1
|
Brezgin S, Parodi A, Kostyusheva A, Ponomareva N, Lukashev A, Sokolova D, Pokrovsky VS, Slatinskaya O, Maksimov G, Zamyatnin AA, Chulanov V, Kostyushev D. Technological aspects of manufacturing and analytical control of biological nanoparticles. Biotechnol Adv 2023; 64:108122. [PMID: 36813011 DOI: 10.1016/j.biotechadv.2023.108122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/19/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived biological nanoparticles that gained great interest for drug delivery. EVs have numerous advantages compared to synthetic nanoparticles, such as ideal biocompatibility, safety, ability to cross biological barriers and surface modification via genetic or chemical methods. On the other hand, the translation and the study of these carriers resulted difficult, mostly because of significant issues in up-scaling, synthesis and impractical methods of quality control. However, current manufacturing advances enable EV packaging with any therapeutic cargo, including DNA, RNA (for RNA vaccines and RNA therapeutics), proteins, peptides, RNA-protein complexes (including gene-editing complexes) and small molecules drugs. To date, an array of new and upgraded technologies have been introduced, substantially improving EV production, isolation, characterization and standardization. The used-to-be "gold standards" of EV manufacturing are now outdated, and the state-of-art requires extensive revision. This review re-evaluates the pipeline for EV industrial production and provides a critical overview of the modern technologies required for their synthesis and characterization.
Collapse
Affiliation(s)
- Sergey Brezgin
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia
| | | | - Anastasiya Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia
| | - Natalia Ponomareva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia
| | - Alexander Lukashev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia
| | - Darina Sokolova
- Sirius University of Science and Technology, Sochi 354340, Russia; Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; People's Friendship University, Moscow 117198, Russia
| | - Vadim S Pokrovsky
- Sirius University of Science and Technology, Sochi 354340, Russia; Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; People's Friendship University, Moscow 117198, Russia
| | - Olga Slatinskaya
- Lomonosov Moscow State University, Faculty of Biology, Moscow 119991, Russia
| | - Georgy Maksimov
- Lomonosov Moscow State University, Faculty of Biology, Moscow 119991, Russia
| | - Andrey A Zamyatnin
- Sirius University of Science and Technology, Sochi 354340, Russia; Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7X, UK
| | - Vladimir Chulanov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia; Department of Infectious Diseases, Sechenov University, Moscow 119048, Russia; National Medical Research Center for Tuberculosis and Infectious Diseases, Moscow 127994, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia.
| |
Collapse
|
2
|
Rao JS, Ivkov R, Sharma A. Nanoparticle-Based Interventions for Liver Transplantation. Int J Mol Sci 2023; 24:7496. [PMID: 37108659 PMCID: PMC10144867 DOI: 10.3390/ijms24087496] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/29/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Liver transplantation is the only treatment for hepatic insufficiency as a result of acute and chronic liver injuries/pathologies that fail to recover. Unfortunately, there remains an enormous and growing gap between organ supply and demand. Although recipients on the liver transplantation waitlist have significantly higher mortality, livers are often not allocated because they are (i) classified as extended criteria or marginal livers and (ii) subjected to longer cold preservation time (>6 h) with a direct correlation of poor outcomes with longer cold ischemia. Downregulating the recipient's innate immune response to successfully tolerate a graft having longer cold ischemia times or ischemia-reperfusion injury through induction of immune tolerance in the graft and the host would significantly improve organ utilization and post-transplant outcomes. Broadly, technologies proposed for development aim to extend the life of the transplanted liver through post-transplant or recipient conditioning. In this review, we focus on the potential benefits of nanotechnology to provide unique pre-transplant grafting and recipient conditioning of extended criteria donor livers using immune tolerance induction and hyperthermic pre-conditioning.
Collapse
Affiliation(s)
- Joseph Sushil Rao
- Division of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anirudh Sharma
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
3
|
Soprano E, Polo E, Pelaz B, del Pino P. Biomimetic cell-derived nanocarriers in cancer research. J Nanobiotechnology 2022; 20:538. [PMID: 36544135 PMCID: PMC9771790 DOI: 10.1186/s12951-022-01748-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Nanoparticles have now long demonstrated capabilities that make them attractive to use in biology and medicine. Some of them, such as lipid nanoparticles (SARS-CoV-2 vaccines) or metallic nanoparticles (contrast agents) are already approved for their use in the clinic. However, considering the constantly growing body of different formulations and the huge research around nanomaterials the number of candidates reaching clinical trials or being commercialized is minimal. The reasons behind being related to the "synthetic" and "foreign" character of their surface. Typically, nanomaterials aiming to develop a function or deliver a cargo locally, fail by showing strong off-target accumulation and generation of adverse responses, which is connected to their strong recognition by immune phagocytes primarily. Therefore, rendering in negligible numbers of nanoparticles developing their intended function. While a wide range of coatings has been applied to avoid certain interactions with the surrounding milieu, the issues remained. Taking advantage of the natural cell membranes, in an approach that resembles a cell transfer, the use of cell-derived surfaces has risen as an alternative to artificial coatings or encapsulation methods. Biomimetic technologies are based on the use of isolated natural components to provide autologous properties to the nanoparticle or cargo being encapsulated, thus, improving their therapeutic behavior. The main goal is to replicate the (bio)-physical properties and functionalities of the source cell and tissue, not only providing a stealthy character to the core but also taking advantage of homotypic properties, that could prove relevant for targeted strategies. Such biomimetic formulations have the potential to overcome the main issues of approaches to provide specific features and identities synthetically. In this review, we provide insight into the challenges of nano-biointerfaces for drug delivery; and the main applications of biomimetic materials derived from specific cell types, focusing on the unique strengths of the fabrication of novel nanotherapeutics in cancer therapy.
Collapse
Affiliation(s)
- Enrica Soprano
- grid.11794.3a0000000109410645Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Ester Polo
- grid.11794.3a0000000109410645Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- grid.11794.3a0000000109410645Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Pablo del Pino
- grid.11794.3a0000000109410645Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| |
Collapse
|
4
|
Biomimetic approaches for targeting tumor inflammation. Semin Cancer Biol 2022; 86:555-567. [DOI: 10.1016/j.semcancer.2022.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/31/2022] [Accepted: 04/20/2022] [Indexed: 02/08/2023]
|
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: 7.3] [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
|
Pereira-Silva M, Chauhan G, Shin MD, Hoskins C, Madou MJ, Martinez-Chapa SO, Steinmetz NF, Veiga F, Paiva-Santos AC. Unleashing the potential of cell membrane-based nanoparticles for COVID-19 treatment and vaccination. Expert Opin Drug Deliv 2021; 18:1395-1414. [PMID: 33944644 PMCID: PMC8182831 DOI: 10.1080/17425247.2021.1922387] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/23/2021] [Indexed: 12/22/2022]
Abstract
Introduction: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a particular coronavirus strain responsible for the coronavirus disease 2019 (COVID-19), accounting for more than 3.1 million deaths worldwide. Several health-related strategies have been successfully developed to contain the rapidly-spreading virus across the globe, toward reduction of both disease burden and infection rates. Particularly, attention has been focused on either the development of novel drugs and vaccines, or by adapting already-existing drugs for COVID-19 treatment, mobilizing huge efforts to block disease progression and to overcome the shortage of effective measures available at this point.Areas covered: This perspective covers the breakthrough of multifunctional biomimetic cell membrane-based nanoparticles as next-generation nanosystems for cutting-edge COVID-19 therapeutics and vaccination, specifically cell membrane-derived nanovesicles and cell membrane-coated nanoparticles, both tailorable cell membrane-based nanosystems enriched with the surface repertoire of native cell membranes, toward maximized biointerfacing, immune evasion, cell targeting and cell-mimicking properties.Expert opinion: Nano-based approaches have received widespread interest regarding enhanced antigen delivery, prolonged blood circulation half-life and controlled release of drugs. Cell membrane-based nanoparticles comprise interesting antiviral multifunctional nanoplatforms for blocking SARS-CoV-2 binding to host cells, reducing inflammation through cytokine neutralization and improving drug delivery toward COVID-19 treatment.
Collapse
Affiliation(s)
- Miguel Pereira-Silva
- Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
| | - Gaurav Chauhan
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey, Mexico
| | - Matthew D. Shin
- Department of Nanoengineering, University of California, San Diego, San Diego, United States
| | - Clare Hoskins
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Marc J. Madou
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey, Mexico
- Department of Mechanical and Aerospace Engineering, University of California Irvine, Engineering Gateway 4200, Irvine, United States
| | | | - Nicole F. Steinmetz
- Department of Nanoengineering, University of California, San Diego, San Diego, United States
- Department of Bioengineering, University of California, San Diego, United States
- Department of Radiology, UC San Diego Health, University of California, San Diego, United States
- Center for Nano-ImmunoEngineering (Nanoie), University of California, San Diego, United States
- Moores Cancer Center, UC San Diego Health, University of California, San Diego, United States
| | - Francisco Veiga
- Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
7
|
Jahromi LP, Shahbazi M, Maleki A, Azadi A, Santos HA. Chemically Engineered Immune Cell-Derived Microrobots and Biomimetic Nanoparticles: Emerging Biodiagnostic and Therapeutic Tools. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002499. [PMID: 33898169 PMCID: PMC8061401 DOI: 10.1002/advs.202002499] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/26/2020] [Indexed: 05/16/2023]
Abstract
Over the past decades, considerable attention has been dedicated to the exploitation of diverse immune cells as therapeutic and/or diagnostic cell-based microrobots for hard-to-treat disorders. To date, a plethora of therapeutics based on alive immune cells, surface-engineered immune cells, immunocytes' cell membranes, leukocyte-derived extracellular vesicles or exosomes, and artificial immune cells have been investigated and a few have been introduced into the market. These systems take advantage of the unique characteristics and functions of immune cells, including their presence in circulating blood and various tissues, complex crosstalk properties, high affinity to different self and foreign markers, unique potential of their on-demand navigation and activity, production of a variety of chemokines/cytokines, as well as being cytotoxic in particular conditions. Here, the latest progress in the development of engineered therapeutics and diagnostics inspired by immune cells to ameliorate cancer, inflammatory conditions, autoimmune diseases, neurodegenerative disorders, cardiovascular complications, and infectious diseases is reviewed, and finally, the perspective for their clinical application is delineated.
Collapse
Affiliation(s)
- Leila Pourtalebi Jahromi
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Pharmaceutical Sciences Research CenterShiraz University of Medical SciencesShiraz71468‐64685Iran
- Present address:
Helmholtz Institute for Pharmaceutical Research SaarlandHelmholtz Centre for Infection ResearchBiogenic Nanotherapeutics GroupCampus E8.1Saarbrücken66123Germany
| | - Mohammad‐Ali Shahbazi
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC)Zanjan University of Medical SciencesZanjan45139‐56184Iran
| | - Aziz Maleki
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC)Zanjan University of Medical SciencesZanjan45139‐56184Iran
| | - Amir Azadi
- Pharmaceutical Sciences Research CenterShiraz University of Medical SciencesShiraz71468‐64685Iran
- Department of PharmaceuticsSchool of PharmacyShiraz University of Medical SciencesShiraz71468‐64685Iran
| | - Hélder A. Santos
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFI‐00014Finland
| |
Collapse
|
8
|
Martinelli C. Smart Nanocarriers for Targeted Cancer Therapy. Anticancer Agents Med Chem 2021; 21:546-557. [PMID: 32560615 DOI: 10.2174/1871520620666200619181425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/27/2020] [Accepted: 04/24/2020] [Indexed: 11/22/2022]
Abstract
Cancer is considered one of the most threatening diseases worldwide. Although many therapeutic approaches have been developed and optimized for ameliorating patient's conditions and life expectancy, however, it frequently remains an incurable pathology. Notably, conventional treatments may reveal inefficient in the presence of metastasis development, multidrug resistance and inability to achieve targeted drug delivery. In the last decades, nanomedicine has gained a prominent role, due to many properties ascribable to nanomaterials. It is worth mentioning their small size, their ability to be loaded with small drugs and bioactive molecules and the possibility to be functionalized for tumor targeting. Natural vehicles have been exploited, such as exosomes, and designed, such as liposomes. Biomimetic nanomaterials have been engineered, by modification with biological membrane coating. Several nanoparticles have already entered clinical trials and some liposomal formulations have been approved for therapeutic applications. In this review, natural and synthetic nanocarriers functionalized for actively targeting cancer cells will be described, focusing on their advantages with respect to conventional treatments. Recent innovations related to biomimetic nanoparticles camouflaged with membranes isolated from different types of cells will be reported, together with their promising applications. Finally, a short overview on the latest advances in carrier-free nanomaterials will be provided.
Collapse
Affiliation(s)
- Chiara Martinelli
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| |
Collapse
|
9
|
Biomimetic Nanoparticles Potentiate the Anti-Inflammatory Properties of Dexamethasone and Reduce the Cytokine Storm Syndrome: An Additional Weapon against COVID-19? NANOMATERIALS 2020; 10:nano10112301. [PMID: 33233748 PMCID: PMC7699958 DOI: 10.3390/nano10112301] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022]
Abstract
Recent studies on coronavirus infectious disease 2019 (COVID-19) pathophysiology indicated the cytokine release syndrome induced by the virus as the main cause of mortality. Patients with severe COVID-19 infection present a systemic hyper inflammation that can lead to lung and multi-organ injuries. Among the most recent treatments, corticosteroids have been identified to be effective in mitigating these catastrophic effects. Our group has recently developed leukocyte-derived nanovesicles, termed leukosomes, able to target in vivo the inflamed vasculature associated with pathological conditions including cancer, cardiovascular diseases, and sepsis. Herein, to gain insights on the anti-inflammatory properties of leukosomes, we investigated their ability to reduce uncontrolled inflammation in a lethal model of lipopolysaccharide (LPS)-induced endotoxemia, recapitulating the cytokine storm syndrome observed in COVID-19 infection after encapsulating dexamethasone. Treated animals showed a significant survival advantage and an improved immune response resolution, as demonstrated by a cytokine array analysis of pro- and anti-inflammatory cytokines, chemokines, and other immune-relevant markers. Our results showed that leukosomes enhance the therapeutic activity of dexamethasone and better control the inflammatory response compared to the free drug. Such an approach could be useful for the development of personalized therapies in the treatment of hyperinflammation related to infectious diseases, including the ones caused by COVID-19.
Collapse
|
10
|
Zinger A, Brozovich A, Pasto A, Sushnitha M, Martinez JO, Evangelopoulos M, Boada C, Tasciotti E, Taraballi F. Bioinspired Extracellular Vesicles: Lessons Learned From Nature for Biomedicine and Bioengineering. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2172. [PMID: 33143238 PMCID: PMC7693812 DOI: 10.3390/nano10112172] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/23/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022]
Abstract
Efficient communication is essential in all layers of the biological chain. Cells exchange information using a variety of signaling moieties, such as small molecules, proteins, and nucleic acids. Cells carefully package these messages into lipid complexes, collectively named extracellular vesicles (EVs). In this work, we discuss the nature of these cell carriers, categorize them by their origin, explore their role in the homeostasis of healthy tissues, and examine how they regulate the pathophysiology of several diseases. This review will also address the limitations of using EVs for clinical applications and discuss novel methods to engineer nanoparticles to mimic the structure, function, and features of EVs. Using lessons learned from nature and understanding how cells use EVs to communicate across distant sites, we can develop a better understanding of how to tailor the fundamental features of drug delivery carriers to encapsulate various cargos and target specific sites for biomedicine and bioengineering.
Collapse
Affiliation(s)
- Assaf Zinger
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ava Brozovich
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Texas A&M College of Medicine, Bryan, TX 77807, USA
| | - Anna Pasto
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Italy
| | - Manuela Sushnitha
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Jonathan O. Martinez
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Michael Evangelopoulos
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Christian Boada
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ennio Tasciotti
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Biotechnology Program, San Raffaele University, Via di Val Cannuta, 247, 00166 Roma RM, Italy
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
11
|
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: 4.8] [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
|
12
|
Arrighetti N, Corbo C, Evangelopoulos M, Pastò A, Zuco V, Tasciotti E. Exosome-like Nanovectors for Drug Delivery in Cancer. Curr Med Chem 2019; 26:6132-6148. [PMID: 30182846 DOI: 10.2174/0929867325666180831150259] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/20/2018] [Accepted: 07/04/2018] [Indexed: 02/07/2023]
Abstract
Cancer treatment still represents a formidable challenge, despite substantial advancements in available therapies being made over the past decade. One major issue is poor therapeutic efficacy due to lack of specificity and low bioavailability. The progress of nanotechnology and the development of a variety of nanoplatforms have had a significant impact in improving the therapeutic outcome of chemotherapeutics. Nanoparticles can overcome various biological barriers and localize at tumor site, while simultaneously protecting a therapeutic cargo and increasing its circulation time. Despite this, due to their synthetic origin, nanoparticles are often detected by the immune system and preferentially sequestered by filtering organs. Exosomes have recently been investigated as suitable substitutes for the shortcomings of nanoparticles due to their biological compatibility and particularly small size (i.e., 30-150 nm). In addition, exosomes have been found to play important roles in cell communication, acting as natural carriers of biological cargoes throughout the body. This review aims to highlight the use of exosomes as drug delivery vehicles for cancer and showcases the various attempts used to exploit exosomes with a focus on the delivery of chemotherapeutics and nucleic acids.
Collapse
Affiliation(s)
- Noemi Arrighetti
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Claudia Corbo
- Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, United States
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, United States
| | - Anna Pastò
- Istituto Oncologico Veneto-IRCCS, Padova, Italy
| | - Valentina Zuco
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, United States.,Houston Methodist Orthopedics & Sports Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, United States
| |
Collapse
|
13
|
Sukumaran A, Coish J, Yeung J, Muselius B, Gadjeva M, MacNeil A, Geddes-McAlister J. Decoding communication patterns of the innate immune system by quantitative proteomics. J Leukoc Biol 2019; 106:1221-1232. [PMID: 31556465 PMCID: PMC7309189 DOI: 10.1002/jlb.2ri0919-302r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 12/15/2022] Open
Abstract
The innate immune system is a collective network of cell types involved in cell recruitment and activation using a robust and refined communication system. Engagement of receptor-mediated intracellular signaling initiates communication cascades by conveying information about the host cell status to surrounding cells for surveillance and protection. Comprehensive profiling of innate immune cells is challenging due to low cell numbers, high dynamic range of the cellular proteome, low abundance of secreted proteins, and the release of degradative enzymes (e.g., proteases). However, recent advances in mass spectrometry-based proteomics provides the capability to overcome these limitations through profiling the dynamics of cellular processes, signaling cascades, post-translational modifications, and interaction networks. Moreover, integration of technologies and molecular datasets provide a holistic view of a complex and intricate network of communications underscoring host defense and tissue homeostasis mechanisms. In this Review, we explore the diverse applications of mass spectrometry-based proteomics in innate immunity to define communication patterns of the innate immune cells during health and disease. We also provide a technical overview of mass spectrometry-based proteomic workflows, with a focus on bottom-up approaches, and we present the emerging role of proteomics in immune-based drug discovery while providing a perspective on new applications in the future.
Collapse
Affiliation(s)
- A. Sukumaran
- Molecular and Cellular Biology Department, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - J.M. Coish
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1
| | - J. Yeung
- Molecular and Cellular Biology Department, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - B. Muselius
- Molecular and Cellular Biology Department, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - M. Gadjeva
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA 02115
| | - A.J. MacNeil
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1
| | - J. Geddes-McAlister
- Molecular and Cellular Biology Department, University of Guelph, Guelph, ON, Canada, N1G 2W1
| |
Collapse
|
14
|
Li A, Zhao J, Fu J, Cai J, Zhang P. Recent advances of biomimetic nano-systems in the diagnosis and treatment of tumor. Asian J Pharm Sci 2019; 16:161-174. [PMID: 33995611 PMCID: PMC8105416 DOI: 10.1016/j.ajps.2019.08.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/22/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022] Open
Abstract
The lack of effective methods of diagnosis and treatment presents a major barrier to combat against tumor. The biomimetic concept is an emerging field that expresses great application potential in tumor fighting. Strategy for combining nano-systems with biomimetic technology has gained increasing attention that is proved bioinspired, environmentally benign, and promising. Herein, we provide an up-to-date review of biomimetic nano-systems as well as their applications in tumor therapy. In addition, the challenges and future directions of biomimetic nano-systems to achieve clinical translation are also pointed out.
Collapse
Affiliation(s)
- Anning Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiawei Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingru Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jia Cai
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
15
|
Pratiwi FW, Kuo CW, Chen BC, Chen P. Recent advances in the use of fluorescent nanoparticles for bioimaging. Nanomedicine (Lond) 2019; 14:1759-1769. [DOI: 10.2217/nnm-2019-0105] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Rapid and recent progress in fluorescence microscopic techniques has allowed for routine discovery and viewing of biological structures and processes in unprecedented spatiotemporal resolution. In these imaging techniques, fluorescent nanoparticles (NPs) play important roles in the improvement of reporting systems. A short overview of recently developed fluorescent NPs used for advanced in vivo imaging will be discussed in this mini-review. The discussion begins with the contribution of fluorescence imaging in exploring the fate of NPs in biological systems. NP applications for in vivo imaging, including cell labeling, multimodal imaging and theranostic agents, are then discussed. Finally, despite all of the advancements in bioimaging, some unsolved challenges will be briefly discussed concerning future research directions.
Collapse
Affiliation(s)
| | - Chiung Wen Kuo
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Bi-Chang Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Peilin Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
16
|
Pasto A, Giordano F, Evangelopoulos M, Amadori A, Tasciotti E. Cell membrane protein functionalization of nanoparticles as a new tumor-targeting strategy. Clin Transl Med 2019; 8:8. [PMID: 30877412 PMCID: PMC6420595 DOI: 10.1186/s40169-019-0224-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
Nanoparticles have seen considerable popularity as effective tools for drug delivery. However, non-specific targeting continues to remain a challenge. Recently, biomimetic nanoparticles have emerged as an innovative solution that exploits biologically-derived components to improve therapeutic potential. Specifically, cell membrane proteins extracted from various cells (i.e., leukocytes, erythrocytes, platelets, mesenchymal stem cells, cancer) have shown considerable promise in bestowing nanoparticles with increased circulation and targeting efficacy. Traditional nanoparticles can be detected and removed by the immune system which significantly hinders their clinical success. Biomimicry has been proposed as a promising approach to overcome these limitations. In this review, we highlight the current trends in biomimetic nanoparticles and describe how they are being used to increase their chemotherapeutic effect in cancer treatment.
Collapse
Affiliation(s)
- Anna Pasto
- Veneto Institute of Oncology-IRCCS, Padua, Italy.,Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Federica Giordano
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Alberto Amadori
- Veneto Institute of Oncology-IRCCS, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA. .,Houston Methodist Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
17
|
Pasto A, Giordano F, Evangelopoulos M, Amadori A, Tasciotti E. Cell membrane protein functionalization of nanoparticles as a new tumor-targeting strategy. Clin Transl Med 2019. [PMID: 30877412 DOI: 10.1186/s40169019-0224-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Nanoparticles have seen considerable popularity as effective tools for drug delivery. However, non-specific targeting continues to remain a challenge. Recently, biomimetic nanoparticles have emerged as an innovative solution that exploits biologically-derived components to improve therapeutic potential. Specifically, cell membrane proteins extracted from various cells (i.e., leukocytes, erythrocytes, platelets, mesenchymal stem cells, cancer) have shown considerable promise in bestowing nanoparticles with increased circulation and targeting efficacy. Traditional nanoparticles can be detected and removed by the immune system which significantly hinders their clinical success. Biomimicry has been proposed as a promising approach to overcome these limitations. In this review, we highlight the current trends in biomimetic nanoparticles and describe how they are being used to increase their chemotherapeutic effect in cancer treatment.
Collapse
Affiliation(s)
- Anna Pasto
- Veneto Institute of Oncology-IRCCS, Padua, Italy
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Federica Giordano
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Alberto Amadori
- Veneto Institute of Oncology-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.
- Houston Methodist Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
18
|
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: 1.8] [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
|
19
|
Molinaro R, Evangelopoulos M, Hoffman JR, Corbo C, Taraballi F, Martinez JO, Hartman KA, Cosco D, Costa G, Romeo I, Sherman M, Paolino D, Alcaro S, Tasciotti E. Design and Development of Biomimetic Nanovesicles Using a Microfluidic Approach. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1702749. [PMID: 29512198 DOI: 10.1002/adma.201702749] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 11/27/2017] [Indexed: 05/17/2023]
Abstract
The advancement of nanotechnology toward more sophisticated bioinspired approaches has highlighted the gap between the advantages of biomimetic and biohybrid platforms and the availability of manufacturing processes to scale up their production. Though the advantages of transferring biological features from cells to synthetic nanoparticles for drug delivery purposes have recently been reported, a standardizable, batch-to-batch consistent, scalable, and high-throughput assembly method is required to further develop these platforms. Microfluidics has offered a robust tool for the controlled synthesis of nanoparticles in a versatile and reproducible approach. In this study, the incorporation of membrane proteins within the bilayer of biomimetic nanovesicles (leukosomes) using a microfluidic-based platform is demonstrated. The physical, pharmaceutical, and biological properties of microfluidic-formulated leukosomes (called NA-Leuko) are characterized. NA-Leuko show extended shelf life and retention of the biological functions of donor cells (i.e., macrophage avoidance and targeting of inflamed vasculature). The NA approach represents a universal, versatile, robust, and scalable tool, which is extensively used for the assembly of lipid nanoparticles and adapted here for the manufacturing of biomimetic nanovesicles.
Collapse
Affiliation(s)
- Roberto Molinaro
- Center of Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
- Nanoinspired Biomedicine Lab, Fondazione Istituto di Ricerca, Pediatrica Città della Speranza, 35127, Padua, Italy
| | - Michael Evangelopoulos
- Center of Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
| | - Jessica R Hoffman
- Center of Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
| | - Claudia Corbo
- Center of Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
| | - Francesca Taraballi
- Center of Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
| | - Jonathan O Martinez
- Center of Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
| | - Kelly A Hartman
- Center of Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
| | - Donato Cosco
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Campus Universitario "S. Venuta,", Viale S. Venuta, Germaneto, I-88100, Catanzaro, Italy
| | - Giosue' Costa
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Campus Universitario "S. Venuta,", Viale S. Venuta, Germaneto, I-88100, Catanzaro, Italy
| | - Isabella Romeo
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Campus Universitario "S. Venuta,", Viale S. Venuta, Germaneto, I-88100, Catanzaro, Italy
| | - Michael Sherman
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Campus Universitario "S. Venuta,", Viale S. Venuta, Germaneto, I-88100, Catanzaro, Italy
| | - Stefano Alcaro
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Campus Universitario "S. Venuta,", Viale S. Venuta, Germaneto, I-88100, Catanzaro, Italy
| | - Ennio Tasciotti
- Center of Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
- Houston Methodist Orthopedic and Sports Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| |
Collapse
|
20
|
Molinaro R, Corbo C, Livingston M, Evangelopoulos M, Parodi A, Boada C, Agostini M, Tasciotti E. Inflammation and Cancer: In Medio Stat Nano. Curr Med Chem 2018; 25:4208-4223. [PMID: 28933296 PMCID: PMC5860929 DOI: 10.2174/0929867324666170920160030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 06/06/2017] [Accepted: 07/02/2017] [Indexed: 12/21/2022]
Abstract
Cancer treatment still remains a challenge due to the several limitations of currently used chemotherapeutics, such as their poor pharmacokinetics, unfavorable chemical properties, as well as inability to discriminate between healthy and diseased tissue. Nanotechnology offered potent tools to overcome these limitations. Drug encapsulation within a delivery system permitted i) to protect the payload from enzymatic degradation/ inactivation in the blood stream, ii) to improve the physicochemical properties of poorly water-soluble drugs, like paclitaxel, and iii) to selectively deliver chemotherapeutics to the cancer lesions, thus reducing the off-target toxicity, and promoting the intracellular internalization. To accomplish this purpose, several strategies have been developed, based on biological and physical changes happening locally and systemically as a consequence of tumorigenesis. Here, we will discuss the role of inflammation in the different steps of tumor development and the strategies based on the use of nanoparticles that exploit the inflammatory pathways in order to selectively target the tumor-associated microenvironment for therapeutic and diagnostic purposes.
Collapse
Affiliation(s)
- Roberto Molinaro
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, United States
| | - Claudia Corbo
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, United States
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, United States
| | - Megan Livingston
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, United States
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, United States
| | - Alessandro Parodi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, United States
| | - Christian Boada
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, United States
- Centro de Biotecnología FEMSA, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey, Nuevo León, 64710, Mexico
| | - Marco Agostini
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, 35124, Italy
- Nanoinspired Biomedicine Laboratory, Institute of Pediatric Research, Fondazione Citta della Speranza, 35129, Padua, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, United States
- Houston Methodist Orthopedics & Sports Medicine, Houston Methodist Hospital, Houston, TX, 77030, United States
| |
Collapse
|
21
|
Shen H, Sun T, Hoang HH, Burchfield JS, Hamilton GF, Mittendorf EA, Ferrari M. Enhancing cancer immunotherapy through nanotechnology-mediated tumor infiltration and activation of immune cells. Semin Immunol 2017; 34:114-122. [PMID: 28947107 PMCID: PMC5705528 DOI: 10.1016/j.smim.2017.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/10/2017] [Accepted: 09/11/2017] [Indexed: 12/11/2022]
Abstract
Cancer immunotherapy has become arguably the most promising advancement in cancer research and therapy in recent years. The efficacy of cancer immunotherapy is critically dependent on specific physiological and physical processes - collectively referred to as transport barriers - including the activation of T cells by antigen presenting cells, T cells migration to and penetration into the tumor microenvironment, and movement of nutrients and other immune cells through the tumor microenvironment. Nanotechnology-based approaches have great potential to help overcome these transport barriers. In this review, we discuss the ways that nanotechnology is being leveraged to improve the efficacy and potency of various cancer immunotherapies.
Collapse
Affiliation(s)
- Haifa Shen
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Tong Sun
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Hanh H Hoang
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Jana S Burchfield
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Gillian F Hamilton
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Elizabeth A Mittendorf
- Department of Breast Surgical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mauro Ferrari
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
22
|
Corbo C, Cromer WE, Molinaro R, Toledano Furman NE, Hartman KA, De Rosa E, Boada C, Wang X, Zawieja DC, Agostini M, Salvatore F, Abraham BP, Tasciotti E. Engineered biomimetic nanovesicles show intrinsic anti-inflammatory properties for the treatment of inflammatory bowel diseases. NANOSCALE 2017; 9:14581-14591. [PMID: 28932838 DOI: 10.1039/c7nr04734g] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Inflammatory bowel disease (IBD), which includes Crohn's disease and ulcerative colitis, is a chronic inflammatory condition of the gastrointestinal (GI) tract. Currently, it is treated with immunosuppressant or biologics that often induce severe adverse effects. Thus, there is an urgent clinical need for more specific treatments. To provide a valid therapeutic tool for IBD therapy, in this work we developed biomimetic nanovesicles by manipulating leukocyte membranes to exploit mechanisms of T-cell recruitment during inflammation. A subset of T-lymphocytes participates in homing to inflamed tissue in the gastrointestinal tract by overexpressing the α4β7 integrin, which is responsible for binding to its receptor on the endothelial membrane, the mucosal addressin cell adhesion molecule 1. Based on this principle, we engineered biomimetic vesicles, referred to as specialized leukosomes (SLKs), which are leukocyte-like carriers 'doped' with the α4β7 integrin over-induced in purified immune cells. We tested SLKs in an in vivo murine model of IBD induced by treatment with dextran sulfate sodium. Notably, treatment of IBD mice with SLKs allowed us to observe a reduction of inflammation (favorable modulation of both pro- and anti-inflammatory genes, as well as reduction of immune cells infiltration into the colon tissue), and a consequent enhanced intestinal repair (low epithelial damage). In this study, we demonstrate that biological-derived nanoparticles can be used not only as naturally targeted drug delivery systems, but also as nano-therapeutics endowed with intrinsic anti-inflammatory properties.
Collapse
Affiliation(s)
- Claudia Corbo
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77002, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Parodi A, Molinaro R, Sushnitha M, Evangelopoulos M, Martinez JO, Arrighetti N, Corbo C, Tasciotti E. Bio-inspired engineering of cell- and virus-like nanoparticles for drug delivery. Biomaterials 2017; 147:155-168. [PMID: 28946131 DOI: 10.1016/j.biomaterials.2017.09.020] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/13/2017] [Accepted: 09/17/2017] [Indexed: 12/25/2022]
Abstract
The engineering of future generations of nanodelivery systems aims at the creation of multifunctional vectors endowed with improved circulation, enhanced targeting and responsiveness to the biological environment. Moving past purely bio-inert systems, researchers have begun to create nanoparticles capable of proactively interacting with the biology of the body. Nature offers a wide-range of sources of inspiration for the synthesis of more effective drug delivery platforms. Because the nano-bio-interface is the key driver of nanoparticle behavior and function, the modification of nanoparticles' surfaces allows the transfer of biological properties to synthetic carriers by imparting them with a biological identity. Modulation of these surface characteristics governs nanoparticle interactions with the biological barriers they encounter. Building off these observations, we provide here an overview of virus- and cell-derived biomimetic delivery systems that combine the intrinsic hallmarks of biological membranes with the delivery capabilities of synthetic carriers. We describe the features and properties of biomimetic delivery systems, recapitulating the distinctive traits and functions of viruses, exosomes, platelets, red and white blood cells. By mimicking these biological entities, we will learn how to more efficiently interact with the human body and refine our ability to negotiate with the biological barriers that impair the therapeutic efficacy of nanoparticles.
Collapse
Affiliation(s)
- Alessandro Parodi
- Department of Pharmacology, University of Illinois, Chicago College of Medicine, Chicago, IL, USA
| | - Roberto Molinaro
- Department of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Manuela Sushnitha
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX, USA; Department of Bioengineering, Rice University, Houston, TX, USA
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX, USA
| | - Jonathan O Martinez
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX, USA
| | - Noemi Arrighetti
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX, USA; Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale per Lo Studio e La Cura Dei Tumori, Milan, Italy
| | - Claudia Corbo
- Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, MA, USA
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX, USA; Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
24
|
Abstract
Transplantation is often the only choice many patients have when suffering from end-stage organ failure. Although the quality of life improves after transplantation, challenges, such as organ shortages, necessary immunosuppression with associated complications, and chronic graft rejection, limit its wide clinical application. Nanotechnology has emerged in the past 2 decades as a field with the potential to satisfy clinical needs in the area of targeted and sustained drug delivery, noninvasive imaging, and tissue engineering. In this article, we provide an overview of popular nanotechnologies and a summary of the current and potential uses of nanotechnology in cell and organ transplantation.
Collapse
|
25
|
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: 4.7] [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
|
26
|
Corbo C, Molinaro R, Taraballi F, Toledano Furman NE, Hartman KA, Sherman MB, De Rosa E, Kirui DK, Salvatore F, Tasciotti E. Unveiling the in Vivo Protein Corona of Circulating Leukocyte-like Carriers. ACS NANO 2017; 11:3262-3273. [PMID: 28264157 DOI: 10.1021/acsnano.7b00376] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Understanding interactions occurring at the interface between nanoparticles and biological components is an urgent challenge in nanomedicine due to their effect on the biological fate of nanoparticles. After the systemic injection of nanoparticles, a protein corona constructed by blood components surrounds the carrier's surface and modulates its pharmacokinetics and biodistribution. Biomimicry-based approaches in nanotechnology attempt to imitate what happens in nature in order to transfer specific natural functionalities to synthetic nanoparticles. Several biomimetic formulations have been developed, showing superior in vivo features as a result of their cell-like identity. We have recently designed biomimetic liposomes, called leukosomes, which recapitulate the ability of leukocytes to target inflamed endothelium and escape clearance by the immune system. To gain insight into the properties of leukosomes, we decided to investigate their protein corona in vivo. So far, most information about the protein corona has been obtained using in vitro experiments, which have been shown to minimally reproduce in vivo phenomena. Here we directly show a time-dependent quantitative and qualitative analysis of the protein corona adsorbed in vivo on leukosomes and control liposomes. We observed that leukosomes absorb fewer proteins than liposomes, and we identified a group of proteins specifically adsorbed on leukosomes. Moreover, we hypothesize that the presence of macrophage receptors on leukosomes' surface neutralizes their protein corona-meditated uptake by immune cells. This work unveils the protein corona of a biomimetic carrier and is one of the few studies on the corona performed in vivo.
Collapse
Affiliation(s)
- Claudia Corbo
- CEINGE-Biotecnologie Avanzate s.c.a r.l. , Via G. Salvatore 486, Naples, 80145, Italy
| | | | | | | | | | - Michael B Sherman
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch , Galveston, Texas 77555, United States
| | | | | | - Francesco Salvatore
- CEINGE-Biotecnologie Avanzate s.c.a r.l. , Via G. Salvatore 486, Naples, 80145, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II , Via Sergio Pansini 5, Naples, 80131, Italy
| | - Ennio Tasciotti
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital , 6565 Fannin Street, Houston, Texas 77030, United States
| |
Collapse
|
27
|
Venuta A, Wolfram J, Shen H, Ferrari M. Post-nano strategies for drug delivery: Multistage porous silicon microvectors. J Mater Chem B 2016; 5:207-219. [PMID: 28670454 DOI: 10.1039/c6tb01978a] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanodelivery systems usually improve the biodistribution of drugs, leading to reduced side effects and enhanced therapeutic efficacy. However, only a small portion of the injected nanoparticle dose accumulates in pathological tissue. Challenges in drug delivery arise due to a multitude of transport obstacles in the body, including the endothelium, the extracellular matrix, and the cell membrane. In general, nanoparticles are designed to overcome only a few biological barriers, making them inadequate for localized drug delivery. Accordingly, a multifunctional and multicomponent systems are required to effectively address a wide variety of transport obstacles. A suitable approach to obtain high levels of multifunctionality is to bring together the nanoscale with the microscale, resulting in post-nano strategies for drug delivery. This review discusses several such post-nano approaches, with an emphasis on the multistage vector (MSV) platform. The MSV consists of three components on different spatial scales, each intended to address biological barriers that exist in a specific compartment in the body. The first stage vector is a microparticle that is designed to navigate in the vascular compartment. The second stage vector consists of nanoparticles that are released from the microparticle into the tissue interstitium, where they address biological barriers in extracellular and intracellular compartments. The final component of the system is a small molecule therapeutic agent. A new generation of microparticle-based strategies with expanded applications has recently been developed, including injectable nanoparticle generators and silicon particles for immunotherapy. Notably, the advantage of incorporating microstructures in drug delivery vehicles is apparent from the observation that superior functionality only appears on the microscale, highlighting the inherent functional limitations of nanostructures.
Collapse
Affiliation(s)
- Alessandro Venuta
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.,Department of Pharmacy, University of Naples Federico II, Naples 80131, Italy
| | - 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 Medicine, New York, NY 10065, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.,Department of Medicine, Weill Cornell Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
28
|
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: 10.6] [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
|
29
|
Molinaro R, Corbo C, Martinez JO, Taraballi F, Evangelopoulos M, Minardi S, Yazdi I, Zhao P, De Rosa E, Sherman M, De Vita A, Furman NT, Wang X, Parodi A, Tasciotti E. Biomimetic proteolipid vesicles for targeting inflamed tissues. NATURE MATERIALS 2016; 15:1037-46. [PMID: 27213956 PMCID: PMC5127392 DOI: 10.1038/nmat4644] [Citation(s) in RCA: 301] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 04/13/2016] [Indexed: 05/13/2023]
Abstract
A multitude of micro- and nanoparticles have been developed to improve the delivery of systemically administered pharmaceuticals, which are subject to a number of biological barriers that limit their optimal biodistribution. Bioinspired drug-delivery carriers formulated by bottom-up or top-down strategies have emerged as an alternative approach to evade the mononuclear phagocytic system and facilitate transport across the endothelial vessel wall. Here, we describe a method that leverages the advantages of bottom-up and top-down strategies to incorporate proteins derived from the leukocyte plasma membrane into lipid nanoparticles. The resulting proteolipid vesicles-which we refer to as leukosomes-retained the versatility and physicochemical properties typical of liposomal formulations, preferentially targeted inflamed vasculature, enabled the selective and effective delivery of dexamethasone to inflamed tissues, and reduced phlogosis in a localized model of inflammation.
Collapse
Affiliation(s)
- R. Molinaro
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - C. Corbo
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- EINGE–Biotecnologie Avanzate s.c.a.r.l., Via G. Salvatore 486, 80145 Naples, Italy
- IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
| | - J. O. Martinez
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - F. Taraballi
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- Pain Therapy Service, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - M. Evangelopoulos
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - S. Minardi
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - I.K. Yazdi
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - P. Zhao
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - E. De Rosa
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - M. Sherman
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555
| | - A. De Vita
- Osteoncology and Rare Tumors Center, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Italy
| | - N.E. Toledano Furman
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - X. Wang
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - A. Parodi
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
| | - E. Tasciotti
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- To whom correspondence should be addressed: Dr. Ennio Tasciotti, Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030,
| |
Collapse
|
30
|
Corbo C, Molinaro R, Taraballi F, Toledano Furman NE, Sherman MB, Parodi A, Salvatore F, Tasciotti E. Effects of the protein corona on liposome-liposome and liposome-cell interactions. Int J Nanomedicine 2016; 11:3049-63. [PMID: 27445473 PMCID: PMC4938145 DOI: 10.2147/ijn.s109059] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A thorough understanding of interactions occurring at the interface between nanocarriers and biological systems is crucial to predict and interpret their biodistribution, targeting, and efficacy, and thus design more effective drug delivery systems. Upon intravenous injection, nanoparticles are coated by a protein corona (PC). This confers a new biological identity on the particles that largely determines their biological fate. Liposomes have great pharmaceutical versatility, so, as proof of concept, their PC has recently been implicated in the mechanism and efficiency of their internalization into the cell. In an attempt to better understand the interactions between nanocarriers and biological systems, we analyzed the plasma proteins adsorbed on the surface of multicomponent liposomes. Specifically, we analyzed the physical properties and ultrastructure of liposome/PC complexes and the aggregation process that occurs when liposomes are dispersed in plasma. The results of combined confocal microscopy and flow cytometry experiments demonstrated that the PC favors liposome internalization by both macrophages and tumor cells. This work provides insights into the effects of the PC on liposomes' physical properties and, consequently, liposome-liposome and liposome-cell interactions.
Collapse
Affiliation(s)
- Claudia Corbo
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA
- CEINGE-Biotecnologie Avanzate s.c.a r.l., Naples, Italy
| | - Roberto Molinaro
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Francesca Taraballi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | | | - Michael B Sherman
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA
| | - Alessandro Parodi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Francesco Salvatore
- CEINGE-Biotecnologie Avanzate s.c.a r.l., Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Department of Orthopedics, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
31
|
Corbo C, Molinaro R, Parodi A, Toledano Furman NE, Salvatore F, Tasciotti E. The impact of nanoparticle protein corona on cytotoxicity, immunotoxicity and target drug delivery. Nanomedicine (Lond) 2016; 11:81-100. [PMID: 26653875 PMCID: PMC4910943 DOI: 10.2217/nnm.15.188] [Citation(s) in RCA: 423] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/29/2015] [Indexed: 12/17/2022] Open
Abstract
In a perfect sequence of events, nanoparticles (NPs) are injected into the bloodstream where they circulate until they reach the target tissue. The ligand on the NP surface recognizes its specific receptor expressed on the target tissue and the drug is released in a controlled manner. However, once injected in a physiological environment, NPs interact with biological components and are surrounded by a protein corona (PC). This can trigger an immune response and affect NP toxicity and targeting capabilities. In this review, we provide a survey of recent findings on the NP-PC interactions and discuss how the PC can be used to modulate both cytotoxicity and the immune response as well as to improve the efficacy of targeted delivery of nanocarriers.
Collapse
Affiliation(s)
- Claudia Corbo
- Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, 77030 Houston, TX, USA
- Fondazione SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Roberto Molinaro
- Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, 77030 Houston, TX, USA
| | - Alessandro Parodi
- Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, 77030 Houston, TX, USA
- Fondazione SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Naama E Toledano Furman
- Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, 77030 Houston, TX, USA
| | - Francesco Salvatore
- CEINGE, Advanced Biotechnology s.c.a.r.l., Via G. Salvatore 486, 80145 Naples, Italy
| | - Ennio Tasciotti
- Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, 77030 Houston, TX, USA
| |
Collapse
|
32
|
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: 4.8] [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
|
33
|
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: 5.2] [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
|
34
|
Parodi A, Corbo C, Cevenini A, Molinaro R, Palomba R, Pandolfi L, Agostini M, Salvatore F, Tasciotti E. Enabling cytoplasmic delivery and organelle targeting by surface modification of nanocarriers. Nanomedicine (Lond) 2015; 10:1923-40. [PMID: 26139126 PMCID: PMC5561781 DOI: 10.2217/nnm.15.39] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nanocarriers are designed to specifically accumulate in diseased tissues. In this context, targeting of intracellular compartments was shown to enhance the efficacy of many drugs and to offer new and more effective therapeutic approaches. This is especially true for therapies based on biologicals that must be encapsulated to favor cell internalization, and to avoid intracellular endosomal sequestration and degradation of the payload. In this review, we discuss specific surface modifications designed to achieve cell cytoplasm delivery and to improve targeting of major organelles; we also discuss the therapeutic applications of these approaches. Last, we describe some integrated strategies designed to sequentially overcome the biological barriers that separate the site of administration from the cell cytoplasm, which is the drug's site of action.
Collapse
Affiliation(s)
- Alessandro Parodi
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
- Fondazione IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Claudia Corbo
- Fondazione IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Armando Cevenini
- Department of Molecular Medicine & Medical Biotechnology, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
- CEINGE, Biotecnologie Avanzate s.c.a.r.l., Via G. Salvatore 486, 80145 Naples, Italy
| | - Roberto Molinaro
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
- Clinica Chirurgica I, Dipartimento di Scienze Chirurgiche Oncologiche e Gastroeterologiche, Università di Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Roberto Palomba
- Fondazione IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Laura Pandolfi
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
- College of Materials Science & Optoelectronic Technology, University of Chinese Academy of Science, 19A Yuquanlu, Beijing, China
| | - Marco Agostini
- Clinica Chirurgica I, Dipartimento di Scienze Chirurgiche Oncologiche e Gastroeterologiche, Università di Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Francesco Salvatore
- Fondazione IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
- CEINGE, Biotecnologie Avanzate s.c.a.r.l., Via G. Salvatore 486, 80145 Naples, Italy
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| |
Collapse
|