Ferreira
- Department of Immunology, Instituto Oswaldo Cruz-FIOCRUZ, Rio de Janeiro, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|