Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Enrica De Rosa is active.

Publication


Featured researches published by Enrica De Rosa.


Lab on a Chip | 2010

A robust nanofluidic membrane with tunable zero-order release for implantable dose specific drug delivery

Daniel Fine; Alessandro Grattoni; Sharath Hosali; Arturas Ziemys; Enrica De Rosa; Jaskaran Gill; Ryan Medema; Lee Hudson; Milos Kojic; Miljan Milosevic; Louis Brousseau; Randy Goodall; Mauro Ferrari; Xuewu Liu

This manuscript demonstrates a mechanically robust implantable nanofluidic membrane capable of tunable long-term zero-order release of therapeutic agents in ranges relevant for clinical applications. The membrane, with nanochannels as small as 5 nm, allows for the independent control of both dosage and mechanical strength through the integration of high-density short nanochannels parallel to the membrane surface with perpendicular micro- and macrochannels for interfacing with the ambient solutions. These nanofluidic membranes are created using precision silicon fabrication techniques on silicon-on-insulator substrates enabling exquisite control over the monodispersed nanochannel dimensions and surface roughness. Zero-order release of analytes is achieved by exploiting molecule to surface interactions which dominate diffusive transport when fluids are confined to the nanoscale. In this study we investigate the nanofluidic membrane performance using custom diffusion and gas testing apparatuses to quantify molecular release rate and process uniformity as well as mechanical strength using a gas based burst test. The kinetics of the constrained zero-order release is probed with molecules presenting a range of sizes, charge states, and structural conformations. Finally, an optimal ratio of the molecular hydrodynamic diameter to the nanochannel dimension is determined to assure zero-order release for each tested molecule.


ACS Nano | 2015

Soft Discoidal Polymeric Nanoconstructs Resist Macrophage Uptake and Enhance Vascular Targeting in Tumors

Jaehong Key; Anna Lisa Palange; Francesco Gentile; Santosh Aryal; Cinzia Stigliano; Daniele Di Mascolo; Enrica De Rosa; Minjung Cho; Yeonju Lee; Jaykrishna Singh; Paolo Decuzzi

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.


Journal of Controlled Release | 2013

Leveraging nanochannels for universal, zero-order drug delivery in vivo

Silvia Ferrati; Daniel Fine; Junping You; Enrica De Rosa; Lee Hudson; Erika Zabre; Sharath Hosali; Li Zhang; Catherine Hickman; Shyam S. Bansal; Andrea M. Cordero-Reyes; Thomas Geninatti; Juliana Sih; Randy Goodall; Ganesh S. Palapattu; Malgorzata Kloc; Rafik M. Ghobrial; Mauro Ferrari; Alessandro Grattoni

Drug delivery is essential to achieve effective therapy. Herein we report on the only implantable nanochannel membrane with geometrically defined channels as small as 2.5 nm that achieves constant drug delivery in vivo. Nanochannels passively control the release of molecules by physico-electrostatic confinement, thereby leading to constant drug diffusion. We utilize a novel design algorithm to select the optimal nanochannel size for each therapeutic agent. Using nanochannels as small as 3.6 and 20 nm, we achieve sustained and constant plasma levels of leuprolide, interferon α-2b, letrozole, Y-27632, octreotide, and human growth hormone, all delivered at clinically-relevant doses. The device was demonstrated in dogs, rats, and mice and was capable of sustaining target doses for up to 70 days. To provide evidence of therapeutic efficacy, we successfully combined nanochannel delivery with a RhoA pathway inhibitor to prevent chronic rejection of cardiac allografts in a rat model. Our results provide evidence that the nanochannel platform has the potential to dramatically improve long-term therapies for chronic conditions.


ACS Applied Materials & Interfaces | 2015

PLGA-Mesoporous Silicon Microspheres for the in Vivo Controlled Temporospatial Delivery of Proteins.

Silvia Minardi; Laura Pandolfi; Francesca Taraballi; Enrica De Rosa; Iman K. Yazdi; Xeuwu Liu; Mauro Ferrari; Ennio Tasciotti

In regenerative medicine, the temporospatially controlled delivery of growth factors (GFs) is crucial to trigger the desired healing mechanisms in the target tissues. The uncontrolled release of GFs has been demonstrated to cause severe side effects in the surrounding tissues. The aim of this study was to optimize a translational approach for the fine temporal and spatial control over the release of proteins, in vivo. Hence, we proposed a newly developed multiscale composite microsphere based on a core consisting of the nanostructured silicon multistage vector (MSV) and a poly(dl-lactide-co-glycolide) acid (PLGA) outer shell. Both of the two components of the resulting composite microspheres (PLGA-MSV) can be independently tailored to achieve multiple release kinetics contributing to the control of the release profile of a reporter protein in vitro. The influence of MSV shape (hemispherical or discoidal) and size (1, 3, or 7 μm) on PLGA-MSVs morphology and size distribution was investigated. Second, the copolymer ratio of the PLGA used to fabricate the outer shell of PLGA-MSV was varied. The composites were fully characterized by optical microscopy, scanning electron microscopy, ζ potential, Fourier transform infrared spectroscopy, and thermogravimetric analysis-differential scanning calorimetry, and their release kinetics over 30 days. PLGA-MSVs biocompatibility was assessed in vitro with J774 macrophages. Finally, the formulation of PLGA-MSV was selected, which concurrently provided the most consistent microsphere size and allowed for a zero-order release kinetic. The selected PLGA-MSVs were injected in a subcutaneous model in mice, and the in vivo release of the reporter protein was followed over 2 weeks by intravital microscopy, to assess if the zero-order release was preserved. PLGA-MSV was able to retain the payload over 2 weeks, avoiding the initial burst release typical of most drug delivery systems. Finally, histological evaluation assessed the biocompatibility of the platform in vivo.


Pharmaceutical Research | 2011

Agarose surface coating influences intracellular accumulation and enhances payload stability of a nano-delivery system.

Enrica De Rosa; Ciro Chiappini; Dongmei Fan; Xuewu Liu; Mauro Ferrari; Ennio Tasciotti

ABSTRACTPurposeProtein therapeutics often require repeated administrations of drug over a long period of time. Protein instability is a major obstacle to the development of systems for their controlled and sustained release. We describe a surface modification of nanoporous silicon particles (NSP) with an agarose hydrogel matrix that enhances their ability to load and release proteins, influencing intracellular delivery and preserving molecular stability.MethodsWe developed and characterized an agarose surface modification of NSP. Stability of the released protein after enzymatic treatment of loaded particles was evaluated with SDS-page and HPLC analysis. FITC-conjugated BSA was chosen as probe protein and intracellular delivery evaluated by fluorescence microscopy.ResultsWe showed that agarose coating does not affect NSP protein release rate, while fewer digestion products were found in the released solution after all the enzymatic treatments. Confocal images show that the hydrogel coating improves intracellular delivery, specifically within the nucleus, without affecting the internalization process.ConclusionsThis modification of porous silicon adds to its tunability, biocompatibility, and biodegradability the ability to preserve protein integrity during delivery without affecting release rates and internalization dynamics. Moreover, it may allow the silicon particles to function as protein carriers that enable control of cell function.


Journal of Functional Biomaterials | 2011

Multi-Composite Bioactive Osteogenic Sponges Featuring Mesenchymal Stem Cells, Platelet-Rich Plasma, Nanoporous Silicon Enclosures, and Peptide Amphiphiles for Rapid Bone Regeneration

Matthew B. Murphy; Daniel Blashki; Rachel M. Buchanan; Dongmei Fan; Enrica De Rosa; Ramille N. Shah; Samuel I. Stupp; Bradley K. Weiner; Paul J. Simmons; Mauro Ferrari; Ennio Tasciotti

A novel bioactive sponge was created with a composite of type I collagen sponges or porous poly(ε-caprolactone) (PCL) scaffolds, platelet-rich plasma (PRP), BMP2-loaded nanoporous silicon enclosure (NSE) microparticles, mineralizing peptide amphiphiles (PA), and mesenchymal stem cells (MSC). Primary MSC from cortical bone (CB) tissue proved to form more and larger colony units, as well as produce more mineral matrix under osteogenic differentiation, than MSC from bone marrow (BM). Coating pre-treatments were optimized for maximum cell adhesion and mineralization, while a PRP-based gel carrier was created to efficiently deliver and retain MSC and microparticles within a porous scaffold while simultaneously promoting cell recruitment, proliferation, and angiogenesis. Components and composite sponges were evaluated for osteogenic differentiation in vitro. Osteogenic sponges were loaded with MSC, PRP, PA, and NSE and implanted subcutaneously in rats to evaluate the formation of bone tissue and angiogenesis in vivo. It was found that the combination of a collagen sponge with CB MSC, PRP, PA, and the BMP2-releasing NSE formed the most bone and was most vascularized by four weeks compared to analogous composites featuring BM MSC or PCL or lacking PRP, PA, and NSE. This study indicates that CB MSC should be considered as an alternative to marrow as a source of stem cells, while the PRP-PA cell and microparticle delivery system may be utilized for diverse tissue engineering applications.


Journal of Micromechanics and Microengineering | 2009

Analysis of a nanochanneled membrane structure through convective gas flow

Alessandro Grattoni; Enrica De Rosa; Silvia Ferrati; Zongxing Wang; Anna Gianesini; Xuewu Liu; Fazle Hussain; Randy Goodall; Mauro Ferrari

Micro- and nano-fluidic devices are under development for a variety of applications including bio-molecular separation, drug delivery, biosensing and cell transplantation. Regulatory approval for the commercialization of these products requires the ability to fabricate a large number of these devices with high reproducibility and precision. Though traditional microscopy and particle rejection characterization techniques provide extremely useful measurements of nano-features, they are expensive and inadequate for quality control purposes. In this study, an agile and non-destructive selection method is presented which combines a predictive theoretical model with experimental analysis of convective nitrogen flow to detect structural defects in complex drug delivery membranes (nDS) combining both micro- and nanochanneled features. The mathematical model developed bridges the fluid dynamics between the micro- and nano-scales. An experimental analysis of gas flow was performed on a total of 250 membranes representing five different channel size configurations. The accuracy and reliability of this test in detecting major and minor defects of various kinds were verified by comparing the experimental results with the theoretical prediction.


ACS Applied Materials & Interfaces | 2017

Enhancing Vascularization through the Controlled Release of Platelet-Derived Growth Factor-BB

Silvia Minardi; Laura Pandolfi; Francesca Taraballi; Xin Wang; Enrica De Rosa; Zachary D. Mills; Xuewu Liu; Mauro Ferrari; Ennio Tasciotti

Using delivery systems to control the in vivo release of growth factors (GFs) for tissue engineering applications is extremely desirable as the clinical use of GFs is limited by their fast in vivo turnover. Hence, the development of effective platforms that are able to finely control the release of GFs in vivo remains a challenge. Herein, we investigated the ability of multiscale microspheres, composed by a nanostructured silicon multistage vector (MSV) core and a poly(dl-lactide-co-glycolide) acid (PLGA) forming outer shell (PLGA-MSV), to release functional platelet-derived growth factor-BB (PDGF-BB) to induce in vivo localized neovascularization. The in vitro release of PDGF-BB was assessed by enzyme-linked immunosorbent assay (ELISA) over 2 weeks and showed a sustained, zero-order release kinetics. The ability to promote in vivo localized neovascularization was investigated in a subcutaneous injection model in BALB/c mice and followed by intravital microscopy up to 2 weeks. Fully functional newly formed vessels were found within the area where PLGA-MSVs were localized and covered 3.0 ± 0.9 and 19 ± 5.1% at 7 and 14 days, respectively, showing a 6-fold increase in 1 week. The distribution of CD31+ and α-SMA+ cells was detected by immunofluorescence on harvested tissues. CD31 was significantly more expressed (4-fold increase) compared to the untreated control. Finally, the level of up-regulation of angiogenesis-associated genes (Vegfa, Vwf, and Col3a1) was assessed by q-PCR, resulting in a significantly higher expression where PLGA-MSVs were localized (Vegfa: 2.32 ± 0.50 at 7 days and 4.37 ± 0.75 at 14 days; Vwf: 4.13 ± 0.82 and 7.74 ± 0.91; Col3a1: 5.43 ± 0.37 and 6.66 ± 0.89). Altogether, our data supported the conclusion that the localized delivery of PDGF-BB from PLGA-MSVs induced the localized de novo formation of fully functional vessels in vivo. With this study, we demonstrated that PLGA-MSV holds promise for accomplishing the controlled localized in vivo release of GFs for the design of innovative tissue engineering strategies.


Theranostics | 2018

Biomimetic nanoparticles with enhanced affinity towards activated endothelium as versatile tools for theranostic drug delivery

Jonathan O. Martinez; Roberto Molinaro; Kelly A. Hartman; Christian Boada; Roman A. Sukhovershin; Enrica De Rosa; Dickson K. Kirui; Shanrong Zhang; Michael Evangelopoulos; Angela M. Carter; James A. Bibb; John P. Cooke; Ennio Tasciotti

Activation of the vascular endothelium is characterized by increased expression of vascular adhesion molecules and chemokines. This activation occurs early in the progression of several diseases and triggers the recruitment of leukocytes. Inspired by the tropism of leukocytes, we investigated leukocyte-based biomimetic nanoparticles (i.e., leukosomes) as a novel theranostic platform for inflammatory diseases. Methods: Leukosomes were assembled by combining phospholipids and membrane proteins from leukocytes. For imaging applications, phospholipids modified with rhodamine and gadolinium were used. Leukosomes incubated with antibodies blocking lymphocyte function-associated antigen 1 (LFA-1) and CD45 were administered to explore their roles in targeting inflammation. In addition, relaxometric assessment of NPs was evaluated. Results: Liposomes and leukosomes were both spherical in shape with sizes ranging from 140-170 nm. Both NPs successfully integrated 8 and 13 µg of rhodamine and gadolinium, respectively, and demonstrated less than 4% variation in physicochemical features. Leukosomes demonstrated a 16-fold increase in breast tumor accumulation relative to liposomes. Furthermore, quantification of leukosomes in tumor vessels demonstrated a 4.5-fold increase in vessel lumens and a 14-fold increase in vessel walls. Investigating the targeting mechanism of action revealed that blockage of LFA-1 on leukosomes resulted in a 95% decrease in tumor accumulation. Whereas blockage of CD45 yielded a 60% decrease in targeting and significant increases in liver and spleen accumulation. In addition, when administered in mice with atherosclerotic plaques, leukosomes exhibited a 4-fold increase in the targeting of inflammatory vascular lesions. Lastly, relaxometric assessment of NPs demonstrated that the incorporation of membrane proteins into leukosomes did not impact the r1 and r2 relaxivities of the NPs, demonstrating 6 and 30 mM-1s-1, respectively. Conclusion: Our study demonstrates the ability of leukosomes to target activated vasculature and exhibit superior accumulation in tumors and vascular lesions. The versatility of the phospholipid backbone within leukosomes permits the incorporation of various contrast agents. Furthermore, leukosomes can potentially be loaded with therapeutics possessing diverse physical properties and thus warrant further investigation toward the development of powerful theranostic agents.


ACS Nano | 2015

Correction to Biodegradable Nanoneedles for Localized Delivery of Nanoparticles in Vivo: Exploring the Biointerface.

Ciro Chiappini; Jonathan O. Martinez; Enrica De Rosa; Carina S. Almeida; Ennio Tasciotti; Molly M. Stevens

Nanoneedles display potential in mediating the delivery of drugs and biologicals, as well as intracellular sensing and single cell stimulation through direct access to the cell cytoplasm. Nanoneedles enable cytosolic delivery, negotiating the cell membrane and the endolysosomal system, thus overcoming these major obstacles to the efficacy of nanotherapeutics. The low toxicity and minimal invasiveness of nanoneedles has a potential for the sustained nonimmunogenic delivery of payloads in vivo, provided that the development of biocompatible nanoneedles with a simple deployment strategy is achieved. Here we present a mesoporous silicon nanoneedle array that achieves a tight interface with the cell, rapidly negotiating local biological barriers to grant temporary access to the cytosol with minimal impact on cell viability. The tightness of this interfacing enables both delivery of cell-impermeant quantum dots in vivo and live intracellular sensing of pH. Dissecting the biointerface over time elucidated the dynamics of cell association and nanoneedle biodegradation, showing rapid interfacing leading to cytosolic payload delivery within less than 30 minutes in vitro. The rapid and simple application of nanoneedles in vivo to the surface of tissues with different architectures invariably resulted in the localized delivery of quantum dots to the superficial cells and their prolonged retention. This investigation provides an understanding of the dynamics of nanoneedles’ biointerface and delivery outlining a strategy for highly local intracellular delivery of nanoparticles and cell-impermeant payloads within live tissues. To whom correspondence should be addressed: Prof. Molly M. Stevens; Professor of Biomedical Materials and Regenerative Medicine, Department of Materials and Department of Bioengineering, Research Director for Biomedical Material Sciences, Institute of Biomedical Engineering, Imperial College London, Prince Consort Rd. SW7 2AZ London, UK [email protected] +44 (0)20 7594 6804 Dr. Ennio Tasciotti; Associate Member, Co-Chair, Department of Nanomedicine, Scientist, Tissue Engineering & Regenerative Medicine Program, Scientific Director, The Surgical Advanced Technology Laboratory, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, United States . Supporting Information Available: Supplementary figures, movies and extended material section are available free of charge via the Internet at http://pubs.acs.org. Europe PMC Funders Group Author Manuscript ACS Nano. Author manuscript; available in PMC 2016 February 01. Published in final edited form as: ACS Nano. 2015 May 26; 9(5): 5500–5509. doi:10.1021/acsnano.5b01490. E uope PM C Fuders A uhor M ancripts E uope PM C Fuders A uhor M ancripts

Collaboration


Dive into the Enrica De Rosa's collaboration.

Top Co-Authors

Avatar

Ennio Tasciotti

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Mauro Ferrari

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Xuewu Liu

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dongmei Fan

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Randy Goodall

University of Texas System

View shared research outputs
Top Co-Authors

Avatar

Roberto Molinaro

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Bradley K. Weiner

Houston Methodist Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge