Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Immunotherapy ; 14(7): 539-552, 2022 05.
Article in English | MEDLINE | ID: mdl-35196877

ABSTRACT

Aim: Epicutaneous immunotherapy (EPIT) with peanut has been demonstrated to be safe but efficacy may be limited by allergen uptake through the skin barrier. To enhance allergen uptake into the skin, the authors used peanut-coated microneedles and compared them with EPIT in a peanut allergy mouse model. Methods: Sensitized mice were treated with peanut-coated microneedles or peanut-EPIT and then challenged with peanut to determine protection. Results: Treatment with peanut-coated microneedles was safe and showed enhanced desensitization to peanut compared with peanut-EPIT administered via a similar schedule. Protection was associated with reduced Th2 immune responses and mast cell accumulation in the intestine. Conclusion: Peanut-coated microneedles have the potential to present a safe method of improving allergen delivery for cutaneous immunotherapy.


Epicutaneous immunotherapy (EPIT) with peanut has been demonstrated to be safe but efficacy has been varied. The tight barrier provided by the skin may limit the amount of allergen taken up through the skin and thus reduce efficacy. The authors evaluated a microneedle-based approach to improve the amount of allergen deposited into the skin to improve efficacy. Mice were made allergic to peanut and then treated with peanut-coated microneedles or peanut-EPIT. Mice were challenged with peanut to determine suppression of allergic reactivity. In mice, treatment with peanut-coated microneedles was safe and enhanced desensitization to peanut compared with peanut-EPIT administered via a similar schedule. Peanut-coated microneedles may present a novel method of improving allergen immunotherapy delivered through the skin.


Subject(s)
Allergens , Peanut Hypersensitivity , Animals , Arachis , Desensitization, Immunologic/methods , Humans , Mice , Peanut Hypersensitivity/therapy , Skin
2.
PLoS Comput Biol ; 17(10): e1009468, 2021 10.
Article in English | MEDLINE | ID: mdl-34648489

ABSTRACT

Understanding how immunological memory lasts a lifetime requires quantifying changes in the number of memory cells as well as how their division and death rates change over time. We address these questions by using a statistically powerful mixed-effects differential equations framework to analyze data from two human studies that follow CD8 T cell responses to the yellow fever vaccine (YFV-17D). Models were first fit to the frequency of YFV-specific memory CD8 T cells and deuterium enrichment in those cells 42 days to 1 year post-vaccination. A different dataset, on the loss of YFV-specific CD8 T cells over three decades, was used to assess out of sample predictions of our models. The commonly used exponential and bi-exponential decline models performed relatively poorly. Models with the cell loss following a power law (exactly or approximately) were most predictive. Notably, using only the first year of data, these models accurately predicted T cell frequencies up to 30 years post-vaccination. Our analyses suggest that division rates of these cells drop and plateau at a low level (0.1% per day, ∼ double the estimated values for naive T cells) within one year following vaccination, whereas death rates continue to decline for much longer. Our results show that power laws can be predictive for T cell memory, a finding that may be useful for vaccine evaluation and epidemiological modeling. Moreover, since power laws asymptotically decline more slowly than any exponential decline, our results help explain the longevity of immune memory phenomenologically.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Yellow Fever Vaccine/immunology , Yellow fever virus/immunology , Computational Biology , Humans , Models, Immunological
3.
Cornea ; 39(3): 362-369, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31724981

ABSTRACT

PURPOSE: Drug delivery directly to the corneal stroma currently relies on microscopic injections that demonstrate low reproducibility and clinician-dependent variability. With use of biological drugs such as adeno-associated viral (AAV) vectors, precise and consistent drug deposition is critical to reduce concerns related to off-target transduction and the host's immune response to the viral capsid and/or transgene-derived product. Therefore, a precise corneal injection (PCI) microneedle was designed to allow accurate depth-specific injections into the corneal stroma in a macroscopic setting. METHODS: High-frequency ultrasound and confocal microscopy demonstrated the consistent ability to predetermine the precise injection depth using PCI needles of varying sizes. Next, a comparison between a standard 31-G needle and PCI needles was performed in vivo using AAV vector gene delivery. RESULTS: Intrastromal corneal injections using the PCI microneedle resulted in less vector leakage at the site of injection and fewer anterior chamber penetrations compared with a standard 31-G needle. Although reporter gene expression appeared similar when the vector was administered with either needle type, a trend toward increased vector genomes was noted in the PCI-injected corneas at the experimental conclusion. As hypothesized, corneal perforation resulted in increased detection of AAV vector genomes in nontarget tissues, highlighting the importance of consistency for biological drug applications in the cornea. CONCLUSIONS: Further development of the PCI microneedle is warranted especially for AAV corneal gene therapy and offers the potential to enhance transduction while significantly reducing safety concerns and intraclinician and interclinician injection variability.


Subject(s)
Corneal Stroma/metabolism , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Green Fluorescent Proteins/genetics , Needles , Animals , Gene Expression , Gene Transfer Techniques , Injections, Intraocular , Male , Microscopy, Confocal , Rabbits , Reproducibility of Results , Swine , Ultrasonography
4.
PLoS One ; 10(3): e0120797, 2015.
Article in English | MEDLINE | ID: mdl-25785935

ABSTRACT

Human papilloma virus-like particles (HPV VLP) serve as the basis of the current licensed vaccines for HPV. We have previously shown that encapsidation of DNA expressing the model antigen M/M2 from respiratory syncytial virus (RSV) in HPV pseudovirions (PsV) is immunogenic when delivered intravaginally. Because the HPV capsids confer tropism for basal epithelium, they represent attractive carriers for vaccination targeted to the skin using microneedles. In this study we asked: 1) whether HPV16 VLP administered by microneedles could induce protective immune responses to HPV16 and 2) whether HPV16 PsV-encapsidated plasmids delivered by microneedles could elicit immune responses to both HPV and the antigen delivered by the transgene. Mice immunized with HPV16 VLP coated microneedles generated robust neutralizing antibody responses and were protected from HPV16 challenge. Microneedle arrays coated with HPV16-M/M2 or HPV16-F protein (genes of RSV) were then tested and dose-dependent HPV and F-specific antibody responses were detected post-immunization, and M/M2-specific T-cell responses were detected post RSV challenge, respectively. HPV16 PsV-F immunized mice were fully protected from challenge with HPV16 PsV and had reduced RSV viral load in lung and nose upon intranasal RSV challenge. In summary, HPV16 PsV-encapsidated DNA delivered by microneedles induced neutralizing antibody responses against HPV and primed for antibody and T-cell responses to RSV antigens encoded by the encapsidated plasmids. Although the immunogenicity of the DNA component was just above the dose response threshold, the HPV-specific immunity was robust. Taken together, these data suggest microneedle delivery of lyophilized HPV PsV could provide a practical, thermostable combined vaccine approach that could be developed for clinical evaluation.


Subject(s)
Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Plasmids/immunology , Skin/immunology , Uterine Cervical Neoplasms/prevention & control , Vaccination , Administration, Cutaneous , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , DNA, Viral/genetics , DNA, Viral/immunology , Female , Gene Expression , Genes, Reporter , Human papillomavirus 16/drug effects , Human papillomavirus 16/genetics , Human papillomavirus 16/immunology , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Microinjections , Needles , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/genetics , Plasmids/administration & dosage , Plasmids/genetics , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/immunology , Transgenes , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Viral Matrix Proteins/administration & dosage , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
5.
PLoS One ; 7(7): e41501, 2012.
Article in English | MEDLINE | ID: mdl-22848514

ABSTRACT

Recent studies have demonstrated the effectiveness of vaccine delivery to the skin by vaccine-coated microneedles; however there is little information on the effects of adjuvants using this approach for vaccination. Here we investigate the use of TLR ligands as adjuvants with skin-based delivery of influenza subunit vaccine. BALB/c mice received 1 µg of monovalent H1N1 subunit vaccine alone or with 1 µg of imiquimod or poly(I:C) individually or in combination via coated microneedle patches inserted into the skin. Poly(I:C) adjuvanted subunit influenza vaccine induced similar antigen-specific immune responses compared to vaccine alone when delivered to the skin by microneedles. However, imiquimod-adjuvanted vaccine elicited higher levels of serum IgG2a antibodies and increased hemagglutination inhibition titers compared to vaccine alone, suggesting enhanced induction of functional antibodies. In addition, imiquimod-adjuvanted vaccine induced a robust IFN-γ cellular response. These responses correlated with improved protection compared to influenza subunit vaccine alone, as well as reduced viral replication and production of pro-inflammatory cytokines in the lungs. The finding that microneedle delivery of imiquimod with influenza subunit vaccine induces improved immune responses compared to vaccine alone supports the use of TLR7 ligands as adjuvants for skin-based influenza vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aminoquinolines/pharmacology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/pharmacology , Poly I-C/pharmacology , Vaccination , Aminoquinolines/immunology , Animals , Antibodies, Viral/immunology , Antiviral Agents/immunology , Antiviral Agents/pharmacology , Female , Imiquimod , Immunoglobulin G/immunology , Influenza Vaccines/immunology , Injections, Intradermal , Mice , Mice, Inbred BALB C , Poly I-C/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacology
6.
Invest Ophthalmol Vis Sci ; 53(8): 4433-41, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22669719

ABSTRACT

PURPOSE: This study seeks to determine the intraocular pharmacokinetics of molecules and particles injected into the suprachoroidal space of the rabbit eye in vivo using a hollow microneedle. METHODS: Suprachoroidal injections of fluorescein and fluorescently tagged dextrans (40 and 250 kDa), bevacizumab, and polymeric particles (20 nm to 10 µm in diameter) were performed using microneedles in New Zealand white rabbits. The fluorescence intensity within the eye was monitored in each animal using an ocular fluorophotometer to determine the distribution of the injected material in the eye over time as compared with intravitreal injection of fluorescein. Fundus photography and histology were performed as well. RESULTS: Molecules and particles injected near the limbus using a microneedle flowed circumferentially around the eye within the suprachoroidal space. By targeting the suprachoroidal space, the concentration of injected materials was at least 10-fold higher in the back of the eye tissues than in anterior tissues. In contrast, intravitreal injection of fluorescein targeted the vitreous humor with no significant selectivity for posterior versus anterior segment tissues. Half-lives in the suprachoroidal space for molecules of molecular weight from 0.3 to 250 kDa ranged from 1.2 to 7.9 hours. In contrast, particles ranging in size from 20 nm to 10 µm remained primarily in the suprachoroidal space and choroid for a period of months and did not clear the eye. No adverse effects of injection into the suprachoroidal space were observed. CONCLUSION: Injection into the suprachoroidal space using a microneedle offers a simple and minimally invasive way to target the delivery of drugs to the choroid and retina.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Choroid/drug effects , Dextrans/administration & dosage , Drug Delivery Systems , Fluorescein/administration & dosage , Fluoresceins/administration & dosage , Posterior Eye Segment/drug effects , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Bevacizumab , Choroid/metabolism , Dextrans/pharmacokinetics , Fluorescein/pharmacokinetics , Fluoresceins/pharmacokinetics , Fluorescence , Fluorophotometry , Injections, Intraocular , Needles , Posterior Eye Segment/metabolism , Rabbits
7.
Sci Rep ; 2: 357, 2012.
Article in English | MEDLINE | ID: mdl-22500210

ABSTRACT

Influenza infection represents a major socio-economic burden worldwide. Novel delivery methods can render influenza vaccination easier and more acceptable by the public, and importantly confer protection equal or superior to that induced by conventional systemic administration. An attractive target for vaccine delivery is the skin. Recent studies have demonstrated improved immune responses after transdermal delivery of inactivated influenza virus with microneedle patches. Here we show that immunization with a licensed influenza subunit vaccine coated on metal microneedles can activate both humoral and cellular arms of the immune response and confer improved long-term protection in the mouse model when compared to the conventional systemic route of delivery. These results demonstrate the promising potential of microneedle delivery of licensed influenza subunit vaccines, that could be beneficial in increasing vaccine coverage and protection and reducing influenza-related mortality worldwide.


Subject(s)
Antibodies, Viral/biosynthesis , Immunity, Cellular , Influenza Vaccines/administration & dosage , Needles , Skin , Animals , Cell Line , Dogs , Enzyme-Linked Immunosorbent Assay , Female , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C
8.
J Control Release ; 160(3): 561-9, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22516089

ABSTRACT

Microneedle delivery of nucleic acids, in particular plasmid DNA (pDNA), to the skin represents a potential new approach for the clinical management of genetic skin diseases and cutaneous cancers, and for intracutaneous genetic immunisation. In this study excised human skin explants were used to investigate and optimise key parameters that will determine stable and effective microneedle-facilitated pDNA delivery. These include (i) high dose-loading of pDNA onto microneedle surfaces, (ii) stability and functionality of the coated pDNA, (iii) skin penetration capability of pDNA-coated microneedles, and (iv) efficient gene expression in human skin. Optimisation of a dip-coating method enabled significant increases in the loading capacity, up to 100µg of pDNA per 5-microneedle array. Coated microneedles were able to reproducibly perforate human skin at low (<1N) insertion forces. The physical stability of the coated pDNA was partially compromised on storage, although this was improved through the addition of saccharide excipients without detriment to the biological functionality of pDNA. The pDNA-coated microneedles facilitated reporter gene expression in viable human skin. The efficiency of gene expression from coated microneedles will depend upon suitable DNA loading, efficient and reproducible skin puncture and rapid in situ dissolution of the plasmid at the site of delivery.


Subject(s)
DNA/administration & dosage , Needles , Skin/metabolism , Transfection/methods , Gene Expression , Green Fluorescent Proteins/genetics , Humans , Microinjections , Plasmids
9.
mBio ; 3(2): e00012-12, 2012.
Article in English | MEDLINE | ID: mdl-22396479

ABSTRACT

UNLABELLED: Microneedle patches (MN) provide a novel method of vaccine delivery to the skin with the objective of targeting the large network of resident antigen-presenting cells to induce an efficient immune response. Our previous reports demonstrated that cutaneous delivery of inactivated influenza virus-coated MN to mice protects against lethal infection. Protection is correlated with sustained levels of anti-influenza virus serum antibodies, hemagglutination inhibition titers, and robust cellular responses that are often stronger than those generated by intramuscular vaccination. Here we dissect the early events occurring in murine skin after microneedle delivery of inactivated influenza virus. We demonstrate correlation of immunization against influenza virus with a local increase of cytokines important for recruitment of neutrophils, monocytes and dendritic cells at the site of immunization. We also observed prolonged antigen deposition, and migration of matured dendritic cells bearing influenza virus antigen from the skin. IMPORTANCE: The immunological mechanisms by which MN vaccination confers protective immunity are not well understood. The present study provides a first analysis of the early immune events after microneedle-based vaccination.


Subject(s)
Immunization/methods , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Injections, Intradermal/methods , Skin/immunology , Animals , Cytokines/metabolism , Dendritic Cells/immunology , Female , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Nude , Monocytes/immunology , Neutrophils/immunology , Skin/pathology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Whole Body Imaging
10.
Int J Pharm ; 423(2): 257-63, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22172290

ABSTRACT

Peptides and polypeptides have important pharmacological properties but only a limited number have been exploited as therapeutics because of problems related to their delivery. Most of these drugs require a parenteral delivery system which introduces the problems of pain, possible infection, and expertise required to carry out an injection. The aim of this study was to develop a transdermal patch containing microneedles (MNs) coated with a peptide drug, salmon calcitonin (sCT), as an alternative to traditional subcutaneous and nasal delivery routes. Quantitative analysis of sCT after coating and drying onto microneedles was performed with a validated HPLC method. In vivo studies were carried out on hairless rats and serum levels of sCT were determined by ELISA. The AUC value of MNs coated with a trehalose-containing formulation (250 ± 83 ng/mL min) was not significantly different as compared to subcutaneous injections (403 ± 253 ng/mL min), but approximately 13 times higher than nasal administration (18.4 ± 14.5 ng/mL min). T(max) (7.5 ± 5 min) values for MN mediated administration were 50% shorter than subcutaneous injections (15 min), possibly due to rapid sCT dissolution and absorption by dermal capillaries. These results suggest that with further optimization of coating formulations, microneedles may enable administration of sCT and other peptides without the need for hypodermic injections.


Subject(s)
Calcitonin/administration & dosage , Coated Materials, Biocompatible , Drug Carriers , Administration, Intranasal , Animals , Biological Availability , Calcitonin/blood , Calcitonin/chemistry , Calcitonin/pharmacokinetics , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Equipment Design , Injections, Intradermal , Injections, Intravenous , Injections, Subcutaneous , Male , Microinjections , Miniaturization , Needles , Rats , Rats, Hairless , Rats, Sprague-Dawley , Solubility , Technology, Pharmaceutical/methods , Transdermal Patch
11.
J Infect Dis ; 204(4): 582-91, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21685355

ABSTRACT

BACKGROUND: A major goal in influenza vaccine development is induction of serological memory and cellular responses to confer long-term protection and limit virus spread after infection. Here, we investigate induction of long-lived immunity against the 2009 H1N1 virus after skin vaccination. METHODS: BALB/c mice received a single dose of 5 µg inactivated A/California/04/09 virus via coated metal microneedles (MN) applied to skin or via subcutaneous injection. RESULTS: MN or subcutaneous vaccination elicited similar serum IgG and hemagglutination inhibition titers and 100% protection against lethal viral challenge 6 weeks after vaccination. Six months after vaccination, the subcutaneous group exhibited a 60% decrease in functional antibody titers and extensive lung inflammation after challenge with 10 × LD(50) of homologous virus. In contrast, the MN group maintained high functional antibody titers and IFN-γ levels, inhibition of viral replication, and no signs of lung inflammation after challenge. MN vaccination conferred complete protection against lethal challenge, whereas subcutaneous vaccination induced only partial protection. These findings were further supported by high numbers of bone marrow plasma cells and spleen antibody-secreting cells detected in the MN group. CONCLUSIONS: A single skin vaccination with MN induced potent long-lived immunity and improved protection against the 2009 H1N1 influenza virus, compared with subcutaneous injection.


Subject(s)
Immunity, Humoral/physiology , Immunity, Mucosal/physiology , Immunologic Memory , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Viral/physiology , Bone Marrow Cells/physiology , Cell Line , Dogs , Female , Influenza Vaccines/administration & dosage , Injections, Intradermal , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Spleen/physiology , Virus Replication
12.
Clin Vaccine Immunol ; 18(4): 647-54, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21288996

ABSTRACT

The emergence of the swine-origin 2009 influenza pandemic illustrates the need for improved vaccine production and delivery strategies. Skin-based immunization represents an attractive alternative to traditional hypodermic needle vaccination routes. Microneedles (MNs) can deliver vaccine to the epidermis and dermis, which are rich in antigen-presenting cells (APC) such as Langerhans cells and dermal dendritic cells. Previous studies using coated or dissolvable microneedles emphasized the use of inactivated influenza virus or virus-like particles as skin-based vaccines. However, most currently available influenza vaccines consist of solubilized viral protein antigens. Here we test the hypothesis that a recombinant subunit influenza vaccine can be delivered to the skin by coated microneedles and can induce protective immunity. We found that mice vaccinated via MN delivery with a stabilized recombinant trimeric soluble hemagglutinin (sHA) derived from A/Aichi/2/68 (H3) virus had significantly higher immune responses than did mice vaccinated with unmodified sHA. These mice were fully protected against a lethal challenge with influenza virus. Analysis of postchallenge lung titers showed that MN-immunized mice had completely cleared the virus from their lungs, in contrast to mice given the same vaccine by a standard subcutaneous route. In addition, we observed a higher ratio of antigen-specific Th1 cells in trimeric sHA-vaccinated mice and a greater mucosal antibody response. Our data therefore demonstrate the improved efficacy of a skin-based recombinant subunit influenza vaccine and emphasize the advantage of this route of vaccination for a protein subunit vaccine.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Vaccination/methods , Animals , Disease Models, Animal , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Immunity, Mucosal , Influenza Vaccines/administration & dosage , Injections, Intradermal/methods , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Survival Analysis , Th1 Cells/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Load
13.
Int J Pharm ; 407(1-2): 95-104, 2011 Apr 04.
Article in English | MEDLINE | ID: mdl-21272628

ABSTRACT

Microdermabrasion has been shown to increase skin permeability for transdermal drug delivery by damaging or removing skin's outer layer, stratum corneum. However, relationships between microdermabrasion parameters and effects on the stratum corneum barrier have not been developed. In this study, we determined the effect of microdermabrasion crystal flow rate, time, and suction pressure applied in both static and dynamic modes on the extent of stratum corneum removal from excised porcine skin. In addition to controlling the depth of tissue removal by microdermabrasion parameters, we also controlled the area of tissue removal by applying a metal mask patterned with 125- or 250-µm holes to selectively expose small spots of tissue to microdermabrasion. We found that the extent of stratum corneum removal depended strongly on the crystal flow rate and exposure time and only weakly on pressure or static/dynamic mode operation. Masking the skin was effective to localize stratum corneum removal to exposed sites. Overall, this study demonstrates that optimized microdermabrasion in combination with a mask can be used to selectively remove stratum corneum with three-dimensional control, which is important to translating this technique into a novel method of transdermal drug delivery.


Subject(s)
Dermabrasion/methods , Drug Delivery Systems , Skin Absorption , Administration, Cutaneous , Animals , Dermabrasion/instrumentation , Permeability , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/metabolism , Skin/metabolism , Swine , Time Factors
14.
Nat Med ; 16(8): 915-20, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20639891

ABSTRACT

Influenza prophylaxis would benefit from a vaccination method enabling simplified logistics and improved immunogenicity without the dangers posed by hypodermic needles. Here we introduce dissolving microneedle patches for influenza vaccination using a simple patch-based system that targets delivery to skin's antigen-presenting cells. Microneedles were fabricated using a biocompatible polymer encapsulating inactivated influenza virus vaccine for insertion and dissolution in the skin within minutes. Microneedle vaccination generated robust antibody and cellular immune responses in mice that provided complete protection against lethal challenge. Compared to conventional intramuscular injection, microneedle vaccination resulted in more efficient lung virus clearance and enhanced cellular recall responses after challenge. These results suggest that dissolving microneedle patches can provide a new technology for simpler and safer vaccination with improved immunogenicity that could facilitate increased vaccination coverage.


Subject(s)
Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Needles , Polymers/administration & dosage , Vaccination/methods , Administration, Cutaneous , Adsorption , Animals , Antibody Formation/physiology , Dosage Forms , Humans , Immunization, Secondary/methods , Influenza Vaccines/pharmacokinetics , Injections, Intradermal , Mice , Needles/statistics & numerical data , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , Polymers/pharmacokinetics , Vaccination/instrumentation
15.
Proc Natl Acad Sci U S A ; 106(19): 7968-73, 2009 May 12.
Article in English | MEDLINE | ID: mdl-19416832

ABSTRACT

Influenza prophylaxis would benefit from a simple method to administer influenza vaccine into skin without the need for hypodermic needles. In this study, solid metal microneedle arrays (MNs) were investigated as a system for cutaneous vaccine delivery using influenza virus antigen. The MNs with 5 monument-shaped microneedles per array were produced and coated with inactivated influenza virus A/PR/8/34 (IIV). As much as 10 microg of viral proteins could be coated onto an array of 5 microneedles, and the coated IIV was delivered into skin at high efficiency within minutes. The coated MNs were used to immunize mice in comparison with conventional intramuscular injection at the same dose. Analysis of immune responses showed that a single immunization with IIV-coated MNs induced strong antibody responses against influenza virus, with significant levels of hemagglutination inhibition activities (>1:40), which were comparable to those induced by conventional intramuscular immunization. Moreover, mice immunized by a single dose of IIV coated on MNs were effectively protected against lethal challenge by a high dose of mouse-adapted influenza virus A/PR/8/34. These results show that MNs are highly effective as a simple method of vaccine delivery to elicit protective immune responses against virus infection.


Subject(s)
Immunization/methods , Influenza Vaccines/administration & dosage , Orthomyxoviridae/immunology , Administration, Cutaneous , Animals , Antibodies/chemistry , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Hemagglutination , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A virus/immunology , Mice , Mice, Inbred BALB C , Vaccines/chemistry
16.
PLoS One ; 4(3): e4773, 2009.
Article in English | MEDLINE | ID: mdl-19274084

ABSTRACT

BACKGROUND: Influenza is a contagious disease caused by a pathogenic virus, with outbreaks all over the world and thousands of hospitalizations and deaths every year. Due to virus antigenic drift and short-lived immune responses, annual vaccination is required. However, vaccine coverage is incomplete, and improvement in immunization is needed. The objective of this study is to investigate a novel method for transdermal delivery using metal microneedle arrays (MN) coated with inactivated influenza virus to determine whether this route is a simpler and safer approach than the conventional immunization, capable to induce robust immune responses and confer protection against lethal virus challenge. METHODOLOGY/PRINCIPAL FINDINGS: Inactivated A/Aichi/2/68 (H3N2) influenza virus was coated on metal microneedle arrays and applied to mice as a vaccine in the caudal dorsal skin area. Substantial antibody titers with hemagglutination inhibition activity were detected in sera collected two and four weeks after a single vaccine dose. Challenge studies in mice with 5 x LD(50) of mouse adapted Aichi virus demonstrated complete protection. Microneedle vaccination induced a broad spectrum of immune responses including CD4+ and CD8+ responses in the spleen and draining lymph node, a high frequency of antigen-secreting cells in the lung and induction of virus-specific memory B-cells. In addition, the use of MN showed a dose-sparing effect and a strong Th2 bias when compared to an intramuscular (IM) reference immunization. CONCLUSIONS/SIGNIFICANCE: The present results show that delivery of inactivated influenza virus through the skin using metal microneedle arrays induced strong humoral and cellular immune responses capable of conferring protection against virus challenge as efficiently as intramuscular immunization, which is the standard vaccination route. In view of the convenience of delivery and the potential for self-administration, vaccine-coated metal microneedles may provide a novel and highly effective immunization method.


Subject(s)
Administration, Cutaneous , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Needles , Vaccination/methods , Animals , Antibody Formation/drug effects , Equipment Design , Immunity, Cellular/drug effects , Mice
17.
Biophys J ; 95(9): 4124-38, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18676653

ABSTRACT

Cells exposed to acoustic cavitation and other mechanical stresses can be transiently permeabilized to permit intracellular uptake of molecules, including drugs, proteins, and genes. Microscopic imaging and other studies suggest that intracellular loading occurs through plasma membrane wounds of submicrometer radius that reseal over time through the aggregation and fusion of lipid vesicles trafficked to the wound site. The goal of this study was to 1), determine the size of membrane wounds as a function of time after in vitro sonication of DU145 prostate cancer cells under conditions that caused extensive acoustic cavitation; and 2), theoretically model transport processes leading to intracellular loading. Our overall hypothesis was that intracellular loading is governed by passive diffusion through porous membrane wounds of up to 300-nm radius containing pores that permit entry of molecules up to at least 28-nm radius over a timescale of minutes. Experimental measurements showed intracellular loading of molecules with radii from 0.6 to 28 nm, where most loading occurred after sonication over a timescale up to minutes and where smaller molecules were taken up to a greater extent and over a longer timescale than larger molecules. Theoretical modeling predicted that membrane wounds would have a 300-nm radius initially and then would shrink, with a half life of 20 to 50 s. Uptake was shown to occur predominantly by diffusion and the increasing levels of uptake with decreasing molecular size was explained primarily by differences in molecular diffusivity and, for the largest molecule, geometrical hindrance within the wound. Mathematical modeling was simplified, because transport through porous wounds of possibly complex internal nanostructure was governed largely by transport at the edge of the wound, and depended only weakly on the size, number, and distribution of nanopores within the wound under the conditions relevant to this study. Overall, this study developed a theoretical framework for analysis of transmembrane transport through cell membrane wounds and thereby provided quantitative estimates of their size and lifetime.


Subject(s)
Acoustics , Cell Membrane/metabolism , Models, Molecular , Cell Line, Tumor , Cell Membrane Permeability , Diffusion , Humans , Intracellular Space/metabolism , Porosity , Stress, Mechanical , Time Factors
18.
Biomed Microdevices ; 10(2): 299-308, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17994280

ABSTRACT

We report on development and experimental characterization of a novel cell manipulation device-the electrosonic ejector microarray-which establishes a pathway for drug and/or gene delivery with control of biophysical action on the length scale of an individual cell. The device comprises a piezoelectric transducer for ultrasound wave generation, a reservoir for storing the sample mixture and a set of acoustic horn structures that form a nozzle array for focused application of mechanical energy. The nozzles are micromachined in silicon or plastic using simple and economical batch fabrication processes. When the device is driven at a particular resonant frequency of the acoustic horn structures, the sample mixture of cells and desired transfection agents/molecules suspended in culture medium is ejected from orifices located at the nozzle tips. During sample ejection, focused mechanical forces (pressure and shear) are generated on a microsecond time scale (dictated by nozzle size/geometry and ejection velocity) resulting in identical "active" microenvironments for each ejected cell. This process enables a number of cellular bioeffects, from uptake of small molecules and gene delivery/transfection to cell lysis. Specifically, we demonstrate successful calcein uptake and transfection of DNA plasmid encoding green fluorescent protein (GFP) into human malignant glioma cells (cell line LN443) using electrosonic microarrays with 36, 45 and 50 mum diameter nozzle orifices and operating at ultrasound frequencies between 0.91 and 0.98 MHz. Our results suggest that efficacy and the extent of bioeffects are mainly controlled by nozzle orifice size and the localized intensity of the applied acoustic field.


Subject(s)
Acoustics/instrumentation , Cell Separation/instrumentation , Drug Delivery Systems/instrumentation , Injections, Jet/instrumentation , Microarray Analysis/instrumentation , Microfluidic Analytical Techniques/instrumentation , Transfection/instrumentation , Cell Separation/methods , Drug Delivery Systems/methods , Equipment Design , Equipment Failure Analysis , Injections, Jet/methods , Microarray Analysis/methods , Micromanipulation/instrumentation , Micromanipulation/methods , Transfection/methods
19.
Technol Cancer Res Treat ; 6(5): 433-42, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17877433

ABSTRACT

Treatment of brain cancer is limited in part by inefficient intracellular delivery of drugs and DNA for chemotherapy and gene therapy, respectively. This study tested the hypothesis that ultrasound may be used to enhance intracellular delivery and efficacy of chemotherapeutics and genes in glioma cells in vitro. First, suitable ultrasound conditions were identified by measuring intracellular uptake of calcein and viability of GS 9L rat gliosarcoma cells after a range of different ultrasound exposures. We selected sonication at 10 J/cm2, which achieved intracellular delivery of approximately 10(6) molecules/cell. Next, glial cells were sonicated with varying concentrations of model chemotherapeutics: BCNU and bleomycin. For both drugs, cytotoxicity was increased in a synergistic manner when accompanied by ultrasound exposure. Finally, expression of a plasmid DNA encoding a GFP reporter was increased up to 30-fold when exposed to ultrasound. Altogether, these findings suggest that ultrasound may be useful to increase the efficacy of chemotherapy and gene therapy of glioma cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Genetic Therapy/methods , Gliosarcoma/therapy , Ultrasonic Therapy , Animals , Bleomycin/administration & dosage , Carmustine/administration & dosage , Cell Survival , Combined Modality Therapy , DNA/administration & dosage , Dose-Response Relationship, Drug , Gene Transfer Techniques , Green Fluorescent Proteins/administration & dosage , Green Fluorescent Proteins/genetics , Rats , Transfection , Tumor Cells, Cultured
20.
Langmuir ; 23(11): 6245-51, 2007 May 22.
Article in English | MEDLINE | ID: mdl-17439250

ABSTRACT

We investigate physical processes taking place during nanoscale mechanosensing of soft biological membranes in liquid environments. Examples include tapping mode imaging by atomic force microscope (AFM) and microscopy based on the Brownian motion of a nanoparticle in an optical-tweezers-controlled trap. The softness and fluidity of the cellular membrane make it difficult to accurately detect (i.e., image) the shape of the cell using traditional mechanosensing methods. The aim of the reported work is to theoretically evaluate whether the drag force acting on the nanoscale mechanical probe due to a combined effect of intra- and extracellular environments can be exploited to develop a new imaging mode suitable for soft cellular interfaces. We approach this problem by rigorous modeling of the fluid mechanics of a complex viscoelastic biosystem in which the probe sensing process is intimately coupled to the membrane biomechanics. The effects of the probe dimensions and elastic properties of the membrane as well as intra- and extracellular viscosities are investigated in detail to establish the structure and evolution of the fluid field as well as the dynamics of membrane deformation. The results of numerical simulations, supported by predictions of the scaling analysis of forces acting on the probe, suggest that viscous drag is the dominant force dictating the probe dynamics as it approaches a biological interface. The increase in the drag force is shown to be measurable, to scale linearly with an increase in the viscosity ratio of the fluids on either side of the membrane, and to be inversely proportional to the probe-to-membrane distance. This leads to the postulation of a new strategy for lipid membrane imaging by AFM or other mechanosensing methods using a variation in the maximum drag force as an indicator of the membrane position.


Subject(s)
Biosensing Techniques/methods , Cell Membrane/chemistry , Biomechanical Phenomena , Cell Membrane/metabolism , Lipid Bilayers/chemistry , Membrane Lipids/chemistry , Microscopy, Atomic Force , Models, Biological , Nanotechnology
SELECTION OF CITATIONS
SEARCH DETAIL
...