Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Control Release ; 367: 649-660, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38295993

RESUMEN

The oral mucosa is an attractive site for immunization due to its accessibility and ability to elicit local and systemic immune responses. However, evaluating oral mucosal immunogenicity has proven challenging due to the physical barriers and immunological complexity of the oral mucosa. Microneedles can overcome these physical barriers, but previous work has been limited in the scope of microneedle delivery site, geometry, and release kinetics, all of which are expected to affect physiological responses. Here, we develop integrated fiber microneedle devices, an oral dosage form with tunable geometries and material configurations capable of both burst and sustained release to controlled depths in the oral mucosa. Integrated fiber microneedles administered to either the buccal or sublingual mucosa result in seroconversion and antigen-specific interferon-γ secretion in splenocytes. The dynamics and magnitude of the resulting immune response can be modulated by tuning microneedle release kinetics. Optimal microneedle geometry is site-specific, with longer microneedles eliciting greater immunogenicity in the buccal mucosa, and shorter microneedles eliciting greater immunogenicity in the sublingual mucosa. The Th1/Th2 phenotype of the resulting immune response is also dependent on integrated fiber microneedle length. Together, these results establish integrated fiber microneedles as a multifunctional delivery system for the oral mucosa and motivate further exploration using tunable delivery systems to better understand oral mucosal immunity.


Asunto(s)
Antígenos , Mucosa Bucal , Vacunación/métodos , Inmunidad Mucosa , Agujas , Sistemas de Liberación de Medicamentos , Administración Cutánea
2.
Adv Drug Deliv Rev ; 198: 114860, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37160248

RESUMEN

Long-acting formulations are designed to reduce dosing frequency and simplify dosing schedules by providing an extended duration of action. One approach to obtain long-acting formulations is to combine long-acting prodrugs (LA-prodrug) with existing or emerging drug delivery technologies (DDS). The design criteria for long-acting prodrugs are distinct from conventional prodrug strategies that alter absorption, distribution, metabolism, and excretion (ADME) parameters. Our review focuses on long-acting prodrug delivery systems (LA-prodrug DDS), which is a subcategory of long-acting formulations where prodrug design enables DDS formulation to achieve an extended duration of action that is greater than the parent drug. Here, we define LA-prodrugs as the conjugation of an active pharmaceutical ingredient (API) to a promoiety group via a cleavable covalent linker, where both the promoiety and linker are selected to enable formulation and administration from a drug delivery system (DDS) to achieve an extended duration of action. These LA-prodrug DDS results in an extended interval where the API is within a therapeutic range without necessarily altering ADME as is typical of conventional prodrugs. The conversion of the LA-prodrug to the API is dependent on linker cleavage, which can occur before or after release from the DDS. The requirement for linker cleavage provides an additional tool to prolong release from these LA-prodrug DDS. In addition, the physicochemical properties of drugs can be tuned by promoiety selection for a particular DDS. Conjugation with promoieties that are carriers or amenable to assembly into carriers can also provide access to formulations designed for extending duration of action. LA-prodrugs have been applied to a wide variety of drug delivery strategies and are categorized in this review by promoiety size and complexity. Small molecule promoieties (typically MW < 1000 Da) have been used to improve encapsulation or partitioning as well as broaden APIs for use with traditional long-acting formulations such as solid drug dispersions. Macromolecular promoieties (typically MW > 1000 Da) have been applied to hydrogels, nanoparticles, micelles, dendrimers, and polymerized prodrug monomers. The resulting LA-prodrug DDS enable extended duration of action for active pharmaceuticals across a wide range of applications, with target release timescales spanning days to years.


Asunto(s)
Profármacos , Humanos , Profármacos/uso terapéutico , Sistemas de Liberación de Medicamentos
3.
ACS Biomater Sci Eng ; 8(7): 2995-3009, 2022 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-35749682

RESUMEN

Medical technology that blocks the fallopian tubes nonsurgically could increase access to permanent contraception and address current unmet needs in family planning. To achieve total occlusion of the fallopian tube via scar tissue formation, acute trauma to the tubal epithelium must first occur followed by a sustained and ultimately fibrotic inflammatory response. Here, we developed drug-eluting fiber-based microparticles that provide tunable dose and release of potent sclerosing agents. This fabrication strategy demonstrates high encapsulation of physicochemically diverse agents and the potential for scalable manufacturing by utilizing free-surface electrospinning to generate material for fiber micronization. Manipulation of nanofiber formulation such as drug loading, drug hydrophobicity, polymer hydrophobicity, and crystallinity allowed for modulation of the sustained release properties of our fiber microparticles. We assessed various fibrous microparticle formulations in vivo using a newly developed and validated guinea pig model for contraception. We found that fiber microparticles with bolus release doxycycline effectively elicited acute trauma and those formulated with highly loaded phenyl benzoate caused sustained inflammation in the target organs. The demonstrated potency of these electrospun microparticles, as well as their embolic size and shape, suggests potential for proximal agglomeration and inflammatory activity in the fallopian tubes following transcervical delivery.


Asunto(s)
Trompas Uterinas , Soluciones Esclerosantes , Animales , Anticoncepción , Femenino , Cobayas , Humanos , Polímeros , Soluciones Esclerosantes/farmacología
4.
Adv Healthc Mater ; 11(9): e2102087, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35137550

RESUMEN

Porosity is an important material feature commonly employed in implants and tissue scaffolds. The presence of material voids permits the infiltration of cells, mechanical compliance, and outward diffusion of pharmaceutical agents. Various studies have confirmed that porosity indeed promotes favorable tissue responses, including minimal fibrous encapsulation during the foreign body reaction (FBR). However, increased biofilm formation and calcification is also described to arise due to biomaterial porosity. Additionally, the relevance of host responses like the FBR, infection, calcification, and thrombosis are dependent on tissue location and specific tissue microenvironment. In this review, the features of porous materials and the implications of porosity in the context of medical devices is discussed. Common methods to create porous materials are also discussed, as well as the parameters that are used to tune pore features. Responses toward porous biomaterials are also reviewed, including the various stages of the FBR, hemocompatibility, biofilm formation, and calcification. Finally, these host responses are considered in tissue specific locations including the subcutis, bone, cardiovascular system, brain, eye, and female reproductive tract. The effects of porosity across the various tissues of the body is highlighted and the need to consider the tissue context when engineering biomaterials is emphasized.


Asunto(s)
Calcificación Fisiológica , Calcinosis , Materiales Biocompatibles , Femenino , Reacción a Cuerpo Extraño , Humanos , Porosidad
5.
Adv Drug Deliv Rev ; 178: 113956, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34481031

RESUMEN

Recent estimates suggest that one in two sexually active individuals will acquire a sexually transmitted infection by age 25, an alarming statistic that amounts to over 1 million new infections per day worldwide. Vaccination against STIs is highly desirable for alleviating this global burden of disease. Vaginal immunization is a promising strategy to combat transmission via the vaginal mucosa. The vagina is typically considered a poor inductive site for common correlates of adaptive immunity. However, emerging evidence suggests that immune tolerance may be overcome by precisely engineered vaccination schemes that orchestrate cell-mediated immunity and establish tissue resident memory immune cells. In this review, we will discuss the unique immunological milieu of the vaginal mucosa and our current understanding of correlates of pathogenesis and protection for several common STIs. We then present a summary of recent vaginal vaccine studies and explore the role that mucosal adjuvants and delivery systems play in enhancing protection according to requisite features of immunity. Finally, we offer perspectives on the challenges and future directions of vaginal vaccine delivery, discussing remaining physiological barriers and innovative vaccine formulations that may overcome them.


Asunto(s)
Vacunas/inmunología , Vagina/inmunología , Inmunidad Adaptativa/inmunología , Femenino , Humanos , Tolerancia Inmunológica/inmunología , Vacunación
6.
Biomaterials ; 273: 120806, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33905960

RESUMEN

Within tissue exposed to the systemic immune system, lymphocytes and fibroblasts act against biomaterials via the development of a fibrous capsule, known as the foreign body reaction (FBR). Inspired by the natural tolerance that the uterine cavity has to foreign bodies, our study explores the role of microenvironment across classical (subcutaneous) and immune privileged (uterine) tissues in the development of the FBR. As a model biomaterial, we used electrospun fibers loaded with sclerosing agents to provoke scar tissue growth. Additionally, we integrated these materials onto an intrauterine device as a platform for intrauterine biomaterial studies. Polyester materials in vitro achieved drug release up to 10 days, greater pro-inflammatory and pro-healing cytokine expression, and the addition of gelatin enabled greater fibroblast attachment. We observed the materials that induced the greatest FBR in the mouse, had no effect when inserted at the utero-tubal junction of non-human primates. These results suggest that the FBR varies across different tissue microenvironments, and a dampened fibrotic response exists in the uterine cavity, possibly due to immune privilege. Further study of immune privileged tissue factors on biomaterials could broaden our understanding of the FBR and inform new methods for achieving biocompatibility in vivo.


Asunto(s)
Materiales Biocompatibles , Reacción a Cuerpo Extraño , Animales , Fibrosis , Reacción a Cuerpo Extraño/etiología , Reacción a Cuerpo Extraño/patología , Ratones , Prótesis e Implantes , Tejido Subcutáneo/patología
7.
Front Oral Health ; 2: 821812, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35224539

RESUMEN

The oral mucosa contains distinct tissue sites with immune niches capable of either immunogenic or tolerogenic responses. However, immune cell compositions within oral mucosal tissues at homeostasis have not been well-characterized in human relevant tissues. Non-human primates (NHP) are a major model for the human immune system and oral anatomy, and therefore improved understanding of NHP oral immune cell populations can provide important insights for studying disease pathologies and developing therapies. Herein, we characterize immune cell types of three sites within the oral cavity (buccal, sublingual, lingual tonsil) sampled by biopsy and cytobrush in pigtail macaques. Tonsil biopsies had more T-cells, dendritic cells (DCs), DC subtypes, and CD4+ T-cells than buccal or sublingual biopsies when normalized by tissue mass. Biopsy proved to collect more immune cells than cytobrushes, however frequencies of CD45+ subpopulations were comparable between methods. Live cells isolated from biopsied tonsils had greater CD45+ leukocyte frequencies (mean 31.6 ± SD 20.4%) than buccal (13.8 ± 4.6%) or sublingual (10.0 ± 5.1%) tissues. T-cells composed more than half of the CD45+ population in sublingual tissue (60.1 ± 9.6%) and the tonsil (54.6 ± 7.5%), but only 31.9 ± 7.2% in buccal samples. CD20+ B-cells composed a greater percentage of CD45+ leukocytes in the tonsil (12.8 ± 9.1%) than buccal (1.2 ± 1.0%) or sublingual tissues (0.8 ± 1.2%). Immune population comparisons are also made between sex and age. These results present an important step for understanding the oral immune environment, oral disease, and site-specific therapy development.

8.
J Control Release ; 329: 782-793, 2021 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-33035616

RESUMEN

Dendritic cells (DCs) play a critical role in shaping adaptive immunity. Systemic transfer of DCs by intravenous injection has been widely investigated to inform the development of immunogenic DCs for use as cellular therapies. Adoptive transfer of DCs to mucosal sites has been limited but serves as a valuable tool to understand the role of the microenvironment on mucosal DC activation, maturation and antigen presentation. Here, we show that chitosan facilitates transmigration of DCs across the vaginal epithelium in the mouse female reproductive tract (FRT). In addition, ex vivo programming of DCs with fms-related tyrosine kinase 3 ligand (Flt3-L) was found to enhance translocation of intravaginally administered DCs to draining lymph nodes (dLNs) and stimulate in vivo proliferation of both antigen-specific CD4+ and CD8+ T cells (cross-presentation). Mucosal priming with chitosan and DC programming may hold great promise to enhance efficacy of DC-based vaccination to the female genital mucosa.


Asunto(s)
Presentación de Antígeno , Células Dendríticas , Animales , Linfocitos T CD8-positivos , Femenino , Genitales , Ratones , Ratones Endogámicos C57BL , Membrana Mucosa
9.
Sci Adv ; 5(3): eaav6322, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30944862

RESUMEN

A proposed strategy to cure HIV uses latency-reversing agents (LRAs) to reactivate latent proviruses for purging HIV reservoirs. A variety of LRAs have been identified, but none has yet proven effective in reducing the reservoir size in vivo. Nanocarriers could address some major challenges by improving drug solubility and safety, providing sustained drug release, and simultaneously delivering multiple drugs to target tissues and cells. Here, we formulated hybrid nanocarriers that incorporate physicochemically diverse LRAs and target lymphatic CD4+ T cells. We identified one LRA combination that displayed synergistic latency reversal and low cytotoxicity in a cell model of HIV and in CD4+ T cells from virologically suppressed patients. Furthermore, our targeted nanocarriers selectively activated CD4+ T cells in nonhuman primate peripheral blood mononuclear cells as well as in murine lymph nodes, and substantially reduced local toxicity. This nanocarrier platform may enable new solutions for delivering anti-HIV agents for an HIV cure.


Asunto(s)
Fármacos Anti-VIH/administración & dosificación , Linfocitos T CD4-Positivos/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Nanoestructuras/administración & dosificación , Latencia del Virus/efectos de los fármacos , Animales , Fármacos Anti-VIH/química , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Fenómenos Químicos , Portadores de Fármacos/química , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/fisiología , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Macaca , Ratones Endogámicos C57BL , Nanoestructuras/química , Latencia del Virus/inmunología
10.
Macromol Biosci ; 19(4): e1800362, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30589222

RESUMEN

Significant efforts have been invested in finding a delivery system that can encapsulate and deliver therapeutics. Core-shell polymer-lipid hybrid nanoparticles have been studied as a promising platform because of their mechanical stability, narrow size distribution, biocompatibility, and ability to co-deliver diverse drugs. Here, novel core-shell nanoparticles based on a poly(lactic-co-glycolic acid) (PLGA) core and multilamellar lipid shell are designed, where the lipid bilayers are crosslinked between the two adjacent bilayers (PLGA-ICMVs). The cross-platform performance of the nanoparticles to other polymer-lipid hybrid platforms is examined, including physicochemical characteristics, ability to encapsulate a variety of therapeutics, biocompatibility, and functionality as a vaccine delivery platform. Differential abilities of nanoparticle systems to encapsulate distinct pharmaceutics are observed, which suggest careful consideration of the platform chosen depending on the therapeutic agent and desired function. The novel PLGA-ICMV platform herein demonstrates great potential in stably encapsulating water-soluble agents and therefore is an attractive platform for therapeutic delivery.


Asunto(s)
Materiales Biocompatibles Revestidos , Sistemas de Liberación de Medicamentos , Lípidos , Ensayo de Materiales , Nanopartículas/química , Animales , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Lípidos/química , Lípidos/farmacología , Ratones , Ratones Transgénicos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología
11.
Adv Healthc Mater ; 8(4): e1801180, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30537400

RESUMEN

The oral mucosa is a minimally invasive and immunologically rich site that is underutilized for vaccination due to physiological and immunological barriers. To develop effective oral mucosal vaccines, key questions regarding vaccine residence time, uptake, adjuvant formulation, dose, and delivery location must be answered. However, currently available dosage forms are insufficient to address all these questions. An ideal oral mucosal vaccine delivery system would improve both residence time and epithelial permeation while enabling efficient delivery of physicochemically diverse vaccine formulations. Microneedles have demonstrated these capabilities for dermal vaccine delivery. Additionally, microneedles enable precise control over delivery properties like depth, uniformity, and dosing, making them an ideal tool to study oral mucosal vaccination. Select studies have demonstrated the feasibility of microneedle-mediated oral mucosal vaccination, but they have only begun to explore the broad functionality of microneedles. This review describes the physiological and immunological challenges related to oral mucosal vaccine delivery and provides specific examples of how microneedles can be used to address these challenges. It summarizes and compares the few existing oral mucosal microneedle vaccine studies and offers a perspective for the future of the field.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Mucosa Bucal/inmunología , Agujas , Vacunación , Vacunas/uso terapéutico , Animales , Humanos , Vacunas/inmunología
12.
Eur J Pharm Biopharm ; 138: 48-63, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-29879528

RESUMEN

The advent of combination antiretroviral therapy (cART) has transformed HIV-1 infection into a controllable chronic disease, but these therapies are incapable of eradicating the virus to bring about an HIV cure. Multiple strategies have been proposed and investigated to eradicate latent viral reservoirs from various biological sanctuaries. However, due to the complexity of HIV infection and latency maintenance, a single drug is unlikely to eliminate all HIV reservoirs and novel strategies may be needed to achieve better efficacy while limiting systemic toxicity. In this review, we describe HIV latency in cellular and anatomical reservoirs, and present an overview of current strategies for HIV cure with a focus on their challenges for clinical translation. Then we provide a summary of nanotechnology solutions that have been used to address challenges in HIV cure by delivering physicochemically diverse agents for combination therapy or targeting HIV reservoir sites. We also review nanocarrier-based gene delivery and immunotherapy used in cancer treatment but may have potential applications in HIV cure.


Asunto(s)
Antirretrovirales/farmacología , Antirretrovirales/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Animales , Humanos , Inmunoterapia/métodos , Nanotecnología/métodos
13.
ACS Biomater Sci Eng ; 5(8): 4013-4022, 2019 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-33117884

RESUMEN

Polymeric nanocarriers have been extensively used to improve the delivery of hydrophobic drugs, but often provide low encapsulation efficiency and percent loading for hydrophilic compounds. In particular, insufficient loading of hydrophilic antiretroviral drugs such as the integrase inhibitor raltegravir (RAL) has limited the development of sustained-release therapeutics or prevention strategies against HIV. To address this, we developed a generalizable prodrug strategy using RAL as a model where loading, release and subsequent hydrolysis can be tuned by promoiety selection. Prodrugs with large partition coefficients increased the encapsulation efficiency up to 25-fold relative to RAL, leading to significant dose reductions in antiviral activity assays. The differential hydrolysis rates of these prodrugs led to distinct patterns of RAL availability and observed antiviral activity. We also developed a method to monitor the temporal distribution of both prodrug and RAL in cells treated with free prodrug or prodrug-NPs. Results of these studies indicated that prodrug-NPs create an intracellular drug reservoir capable of sustained intracellular drug release. Overall, our results suggest that the design of prodrugs for specific polymeric nanocarrier systems could provide a more generalized strategy to formulate physicochemically diverse hydrophilic drugs with a number of biomedical applications.

14.
Nanomedicine ; 14(7): 2143-2153, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29964219

RESUMEN

A major sanctuary site for HIV infection is the gut-associated lymphoid tissue (GALT). The α4ß7 integrin gut homing receptor is a promising therapeutic target for the virus reservoir because it leads to migration of infected cells to the GALT and facilitates HIV infection. Here, we developed a core-shell nanoparticle incorporating the α4ß7 monoclonal antibody (mAb) as a dual-functional ligand for selectively targeting a protease inhibitor (PI) to gut-homing T cells in the GALT while simultaneously blocking HIV infection. Our nanoparticles significantly reduced cytotoxicity of the PI and enhanced its in vitro antiviral activity in combination with α4ß7 mAb. We demonstrate targeting function of our nanocarriers in a human T cell line and primary cells isolated from macaque ileum, and observed higher in vivo biodistribution to the murine small intestines where they accumulate in α4ß7+ cells. Our LCNP shows the potential to co-deliver ARVs and mAbs for eradicating HIV reservoirs.


Asunto(s)
Fármacos Anti-VIH/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Integrinas/inmunología , Intestino Delgado/efectos de los fármacos , Nanopartículas/administración & dosificación , Inhibidores de Proteasas/química , Linfocitos T/efectos de los fármacos , Animales , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/inmunología , Humanos , Íleon/efectos de los fármacos , Íleon/inmunología , Íleon/virología , Intestino Delgado/inmunología , Intestino Delgado/virología , Macaca mulatta , Ratones , Nanopartículas/química , Piridinas/administración & dosificación , Piridinas/química , Piridinas/farmacología , Pironas/administración & dosificación , Pironas/química , Pironas/farmacología , Sulfonamidas , Linfocitos T/inmunología , Linfocitos T/virología
15.
J Biomed Mater Res A ; 106(5): 1177-1188, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29271128

RESUMEN

Monoclonal antibodies and peptides are conjugated to the surface of nanocarriers (NCs) for targeting purposes in numerous applications. However, targeting efficacy may vary with their specificity, affinity, or avidity when linked to NCs. The physicochemical properties of NCs may also affect targeting. We compared the targeting efficacy of the CD4 binding peptide BP4 and an anti-CD4 monoclonal antibody (CD4 mAb) and its fragments, when conjugated to lipid-coated poly(lactic-co-glycolic) acid nanoparticles (LCNPs). Negatively charged LCNPs with cholesteryl butyrate in the lipid layer (cbLCNPs) dramatically reduced nonspecific binding, leading to higher targeting specificity, compared to neutral or positively charged LCNPs with DOTAP (dtLCNP). cbLCNPs surface conjugated with a CD4 antibody (CD4-cbLCNPs) or its fragments (fCD4-cbLCNPs), but not BP4, showed high binding in vitro to the human T cell line 174xCEM, and preferential binding to CD3+ CD14-CD8- cells from pigtail macaque peripheral blood mononuclear cells. CD4-cbLCNPs showed 10-fold higher binding specificity for CD4+ than CD8+ T cells, while fCD4-cbLCNPs demonstrated the highest binding level overall, but only three-fold higher binding specificity. This study demonstrates the importance of ζ-potential on NC targeting and indicates that CD4 mAb and its fragments are the best candidates for delivery of therapeutic agents to CD4+ T cells. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1177-1188, 2018.


Asunto(s)
Antígenos CD4/metabolismo , Lípidos/química , Nanopartículas/química , Polímeros/química , Animales , Línea Celular , Humanos , Leucocitos Mononucleares/metabolismo , Ligandos , Macaca , Tamaño de la Partícula , Péptidos/metabolismo
16.
Int J Pharm ; 533(1): 99-110, 2017 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-28941831

RESUMEN

Biologics are the most rapidly growing class of therapeutics, but commonly suffer from low stability. Peroral administration of these therapeutics is an attractive delivery route; however, this route introduces unique physiological challenges that increase the susceptibility of proteins to lose function. Formulation of proteins into biomaterials, such as electrospun fibers, is one strategy to overcome these barriers, but such platforms need to be optimized to ensure protein stability and maintenance of bioactivity during the formulation process. This work develops an emulsion electrospinning method to load proteins into Eudragit® L100 fibers for peroral delivery. Horseradish peroxidase and alkaline phosphatase are encapsulated with high efficiency into fibers and released with pH-specificity. Recovery of protein bioactivity is enhanced through reduction of the emulsion aqueous phase and the inclusion of a hydrophilic polymer excipient. Finally, we show that formulation of proteins in lyophilized electrospun fibers extends the therapeutic shelf life compared to aqueous storage. Thus, this platform shows promise as a novel dosage form for the peroral delivery of biotherapeutics.


Asunto(s)
Fosfatasa Alcalina/química , Sistemas de Liberación de Medicamentos , Peroxidasa de Rábano Silvestre/química , Nanofibras/química , Administración Oral , Productos Biológicos/química , Liberación de Fármacos , Emulsiones , Liofilización , Concentración de Iones de Hidrógeno , Ácidos Polimetacrílicos/química
17.
Biomaterials ; 144: 1-16, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28802690

RESUMEN

Current approaches for topical vaginal administration of nanoparticles result in poor retention and extensive leakage. To overcome these challenges, we developed a nanoparticle-releasing nanofiber delivery platform and evaluated its ability to improve nanoparticle retention in a murine model. We individually tailored two components of this drug delivery system for optimal interaction with mucus, designing (1) mucoadhesive fibers for better retention in the vaginal tract, and (2) PEGylated nanoparticles that diffuse quickly through mucus. We hypothesized that this novel dual-functioning (mucoadhesive/mucus-penetrating) composite material would provide enhanced retention of nanoparticles in the vaginal mucosa. Equivalent doses of fluorescent nanoparticles were vaginally administered to mice in either water (aqueous suspension) or fiber composites, and fluorescent content was quantified in cervicovaginal mucus and vaginal tissue at time points from 24 h to 7d. We also fabricated composite fibers containing etravirine-loaded nanoparticles and evaluated the pharmacokinetics over 7d. We found that our composite materials provided approximately 30-fold greater retention of nanoparticles in the reproductive tract at 24 h compared to aqueous suspensions. Compared to nanoparticles in aqueous suspension, the nanoparticles in fiber composites exhibited sustained and higher etravirine concentrations after 24 h and up to 7d, demonstrating the capabilities of this new delivery platform to sustain nanoparticle release out to 3d and drug retention out to one week after a single administration. This is the first report of nanoparticle-releasing fibers for vaginal drug delivery, as well as the first study of a single delivery system that combines two components uniquely engineered for complementary interactions with mucus.


Asunto(s)
Preparaciones de Acción Retardada/química , Colorantes Fluorescentes/administración & dosificación , Nanofibras/química , Nanopartículas/administración & dosificación , Piridazinas/administración & dosificación , Inhibidores de la Transcriptasa Inversa/administración & dosificación , Vagina/metabolismo , Administración Intravaginal , Animales , Sistemas de Liberación de Medicamentos/métodos , Femenino , Colorantes Fluorescentes/farmacocinética , Ratones , Ratones Endogámicos C57BL , Membrana Mucosa/metabolismo , Nanofibras/ultraestructura , Nanopartículas/análisis , Nitrilos , Piridazinas/farmacocinética , Pirimidinas , Inhibidores de la Transcriptasa Inversa/farmacocinética
18.
Drug Deliv Transl Res ; 7(6): 796-804, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28497376

RESUMEN

Here, we present the current challenges in women's reproductive health and the current state-of-the-art treatment and prevention options for STI prevention, contraception, and treatment of infections. We discuss how the versatile platform of electrospun fibers can be applied to each challenge, and postulate at how these technologies could be improved. The void of approved electrospun fiber-based products yields the potential to apply this useful technology to a number of medical applications, many of which are relevant to women's reproductive health. Given the ability to tune drug delivery characteristics and three-dimensional geometry, there are many opportunities to pursue new product designs and routes of administration for electrospun fibers. For each application, we provide an overview of the versatility of electrospun fibers as a novel dosage form and summarize their advantages in clinical applications. We also provide a perspective on why electrospun fibers are well-suited for a variety of applications within women's reproductive health and identify areas that could greatly benefit from innovations with electrospun fiber-based approaches.


Asunto(s)
Sistemas de Liberación de Medicamentos/instrumentación , Enfermedades de Transmisión Sexual/tratamiento farmacológico , Enfermedades de Transmisión Sexual/prevención & control , Anticoncepción/métodos , Femenino , Humanos , Salud Reproductiva , Ingeniería de Tejidos , Salud de la Mujer
19.
Nanomedicine ; 13(6): 2015-2025, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28435136

RESUMEN

To prime adaptive immune responses from the female reproductive tract (FRT), particulate antigens must be transported to draining lymph nodes (dLNs) since there are no local organized lymphoid structures equivalent to those found in the respiratory or gastrointestinal tracts. However, little is known about how to safely and effectively navigate successive barriers to transport such as crossing the epithelium and gaining access to migratory cells and lymphatic drainage that provide entry into dLNs. Here, we demonstrate that intravaginal pre-treatment with chitosan significantly facilitates translocation of nanoparticles (NPs) across the multilayered vaginal epithelium to target dLNs. In addition, chitosan pre-treatment was found to enhance NP associations with immunogenic antigen presenting cells in the vaginal submucosa. These observations indicate that chitosan may have great potential as an adjuvant for both local and systemic protective immunity against viral infections in the FRT.


Asunto(s)
Adyuvantes Inmunológicos , Antígenos/inmunología , Quitosano/administración & dosificación , Sistemas de Liberación de Medicamentos , Tejido Linfoide/inmunología , Nanopartículas/administración & dosificación , Vagina/inmunología , Animales , Quitosano/química , Células Dendríticas , Femenino , Ratones Endogámicos C57BL , Nanopartículas/química
20.
Acta Biomater ; 55: 183-193, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28365480

RESUMEN

Self-assembling peptides programed by sequence design to form predefined nanostructures are useful for a variety of biomedical applications. However, assemblies of classic ionic self-complementary peptides are unstable in neutral pH, while charged peptide hydrogels have low mechanical strength. Here, we report on the rational design of a self-assembling peptide system with optimized charge distribution and density for bioscaffold development. Our designer peptides employs a sequence pattern that undergoes salt triggered self-assembly into ß-sheet rich cationic nanofibers in the full pH range (pH 0-14). Our peptides form nanofibrils in physiological condition at a minimum concentration that is significantly lower than has been reported for self-assembly of comparable peptides. The robust fiber-forming ability of our peptides results in the rapid formation of hydrogels in physiological conditions with strong mechanical strength. Moreover, fiber structure is maintained even upon dense conjugation with a model bioactive cargo OVA257-264 peptide. Nanofibers carrying OVA257-264 significantly enhanced CD8+ T cell activation in vitro. Subcutaneous immunization of our peptide fiber vaccine also elicited robust CD8+ T cell activation and proliferation in vivo. Our self-assembling peptides are expected to provide a versatile platform to construct diverse biomaterials. STATEMENT OF SIGNIFICANCE: This work is an attempt of rational design of materials from molecular level for targeted properties and an exploration in molecular self-assembly. Current widely studied self-assembling peptides do not have stable nanofiber structures and form weak hydrogels under physiological conditions. To address this issue, we develop charged self-assembling peptides with a novel sequence pattern for strong fiber-forming ability under physiological conditions. Our designer peptides can undergo salt-triggered self-assembly into nanofibers that are ultrastable in extreme pH (0-14) and dilute solutions, and into hydrogels with strong mechanical strength. Upon conjugation with a model bioactive cargo, our self-assembled peptides exhibit great potential as bioscaffolds for multiple applications.


Asunto(s)
Hidrogeles/química , Nanofibras/química , Péptidos/química , Andamios del Tejido/química , Concentración de Iones de Hidrógeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...