Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
NPJ Vaccines ; 9(1): 2, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38167387

ABSTRACT

Diarrhea caused by Shigella has been associated with high morbidity and mortality in young children worldwide. There are no licensed vaccines, and those clinically advanced have restricted coverage as they elicit serotype-specific immunity while disease is caused by multiple circulating serotypes. Our group had previously reported a close association between serum antibodies to the Shigella virulence factor VirG (or IcsA) and clinical protection in infected individuals. VirG is highly conserved among Shigella strains and appealing as a broad-spectrum vaccine candidate. In this study, we investigated the immunogenicity and protective capacity of VirG as a subunit vaccine in mice. The surface-exposed alpha (α) domain of VirG (VirGα) was produced as a recombinant protein. This region has almost identical immune reactivity to full-length VirG. Administered intramuscularly with alum, VirGα elicited robust immune responses and high protective efficacy against S. flexneri 2a and S. sonnei. Almost complete protection was afforded by VirGα given intranasally with the E. coli double mutant heat-labile toxin (dmLT). VirGα-specific antibodies recognized VirG expressed on live Shigella, and blocked Shigella adhesion and invasion to human colonic cells. These results show for the first time that VirGα is a promising cross-protective vaccine candidate to prevent Shigella infection.

2.
Vaccine ; 41(42): 6261-6271, 2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37666695

ABSTRACT

Shigella species cause severe disease among travelers to, and children living in, endemic countries. Although significant efforts have been made to improve sanitation, increased antibiotic resistance and other factors suggest an effective vaccine is a critical need. Artificial Invaplex (InvaplexAR) is a subunit vaccine approach complexing Shigella LPS with invasion plasmid antigens. In pre-clinical studies, the InvaplexAR vaccine demonstrated increased immunogenicity as compared to the first generation product and was subsequently manufactured under cGMP for clinical testing in a first-in-human Phase 1 study. The primary objective of this study was the safety of S. flexneri 2a InvaplexAR given by intranasal (IN) immunization (without adjuvant) in a single-center, open-label, dose-escalating Phase 1 trial and secondarily to assess immunogenicity to identify a dose of InvaplexAR for subsequent clinical evaluations. Subjects received three IN immunizations of InvaplexAR, two weeks apart, in increasing dose cohorts (10 µg, 50 µg, 250 µg, and 500 µg). Adverse events were monitored using symptom surveillance, memory aids, and targeted physical exams. Samples were collected throughout the study to investigate vaccine-induced systemic and mucosal immune responses. There were no adverse events that met vaccination-stopping criteria. The majority (96%) of vaccine-related adverse events were mild in severity (most commonly nasal congestion, rhinorrhea, and post-nasal drip). Vaccination with InvaplexAR induced anti-LPS serum IgG responses and anti-Invaplex IgA and IgG antibody secreting cell (ASC) responses at vaccine doses ≥250 µg. Additionally, mucosal immune responses and functional antibody responses were seen from the serum bactericidal assay measurements. Notably, the responder rates and the kinetics of ASCs and antibody lymphocyte secretion (ALS) were similar, suggesting that either assay may be employed to identify IgG and IgA secreting cells. Further studies with InvaplexAR will evaluate alternative immunization routes, vaccination schedules and formulations to further optimize immunogenicity. (Clinical Trial Registry Number NCT02445963).

3.
mSphere ; 8(4): e0007323, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37389412

ABSTRACT

The Shigella artificial invasin complex (InvaplexAR) vaccine is a subunit approach that effectively induces robust immunogenicity directed to serotype-specific lipopolysaccharide and the broadly conserved IpaB and IpaC proteins. One advantage of the vaccine approach is the ability to adjust the constituents to address suboptimal immunogenicity and to change the Shigella serotype targeted by the vaccine. As the vaccine moves through the product development pipeline, substantial modifications have been made to address manufacturing feasibility, acceptability to regulatory authorities, and developing immunogenic and effective products for an expanded list of Shigella serotypes. Modifications of the recombinant clones used to express affinity tag-free proteins using well-established purification methods, changes to detergents utilized in the assembly process, and in vitro and in vivo evaluation of different Invaplex formulations have led to the establishment of a scalable, reproducible manufacturing process and enhanced immunogenicity of Invaplex products designed to protect against four of the most predominant Shigella serotypes responsible for global morbidity and mortality. These adjustments and improvements provide the pathway for the manufacture and clinical testing of a multivalent Invaplex vaccine. IMPORTANCE Shigella species are a major global health concern that cause severe diarrhea and dysentery in children and travelers to endemic areas of the world. Despite significant advancements in access to clean water, the increases in antimicrobial resistance and the risk of post-infection sequelae, including cognitive and physical stunting in children, highlight the urgent need for an efficacious vaccine. One promising vaccine approach, artificial Invaplex, delivers key antigens recognized by the immune system during infection, which results in increased resistance to re-infection. The work presented here describes novel modifications to a previously described vaccine approach resulting in improved methods for manufacturing and regulatory approvals, expansion of the breadth of coverage to all major Shigella serotypes, and an increase in the potency of artificial Invaplex.


Subject(s)
Shigella Vaccines , Shigella , Vaccines , Child , Humans , Shigella flexneri , Lipopolysaccharides
4.
mSphere ; 8(3): e0001923, 2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37017547

ABSTRACT

Shigella is responsible for high burdens of diarrhea and dysentery globally. Children living in areas of endemicity are the most affected, and currently, there are no licensed vaccines to prevent shigellosis. Vaccine approaches have traditionally targeted the bacterial lipopolysaccharide as a protective antigen. Shigella O-polysaccharide (OPS) conjugated to recombinant Pseudomonas aeruginosa exotoxin A (rEPA) or tetanus toxoid (TT) is advanced in clinical evaluation. Adequate efficacy of these vaccines, particularly in the infant target group, remains to be demonstrated. A major limitation of the OPS-glycoconjugate concept is its limited coverage, since immunity to the O antigen is serotype specific, and there are multiple disease-causing serotypes. Another concern is the use of protein carriers already included in multiple other childhood vaccines. This study reports a novel Shigella OPS conjugate vaccine that uses the Shigella invasion plasmid antigen B (IpaB) as the carrier protein. IpaB is a virulence factor component of the Shigella type III secretion system and highly conserved among Shigella serotypes. It is robustly immunogenic and a protective antigen. IpaB and IpaB containing nonnative amino acids (nnAA) were produced at large scale using cell-free protein synthesis. Incorporation of nnAA enabled site-specific conjugation of IpaB to Shigella flexneri 2a OPS using click chemistry, yielding OPS-IpaB glycoconjugate. Parenteral immunization of mice with the OPS-IpaB vaccine resulted in high levels of OPS- and IpaB-specific serum IgG and robust protection against lethal S. flexneri 2a or Shigella sonnei challenge. The OPS-IpaB vaccine is a promising new vaccine candidate with the capacity to confer broad protection against clinically relevant Shigella serotypes. IMPORTANCE Diarrhea caused by Shigella species results in long-term disability and mortality globally, disproportionally affecting younger children living in poor countries. Although it is treatable by antibiotics, the rapid and widespread emergence of resistant strains and the highly contagious nature of the disease compel the development of preventive tools. Currently, several Shigella OPS conjugate vaccines are being evaluated in clinical studies, but these rely exclusively on immunity against the bacterial O antigen, which limits their coverage to only the immunizing serotype; multivalent vaccines are needed to protect against the most prevalent serotypes. This is the first report of a novel Shigella OPS-conjugate vaccine that uses Shigella IpaB as a carrier and protective antigen. This vaccine, administered parenterally, elicited robust immunity and protected mice against lethal infection by S. flexneri 2a or S. sonnei. The OPS-IpaB vaccine is a promising candidate for evaluation in vulnerable populations.


Subject(s)
Shigella Vaccines , Shigella , Animals , Mice , Vaccines, Conjugate , Serogroup , Antibody Formation , Lipopolysaccharides , O Antigens , Pseudomonas aeruginosa Exotoxin A
5.
Vaccines (Basel) ; 10(4)2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35455297

ABSTRACT

The Shigella invasin complex or Invaplex vaccine is a unique subunit approach to generate a protective immune response. Invaplex is a large, macromolecular complex consisting of the major Shigella antigens: lipopolysaccharide (LPS) and the invasion plasmid antigen (Ipa) proteins B and C. Over the past several decades, the vaccine has progressed from initial observations through pre-clinical studies to cGMP manufacture and clinical evaluations. The Invaplex product maintains unique biological properties associated with the invasiveness of virulent shigellae and also presents both serotype-specific epitopes, as well as highly conserved invasin protein epitopes, to the immunized host. The vaccine product has evolved from a native product isolated from wild-type shigellae (native Invaplex) to a more defined vaccine produced from purified LPS and recombinant IpaB and IpaC (artificial Invaplex). Each successive "generation" of the vaccine is derived from earlier versions, resulting in improved immunogenicity, homogeneity and effectiveness. The current vaccine, detoxified artificial Invaplex (InvaplexAR-Detox), was developed for parenteral administration by incorporating LPS with under-acylated lipid A. InvaplexAR-Detox has demonstrated an excellent safety and immunogenicity profile in initial clinical studies and is advancing toward evaluations in the target populations of children and travelers to endemic countries.

6.
Appl Microbiol Biotechnol ; 106(1): 401-414, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34932164

ABSTRACT

Shigella spp. invade the colonic epithelium and cause bacillary dysentery in humans. Individuals living in areas that lack access to clean water and sanitation are the most affected. Even though infection can be treated with antibiotics, Shigella antimicrobial drug resistance complicates clinical management. Despite decades of effort, there are no licensed vaccines to prevent shigellosis. The highly conserved invasion plasmid antigens (Ipa), which are components of the Shigella type III secretion system, participate in bacterial epithelial cell invasion and have been pursued as vaccine targets. However, expression and purification of these proteins in conventional cell-based systems have been challenging due to solubility issues and extremely low recovery yields. These difficulties have impeded manufacturing and clinical advancement. In this study, we describe a new method to express Ipa proteins using the Xpress+TM cell-free protein synthesis (CFPS) platform. Both IpaB and the C-terminal domain of IpaH1.4 (IpaH-CTD) were efficiently produced with this technology at yields > 200 mg/L. Furthermore, the expression was linearly scaled in a bioreactor under controlled conditions, and proteins were successfully purified using multimode column chromatography to > 95% purity as determined by SDS-PAGE. Biophysical characterization of the cell-free synthetized IpaB and IpaH-CTD using SEC-MALS analysis showed well-defined oligomeric states of the proteins in solution. Functional analysis revealed similar immunoreactivity as compared to antigens purified from E. coli. These results demonstrate the efficiency of CFPS for Shigella protein production; the practicality and scalability of this method will facilitate production of antigens for Shigella vaccine development and immunological analysis. KEY POINTS : • First report of Shigella IpaB and IpaH produced at high purity and yield using CFPS • CFPS-IpaB and IpaH perform similarly to E. coli-produced proteins in immunoassays • CFPS-IpaB and IpaH react with Shigella-specific human antibodies and are immunogenic in mice.


Subject(s)
Escherichia coli , Shigella , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Escherichia coli/genetics , Mice , Plasmids/genetics , Shigella flexneri , Vaccine Development
7.
Vaccine X ; 8: 100105, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34258576

ABSTRACT

Development of intranasal vaccines for HIV-1 and other mucosal pathogens has been hampered by the lack of adjuvants that can be given safely to humans. We have found that an intranasal Shigella vaccine (Invaplex) which is well tolerated in humans can also function as an adjuvant for intranasal protein and DNA vaccines in mice. To determine whether Invaplex could potentially adjuvant similar vaccines in humans, we simultaneously administered a simian immunodeficiency virus (SIV) envelope (Env) protein and DNA encoding simian-human immunodeficiency virus (SHIV) with or without Invaplex in the nasal cavity of female rhesus macaques. Animals were intranasally boosted with adenoviral vectors expressing SIV env or gag,pol to evaluate memory responses. Anti-SIV antibodies in sera and nasal, genital tract and rectal secretions were quantitated by ELISA. Intracellular cytokine staining was used to measure Th1-type T cells in blood. Macaques given DNA/protein immunizations with 0.5 mg Invaplex developed greater serum IgG, nasal IgA and cervicovaginal IgA responses to SIV Env and SHIV Gag,Pol proteins when compared to non-adjuvanted controls. Rectal IgA responses to Env were only briefly elevated and not observed to Gag,Pol. Invaplex increased frequencies of IFNγ-producing CD4 and CD8 T cells to the Env protein, but not T cell responses induced by the DNA. Ad-SIV boosting increased Env-specific polyfunctional T cells and Env- and Gag,Pol-specific antibodies in serum and all secretions. The data suggest that Invaplex could be highly effective as an adjuvant for intranasal protein vaccines in humans, especially those intended to prevent infections in the genital or respiratory tract.

8.
mSphere ; 3(2)2018.
Article in English | MEDLINE | ID: mdl-29600284

ABSTRACT

The native Invaplex (InvaplexNAT) vaccine and adjuvant is an ion exchange-purified product derived from the water extract of virulent Shigella species. The key component of InvaplexNAT is a high-molecular-mass complex (HMMC) consisting of the Shigella lipopolysaccharide (LPS) and the invasin proteins IpaB and IpaC. To improve product purity and immunogenicity, artificial Invaplex (InvaplexAR) was developed using recombinant IpaB and IpaC proteins and purified Shigella LPS to assemble an HMMC consisting of all three components. Characterization of InvaplexAR by various methods demonstrated similar characteristics as the previously reported HMMC in InvaplexNAT. The well-defined InvaplexAR vaccine consistently contained greater quantities of IpaB, IpaC, and LPS than InvaplexNAT. InvaplexAR and InvaplexNAT immunogenicities were compared in mouse and guinea pig dose escalation studies. In both models, immunization induced antibody responses specific for InvaplexNAT and LPS while InvaplexAR induced markedly higher anti-IpaB and -IpaC serum IgG and IgA endpoint titers. In the murine model, homologous protection was achieved with 10-fold less InvaplexAR than InvaplexNAT and mice receiving InvaplexAR lost significantly less weight than mice receiving the same amount of InvaplexNAT. Moreover, mice immunized with InvaplexAR were protected from challenge with both homologous and heterologous Shigella serotypes. Guinea pigs receiving approximately 5-fold less InvaplexAR compared to cohorts immunized with InvaplexNAT were protected from ocular challenge. Furthermore, adjuvanticity previously attributed to InvaplexNAT was retained with InvaplexAR. The second-generation Shigella Invaplex vaccine, InvaplexAR, offers significant advantages over InvaplexNAT in reproducibility, flexible yet defined composition, immunogenicity, and protective efficacy. IMPORTANCEShigella species are bacteria that cause severe diarrheal disease worldwide, primarily in young children. Treatment of shigellosis includes oral fluids and antibiotics, but the high burden of disease, increasing prevalence of antibiotic resistance, and long-term health consequences clearly warrant the development of an effective vaccine. One Shigella vaccine under development is termed the invasin complex or Invaplex and is designed to drive an immune response to specific antigens of the bacteria in an effort to protect an individual from infection. The work presented here describes the production and evaluation of a new generation of Invaplex. The improved vaccine stimulates the production of antibodies in immunized mice and guinea pigs and protects these animals from Shigella infection. The next step in the product's development will be to test the safety and immune response induced in humans immunized with Invaplex.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Dysentery, Bacillary/prevention & control , Immunogenicity, Vaccine , Shigella Vaccines/immunology , Shigella/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Dysentery, Bacillary/immunology , Guinea Pigs , Immunization , Immunoglobulin A/blood , Immunoglobulin G/blood , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred BALB C , Reproducibility of Results
9.
Vaccine ; 35(49 Pt A): 6798-6802, 2017 12 14.
Article in English | MEDLINE | ID: mdl-28890194

ABSTRACT

In order to avoid expensive clinical failures, better and more predictive animal models of vaccine efficacy are needed to screen Shigella and ETEC vaccine candidates for protective efficacy. The 2016 Vaccines Against Shigella and ETEC (VASE) Conference included a workshop focused on the strengths and weaknesses of current models, particularly in terms of the correlation to vaccine efficacy in human clinical trials. Workshop presenters shared information on existing preclinical animal models for assessing the immunogenicity and protective efficacy of Shigella and ETEC vaccines. The presentations were followed by a discussion about how to best utilize these models, how the models can be improved, and best practices for Shigella and ETEC vaccine developers. The workshop concluded with three major recommendations for the field: (1) develop better and more consistent reagents for animal studies and make them widely available, (2) prioritize harmonization of animal models and immunology assays, and (3) develop preclinical correlates of protection, which will be key in selecting the best vaccine candidates for further clinical development.


Subject(s)
Disease Models, Animal , Drug Evaluation, Preclinical , Treatment Outcome , Animals , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/prevention & control , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli Vaccines/administration & dosage , Humans , Reproducibility of Results , Shigella/immunology , Shigella Vaccines/administration & dosage
10.
Infect Immun ; 82(5): 2027-36, 2014 May.
Article in English | MEDLINE | ID: mdl-24595138

ABSTRACT

Several animal models exist to evaluate the immunogenicity and protective efficacy of candidate Shigella vaccines. The two most widely used nonprimate models for vaccine development include a murine pulmonary challenge model and a guinea pig keratoconjunctivitis model. Nonhuman primate models exhibit clinical features and gross and microscopic colonic lesions that mimic those induced in human shigellosis. Challenge models for enterotoxigenic Escherichia coli (ETEC) and Campylobacter spp. have been successfully developed with Aotus nancymaae, and the addition of a Shigella-Aotus challenge model would facilitate the testing of combination vaccines. A series of experiments were designed to identify the dose of Shigella flexneri 2a strain 2457T that induces an attack rate of 75% in the Aotus monkey. After primary challenge, the dose required to induce an attack rate of 75% was calculated to be 1 × 10(11) CFU. Shigella-specific immune responses were low after primary challenge and subsequently boosted upon rechallenge. However, preexisting immunity derived from the primary challenge was insufficient to protect against the homologous Shigella serotype. A successive study in A. nancymaae evaluated the ability of multiple oral immunizations with live-attenuated Shigella vaccine strain SC602 to protect against challenge. After three oral immunizations, animals were challenged with S. flexneri 2a 2457T. A 70% attack rate was demonstrated in control animals, whereas animals immunized with vaccine strain SC602 were protected from challenge (efficacy of 80%; P = 0.05). The overall study results indicate that the Shigella-Aotus nancymaae challenge model may be a valuable tool for evaluating vaccine efficacy and investigating immune correlates of protection.


Subject(s)
Aotidae , Dysentery, Bacillary/prevention & control , Shigella Vaccines/immunology , Administration, Oral , Animals , Antibodies, Bacterial/blood , Diarrhea/microbiology , Diarrhea/prevention & control , Disease Models, Animal , Immunoglobulin A/blood , Immunoglobulin G/blood , Shigella Vaccines/administration & dosage , Shigella Vaccines/adverse effects
12.
J Immunol Methods ; 393(1-2): 18-29, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23583496

ABSTRACT

Infection with Shigella spp. results in bacillary dysentery and a systemic and mucosal antibody response. The immune response is directed at multiple antigens, including LPS and the invasin plasmid antigen (Ipa) proteins, and is capable of conferring short-term, serotype-specific protection. Both live-attenuated and several subunit vaccine approaches have focused on inducing a pronounced mucosal immune response directed to the same antigens recognized after natural infection. Traditionally, Shigella-specific antibody responses are measured using ELISA. Although ELISAs are robust immunological assays, limited sample volume and assay costs often precludes its use for immunological evaluation against multiple antigens. To overcome these shortcomings, a novel assay has been developed to simultaneously measure specific antibody levels to six Shigella antigens, including three serotype-specific LPS preparations and three conserved protein antigens in a Luminex™-based system. Coupling methods were optimized to covalently link recombinant Ipa proteins (IpaB, IpaC, and IpaD) and purified LPS from Shigella sonnei, Shigella flexneri 2a, and Shigella dysenteriae 1 to unique bead sets. The antigen-coupled beads maintained stable reactivity with monoclonal antibodies (mAbs) for 6 weeks (protein) to 3 months (LPS). The specificity of the Luminex assay was similar to an ELISA, with the multiplexed assay providing a larger dynamic range. Comparable levels of antigen-specific reactivity were attained in monoplex or multiplex formats indicating limited interference. The correlations (R(2)) between the ELISA and the multiplexed assay along with the repeatability and reproducibility of the assay were very high. Minor changes in species-specific conjugated secondary antibodies allowed the optimized multiplexed assays to be used to assess antigen-specific antibodies in serum or blood eluted from filter paper isolated from mice and guinea pigs highlighting applicability of the assay for seroepidemiology and pre-clinical/clinical vaccine studies.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Dysentery, Bacillary/immunology , Immunoassay/methods , Shigella/immunology , Animals , Antibody Formation , Bacterial Proteins/immunology , Guinea Pigs , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Lipopolysaccharides/immunology , Mice , Reproducibility of Results , Sensitivity and Specificity
13.
Trans R Soc Trop Med Hyg ; 107(2): 83-90, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23222955

ABSTRACT

BACKGROUND: Acute diarrhea remains a major public health problem in East African nations such as Kenya. Surveillance for a broad range of enteric pathogens is necessary to accurately predict the frequency of pathogens and potential changes in antibiotic resistance patterns. METHOD: Stool samples were collected from September 2009 to September 2011; 193 and 239 samples, from age-matched cases and asymptomatic controls, were collected, respectively, from Kericho and Kisumu District Hospitals in western Kenya. Bacterial pathogens were identified by conventional microbiological methods; antibiotic susceptibility of bacterial isolates was ascertained using the MicroScan WalkAway 40 Plus. An enzyme immunoassay kit was used to detect rotavirus, and ova and parasite examination was conducted by microscopy and an enzyme immunoassay. RESULTS: Rotavirus (10.2% and 10.5%) and Shigella (11% and 8%) were isolated significantly more often in the cases than the controls from Kericho and Kisumu District Hospitals respectively. The diarrheagenic Escherichia coli, Campylobacter jejuni and Salmonella were found most often in the cases while Giardia lamblia and Entamoeba histolytica/E. dispar were found more often in the controls. Most pathogens were isolated from children under 5 years old. More than 50% of the Shigella, Salmonella and diarrheagenic E. coli isolates were multidrug resistant to ampicillin, tetracycline and trimethoprim/sulfamethoxazole with several enteroaggregative and enterotoxigenic E. coli isolates producing extended-spectrum beta-lactamases. CONCLUSION: Accurate epidemiologic information on acute diarrheal illness in Kenya will be critical for augmenting existing diarrhea management policies in terms of treatment and to strengthen future community awareness and health promotion programs.


Subject(s)
Diarrhea/microbiology , Diarrhea/parasitology , Acute Disease , Adolescent , Adult , Anti-Bacterial Agents/pharmacology , Case-Control Studies , Child , Child, Preschool , Diarrhea/virology , Enterobacteriaceae/drug effects , Enterobacteriaceae/isolation & purification , Enterobacteriaceae Infections/microbiology , Feces/microbiology , Feces/parasitology , Feces/virology , Female , Giardia lamblia/isolation & purification , Humans , Kenya , Male , Microbial Sensitivity Tests , Population Surveillance , Rotavirus/isolation & purification , Young Adult
14.
Vaccine ; 29(40): 7009-19, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21787825

ABSTRACT

BACKGROUND: Shigella flexneri 2a lipopolysaccharide 50 is a nasally delivered subunit vaccine consisting of a macromolecular complex composed of LPS, IpaB, IpaC and IpaD. The current study examined vaccine safety and immunogenicity across a dose range and the clinical performance of a new intranasal delivery device. METHODS: Volunteers (N=36) were randomized to receive vaccine via the Dolphin™ (Valois of America, Congers, New York) intranasal spray device at one of three doses (240, 480, and 690 µg) on days 0, 14, and 28. Another group (N=8) received the 240 µg dose via pipette. Vaccine safety was actively monitored and antigen-specific humoral and mucosal immune responses were determined. RESULTS: There were no serious adverse events and the majority of adverse events (98%) were mild. Antibody secreting cells (ASC), plasma, and mucosal immune responses to Shigella antigens were detected at all three dose levels with the 690 µg dose inducing the highest magnitude and frequency of responses. Vaccination with comparable doses of Invaplex 50 via the Dolphin™ resulted in higher plasma and ASC immune responses as compared to pipette delivery. CONCLUSION: In this trial the S. flexneri 2a Invaplex 50 vaccine was safe, well-tolerated and induced robust levels of antigen-specific intestinal IgA and ASC responses. The spray device performed well and offered an advantage over pipette intranasal delivery.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Immunity, Mucosal/immunology , Lipopolysaccharides/immunology , Shigella Vaccines/administration & dosage , Shigella Vaccines/immunology , Shigella flexneri/immunology , Administration, Intranasal , Adolescent , Adult , Animals , Antibodies, Bacterial/blood , Antibody-Producing Cells/immunology , Double-Blind Method , Drug Administration Routes , Female , Guinea Pigs , Humans , Immunity, Humoral/immunology , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/adverse effects , Male , Mice , Middle Aged , Nasal Sprays , Shigella Vaccines/adverse effects , Vaccination/methods , Young Adult
15.
BMC Public Health ; 11 Suppl 2: S7, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21388567

ABSTRACT

The mission of the Armed Forces Health Surveillance Center, Division of Global Emerging Infections Surveillance and Response System (AFHSC-GEIS) is to support global public health and to counter infectious disease threats to the United States Armed Forces, including newly identified agents or those increasing in incidence. Enteric diseases are a growing threat to U.S. forces, which must be ready to deploy to austere environments where the risk of exposure to enteropathogens may be significant and where routine prevention efforts may be impractical. In this report, the authors review the recent activities of AFHSC-GEIS partner laboratories in regards to enteric disease surveillance, prevention and response. Each partner identified recent accomplishments, including support for regional networks. AFHSC/GEIS partners also completed a Strengths, Weaknesses, Opportunities and Threats (SWOT) survey as part of a landscape analysis of global enteric surveillance efforts. The current strengths of this network include excellent laboratory infrastructure, equipment and personnel that provide the opportunity for high-quality epidemiological studies and test platforms for point-of-care diagnostics. Weaknesses include inconsistent guidance and a splintered reporting system that hampers the comparison of data across regions or longitudinally. The newly chartered Enterics Surveillance Steering Committee (ESSC) is intended to provide clear mission guidance, a structured project review process, and central data management and analysis in support of rationally directed enteric disease surveillance efforts.


Subject(s)
Disease Outbreaks/prevention & control , Gastrointestinal Diseases/epidemiology , Global Health , Military Medicine , Sentinel Surveillance , Communicable Diseases/epidemiology , Forecasting , Humans , Incidence , Infection Control , Laboratories , United States
16.
Expert Rev Vaccines ; 8(12): 1693-704, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19943764

ABSTRACT

Shigellosis remains a formidable disease globally, with children of the developing world bearing the greatest number of infections. The need for an affordable, safe and efficacious vaccine has persisted for decades. Vaccines to prevent shigellosis can be divided into living and nonliving approaches. Several nonliving Shigella vaccines are currently at different stages of development and show substantial promise. Outlined here is an overview of multiple nonliving vaccine technologies, highlighting their current status and recent advances in testing. In addition, gaps in the knowledge base regarding immune mechanisms of protection are explored.


Subject(s)
Dysentery, Bacillary/prevention & control , Shigella Vaccines , Vaccines, Inactivated , Vaccines, Subunit , Bacterial Proteins/immunology , Humans , Lipopolysaccharides/immunology , Shigella Vaccines/administration & dosage , Shigella Vaccines/genetics , Shigella Vaccines/immunology , Shigella dysenteriae/immunology , Shigella flexneri/immunology , Shigella sonnei/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
17.
Infect Immun ; 77(5): 1992-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19289511

ABSTRACT

The sit-encoded iron transport system is present within pathogenicity islands in all Shigella spp. and some pathogenic Escherichia coli strains. The islands contain numerous insertion elements and sequences with homology to bacteriophage genes. The Shigella flexneri sit genes can be lost as a result of deletion within the island. The formation of deletions was dependent upon RecA and occurred at relatively high frequency. This suggests that the sit region is inherently unstable, yet sit genes are maintained in all of the clinical isolates tested. Characterization of the sitABCD genes in S. flexneri indicates that they encode a ferrous iron transport system, although the genes are induced aerobically. The sit genes provide a competitive advantage to S. flexneri growing within epithelial cells, and a sitA mutant is outcompeted by the wild type in cultured epithelial cells. The Sit system is also required for virulence in a mouse lung model. The sitA mutant was able to infect the mice and induce a protective immune response but was avirulent compared to its wild-type parent strain.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Iron/metabolism , Shigella flexneri/genetics , Shigella flexneri/pathogenicity , Virulence Factors/genetics , Virulence Factors/metabolism , Animals , Cell Line , Dysentery, Bacillary/microbiology , Epithelial Cells/microbiology , Female , Gene Deletion , Humans , Lung/microbiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred BALB C , Virulence
18.
Clin Vaccine Immunol ; 16(4): 574-86, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19225079

ABSTRACT

Protection against many infectious diseases may require the induction of cell-mediated and mucosal immunity. Immunization with plasmid DNA-based vaccines has successfully induced cell-mediated immune responses in small animals but is less potent in humans. Therefore, several methods are under investigation to augment DNA vaccine immunogenicity. In the current study, a mucosal adjuvant consisting of an invasin protein-lipopolysaccharide complex (Invaplex) isolated from Shigella spp. was evaluated as an adjuvant for DNA-based vaccines. Coadministration of plasmid DNA encoding the Orientia tsutsugamushi r56Karp protein with Invaplex resulted in enhanced cellular and humoral responses in intranasally immunized mice compared to immunization with DNA without adjuvant. Mucosal immunoglobulin A, directed to plasmid-encoded antigen, was detected in lung and intestinal compartments after Invaplex-DNA immunization followed by a protein booster. Moreover, immunization with Invaplex elicited Shigella-specific immune responses, highlighting its potential use in a combination vaccine strategy. The capacity of Invaplex to enhance the immunogenicity of plasmid-encoded genes suggested that Invaplex promoted the uptake and expression of the delivered genes. To better understand the native biological activities of Invaplex related to its adjuvanticity, interactions between Invaplex and mammalian cells were characterized. Invaplex rapidly bound to and was internalized by nonphagocytic, eukaryotic cells in an endocytic process dependent on actin polymerization and independent of microtubule formation. Invaplex also mediated transfection with several plasmid DNA constructs, which could be inhibited with monoclonal antibodies specific for IpaB and IpaC or Invaplex-specific polyclonal sera. The cellular binding and transport capabilities of Invaplex likely contribute to the adjuvanticity and immunogenicity of Invaplex.


Subject(s)
Adhesins, Bacterial/administration & dosage , Adjuvants, Immunologic/administration & dosage , Lipopolysaccharides/administration & dosage , Shigella/chemistry , Vaccines, DNA/immunology , Adhesins, Bacterial/isolation & purification , Adhesins, Bacterial/pharmacology , Adjuvants, Immunologic/isolation & purification , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Antibodies, Bacterial/analysis , Cell Line , Cricetinae , Endocytosis , Immunization, Secondary , Immunoglobulin A/analysis , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/isolation & purification , Lipopolysaccharides/pharmacology , Lung/immunology , Mice , Mice, Inbred BALB C , Mucous Membrane/immunology , Orientia tsutsugamushi/genetics , Orientia tsutsugamushi/immunology
19.
Vaccine ; 26(10): 1353-64, 2008 Mar 04.
Article in English | MEDLINE | ID: mdl-18276045

ABSTRACT

Development of a subunit vaccine for shigellosis requires identification of protective antigens and delivering these antigens in a manner that stimulates immunity comparable to that induced by natural infection. The Shigella invasin complex (Invaplex) vaccine is an ion-exchange-purified extract from virulent Shigella that consists of LPS and several other proteins, including the invasins IpaB and IpaC. Intranasal delivery of Invaplex stimulates protective immunity in small animal models for shigellosis. To identify the active component(s) of Invaplex responsible for its immunogenicity and efficacy, size-exclusion chromatography (SEC) was used to separate Invaplex into several different fractions. A high-molecular mass complex with a molecular mass between 669 MDa and 2 MDa consisted primarily of LPS, IpaB and IpaC and was considered to be a highly purified (HP) form of Invaplex. Using the mouse lung model to evaluate the immunogenicity and efficacy of the SEC fractions it was clearly demonstrated that the high-molecular mass complex of the invasins and LPS was responsible for the protective capacity of parent native Invaplex. Other smaller mass SEC fractions were mostly non-immunogenic and did not stimulate solid protection. In guinea pigs, the HP Invaplex stimulated an enhanced immune response as compared to the parent Invaplex and was fully protective. Isolation and characterization of the immunogenic and protective moiety within Invaplex will allow better standardization of the Invaplex product and may allow future development of an Invaplex assembled from purified components.


Subject(s)
Adhesins, Bacterial/immunology , Shigella Vaccines/immunology , Shigella flexneri/immunology , Adhesins, Bacterial/isolation & purification , Animals , Blotting, Western , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Guinea Pigs , Immunoassay , Immunoglobulin A/biosynthesis , Immunoglobulin A/genetics , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Keratoconjunctivitis/immunology , Keratoconjunctivitis/microbiology , Keratoconjunctivitis/prevention & control , Lipopolysaccharides/immunology , Molecular Weight
20.
Infect Immun ; 74(8): 4391-400, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16861624

ABSTRACT

Shigella flexneri, the causative agent of shigellosis, is a gram-negative bacterial pathogen that initiates infection by invading cells within the colonic epithelium. Contact with host cell surfaces induces a rapid burst of protein secretion via the Shigella type III secretion system (TTSS). The first proteins secreted are IpaD, IpaB, and IpaC, with IpaB and IpaC being inserted into the host cell membrane to form a pore for translocating late effectors into the target cell cytoplasm. The resulting pathogen-host cross talk results in localized actin polymerization, membrane ruffling, and, ultimately, pathogen entry. IpaD is essential for host cell invasion, but its role in this process is just now coming to light. IpaD is a multifunctional protein that controls the secretion and presentation of IpaB and IpaC at the pathogen-host interface. We show here that antibodies recognizing the surface-exposed N terminus of IpaD neutralize Shigella's ability to promote pore formation in erythrocyte membranes. We further show that MxiH and IpaD colocalize on the bacterial surface. When TTSS needles were sheared from the Shigella surface, IpaD was found at only the needle tips. Consistent with this, IpaD localized to the exposed tips of needles that were still attached to the bacterium. Molecular analyses then showed that the IpaD C terminus is required for this surface localization and function. Furthermore, mutations that prevent IpaD surface localization also eliminate all IpaD-related functions. Thus, this study demonstrates that IpaD localizes to the TTSA needle tip, where it functions to control the secretion and proper insertion of translocators into host cell membranes.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Shigella flexneri/pathogenicity , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Erythrocytes/microbiology , Gene Expression Regulation, Bacterial , Hemolysis , Mutation , Neutralization Tests , Shigella flexneri/genetics , Shigella flexneri/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...