Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Langmuir ; 38(45): 13983-13994, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36318182

RESUMO

Delivery of small molecules and anticancer agents to malignant cells or specific regions within a tumor is limited by penetration depth and poor spatial drug distribution, hindering anticancer efficacy. Herein, we demonstrate control over gold nanoparticle (GNP) penetration and spatial distribution across solid tumors by administering GNPs with different surface chemistries at a constant injection rate via syringe pump. A key finding in this study is the discovery of different zone-specific accumulation patterns of intratumorally injected nanoparticles dependent on surface functionalization. Computed tomography (CT) imaging performed in vivo of C57BL/6 mice harboring Lewis lung carcinoma (LLC) tumors on their flank and gross visualization of excised tumors consistently revealed that intratumorally administered citrate-GNPs accumulate in particle clusters in central areas of the tumor, while GNPs functionalized with thiolated phosphothioethanol (PTE-GNPs) and thiolated polyethylene glycol (PEG-GNPs) regularly accumulate in the tumor periphery. Further, PEG functionalization resulted in larger tumoral surface coverage than PTE, reaching beyond the outer zone of the tumor mass and into the surrounding stroma. To understand the dissimilarities in spatiotemporal evolution across the different GNP surface chemistries, we modeled their intratumoral transport with reaction-diffusion equations. Our results suggest that GNP surface passivation affects nanoparticle reactivity with the tumor microenvironment, leading to differential transport behavior across tumor zones. The present study provides a mechanistic understanding of the factors affecting spatiotemporal distribution of nanoparticles in the tumor. Our proof of concept of zonal delivery within the tumor may prove useful for directing anticancer therapies to regions of biomarker overexpression.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Animais , Camundongos , Ouro , Camundongos Endogâmicos C57BL , Polietilenoglicóis , Ácido Cítrico
2.
Nanomedicine ; 34: 102387, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33753283

RESUMO

A large majority of cardiovascular nanomedicine research has focused on fabricating designer nanoparticles for improved targeting as a means to overcome biological barriers. For cardiac related disorders, such as atherosclerosis, hypertension, and myocardial infarction, designer micro or nanoparticles are often administered into the vasculature or targeted vessel with the hope to circumvent problems associated with conventional drug delivery, including negative systemic side effects. Additionally, novel nano-drug carriers that enter circulation can be selectively uptaken by immune cells with the intended purpose that they modulate inflammatory processes and migrate locally to plaque for therapeutic payload delivery. Indeed, innovative design in nanoparticle composition, formulation, and functionalization has advanced the field as a means to achieve therapeutic efficacy for a variety of cardiac disease indications. This perspective aims to discuss these advances and provide new interpretations of how nanotechnology can be best applied to aid in cardiovascular disease treatment. In an effort to spark discussions on where the field of research should go, we share our outlook in new areas of nanotechnological inclusion and integration, such as in vascular, implantable, or wearable device technologies as well as nanocomposites and nanocoatings. Further, as cardiovascular diseases (CVD) increasingly claim a number of lives globally, we propose more attention should be placed by researchers on nanotechnological approaches for risk factor treatment to aid in early prevention and treatment of CVD.


Assuntos
Doenças Cardiovasculares/terapia , Nanomedicina , Doenças Cardiovasculares/prevenção & controle , Portadores de Fármacos , Humanos , Fatores de Risco
3.
Nanomedicine ; 16: 1-9, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30468870

RESUMO

Globally, 145.2 million people suffer from moderate to severe vision impairment or blindness due to preventable or treatable causes. However, patient adherence to topical or intravitreal treatment is a leading cause of poor outcomes. To address this issue, we designed an intraocularly implantable device called the nanofluidic Vitreal System for Therapeutic Administration (nViSTA) for continuous and controlled drug release based on a nanochannel membrane that obviates the need for pumps or actuation. In vitro release analysis demonstrated that our device achieves sustained release of bimatoprost (BIM) and dexamethasone (DEX) at concentrations within clinically relevant therapeutic window. In this proof of concept study, we constructed an anatomically similar in silico human eye model to simulate DEX release from our implant and gain insight into intraocular pharmacokinetics profile. Overall, our drug-agnostic intraocular implant represents a potentially viable platform for long-term treatment of various chronic ophthalmologic diseases, including diabetic macular edema and uveitis.


Assuntos
Dexametasona/administração & dosagem , Implante de Lente Intraocular/métodos , Edema Macular/tratamento farmacológico , Edema Macular/cirurgia , Sistemas Microeletromecânicos/métodos , Nanotecnologia/métodos , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/cirurgia , Implantes de Medicamento/uso terapêutico , Humanos , Uveíte/tratamento farmacológico , Uveíte/cirurgia
4.
Biomed Microdevices ; 20(2): 49, 2018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29916059

RESUMO

With nearly 40% of U.S. adults obese, and childhood and adolescent rates rising, obesity and associated comorbidities are serious public health concerns with massive societal costs. Often, lifestyle interventions do not offer sufficient weight loss to improve health, requiring surgery and medications as adjunct management strategies. Here, we present a 4-month case study in which the sustained, low-dose, and constant administration of the thyroid receptor ß selective agonist GC-1 (sobetirome) from a novel nanochannel membrane implant was assessed in an obese, pre-diabetic rhesus macaque. Dramatic loss of white adipose tissue in the abdomen from 36 to 18% was observed via magnetic resonance imaging in conjunction with normalized serum insulin and glycemia, with no signs of cardiotoxicity shown. The non-human primate study highlights sustained low-dose delivery of GC-1 from our minimally invasive subcutaneous implant as a valuable approach to induce weight loss and manage obesity and comorbidities, including type 2 diabetes.


Assuntos
Acetatos/metabolismo , Sistemas de Liberação de Medicamentos/instrumentação , Nanotecnologia/instrumentação , Obesidade/metabolismo , Fenóis/metabolismo , Animais , Macaca mulatta
5.
Nanomedicine ; 13(5): 1739-1744, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28259802

RESUMO

This study demonstrated a nanochannel membrane device (NMD) for controlled and sustained release of GC-1 in rats, in the context of the treatment of metabolic syndrome. Release profiles were established in vitro both with and without 5% labrasol for over 2 months. In vivo pharmacokinetic evaluation showed effective GC-1 plasma concentrations, which resulted in significant reductions in body weight after just one week of treatment when compared to the NMD releasing vehicle only (PBS). We also provided evidence that rats treated with NMD-GC-1 present sub-active thyroids and clear differences in the morphology of the epithelium and follicles as compared to the controls, while the heart showed changes in weight. Moreover, body temperatures remained stable throughout treatment, and glucose, pancreatic islet size, and liver histology appeared similar between the treated and control groups. Prolonged constant administration of GC-1 from the NMD proved to be a valid strategy to facilitate weight loss.


Assuntos
Acetatos/farmacocinética , Nanotecnologia , Fenóis/farmacocinética , Acetatos/administração & dosagem , Animais , Peso Corporal , Fígado , Fenóis/administração & dosagem , Ratos , Ratos Endogâmicos F344
6.
Biochem Biophys Res Commun ; 464(3): 718-23, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26168725

RESUMO

Thiazolidinediones (TZDs) are peroxisome proliferator-activated receptor gamma (PPARγ) agonists that improve insulin resistance but trigger side effects such as weight gain, edema, congestive heart failure and bone loss. GQ-16 is a PPARγ partial agonist that improves glucose tolerance and insulin sensitivity in mouse models of obesity and diabetes without inducing weight gain or edema. It is not clear whether GQ-16 acts as a partial agonist at all PPARγ target genes, or whether it displays gene-selective actions. To determine how GQ-16 influences PPARγ activity on a gene by gene basis, we compared effects of rosiglitazone (Rosi) and GQ-16 in mature 3T3-L1 adipocytes using microarray and qRT-PCR. Rosi changed expression of 1156 genes in 3T3-L1, but GQ-16 only changed 89 genes. GQ-16 generally showed weak effects upon Rosi induced genes, consistent with partial agonist actions, but a subset of modestly Rosi induced and strongly repressed genes displayed disproportionately strong GQ-16 responses. PPARγ partial agonists MLR24 and SR1664 also exhibit disproportionately strong effects on transcriptional repression. We conclude that GQ-16 displays a continuum of weak partial agonist effects but efficiently represses some negatively regulated PPARγ responsive genes. Strong repressive effects could contribute to physiologic actions of GQ-16.


Assuntos
Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Células 3T3-L1 , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Expressão Gênica/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Resistência à Insulina/genética , Camundongos , Rosiglitazona
7.
Heliyon ; 10(5): e26354, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38434281

RESUMO

The biomechanical and biochemical processes in the biological systems of living organisms are extremely complex. Advances in understanding these processes are mainly achieved by laboratory and clinical investigations, but in recent decades they are supported by computational modeling. Besides enormous efforts and achievements in this modeling, there still is a need for new methods that can be used in everyday research and medical practice. In this report, we give a view of the generality of the finite element methodology introduced by the first author and supported by his collaborators. It is based on the multiscale smeared physical fields, termed as Kojic Transport Model (KTM), published in several journal papers and summarized in a recent book (Kojic et al., 2022) [1]. We review relevant literature to demonstrate the distinctions and advantages of our methodology and indicate possible further applications. We refer to our published results by a selection of a few examples which include modeling of partitioning, blood flow, molecular transport within the pancreas, multiscale-multiphysics model of coupling electrical field and ion concentration, and a model of convective-diffusive transport within the lung parenchyma. Two new examples include a model of convective-diffusive transport within a growing tumor, and drug release from nanofibers with fiber degradation.

8.
Osteoarthr Cartil Open ; 4(2): 100259, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36475290

RESUMO

Objective: To demonstrate an ultra-high field (UHF) 7 â€‹T delayed gadolinium-enhanced MRI of cartilage (dGEMRIC) protocol for quantitative post-traumatic osteoarthritis (PTOA) detection and monitoring in a rabbit anterior cruciate ligament transection (ACLT) model. Design: ACL transections were performed unilaterally in 5 rabbits (33-weeks-old, 3.5 â€‹± â€‹0.5 â€‹kg) to induce PTOA. MRI exams were performed at 7 â€‹T prior to and 2, 4, 7 and 10-weeks after ACLT using a modified dGEMRIC protocol. Voxel-based T1 and T2 maps were created over manually drawn femoral cartilage ROIs from the center of the tibial plateau to the posterior meniscus. Femoral, tibial, and patellar epiphyses were harvested 10-weeks post-surgery and processed for µCT imaging and histology. Results: Quantitative analysis revealed a 35% and 39% decrease in dGEMRIC index in the medial ACLT knee compartment 7- and 10-weeks post-surgery, respectively (p â€‹= â€‹0.009 and p â€‹= â€‹0.006) when compared to baseline. There was no significant change in the lateral ACLT compartment or in either compartment of the control knees. Visual inspection of histology confirmed PTOA in the ACLT knees. Osteophytes were found only in ACLT knees (osteophyte volume in femur: 94.53 â€‹± â€‹44.08 â€‹mm3, tibia: 29.35 â€‹± â€‹13.79 â€‹mm3, and patella: 3.84 â€‹± â€‹0.92 â€‹mm3) and were significantly larger in the medial compartments of the femur than lateral (p â€‹= â€‹0.0312). Conclusion: The dGEMRIC technique quantitatively applied at 7 â€‹T UHF-MRI demonstrates site-specific cartilage degeneration in a large animal PTOA model. This should encourage further investigation, with potential applications in drug and therapeutic animal trials as well as human studies.

9.
Biomedicines ; 9(11)2021 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-34829790

RESUMO

Biofouling is the unwanted adsorption of cells, proteins, or intracellular and extracellular biomolecules that can spontaneously occur on the surface of metal nanocomplexes. It represents a major issue in bioinorganic chemistry because it leads to the creation of a protein corona, which can destabilize a colloidal solution and result in undesired macrophage-driven clearance, consequently causing failed delivery of a targeted drug cargo. Hyaluronic acid (HA) is a bioactive, natural mucopolysaccharide with excellent antifouling properties, arising from its hydrophilic and polyanionic characteristics in physiological environments which prevent opsonization. In this study, hyaluronate-thiol (HA-SH) (MW 10 kDa) was used to surface-passivate gold nanoparticles (GNPs) synthesized using a citrate reduction method. HA functionalized GNP complexes (HA-GNPs) were characterized using absorption spectroscopy, scanning electron microscopy, zeta potential, and dynamic light scattering. GNP cellular uptake and potential dose-dependent cytotoxic effects due to treatment were evaluated in vitro in HeLa cells using inductively coupled plasma-optical emission spectrometry (ICP-OES) and trypan blue and MTT assays. Further, we quantified the in vivo biodistribution of intratumorally injected HA functionalized GNPs in Lewis Lung carcinoma (LLC) solid tumors grown on the flank of C57BL/6 mice and compared localization and retention with nascent particles. Our results reveal that HA-GNPs show overall greater peritumoral distribution (** p < 0.005, 3 days post-intratumoral injection) than citrate-GNPs with reduced biodistribution in off-target organs. This property represents an advantageous step forward in localized delivery of metal nano-complexes to the infiltrative region of a tumor, which may improve the application of nanomedicine in the diagnosis and treatment of cancer.

10.
Pharmaceutics ; 13(2)2021 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-33562434

RESUMO

The heterogeneous distribution of delivery or treatment modalities within the tumor mass is a crucial limiting factor for a vast range of theranostic applications. Understanding the interactions between a nanomaterial and the tumor microenvironment will help to overcome challenges associated with tumor heterogeneity, as well as the clinical translation of nanotheranostic materials. This study aims to evaluate the influence of protein surface adsorption on gold nanoparticle (GNP) biodistribution using high-resolution computed tomography (CT) preclinical imaging in C57BL/6 mice harboring Lewis lung carcinoma (LLC) tumors. LLC provides a valuable model for study due to its highly heterogenous nature, which makes drug delivery to the tumor challenging. By controlling the adsorption of proteins on the GNP surface, we hypothesize that we can influence the intratumoral distribution pattern and particle retention. We performed an in vitro study to evaluate the uptake of GNPs by LLC cells and an in vivo study to assess and quantify the GNP biodistribution by injecting concentrated GNPs citrate-stabilized or passivated with bovine serum albumin (BSA) intratumorally into LLC solid tumors. Quantitative CT and inductively coupled plasma optical emission spectrometry (ICP-OES) results both confirm the presence of particles in the tumor 9 days post-injection (n = 8 mice/group). A significant difference is highlighted between citrate-GNP and BSA-GNP groups (** p < 0.005, Tukey's multiple comparisons test), confirming that the protein corona of GNPs modifies intratumoral distribution and retention of the particles. In conclusion, our investigations show that the surface passivation of GNPs influences the mechanism of cellular uptake and intratumoral distribution in vivo, highlighting the spatial heterogeneity of the solid tumor.

11.
Nanomaterials (Basel) ; 10(9)2020 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-32872626

RESUMO

It has been suggested that particle size plays an important role in determining the genotoxicity of gold nanoparticles (GNPs). The purpose of this study was to compare the potential radio-sensitization effects of two different sized GNPs (3.9 and 37.4 nm) fabricated and examined in vitro in Lewis lung carcinoma (LLC) as a model of non-small cell lung cancer through use of comet and clonogenic assays. After treatment with 2Gy X-ray irradiation, both particle sizes demonstrated increased DNA damage when compared to treatment with particles only and radiation alone. This radio-sensitization was further translated into a reduction in cell survival demonstrated by clonogenicity. This work indicates that GNPs of both sizes induce DNA damage in LLC cells at the tested concentrations, whereas the 37.4 nm particle size treatment group demonstrated greater significance in vitro. The presented data aids in the evaluation of the radiobiological response of Lewis lung carcinoma cells treated with gold nanoparticles.

12.
ACS Chem Biol ; 15(11): 2916-2928, 2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33074669

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is the predominant form of pancreatic cancer. PDACs harbor oncogenic mutations in the KRAS gene, and ongoing efforts to directly target its mutant protein product to inhibit tumor growth are a priority not only in pancreatic cancer but in other malignancies such as lung and colorectal cancers where KRAS is also commonly mutated. An alternative strategy to directly targeting KRAS is to identify and target druggable receptors involved in dysregulated cancer hallmarks downstream of KRAS dysregulation. Liver X receptors (LXRs) are members of the nuclear receptor family of ligand-modulated transcription factors and are involved in the regulation of genes which function in key cancer-related processes, including cholesterol transport, lipid and glucose metabolism, and inflammatory and immune responses. Modulation of LXRs via small molecule ligands has emerged as a promising approach for directly targeting tumor cells or the stromal and immune cells within the tumor microenvironment. We have previously shown that only one of the two LXR subtypes (LXRß) is expressed in pancreatic cancer cells, and targeting LXR with available synthetic ligands blocked the proliferation of PDAC cells and tumor formation. In a screen of a focused library of drug-like small molecules predicted to dock in the ligand-binding pocket of LXRß, we identified two novel LXR ligands with more potent antitumor activity than current LXR agonists used in our published studies. Characterization of the two lead compounds (GAC0001E5 and GAC0003A4) indicates that they function as LXR inverse agonists which inhibit their transcriptional activity. Prolonged treatments with novel ligands further revealed their function as LXR "degraders" which significantly reduced LXR protein levels in all three PDAC cell lines tested. These findings support the utility of these novel inhibitors in basic research on ligand design, allosteric mechanisms, and LXR functions and their potential application as treatments for advanced pancreatic cancer and other recalcitrant malignancies.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Receptores X do Fígado/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , Antineoplásicos/química , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Agonismo Inverso de Drogas , Humanos , Ligantes , Receptores X do Fígado/agonistas , Neoplasias Pancreáticas/metabolismo , Proteólise/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química
13.
Adv Drug Deliv Rev ; 151-152: 222-232, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30797957

RESUMO

The pericardium, which surrounds the heart, provides a unique enclosed volume and a site for the delivery of agents to the heart and coronary arteries. While strategies for targeting the delivery of therapeutics to the heart are lacking, various technologies and nanodelivery approaches are emerging as promising methods for site specific delivery to increase therapeutic myocardial retention, efficacy, and bioactivity, while decreasing undesired systemic effects. Here, we provide a literature review of various approaches for intrapericardial delivery of agents. Emphasis is given to sustained delivery approaches (pumps and catheters) and localized release (patches, drug eluting stents, and support devices and meshes). Further, minimally invasive access techniques, pericardial access devices, pericardial washout and fluid analysis, as well as therapeutic and cell delivery vehicles are presented. Finally, several promising new therapeutic targets to treat heart diseases are highlighted.


Assuntos
Cardiotônicos/uso terapêutico , Sistemas de Liberação de Medicamentos , Cardiopatias/tratamento farmacológico , Animais , Cardiotônicos/administração & dosagem , Humanos , Injeções Intraperitoneais
14.
Lab Chip ; 19(13): 2192-2204, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31169840

RESUMO

Chronic diseases such as hypertension and rheumatoid arthritis are persistent ailments that require personalized lifelong therapeutic management. However, the difficulty of adherence to strict dosing schedule compromises therapeutic efficacy and safety. Moreover, the conventional one-size-fits-all treatment approach is increasingly challenged due to the intricacies of inter- and intra-individual variabilities. While accelerated technological advances have led to sophisticated implantable drug delivery devices, flexibility in dosage and timing modulation to tailor precise treatment to individual needs remains an elusive goal. Here we describe the development of a subcutaneously implantable remote-controlled nanofluidic device capable of sustained drug release with adjustable dosing and timing. By leveraging a low intensity electric field to modify the concentration driven diffusion across a nanofluidic membrane, the rate of drug administration can be increased, decreased or stopped via Bluetooth remote command. We demonstrate in vitro the release modulation of enalapril and methotrexate, first-line therapeutics for treatment of hypertension and rheumatoid arthritis, respectively. Further, we show reliable remote communication and device biocompatibility via in vivo studies. Unlike a pulsatile release regimen typical of some conventional controlled delivery systems, our implant offers a continuous drug administration that avoids abrupt fluctuations, which could affect response and tolerability. Our system could set the foundation for an on-demand delivery platform technology for long term management of chronic diseases.


Assuntos
Sistemas de Liberação de Medicamentos , Técnicas Analíticas Microfluídicas , Nanotecnologia , Sistemas de Liberação de Medicamentos/instrumentação , Desenho de Equipamento , Técnicas Analíticas Microfluídicas/instrumentação , Nanotecnologia/instrumentação
15.
J Control Release ; 285: 23-34, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30008369

RESUMO

Conventional systemic immunotherapy administration often results in insufficient anti-tumor immune response and adverse side effects. Delivering immunotherapeutics intratumorally could maximize tumor exposure, elicit efficient anti-tumor immune response, and minimize toxicity. To fulfill the unmet clinical need for sustained local drug delivery and to avoid repeated intratumoral injections, we developed a nanofluidic-based device for intratumoral drug delivery called the nanofluidic drug-eluting seed (NDES). The NDES is inserted intratumorally using a minimally invasive trocar method similar to brachytherapy seed insertion and offers a clinical advantage of drug elution. Drug diffusion from the NDES is regulated by physical and electrostatic nanoconfinement, thereby resulting in constant and sustained immunotherapeutic delivery without the need for injections or clinician intervention. In this study, the NDES was used to deliver immunotherapeutics intratumorally in the 4 T1 orthotopic murine mammary carcinoma model, which recapitulates triple negative breast cancer. We demonstrated that NDES-mediated intratumoral release of agonist monoclonal antibodies, OX40 and CD40, resulted in potentiation of local and systemic anti-tumor immune response and inhibition of tumor growth compared to control mice. Further, mice treated with NDES-CD40 demonstrated minimal liver damage compared to systemically treated mice. Collectively, our study highlights the NDES as an effective platform for sustained intratumoral immunotherapeutic delivery. The potential clinical impact is tremendous given that the NDES is applicable to a broad spectrum of drugs and solid tumors.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Sistemas de Liberação de Medicamentos/instrumentação , Implantes de Medicamento , Imunoterapia/instrumentação , Neoplasias de Mama Triplo Negativas/terapia , Animais , Antineoplásicos Imunológicos/uso terapêutico , Implantes de Medicamento/química , Desenho de Equipamento , Feminino , Camundongos Endogâmicos BALB C , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/patologia
16.
J Control Release ; 286: 315-325, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30092254

RESUMO

Pre-exposure prophylaxis (PrEP) with antiretroviral (ARV) drugs are effective at preventing human immunodeficiency virus (HIV) transmission. However, implementation of PrEP presents significant challenges due to poor user adherence, low accessibility to ARVs and multiple routes of HIV exposure. To address these challenges, we developed the nanochannel delivery implant (NDI), a subcutaneously implantable device for sustained and constant delivery of tenofovir alafenamide (TAF) and emtricitabine (FTC) for HIV PrEP. Unlike existing drug delivery platforms with finite depots, the NDI incorporates ports allowing for transcutaneous refilling upon drug exhaustion. NDI-mediated drug delivery in rhesus macaques resulted in sustained release of both TAF and FTC for 83 days, as indicated by concentrations of TAF, FTC and their respectively metabolites in plasma, PBMCs, rectal mononuclear cells and tissues associated with HIV transmission. Notably, clinically relevant preventative levels of tenofovir diphosphate were achieved as early as 3 days after NDI implantation. We also demonstrated the feasibility of transcutaneous drug refilling to extend the duration of PrEP drug delivery in NHPs. Overall, the NDI represents an innovative strategy for long-term HIV PrEP administration in both developed and developing countries.


Assuntos
Adenina/análogos & derivados , Antivirais/administração & dosagem , Sistemas de Liberação de Medicamentos/instrumentação , Emtricitabina/administração & dosagem , Infecções por HIV/prevenção & controle , Bombas de Infusão Implantáveis , Dispositivos Lab-On-A-Chip , Organofosfatos/administração & dosagem , Adenina/administração & dosagem , Adenina/sangue , Adenina/farmacocinética , Administração Cutânea , Animais , Antivirais/sangue , Antivirais/farmacocinética , Emtricitabina/sangue , Emtricitabina/farmacocinética , Desenho de Equipamento , Humanos , Macaca mulatta , Organofosfatos/sangue , Organofosfatos/farmacocinética , Profilaxia Pré-Exposição
18.
Nanoscale ; 8(44): 18718-18725, 2016 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-27787528

RESUMO

We report an electro-nanofluidic membrane for tunable, ultra-low power drug delivery employing an ionic field effect transistor. Therapeutic release from a drug reservoir was successfully modulated, with high energy efficiency, by actively adjusting the surface charge of slit-nanochannels 50, 110, and 160 nm in size, by the polarization of a buried gate electrode and the consequent variation of the electrical double layer in the nanochannel. We demonstrated control over the transport of ionic species, including two relevant hypertension drugs, atenolol and perindopril, that could benefit from such modulation. By leveraging concentration-driven diffusion, we achieve a 2 to 3 order of magnitude reduction in power consumption as compared to other electrokinetic phenomena. The application of a small gate potential (±5 V) in close proximity (150 nm) of 50 nm nanochannels generated a sufficiently strong electric field, which doubled or blocked the ionic flux depending on the polarity of the voltage applied. These compelling findings can lead to next generation, more reliable, smaller, and longer lasting drug delivery implants with ultra-low power consumption.

19.
J Tissue Eng ; 7: 2041731416638198, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27152147

RESUMO

Diabetes is one of the most prevalent, costly, and debilitating diseases in the world. Pancreas and islet transplants have shown success in re-establishing glucose control and reversing diabetic complications. However, both are limited by donor availability, need for continuous immunosuppression, loss of transplanted tissue due to dispersion, and lack of vascularization. To overcome the limitations of poor islet availability, here, we investigate the potential of bone marrow-derived mesenchymal stem cells differentiated into islet-like insulin-producing aggregates. Islet-like insulin-producing aggregates, characterized by gene expression, are shown to be similar to pancreatic islets and display positive immunostaining for insulin and glucagon. To address the limits of current encapsulation systems, we developed a novel three-dimensional printed, scalable, and potentially refillable polymeric construct (nanogland) to support islet-like insulin-producing aggregates' survival and function in the host body. In vitro studies showed that encapsulated islet-like insulin-producing aggregates maintained viability and function, producing steady levels of insulin for at least 4 weeks. Nanogland-islet-like insulin-producing aggregate technology here investigated as a proof of concept holds potential as an effective and innovative approach for diabetes cell therapy.

20.
Nucl Recept Signal ; 13: e004, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26445566

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) display anti-inflammatory, antipyretic and analgesic properties by inhibiting cyclooxygenases and blocking prostaglandin production. Previous studies, however, suggested that some NSAIDs also modulate peroxisome proliferator activated receptors (PPARs), raising the possibility that such off target effects contribute to the spectrum of clinically relevant NSAID actions. In this study, we set out to understand how peroxisome proliferator activated receptor-γ (PPARγ/PPARG) interacts with NSAIDs using X-ray crystallography and to relate ligand binding modes to effects on receptor activity. We find that several NSAIDs (sulindac sulfide, diclofenac, indomethacin and ibuprofen) bind PPARγ and modulate PPARγ activity at pharmacologically relevant concentrations. Diclofenac acts as a partial agonist and binds to the PPARγ ligand binding pocket (LBP) in typical partial agonist mode, near the ß-sheets and helix 3. By contrast, two copies of indomethacin and sulindac sulfide bind the LBP and, in aggregate, these ligands engage in LBP contacts that resemble agonists. Accordingly, both compounds, and ibuprofen, act as strong partial agonists. Assessment of NSAID activities in PPARγ-dependent 3T3-L1 cells reveals that NSAIDs display adipogenic activities and exclusively regulate PPARγ-dependent target genes in a manner that is consistent with their observed binding modes. Further, PPARγ knockdown eliminates indomethacin activities at selected endogenous genes, confirming receptor-dependence of observed effects. We propose that it is important to consider how individual NSAIDs interact with PPARγ to understand their activities, and that it will be interesting to determine whether high dose NSAID therapies result in PPAR activation.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , PPAR gama/metabolismo , Células 3T3-L1 , Adipogenia/efeitos dos fármacos , Animais , Agonismo Parcial de Drogas , Células HeLa , Humanos , Camundongos , Modelos Moleculares , PPAR gama/agonistas , PPAR gama/química , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA