Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
Expert Opin Drug Deliv ; 21(4): 537-551, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38568159

ABSTRACT

INTRODUCTION: The nose has been receiving increased attention as a route for drug delivery. As the site of deposition constitutes the first point of contact of the body with the drug, characterization of the regional deposition of intranasally delivered droplets or particles is paramount to formulation and device design of new products. AREAS COVERED: This review article summarizes the recent literature on intranasal regional drug deposition evaluated in vivo, in vitro and in silico, with the aim of correlating parameters measured in vitro with formulation and device performance. We also highlight the relevance of regional deposition to two emerging applications: nose-to-brain drug delivery and intranasal vaccines. EXPERT OPINION: As in vivo studies of deposition can be costly and time-consuming, researchers have often turned to predictive in vitro and in silico models. Variability in deposition is high due in part to individual differences in nasal geometry, and a complete predictive model of deposition based on spray characteristics remains elusive. Carefully selected or idealized geometries capturing population average deposition can be useful surrogates to in vivo measurements. Continued development of in vitro and in silico models may pave the way for development of less variable and more effective intranasal drug products.


Subject(s)
Administration, Intranasal , Computer Simulation , Drug Delivery Systems , Humans , Animals , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Vaccines/administration & dosage , Vaccines/pharmacokinetics , Nasal Mucosa/metabolism , Equipment Design , Models, Biological , Chemistry, Pharmaceutical/methods , Tissue Distribution , Nasal Cavity/metabolism
2.
ACS Appl Mater Interfaces ; 14(9): 11124-11143, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35227057

ABSTRACT

Antigen delivery through an oral route requires overcoming multiple challenges, including gastrointestinal enzymes, mucus, and epithelial tight junctions. Although each barrier has a crucial role in determining the final efficiency of the oral vaccination, transcytosis of antigens through follicle-associated epithelium (FAE) represents a major challenge. Most of the research is focused on delivering an antigen to the M-cell for FAE transcytosis because M-cells can easily transport the antigen from the luminal site. However, the fact is that the M-cell population is less than 1% of the total gastrointestinal cells, and most of the oral vaccines have failed to show any effect in clinical trials. To challenge the current dogma of M-cell targeting, in this study, we designed a novel tandem peptide with a FAE-targeting peptide at the front position and a cell-penetrating peptide at the back position. The tandem peptide was attached to a smart delivery system, which overcomes the enzymatic barrier and the mucosal barrier. The result showed that the engineered system could target the FAE (enterocytes and M-cells) and successfully penetrate the enterocytes to reach the dendritic cells located at the subepithelium dome. There was successful maturation and activation of dendritic cells in vitro confirmed by a significant increase in maturation markers such as CD40, CD86, presentation marker MHC I, and proinflammatory cytokines (TNF-α, IL-6, and IL-10). The in vivo results showed a high production of CD4+ T-lymphocytes (helper T-cell) and a significantly higher production of CD8+ T-lymphocytes (killer T-cell). Finally, the production of mucosal immunity (IgA) in the trachea, intestine, and fecal extracts and systemic immunity (IgG, IgG1, and IgG2a) was successfully confirmed. To the best of our knowledge, this is the first study that designed a novel tandem peptide to target the FAE, which includes M-cells and enterocytes rather than M-cell targeting and showed that a significant induction of both the mucosal and systemic immune response was achieved compared to M-cell targeting.


Subject(s)
Immunity, Mucosal/drug effects , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Administration, Oral , Animals , Antigens/immunology , Cell Line , Cell Survival/drug effects , Cytokines/metabolism , Female , Humans , Hydrogen-Ion Concentration , Immunity , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Mice, Inbred C57BL , Nanoparticles/toxicity , Ovalbumin/immunology , Peyer's Patches/immunology , Spleen/drug effects , Th1 Cells/metabolism , Th2 Cells , Vaccines/administration & dosage , Vaccines/chemical synthesis , Vaccines/chemistry , Vaccines/pharmacokinetics
3.
J Nanobiotechnology ; 19(1): 394, 2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34838057

ABSTRACT

BACKGROUND: Poly(D, L-lactic-co-glycolic acid) (PLGA) nanoparticles have potential applications as a vaccine adjuvant and delivery system due to its unique advantages as biodegradability and biocompatibility. EXPERIMENTAL: We fabricated cationic solid lipid nanoparticles using PLGA and dimethyl-dioctadecyl-ammonium bromide (DDAB), followed by loading of model antigen OVA (antigen ovalbumin, OVA257-264) to form an OVA@DDAB/PLGA nano-vaccine. And we investigated the intracellular signaling pathway in dendritic cells in vitro and antigen transport pathway and immune response in vivo mediated by an OVA@DDAB/PLGA nano-vaccine. RESULTS: In vitro experiments revealed that the antigen uptake of BMDCs after nanovaccine incubation was two times higher than pure OVA or OVA@Al at 12 h. The BMDCs were well activated by p38 MAPK signaling pathway. Furthermore, the nano-vaccine induced antigen escape from lysosome into cytoplasm with 10 times increased cross-presentation activity than those of OVA or OVA@Al. Regarding the transport of antigen into draining lymph nodes (LNs), the nano-vaccine could rapidly transfer antigen to LNs by passive lymphatic drainage and active DC transport. The antigen+ cells in inguinal/popliteal LNs for the nano-vaccine were increased over two folds comparing to OVA@Al and OVA at 12 h. Moreover, the antigen of nano-vaccine stayed in LNs for over 7 days, germinal center formation over two folds higher than those of OVA@Al and OVA. After immunization, the nano-vaccine induced a much higher ratio of IgG2c/IgG1 than OVA@Al. It also effectively activated CD4+ T, CD8+ T and B cells for immune memory with a strong cellular response. CONCLUSION: These results indicated that DDAB/PLGA NP was a potent platform to improve vaccine immunogenicity by p38 signaling pathway in BMDCs, enhancing transport of antigens to LNs, and higher immunity response.


Subject(s)
Antigen Presentation , Dendritic Cells , Nanostructures/chemistry , Signal Transduction , Vaccines , Adjuvants, Vaccine/chemistry , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Mice , Mice, Inbred BALB C , Ovalbumin/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Quaternary Ammonium Compounds/chemistry , Signal Transduction/drug effects , Signal Transduction/immunology , Vaccines/chemistry , Vaccines/immunology , Vaccines/pharmacokinetics , Vaccines/pharmacology
4.
J Pharm Pharm Sci ; 24: 267-276, 2021.
Article in English | MEDLINE | ID: mdl-34062113

ABSTRACT

PURPOSE: α-Pyrrolidinovalerophenone (α-PVP) is a second-generation synthetic cathinone which acts as an inhibitor at the dopamine and norepinephrine transporters in the brain. These novel studies determined the pharmacokinetics (PK) of α-PVP in rats and then evaluated the effects of an α-PVP vaccine on the PK profile. METHODS: Adult male Sprague-Dawley rats were randomly divided into treatment groups (n = 24/group) in which the vaccinated rats received an initial and two booster immunizations of the α-PVP vaccine at 0, 3, and 9 wks. Control rats received saline injections. α-PVP (0.56, 1, 3 mg/kg, sc) was then administered to both groups between 11-12 weeks and serum samples were collected for determination of α-PVP serum concentrations by LC-MS/MS (n=6 rats/treatment/time). At 13 weeks, brain, heart and kidney concentrations of α-PVP were determined by LC-MS/MS after administration of 1 mg/kg α-PVP (n=4-5 rats/treatment/time). RESULTS: PK values in control rats showed dose-dependent increases in maximum serum concentrations (Cmax) and area under the curve (AUCinf) values with an elimination half-life (t1/2) of approximately 2.1 h. α-PVP exhibited linear PK profile in control rats. Vaccinated rats had significantly (p<0.05) higher serum Cmax and AUCinf values than controls, and significantly reduced total body clearance, volume of distribution and t1/2 values. Vaccinated rats had significantly lower α-PVP concentrations in the brain, heart, and kidney in comparison to control rats at early time points. CONCLUSION: Vaccination with the novel α-PVP vaccine significantly altered serum PK leading to a time-dependent reduction in brain, kidney and heart concentrations of α-PVP compared to controls.


Subject(s)
Pyrrolidines/pharmacokinetics , Vaccines/pharmacokinetics , Animals , Brain/metabolism , Kidney/metabolism , Male , Myocardium/metabolism , Pyrrolidines/blood , Rats, Sprague-Dawley , Vaccination , Vaccines/blood
5.
J Nanobiotechnology ; 19(1): 106, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33858436

ABSTRACT

As a neglected tropical disease, Leishmaniasis is significantly instigating morbidity and mortality across the globe. Its clinical spectrum varies from ulcerative cutaneous lesions to systemic immersion causing hyperthermic hepato-splenomegaly. Curbing leishmanial parasite is toughly attributable to the myriad obstacles in existing chemotherapy and immunization. Since the 1990s, extensive research has been conducted for ameliorating disease prognosis, by resolving certain obstacles of conventional therapeutics viz. poor efficacy, systemic toxicity, inadequate drug accumulation inside the macrophage, scarce antigenic presentation to body's immune cells, protracted length and cost of the treatment. Mentioned hurdles can be restricted by designing nano-drug delivery system (nano-DDS) of extant anti-leishmanials, phyto-nano-DDS, surface modified-mannosylated and thiolated nano-DDS. Likewise, antigen delivery with co-transportation of suitable adjuvants would be achievable through nano-vaccines. In the past decade, researchers have engineered nano-DDS to improve the safety profile of existing drugs by restricting their release parameters. Polymerically-derived nano-DDS were found as a suitable option for oral delivery as well as SLNs due to pharmacokinetic re-modeling of drugs. Mannosylated nano-DDS have upgraded macrophage internalizing of nanosystem and the entrapped drug, provided with minimal toxicity. Cutaneous Leishmaniasis (CL) was tackling by the utilization of nano-DDS designed for topical delivery including niosomes, liposomes, and transfersomes. Transfersomes, however, appears to be superior for this purpose. The nanotechnology-based solution to prevent parasitic resistance is the use of Thiolated drug-loaded and multiple drugs loaded nano-DDS. These surfaces amended nano-DDS possess augmented IC50 values in comparison to conventional drugs and un-modified nano-DDS. Phyto-nano-DDS, another obscure horizon, have also been evaluated for their anti-leishmanial response, however, more intense assessment is a prerequisite. Impoverished Cytotoxic T-cells response followed by Leishmanial antigen proteins delivery have also been vanquished using nano-adjuvants. The eminence of nano-DDS for curtailment of anti-leishmanial chemotherapy and immunization associated challenges are extensively summed up in this review. This expedited approach is ameliorating the Leishmaniasis management successfully. Alongside, total to partial eradication of this disease can be sought along with associated co-morbidities.


Subject(s)
Antiprotozoal Agents/pharmacology , Leishmania/drug effects , Leishmaniasis/drug therapy , Nanotechnology/methods , Animals , Antiprotozoal Agents/therapeutic use , Drug Carriers/pharmacokinetics , Drug Delivery Systems , Drug Therapy , Humans , Liposomes/therapeutic use , Nanoparticles , Vaccination , Vaccines/pharmacokinetics
7.
Pharm Res ; 38(1): 141-153, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33449250

ABSTRACT

PURPOSE: Nasal delivery is a favorable route for vaccination against most respiratory infections, as antigen deposited in the nasal turbinate and Waldeyer's ring areas induce mucosal and systemic immune responses. However, little is known about the nasal distribution of the vaccines, specifically for infants. METHODS: Anatomical nasal replicas of five subjects, 3-24 months, were developed to assess local intranasal vaccine delivery using MAD Nasal™ device, and understand impact of breathing conditions and administration parameters. High performance liquid chromatography was used to quantify the deposition pattern and determine the delivery efficiency. RESULTS: The delivery efficiency on average for all models was found to be 86.57±14.23%. There were no significant differences in the total delivery efficiency between the models in all cases. However, the regional deposition pattern was altered based on the model and subsequent administration. Furthermore, removing the foam tip from the MAD Nasal™ device, to study the impact of insertion length, did not significantly increase the efficiency within the two models tested, 5- and 16-month. CONCLUSION: Incorporating nasal replicas in testing provided a benchmark to determine the efficiency of a common intranasal vaccine delivery combination product. This proposed platform would allow comparing other potential nasal vaccine delivery devices.


Subject(s)
Models, Anatomic , Nasal Mucosa/metabolism , Vaccination/methods , Vaccines/pharmacokinetics , Administration, Intranasal , Child, Preschool , Drug Evaluation, Preclinical/methods , Humans , Infant , Nose/anatomy & histology , Nose/diagnostic imaging , Printing, Three-Dimensional , Tomography, X-Ray Computed , Vaccines/administration & dosage
8.
Goiânia; SES-GO; 05 jan. 2021. 1-6 p. fig, ilus.
Non-conventional in Portuguese | LILACS, Coleciona SUS, CONASS, SES-GO | ID: biblio-1224581

ABSTRACT

Diversos países já aprovaram, em caráter emergencial ou definitivo, o uso de imunizantes para vacinação da população contra o novo coronavirus. Até o dia 05 de janeiro de 2021, ao menos 48 países já começaram a imunizar suas populações. Os últimos a entrarem na lista foram: República de Palau, Islândia, Singapura, Irlanda, Belarus e a Argentina (CNNBRASIL, 2021a). Com a vacinação da população já iniciada em diferentes países, também é relevante o acompanhamento das doses já administradas em razão da população do país e em números absolutos.


Several countries have already approved, on an emergency or definitive basis, the use of immunizations to vaccination of the population against the new coronavirus. As of January 5, 2021, at least 48 countries have begun immunizing their populations. The last to enter the list were: Republic of Palau, Iceland, Singapore, Ireland, Belarus and Argentina (CNNBRASIL, 2021a). With the vaccination of the population already initiated in different countries, it is also relevant the monitoring of doses already administered due to the population of the country and in absolute numbers.


Subject(s)
Humans , Male , Female , Pregnancy , Infant, Newborn , Infant , Child, Preschool , Child , Adolescent , Adult , Middle Aged , Aged , Aged, 80 and over , Young Adult , Vaccines/analysis , Vaccines/standards , Vaccines/pharmacokinetics , Coronavirus Infections/prevention & control
9.
Crit Rev Ther Drug Carrier Syst ; 37(2): 183-204, 2020.
Article in English | MEDLINE | ID: mdl-32865905

ABSTRACT

Nanotechnology provides an excellent platform for the development of a new generation of vaccines. These are based on purified subunit proteins or polysaccharides, recombinant proteins, synthetic peptides, or nucleic acids. These types of vaccines may be insufficiently immunogenic, thus requiring adjuvants that augment their immunogenicity. Nanoparticles (NPs) can act as adjuvants for vaccines, hence they are referred to as a nano-adjuvant (NA). NPs can either encapsulate or adsorb the vaccine antigen or DNA in an appropriate formulation, thus increasing stability, cellular uptake, and immunogenicity. In addition, the biodistribution and systemic release of a vaccine can also be controlled by different NA formulations. This review provides an overview of the classification of NAs and also addresses factors influencing the stability, release, and immunogenicity of the formulated vaccine. A basic understanding of these factors enables a more rational design of NA formulations. Applications of NAs and key challenges in their formulation development are also discussed.


Subject(s)
Adjuvants, Immunologic/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Vaccines/administration & dosage , Drug Development/methods , Drug Liberation , Drug Stability , Humans , Immunogenicity, Vaccine , Nanomedicine/methods , Tissue Distribution , Vaccines/immunology , Vaccines/pharmacokinetics
10.
Int J Mol Sci ; 21(14)2020 Jul 16.
Article in English | MEDLINE | ID: mdl-32708704

ABSTRACT

In an attempt to develop drug delivery systems that bypass the blood-brain barrier (BBB) and prevent liver and intestinal degradation, it was concluded that nasal medication meets these criteria and can be used for drugs that have these drawbacks. The aim of this review is to present the influence of the properties of chitosan and its derivatives (mucoadhesion, permeability enhancement, surface tension, and zeta potential) on the development of suitable nasal drug delivery systems and on the nasal bioavailability of various active pharmaceutical ingredients. Interactions between chitosan and proteins, lipids, antigens, and other molecules lead to complexes that have their own applications or to changing characteristics of the substances involved in the bond (conformational changes, increased stability or solubility, etc.). Chitosan and its derivatives have their own actions (antibacterial, antifungal, immunostimulant, antioxidant, etc.) and can be used as such or in combination with other molecules from the same class to achieve a synergistic effect. The applicability of the properties is set out in the second part of the paper, where nasal formulations based on chitosan are described (vaccines, hydrogels, nanoparticles, nanostructured lipid carriers (NLC), powders, emulsions, etc.).


Subject(s)
Chitosan/chemistry , Drug Carriers/chemistry , Drug Delivery Systems/methods , Pharmaceutical Preparations/administration & dosage , Vaccines/administration & dosage , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Biological Availability , Chitosan/metabolism , Chitosan/pharmacology , Drug Carriers/metabolism , Drug Carriers/pharmacology , Drug Compounding/methods , Humans , Vaccination/methods , Vaccines/pharmacokinetics
11.
Adv Drug Deliv Rev ; 158: 91-115, 2020.
Article in English | MEDLINE | ID: mdl-32598970

ABSTRACT

Vaccines are one of the most powerful technologies supporting public health. The adaptive immune response induced by immunization arises following appropriate activation and differentiation of T and B cells in lymph nodes. Among many parameters impacting the resulting immune response, the presence of antigen and inflammatory cues for an appropriate temporal duration within the lymph nodes, and further within appropriate subcompartments of the lymph nodes- the right timing and location- play a critical role in shaping cellular and humoral immunity. Here we review recent advances in our understanding of how vaccine kinetics and biodistribution impact adaptive immunity, and the underlying immunological mechanisms that govern these responses. We discuss emerging approaches to engineer these properties for future vaccines, with a focus on subunit vaccines.


Subject(s)
Vaccines/immunology , Vaccines/pharmacokinetics , Adjuvants, Immunologic/pharmacology , B-Lymphocytes/immunology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Humans , Immunity, Humoral/immunology , Inflammation Mediators/metabolism , Liposomes/metabolism , Lymph Nodes/immunology , Nanoparticles/metabolism , Plasmids/pharmacokinetics , RNA, Messenger/pharmacokinetics , T-Lymphocytes/immunology , Tissue Distribution
12.
Biomolecules ; 10(2)2020 02 17.
Article in English | MEDLINE | ID: mdl-32079263

ABSTRACT

: Antisense oligonucleotides (ASOs) are synthetically prepared short single-stranded deoxynucleotide sequences that have been validated as therapeutic agents and as a valuable tool in molecular driving biology. ASOs can block the expression of specific target genes via complementary hybridization to mRNA. Due to their high specificity and well-known mechanism of action, there has been a growing interest in using them for improving vaccine efficacy. Several studies have shown that ASOs can improve the efficacy of vaccines either by inducing antigen modification such as enhanced expression of immunogenic molecules or by targeting certain components of the host immune system to achieve the desired immune response. However, despite their extended use, some problems such as insufficient stability and low cellular delivery have not been sufficiently resolved to achieve effective and safe ASO-based vaccines. In this review, we analyze the molecular bases and the research that has been conducted to demonstrate the potential use of ASOs in vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity/drug effects , Oligonucleotides, Antisense/pharmacology , Adjuvants, Immunologic/pharmacokinetics , Animals , Humans , Oligonucleotides, Antisense/immunology , Oligonucleotides, Antisense/pharmacokinetics , Vaccination , Vaccines/immunology , Vaccines/pharmacokinetics , Vaccines/pharmacology
14.
Ther Deliv ; 10(11): 697-717, 2019 11.
Article in English | MEDLINE | ID: mdl-31789106

ABSTRACT

Nanogels have attracted considerable attention as nanoscopic drug carriers, particularly for site-specific or time-controlled delivery of bioactive mediators. A high diversity of polymer systems and the simple modification of their physicochemical features have provided multipurpose forms of nanogel preparations. Nanogels have outstandingly high stability, drug loading ability, biologic consistence, good permeation capability and can be responsive to environmental stimuli. Great potential has been shown by nanogels in many fields including delivery of genes, chemotherapy drugs, diagnosis, targeting of specific organs and several others. This review focuses mainly on different types of nanogels, methods of preparation including methods of drug loading, different modes of biodegradation mechanisms as well as main mechanisms of drug release from nanogels. Recent applications of nanogels are also briefly discussed and exemplified.


Subject(s)
Biocompatible Materials/chemistry , Drug Carriers/chemistry , Drug Compounding/methods , Nanogels/chemistry , Animals , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/pharmacokinetics , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Chemistry, Pharmaceutical , Cross-Linking Reagents/chemistry , Drug Liberation , Drug Stability , Emulsions , Gene Transfer Techniques , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , Micelles , Models, Animal , Particle Size , Polymerization , Solubility , Vaccines/administration & dosage , Vaccines/pharmacokinetics
15.
Arch Toxicol ; 93(10): 2787-2796, 2019 10.
Article in English | MEDLINE | ID: mdl-31522239

ABSTRACT

Aluminium (Al) toxicokinetics after intramuscular (IM) injection of Al-adjuvanted vaccines is unknown. Since animal data are required for modeling and extrapolation, a rat study was conducted measuring Al in plasma and tissues after IM injection of either plain Al-hydroxide (pAH) or Al-phosphate (pAP) adjuvant (Al dose 1.25 mg), single human doses of three Al-adjuvanted vaccines (V1, V2, and V3; Al doses 0.5-0.82 mg), or vehicle (saline). A significant increase in Al plasma levels compared to controls was observed after pAP (AUC(0-80 d), mean ± SD: 2424 ± 496 vs. 1744 ± 508 µg/L*d). Percentage of Al dose released from injected muscle until day 80 was higher after pAP (66.9%) and AP-adjuvanted V3 (85.5%) than after pAH and AH-adjuvanted V1 (0 and 22.3%, resp.). Estimated absolute Al release was highest for pAP (836.8 µg per rat). Al concentration in humerus bone was increased in all groups, again strongest in the pAP group [3.35 ± 0.39 vs. 0.05 ± 0.06 µg/g wet weight (ww)]. Extrapolated amounts in whole skeleton corresponded to 5-12% of the released Al dose. Very low brain Al concentrations were observed in all groups (adjuvant group means 0.14-0.29 µg/g ww; control 0.13 ± 0.04 µg/g ww). The results demonstrate systemically available Al from marketed vaccines in rats being mainly detectable in bone. Al release appears to be faster from AP- than AH-adjuvants. Dose scaling to human adults suggests that increase of Al in plasma and tissues after single vaccinations will be indistinguishable from baseline levels.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Compounds/administration & dosage , Aluminum Hydroxide/administration & dosage , Phosphates/administration & dosage , Vaccines/administration & dosage , Adjuvants, Immunologic/pharmacokinetics , Aluminum Compounds/pharmacokinetics , Aluminum Hydroxide/pharmacokinetics , Animals , Area Under Curve , Humans , Injections, Intramuscular , Male , Phosphates/pharmacokinetics , Rats , Rats, Wistar , Tissue Distribution , Vaccines/pharmacokinetics
16.
Adv Healthc Mater ; 8(15): e1900035, 2019 08.
Article in English | MEDLINE | ID: mdl-31165556

ABSTRACT

Efficient and timely delivery of vaccine antigens to the secondary lymphoid tissue is crucial to induce protective immune responses by vaccination. However, determining the longitudinal biodistribution of injected vaccines in the body has been a challenge. Here, the near-infrared (NIR) fluorescence imaging is reported that can efficiently enable the trafficking and biodistribution of vaccines in real time. Zwitterionic NIR fluorophores are conjugated on the surface of model vaccines and tracked the fate of bioconjugated vaccines after intradermal administration. Using an NIR fluorescence imaging system, it is possible to obtain time-course imaging of vaccine trafficking through the lymphatics, observing notable uptake in lymph nodes with minimal nonspecific tissue interactions. Flow cytometry analysis confirmed that the uptake in lymph nodes by antigen presenting cells was highly dependent on the hydrodynamic diameter of vaccines. These results demonstrate that the combination of a real-time NIR fluorescence imaging system and zwitterionic fluorophores is a powerful tool to determine the fate of vaccine antigens. Since such non-specific vaccine uptake causes serious adverse reactions, this method is not only useful for optimization of vaccine design, but also for safety evaluation of clinical vaccine candidates.


Subject(s)
Nanoparticles/chemistry , Spectroscopy, Near-Infrared/methods , Vaccines/pharmacokinetics , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/metabolism , Fluorescent Dyes/metabolism , Ions , Lymph Nodes/metabolism , Lymph Nodes/pathology , Mice , Models, Biological , Ovalbumin/chemistry , Ovalbumin/immunology , Quaternary Ammonium Compounds/chemistry , Silicon Dioxide/chemistry , Sulfonic Acids/chemistry , Tissue Distribution , Vaccines/chemistry , Vaccines/immunology
17.
Drug Dev Ind Pharm ; 45(6): 869-881, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30767577

ABSTRACT

Among lipid-based nanocarriers, multi-layered cochleates emerge as a novel delivery system because of prevention of oxidation of hydrophobic and hydrophilic drugs, enhancement in permeability, and reduction in dose of drugs. It also improves oral bioavailability and increases the safety of a drug by targeting at a specific site with less side effects. Nanostructured cochleates are used as a carrier for the delivery of water-insoluble or hydrophobic drugs of anticancer, antiviral and anti-inflammatory action. This review article focuses on different methods for preparation of cochleates, mechanism of formation of cochleates, mechanism of action like cochleate undergoes macrophagic endocytosis and release the drug into the systemic circulation by acting on membrane proteins, phospholipids, and receptors. Advanced methods such as calcium-substituted and ß-cyclodextrin-based cochleates, novel techniques include microfluidic and modified trapping method. Cochleates showed enhancement in oral bioavailability of amphotericin B, delivery of factor VII, oral mucosal vaccine adjuvant-delivery system, and delivery of volatile oil. In near future, cochleate will be one of the interesting delivery systems to overcome the stability and encapsulation efficiency issues associated with liposomes. The current limiting factors for commercial preparation of cochleates involve high cost of manufacturing, lack of standardization, and specialized equipments.


Subject(s)
Drug Compounding/methods , Drug Delivery Systems/methods , Lipid Bilayers/chemistry , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacokinetics , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Biological Availability , Calcium/chemistry , Chemistry, Pharmaceutical/methods , Drug Compounding/economics , Drug Stability , Microfluidics/methods , Particle Size , Vaccines/administration & dosage , Vaccines/chemistry , Vaccines/pharmacokinetics , beta-Cyclodextrins/chemistry
18.
Drug Dev Ind Pharm ; 45(6): 882-894, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30767591

ABSTRACT

Pulmonary infections are the most common cause of death globally. However, the development of mucosal vaccines that provide protective immunity against respiratory pathogens are limited. In contrast to needle-based vaccines, efficient vaccines that are delivered via noninvasive mucosal routes (such as via the lungs and nasal passage) produce both antigen-specific local mucosal IgA and systemic IgG protective antibodies. One major challenge in the development of pulmonary vaccines using subunit antigens however, is the production of optimal immune responses. Subunit vaccines therefore rely upon use of adjuvants to potentiate immune responses. While the lack of suitable mucosal adjuvants has hindered progress in the development of efficient pulmonary vaccines, particle-based systems can provide an alternative approach for the safe and efficient delivery of subunit vaccines. In particular, the rational engineering of particulate vaccines with optimal physicochemical characteristics can produce long-term protective immunity. These protect antigens against enzymatic degradation, target antigen presenting cells and initiate optimal humoral and cellular immunity. This review will discuss our current understanding of pulmonary immunology and developments in fabricating particle characteristics that may evoke potent and durable pulmonary immunity.


Subject(s)
Drug Delivery Systems/methods , Lung/metabolism , Respiratory Mucosa/metabolism , Respiratory Tract Infections/prevention & control , Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacokinetics , Administration, Inhalation , Administration, Intranasal/instrumentation , Animals , Dry Powder Inhalers , Feasibility Studies , Humans , Immunogenicity, Vaccine , Lung/immunology , Nanoparticles/chemistry , Particle Size , Permeability , Respiratory Mucosa/immunology , Respiratory Tract Infections/microbiology , Vaccines/immunology , Vaccines/pharmacokinetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics
19.
J Theor Biol ; 465: 51-55, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30639297

ABSTRACT

Current methods to optimize vaccine dose are purely empirically based, whereas in the drug development field, dosing determinations use far more advanced quantitative methodology to accelerate decision-making. Applying these established methods in the field of vaccine development may reduce the currently large clinical trial sample sizes, long time frames, high costs, and ultimately have a better potential to save lives. We propose the field of immunostimulation/immunodynamic (IS/ID) modelling, which aims to translate mathematical frameworks used for drug dosing towards optimizing vaccine dose decision-making. Analogous to Pharmacokinetic/Pharmacodynamic (PK/PD) modelling, the mathematical description of drug distribution (PK) and effect (PD) in host, IS/ID modelling approaches apply mathematical models to describe the underlying mechanisms by which the immune response is stimulated by vaccination (IS) and the resulting measured immune response dynamics (ID). To move IS/ID modelling forward, existing datasets and further data on vaccine allometry and dose-dependent dynamics need to be generated and collate, requiring a collaborative environment with input from academia, industry, regulators, governmental and non-governmental agencies to share modelling expertise, and connect modellers to vaccine data.


Subject(s)
Immunogenicity, Vaccine/immunology , Models, Immunological , Vaccination/methods , Vaccines/pharmacokinetics , Animals , Computer Simulation , Dose-Response Relationship, Drug , Humans , Reproducibility of Results , Vaccines/administration & dosage
20.
Biomaterials ; 192: 75-94, 2019 02.
Article in English | MEDLINE | ID: mdl-30439573

ABSTRACT

Vaccines are one of the greatest medical interventions of all time and have been successful in controlling and eliminating a myriad of diseases over the past two centuries. Among several vaccination strategies, mucosal vaccines have wide clinical applications and attract considerable interest in research, showing potential as innovative and novel therapeutics. In mucosal vaccination, targeting (microfold) M cells is a frontline prerequisite for inducing effective antigen-specific immunostimulatory effects. In this review, we primarily focus on materials engineered for use as vaccine delivery platforms to target M cells. We also describe potential M cell targeting areas, methods to overcome current challenges and limitations of the field. Furthermore, we present the potential of biomaterials engineering as well as various natural and synthetic delivery technologies to overcome the challenges of M cell targeting, all of which are absent in current literature. Finally, we briefly discuss manufacturing and regulatory processes to bring a robust perspective on the feasibility and potential of this next-generation vaccine technology.


Subject(s)
Drug Delivery Systems/methods , Intestinal Mucosa/metabolism , Peyer's Patches/metabolism , Vaccination/methods , Vaccines/administration & dosage , Animals , Biocompatible Materials/chemistry , Drug Carriers/chemistry , Humans , Vaccines/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...