Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 617, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38778159

ABSTRACT

The question of whether material stiffness enhances cell adhesion and clustering is still open to debate. Results from the literature are seemingly contradictory, with some reports illustrating that adhesion increases with surface stiffness and others suggesting that the performance of a system of cells is curbed by high values of elasticity. To address the role of elasticity as a regulator in neuronal cell adhesion and clustering, we investigated the topological characteristics of networks of neurons on polydimethylsiloxane (PDMS) surfaces - with values of elasticity (E) varying in the 0.55-2.65 MPa range. Results illustrate that, as elasticity increases, the number of neurons adhering on the surface decreases. Notably, the small-world coefficient - a topological measure of networks - also decreases. Numerical simulations and functional multi-calcium imaging experiments further indicated that the activity of neuronal cells on soft surfaces improves for decreasing E. Experimental findings are supported by a mathematical model, that explains adhesion and clustering of cells on soft materials as a function of few parameters - including the Young's modulus and roughness of the material. Overall, results indicate that - in the considered elasticity interval - increasing the compliance of a material improves adhesion, improves clustering, and enhances communication of neurons.


Subject(s)
Cell Adhesion , Elasticity , Neurons , Neurons/physiology , Animals , Dimethylpolysiloxanes/chemistry , Surface Properties , Elastic Modulus , Cells, Cultured , Rats
2.
Int J Mol Sci ; 24(12)2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37373273

ABSTRACT

The deposition of stem cells at sites of injury is a clinically relevant approach to facilitate tissue repair and angiogenesis. However, insufficient cell engraftment and survival require the engineering of novel scaffolds. Here, a regular network of microscopic poly(lactic-co-glycolic acid) (PLGA) filaments was investigated as a promising biodegradable scaffold for human Adipose-Derived Stem Cell (hADSC) tissue integration. Via soft lithography, three different microstructured fabrics were realized where 5 × 5 and 5 × 3 µm PLGA 'warp' and 'weft' filaments crossed perpendicularly with pitch distances of 5, 10 and 20 µm. After hADSC seeding, cell viability, actin cytoskeleton, spatial organization and the secretome were characterized and compared to conventional substrates, including collagen layers. On the PLGA fabric, hADSC re-assembled to form spheroidal-like structures, preserving cell viability and favoring a nonlinear actin organization. Moreover, the secretion of specific factors involved in angiogenesis, the remodeling of the extracellular matrix and stem cell homing was favored on the PLGA fabric as compared to that which occurred on conventional substrates. The paracrine activity of hADSC was microstructure-dependent, with 5 µm PLGA fabric enhancing the expression of factors involved in all three processes. Although more studies are needed, the proposed PLGA fabric would represent a promising alternative to conventional collagen substrates for stem cell implantation and angiogenesis induction.


Subject(s)
Polyglycolic Acid , Tissue Scaffolds , Humans , Polylactic Acid-Polyglycolic Acid Copolymer , Tissue Scaffolds/chemistry , Polyglycolic Acid/chemistry , Lactic Acid/chemistry , Tissue Engineering , Cells, Cultured , Collagen/chemistry , Stem Cells/ultrastructure
3.
ACS Nano ; 17(15): 14572-14585, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37379253

ABSTRACT

Modest tissue penetrance, nonuniform distribution, and suboptimal release of drugs limit the potential of intracranial therapies against glioblastoma. Here, a conformable polymeric implant, µMESH, is realized by intercalating a micronetwork of 3 × 5 µm poly(lactic-co-glycolic acid) (PLGA) edges over arrays of 20 × 20 µm polyvinyl alcohol (PVA) pillars for the sustained delivery of potent chemotherapeutic molecules, docetaxel (DTXL) and paclitaxel (PTXL). Four different µMESH configurations were engineered by encapsulating DTXL or PTXL within the PLGA micronetwork and nanoformulated DTXL (nanoDTXL) or PTXL (nanoPTXL) within the PVA microlayer. All four µMESH configurations provided sustained drug release for at least 150 days. However, while a burst release of up to 80% of nanoPTXL/nanoDTXL was documented within the first 4 days, molecular DTXL and PTXL were released more slowly from µMESH. Upon incubation with U87-MG cell spheroids, DTXL-µMESH was associated with the lowest lethal drug dose, followed by nanoDTXL-µMESH, PTXL-µMESH, and nanoPTXL-µMESH. In orthotopic models of glioblastoma, µMESH was peritumorally deposited at 15 days post-cell inoculation and tumor proliferation was monitored via bioluminescence imaging. The overall animal survival increased from ∼30 days of the untreated controls to 75 days for nanoPTXL-µMESH and 90 days for PTXL-µMESH. For the DTXL groups, the overall survival could not be defined as 80% and 60% of the animals treated with DTXL-µMESH and nanoDTXL-µMESH were still alive at 90 days, respectively. These results suggest that the sustained delivery of potent drugs properly encapsulated in conformable polymeric implants could halt the proliferation of aggressive brain tumors.


Subject(s)
Glioblastoma , Nanoparticles , Animals , Glioblastoma/drug therapy , Glioblastoma/pathology , Pharmaceutical Preparations , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Docetaxel/therapeutic use , Polymers/therapeutic use , Polyvinyl Alcohol , Cell Line, Tumor
5.
Eur J Pharm Biopharm ; 174: 90-100, 2022 May.
Article in English | MEDLINE | ID: mdl-35358697

ABSTRACT

Maximizing loading while modulating the release of therapeutic molecules from nanoparticles and implantable drug delivery systems is the key to successfully address deadly diseases like brain cancer. Here, four different conjugates of the potent chemotherapeutic molecule docetaxel (DTXL)were realized to optimize the pharmacological properties of 1,000 × 400 nmDiscoidal PolymericNanoconstructs(DPNs). DTXL was covalently linked to poly-(ethylene) glycol(PEG)chains of different molecular weights, namely 350, 550 and 1,000 Da, and oleic acid (OA). After extensive physico-chemical and pharmacological characterizations, the conjugate PEG550-DTXL showedan optimal compromise between loading and sustained release out of DPNs, as opposed to the insufficient loading of PEG1000-DTXL and PEG350-DTXL and the excessively slow release of OA-DTXL. Not surprisingly, viability tests conducted on U87-MG cells showed a delay in cytotoxic activity for the DTXL conjugates compared to free DTXL within the first 48 h. However, PEG550-DTXL returned an IC50 value of âˆ¼ 10 nMat 72 h, which is comparable to free DTXL.In mice bearing orthotopically implanted U87-MG cells, the intravenous administration of PEG550-DTXL loaded DPNs doubled the overall animal survival (52.5 days) as compared to temozolomide (27 days) and the untreated controls (32 days). Collectively, these results continue to demonstrate that the therapeutic efficacy of nanoparticles can be boosted by rationally designing drug conjugates-particle complexes for optimal loading and release profiles.


Subject(s)
Antineoplastic Agents , Glioblastoma , Nanoparticles , Animals , Docetaxel , Glioblastoma/drug therapy , Mice , Nanoparticles/chemistry , Oleic Acid , Polyethylene Glycols/chemistry , Taxoids/pharmacology
6.
Drug Deliv Transl Res ; 12(8): 2019-2037, 2022 08.
Article in English | MEDLINE | ID: mdl-35284984

ABSTRACT

The storied history of controlled the release systems has evolved over time; from degradable drug-loaded sutures to monolithic zero-ordered release devices and nano-sized drug delivery formulations. Scientists have tuned the physico-chemical properties of these drug carriers to optimize their performance in biomedical/pharmaceutical applications. In particular, particle drug delivery systems at the micron size regime have been used since the 1980s. Recent advances in micro and nanofabrication techniques have enabled precise control of particle size and geometry-here we review the utility of microplates and discoidal polymeric particles for a range of pharmaceutical applications. Microplates are defined as micrometer scale polymeric local depot devices in cuboid form, while discoidal polymeric nanoconstructs are disk-shaped polymeric particles having a cross-sectional diameter in the micrometer range and a thickness in the hundreds of nanometer range. These versatile particles can be used to treat several pathologies such as cancer, inflammatory diseases and vascular diseases, by leveraging their size, shape, physical properties (e.g., stiffness), and component materials, to tune their functionality. This review highlights design and fabrication strategies for these particles, discusses their applications, and elaborates on emerging trends for their use in formulations.


Subject(s)
Drug Carriers , Drug Delivery Systems , Drug Carriers/chemistry , Drug Compounding , Drug Delivery Systems/methods , Particle Size , Polymers/chemistry
7.
ACS Appl Mater Interfaces ; 13(45): 53618-53629, 2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34751556

ABSTRACT

Type-1 diabetes (T1DM) is a chronic metabolic disorder resulting from the autoimmune destruction of ß cells. The current standard of care requires multiple, daily injections of insulin and accurate monitoring of blood glucose levels (BGLs); in some cases, this results in diminished patient compliance and increased risk of hypoglycemia. Herein, we engineered hierarchically structured particles comprising a poly(lactic-co-glycolic) acid (PLGA) prismatic matrix, with a 20 × 20 µm base, encapsulating 200 nm insulin granules. Five configurations of these insulin-microPlates (INS-µPLs) were realized with different heights (5, 10, and 20 µm) and PLGA contents (10, 40, and, 60 mg). After detailed physicochemical and biopharmacological characterizations, the tissue-compliant 10H INS-µPL, realized with 10 mg of PLGA, presented the most effective release profile with ∼50% of the loaded insulin delivered at 4 weeks. In diabetic mice, a single 10H INS-µPL intraperitoneal deposition reduced BGLs to that of healthy mice within 1 h post-implantation (167.4 ± 49.0 vs 140.0 ± 9.2 mg/dL, respectively) and supported normoglycemic conditions for about 2 weeks. Furthermore, following the glucose challenge, diabetic mice implanted with 10H INS-µPL successfully regained glycemic control with a significant reduction in AUC0-120min (799.9 ± 134.83 vs 2234.60 ± 82.72 mg/dL) and increased insulin levels at 7 days post-implantation (1.14 ± 0.11 vs 0.38 ± 0.02 ng/mL), as compared to untreated diabetic mice. Collectively, these results demonstrate that INS-µPLs are a promising platform for the treatment of T1DM to be further optimized with the integration of smart glucose sensors.


Subject(s)
Biocompatible Materials/pharmacology , Blood Glucose/analysis , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Polyglycolic Acid/pharmacology , Animals , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/metabolism , Dose-Response Relationship, Drug , Hypoglycemia/chemically induced , Hypoglycemia/drug therapy , Hypoglycemia/metabolism , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Insulin/chemistry , Male , Mice , Mice, Inbred C57BL , Particle Size , Polyglycolic Acid/chemistry , Streptozocin
8.
ACS Appl Mater Interfaces ; 13(27): 31379-31392, 2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34197081

ABSTRACT

Osteoarthritis (OA) is treated with the intra-articular injection of steroids such as dexamethasone (DEX) to provide short-term pain management. However, DEX treatment suffers from rapid joint clearance. Here, 20 × 10 µm, shape-defined poly(d,l-lactide-co-glycolide)acid microPlates (µPLs) are created and intra-articularly deposited for the sustained release of DEX. Under confined conditions, DEX release is projected to persist for several months, with only ∼20% released in the first month. In a highly rigorous murine knee overload injury model (post-traumatic osteoarthritis), a single intra-articular injection of Cy5-µPLs is detected in the cartilage surface, infrapatellar fat pad/synovium, joint capsule, and posterior joint space up to 30 days. One intra-articular injection of DEX-µPL (1 mg kg-1) decreased the expression of interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, IL-6, and matrix metalloproteinase (MMP)-13 by approximately half compared to free DEX at 4 weeks post-treatment. DEX-µPL also reduced load-induced histological changes in the articular cartilage and synovial tissues relative to saline or free DEX. In sum, the µPLs provide sustained drug release along with the capability to precisely control particle geometry and mechanical properties, yielding long-lasting benefits in overload-induced OA. This work motivates further study and development of particles that provide combined pharmacological and mechanical benefits.


Subject(s)
Cartilage, Articular/metabolism , Dexamethasone/chemistry , Dexamethasone/metabolism , Drug Carriers/chemistry , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Animals , Biomarkers/metabolism , Delayed-Action Preparations , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Gene Expression Regulation/drug effects , Injections, Intra-Articular , Mice , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Stress, Mechanical
9.
Nat Nanotechnol ; 16(7): 820-829, 2021 07.
Article in English | MEDLINE | ID: mdl-33795849

ABSTRACT

The poor transport of molecular and nanoscale agents through the blood-brain barrier together with tumour heterogeneity contribute to the dismal prognosis in patients with glioblastoma multiforme. Here, a biodegradable implant (µMESH) is engineered in the form of a micrometre-sized poly(lactic-co-glycolic acid) mesh laid over a water-soluble poly(vinyl alcohol) layer. Upon poly(vinyl alcohol) dissolution, the flexible poly(lactic-co-glycolic acid) mesh conforms to the resected tumour cavity as docetaxel-loaded nanomedicines and diclofenac molecules are continuously and directly released into the adjacent tumour bed. In orthotopic brain cancer models, generated with a conventional, reference cell line and patient-derived cells, a single µMESH application, carrying 0.75 mg kg-1 of docetaxel and diclofenac, abrogates disease recurrence up to eight months after tumour resection, with no appreciable adverse effects. Without tumour resection, the µMESH increases the median overall survival (∼30 d) as compared with the one-time intracranial deposition of docetaxel-loaded nanomedicines (15 d) or 10 cycles of systemically administered temozolomide (12 d). The µMESH modular structure, for the independent coloading of different molecules and nanomedicines, together with its mechanical flexibility, can be exploited to treat a variety of cancers, realizing patient-specific dosing and interventions.


Subject(s)
Absorbable Implants , Antineoplastic Combined Chemotherapy Protocols , Brain Neoplasms/drug therapy , Polylactic Acid-Polyglycolic Acid Copolymer , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line , Diclofenac/pharmacokinetics , Diclofenac/pharmacology , Docetaxel/pharmacokinetics , Docetaxel/pharmacology , Drug Implants/pharmacokinetics , Drug Implants/pharmacology , Female , Humans , Mice , Mice, Nude , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Xenograft Model Antitumor Assays
10.
Biofabrication ; 13(3)2021 04 07.
Article in English | MEDLINE | ID: mdl-33455953

ABSTRACT

Islet transplantation is a promising approach to enable type 1 diabetic patients to attain glycemic control independent of insulin injections. However, up to 60% of islets are lost immediately following transplantation. To improve this outcome, islets can be transplanted within bioscaffolds, however, synthetic bioscaffolds induce an intense inflammatory reaction which can have detrimental effects on islet function and survival. In the present study, we first improved the biocompatibility of polydimethylsiloxane (PDMS) bioscaffolds by coating them with collagen. To reduce the inflammatory response to PDMS bioscaffolds, we then enriched the bioscaffolds with dexamethasone-loaded microplates (DEX-µScaffolds). These DEX-microplates have the ability to release DEX in a sustained manner over 7 weeks within a therapeutic range that does not affect the glucose responsiveness of the islets but which minimizes inflammation in the surrounding microenvironment. The bioscaffold showed excellent mechanical properties that enabled it to resist pore collapse thereby helping to facilitate islet seeding and its handling for implantation, and subsequent engraftment, within the epididymal fat pad (EFP). Following the transplantation of islets into the EFP of diabetic mice using DEX-µScaffolds there was a return in basal blood glucose to normal values by day 4, with normoglycemia maintained for 30 d. Furthermore, these animals demonstrated a normal dynamic response to glucose challenges with histological evidence showing reduced pro-inflammatory cytokines and fibrotic tissue surrounding DEX-µScaffolds at the transplantation site. In contrast, diabetic animals transplanted with either islets alone or islets in bioscaffolds without DEX microplates were not able to regain glycemic control during basal conditions with overall poor islet function. Taken together, our data show that coating PDMS bioscaffolds with collagen, and enriching them with DEX-microplates, significantly prolongs and enhances islet function and survival.


Subject(s)
Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Islets of Langerhans , Animals , Collagen/pharmacology , Dexamethasone/pharmacology , Diabetes Mellitus, Experimental/therapy , Dimethylpolysiloxanes , Humans , Insulin , Mice
11.
Comput Biol Med ; 123: 103820, 2020 08.
Article in English | MEDLINE | ID: mdl-32658778

ABSTRACT

We address the problem of determining from laboratory experiments the data necessary for a proper modeling of drug delivery and efficacy in anticancer therapy. There is an inherent difficulty in extracting the necessary parameters, because the experiments often yield an insufficient quantity of information. To overcome this difficulty, we propose to combine real experiments, numerical simulation, and Machine Learning (ML) based on Artificial Neural Networks (ANN), aiming at a reliable identification of the physical model factors, e.g. the killing action of the drug. To this purpose, we exploit the employed mathematical-numerical model for tumor growth and drug delivery, together with the ANN - ML procedure, to integrate the results of the experimental tests and feed back the model itself, thus obtaining a reliable predictive tool. The procedure represents a hybrid data-driven, physics-informed approach to machine learning. The physical and mathematical model employed for the numerical simulations is without extracellular matrix (ECM) and healthy cells because of the experimental conditions we reproduce.


Subject(s)
Machine Learning , Pharmaceutical Preparations , Computer Simulation , Models, Theoretical , Neural Networks, Computer
12.
J Control Release ; 319: 201-212, 2020 03 10.
Article in English | MEDLINE | ID: mdl-31899267

ABSTRACT

Over the years, nanoparticles, microparticles, implants of poly(D,l-lactide-co-glycolide) (PLGA) have been demonstrated for diverse biomedical applications. Yet, initial burst release and optimal modulation of the release profiles limit their clinical use. Here, shape-defined PLGA microPlates (µPLs) were realized for the sustained release of two anti-inflammatory molecules, the natural polyphenol curcumin (CURC) and the corticosteroid dexamethasone (DEX). Under the electron microscope, µPLs appeared as square prisms with an edge length of 20 µm. The top-down fabrication process allowed the authors to vary, readily and systematically, the µPL height from 5 to 10 µm and the PLGA mass from 1 to 5, 10 and 20 mg. 'Taller' particles realized with higher PLGA concentrations encapsulated more drug reaching on average values of about 150 pg/µPL, for both CURC and DEX. The µPL height and PLGA concentration had major effects on drug release, too. Under sink conditions, DEX release from tall µPLs at 1 h reduced from 50% to 10% and 2% for the 5, 10 and 20 mg PLGA configurations, respectively. Also, DEX was released more slowly from taller as compared to short µPLs. The opposite trend was observed for CURC, possibly for its lower hydrophobicity and molecular weight as compared to DEX. This was also confirmed by quantifying the free energy of translocation for the two drugs via molecular dynamics simulations. Finally, the anti-inflammatory activity of µPLs was tested in vitro on LPS-stimulated rat monocytes and in vivo on a murine model of UVB-induced skin burns. Both in vitro and in vivo, the expression of pro-inflammatory cytokines (IL-6, IL-1ß, and TNF-α) was significantly reduced by the application of µPLs as compared to the free compounds. In vivo, one single topical deposition of CURC-µPLs outperformed multiple, free CURC applications. This work demonstrates that geometry and polymer density can be effectively used to modulate the pharmacological performance of microparticles and mitigate the initial burst release.


Subject(s)
Lactic Acid , Polyglycolic Acid , Animals , Anti-Inflammatory Agents , Delayed-Action Preparations , Drug Liberation , Mice , Particle Size , Rats
13.
Nanoscale Adv ; 1(4): 1541-1552, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-31304459

ABSTRACT

Polymeric micro and nanoconstructs are emerging as promising delivery systems for therapeutics and contrast agents in microcirculation. Excellent assets associated with polymeric particulates of tunable shape, size, mechanical and chemical properties may improve the efficiency of delivery and represent the basis of personalized medicine and treatment. Nevertheless, lack of effective techniques of analysis may limit their use in biomedicine and bioengineering. In this paper, we demonstrated Raman Spectroscopy for quantitative characterization of poly lactic-co-glycolic acid (PLGA) micro-plate drug delivery systems. To do so, we (i) acquired bi-dimensional Raman maps of PLGA micro-plates loaded with curcumin at various times of release over multiple particles. We (ii) realized an exploratory analysis of data using the principal component analysis (PCA) technique to find hidden patterns in the data and reduce the dimensionality of the system. Then, we (iii) used an innovative univariate method of analysis of the reduced system to derive quantitative drug release profiles. High performance liquid chromatography (HPLC), the consolidated method of analysis of macro-sized systems, was used for comparison. We found that our system is as efficient as HPLC but, differently from HPLC, it enables quantitative analysis of systems at the single particle level.

14.
Cancers (Basel) ; 12(1)2019 Dec 31.
Article in English | MEDLINE | ID: mdl-31906080

ABSTRACT

Aminobisphosphonates, such as zoledronic acid (ZA), have shown potential in the treatment of different malignancies, including colorectal carcinoma (CRC). Yet, their clinical exploitation is limited by their high bone affinity and modest bioavailability. Here, ZA is encapsulated into the aqueous core of spherical polymeric nanoparticles (SPNs), whose size and architecture resemble that of biological vesicles. On Vδ2 T cells, derived from the peripheral blood of healthy donors and CRC patients, ZA-SPNs induce proliferation and trigger activation up to three orders of magnitude more efficiently than soluble ZA. These activated Vδ2 T cells kill CRC cells and tumor spheroids, and are able to migrate toward CRC cells in a microfluidic system. Notably, ZA-SPNs can also stimulate the proliferation of Vδ2 T cells from the tumor-infiltrating lymphocytes of CRC patients and boost their cytotoxic activity against patients' autologous tumor organoids. These data represent a first step toward the use of nanoformulated ZA for immunotherapy in CRC patients.

15.
ACS Appl Mater Interfaces ; 10(11): 9280-9289, 2018 Mar 21.
Article in English | MEDLINE | ID: mdl-29481038

ABSTRACT

A variety of microparticles have been proposed for the sustained and localized delivery of drugs with the objective of increasing therapeutic indexes by circumventing filtering organs and biological barriers. Yet, the geometrical, mechanical, and therapeutic properties of such microparticles cannot be simultaneously and independently tailored during the fabrication process to optimize their performance. In this work, a top-down approach is employed to realize micron-sized polymeric particles, called microplates (µPLs), for the sustained release of therapeutic agents. µPLs are square hydrogel particles, with an edge length of 20 µm and a height of 5 µm, made out of poly(lactic- co-glycolic acid) (PLGA). During the synthesis process, the µPL Young's modulus can be varied from 0.6 to 5 MPa by changing the PLGA amounts from 1 to 7.5 mg, without affecting the µPL geometry while matching the properties of the surrounding tissue. Within the porous µPL matrix, different classes of therapeutic payloads can be incorporated including molecular agents, such as anti-inflammatory dexamethasone (DEX), and nanoparticles containing imaging and therapeutic molecules themselves, thus originating a truly hierarchical platform. As a proof of principle, µPLs are loaded with free DEX and 200 nm spherical polymeric nanoparticles, carrying DEX molecules (DEX-SPNs). Electron and fluorescent confocal microscopy analyses document the uniform distribution and stability of molecular and nanoagents within the µPL matrix. This multiscale, hierarchical microparticle releases DEX for at least 10 days. The inclusion of DEX-SPNs serves to minimize the initial burst release and modulate the diffusion of DEX molecules out of the µPL matrix. The biopharmacological and therapeutic properties together with the fine tuning of geometry and mechanical stiffness make µPLs a unique polymeric depot for the potential treatment of cancer, cardiovascular, and chronic, inflammatory diseases.


Subject(s)
Drug Design , Dexamethasone , Lactic Acid , Nanoparticles , Polyglycolic Acid , Porosity
16.
Front Immunol ; 8: 1411, 2017.
Article in English | MEDLINE | ID: mdl-29163489

ABSTRACT

Inflammation is a common hallmark in several diseases, including atherosclerosis, cancer, obesity, and neurodegeneration. In Alzheimer's disease (AD), growing evidence directly correlates neuronal damage with inflammation of myeloid brain cells, such as microglia. Here, polymeric nanoparticles were engineered and characterized for the delivery of anti-inflammatory molecules to macrophages stimulated via direct incubation with amyloid-ß fibers. 200 nm spherical polymeric nanoconstructs (SPNs) and 1,000 nm discoidal polymeric nanoconstructs (DPNs) were synthesized using poly(lactic-co-glycolic acid) (PLGA), polyethylene glycol (PEG), and lipid chains as building blocks. First, the internalization propensity in macrophages of both nanoparticles was assessed via cytofluorimetric and confocal microscopy analyses, demonstrating that SPNs are by far more rapidly taken up as compared to DPNs (99.6 ± 0.11 vs 14.4 ± 0.06%, within 24 h). Then, Curcumin-loaded SPNs (Curc-SPNs) were realized by encapsulating Curcumin, a natural anti-inflammatory molecule, within the PLGA core of SPNs. Finally, Curc-SPNs were shown to diminish up to 6.5-fold the production of pro-inflammatory cytokines-IL-1ß; IL-6, and TNF-α-in macrophages stimulated via amyloid-ß fibers. Although more sophisticated in vitro models and systematic analyses on the blood-brain barrier permeability are critically needed, these findings hold potential in the development of nanoparticles for modulating inflammation in AD.

17.
Nanomedicine ; 12(7): 2139-2147, 2016 10.
Article in English | MEDLINE | ID: mdl-27247189

ABSTRACT

Nanoparticles can simultaneously deliver multiple agents to cancerous lesions enabling de facto combination therapies. Here, spherical polymeric nanoconstructs (SPNs) are loaded with anti-cancer - docetaxel (DTXL) - and anti-inflammatory - diclofenac (DICL) - molecules. In vitro, combination SPNs kill U87-MG cells twice as efficiently as DTXL SPNs, achieving a IC50 of 90.5nM at 72h. Isobologram analysis confirms a significant synergy (CI=0.56) between DTXL and DICL. In mice bearing non-orthotopic glioblastoma multiforme tumors, combination SPNs demonstrate higher inhibition in disease progression. At 70days post treatment, the survival rate of mice treated with combination SPNs is of about 70%, against a 40% for DTXL SPNs and 0% for free DTXL. Combination SPNs dramatically inhibit COX-2 expression, modulating the local inflammatory status, and increase Caspase-3 expression, which is directly related to cell death. These results suggest that the combination of anti-cancer and anti-inflammatory molecules constitutes a potent strategy for inhibiting tumor growth.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Glioblastoma/drug therapy , Nanoparticles , Animals , Caspase 3 , Cell Death , Mice , Polymers
18.
ACS Nano ; 9(12): 11628-41, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26488177

ABSTRACT

Most nanoparticles for biomedical applications originate from the self-assembling of individual constituents through molecular interactions and possess limited geometry control and stability. Here, 1000 × 400 nm discoidal polymeric nanoconstructs (DPNs) are demonstrated by mixing hydrophobic and hydrophilic polymers with lipid chains and curing the resulting paste directly within silicon templates. By changing the paste composition, soft- and rigid-DPNs (s- and r-DPNs) are synthesized exhibiting the same geometry, a moderately negative surface electrostatic charge (-14 mV), and different mechanical stiffness (∼1.3 and 15 kPa, respectively). Upon injection in mice bearing nonorthotopic brain or skin cancers, s-DPNs exhibit ∼24 h circulation half-life and accumulate up to ∼20% of the injected dose per gram tumor, detecting malignant masses as small as ∼0.1% the animal weight via PET imaging. This unprecedented behavior is ascribed to the unique combination of geometry, surface properties, and mechanical stiffness which minimizes s-DPN sequestration by the mononuclear phagocyte system. Our results could boost the interest in using less conventional delivery systems for cancer theranosis.


Subject(s)
Macrophages/metabolism , Molecular Imaging/methods , Nanoparticles/chemistry , Neovascularization, Pathologic/pathology , Polymers/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Female , Ferric Compounds , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Nude , Mice, Transgenic , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/metabolism
19.
Nanomedicine ; 10(5): 991-1002, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24566270

ABSTRACT

Vascular adhesion and endothelial transmigration are critical steps in the establishment of distant metastasis by circulating tumor cells (CTCs). Also, vascular inflammation plays a pivotal role in steering CTCs out of the blood stream. Here, long circulating lipid-polymer nanoparticles encapsulating curcumin (NANOCurc) are proposed for modulating the vascular deposition of CTCs. Upon treatment with NANOCurc, the adhesion propensity of highly metastatic breast cancer cells (MDA-MB-231) onto TNF-α stimulated endothelial cells (HUVECs) reduces by ~70%, in a capillary flow. Remarkably, the CTCs vascular deposition already reduces up to ~50% by treating solely the inflamed HUVECs. The CTCs arrest is mediated by the interaction between ICAM-1 on HUVECs and MUC-1 on cancer cells, and moderate doses of curcumin down-regulate the expression of both molecules. This suggests that NANOCurc could prevent metastasis and limit the progression of the disease by modulating vascular inflammation and impairing the CTCs arrest. FROM THE CLINICAL EDITOR: In this novel study, lipid nanoparticles encapsulating curcumin were able to prevent metastasis formation and limited the progression of the disease by modulating vascular inflammation and impairing the circulating tumor cells' arrest as a result of down-regulation of ICAM1 and MUC1 in a highly metastatic breast cancer cell line model.


Subject(s)
Curcumin/chemistry , Curcumin/pharmacology , Lipids/chemistry , Nanoparticles/chemistry , Polymers/chemistry , Cell Adhesion/drug effects , Cell Line, Tumor , Female , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Adhesion Molecule-1/metabolism , Tumor Necrosis Factor-alpha/pharmacology
20.
Adv Funct Mater ; 24(29): 4584-4594, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-26167143

ABSTRACT

Iron oxide nanoparticles are formidable multifunctional systems capable of contrast enhancement in magnetic resonance imaging; guidance under remote fields; heat generation; and biodegradation. Yet, this potential is underutilized in that each function manifests at different nanoparticle sizes. Here, sub-micrometer discoidal magnetic nanoconstructs are realized by confining 5 nm ultra-small super-paramagnetic iron oxide nanoparticles (USPIOs) within two different mesoporous structures, made out of silicon and polymers. These nanoconstructs exhibit transversal relaxivities up to ~10 times (r2 ~ 835 (mM·s)-1) higher than conventional USPIOs and, under external magnetic fields, collectively cooperate to amplify tumor accumulation. The boost in r2 relaxivity arises from the formation of mesoscopic USPIO clusters within the porous matrix, inducing a local reduction in water molecule mobility as demonstrated via molecular dynamics simulations. The cooperative accumulation under static magnetic field derives from the large amount of iron that can be loaded per nanoconstuct (up to ~ 65 fg) and the consequent generation of significant inter-particle magnetic dipole interactions. In tumor bearing mice, the silicon-based nanoconstructs provide MRI contrast enhancement at much smaller doses of iron (~ 0.5 mg of Fe/kg animal) as compared to current practice.

SELECTION OF CITATIONS
SEARCH DETAIL
...