Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Postepy Biochem ; 68(2): 179-188, 2022 06 30.
Article in Polish | MEDLINE | ID: mdl-35792641

ABSTRACT

Most pathogens enter the body through the surfaces of the mucous membranes, e.g. the nose or the intestines. The mucosal immune response is essential for the effective elimination of invading pathogens. Unfortunately, most vaccines which are administered intramuscularly by injection do not induce an adequate protective immune response on mucous membranes. For example, after intramuscular injection, the level of secretory IgA antibodies is low and often insufficient to successfully combat the pathogen. On the other hand, mucosal-induced immunity produces a long-lasting effect in the form of a local and systemic response to the pathogen. Moreover, the administration of such vaccines does not generate hazardous medical waste and their application does not require the presence of qualified medical personnel. Therefore, intensive research into vaccines administered via the mucosal route is ongoing. An obstacle in the development of mucosal vaccines is the natural defense mechanisms of the mucosa, the overcoming of which requires the use of specialized adjuvants. Currently, there are no such formulations on the market.


Subject(s)
Mucous Membrane , Vaccines
2.
Vaccines (Basel) ; 9(3)2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33800507

ABSTRACT

Many conventional vaccines are administered via a needle injection, while most pathogens primarily invade the host via mucosal surfaces. Moreover, protective IgA antibodies are insufficiently induced by parenteral vaccines. Mucosal immunity induces both local and systemic response to pathogens and typically lasts for long periods of time. Therefore, vaccination via mucosal routes has been increasingly explored. However, mucosal vaccines require potent adjuvants to become efficacious. Despite many efforts to develop safe and robust adjuvants for mucosal vaccines, only a few have been approved for use in human formulations. The aim of our study was to design, develop and characterize new silicone oil-based nanoadjuvant candidates for intranasal vaccines with potential to become mucosal adjuvants. We have developed an array of nanoadjuvant candidates (NACs), based on well-defined ingredients. NAC1, 2 and 3 are based on silicone oil, but differ in the used detergents and organic solvents, which results in variations in their droplet size and zeta potential. NACs' cytotoxicity, Tumor Necrosis Factor α (TNF-α) induction and their effect on antigen engulfment by immune cells were tested in vitro. Adjuvant properties of NACs were verified by intranasal vaccination of mice together with ovalbumin (OVA). NACs show remarkable stability and do not require any special storage conditions. They exhibit bio-adhesiveness and influence the degree of model protein engulfment by epithelial cells. Moreover, they induce high specific anti-OVA IgG antibody titers after two intranasal administrations. Nanoadjuvant candidates composed of silicone oil and cationic detergents are stable, exhibit remarkable adjuvant properties and can be used as adjuvants for intranasal immunization.

3.
Cells ; 9(5)2020 05 07.
Article in English | MEDLINE | ID: mdl-32392707

ABSTRACT

Clostridioides difficile (CD) is a Gram-positive pathogen responsible for CD-associated disease (CDAD), which is characterized by symptoms ranging from mild diarrhea to pseudomembranous colitis. This work is an attempt to respond to the need of novel methods for CD infection (CDI) prevention, since the number of CDI cases is still rising. A bioinformatics approach was applied to design twenty-one peptides consisting of in silico predicted linear B-cell and T-cell epitopes of aminopeptidase M24 from CD. These peptides were mapped for epitopes exploiting PEPSCAN procedure and using sera obtained from CD infected patients, umbilical cord blood, and healthy volunteers. Two new CD epitopes, 131KKGIK135 and 184KGTSTHVIT192, were identified and characterized. Immunoreactivity of the synthetic biotinylated 131KKGIK135 epitope was significantly higher compared to 184KGTSTHVIT192 epitope in Enzyme-Linked Immunosorbent Assay (ELISA) with umbilical cord blood and CDI patients' sera. Hereafter, the conjugate of bovine serum albumin and epitope 131KKGIK135 was evaluated in vitro on lung epithelial cell line. In vitro, a significant induction of IL-6 by conjugate was observed, thereby we postulate that this new 131KKGIK135 epitope possesses immunostimulating properties suggesting possibility of its use in a vaccine against Clostridioides difficile.


Subject(s)
Aminopeptidases/chemistry , Clostridioides difficile/enzymology , Computational Biology , Peptides/chemistry , Peptides/immunology , Amino Acid Sequence , Cell Line , Clostridium Infections/immunology , Clostridium Infections/microbiology , Epitopes/chemistry , Epitopes/immunology , Humans , Interleukin-6/biosynthesis , Models, Molecular
4.
Microorganisms ; 7(11)2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31739602

ABSTRACT

Clostridioides difficile (CD) cause a severe diarrhea which can lead to pseudomembranous colitis and even patient death. CD infection (CDI) is connected mainly with changes in intestinal microbiota as a consequence of antibiotic treatment. The growing resistance to antibiotics, justifies the search for new methods of combating CD. Despite of ongoing research on the immunity against the pathogen, there is still lack of any reliable vaccine. Most recently, Cwp22, that is a cross-linking enzyme involved in the production of CD peptidoglycan, seems to be a promising target to prevent CDI in high-risk patients. In this paper, the Cwp22 protein polypeptide-specific epitopes were mapped in silico and using PEPSCAN procedure. They were recognized not only by antibodies from CDI patients' but also by umbilical cord blood sera. We identified three epitopes 54EFRVAT59, 201KVNGKM206 and 268WQEKNGKKYY277 of Cwp22 protein. Since Cwp22 protein has key functionality and the described above epitopes are also recognized by umbilical cord blood serum, we postulate that they could have important protective properties. In this paper, we propose Cwp22 protein as a good antigen candidate for CDI preventive vaccine. Our results open the possibility to use 54EFRVAT59, 201KVNGKM206 and 268WQEKNGKKYY277, epitopes as suitable anti-CD vaccine antigens.

5.
Sci Rep ; 8(1): 13946, 2018 09 17.
Article in English | MEDLINE | ID: mdl-30224677

ABSTRACT

Clostridium difficile (CD) infections are a growing threat due to the strain resistance to antibiotic treatment and the emergence of hypervirulent strains. One solution to this problem is the search for new vaccine antigens, preferably surface-localized that will be recognized by antibodies at an early stage of colonization. The purpose of the study was to assess the usefulness of novel immunoreactive surface proteins (epitopes) as potential vaccine antigens. Such approach might be tough to pursue since pathogens have acquired strategies to subvert adaptive immune response to produce humoral response against non-essential proteins for their survival. In this study CD surface proteins were isolated, immunoreactive proteins identified and mapped to select potential epitopes. The results of the study exclude the use of CD glyceraldehyde 3-phosphate dehydrogenase as a vaccine antigen, especially as a whole protein. Sequences P9 (201AAGNIVPNTTGAAKAI218) and P10 (224KGKLDGAAQRVPVVTG241) recognized by patients sera are conserved and widespread among CD strains. They show cross-reactivity with sera of people suffering from other bacterial infections and are recognized by sera of autoimmune disease patients. Our study documents that special care in analyzing the sequence of new epitope should be taken to avoid side effects prior to consider it as a vaccine antigen.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Clostridioides difficile/immunology , Clostridium Infections/immunology , Epitopes/immunology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/immunology , Adaptive Immunity/immunology , Adolescent , Adult , Amino Acid Sequence , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Autoimmune Diseases/immunology , Cross Reactions/immunology , Epitope Mapping/methods , Female , Humans , Membrane Proteins/immunology , Pregnancy , Sequence Alignment , Vaccines/immunology , Young Adult
6.
Article in English | MEDLINE | ID: mdl-29597292

ABSTRACT

Commensal Escherichia coli population is a dynamic structure which may be important in the pathogenesis of extraintestinal infections. The aim of this study was the comparison of genetic diversity of commensal E. coli isolates from two age group-adults and young children. E. coli strains were isolated on MacConkey agar and identified by biochemical tests. Determination of four major phylogenetic groups, identification of virulence genes and antimicrobial resistance determinants were performed by using multiplex or simplex PCR. Phenotypic analysis of resistance was based on disc-diffusion method. The prevalence of virulence genes was significantly higher among isolates from adults than from young children. Phylogroup B2 predominated among E. coli from adults, whereas phylogroup A was the most common in isolates from young children. The analyses of antimicrobial resistance revealed that resistance to at least one antimicrobial agent and multidrug-resistance were detected significantly more frequent in the isolates from adults than from young children. This study documented that the commensal E. coli isolates from adults showed greater genetic diversity than from young children and constitutes a substantial reservoir of the virulence genes typical for extraintestinal pathogenic E. coli.


Subject(s)
Drug Resistance, Bacterial/genetics , Escherichia coli , Gastrointestinal Microbiome , Genetic Variation , Phylogeny , Symbiosis , Virulence/genetics , Adolescent , Adult , Age Factors , Anti-Bacterial Agents/pharmacology , Child, Preschool , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli/pathogenicity , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Humans , Infant , Male , Middle Aged , Phenotype , Poland , Virulence Factors/genetics , Young Adult
7.
PLoS One ; 10(5): e0126120, 2015.
Article in English | MEDLINE | ID: mdl-25962136

ABSTRACT

Vaccine adjuvants have been reported to induce both mucosal and systemic immunity when applied to mucosal surfaces and this dual response appears important for protection against certain pathogens. Despite the potential advantages, however, no mucosal adjuvants are currently approved for human use. Evaluating compounds as mucosal adjuvants is a slow and costly process due to the need for lengthy animal immunogenicity studies. We have constructed a library of 112 intranasal adjuvant candidate formulations consisting of oil-in-water nanoemulsions that contain various cationic and nonionic surfactants. To facilitate adjuvant development we first evaluated this library in a series of high-throughput, in vitro assays for activities associated with innate and adaptive immune activation in vivo. These in vitro assays screened for the ability of the adjuvant to bind to mucin, induce cytotoxicity, facilitate antigen uptake in epithelial and dendritic cells, and activate cellular pathways. We then sought to determine how these parameters related to adjuvant activity in vivo. While the in vitro assays alone were not enough to predict the in vivo adjuvant activity completely, several interesting relationships were found with immune responses in mice. Furthermore, by varying the physicochemical properties of the surfactant components (charge, surfactant polar head size and hydrophobicity) and the surfactant blend ratio of the formulations, the strength and type of the immune response generated (TH1, TH2, TH17) could be modulated. These findings suggest the possibility of using high-throughput screens to aid in the design of custom adjuvants with unique immunological profiles to match specific mucosal vaccine applications.


Subject(s)
Adjuvants, Immunologic/chemistry , Vaccines/administration & dosage , Vaccines/chemistry , Adjuvants, Immunologic/toxicity , Administration, Intranasal , Animals , Cell Line , Chemistry, Pharmaceutical , Cytokines/biosynthesis , Emulsions , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , High-Throughput Screening Assays , Immunity, Cellular , Immunity, Humoral , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Nanotechnology
8.
Postepy Hig Med Dosw (Online) ; 69: 598-611, 2015 May 05.
Article in Polish | MEDLINE | ID: mdl-25983298

ABSTRACT

Clostridium difficile pathogen is a cause of the most frequent nosocomial infection, which is antibiotic-associated diarrhea. Antibiotic treatment causes disruption of the microbiome balance, which makes the gut a friendly environment for the pathogen. It leads to pseudomembranous colitis, toxic megacolon and even death. Clostridium difficile infection (CDI) is particularly dangerous to elderly patients, leading to the highest mortality rate. C. difficile is equipped with many virulence factors such as toxin A and B, binary toxin CDT, flagellum, S-layer proteins, Cwp66 and GroEL proteins, protease Cwp84, fibronectin-binding protein and the ability to form biofilm and spores. Problems with anti-CDI therapy prompt researchers and clinicians to seek alternative ways of therapy. Identification of immunological epitopes in outer layer proteins and the use of them as antigens for anti-CDI vaccines would be a rational approach to prevent the disease, but unfortunately such vaccines are not available yet. In this article we review the course of the disease, virulence and risk factors. We summarize briefly epidemiological data and the latest achievements in CDI treatment.


Subject(s)
Clostridioides difficile/pathogenicity , Clostridium Infections/physiopathology , Virulence Factors , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Clostridium Infections/therapy , Humans , Risk Factors , Virulence
9.
Vaccine ; 33(19): 2289-2296, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25817825

ABSTRACT

Nanoemulsions (NEs) are adjuvants that enhance antigen penetration of the nasal mucosa, increase cellular uptake of antigens by both epithelial and dendritic cells, and promote the migration of antigen-loaded dendritic cells to regional lymph nodes within 24-h of vaccine administration. The objective of this study was to elucidate cell death caused by W805EC NE and identify caspases and genes associated with death pathways. Consistent with this aim, we show that exposure of human epithelial cells (EC), both RPMI 2650 and FaDu, to NE results in the activation of caspases (1, 3/7, 6, 8, and 9) and the expression of genes involved in apoptotic as well as authophagy and necrosis pathways. Interestingly, the NE activates caspase 8 which promotes "immunogenic apoptosis". The rescue assay was employed to investigate the fate of RPMI 2650 cells treated with W805EC NE. After four-hour treatment with as little as 0.03% of NE no cells were rescued at 72h. Remarkably, immediately after four-hour treatment, the cells morphologically resembled untreated cells and most of the cells were alive. Altogether, these results suggest that NE induces death of human ECs through multiple pathways. Epithelial cell death caused by W805EC may have further implications on antigen uptake, processing, and presentation by DC's.


Subject(s)
Adjuvants, Immunologic/toxicity , Apoptosis , Emulsions/toxicity , Epithelial Cells/drug effects , Epithelial Cells/physiology , Nanoparticles/toxicity , Cell Line , Humans
10.
Hum Vaccin Immunother ; 10(3): 615-22, 2014.
Article in English | MEDLINE | ID: mdl-24326268

ABSTRACT

Respiratory Syncytial Virus (RSV) is a ubiquitous virus that infects almost all people by age two and is a major source of respiratory illness in infants, the elderly and others with compromised immune systems. Currently there is no available vaccine. Prior efforts using formalin-inactivated RSV (FI-RSV) were associated with enhanced respiratory disease upon viral exposure following clinical vaccine trials. Several researchers and pharmaceutical companies have utilized vector-associated live attenuated RSV vaccines in pre-clinical and clinical studies. Another attractive approach, however, is a subunit vaccine which would be easier to produce and quality control. Our group has previously demonstrated in a murine model of infection that intranasal immunization with nanoemulsion-inactivated and adjuvanted RSV induces humoral and cellular immune responses, resulting in protection against RSV infection. The present studies characterize the immune responses elicited by intranasal RSV F protein adjuvanted with nanoemulsion. Intranasal application of nanoemulsion adjuvanted F protein induced a rapid and robust systemic and mucosal antibody response, as well as protection against subsequent RSV challenge. Importantly, RSV challenge in immunized animals did not elicit airway hyper-reactivity, a Th2-skewed immune response or immunopathology associated with hypersensitivity reactions with formalin-inactivated vaccine. These results suggest that RSV F protein adjuvanted with nanoemulsion may be a good mucosal vaccine candidate. Formulating RSV F protein in nanoemulsion creates a well-defined and well-controlled vaccine that can be delivered intranasally to induce T cell mediated immunity without inducing enhanced disease associated with the mouse model of FI-RSV vaccination and infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Immunization/methods , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Administration, Intranasal , Animals , Antibodies, Viral/analysis , Antibodies, Viral/blood , Blood/immunology , Disease Models, Animal , Emulsions/administration & dosage , Female , Immunity, Mucosal , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Viruses/isolation & purification , Th2 Cells/immunology , Treatment Outcome , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Fusion Proteins/immunology
11.
Cell Immunol ; 282(2): 79-84, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23770715

ABSTRACT

OT-II mice were evaluated as a transgenic strain-specific model to assess T-cell help for B-cell responses. OT-II CD4(+) T-cells express transgenic OVA-specific αß-TCRs. This high frequency of antigen-specific helper T-lymphocytes may augment induction of B-cell responses. Unexpectedly, OT-II mice did not produce OVA-specific antibodies after intranasal immunization. However, B-cells expressed normal antigen-presenting function in vitro for activation of OVA-specific T-cell responses. These OT-II T-cell responses produced a Th1-type cytokine profile with significantly reduced Th2 or Th17 responses. These data suggest that OT-II B-cells are not defective as APCs, however, downstream antibody responses are abrogated in this transgenic strain.


Subject(s)
Antibodies/immunology , Ovalbumin/immunology , Receptors, Antigen, T-Cell/immunology , Vaccination/methods , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Cholera Toxin/immunology , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism
12.
Vaccine ; 31(7): 1072-9, 2013 Feb 04.
Article in English | MEDLINE | ID: mdl-23273511

ABSTRACT

Nanoemulsions are adjuvants that enhance antigen penetration in the nasal mucosa, increase cellular uptake of antigens by both epithelial dendritic cells, and promote migration of antigen-loaded dendritic cells to regional lymph nodes within a day of vaccine administration. The objective of this study was to determine whether the W(80)5EC nanoemulsion adjuvant enhances immune response not only by direct uptake of antigen by dendritic cells, but also indirectly, by phagocytosis of antigen-primed, apoptotic, epithelial cells. Consistent with this, we show that exposure of both epithelial cells (TC-1s) and dendritic cells (JAWS II or bone marrow derived dendritic cells (BMDCs)) to nanoemulsion exhibited augmented antigen uptake in cell culture. TC-1 cells subsequently underwent G(2)/M cell cycle arrest and apoptosis, and when co-cultured with JAWS II or BMDCs were rapidly engulfed by the dendritic cells, which responded by up-regulating dendritic cell maturation marker CD86. Altogether these results suggest that the effectiveness of nanoemulsions as adjuvants stems, at least in part, from the engulfment of antigen-loaded epithelial cells, leading to enhanced antigen processing and a strong and balanced mucosal and systemic immune response.


Subject(s)
Adjuvants, Immunologic/metabolism , Antigens/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Emulsions/metabolism , Epithelial Cells/immunology , Phagocytosis/drug effects , Animals , Antigens/metabolism , Female , Mice , Mice, Inbred C57BL
13.
ACS Nano ; 7(1): 214-28, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23259666

ABSTRACT

Vancomycin represents the preferred ligand for bacteria-targeting nanosystems. However, it is inefficient for emerging vancomycin-resistant species because of its poor affinity to the reprogrammed cell wall structure. This study demonstrates the use of a multivalent strategy as an effective way for overcoming such an affinity limitation in bacteria targeting. We designed a series of fifth generation (G5) poly(amidoamine) (PAMAM) dendrimers tethered with vancomycin at the C-terminus at different valencies. We performed surface plasmon resonance (SPR) studies to determine their binding avidity to two cell wall models, each made with either a vancomycin-susceptible (D)-Ala-(D)-Ala or vancomycin-resistant (D)-Ala-(D)-Lac cell wall precursor. These conjugates showed remarkable enhancement in avidity in the cell wall models tested, including the vancomycin-resistant model, which had an increase in avidity of four to five orders of magnitude greater than free vancomycin. The tight adsorption of the conjugate to the model surface corresponded with its ability to bind vancomycin-susceptible Staphylococcus aureus bacterial cells in vitro as imaged by confocal fluorescent microscopy. This vancomycin platform was then used to fabricate the surface of iron oxide nanoparticles by coating them with the dendrimer conjugates, and the resulting dendrimer-covered magnetic nanoparticles were demonstrated to rapidly sequester bacterial cells. In summary, this article investigates the biophysical basis of the tight, multivalent association of dendrimer-based vancomycin conjugates to the bacterial cell wall, and proposes a potential new use of this nanoplatform in targeting Gram-positive bacteria.


Subject(s)
Dendrimers/chemistry , Gram-Positive Bacteria/drug effects , Nanocapsules/administration & dosage , Vancomycin/administration & dosage , Vancomycin/chemistry , Anti-Bacterial Agents/administration & dosage , Apoptosis/drug effects , Gram-Positive Bacteria/cytology , Gram-Positive Bacteria/physiology , Materials Testing , Vancomycin Resistance
14.
Vaccine ; 30(48): 6871-7, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-22989689

ABSTRACT

Currently available influenza vaccines provide suboptimal protection. In order to improve the quality of protective immune responses elicited following vaccination, we developed an oil-in-water nanoemulsion (NE)-based adjuvant for an intranasally-delivered inactivated influenza vaccine. Using a prime-boost vaccination regimen, we show that intranasal vaccines containing the W(80)5EC NE elicited higher titers of serum hemagglutination inhibiting (HAI) antibody and influenza-specific IgG and IgA titers compared to vaccines that did not contain the NE. Similarly, vaccines containing the W(80)5EC NE resulted in higher influenza-specific IgA levels in the bronchoalveolar lavage (BAL) fluid and nasal wash when compared to vaccines formulated without NE. The higher antibody titers in mice immunized with the NE-containing vaccines correlated with reduced viral loads in the lungs and nasal turbinates following a high dose viral challenge. Mice immunized with vaccines containing the W(80)5EC NE also showed a reduction in body weight loss following challenge compared to mice immunized with equivalent vaccines produced without NE. Taken together, our results show that the W(80)5EC NE substantially improves the magnitude of protective influenza-specific antibody responses and is a promising mucosal adjuvant for influenza vaccines and vaccines against other mucosal pathogens.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Emulsions/administration & dosage , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Nanostructures/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/blood , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/virology , Disease Models, Animal , Female , Hemagglutination Inhibition Tests , Immunoglobulin A/blood , Immunoglobulin G/blood , Mice , Nasal Mucosa/immunology , Nasal Mucosa/virology , Orthomyxoviridae Infections/prevention & control , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
15.
Arch Immunol Ther Exp (Warsz) ; 59(4): 249-59, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21644030

ABSTRACT

The idea that vaccination can be used to fight cancer is not new. Approximately 100 years ago, researchers attempted to stimulate a tumor-specific, therapeutic immune response to tumors by injecting patients with cells and extracts from their own tumors, or tumors of the same type from different individuals. During the last decade, great efforts have been made to develop immunotherapeutic approaches for the treatment of malignant diseases as alternatives to traditional chemo- and radiotherapy. A quintessential goal of immunotherapy in cancer is treatment with vaccines that elicit potent anti-tumor immune responses without side effects. In this article, we have attempted to review some of the most problematic issues facing the development of cancer vaccines. With the prospect of immunosuppression, an ill-designed cancer vaccine can be more harmful than a no-benefit therapy. We have noted that "immunoediting" and "immunodominance" are the premier setbacks in peptide-based vaccines and therefore it appears necessary not only to manipulate the activity of a vast number of principal components but also to finely tune their concentrations in time and space. In the face of all these quandaries, it is at least doubtful that any reliable anti-cancer vaccine strategy will emerge in the near future.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines , Immunotherapy/methods , Neoplasms/immunology , Adjuvants, Immunologic , Animals , Antigen-Presenting Cells/immunology , Antineoplastic Agents/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Clinical Trials as Topic , Disease Models, Animal , Humans , Immune Tolerance/immunology , Mice , Models, Immunological , Neoplasms/therapy , T-Lymphocytes/immunology , Vaccines, Subunit/immunology
17.
Hum Vaccin ; 6(7): 585-94, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20421727

ABSTRACT

Mutations of influenza virus increase concerns of worldwide epidemics resulting from the newly emergent strains. Current influenza vaccines are inefficient and require annual vaccinations. W805EC adjuvant is an oil-in-water emulsion composed of nanodroplets with an average diameter of approximately 400 nm. The nanoemulsion adjuvant has been used successfully to stimulate the immune response when mixed with several other antigens in animal models. In this study, W805EC nanoemulsion adjuvant activity was evaluated using nasal influenza vaccination in a murine model. Five to twenty percent W805EC adjuvant was used to inactivate influenza A/Puerto Rico/8/34/05 (H1N1). Mice immunized with the nanoemulsion adjuvanted influenza virus intranasally showed a robust specific humoral immune response as demonstrated using ELISA and HAI assays. Serum HAI titers were more than 104 following two vaccinations. Vaccinated mice were also protected against challenge with an LD80 of live influenza virus. Splenocytes from vaccinated mice were assayed for cytokine production following virus stimulation. The cytokine profile demonstrated a robust cellular immune response with enhanced Th1 and Th17 immunity that provided balanced immunity against both intracellular and extracellular forms of the virus. Additionally, the vaccine preparations showed minimal protein degradation but remained potent when stored at 4°C for up to three months. This work demonstrates that W805EC nanoemulsion adjuvant can effectively enhance the immunogenicity of influenza hemagglutinin antigen. The nanoemulsion adjuvant can result in antigen sparing and cross-protection. The potential exists for a nasally administered influenza vaccine that may require little or no refrigerated storage.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Emulsions/administration & dosage , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Adjuvants, Immunologic/adverse effects , Administration, Intranasal , Animals , Antibodies, Viral/blood , Cytokines/metabolism , Emulsions/adverse effects , Enzyme-Linked Immunosorbent Assay , Female , Hemagglutination Inhibition Tests , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Leukocytes, Mononuclear/immunology , Male , Mice , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/prevention & control , Puerto Rico , Rodent Diseases/mortality , Rodent Diseases/prevention & control , Spleen/immunology , Survival Analysis , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/immunology
18.
J Aerosol Med Pulm Drug Deliv ; 23(2): 77-89, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19778268

ABSTRACT

BACKGROUND: Many infectious diseases that cause significant morbidity and mortality, especially in the developing world, could be preventable through vaccination. The effort to produce safe, thermally stable, and needle-free mucosal vaccines has become increasingly important for global health considerations. We have previously demonstrated that a thermally stable nanoemulsion, a mucosal adjuvant for needle-free nasal immunization, is safe and induces protective immunity with a variety of antigens, including recombinant protein. The successful use of nanoemulsion-based vaccines, however, poses numerous challenges. Among the challenges is optimization of the formulation to maintain thermal stability and potency and another is accuracy and efficiency of dispensing the vaccines to the nasal mucosa in the anterior and turbinate region of the nasal cavity or potentially to the nasopharynx-associated lymphoid tissue. METHODS: We have examined the effects of different diluents [phosphate-buffered saline (PBS) and 0.9% NaCl] on the stability and potency of nanoemulsion-based vaccines. In addition, we have determined the efficiency of delivering them using commercially available nasal spray devices (Pfeiffer SAP-62602 multidose pump and the BD Hypak SCF 0.5 ml unit dose Accuspray(TM)). RESULTS: We report the stability and potency of PBS-diluted ovalbumin-nanomeulsion mixtures for up to 8 months and NaCl-diluted mixtures up to 6 months when stored at room temperature. Significant differences in spray characteristics including droplet size, spray angle, plume width, and ovality ratios were observed between the two pumps. Further, we have demonstrated that the nanoemulsion-based vaccines are not physically or chemically altered and retain potency following actuation with nasal spray devices. Using either device, the measured spray characteristics suggest deposition of nanoemulsion-based vaccines in inductive tissues located in the anterior region of the nasal cavity. CONCLUSIONS: The results of this study suggest that nanoemulsion-based vaccines do not require specially engineered delivery devices and support their potential use as nasopharyngeal vaccine adjuvants.


Subject(s)
Nanoparticles , Nebulizers and Vaporizers , Ovalbumin/administration & dosage , Vaccines/administration & dosage , Administration, Intranasal , Aerosols , Alkaline Phosphatase/administration & dosage , Alkaline Phosphatase/chemistry , Alkaline Phosphatase/immunology , Animals , Drug Stability , Drug Storage , Emulsions , Excipients/chemistry , Female , Hepatitis B Surface Antigens/administration & dosage , Hepatitis B Surface Antigens/chemistry , Hepatitis B Surface Antigens/immunology , Mice , Ovalbumin/chemistry , Ovalbumin/immunology , Particle Size , Sodium Chloride/chemistry , Swine , Vaccines/immunology
19.
Anticancer Drugs ; 21(2): 186-92, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20010426

ABSTRACT

Our earlier studies have shown the in vitro and in vivo targeting of a generation 5 (G5) dendrimer-based multifunctional conjugate that contained folic acid (FA) as the targeting agent and methotrexate (MTX) as the chemotherapeutic drug. To clinically apply the synthesized G5-FA-MTX nanotherapeutic, it is important that the anticancer conjugate elicits cytotoxicity specifically and consistently. Toward this objective, we evaluated the large-scale synthesis of a G5-FA-MTX conjugate (Lot # 123-34) for its cytotoxic potential and specificity in vitro and in vivo. The cytotoxicity and specificity were tested by using a coculture assay in which FA receptor-expressing and nonexpressing cells (KB and SK-BR-3 cells, respectively) were cultured together and preferential killing was examined. The in-vitro data were compared with the in-vivo data obtained from a heterogeneous xenograft tumor model. The animal model of the artificial heterogeneous xenograft tumor showed that the nanotherapeutic was preferentially cytotoxic to KB cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Dendrimers/chemistry , Folic Acid/metabolism , Methotrexate/pharmacology , Nanostructures/chemistry , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Carriers/chemical synthesis , Drug Carriers/chemistry , Drug Delivery Systems , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , KB Cells/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
20.
Proc SPIE Int Soc Opt Eng ; 7188: 71880B-71880B12, 2009 Feb 16.
Article in English | MEDLINE | ID: mdl-20700480

ABSTRACT

A novel optical biosensor using a one-dimensional photonic crystal structure in a total-internal-reflection geometry (PC-TIR) is presented and investigated for label-free biosensing applications. This simple configuration forms a micro Fabry-Perot resonator in the top layer which provides a narrow optical resonance to enable label-free, highly sensitive measurements for the presence of analytes on the sensing surface or the refractive index change of the surrounding medium in the enhanced evanescent field; and at the same time it employs an open sensing surface for real-time biomolecular binding detection. The high sensitivity of the sensor was experimentally demonstrated by bulk solvent refractive index changes, ultrathin molecular films adsorbed on the sensing surface, and real-time analytes binding, measuring both the spectral shift of the photonic crystal resonance and the change of the intensity ratio in a differential reflectance measurement. Detection limits of 7x10(-8) RIU for bulk solvent refractive index, 6x10(-5) nm for molecular layer thickness and 24 fg/mm(2) for mass density were obtained, which represent a significant improvement relative to state-of-the-art surface-plasmon-resonance (SPR)-based systems. The PC-TIR sensor is thus seen to be a promising technology platform for high sensitivity and accurate biomolecular detection.

SELECTION OF CITATIONS
SEARCH DETAIL
...