Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Langmuir ; 33(3): 773-782, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28006902

RESUMO

Immunoassays are ubiquitous across research and clinical laboratories, yet little attention is paid to the effect of the substrate material on the assay performance characteristics. Given the emerging interest in wearable immunoassay formats, investigations into substrate materials that provide an optimal mix of mechanical and bioanalytical properties are paramount. In the course of our research in developing wearable immunoassays which can penetrate skin to selectively capture disease antigens from the underlying blood vessels, we recently identified significant differences in immunoassay performance between gold and polycarbonate surfaces, even with a consistent surface modification procedure. We observed significant differences in PEG density, antibody immobilization, and nonspecific adsorption between the two substrates. Despite a higher PEG density formed on gold-coated surfaces than on amine-functionalized polycarbonate, the latter revealed a higher immobilized capture antibody density and lower nonspecific adsorption, leading to improved signal-to-noise ratios and assay sensitivities. The major conclusion from this study is that in designing wearable bioassays or biosensors, the design and its effect on the antifouling polymer layer can significantly affect the assay performance in terms of analytical specificity and sensitivity.


Assuntos
Ensaio de Imunoadsorção Enzimática/instrumentação , Polietilenoglicóis/química , Adsorção , Animais , Ouro/química , Imunoglobulina G/química , Camundongos , Cimento de Policarboxilato/química , Silício/química , Propriedades de Superfície
2.
Anal Chem ; 86(20): 10474-83, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25232916

RESUMO

Herein we demonstrate the use of a wearable device that can selectively capture two distinct circulating protein biomarkers (recombinant P. falciparum rPfHRP2 and total IgG) from the intradermal fluid of live mice in situ, for subsequent detection in vitro. The device comprises a microprojection array that, when applied to the skin, penetrates the outer skin layers to interface directly with intradermal fluid. Because of the complexity of the biological fluid being sampled, we investigated the effects of solution conditions on the attachment of capture antibodies, to optimize the assay detection limit both in vitro and on live mice. For detection of the target antigen diluted in 20% serum, immobilization conditions favoring high antibody surface density (low pH, low ionic strength) resulted in 100-fold greater sensitivity in comparison to standard conditions, yielding a detection limit equivalent to the plate enzyme-linked immunosorbent assay (ELISA). We also show that blocking the device surface to reduce nonspecific adsorption of target analyte and host proteins does not significantly change sensitivity. After injecting mice with rPfHRP2 via the tail vein, we compared analyte levels in both plasma and skin biopsies (cross-sectional area same as the microprojection array), observing that skin samples contained the equivalent of ∼8 µL of analyte-containing plasma. We then applied the arrays to mice, showing that surfaces coated with a high density of antibodies captured a significant amount of the rPfHRP2 target while the standard surface showed no capture in comparison to the negative control. Next, we applied a triplex device to both control and rPfHRP2-treated mice, simultaneously capturing rPfHRP2 and total IgG (as a positive control for skin penetration) in comparison to a negative control device. We conclude that such devices can be used to capture clinically relevant, circulating protein biomarkers of infectious disease via the skin, with potential applications as a minimally invasive and lab-free biomarker detection platform.


Assuntos
Biomarcadores/sangue , Análise Química do Sangue/métodos , Malária Falciparum/diagnóstico , Plasmodium falciparum/química , Adesivo Transdérmico , Animais , Ensaio de Imunoadsorção Enzimática , Camundongos
3.
Anal Chem ; 85(21): 10196-204, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24083844

RESUMO

Herein, we report the fabrication, characterization, and testing of a polymer microprojection array, for the direct and selective capture of circulating biomarkers from the skin of live mice. First, we modified polycarbonate wafers using an electrophilic aromatic substitution reaction with nitric acid to insert aromatic nitro-groups into the benzene rings, followed by treatment with sodium borohydride to reduce the nitro-groups to primary amines. Initial characterization by ultraviolet-visible (UV-vis) spectroscopy suggested that increasing acid concentration led to increased depth of material modification and that this was associated with decreased surface hardness and slight changes in surface roughness. Chemical analysis with X-ray photoelectron spectroscopy (XPS) and attenuated total reflectance fourier transform infrared (ATR-FT-IR) spectroscopy showed nitrogen species present at the surface for all acid concentrations used, but subsurface nitrogen species were only observed at acid concentrations >35%. The nitrogen species were identified as a mixture of nitro, imine, and amine groups, and following reduction, there was sufficient amounts of primary amine groups for covalent attachment of a polyethylene glycol antifouling layer and protein capture probes, as determined by colorimetric and radiometric assays. Finally, the modification scheme was applied to polycarbonate microprojection arrays, and we show that these devices achieve flank skin penetration depths and biomarker yields comparable with our previously reported gold-coated silicon arrays, with very low nonspecific binding even in 10% mouse serum (in vitro) or directly in mouse skin (in vivo). This study is the first demonstration showing the potential utility of polymer microprojections in immunodiagnostics applications.


Assuntos
Biomarcadores/sangue , Animais , Biomarcadores/metabolismo , Ensaio de Imunoadsorção Enzimática , Camundongos , Espectroscopia Fotoeletrônica , Pele/metabolismo , Espectrofotometria Ultravioleta , Espectroscopia de Infravermelho com Transformada de Fourier , Propriedades de Superfície
4.
Lab Chip ; 10(20): 2655-8, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20820632

RESUMO

Minimally invasive biosensors are of great interest for rapid detection of disease biomarkers for diagnostic screening at the point-of-care. Here we introduce a device which extracts disease-specific biomarkers directly from the upper dermis, without the needle and syringe or resource-intensive blood processing. Using antigen-specific antibodies raised in mice as a model system, we confirm the analytical specificity and sensitivity of the antibody capture and extraction in comparison to the conventional methods based on needle/syringe blood draw followed by processing and antigen-specific ELISAs.


Assuntos
Biomarcadores/análise , Técnicas Biossensoriais/instrumentação , Coleta de Amostras Sanguíneas/instrumentação , Imunoensaio/instrumentação , Técnicas Analíticas Microfluídicas/instrumentação , Análise Serial de Proteínas/instrumentação , Pele/metabolismo , Animais , Ação Capilar , Desenho de Equipamento , Análise de Falha de Equipamento , Camundongos , Camundongos Endogâmicos C57BL
5.
Small ; 6(16): 1785-93, 2010 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-20665628

RESUMO

Targeting of vaccines to abundant immune cell populations within our outer thin skin layers using miniaturized devices-much thinner than a needle and syringe, could improve the efficacy of vaccines (and other immunotherapies). To meet this goal, a densely packed dissolving microprojection array (dissolving Nanopatch) is designed, achieving functional miniaturization by 1) formulating small microneedles (two orders of magnitude smaller than a standard needle and syringe) and 2) multiple layering of the payload within microprojections with tight tolerances (of the order of a micrometer). The formulation method is suitable to many vaccines because it is without harsh or complex chemical processes, and it is performed at low temperatures and at a neutral pH. When the formulated dNPs are applied to skin, consistent and robust penetration is achieved, rapidly targeting the skin strata of interest (<5 min; significantly faster than larger dissolving microneedles that have been previously reported). Resultant diffusion is significantly enhanced within the dermis compared with the epidermis. Using two different antigens (ovalbumin and a commercial trivalent influenza vaccine [Fluvax2008]), the administration of these dissolving patches generate robust systemic immune responses in a mouse model. To the authors' knowledge, this is the first report of successful vaccination with any form of dissolving microneedles. The patches made by this method therefore have the potential for pain-free, needle-free, and effective vaccination in humans.


Assuntos
Nanopartículas/química , Vacinação/métodos , Administração Cutânea , Animais , Carboximetilcelulose Sódica/química , Química Farmacêutica , Dimetilpolisiloxanos/química , Feminino , Vacinas contra Influenza/química , Vacinas contra Influenza/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/química
6.
Small ; 6(16): 1776-84, 2010 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-20665754

RESUMO

The 'Nanopatch' (NP) comprises arrays of densely packed projections with a defined geometry and distribution designed to physically target vaccines directly to thousands of epidermal and dermal antigen presenting cells (APCs). These miniaturized arrays are two orders of magnitude smaller than standard needles-which deliver most vaccines-and are also much smaller than current microneedle arrays. The NP is dry-coated with antigen, adjuvant, and/or DNA payloads. After the NP was pressed onto mouse skin, a protein payload co-localized with 91.4 + or - 4.1 APC mm(-2) (or 2925 in total) representing 52% of the delivery sites within the NP contact area, agreeing well with a probability-based model used to guide the device design; it then substantially increases as the antigen diffuses in the skin to many more cells. APC co-localizing with protein payloads rapidly disappears from the application area, suggesting APC migration. The NP also delivers DNA payloads leading to cutaneous expression of encoded proteins within 24 h. The efficiency of NP immunization is demonstrated using an inactivated whole chikungunya virus vaccine and a DNA-delivered attenuated West Nile virus vaccine. The NP thus offers a needle-free, versatile, highly effective vaccine delivery system that is potentially inexpensive and simple to use.


Assuntos
Vírus Chikungunya/imunologia , Nanoestruturas/química , Vacinação/métodos , Vacinas Virais/administração & dosagem , Vacinas contra o Vírus do Nilo Ocidental/administração & dosagem , Administração Cutânea , Infecções por Alphavirus/prevenção & controle , Animais , Células Apresentadoras de Antígenos/imunologia , Febre de Chikungunya , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de DNA/administração & dosagem , Febre do Nilo Ocidental/prevenção & controle , Vacinas contra o Vírus do Nilo Ocidental/genética , Vacinas contra o Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia
7.
Handb Exp Pharmacol ; (197): 193-219, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20217531

RESUMO

Millions of people die each year from infectious disease, with a main stumbling block being our limited ability to deliver vaccines to optimal sites in the body. Specifically, effective methods to deliver vaccines into outer skin and mucosal layers--sites with immunological, physical and practical advantages that cannot be targeted via traditional delivery methods--are lacking. This chapter investigates the challenge for physical delivery approaches that are primarily needle-free. We examine the skin's structural and immunogenic properties in the context of the physical cell targeting requirements of the viable epidermis, and we review selected current physical cell targeting technologies engineered to meet these needs: needle and syringe, diffusion patches, liquid jet injectors, and microneedle arrays/patches. We then focus on biolistic particle delivery: we first analyze engineering these systems to meet demanding clinical needs, we then examine the interaction of biolistic devices with the skin, focusing on the mechanical interactions of ballistic impact and cell death, and finally we discuss the current clinical outcomes of one key application of engineered delivery devices--DNA vaccines.


Assuntos
Biolística/métodos , Sistemas de Liberação de Medicamentos , Vacinas/administração & dosagem , Animais , Humanos , Injeções a Jato , Microesferas , Mucosa/metabolismo , Pele/metabolismo , Vacinas de DNA/administração & dosagem
8.
NPJ Vaccines ; 4: 41, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31632742

RESUMO

Chemical adjuvants are typically used to improve immune responses induced by immunisation with protein antigens. Here we demonstrate an approach to enhance immune responses that does not require chemical adjuvants. We applied microprojection arrays to the skin, producing a range of controlled mechanical energy to invoke localised inflammation, while administering influenza split virus protein antigen. We used validated computational modelling methods to identify links between mechanical stress and energy generated within the skin strata and resultant cell death. We compared induced immune responses to those induced by needle-based intradermal antigen delivery and used a systems biology approach to examine the nature of the induced inflammatory response, and correlated this with markers of cell stress and death. Increasing the microprojection array application energy and the addition of QS-21 adjuvant were each associated with enhanced antibody response to delivered antigen and with induction of gene transcriptions associated with TNF and NF-κB signalling pathways. We concluded that microprojection intradermal antigen delivery inducing controlled local cell death could potentially replace chemical adjuvants to enhance the immune response to protein antigen.

9.
J Control Release ; 302: 190-200, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-30940498

RESUMO

In a low inflammatory skin environment, Langerhans cells (LCs) - but not dermal dendritic cells (dDCs) - contribute to the pivotal process of tolerance induction. Thus LCs are a target for specific-tolerance therapies. LCs reside just below the stratum corneum, within the skin's viable epidermis. One way to precisely deliver immunotherapies to LCs while remaining minimally invasive is with a skin delivery device such as a microprojection arrays (MPA). Today's MPAs currently achieve rapid delivery (e.g. within minutes of application), but are focussed primarily at delivery of therapeutics to the dermis, deeper within the skin. Indeed, no MPA currently delivers specifically to the epidermal LCs of mouse skin. Without any convenient, pre-clinical device available, advancement of LC-targeted therapies has been limited. In this study, we designed and tested a novel MPA that delivers ovalbumin to the mouse epidermis (eMPA) while maintaining a low, local inflammatory response (as defined by low erythema after 24 h). In comparison to available dermal-targeted MPAs (dMPA), only eMPAs with larger projection tip surface areas achieved shallow epidermal penetration at a low application energy. The eMPA characterised here induced significantly less erythema after 24 h (p = 0.0004), less epidermal swelling after 72 h (p < 0.0001) and 52% less epidermal cell death than the dMPA. Despite these differences in skin inflammation, the eMPA and dMPA promoted similar levels of LC migration out of the skin. However, only the eMPA promoted LCs to migrate with a low MHC II expression and in the absence of dDC migration. Implementing this more mouse-appropriate and low-inflammatory eMPA device to deliver potential immunotherapeutics could improve the practicality and cell-specific targeting of such therapeutics in the pre-clinical stage. Leading to more opportunities for LC-targeted therapeutics such as for allergy immunotherapy and asthma.


Assuntos
Derme/química , Portadores de Fármacos/química , Epiderme/efeitos dos fármacos , Inflamação/prevenção & controle , Células de Langerhans/metabolismo , Ovalbumina/química , Animais , Movimento Celular , Sobrevivência Celular/efeitos dos fármacos , Liberação Controlada de Fármacos , Células Epidérmicas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Teóricos , Ovalbumina/administração & dosagem , Cimento de Policarboxilato/química , Silício/química , Pele , Adesivo Transdérmico
10.
Vaccines (Basel) ; 7(4)2019 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-31756967

RESUMO

Dengue virus is the most important arbovirus impacting global human health, with an estimated 390 million infections annually, and over half the world's population at risk of infection. While significant efforts have been made to develop effective vaccines to mitigate this threat, the task has proven extremely challenging, with new approaches continually being sought. The majority of protective, neutralizing antibodies induced during infection are targeted by the envelope (E) protein, making it an ideal candidate for a subunit vaccine approach. Using truncated, recombinant, secreted E proteins (sE) of all 4 dengue virus serotypes, we have assessed their immunogenicity and protective efficacy in mice, with or without Quil-A as an adjuvant, and delivered via micropatch array (MPA) to the skin in comparison with more traditional routes of immunization. The micropatch contains an ultra-high density array (21,000/cm2) of 110 µm microprojections. Mice received 3 doses of 1 µg (nanopatch, intradermal, subcutaneous, or intra muscular injection) or 10 µg (intradermal, subcutaneous, or intra muscular injection) of tetravalent sE spaced 4 weeks apart. When adjuvanted with Quil-A, tetravalent sE vaccination delivered via MPA resulted in earlier induction of virus-neutralizing IgG antibodies for all four serotypes when compared with all of the other vaccination routes. Using the infectious dengue virus AG129 mouse infectious dengue model, these neutralizing antibodies protected all mice from lethal dengue virus type 2 D220 challenge, with protected animals showing no signs of disease or circulating virus. If these results can be translated to humans, MPA-delivered sE represents a promising approach to dengue virus vaccination.

11.
Vaccine X ; 2: 100030, 2019 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-31384745

RESUMO

The human papillomavirus (HPV) 9-valent, recombinant vaccine (Gardasil™9) helps protect young adults (males and females) against anogenital cancers and genital warts caused by certain HPV genotypes (ref. Gardasil™9 insert). This vaccine is administered intramuscularly (IM). The aim of this study was to determine preclinically whether intradermal (ID) vaccination with an unadjuvanted 9-valent recombinant HPV vaccine using a first-generation ID delivery device, the Nanopatch™, could enhance vaccine immunogenicity compared with the traditional ID route (Mantoux technique). IM injection of HPV VLPs formulated with Merck & Co., Inc., Kenilworth, NJ, USA Alum Adjuvant (MAA) were included in the rhesus study for comparison. The Nanopatch™ prototype contains a high-density array comprised of 10,000 microprojections/cm2, each 250 µm long. It was hypothesized the higher density array with shallower ID delivery may be superior to the Mantoux technique. To test this hypothesis, HPV VLPs without adjuvant were coated on the Nanopatch™, stability of the Nanopatch™ with unadjuvanted HPV VLPs were evaluated under accelerated conditions, skin delivery was verified using radiolabelled VLPs or FluoSpheres®, and the immune response and skin site reaction with the Nanopatch™ was evaluated in rhesus macaques. The immune response induced by Nanopatch™ administration, measured as HPV-specific binding antibodies, was similar to that induced using the Mantoux technique. It was also observed that a lower dose of unadjuvanted HPV VLPs delivered with the first-generation Nanopatch™ and applicator or Mantoux technique resulted in an immune response that was significantly lower compared to a higher-dose of alum adjuvanted HPV VLPs delivered IM in rhesus macaques. The study also indicated unadjuvanted HPV VLPs could be delivered with the first-generation Nanopatch™ and applicator to the skin in 15 s with a transfer efficiency of approximately 20%. This study is the first demonstration of patch administration in non-human primates with a vaccine composed of HPV VLPs.

12.
J Control Release ; 306: 59-68, 2019 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-31121279

RESUMO

Skin-targeting microscale medical devices are becoming popular for therapeutic delivery and diagnosis. We used cryo-SEM, fluorescence lifetime imaging microscopy (FLIM), autofluorescence imaging microscopy and inflammatory response to study the puncturing and recovery of human skin ex vivo and in vivo after discretised puncturing by a microneedle array (Nanopatch®). Pores induced by the microprojections were found to close by ~25% in diameter within the first 30 min, and almost completely close by ~6 h. FLIM images of ex vivo viable epidermis showed a stable fluorescence lifetime for unpatched areas of ~1000 ps up to 24 h. Only the cells in the immediate puncture zones (in direct contact with projections) showed a reduction in the observed fluorescence lifetimes to between ~518-583 ps. The ratio of free-bound NAD(P)H (α1/α2) in unaffected areas of the viable epidermis was ~2.5-3.0, whereas the ratio at puncture holes was almost double at ~4.2-4.6. An exploratory pilot in vivo study also suggested similar closure rate with histamine administration to the forearms of human volunteers after Nanopatch® treatment, although a prolonged inflammation was observed with Tissue Viability Imaging. Overall, this work shows that the pores created by the microneedle-type medical device, Nanopatch®, are transient, with the skin recovering rapidly within 1-2 days in the epidermis after application.


Assuntos
Sistemas de Liberação de Medicamentos , Pele/metabolismo , Adulto , Idoso , Feminino , Humanos , Masculino , Microscopia de Fluorescência por Excitação Multifotônica , Pessoa de Meia-Idade , Agulhas
13.
Biomaterials ; 170: 49-57, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29649748

RESUMO

Microprojection array (MPA) patches are an attractive approach to selectively capture circulating proteins from the skin with minimal invasiveness for diagnostics at the point-of-care or in the home. A key challenge to develop this technology is to extract sufficient quantities of specific proteins from within the skin to enable high diagnostic sensitivity within a convenient amount of time. To achieve this, we investigated the effect of MPA geometry (i.e. projection density, length and array size) on protein capture. We hypothesised that the penetrated surface area of MPAs is a major determinant of protein capture however it was not known if simultaneously increasing projection density, length and array size is possible without adversely affecting penetration and/or tolerability. We show that increasing the projection density (5000-30,000 proj. cm-2) and array size (4-36 mm2) significantly increases biomarker capture whilst maintaining of a similar level tolerability, which supports previous literature for projection length (40-190 µm). Ultimately, we designed a high surface area MPA (30,000 proj. cm-2, 36 mm2, 140 µm) with a 4.5-fold increase in penetrated surface area compared to our standard MPA design (20,408 proj. cm-2, 16 mm2, 100 µm). The high surface area MPA captured antigen-specific IgG from mice in 30 s with 100% diagnostic sensitivity compared with 10-30 min for previous MPA immunoassay patches, which is over an order of magnitude reduction in wear time. This demonstrates for the first time that MPAs may be used for ultra-rapid (<1 min) protein capture from skin in a time competitive with standard clinical procedures like the needle and lancet, which has broad implications for minimally invasive and point-of-care diagnostics.


Assuntos
Imunoensaio/métodos , Imunoglobulina G/isolamento & purificação , Pele/metabolismo , Dispositivos Eletrônicos Vestíveis , Animais , Camundongos , Propriedades de Superfície , Fatores de Tempo
14.
Sci Rep ; 8(1): 17759, 2018 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-30531828

RESUMO

Microscale medical devices are being developed for targeted skin delivery of vaccines and the extraction of biomarkers, with the potential to revolutionise healthcare in both developing and developed countries. The effective clinical development of these devices is dependent on understanding the macro-molecular diffusion properties of skin. We hypothesised that diffusion varied according to specific skin layers. Using three different molecular weights of rhodamine dextran (RD) (MW of 70, 500 and 2000 kDa) relevant to the vaccine and therapeutic scales, we deposited molecules to a range of depths (0-300 µm) in ex vivo human skin using the Nanopatch device. We observed significant dissipation of RD as diffusion with 70 and 500 kDa within the 30 min timeframe, which varied with MW and skin layer. Using multiphoton microscopy, image analysis and a Fick's law analysis with 2D cartesian and axisymmetric cylindrical coordinates, we reported experimental trends of epidermal and dermal diffusivity values ranging from 1-8 µm2 s-1 to 1-20 µm2 s-1 respectively, with a significant decrease in the dermal-epidermal junction of 0.7-3 µm2 s-1. In breaching the stratum corneum (SC) and dermal-epidermal junction barriers, we have demonstrated practical application, delivery and targeting of macromolecules to both epidermal and dermal antigen presenting cells, providing a sound knowledge base for future development of skin-targeting clinical technologies in humans.


Assuntos
Derme/metabolismo , Epiderme/metabolismo , Administração Cutânea , Adulto , Dextranos/farmacologia , Difusão , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Cinética , Peso Molecular , Agulhas , Rodaminas/farmacologia , Absorção Cutânea , Vacinas/farmacologia
15.
Biomaterials ; 28(33): 4968-77, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17720240

RESUMO

A challenge in combating many major diseases is breaching the skin's tough outer layer (the stratum corneum (SC)) and delivering drugs and genes into the underlying abundant immunologically sensitive viable epidermal cells with safe, practical physical technologies. To achieve this effectively and accurately, design information is needed on key skin mechanical properties when pushing into and through epidermal skin cells. We measure these important mechanical properties by penetrating through the intact SC and viable epidermis (VE) of freshly excised murine skin with a NANO-indenter, using custom tungsten probes fabricated with nominally 5 and 2 microm diameters (with nanoscale tips). We show the skin Young's modulus, storage modulus and stress all dramatically decreased through the SC. Also, for a given penetration depth, decreasing the probe size significantly increases the storage modulus. Biological variation in penetrating the skin was shown. These collective findings advance the rational design of physical approaches for delivering genes and drugs within key cells of the VE.


Assuntos
Sistemas de Liberação de Medicamentos , Epiderme/fisiologia , Técnicas de Transferência de Genes , Animais , Fenômenos Biomecânicos , Citoesqueleto/metabolismo , Epiderme/anatomia & histologia , Epiderme/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Pele/metabolismo , Estresse Mecânico , Propriedades de Superfície , Tungstênio/química
16.
Comput Methods Programs Biomed ; 85(2): 124-8, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17141915

RESUMO

A unique hand-held needle-free powder injection system, using a transient shock-tube flow to deliver powder genes and drugs into human skin for a wide range of treatments, has been proposed. In the development of such devices, a strong non-linear phenomenon, possibly shock process instead of unsteady expansion waves, was observed in the driver portion of the shock-tube flow in the presence of a gas micro-cylinder. In this paper, we further investigate effects of a model micro-cylinder in the driver on the gas dynamics of a prototype clinical device numerically. To accurately simulate such complex shock-tube flows, an efficient numerical solver, MIFVS, is extended to incorporate with a transition-modified turbulence model. Comparison with experimental measurements shows that the extended MIFVS accurately predicts pressure traces in both laminar and turbulent regimes. The separation zone due to a strong non-linear process is properly captured via such transition-modified turbulence model. Numerical investigations and discoveries are presented and discussed.


Assuntos
Sistemas de Liberação de Medicamentos/instrumentação , Desenho de Equipamento , Humanos , Injeções Epidurais/instrumentação , Modelos Estatísticos , Pós , Ultrassom
17.
Acta Biomater ; 48: 341-356, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27746361

RESUMO

In-depth understanding of skin elastic and rupture behavior is fundamental to enable next-generation biomedical devices to directly access areas rich in cells and biomolecules. However, the paucity of skin mechanical characterization and lack of established fracture models limits their rational design. We present an experimental and numerical study of skin mechanics during dynamic interaction with individual and arrays of micro-penetrators. Initially, micro-indentation of individual skin strata revealed hyperelastic moduli were dramatically rate-dependent, enabling extrapolation of stiffness properties at high velocity regimes (>1ms-1). A layered finite-element model satisfactorily predicted the penetration of micro-penetrators using characteristic fracture energies (∼10pJµm-2) significantly lower than previously reported (≫100pJµm-2). Interestingly, with our standard application conditions (∼2ms-1, 35gpistonmass), ∼95% of the application kinetic energy was transferred to the backing support rather than the skin ∼5% (murine ear model). At higher velocities (∼10ms-1) strain energy accumulated in the top skin layers, initiating fracture before stress waves transmitted deformation to the backing material, increasing energy transfer efficiency to 55%. Thus, the tools developed provide guidelines to rationally engineer skin penetrators to increase depth targeting consistency and payload delivery across patients whilst minimizing penetration energy to control skin inflammation, tolerability and acceptability. STATEMENT OF SIGNIFICANCE: The mechanics of skin penetration by dynamically-applied microscopic tips is investigated using a combined experimental-computational approach. A FE model of skin is parameterized using indentation tests and a ductile-failure implementation validated against penetration assays. The simulations shed light on skin elastic and fracture properties, and elucidate the interaction with microprojection arrays for vaccine delivery allowing rational design of next-generation devices.


Assuntos
Elasticidade , Microscopia/métodos , Fenômenos Fisiológicos da Pele , Animais , Fenômenos Biomecânicos , Feminino , Análise de Elementos Finitos , Camundongos Endogâmicos BALB C , Modelos Animais , Modelos Teóricos , Análise Numérica Assistida por Computador , Permeabilidade , Reprodutibilidade dos Testes , Estresse Mecânico , Viscosidade
18.
Sci Rep ; 7(1): 15885, 2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29162871

RESUMO

Emerging micro-scale medical devices are showing promise, whether in delivering drugs or extracting diagnostic biomarkers from skin. In progressing these devices through animal models towards clinical products, understanding the mechanical properties and skin tissue structure with which they interact will be important. Here, through measurement and analytical modelling, we advanced knowledge of these properties for commonly used laboratory animals and humans (~30 g to ~150 kg). We hypothesised that skin's stiffness is a function of the thickness of its layers through allometric scaling, which could be estimated from knowing a species' body mass. Results suggest that skin layer thicknesses are proportional to body mass with similar composition ratios, inter- and intra-species. Experimental trends showed elastic moduli increased with body mass, except for human skin. To interpret the relationship between species, we developed a simple analytical model for the bulk elastic moduli of skin, which correlated well with experimental data. Our model suggest that layer thicknesses may be a key driver of structural stiffness, as the skin layer constituents are physically and therefore mechanically similar between species. Our findings help advance the knowledge of mammalian skin mechanical properties, providing a route towards streamlined micro-device research and development onto clinical use.


Assuntos
Elasticidade , Equipamentos e Provisões , Pele/anatomia & histologia , Adulto , Animais , Fenômenos Biomecânicos , Módulo de Elasticidade , Feminino , Humanos , Modelos Lineares , Masculino , Camundongos , Modelos Biológicos , Coelhos , Ratos , Pele/citologia , Dobras Cutâneas , Suínos , Viscosidade
19.
Sci Rep ; 7(1): 12644, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28974777

RESUMO

To secure a polio-free world, the live attenuated oral poliovirus vaccine (OPV) will eventually need to be replaced with inactivated poliovirus vaccines (IPV). However, current IPV delivery is less suitable for campaign use than OPV, and more expensive. We are progressing a microarray patch delivery platform, the Nanopatch, as an easy-to-use device to administer vaccines, including IPV. The Nanopatch contains an ultra-high density array (10,000/cm2) of short (~230 µm) microprojections that delivers dry coated vaccine into the skin. Here, we compare the relative immunogenicity of Nanopatch immunisation versus intramuscular injection in rats, using monovalent and trivalent formulations of IPV. Nanopatch delivery elicits faster antibody response kinetics, with high titres of neutralising antibody after just one (IPV2) or two (IPV1 and IPV3) immunisations, while IM injection requires two (IPV2) or three (IPV1 and IPV3) immunisations to induce similar responses. Seroconversion to each poliovirus type was seen in 100% of rats that received ~1/40th of a human dose of IPV delivered by Nanopatch, but not in rats given ~1/8th or ~1/40th dose by IM injection. Ease of administration coupled with dose reduction observed in this study suggests the Nanopatch could facilitate inexpensive IPV vaccination in campaign settings.


Assuntos
Anticorpos Neutralizantes/imunologia , Formação de Anticorpos/imunologia , Poliomielite/prevenção & controle , Vacina Antipólio de Vírus Inativado/imunologia , Animais , Anticorpos Antivirais/imunologia , Humanos , Poliomielite/imunologia , Poliomielite/virologia , Poliovirus/imunologia , Poliovirus/patogenicidade , Vacina Antipólio de Vírus Inativado/administração & dosagem , Vacina Antipólio Oral/administração & dosagem , Ratos , Pele/efeitos dos fármacos , Pele/imunologia , Vacinação
20.
J Invest Dermatol ; 126(7): 1541-8, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16645596

RESUMO

Langerhans cells (LCs) can be targeted with DNA-coated gold micro-projectiles ("Gene Gun") to induce potent cellular and humoral immune responses. It is likely that the relative volumetric distribution of LCs and keratinocytes within the epidermis impacts on the efficacy of Gene Gun immunization protocols. This study quantified the three-dimensional (3D) distribution of LCs and keratinocytes in the mouse skin model with a near-infrared multiphoton laser-scanning microscope (NIR-MPLSM). Stratum corneum (SC) and viable epidermal thickness measured with MPLSM was found in close agreement with conventional histology. LCs were located in the vertical plane at a mean depth of 14.9 microm, less than 3 mum above the dermo-epidermal boundary and with a normal histogram distribution. This likely corresponds to the fact that LCs reside in the suprabasal layer (stratum germinativum). The nuclear volume of keratinocytes was found to be approximately 1.4 times larger than that of resident LCs (88.6 microm3). Importantly, the ratio of LCs to keratinocytes in mouse ear skin (1:15) is more than three times higher than that reported for human breast skin (1:53). Accordingly, cross-presentation may be more significant in clinical Gene Gun applications than in pre-clinical mouse studies. These interspecies differences should be considered in pre-clinical trials using mouse models.


Assuntos
Imunidade Celular , Queratinócitos/imunologia , Células de Langerhans/imunologia , Modelos Animais , Pele/imunologia , Vacinação/métodos , Animais , Biolística/métodos , Mama/citologia , Células Epidérmicas , Humanos , Imageamento Tridimensional , Imunidade Celular/imunologia , Imuno-Histoquímica , Queratinócitos/citologia , Células de Langerhans/citologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência por Excitação Multifotônica , Pele/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA