1
|
Spiteri C, Caprettini V, Wang Y, Dominguez‐Gil S, Kaasalainen M, Wang C, Martella DA, McLennan S, Vashisth P, Gary‐Bobo M, Nguyen C, Bergholt M, Durand J, Cunin F, Chiappini C. Spatially-Resolved Organoid Transfection by Porous Silicon-Mediated Optoporation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407650. [PMID: 39420545 PMCID: PMC11619231 DOI: 10.1002/adma.202407650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/27/2024] [Indexed: 10/19/2024]
Abstract
Engineering the spatial organisation of organotypic cultures is pivotal for refining tissue models that are useful for gaining deeper insights into complex, non-cell autonomous processes. These advanced models are key to improving the understanding of fundamental biological mechanisms and therapeutic strategies. Controlling gene regulation through spatially-resolved delivery of nucleic acids provides an attractive approach to produce such tissue models. An emerging strategy for spatially-resolved transfection uses photosensitizing nanoparticles coupled with laser pulses to optoporate cells in culture and locally mediate gene delivery. However, localized optoporation in 3D systems remains challenging. Here we propose a solution to this longstanding hurdle, demonstrating that porous silicon nanoparticles are a safe and bioresorbable photosensitising nanomaterial capable of spatially-resolved transfection of mRNA in MCF-7 organoids by near-infrared two-photon optoporation. Functionalization with an azobenzene-lysine photo-switchable moiety enhances the transfection efficiency of the nanoparticles up to 84% in a 2D cell system. Moreover, the nanoparticles enable spatially selective mRNA transfection to MCF-7 spheroids, demonstrating targeted gene delivery in complex 3D cellular environments. The approach for spatially-resolved 3D optoporation offers a way forward for the design of tailored spheroids and organoids by spatially selective nucleic acids delivery.
Collapse
Affiliation(s)
- Chantelle Spiteri
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Valeria Caprettini
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Yikai Wang
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | | | - Martti Kaasalainen
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Cong Wang
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Davide Alessandro Martella
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Samuel McLennan
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Priya Vashisth
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
| | | | | | - Mads Bergholt
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
| | | | | | - Ciro Chiappini
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| |
Collapse
|
2
|
Bruce G, Bagherpour S, Duch M, Plaza JA, Stolnik S, Pérez-García L. Cuboids Prevail When Unraveling the Influence of Microchip Geometry on Macrophage Interactions and Metabolic Responses. ACS Biomater Sci Eng 2024; 10:5689-5700. [PMID: 39167686 PMCID: PMC11388147 DOI: 10.1021/acsbiomaterials.4c00849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Drug delivery advances rely on using nano- and microsized carriers to transfer therapeutic molecules, although challenges persist in increasing the availability of new and even approved pharmaceutical products. Particle shape, a critical determinant in how these carriers distribute within the body after administration, raises opportunities of using, for instance, micrometer-sized nonspherical particles for vascular targeting and thereby creating new prospects for precise drug delivery to specific targeted areas. The versatility of polycrystalline silicon microfabrication allows for significant variation in the size and shape of microchips, and so, in the current work, photolithography was employed to create differently shaped polysilicon microchips, including cuboids, cubes, bars, and cylinders, to explore the influence of particle shape on cellular interactions. These microchips with different shapes and lateral dimensions, accounting for surface areas in the range of ca. 15 to 120 μm2 and corresponding total volumes of 0.4 to 27 μm3, serve as ideal models for investigating their interactions with macrophages with diameters of ca. 20 μm. Side-scattering imaging flow cytometry was employed for studying the interaction of label-free prepared microchips with RAW 264.7 macrophages. Using a dose of 3 microchips per cell, results show that cuboids exhibit the highest cellular association (ca. 25%) and uptake (ca. 20%), suggesting their potential as efficient carriers for targeted drug delivery to macrophages. Conversely, similarly sized cylinders and bar-shaped microchips exhibit lower uptakes of about 8% and about 6%, respectively, indicating potential benefits in evading macrophage recognition. On average, 1-1.5 microchips were internalized, and ca. 1 microchip was surface-bound per cell, with cuboids showing the higher values overall. Macrophages respond to microchips by increasing their metabolic activity and releasing low levels of intracellular enzymes, indicating reduced toxicity. Interestingly, increasing the particle dose enhances macrophage metabolic activity without significantly affecting enzyme release.
Collapse
Affiliation(s)
- Gordon Bruce
- Division of Advanced Materials and Healthcare Technologies, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Saman Bagherpour
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Av. Joan XXIII 27-31, 08028 Barcelona, Spain
- Institut de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Marta Duch
- Instituto de Microelectrónica de Barcelona IMB-CNM (CSIC), Campus UAB, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - José Antonio Plaza
- Instituto de Microelectrónica de Barcelona IMB-CNM (CSIC), Campus UAB, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - Snow Stolnik
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Lluïsa Pérez-García
- Division of Advanced Materials and Healthcare Technologies, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Av. Joan XXIII 27-31, 08028 Barcelona, Spain
- Institut de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona (UB), 08028 Barcelona, Spain
| |
Collapse
|
3
|
Wang C, Gu C, Popp C, Vashisth P, Mustfa SA, Martella DA, Spiteri C, McLennan S, Sun N, Riddle M, Eide CR, Parsons M, Tolar J, McGrath JA, Chiappini C. Integrating Porous Silicon Nanoneedles within Medical Devices for Nucleic Acid Nanoinjection. ACS NANO 2024; 18:14938-14953. [PMID: 38726598 PMCID: PMC11171749 DOI: 10.1021/acsnano.4c00206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 06/12/2024]
Abstract
Porous silicon nanoneedles can interface with cells and tissues with minimal perturbation for high-throughput intracellular delivery and biosensing. Typically, nanoneedle devices are rigid, flat, and opaque, which limits their use for topical applications in the clinic. We have developed a robust, rapid, and precise substrate transfer approach to incorporate nanoneedles within diverse substrates of arbitrary composition, flexibility, curvature, transparency, and biodegradability. With this approach, we integrated nanoneedles on medically relevant elastomers, hydrogels, plastics, medical bandages, catheter tubes, and contact lenses. The integration retains the mechanical properties and transfection efficiency of the nanoneedles. Transparent devices enable the live monitoring of cell-nanoneedle interactions. Flexible devices interface with tissues for efficient, uniform, and sustained topical delivery of nucleic acids ex vivo and in vivo. The versatility of this approach highlights the opportunity to integrate nanoneedles within existing medical devices to develop advanced platforms for topical delivery and biosensing.
Collapse
Affiliation(s)
- Cong Wang
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
- London
Centre for Nanotechnology, King’s
College London, WC2R 2LS London, U.K.
| | - Chenlei Gu
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
- London
Centre for Nanotechnology, King’s
College London, WC2R 2LS London, U.K.
| | - Courtney Popp
- Department
of Pediatrics, Medical School, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Priya Vashisth
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
| | - Salman Ahmad Mustfa
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
| | - Davide Alessandro Martella
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
- London
Centre for Nanotechnology, King’s
College London, WC2R 2LS London, U.K.
| | - Chantelle Spiteri
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
| | - Samuel McLennan
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
| | - Ningjia Sun
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
| | - Megan Riddle
- Department
of Pediatrics, Medical School, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Cindy R. Eide
- Department
of Pediatrics, Medical School, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Maddy Parsons
- Randall
Centre for Cell and Molecular Biophysics, King’s College London, SE1 1UL London, U.K.
| | - Jakub Tolar
- Department
of Pediatrics, Medical School, University
of Minnesota, Minneapolis, Minnesota 55455, United States
- Stem
Cell Institute, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - John A. McGrath
- St
John’s
Institute of Dermatology, King’s
College London, SE1 7EP London, U.K.
| | - Ciro Chiappini
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
- London
Centre for Nanotechnology, King’s
College London, WC2R 2LS London, U.K.
| |
Collapse
|
4
|
Chen L, Zhang S, Duan Y, Song X, Chang M, Feng W, Chen Y. Silicon-containing nanomedicine and biomaterials: materials chemistry, multi-dimensional design, and biomedical application. Chem Soc Rev 2024; 53:1167-1315. [PMID: 38168612 DOI: 10.1039/d1cs01022k] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The invention of silica-based bioactive glass in the late 1960s has sparked significant interest in exploring a wide range of silicon-containing biomaterials from the macroscale to the nanoscale. Over the past few decades, these biomaterials have been extensively explored for their potential in diverse biomedical applications, considering their remarkable bioactivity, excellent biocompatibility, facile surface functionalization, controllable synthesis, etc. However, to expedite the clinical translation and the unexpected utilization of silicon-composed nanomedicine and biomaterials, it is highly desirable to achieve a thorough comprehension of their characteristics and biological effects from an overall perspective. In this review, we provide a comprehensive discussion on the state-of-the-art progress of silicon-composed biomaterials, including their classification, characteristics, fabrication methods, and versatile biomedical applications. Additionally, we highlight the multi-dimensional design of both pure and hybrid silicon-composed nanomedicine and biomaterials and their intrinsic biological effects and interactions with biological systems. Their extensive biomedical applications span from drug delivery and bioimaging to therapeutic interventions and regenerative medicine, showcasing the significance of their rational design and fabrication to meet specific requirements and optimize their theranostic performance. Additionally, we offer insights into the future prospects and potential challenges regarding silicon-composed nanomedicine and biomaterials. By shedding light on these exciting research advances, we aspire to foster further progress in the biomedical field and drive the development of innovative silicon-composed nanomedicine and biomaterials with transformative applications in biomedicine.
Collapse
Affiliation(s)
- Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
5
|
Guo M, Xia C, Wu Y, Zhou N, Chen Z, Li W. Research Progress on Cell Membrane-Coated Biomimetic Delivery Systems. Front Bioeng Biotechnol 2021; 9:772522. [PMID: 34869288 PMCID: PMC8636778 DOI: 10.3389/fbioe.2021.772522] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/29/2021] [Indexed: 01/12/2023] Open
Abstract
Cell membrane-coated biomimetic nanoplatforms have many inherent properties, such as bio-interfacing abilities, self-identification, and signal transduction, which enable the biomimetic delivery system to escape immune clearance and opsonization. This can also maximize the drug delivery efficiency of synthetic nanoparticles (NPs) and functional cell membranes. As a new type of delivery system, cell membrane-coated biomimetic delivery systems have broadened the prospects for biomedical applications. In this review, we summarize research progress on cell membrane biomimetic technology from three aspects, including sources of membrane, modifications, and applications, then analyze their limitations and propose future research directions.
Collapse
Affiliation(s)
- Mengyu Guo
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenjie Xia
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Nong Zhou
- The Chongqing Engineering Laboratory for Green Cultivation and Deep Processing of Three Gorges Reservoir Area's Medicinal Herbs, College of Food and Biology Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Zhipeng Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weidong Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
6
|
Chiappini C, Chen Y, Aslanoglou S, Mariano A, Mollo V, Mu H, De Rosa E, He G, Tasciotti E, Xie X, Santoro F, Zhao W, Voelcker NH, Elnathan R. Tutorial: using nanoneedles for intracellular delivery. Nat Protoc 2021; 16:4539-4563. [PMID: 34426708 DOI: 10.1038/s41596-021-00600-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 06/30/2021] [Indexed: 02/08/2023]
Abstract
Intracellular delivery of advanced therapeutics, including biologicals and supramolecular agents, is complex because of the natural biological barriers that have evolved to protect the cell. Efficient delivery of therapeutic nucleic acids, proteins, peptides and nanoparticles is crucial for clinical adoption of emerging technologies that can benefit disease treatment through gene and cell therapy. Nanoneedles are arrays of vertical high-aspect-ratio nanostructures that can precisely manipulate complex processes at the cell interface, enabling effective intracellular delivery. This emerging technology has already enabled the development of efficient and non-destructive routes for direct access to intracellular environments and delivery of cell-impermeant payloads. However, successful implementation of this technology requires knowledge of several scientific fields, making it complex to access and adopt by researchers who are not directly involved in developing nanoneedle platforms. This presents an obstacle to the widespread adoption of nanoneedle technologies for drug delivery. This tutorial aims to equip researchers with the knowledge required to develop a nanoinjection workflow. It discusses the selection of nanoneedle devices, approaches for cargo loading and strategies for interfacing to biological systems and summarises an array of bioassays that can be used to evaluate the efficacy of intracellular delivery.
Collapse
Affiliation(s)
- Ciro Chiappini
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.
- London Centre for Nanotechnology, King's College London, London, UK.
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia
| | - Stella Aslanoglou
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia
- CSIRO Manufacturing, Clayton, Victoria, Australia
| | - Anna Mariano
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
| | - Valentina Mollo
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
| | - Huanwen Mu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Enrica De Rosa
- Center for Musculoskeletal Regeneration, Orthopedics & Sports Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, China
| | - Ennio Tasciotti
- IRCCS San Raffaele Pisana Hospital, Rome, Italy
- San Raffaele University, Rome, Italy
- Sclavo Pharma, Siena, Italy
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, China.
| | - Francesca Santoro
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy.
| | - Wenting Zhao
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
- CSIRO Manufacturing, Clayton, Victoria, Australia.
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia.
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
7
|
Leonardi AA, Lo Faro MJ, Irrera A. Biosensing platforms based on silicon nanostructures: A critical review. Anal Chim Acta 2021; 1160:338393. [PMID: 33894957 DOI: 10.1016/j.aca.2021.338393] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/21/2022]
Abstract
Biosensors are revolutionizing the health-care systems worldwide, permitting to survey several diseases, even at their early stage, by using different biomolecules such as proteins, DNA, and other biomarkers. However, these sensing approaches are still scarcely diffused outside the specialized medical and research facilities. Silicon is the undiscussed leader of the whole microelectronics industry, and novel sensors based on this material may completely change the health-care scenario. In this review, we will show how novel sensing platforms based on Si nanostructures may have a disruptive impact on applications with a real commercial transfer. A critical study for the main Si-based biosensors is herein presented with a comparison of their advantages and drawbacks. The most appealing sensing devices are discussed, starting from electronic transducers, with Si nanowires field-effect transistor (FET) and porous Si, to their optical alternatives, such as effective optical thickness porous silicon, photonic crystals, luminescent Si quantum dots, and finally luminescent Si NWs. All these sensors are investigated in terms of working principle, sensitivity, and selectivity with a specific focus on the possibility of their industrial transfer, and which ones may be preferred for a medical device.
Collapse
Affiliation(s)
- Antonio Alessio Leonardi
- Dipartimento di Fisica e Astronomia "Ettore Majorana", Università di Catania, Via Santa Sofia 64, 95123, Catania, Italy; CNR-IMM UoS Catania, Istituto per La Microelettronica e Microsistemi, Via Santa Sofia 64, Italy; CNR-IPCF, Istituto per I Processi Chimico-Fisici, Viale F. Stagno D'Alcontres 37, 98158, Messina, Italy
| | - Maria José Lo Faro
- Dipartimento di Fisica e Astronomia "Ettore Majorana", Università di Catania, Via Santa Sofia 64, 95123, Catania, Italy; CNR-IMM UoS Catania, Istituto per La Microelettronica e Microsistemi, Via Santa Sofia 64, Italy
| | - Alessia Irrera
- CNR-IPCF, Istituto per I Processi Chimico-Fisici, Viale F. Stagno D'Alcontres 37, 98158, Messina, Italy.
| |
Collapse
|
8
|
Hendrickson T, Mancino C, Whitney L, Tsao C, Rahimi M, Taraballi F. Mimicking cardiac tissue complexity through physical cues: A review on cardiac tissue engineering approaches. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 33:102367. [PMID: 33549819 DOI: 10.1016/j.nano.2021.102367] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/06/2021] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
Cardiovascular diseases are the number one killer in the world.1,2 Currently, there are no clinical treatments to regenerate damaged cardiac tissue, leaving patients to develop further life-threatening cardiac complications. Cardiac tissue has multiple functional demands including vascularization, contraction, and conduction that require many synergic components to properly work. Most of these functions are a direct result of the cardiac tissue structure and composition, and, for this reason, tissue engineering strongly proposed to develop substitute engineered heart tissues (EHTs). EHTs usually have combined pluripotent stem cells and supporting scaffolds with the final aim to repair or replace the damaged native tissue. However, as simple as this idea is, indeed, it resulted, after many attempts in the field, to be very challenging. Without design complexity, EHTs remain unable to mature fully and integrate into surrounding heart tissue resulting in minimal in vivo effects.3 Lately, there has been a growing body of evidence that a complex, multifunctional approach through implementing scaffold designs, cellularization, and molecular release appears to be essential in the development of a functional cardiac EHTs.4-6 This review covers the advancements in EHTs developments focusing on how to integrate contraction, conduction, and vascularization mimics and how combinations have resulted in improved designs thus warranting further investigation to develop a clinically applicable treatment.
Collapse
Affiliation(s)
- Troy Hendrickson
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA; Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA; Texas A&M MD/PhD Program, Texas A&M Health Science Center, College Station, TX, USA
| | - Chiara Mancino
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA; Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, (MI), Italy
| | - Lauren Whitney
- Texas A&M Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Chris Tsao
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA
| | - Maham Rahimi
- Department of Cardiovascular Surgery, Houston Methodist, Houston, TX, USA
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA; Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
9
|
Palma-Chavez JA, Fuentes K, Applegate BE, Jo JA, Charoenphol P. Development and Characterization of PLGA-Based Multistage Delivery System for Enhanced Payload Delivery to Targeted Vascular Endothelium. Macromol Biosci 2021; 21:e2000377. [PMID: 33393217 DOI: 10.1002/mabi.202000377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/18/2020] [Indexed: 01/06/2023]
Abstract
Vascular-targeted drug delivery remains an attractive platform for therapeutic and diagnostic interventions in human diseases. This work focuses on the development of a poly-lactic-co-glycolic-acid (PLGA)-based multistage delivery system (MDS). MDS consists of two stages: a micron-sized PLGA outer shell and encapsulated drug-loaded PLGA nanoparticles. Nanoparticles with average diameters of 76, 119, and 193 nm are successfully encapsulated into 3-6 µm MDS. Sustained in vitro release of nanoparticles from MDS is observed for up to 7 days. Both MDS and nanoparticles arebiocompatible with human endothelial cells. Sialyl-Lewis-A (sLeA ) is successfully immobilized on the MDS and nanoparticle surfaces to enable specific targeting of inflamed endothelium. Functionalized MDS demonstrates a 2.7-fold improvement in endothelial binding compared to PLGA nanoparticles from human blood laminar flow. Overall, the presented results demonstrate successful development and characterization of MDS and suggest that MDS can serve as an effective drug carrier, which can enhance the margination of nanoparticles to the targeted vascular wall.
Collapse
Affiliation(s)
- Jorge A Palma-Chavez
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Kevin Fuentes
- Department of Mechanical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Brian E Applegate
- Prof. B. E. Applegate, Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Javier A Jo
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Phapanin Charoenphol
- Department of Mechanical Engineering, Texas A&M University, College Station, TX, 77843, USA
| |
Collapse
|
10
|
Xue Y, Bai H, Peng B, Fang B, Baell J, Li L, Huang W, Voelcker NH. Stimulus-cleavable chemistry in the field of controlled drug delivery. Chem Soc Rev 2021; 50:4872-4931. [DOI: 10.1039/d0cs01061h] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This review comprehensively summarises stimulus-cleavable linkers from various research areas and their cleavage mechanisms, thus provides an insightful guideline to extend their potential applications to controlled drug release from nanomaterials.
Collapse
Affiliation(s)
- Yufei Xue
- Frontiers Science Center for Flexible Electronics
- Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering
- Northwestern Polytechnical University
- 127 West Youyi Road
- Xi'an 710072
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics
- Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering
- Northwestern Polytechnical University
- 127 West Youyi Road
- Xi'an 710072
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics
- Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering
- Northwestern Polytechnical University
- 127 West Youyi Road
- Xi'an 710072
| | - Bin Fang
- Frontiers Science Center for Flexible Electronics
- Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering
- Northwestern Polytechnical University
- 127 West Youyi Road
- Xi'an 710072
| | - Jonathan Baell
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton
- Victoria 3168
- Australia
| | - Lin Li
- Frontiers Science Center for Flexible Electronics
- Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering
- Northwestern Polytechnical University
- 127 West Youyi Road
- Xi'an 710072
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics
- Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering
- Northwestern Polytechnical University
- 127 West Youyi Road
- Xi'an 710072
| | - Nicolas Hans Voelcker
- Frontiers Science Center for Flexible Electronics
- Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering
- Northwestern Polytechnical University
- 127 West Youyi Road
- Xi'an 710072
| |
Collapse
|
11
|
Lumen D, Wang S, Mäkilä E, Imlimthan S, Sarparanta M, Correia A, Westerveld Haug C, Hirvonen J, Santos HA, Airaksinen AJ, Filtvedt W, Salonen J. Investigation of silicon nanoparticles produced by centrifuge chemical vapor deposition for applications in therapy and diagnostics. Eur J Pharm Biopharm 2020; 158:254-265. [PMID: 33279602 DOI: 10.1016/j.ejpb.2020.11.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 10/23/2020] [Accepted: 11/27/2020] [Indexed: 11/30/2022]
Abstract
Porous silicon (PSi) is a biocompatible and biodegradable material, which can be utilized in biomedical applications. It has several favorable properties, which makes it an excellent material for building engineered nanosystems for drug delivery and diagnostic purposes. One significant hurdle for commercial applications of PSi is the lack of industrial scale production of nanosized PSi particles. Here, we report a novel two-step production method for PSi nanoparticles. The method is based on centrifuge chemical vapor deposition (cCVD) of elemental silicon in an industrial scale reactor followed by electrochemical post-processing to porous particles. Physical properties, biocompatibility and in vivo biodistribution of the cCVD produced nanoparticles were investigated and compared to PSi nanoparticles conventionally produced from silicon wafers by pulse electrochemical etching. Our results demonstrate that the cCVD production provides PSi nanoparticles with comparable physical and biological quality to the conventional method. This method may circumvent several limitations of the conventional method such as the requirements for high purity monocrystalline silicon substrates as starting material and the material losses during the top-down milling process of the pulse-etched films to porous nanoparticles. However, the electroless etching required for the porosification of cCVD-produced nanoparticles limited control over the pore size, but is amenable for scaling of the production to industrial requirements.
Collapse
Affiliation(s)
- Dave Lumen
- Department of Chemistry, Radiochemistry, University of Helsinki, FI-00014 Helsinki, Finland
| | - Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Surachet Imlimthan
- Department of Chemistry, Radiochemistry, University of Helsinki, FI-00014 Helsinki, Finland
| | - Mirkka Sarparanta
- Department of Chemistry, Radiochemistry, University of Helsinki, FI-00014 Helsinki, Finland
| | - Alexandra Correia
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | | | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland.
| | - Anu J Airaksinen
- Department of Chemistry, Radiochemistry, University of Helsinki, FI-00014 Helsinki, Finland.
| | | | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland.
| |
Collapse
|
12
|
Cevenini A, Celia C, Orrù S, Sarnataro D, Raia M, Mollo V, Locatelli M, Imperlini E, Peluso N, Peltrini R, De Rosa E, Parodi A, Del Vecchio L, Di Marzio L, Fresta M, Netti PA, Shen H, Liu X, Tasciotti E, Salvatore F. Liposome-Embedding Silicon Microparticle for Oxaliplatin Delivery in Tumor Chemotherapy. Pharmaceutics 2020; 12:pharmaceutics12060559. [PMID: 32560359 PMCID: PMC7355455 DOI: 10.3390/pharmaceutics12060559] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 12/16/2022] Open
Abstract
Mesoporous silicon microparticles (MSMPs) can incorporate drug-carrying nanoparticles (NPs) into their pores. An NP-loaded MSMP is a multistage vector (MSV) that forms a Matryoshka-like structure that protects the therapeutic cargo from degradation and prevents its dilution in the circulation during delivery to tumor cells. We developed an MSV constituted by 1 µm discoidal MSMPs embedded with PEGylated liposomes containing oxaliplatin (oxa) which is a therapeutic agent for colorectal cancer (CRC). To obtain extra-small liposomes able to fit the 60 nm pores of MSMP, we tested several liposomal formulations, and identified two optimal compositions, with a prevalence of the rigid lipid 1,2-distearoyl-sn-glycero-3-phosphocholine and of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000]. To improve the MSV assembly, we optimized the liposome-loading inside the MSMP and achieved a five-fold increase of the payload using an innovative lyophilization approach. This procedure also increased the load and limited dimensional changes of the liposomes released from the MSV in vitro. Lastly, we found that the cytotoxic efficacy of oxa-loaded liposomes and-oxa-liposome-MSV in CRC cell culture was similar to that of free oxa. This study increases knowledge about extra-small liposomes and their loading into porous materials and provides useful hints about alternative strategies for designing drug-encapsulating NPs.
Collapse
Affiliation(s)
- Armando Cevenini
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
| | - Christian Celia
- Department of Pharmacy, University of Chieti—Pescara “G. d’Annuzio”, 66100 Chieti, Italy; (C.C.); (M.L.); (L.D.M.)
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (E.D.R.); (H.S.); (X.L.)
| | - Stefania Orrù
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
- Dipartimento di Scienze Motorie e del Benessere, Università “Parthenope”, 80133 Napoli, Italy
- IRCCS SDN, 80143 Napoli, Italy; (E.I.); (A.P.)
| | - Daniela Sarnataro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
| | - Maddalena Raia
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
| | - Valentina Mollo
- Italian Institute of Technology@CRIB Center for Advanced Biomaterials for Health Care, 80125 Napoli, Italy; (V.M.); (P.A.N.)
| | - Marcello Locatelli
- Department of Pharmacy, University of Chieti—Pescara “G. d’Annuzio”, 66100 Chieti, Italy; (C.C.); (M.L.); (L.D.M.)
| | | | - Nicoletta Peluso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
| | - Rosa Peltrini
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
- Department of Respiratory Sciences, College of Life Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Enrica De Rosa
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (E.D.R.); (H.S.); (X.L.)
| | - Alessandro Parodi
- IRCCS SDN, 80143 Napoli, Italy; (E.I.); (A.P.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Luigi Del Vecchio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
| | - Luisa Di Marzio
- Department of Pharmacy, University of Chieti—Pescara “G. d’Annuzio”, 66100 Chieti, Italy; (C.C.); (M.L.); (L.D.M.)
| | - Massimo Fresta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S. Venuta”, I-88100 Catanzaro, Italy;
| | - Paolo Antonio Netti
- Italian Institute of Technology@CRIB Center for Advanced Biomaterials for Health Care, 80125 Napoli, Italy; (V.M.); (P.A.N.)
- Department of Chemical, Materials & Industrial Production Engineering, University of Naples Federico II, 80125 Naples, Italy
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (E.D.R.); (H.S.); (X.L.)
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (E.D.R.); (H.S.); (X.L.)
| | - Ennio Tasciotti
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX 77030, USA
- Houston Methodist Orthopedics & Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Correspondence: (E.T.); (F.S.)
| | - Francesco Salvatore
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
- Correspondence: (E.T.); (F.S.)
| |
Collapse
|
13
|
Design and in vitro characterization of multistage silicon-PLGA budesonide particles for inflammatory bowel disease. Eur J Pharm Biopharm 2020; 151:61-72. [DOI: 10.1016/j.ejpb.2020.03.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 12/28/2022]
|
14
|
Kaushik M, Raghunand R, Maheshwari S. Exploring Promises of siRNA in Cancer Therapeutics. CURRENT CANCER THERAPY REVIEWS 2020. [DOI: 10.2174/1573394715666190207130128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the discovery of the RNA interference (RNAi) in 2006, several attempts have
been made to use it for designing and developing drug treatments for a variety of diseases, including
cancer. In this mini-review, we focus on the potential of small interfering RNAs (siRNA) in
anticancer treatment. We first describe the significant barriers that exist on the path to clinical application
of siRNA drugs. Then the current delivery approaches of siRNAs using lipids, polymers,
and, in particular, polymeric carriers that overcome the aforementioned obstacles have been reviewed.
Also, few siRNA mediated drugs currently in clinical trials for cancer therapy, and a collated
list of siRNA databases having a qualitative and/ or quantitative summary of the data in each
database have been briefly mentioned. This mini review aims to facilitate our understanding about
the siRNA, their delivery systems and the possible barriers in their in vivo usage for biomedical
applications.
Collapse
Affiliation(s)
- Mahima Kaushik
- Cluster Innovation Center, University of Delhi, Delhi, India
| | | | | |
Collapse
|
15
|
Site-Specific 111In-Radiolabeling of Dual-PEGylated Porous Silicon Nanoparticles and Their In Vivo Evaluation in Murine 4T1 Breast Cancer Model. Pharmaceutics 2019; 11:pharmaceutics11120686. [PMID: 31861119 PMCID: PMC6969933 DOI: 10.3390/pharmaceutics11120686] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/05/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022] Open
Abstract
Polyethylene glycol (PEG) has been successfully used for improving circulation time of several nanomaterials but prolonging the circulation of porous silicon nanoparticles (PSi NPs) has remained challenging. Here, we report a site specific radiolabeling of dual-PEGylated thermally oxidized porous silicon (DPEG-TOPSi) NPs and investigation of influence of the PEGylation on blood circulation time of TOPSi NPs. Trans-cyclooctene conjugated DPEG-TOPSi NPs were radiolabeled through a click reaction with [111In]In-DOTA-PEG4-tetrazine (DOTA = 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) and the particle behavior was evaluated in vivo in Balb/c mice bearing 4T1 murine breast cancer allografts. The dual-PEGylation significantly prolonged circulation of [111In]In-DPEG-TOPSi particles when compared to non-PEGylated control particles, yielding 10.8 ± 1.7% of the injected activity/g in blood at 15 min for [111In]In-DPEG-TOPSi NPs. The improved circulation time will be beneficial for the accumulation of targeted DPEG-TOPSi to tumors.
Collapse
|
16
|
Nizzero S, Li F, Zhang G, Venuta A, Borsoi C, Mai J, Shen H, Wolfram J, Li Z, Blanco E, Ferrari M. Systematic comparison of methods for determining the in vivo biodistribution of porous nanostructured injectable inorganic particles. Acta Biomater 2019; 97:501-512. [PMID: 31386927 DOI: 10.1016/j.actbio.2019.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 08/01/2019] [Accepted: 08/01/2019] [Indexed: 12/19/2022]
Abstract
With a wide variety of biodistribution measurement techniques reported in the literature, it is important to perform side-by-side comparisons of results obtained with different methods on the same particle platform, to determine differences across methods, highlight advantages and disadvantages, and inform methods selection according to specific applications. Inorganic nanostructured particles (INPs) have gained a central role in the development of injectable delivery vectors thanks to their controllable design, biocompatibility, and favorable degradation kinetic. Thus, accurate determination of in vivo biodistribution of INPs is a key aspect of developing and optimizing this class of delivery vectors. In this study, a systematic comparison of spectroscopy (inductively coupled plasma optical emission spectroscopy), fluorescence (in vivo imaging system, confocal microscopy, and plate reader), and radiolabeling (gamma counter)-based techniques is performed to assess the accuracy and sensitivity of biodistribution measurements in mice. Each method is evaluated on porous silicon particles, an established and versatile injectable delivery platform. Biodistribution is evaluated in all major organs and compared in terms of absolute results (%ID/g and %ID/organ when possible) and sensitivity (σ%). Finally, we discuss how these results can be extended to inform method selection for other platforms and specific applications, with an outlook to potential benefit for pre-clinical and clinical studies. Overall, this study presents a new practical guide for selection of in vivo biodistribution methods that yield quantitative results. STATEMENT OF SIGNIFICANCE: The significance of this work lies in the use of a single platform to test performances of different biodistribution methods in vivo, with a strict quantitative metric. These results, united with the qualitative comparison of advantages and disadvantages of each technique, are aimed at supporting the rational choice of each different method according to the specific application, to improve the quantitative description of biodistribution results that will be published by others in the future.
Collapse
|
17
|
Park Y, Yoo J, Kang MH, Kwon W, Joo J. Photoluminescent and biodegradable porous silicon nanoparticles for biomedical imaging. J Mater Chem B 2019; 7:6271-6292. [DOI: 10.1039/c9tb01042d] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A set of unique properties including biodegradability, intrinsic photoluminescence, and mesoporous structure allows porous silicon nanoparticles to address current challenges of translational nanomedicine, especially in biomedical imaging.
Collapse
Affiliation(s)
- Yoonsang Park
- Department of Chemical Engineering
- Pohang University of Science and Technology (POSTECH)
- Pohang 37673
- Republic of Korea
| | - Jounghyun Yoo
- Department of Chemical Engineering
- Pohang University of Science and Technology (POSTECH)
- Pohang 37673
- Republic of Korea
| | - Myoung-Hee Kang
- Department of Biomedical Engineering
- School of Life Sciences
- Ulsan National Institute of Science and Technology (UNIST)
- Ulsan 44919
- Republic of Korea
| | - Woosung Kwon
- Department of Chemical and Biological Engineering
- Sookmyung Women's University
- Seoul 04310
- Republic of Korea
| | - Jinmyoung Joo
- Department of Biomedical Engineering
- School of Life Sciences
- Ulsan National Institute of Science and Technology (UNIST)
- Ulsan 44919
- Republic of Korea
| |
Collapse
|
18
|
Tieu T, Alba M, Elnathan R, Cifuentes‐Rius A, Voelcker NH. Advances in Porous Silicon–Based Nanomaterials for Diagnostic and Therapeutic Applications. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800095] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Terence Tieu
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Maria Alba
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
| | - Anna Cifuentes‐Rius
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- Prof. N. H. Voelcker Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton Victoria 3168 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| |
Collapse
|
19
|
Evangelopoulos M, Parodi A, Martinez JO, Tasciotti E. Trends towards Biomimicry in Theranostics. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E637. [PMID: 30134564 PMCID: PMC6164646 DOI: 10.3390/nano8090637] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/27/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022]
Abstract
Over the years, imaging and therapeutic modalities have seen considerable progress as a result of advances in nanotechnology. Theranostics, or the marrying of diagnostics and therapy, has increasingly been employing nano-based approaches to treat cancer. While first-generation nanoparticles offered considerable promise in the imaging and treatment of cancer, toxicity and non-specific distribution hindered their true potential. More recently, multistage nanovectors have been strategically designed to shield and carry a payload to its intended site. However, detection by the immune system and sequestration by filtration organs (i.e., liver and spleen) remains a major obstacle. In an effort to circumvent these biological barriers, recent trends have taken inspiration from biology. These bioinspired approaches often involve the use of biologically-derived cellular components in the design and fabrication of biomimetic nanoparticles. In this review, we provide insight into early nanoparticles and how they have steadily evolved to include bioinspired approaches to increase their theranostic potential.
Collapse
Affiliation(s)
- Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Alessandro Parodi
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60607, USA.
| | - Jonathan O Martinez
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
- Department of Orthopedics & Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA.
| |
Collapse
|
20
|
Li B, Wang F, Gui L, He Q, Yao Y, Chen H. The potential of biomimetic nanoparticles for tumor-targeted drug delivery. Nanomedicine (Lond) 2018; 13:2099-2118. [DOI: 10.2217/nnm-2018-0017] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Bowen Li
- Department of Bioengineering, University of Washington, Seattle, Washington WA 98195, USA
| | - Fei Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, PR China
| | - Lijuan Gui
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, PR China
| | - Qing He
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, PR China
| | - Yuxin Yao
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, PR China
| | - Haiyan Chen
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, PR China
| |
Collapse
|
21
|
Top-down fabrication of shape-controlled, monodisperse nanoparticles for biomedical applications. Adv Drug Deliv Rev 2018; 132:169-187. [PMID: 30009884 DOI: 10.1016/j.addr.2018.07.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 06/08/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023]
Abstract
Nanoparticles for biomedical applications are generally formed by bottom-up approaches such as self-assembly, emulsification and precipitation. But these methods usually have critical limitations in fabrication of nanoparticles with controllable morphologies and monodispersed size. Compared with bottom-up methods, top-down nanofabrication techniques offer advantages of high fidelity and high controllability. This review focuses on top-down nanofabrication techniques for engineering particles along with their biomedical applications. We present several commonly used top-down nanofabrication techniques that have the potential to fabricate nanoparticles, including photolithography, interference lithography, electron beam lithography, mold-based lithography (nanoimprint lithography and soft lithography), nanostencil lithography, and nanosphere lithography. Varieties of current and emerging applications are also covered: (i) targeting, (ii) drug and gene delivery, (iii) imaging, and (iv) therapy. Finally, a future perspective of the nanoparticles fabricated by the top-down techniques in biomedicine is also addressed.
Collapse
|
22
|
Karaman DŞ, Sarparanta MP, Rosenholm JM, Airaksinen AJ. Multimodality Imaging of Silica and Silicon Materials In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1703651. [PMID: 29388264 DOI: 10.1002/adma.201703651] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/15/2017] [Indexed: 05/29/2023]
Abstract
Recent progress in the development of silica- and silicon-based multimodality imaging nanoprobes has advanced their use in image-guided drug delivery, and the development of novel systems for nanotheranostic and diagnostic applications. As biocompatible and flexibly tunable materials, silica and silicon provide excellent platforms with high clinical potential in nanotheranostic and diagnostic probes with well-defined morphology and surface chemistry, yielding multifunctional properties. In vivo imaging is of great value in the exploration of methods for improving site-specific nanotherapeutic delivery by silica- and silicon-based drug-delivery systems. Multimodality approaches are essential for understanding the biological interactions of nanotherapeutics in the physiological environment in vivo. The aim here is to describe recent advances in the development of in vivo imaging tools based on nanostructured silica and silicon, and their applications in single and multimodality imaging.
Collapse
Affiliation(s)
- Didem Şen Karaman
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, BioCity, Tykistökatu 6A, FI, 20520, Turku, Finland
| | - Mirkka P Sarparanta
- Department of Chemistry-Radiochemistry, Faculty of Science, University of Helsinki, POB 55, FI-00014, University of Helsinki, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, BioCity, Tykistökatu 6A, FI, 20520, Turku, Finland
| | - Anu J Airaksinen
- Department of Chemistry-Radiochemistry, Faculty of Science, University of Helsinki, POB 55, FI-00014, University of Helsinki, Finland
| |
Collapse
|
23
|
Li W, Liu Z, Fontana F, Ding Y, Liu D, Hirvonen JT, Santos HA. Tailoring Porous Silicon for Biomedical Applications: From Drug Delivery to Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1703740. [PMID: 29534311 DOI: 10.1002/adma.201703740] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/16/2017] [Indexed: 05/24/2023]
Abstract
In the past two decades, porous silicon (PSi) has attracted increasing attention for its potential biomedical applications. With its controllable geometry, tunable nanoporous structure, large pore volume/high specific surface area, and versatile surface chemistry, PSi shows significant advantages over conventional drug carriers. Here, an overview of recent progress in the use of PSi in drug delivery and cancer immunotherapy is presented. First, an overview of the fabrication of PSi with various geometric structures is provided, with particular focus on how the unique geometry of PSi facilitates its biomedical applications, especially for drug delivery. Second, surface chemistry and modification of PSi are discussed in relation to the strengthening of its performance in drug delivery and bioimaging. Emerging technologies for engineering PSi-based composites are then summarized. Emerging PSi advances in the context of cancer immunotherapy are also highlighted. Overall, very promising research results encourage further exploration of PSi for biomedical applications, particularly in drug delivery and cancer immunotherapy, and future translation of PSi into clinical applications.
Collapse
Affiliation(s)
- Wei Li
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Zehua Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Yaping Ding
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Dongfei Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014, Helsinki, Finland
| | - Jouni T Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014, Helsinki, Finland
| |
Collapse
|
24
|
Salonen J, Mäkilä E. Thermally Carbonized Porous Silicon and Its Recent Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1703819. [PMID: 29484727 DOI: 10.1002/adma.201703819] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/08/2017] [Indexed: 06/08/2023]
Abstract
Recent progress in research on thermally carbonized porous silicon (TCPSi) and its applications is reported. Despite a slow start, thermal carbonization has now started to gain interest mainly due to new emerging areas for applications. These new areas, such as optical sensing, drug delivery, and energy storage, require stable surface chemistry and physical properties. TCPSi is known to have all of these desired properties. Herein, the above-listed properties of TCPSi are summarized, and the carbonization processes, functionalization, and characterization of TCPSi are reviewed. Moreover, some of the emerging fields of TCPSi applications are discussed and recent advances in the fields are introduced.
Collapse
Affiliation(s)
- Jarno Salonen
- Industrial Physics Laboratory, Department of Physics and Astronomy, University of Turku, FI-20014, Turku, Finland
| | - Ermei Mäkilä
- Industrial Physics Laboratory, Department of Physics and Astronomy, University of Turku, FI-20014, Turku, Finland
| |
Collapse
|
25
|
McInnes SJP, Santos A, Kumeria T. Porous Silicon Particles for Cancer Therapy and Bioimaging. NANOONCOLOGY 2018. [DOI: 10.1007/978-3-319-89878-0_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
26
|
Fernandez-Moure JS, Evangelopoulos M, Colvill K, Van Eps JL, Tasciotti E. Nanoantibiotics: a new paradigm for the treatment of surgical infection. Nanomedicine (Lond) 2017; 12:1319-1334. [PMID: 28520517 DOI: 10.2217/nnm-2017-0401] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Infections following orthopedic device implantations often impose a substantial health burden and result in high medical costs. Currently, preventative methods are often employed following an orthopedic implant to reduce risk of infection; however, contamination of the surgical site can still occur. Although antibiotics have demonstrated a substantial reduction in bacterial growth and maintenance, biofilm formation around the implant can often minimize efficacy of the antibiotic. Recently, nanotechnology has garnered significant interest, resulting in the development of several antibiotic delivery strategies that exhibit extended release and increased efficacy. In this review, treatment methods of orthopedic-device-related infections will be discussed and an overview of antimicrobial-based nanotechnologies will be provided. Specifically, nonmetal-, metal- and oxide-based nanotechnologies, incorporating antibacterial strategies, will be discussed.
Collapse
Affiliation(s)
- Joseph S Fernandez-Moure
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA.,Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
| | | | - Kayla Colvill
- University of Texas McGovern Medical School, Houston, TX, USA
| | - Jeffrey L Van Eps
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA.,Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
| | - Ennio Tasciotti
- Department of Orthopedics & Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
27
|
Minardi S, Pandolfi L, Taraballi F, Wang X, De Rosa E, Mills ZD, Liu X, Ferrari M, Tasciotti E. Enhancing Vascularization through the Controlled Release of Platelet-Derived Growth Factor-BB. ACS APPLIED MATERIALS & INTERFACES 2017; 9:14566-14575. [PMID: 28393518 DOI: 10.1021/acsami.6b13760] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
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.
Collapse
Affiliation(s)
| | - Laura Pandolfi
- College of Materials Science and Engineering, University of Chinese Academy of Science , 19A Yuquanlu, Beijing 100049, China
| | | | | | | | | | | | | | - Ennio Tasciotti
- Department of Orthopedics, Houston Methodist Hospital , 6565 Fannin Street, Houston, Texas 77030, United States
| |
Collapse
|
28
|
Balasubramanian V, Correia A, Zhang H, Fontana F, Mäkilä E, Salonen J, Hirvonen J, Santos HA. Biomimetic Engineering Using Cancer Cell Membranes for Designing Compartmentalized Nanoreactors with Organelle-Like Functions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1605375. [PMID: 28112838 DOI: 10.1002/adma.201605375] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/08/2016] [Indexed: 05/18/2023]
Abstract
A new biomimetic nanoreactor design is presented based on cancer cell membrane material in combination with porous silicon nanoparticles. This cellular nanoreactor features a biocompartment enclosed by a cell membrane and readily integrated with cells and supplementing the cellular functions under oxidative stress. The study demonstrates the impact of the nanoreactors on improving cellular functions with a potential to serve as artificial organelles.
Collapse
Affiliation(s)
- Vimalkumar Balasubramanian
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI, 00014, Helsinki, Finland
| | - Alexandra Correia
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI, 00014, Helsinki, Finland
| | - Hongbo Zhang
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI, 00014, Helsinki, Finland
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Flavia Fontana
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI, 00014, Helsinki, Finland
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, University of Turku, FI, 20014, Turku, Finland
| | - Jarno Salonen
- Laboratory of Industrial Physics, University of Turku, FI, 20014, Turku, Finland
| | - Jouni Hirvonen
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI, 00014, Helsinki, Finland
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI, 00014, Helsinki, Finland
| |
Collapse
|
29
|
|
30
|
Biomimetic carriers mimicking leukocyte plasma membrane to increase tumor vasculature permeability. Sci Rep 2016; 6:34422. [PMID: 27703233 PMCID: PMC5050497 DOI: 10.1038/srep34422] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 09/13/2016] [Indexed: 12/05/2022] Open
Abstract
Recent advances in the field of nanomedicine have demonstrated that biomimicry can further improve targeting properties of current nanotechnologies while simultaneously enable carriers with a biological identity to better interact with the biological environment. Immune cells for example employ membrane proteins to target inflamed vasculature, locally increase vascular permeability, and extravasate across inflamed endothelium. Inspired by the physiology of immune cells, we recently developed a procedure to transfer leukocyte membranes onto nanoporous silicon particles (NPS), yielding Leukolike Vectors (LLV). LLV are composed of a surface coating containing multiple receptors that are critical in the cross-talk with the endothelium, mediating cellular accumulation in the tumor microenvironment while decreasing vascular barrier function. We previously demonstrated that lymphocyte function-associated antigen (LFA-1) transferred onto LLV was able to trigger the clustering of intercellular adhesion molecule 1 (ICAM-1) on endothelial cells. Herein, we provide a more comprehensive analysis of the working mechanism of LLV in vitro in activating this pathway and in vivo in enhancing vascular permeability. Our results suggest the biological activity of the leukocyte membrane can be retained upon transplant onto NPS and is critical in providing the particles with complex biological functions towards tumor vasculature.
Collapse
|
31
|
Martinez JO, Evangelopoulos M, Bhavane R, Acciardo S, Salvatore F, Liu X, Ferrari M, Tasciotti E. Multistage Nanovectors Enhance the Delivery of Free and Encapsulated Drugs. Curr Drug Targets 2016; 16:1582-90. [PMID: 25316273 DOI: 10.2174/1389450115666141015113914] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 10/03/2014] [Indexed: 02/08/2023]
Abstract
Nanoparticles have considerable potential for cancer imaging and therapy due to their small size and prolonged circulation. However, biological barriers can impede the delivery of a sufficient dose of a drug to the target site, thereby also resulting in the accumulation of toxic compounds within healthy tissues, and systemic toxicity. Multistage nanovectors (MSV) preferentially accumulate on inflamed endothelium, and can thus serve as carriers for drugs and nanoparticles. Herein, we describe the loading of free (i.e., melittin) and nano-encapsulated (i.e., doxorubicin-loaded micelles) drugs into MSV, and report the impact of surface charge and pore size on drug loading. For both drug formulations, negatively charged MSV (i.e., oxidized) with larger pores were shown to retain higher concentrations of payloads compared to positively charged (i.e., APTES-modified) MSV with small pores. Treatment of human umbilical vein endothelial cells (HUVEC) with melittin-loaded MSV (MEL@MSV) resulted in an 80% reduction in cell viability after 3 days. Furthermore, MEL@MSV conjugated with antivascular endothelial growth factor receptor 2 (VEGFR2) antibodies displayed preferential targeting and delivery of MEL to activated HUVEC expressing VEGFR2. Treatment of HUVEC and MCF7 cells with doxorubicin-loaded micelles (DOXNP@MSV) resulted in a 23% and 47% reduction in cell viability, respectively. Taken together, these results demonstrate increased loading of a payload in oxidized, large pore MSV, and effective delivery of free and nano-encapsulated drugs to endothelial and cancer cells.
Collapse
Affiliation(s)
- Jonathan O Martinez
- Houston Methodist Research Institute, 6670 Bertner Avenue MS R7-414, Houston, TX 77030-2602, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Li L, Liu C, Cao X, Wang Y, Dong J, Qian W. Determination of Carcinoembryonic Antigen by Surface-Enhanced Raman Spectroscopy Using Gold Nanobowl Arrays. ANAL LETT 2016. [DOI: 10.1080/00032719.2016.1205080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
33
|
Yanagishita T, Ueno S, Taniguchi K, Masuda H. Fabrication of Porous Si Particles by Barrel Anode Etching. CHEM LETT 2016. [DOI: 10.1246/cl.160248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
34
|
Shevchuk A, Tokar S, Gopal S, Sanchez-Alonso JL, Tarasov AI, Vélez-Ortega AC, Chiappini C, Rorsman P, Stevens MM, Gorelik J, Frolenkov GI, Klenerman D, Korchev YE. Angular Approach Scanning Ion Conductance Microscopy. Biophys J 2016; 110:2252-65. [PMID: 27224490 PMCID: PMC4880884 DOI: 10.1016/j.bpj.2016.04.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 11/16/2022] Open
Abstract
Scanning ion conductance microscopy (SICM) is a super-resolution live imaging technique that uses a glass nanopipette as an imaging probe to produce three-dimensional (3D) images of cell surface. SICM can be used to analyze cell morphology at nanoscale, follow membrane dynamics, precisely position an imaging nanopipette close to a structure of interest, and use it to obtain ion channel recordings or locally apply stimuli or drugs. Practical implementations of these SICM advantages, however, are often complicated due to the limitations of currently available SICM systems that inherited their design from other scanning probe microscopes in which the scan assembly is placed right above the specimen. Such arrangement makes the setting of optimal illumination necessary for phase contrast or the use of high magnification upright optics difficult. Here, we describe the designs that allow mounting SICM scan head on a standard patch-clamp micromanipulator and imaging the sample at an adjustable approach angle. This angle could be as shallow as the approach angle of a patch-clamp pipette between a water immersion objective and the specimen. Using this angular approach SICM, we obtained topographical images of cells grown on nontransparent nanoneedle arrays, of islets of Langerhans, and of hippocampal neurons under upright optical microscope. We also imaged previously inaccessible areas of cells such as the side surfaces of the hair cell stereocilia and the intercalated disks of isolated cardiac myocytes, and performed targeted patch-clamp recordings from the latter. Thus, our new, to our knowledge, angular approach SICM allows imaging of living cells on nontransparent substrates and a seamless integration with most patch-clamp setups on either inverted or upright microscopes, which would facilitate research in cell biophysics and physiology.
Collapse
Affiliation(s)
- Andrew Shevchuk
- Department of Medicine, Imperial College London, London, United Kingdom.
| | - Sergiy Tokar
- Rayne Institute, King's College London, London, United Kingdom
| | - Sahana Gopal
- Department of Medicine, Imperial College London, London, United Kingdom; Department of Materials and Department of Bioengineering and Institute for Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Jose L Sanchez-Alonso
- National Heart and Lung Institute and Department of Cardiac Medicine, Imperial Center for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
| | - Andrei I Tarasov
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | | | - Ciro Chiappini
- Department of Materials and Department of Bioengineering and Institute for Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Molly M Stevens
- Department of Materials and Department of Bioengineering and Institute for Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Julia Gorelik
- National Heart and Lung Institute and Department of Cardiac Medicine, Imperial Center for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
| | | | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Yuri E Korchev
- Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
35
|
Zhu G, Liu JT, Wang Y, Zhang D, Guo Y, Tasciotti E, Hu Z, Liu X. In Situ Reductive Synthesis of Structural Supported Gold Nanorods in Porous Silicon Particles for Multifunctional Nanovectors. ACS APPLIED MATERIALS & INTERFACES 2016; 8:11881-11891. [PMID: 27123698 DOI: 10.1021/acsami.6b03008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Porous silicon nanodisks (PSD) were fabricated by the combination of photolithography and electrochemical etching of silicon. By using PSD as a reducing agent, gold nanorods (AuNR) were in situ synthesized in the nanopores of PSD, forming PSD-supported-AuNR (PSD/AuNR) hybrid particles. The formation mechanism of AuNR in porous silicon (pSi) was revealed by exploring the role of pSi reducibility and each chemical in the reaction. With the PSD support, AuNR exhibited a stable morphology without toxic surface ligands (CTAB). The PSD/AuNR hybrid particles showed enhanced plasmonic property compared to free AuNR. Because high-density "hot spots" can be generated by controlling the distribution of AuNR supported in PSD, surface-enhanced raman scattering (SERS) using PSD/AuNR as particle substrates was demonstrated. A multifunctional vector, PSD/AuNR/DOX, composed of doxorubicin (DOX)-loaded PSD/AuNR capped with agarose (agar), was developed for highly efficient, combinatorial cancer treatment. Their therapeutic efficacy was examined using two pancreatic cancer cell lines, PANC-1 and MIA PaCa-2. PSD/AuNR/DOX (20 μg Au and 1.25 μg DOX/mL) effectively destroyed these cells under near-IR laser irradiation (810 nm, 15 J·cm(-2) power, 90 s). Overall, we envision that PSD/AuNR may be a promising injectable, multifunctional nanovector for biomedical application.
Collapse
Affiliation(s)
- Guixian Zhu
- College of Materials Sciences and Optoelectronics, University of Chinese Academy of Sciences , Beijing 100049, China
- Department of Nanomedicine, Houston Methodist Research Institute , Houston, Texas 77030, United States
| | - Jen-Tsai Liu
- College of Materials Sciences and Optoelectronics, University of Chinese Academy of Sciences , Beijing 100049, China
| | - Yuzhen Wang
- Department of Nanomedicine, Houston Methodist Research Institute , Houston, Texas 77030, United States
| | - Dechen Zhang
- Department of Nanomedicine, Houston Methodist Research Institute , Houston, Texas 77030, United States
| | - Yi Guo
- Department of Nanomedicine, Houston Methodist Research Institute , Houston, Texas 77030, United States
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute , Houston, Texas 77030, United States
| | - Zhongbo Hu
- College of Materials Sciences and Optoelectronics, University of Chinese Academy of Sciences , Beijing 100049, China
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute , Houston, Texas 77030, United States
| |
Collapse
|
36
|
Minardi S, Corradetti B, Taraballi F, Byun JH, Cabrera F, Liu X, Ferrari M, Weiner BK, Tasciotti E. IL-4 Release from a Biomimetic Scaffold for the Temporally Controlled Modulation of Macrophage Response. Ann Biomed Eng 2016; 44:2008-19. [DOI: 10.1007/s10439-016-1580-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/26/2016] [Indexed: 12/22/2022]
|
37
|
Guinan TM, Kobus H, Lu Y, Sweetman M, McInnes SJP, Kirkbride KP, Voelcker NH. Nanostructured Silicon-Based Fingerprint Dusting Powders for Enhanced Visualization and Detection by Mass Spectrometry. Chempluschem 2016; 81:258-261. [PMID: 31968783 DOI: 10.1002/cplu.201500546] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 01/07/2016] [Indexed: 02/04/2023]
Abstract
Porous silicon microparticles (pSi MPs) functionalized with fluorescent dyes (lissamine and carboxy-5-fluorescein) and intrinsically luminescent pSi MPs were explored as novel fingerprint dusting powders. The versatility of luminescent pSi MPs is demonstrated through time-gated imaging of their long-lived (lifetime>28 μs) near-IR emission, and mass spectrometry analysis of fingerprints dusted with luminescent pSi MPs to provide further information on exogenous small molecules present in latent fingerprints.
Collapse
Affiliation(s)
- Taryn M Guinan
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia
| | - Hilton Kobus
- School of Physical and Chemical Sciences, Flinders University, Bedford Park, SA, 5042, Australia
| | - Yiqing Lu
- Department of Physics and Astronomy, Faculty of Science and Engineering, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Martin Sweetman
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia
| | - Steven J P McInnes
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia
| | - K Paul Kirkbride
- School of Physical and Chemical Sciences, Flinders University, Bedford Park, SA, 5042, Australia
| | - Nicolas H Voelcker
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia
| |
Collapse
|
38
|
The Importance of Particle Geometry in Design of Therapeutic and Imaging Nanovectors. ADVANCES IN DELIVERY SCIENCE AND TECHNOLOGY 2016. [DOI: 10.1007/978-1-4939-3634-2_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
39
|
Pandolfi L, Minardi S, Taraballi F, Liu X, Ferrari M, Tasciotti E. Composite microsphere-functionalized scaffold for the controlled release of small molecules in tissue engineering. J Tissue Eng 2016; 7:2041731415624668. [PMID: 26977286 PMCID: PMC4765809 DOI: 10.1177/2041731415624668] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/20/2015] [Indexed: 12/17/2022] Open
Abstract
Current tissue engineering strategies focus on restoring damaged tissue architectures using biologically active scaffolds. The ideal scaffold would mimic the extracellular matrix of any tissue of interest, promoting cell proliferation and de novo extracellular matrix deposition. A plethora of techniques have been evaluated to engineer scaffolds for the controlled and targeted release of bioactive molecules to provide a functional structure for tissue growth and remodeling, as well as enhance recruitment and proliferation of autologous cells within the implant. Recently, novel approaches using small molecules, instead of growth factors, have been exploited to regulate tissue regeneration. The use of small synthetic molecules could be very advantageous because of their stability, tunability, and low cost. Herein, we propose a chitosan-gelatin scaffold functionalized with composite microspheres consisting of mesoporous silicon microparticles and poly(dl-lactic-co-glycolic acid) for the controlled release of sphingosine-1-phospate, a small molecule of interest. We characterized the platform with scanning electron microscopy, Fourier transform infrared spectroscopy, and confocal microscopy. Finally, the biocompatibility of this multiscale system was analyzed by culturing human mesenchymal stem cells onto the scaffold. The presented strategy establishes the basis of a versatile scaffold for the controlled release of small molecules and for culturing mesenchymal stem cells for regenerative medicine applications.
Collapse
Affiliation(s)
- Laura Pandolfi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
- College of Materials Science and Engineering, University of Chinese Academy of Science, Beijing, China
| | - Silvia Minardi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Francesca Taraballi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Xeuwu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
40
|
|
41
|
Wolfram J, Shen H, Ferrari M. Multistage vector (MSV) therapeutics. J Control Release 2015; 219:406-415. [PMID: 26264836 PMCID: PMC4656100 DOI: 10.1016/j.jconrel.2015.08.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/01/2015] [Accepted: 08/04/2015] [Indexed: 12/21/2022]
Abstract
One of the greatest challenges in the field of medicine is obtaining controlled distribution of systemically administered therapeutic agents within the body. Indeed, biological barriers such as physical compartmentalization, pressure gradients, and excretion pathways adversely affect localized delivery of drugs to pathological tissue. The diverse nature of these barriers requires the use of multifunctional drug delivery vehicles that can overcome a wide range of sequential obstacles. In this review, we explore the role of multifunctionality in nanomedicine by primarily focusing on multistage vectors (MSVs). The MSV is an example of a promising therapeutic platform that incorporates several components, including a microparticle, nanoparticles, and small molecules. In particular, these components are activated in a sequential manner in order to successively address transport barriers.
Collapse
Affiliation(s)
- Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
42
|
Evangelopoulos M, Parodi A, Martinez JO, Yazdi IK, Cevenini A, van de Ven AL, Quattrocchi N, Boada C, Taghipour N, Corbo C, Brown BS, Scaria S, Liu X, Ferrari M, Tasciotti E. Cell source determines the immunological impact of biomimetic nanoparticles. Biomaterials 2015; 82:168-77. [PMID: 26761780 DOI: 10.1016/j.biomaterials.2015.11.054] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/17/2015] [Accepted: 11/28/2015] [Indexed: 12/24/2022]
Abstract
Recently, engineering the surface of nanotherapeutics with biologics to provide them with superior biocompatibility and targeting towards pathological tissues has gained significant popularity. Although the functionalization of drug delivery vectors with cellular materials has been shown to provide synthetic particles with unique biological properties, these approaches may have undesirable immunological repercussions upon systemic administration. Herein, we comparatively analyzed unmodified multistage nanovectors and particles functionalized with murine and human leukocyte cellular membrane, dubbed Leukolike Vectors (LLV), and the immunological effects that may arise in vitro and in vivo. Previously, LLV demonstrated an avoidance of opsonization and phagocytosis, in addition to superior targeting of inflammation and prolonged circulation. In this work, we performed a comprehensive evaluation of the importance of the source of cellular membrane in increasing their systemic tolerance and minimizing an inflammatory response. Time-lapse microscopy revealed LLV developed using a cellular coating derived from a murine (i.e., syngeneic) source resulted in an active avoidance of uptake by macrophage cells. Additionally, LLV composed of a murine membrane were found to have decreased uptake in the liver with no significant effect on hepatic function. As biomimicry continues to develop, this work demonstrates the necessity to consider the source of biological material in the development of future drug delivery carriers.
Collapse
Affiliation(s)
- Michael Evangelopoulos
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Alessandro Parodi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan 20133, Italy
| | - Jonathan O Martinez
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Iman K Yazdi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Armando Cevenini
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy; CEINGE-Biotecnologie Avanzate s.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Anne L van de Ven
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Nicoletta Quattrocchi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Christian Boada
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Nuevo Leon, Mexico
| | - Nima Taghipour
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Claudia Corbo
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Brandon S Brown
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Shilpa Scaria
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| |
Collapse
|
43
|
Scavo MP, Gentile E, Wolfram J, Gu J, Barone M, Evangelopoulos M, Martinez JO, Liu X, Celia C, Tasciotti E, Vilar E, Shen H. Multistage vector delivery of sulindac and silymarin for prevention of colon cancer. Colloids Surf B Biointerfaces 2015; 136:694-703. [PMID: 26513752 DOI: 10.1016/j.colsurfb.2015.10.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/09/2015] [Accepted: 10/04/2015] [Indexed: 02/07/2023]
Abstract
Familial adenomatous polyposis (FAP) is an inherited condition secondary to germline mutations in the APC gene, thus resulting in the formation of hundreds of colonic adenomas that eventually progress into colon cancer. Surgical removal of the colon remains the only treatment option to avoid malignancy, as long-term exposure to chemopreventive agents such as sulindac (a non-steroidal anti-inflammatory drug) and silymarin (phytoestrogen) is not feasible. Here, we have developed a multistage silicon-based drug delivery platform for sulindac and silymarin that preferentially interacts with colon cancer cells as opposed to normal intestinal mucosa. Preferential binding and internalization of these drugs into colon cancer cells was obtained using a targeting strategy against the protein meprin A, which we demonstrate is overexpressed in human colon cancer cells and in the small intestine of Apc(Min/+) mice. We propose that this delivery system could potentially be used to reduce drug-induced side effects in FAP patients, thus enabling long-term prevention of adenoma formation.
Collapse
Affiliation(s)
- Maria Principia Scavo
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Emanuela Gentile
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Thoracic & Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, Beijing 100190, China
| | - Jianhua Gu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Michele Barone
- Gastroentrology Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari 70121, Italy
| | - Michael Evangelopoulos
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jonathan O Martinez
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Christian Celia
- Department of Pharmacy, University G. d'Annunzio of Chieti, Pescara 66013, Italy
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
44
|
Yazdi IK, Ziemys A, Evangelopoulos M, Martinez JO, Kojic M, Tasciotti E. Physicochemical properties affect the synthesis, controlled delivery, degradation and pharmacokinetics of inorganic nanoporous materials. Nanomedicine (Lond) 2015; 10:3057-3075. [DOI: 10.2217/nnm.15.133] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Controlling size, shape and uniformity of porous constructs remains a major focus of the development of porous materials. Over the past two decades, we have seen significant developments in the fabrication of new, porous-ordered structures using a wide range of materials, resulting in properties well beyond their traditional use. Porous materials have been considered appealing, due to attractive properties such as pore size length, morphology and surface chemistry. Furthermore, their utilization within the life sciences and medicine has resulted in significant developments in pharmaceutics and medical diagnosis. This article focuses on various classes of porous materials, providing an overview of principle concepts with regard to design and fabrication, surface chemistry and loading and release kinetics. Furthermore, predictions from a multiscale mathematical model revealed the role pore length and diameter could have on payload release kinetics.
Collapse
Affiliation(s)
- Iman K Yazdi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | - Arturas Ziemys
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Michael Evangelopoulos
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jonathan O Martinez
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Milos Kojic
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
45
|
D'Apolito R, Tomaiuolo G, Taraballi F, Minardi S, Kirui D, Liu X, Cevenini A, Palomba R, Ferrari M, Salvatore F, Tasciotti E, Guido S. Red blood cells affect the margination of microparticles in synthetic microcapillaries and intravital microcirculation as a function of their size and shape. J Control Release 2015; 217:263-72. [PMID: 26381900 DOI: 10.1016/j.jconrel.2015.09.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/05/2015] [Accepted: 09/09/2015] [Indexed: 12/29/2022]
Abstract
A key step in particle-based drug delivery throughmicrocirculation is particlemigration from blood flow to vesselwalls, also known as “margination”,which promotes particle contact and adhesion to the vesselwall. Margination and adhesion should be independently addressed as two distinct phenomena, considering that the former is a fundamental prerequisite to achieve particle adhesion and subsequent extravasation. Although margination has beenmodeled by numerical simulations and investigated inmodel systems in vitro, experimental studies including red blood cells (RBCs) are lacking. Here, we evaluate the effect of RBCs on margination through microfluidic studies in vitro and by intravital microscopy in vivo.We showthatmargination,which is almost absent when particles are suspended in a cell-free medium, is drastically enhanced by RBCs. This effect is size- and shape-dependent, larger spherical/discoid particles being more effectively marginated both in vitro and in vivo. Our findings can be explained by the collision of particles with RBCs that induces the drifting of the particles towards the vessel walls where they become trapped in the cell-free layer. These results are relevant for the design of drug delivery strategies based on systemically administered carriers.
Collapse
Affiliation(s)
- Rosa D'Apolito
- Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, Università di Napoli Federico II, Italy; CEINGE Biotecnologie avanzate, Napoli, Italy
| | - Giovanna Tomaiuolo
- Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, Università di Napoli Federico II, Italy; CEINGE Biotecnologie avanzate, Napoli, Italy.
| | - Francesca Taraballi
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Silvia Minardi
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Dickson Kirui
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, TX, USA; Naval Medical Research Unit, San Antonio, TX, USA
| | - Xuewu Liu
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Armando Cevenini
- Department of Molecular Medicine and Medical Biotechnology, Università di Napoli Federico II, Italy
| | - Roberto Palomba
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Mauro Ferrari
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Francesco Salvatore
- Department of Molecular Medicine and Medical Biotechnology, Università di Napoli Federico II, Italy; CEINGE Biotecnologie avanzate, Napoli, Italy
| | - Ennio Tasciotti
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Stefano Guido
- Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, Università di Napoli Federico II, Italy; CEINGE Biotecnologie avanzate, Napoli, Italy
| |
Collapse
|
46
|
Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol 2015; 33:941-51. [PMID: 26348965 PMCID: PMC4978509 DOI: 10.1038/nbt.3330] [Citation(s) in RCA: 4276] [Impact Index Per Article: 427.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 07/29/2015] [Indexed: 12/11/2022]
Abstract
Biological barriers to drug transport prevent successful accumulation of nanotherapeutics specifically at diseased sites, limiting efficacious responses in disease processes ranging from cancer to inflammation. Although substantial research efforts have aimed to incorporate multiple functionalities and moieties within the overall nanoparticle design, many of these strategies fail to adequately address these barriers. Obstacles, such as nonspecific distribution and inadequate accumulation of therapeutics, remain formidable challenges to drug developers. A reimagining of conventional nanoparticles is needed to successfully negotiate these impediments to drug delivery. Site-specific delivery of therapeutics will remain a distant reality unless nanocarrier design takes into account the majority, if not all, of the biological barriers that a particle encounters upon intravenous administration. By successively addressing each of these barriers, innovative design features can be rationally incorporated that will create a new generation of nanotherapeutics, realizing a paradigmatic shift in nanoparticle-based drug delivery.
Collapse
Affiliation(s)
- Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
47
|
Alba M, Delalat B, Formentín P, Rogers ML, Marsal LF, Voelcker NH. Silica Nanopills for Targeted Anticancer Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:4626-4631. [PMID: 26097092 DOI: 10.1002/smll.201402930] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 04/05/2015] [Indexed: 06/04/2023]
Abstract
Multifunctional SiO2 microtubes for targeted drug delivery are produced with precise control over shape and size by combining lithography and electrochemical etching. The hollow core is loaded with a lipophilic anticancer drug generating nanopills and an antibody is conjugated to the external surface for cancer cell targeting. Results demonstrate selective killing of neuroblastoma cells that express the cognate receptor.
Collapse
Affiliation(s)
- María Alba
- Departament d'Enginyeria ElectrònicaElèctrica i Automàtica, Universitat Rovira i Virgili, Avda Països Catalans 26, Tarragona, 43007, Spain
| | - Bahman Delalat
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Mawson Institute, University of South Australia, Mawson Lakes, SA, 5001, Australia
| | - Pilar Formentín
- Departament d'Enginyeria ElectrònicaElèctrica i Automàtica, Universitat Rovira i Virgili, Avda Països Catalans 26, Tarragona, 43007, Spain
| | - Mary-Louise Rogers
- Department of Human Physiology, Centre for Neuroscience, Flinders University, Bedford Park, SA, 5042, Australia
| | - Lluís F Marsal
- Departament d'Enginyeria ElectrònicaElèctrica i Automàtica, Universitat Rovira i Virgili, Avda Països Catalans 26, Tarragona, 43007, Spain
| | - Nicolas H Voelcker
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Mawson Institute, University of South Australia, Mawson Lakes, SA, 5001, Australia
| |
Collapse
|
48
|
Minardi S, Pandolfi L, Taraballi F, De Rosa E, Yazdi IK, Liu X, Ferrari M, Tasciotti E. PLGA-Mesoporous Silicon Microspheres for the in Vivo Controlled Temporospatial Delivery of Proteins. ACS APPLIED MATERIALS & INTERFACES 2015; 7:16364-16373. [PMID: 26108253 DOI: 10.1021/acsami.5b03464] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
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-MSV's 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-MSV's 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.
Collapse
Affiliation(s)
- Silvia Minardi
- †Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
- ‡Institute of Science and Technology for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Ravenna, Italy
| | - Laura Pandolfi
- †Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
- §College of Materials Science and Engineering, University of Chinese Academy of Science, 19A Yuquanlu, Beijing 100049, China
| | - Francesca Taraballi
- †Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| | - Enrica De Rosa
- †Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| | - Iman K Yazdi
- †Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| | - Xeuwu Liu
- †Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| | - Mauro Ferrari
- †Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| | - Ennio Tasciotti
- †Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| |
Collapse
|
49
|
Joo J, Liu X, Kotamraju VR, Ruoslahti E, Nam Y, Sailor MJ. Gated Luminescence Imaging of Silicon Nanoparticles. ACS NANO 2015; 9:6233-41. [PMID: 26034817 PMCID: PMC4931905 DOI: 10.1021/acsnano.5b01594] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The luminescence lifetime of nanocrystalline silicon is typically on the order of microseconds, significantly longer than the nanosecond lifetimes exhibited by fluorescent molecules naturally present in cells and tissues. Time-gated imaging, where the image is acquired at a time after termination of an excitation pulse, allows discrimination of a silicon nanoparticle probe from these endogenous signals. Because of the microsecond time scale for silicon emission, time-gated imaging is relatively simple to implement for this biocompatible and nontoxic probe. Here a time-gated system with ∼10 ns resolution is described, using an intensified CCD camera and pulsed LED or laser excitation sources. The method is demonstrated by tracking the fate of mesoporous silicon nanoparticles containing the tumor-targeting peptide iRGD, administered by retro-orbital injection into live mice. Imaging of such systemically administered nanoparticles in vivo is particularly challenging because of the low concentration of probe in the targeted tissues and relatively high background signals from tissue autofluorescence. Contrast improvements of >100-fold (relative to steady-state imaging) is demonstrated in the targeted tissues.
Collapse
Affiliation(s)
- Jinmyoung Joo
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, mc 0358, La Jolla, California 92093, United States
| | - Xiangyou Liu
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, United States
| | - Venkata Ramana Kotamraju
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, United States
| | - Erkki Ruoslahti
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, United States
- Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California 93106-9610, United States
| | - Yoonkey Nam
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Michael J. Sailor
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, mc 0358, La Jolla, California 92093, United States
- Address correspondence to
| |
Collapse
|
50
|
Petersen RS, Keller SS, Boisen A. Hot punching of high-aspect-ratio 3D polymeric microstructures for drug delivery. LAB ON A CHIP 2015; 15:2576-2579. [PMID: 25976735 DOI: 10.1039/c5lc00372e] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Hot punching with two different strategies has been demonstrated as a new method of fabricating high aspect ratio 3D microstructures for drug delivery. It has been shown that this process is highly versatile with good replication fidelity and yield.
Collapse
Affiliation(s)
- Ritika S Petersen
- Department of Micro- and Nanotechnology, Technical University of Denmark, Building 345B, Kongens Lyngby DK-2800, Denmark.
| | | | | |
Collapse
|