1
|
Gomez-Garcia MJ, Abdelkarim M, Cramb DT, Childs SJ, Rinker KD, Labouta HI. Blood vessel wall shear stress determines regions of liposome accumulation in angiogenic vasculature. Drug Deliv Transl Res 2024; 14:3608-3620. [PMID: 39042244 DOI: 10.1007/s13346-024-01671-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Nanoparticles used for drug delivery often require intravenous administration exposing them to fluid forces within the vasculature, yet the impact of blood flow on nanoparticle delivery remains incompletely understood. Here, we utilized transgenic zebrafish embryos to investigate the relationship between the accumulation of fluorescently labeled PEGylated liposomes and various hemodynamic factors (such as flow velocity, wall shear stress (WSS), and flow pattern) across a wide range of angiogenic blood vessels. We reconstructed 3D models of vascular structures from confocal images and used computational fluid dynamics to calculate local WSS, velocities, and define flow patterns. The spatial distribution of fluorescently labeled liposomes was subsequently mapped within the same 3D space and correlated with local hemodynamic parameters. Through the integration of computational fluid dynamics and in vivo experimentation, we show that liposomes accumulated in vessel regions with WSS between 0.1-0.8 Pa, displaying an inverse linear correlation (R2 > 0.85) between time-averaged wall shear stress and liposome localization in vivo. Interestingly, flow pattern did not appear to impact liposome accumulation. Collectively, our findings suggest the potential of stealth liposomes for passive targeting of low-flow vasculature, including capillaries and intricate angiogenic vasculature resembling that of tumor vessel networks.
Collapse
Affiliation(s)
- M Juliana Gomez-Garcia
- Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto, ON, Canada
- Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Mahmoud Abdelkarim
- Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
| | - David T Cramb
- Department of Chemistry and Biology, Faculty of Science, Toronto Metropolitan University, Toronto, ON, Canada
| | - Sarah J Childs
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kristina D Rinker
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada.
| | - Hagar I Labouta
- Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto, ON, Canada.
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Reza Sayah M, Ebrahimi S, Mirafzal I, Shamloo A. Investigation of the size and shape of nano-microcarriers for targeted drug delivery to atherosclerotic plaque in ischemic stroke prevention. Int J Pharm 2024; 662:124469. [PMID: 39004292 DOI: 10.1016/j.ijpharm.2024.124469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Recognizing the significance of drug carriers in the treatment of atherosclerotic plaque is crucial in light of the worldwide repercussions of ischemic stroke. Conservative methodologies, specifically targeted drug delivery, present encouraging substitutes that mitigate the hazards linked to invasive procedures. With the intention of illuminating their considerable significance and prospective benefits, this study examines the impact of the geometry and dimensions of drug-loaded nano-microcarriers on atherosclerotic plaque. The research utilizes a finite element approach to simulate the motion and fluid dynamics of nano-microcarriers loaded with drugs within the carotid arteries. Carriers are available in a variety of shapes and sizes to accommodate patient-specific geometries, pulsatile fluid flow, and non-Newtonian blood properties. Optimization of drug delivery is achieved through the examination of carrier interaction with the inner wall. The results demonstrated that the interaction data between particles and the inner wall of atherosclerotic plaques exhibits micro- and nanoscale patterns that are distinct. Symmetric plaques demonstrate that nanoparticles with a 0.4 shape factor and diameters below 200 nm show the highest interaction rate. Conversely, larger particles (200 and 500 nm) with shape factors of 1 demonstrate comparatively elevated interaction rates. The optimal shape factor for drug-loaded microparticles has been determined to be one, and the number of interactions increases as the diameter of the nanoparticles increases, with a significant increase observed at a shape factor of one. Asymmetric plaques exhibit the maximum interaction rates among particles that have a shape factor of 0.4 and have diameters smaller than 500 µm. The findings establish a foundation for novel therapeutic strategies, establishing nano-microparticles as auspicious contenders for accurate and efficacious drug delivery systems that inhibit plaque proliferation.
Collapse
Affiliation(s)
- Mohammad Reza Sayah
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Sina Ebrahimi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Iman Mirafzal
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Amir Shamloo
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
3
|
Elblová P, Lunova M, Dejneka A, Jirsa M, Lunov O. Impact of mechanical cues on key cell functions and cell-nanoparticle interactions. DISCOVER NANO 2024; 19:106. [PMID: 38907808 PMCID: PMC11193707 DOI: 10.1186/s11671-024-04052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
In recent years, it has been recognized that mechanical forces play an important regulative role in living organisms and possess a direct impact on crucial cell functions, ranging from cell growth to maintenance of tissue homeostasis. Advancements in mechanobiology have revealed the profound impact of mechanical signals on diverse cellular responses that are cell type specific. Notably, numerous studies have elucidated the pivotal role of different mechanical cues as regulatory factors influencing various cellular processes, including cell spreading, locomotion, differentiation, and proliferation. Given these insights, it is unsurprising that the responses of cells regulated by physical forces are intricately linked to the modulation of nanoparticle uptake kinetics and processing. This complex interplay underscores the significance of understanding the mechanical microenvironment in shaping cellular behaviors and, consequently, influencing how cells interact with and process nanoparticles. Nevertheless, our knowledge on how localized physical forces affect the internalization and processing of nanoparticles by cells remains rather limited. A significant gap exists in the literature concerning a systematic analysis of how mechanical cues might bias the interactions between nanoparticles and cells. Hence, our aim in this review is to provide a comprehensive and critical analysis of the existing knowledge regarding the influence of mechanical cues on the complicated dynamics of cell-nanoparticle interactions. By addressing this gap, we would like to contribute to a detailed understanding of the role that mechanical forces play in shaping the complex interplay between cells and nanoparticles.
Collapse
Affiliation(s)
- Petra Elblová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16, Prague 2, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic.
| |
Collapse
|
4
|
Gurrola TE, Effah SN, Sariyer IK, Dampier W, Nonnemacher MR, Wigdahl B. Delivering CRISPR to the HIV-1 reservoirs. Front Microbiol 2024; 15:1393974. [PMID: 38812680 PMCID: PMC11133543 DOI: 10.3389/fmicb.2024.1393974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/22/2024] [Indexed: 05/31/2024] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection is well known as one of the most complex and difficult viral infections to cure. The difficulty in developing curative strategies arises in large part from the development of latent viral reservoirs (LVRs) within anatomical and cellular compartments of a host. The clustered regularly interspaced short palindromic repeats/ CRISPR-associated protein 9 (CRISPR/Cas9) system shows remarkable potential for the inactivation and/or elimination of integrated proviral DNA within host cells, however, delivery of the CRISPR/Cas9 system to infected cells is still a challenge. In this review, the main factors impacting delivery, the challenges for delivery to each of the LVRs, and the current successes for delivery to each reservoir will be discussed.
Collapse
Affiliation(s)
- Theodore E. Gurrola
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Samuel N. Effah
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ilker K. Sariyer
- Department of Microbiology, Immunology, and Inflammation and Center for Neurovirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Will Dampier
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Michael R. Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
5
|
Chan WJ, Li H. Recent advances in nano/micro systems for improved circulation stability, enhanced tumor targeting, penetration, and intracellular drug delivery: a review. Biomed Phys Eng Express 2024; 10:022001. [PMID: 38086099 DOI: 10.1088/2057-1976/ad14f0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 12/12/2023] [Indexed: 01/17/2024]
Abstract
In recent years, nanoparticles (NPs) have been extensively developed as drug carriers to overcome the limitations of cancer therapeutics. However, there are several biological barriers to nanomedicines, which include the lack of stability in circulation, limited target specificity, low penetration into tumors and insufficient cellular uptake, restricting the active targeting toward tumors of nanomedicines. To address these challenges, a variety of promising strategies were developed recently, as they can be designed to improve NP accumulation and penetration in tumor tissues, circulation stability, tumor targeting, and intracellular uptake. In this Review, we summarized nanomaterials developed in recent three years that could be utilized to improve drug delivery for cancer treatments.
Collapse
Affiliation(s)
- Wei-Jen Chan
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| | - Huatian Li
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| |
Collapse
|
6
|
Abstract
Primary brain cancer or brain cancer is the overgrowth of abnormal or malignant cells in the brain or its nearby tissues that form unwanted masses called brain tumors. People with malignant brain tumors suffer a lot, and the expected life span of the patients after diagnosis is often only around 14 months, even with the most vigorous therapies. The blood-brain barrier (BBB) is the main barrier in the body that restricts the entry of potential chemotherapeutic agents into the brain. The chances of treatment failure or low therapeutic effects are some significant drawbacks of conventional treatment methods. However, recent advancements in nanotechnology have generated hope in cancer treatment. Nanotechnology has shown a vital role starting from the early detection, diagnosis, and treatment of cancer. These tiny nanomaterials have great potential to deliver drugs across the BBB. Beyond just drug delivery, nanomaterials can be simulated to generate fluorescence to detect tumors. The current Review discusses in detail the challenges of brain cancer treatment and the application of nanotechnology to overcome those challenges. The success of chemotherapeutic treatment or the surgical removal of tumors requires proper imaging. Nanomaterials can provide imaging and therapeutic benefits for cancer. The application of nanomaterials in the diagnosis and treatment of brain cancer is discussed in detail by reviewing past studies.
Collapse
Affiliation(s)
- Yogita Ale
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Prem Nagar, Dehradun, Uttarakhand 248007, India
| | - Nidhi Nainwal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Prem Nagar, Dehradun, Uttarakhand 248007, India
| |
Collapse
|
7
|
Sorrin AJ, Zhou K, May K, Liu C, McNaughton K, Rahman I, Liang BJ, Rizvi I, Roque DM, Huang HC. Transient fluid flow improves photoimmunoconjugate delivery and photoimmunotherapy efficacy. iScience 2023; 26:107221. [PMID: 37520715 PMCID: PMC10372742 DOI: 10.1016/j.isci.2023.107221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/01/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023] Open
Abstract
Circulating drugs in the peritoneal cavity is an effective strategy for advanced ovarian cancer treatment. Photoimmunotherapy, an emerging modality with potential for the treatment of ovarian cancer, involves near-infrared light activation of antibody-photosensitizer conjugates (photoimmunoconjugates) to generate cytotoxic reactive oxygen species. Here, a microfluidic cell culture model is used to study how fluid flow-induced shear stress affects photoimmunoconjugate delivery to ovarian cancer cells. Photoimmunoconjugates are composed of the antibody, cetuximab, conjugated to the photosensitizer, and benzoporphyrin derivative. Longitudinal tracking of photoimmunoconjugate treatment under flow conditions reveals enhancements in subcellular photosensitizer accumulation. Compared to static conditions, fluid flow-induced shear stress at 0.5 and 1 dyn/cm2 doubled the cellular delivery of photoimmunoconjugates. Fluid flow-mediated treatment with three different photosensitizer formulations (benzoporphyrin derivative, photoimmunoconjugates, and photoimmunoconjugate-coated liposomes) led to enhanced phototoxicity compared to static conditions. This study confirms the fundamental role of fluid flow-induced shear stress in the anti-cancer effects of photoimmunotherapy.
Collapse
Affiliation(s)
- Aaron J. Sorrin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Keri Zhou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Katherine May
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Cindy Liu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Kathryn McNaughton
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Idrisa Rahman
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Barry J. Liang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, North Carolina State University, Raleigh, NC 27599, USA
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dana M. Roque
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
8
|
Karaz S, Senses E. Liposomes Under Shear: Structure, Dynamics, and Drug Delivery Applications. ADVANCED NANOBIOMED RESEARCH 2023. [DOI: 10.1002/anbr.202200101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Selcan Karaz
- Department of Chemical and Biological Engineering Koç University Istanbul 34450 Turkey
| | - Erkan Senses
- Department of Chemical and Biological Engineering Koç University Istanbul 34450 Turkey
| |
Collapse
|
9
|
Precise Design Strategies of Nanotechnologies for Controlled Drug Delivery. J Funct Biomater 2022; 13:jfb13040188. [PMID: 36278656 PMCID: PMC9590086 DOI: 10.3390/jfb13040188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/03/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
Rapid advances in nanotechnologies are driving the revolution in controlled drug delivery. However, heterogeneous barriers, such as blood circulation and cellular barriers, prevent the drug from reaching the cellular target in complex physiologic environments. In this review, we discuss the precise design of nanotechnologies to enhance the efficacy, quality, and durability of drug delivery. For drug delivery in vivo, drugs loaded in nanoplatforms target particular sites in a spatial- and temporal-dependent manner. Advances in stimuli-responsive nanoparticles and carbon-based drug delivery platforms are summarized. For transdermal drug delivery systems, specific strategies including microneedles and hydrogel lead to a sustained release efficacy. Moreover, we highlight the current limitations of clinical translation and an incentive for the future development of nanotechnology-based drug delivery.
Collapse
|
10
|
Mittal D, Ali SA. Use of Nanomaterials for Diagnosis and Treatment: The Advancement of Next-Generation Antiviral Therapy. Microb Drug Resist 2022; 28:670-697. [PMID: 35696335 DOI: 10.1089/mdr.2021.0281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Globally, viral illness propagation is the leading cause of morbidity and death, causing wreaking havoc on socioeconomic development and health care systems. The rise of infected individuals has outpaced the existing critical care facilities. Early and sophisticated methods are desperately required in this respect to halt the spread of the infection. Therefore, early detection of infectious agents and an early treatment approach may help minimize viral outbreaks. Conventional point-of-care diagnostic techniques such as computed tomography scan, quantitative real time polymerase chain reaction (qRT-PCR), X-ray, and immunoassay are still deemed valuable. However, the labor demanding, low sensitivity, and complex infrastructure needed for these methods preclude their use in distant areas. Nanotechnology has emerged as a potentially transformative technology due to its promise as an effective theranostic platform for diagnosing and treating viral infection, circumventing the limits of traditional techniques. Their unique physical and chemical characteristics make nanoparticles (NPs) advantageous for drug delivery platforms due to their size, encapsulation efficiency, improved bioavailability, effectiveness, immunogenicity, and antiviral response. This study discusses the recent research on nanotechnology-based treatments designed to combat new viruses.
Collapse
Affiliation(s)
- Deepti Mittal
- Nanosafety Lab, Division of Biochemistry, ICAR-NDRI, Karnal, Haryana, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, Karnal, Haryana, India
| |
Collapse
|
11
|
Jansman MMT, Coll-Satue C, Liu X, Kempen PJ, Andresen TL, Thulstrup PW, Hosta-Rigau L. Hemoglobin-based oxygen carriers camouflaged with membranes extracted from red blood cells: Optimization and assessment of functionality. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 134:112691. [DOI: 10.1016/j.msec.2022.112691] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/13/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022]
|
12
|
Tacchi F, Orozco-Aguilar J, Gutiérrez D, Simon F, Salazar J, Vilos C, Cabello-Verrugio C. Scaffold biomaterials and nano-based therapeutic strategies for skeletal muscle regeneration. Nanomedicine (Lond) 2021; 16:2521-2538. [PMID: 34743611 DOI: 10.2217/nnm-2021-0224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Skeletal muscle is integral to the functioning of the human body. Several pathological conditions, such as trauma (primary lesion) or genetic diseases such as Duchenne muscular dystrophy (DMD), can affect and impair its functions or exceed its regeneration capacity. Tissue engineering (TE) based on natural, synthetic and hybrid biomaterials provides a robust platform for developing scaffolds that promote skeletal muscle regeneration, strength recovery, vascularization and innervation. Recent 3D-cell printing technology and the use of nanocarriers for the release of drugs, peptides and antisense oligonucleotides support unique therapeutic alternatives. Here, the authors present recent advances in scaffold biomaterials and nano-based therapeutic strategies for skeletal muscle regeneration and perspectives for future endeavors.
Collapse
Affiliation(s)
- Franco Tacchi
- Department of Biological Sciences, Laboratory of Muscle Pathology, Fragility & Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile.,Millennium Institute on Immunology & Immunotherapy, Santiago, 8370146, Chile.,Center for The Development of Nanoscience & Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, 8350709, Chile
| | - Josué Orozco-Aguilar
- Department of Biological Sciences, Laboratory of Muscle Pathology, Fragility & Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile.,Millennium Institute on Immunology & Immunotherapy, Santiago, 8370146, Chile.,Center for The Development of Nanoscience & Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, 8350709, Chile
| | - Danae Gutiérrez
- Department of Biological Sciences, Laboratory of Muscle Pathology, Fragility & Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile.,Millennium Institute on Immunology & Immunotherapy, Santiago, 8370146, Chile.,Center for The Development of Nanoscience & Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, 8350709, Chile
| | - Felipe Simon
- Millennium Institute on Immunology & Immunotherapy, Santiago, 8370146, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD),Universidad de Chile, Santiago, 8370146, Chile.,Department of Biological Sciences, Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
| | - Javier Salazar
- Center for The Development of Nanoscience & Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, 8350709, Chile.,Laboratory of Nanomedicine & Targeted Delivery, Center for Medical Research, School of Medicine, Universidad de Talca, Talca, 3460000, Chile
| | - Cristian Vilos
- Center for The Development of Nanoscience & Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, 8350709, Chile.,Laboratory of Nanomedicine & Targeted Delivery, Center for Medical Research, School of Medicine, Universidad de Talca, Talca, 3460000, Chile
| | - Claudio Cabello-Verrugio
- Department of Biological Sciences, Laboratory of Muscle Pathology, Fragility & Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile.,Millennium Institute on Immunology & Immunotherapy, Santiago, 8370146, Chile.,Center for The Development of Nanoscience & Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, 8350709, Chile
| |
Collapse
|
13
|
Kamali Shahri SM, Sharifi S, Mahmoudi M. Interdependency of influential parameters in therapeutic nanomedicine. Expert Opin Drug Deliv 2021; 18:1379-1394. [PMID: 33887999 DOI: 10.1080/17425247.2021.1921732] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction:Current challenges to successful clinical translation of therapeutic nanomedicine have discouraged many stakeholders, including patients. Significant effort has been devoted to uncovering the reasons behind the less-than-expected success, beyond failures or ineffectiveness, of therapeutic nanomedicine products (e.g. cancer nanomedicine). Until we understand and address the factors that limit the safety and efficacy of NPs, both individually and in combination, successful clinical development will lag.Areas covered:This review highlights the critical roles of interdependent factors affecting the safety and therapeutic efficacy of therapeutic NPs for drug delivery applications.Expert opinion:Deep analysis of the current nanomedical literature reveals ahistory of unanticipated complexity by awide range of stakeholders including researchers. In the manufacture of nanomedicines themselves, there have been persistent difficulties with reproducibility and batch-to-batch variation. The unanticipated complexity and interdependency of nano-bio parameters has delayed our recognition of important factors affecting the safety and therapeutic efficacy of nanomedicine products. These missteps have had many factors including our lack of understanding of the interdependency of various factors affecting the biological identity and fate of NPs and biased interpretation of data. All these issues could raise significant concern regarding the reproducibility- or even the validity- of past publications that in turn formed the basis of many clinical trials of therapeutic nanomedicines. Therefore, the individual and combined effects of previously overlooked factors on the safety and therapeutic efficacy of NPs need to be fully considered in nanomedicine reports and product development.
Collapse
Affiliation(s)
- Seyed Mehdi Kamali Shahri
- Department of Radiology and Precision Health Program, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Shahriar Sharifi
- Department of Radiology and Precision Health Program, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
14
|
Bichet MC, Chin WH, Richards W, Lin YW, Avellaneda-Franco L, Hernandez CA, Oddo A, Chernyavskiy O, Hilsenstein V, Neild A, Li J, Voelcker NH, Patwa R, Barr JJ. Bacteriophage uptake by mammalian cell layers represents a potential sink that may impact phage therapy. iScience 2021; 24:102287. [PMID: 33855278 PMCID: PMC8024918 DOI: 10.1016/j.isci.2021.102287] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/15/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
It is increasingly apparent that bacteriophages, viruses that infect bacteria and more commonly referred to as simply phages, have tropisms outside their bacterial hosts. Using live tissue culture cell imaging, we demonstrate that cell type, phage size, and morphology play a major role in phage internalization. Uptake was validated under physiological conditions using a microfluidic device. Phages adhered to mammalian tissues, with adherent phages being subsequently internalized by macropinocytosis, with functional phages accumulating intracellularly. We incorporated these results into a pharmacokinetic model demonstrating the potential impact of phage accumulation by cell layers, which represents a potential sink for circulating phages in the body. During phage therapy, high doses of phages are directly administered to a patient in order to treat a bacterial infection, thereby facilitating broad interactions between phages and mammalian cells. Understanding these interactions will have important implications on innate immune responses, phage pharmacokinetics, and the efficacy of phage therapy.
Collapse
Affiliation(s)
- Marion C. Bichet
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Wai Hoe Chin
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - William Richards
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Yu-Wei Lin
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Laura Avellaneda-Franco
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Catherine A. Hernandez
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Arianna Oddo
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, VIC, 3800, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, 3168, Australia
| | | | - Volker Hilsenstein
- Monash Micro Imaging, Monash University, Clayton Campus, Clayton, VIC, 3800, Australia
| | - Adrian Neild
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton Campus, Clayton, VIC 3800, Australia
| | - Jian Li
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Nicolas Hans Voelcker
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, VIC, 3800, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, 3168, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC 3168, Australia
| | - Ruzeen Patwa
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Jeremy J. Barr
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| |
Collapse
|
15
|
Shojaei S, Ali MS, Suresh M, Upreti T, Mogourian V, Helewa M, Labouta HI. Dynamic placenta-on-a-chip model for fetal risk assessment of nanoparticles intended to treat pregnancy-associated diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166131. [PMID: 33766738 DOI: 10.1016/j.bbadis.2021.166131] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022]
Abstract
Pregnant women often have to take medication either for pregnancy-related diseases or for previously existing medical conditions. Current maternal medications pose fetal risks due to off target accumulation in the fetus. Nanoparticles, engineered particles in the nanometer scale, have been used for targeted drug delivery to the site of action without off-target effects. This has opened new avenues for treatment of pregnancy-associated diseases while minimizing risks on the fetus. It is therefore instrumental to study the potential transfer of nanoparticles from the mother to the fetus. Due to limitations of in vivo and ex vivo models, an in vitro model mimicking the in vivo situation is essential. Placenta-on-a-chip provides a microphysiological recapitulation of the human placenta. Here, we reviewed the fetal risks associated with current therapeutic approaches during pregnancy, analyzed the advantages and limitations of current models used for nanoparticle assessment, and highlighted the current need for using dynamic placenta-on-a-chip models for assessing the safety of novel nanoparticle-based therapies during pregnancy.
Collapse
Affiliation(s)
- Shahla Shojaei
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| | - Moustafa S Ali
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada.
| | - Madhumita Suresh
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| | - Tushar Upreti
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| | - Victoria Mogourian
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| | - Michael Helewa
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Manitoba, Winnipeg, Canada.
| | - Hagar I Labouta
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada; Biomedical Engineering, University of Manitoba, Winnipeg, Canada; Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
16
|
Oddo A, Morozesk M, Lombi E, Schmidt TB, Tong Z, Voelcker NH. Risk assessment on-a-chip: a cell-based microfluidic device for immunotoxicity screening. NANOSCALE ADVANCES 2021; 3:682-691. [PMID: 36133829 PMCID: PMC9416880 DOI: 10.1039/d0na00857e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/17/2020] [Indexed: 06/13/2023]
Abstract
Nanomaterials are widely used in industrial and clinical settings due to their unique physical and chemical properties. However, public health and environmental concerns have emerged owing to their undesired toxicity and ability to trigger immune responses. This paper presents the development of a microfluidic-based cell biochip device that enables the administration of nanoparticles under laminar flow to cells of the immune system to assess their cytotoxicity. The exposure of human B lymphocytes to 10 nm silver nanoparticles under fluid flow led to a 3-fold increase in toxicity compared to static conditions, possibly indicating enhanced cell-nanoparticle interactions. To investigate whether the administration under flow was the main contributing factor, we compared and validated the cytotoxicity of the same nanoparticles in different platforms, including the conventional well plate format and in-house fabricated microfluidic devices under both static and dynamic flow conditions. Our results suggest that commonly employed static platforms might not be well-suited to perform toxicological screening of nanomaterials and may lead to an underestimation of cytotoxic responses. The simplicity of the developed flow system makes this setup a valuable tool to preliminary screen nanomaterials.
Collapse
Affiliation(s)
- Arianna Oddo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria 3052 Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Clayton Victoria 3168 Australia
| | - Mariana Morozesk
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria 3052 Australia
- Universidade Federal de São Carlos, Departamento de Ciências Fisiológicas Rod. Washington Luiz, Km 235, São Carlos 13565-905 São Paulo Brazil
| | - Enzo Lombi
- Future Industries Institute and UniSA STEM, University of South Australia Mawson Lakes 5095 South Australia Australia
| | - Tobias Benedikt Schmidt
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria 3052 Australia
- Department of Applied Chemistry, Reutlingen University Alteburgstraße 150 72762 Reutlingen Germany
| | - Ziqiu Tong
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria 3052 Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Clayton Victoria 3168 Australia
| | - Nicolas Hans Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria 3052 Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Clayton Victoria 3168 Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility Clayton Victoria 3168 Australia
- Department of Materials Science & Engineering, Monash University Clayton Victoria 3168 Australia
| |
Collapse
|
17
|
Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov 2020; 20:101-124. [PMID: 33277608 PMCID: PMC7717100 DOI: 10.1038/s41573-020-0090-8] [Citation(s) in RCA: 2965] [Impact Index Per Article: 741.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
In recent years, the development of nanoparticles has expanded into a broad range of clinical applications. Nanoparticles have been developed to overcome the limitations of free therapeutics and navigate biological barriers — systemic, microenvironmental and cellular — that are heterogeneous across patient populations and diseases. Overcoming this patient heterogeneity has also been accomplished through precision therapeutics, in which personalized interventions have enhanced therapeutic efficacy. However, nanoparticle development continues to focus on optimizing delivery platforms with a one-size-fits-all solution. As lipid-based, polymeric and inorganic nanoparticles are engineered in increasingly specified ways, they can begin to be optimized for drug delivery in a more personalized manner, entering the era of precision medicine. In this Review, we discuss advanced nanoparticle designs utilized in both non-personalized and precision applications that could be applied to improve precision therapies. We focus on advances in nanoparticle design that overcome heterogeneous barriers to delivery, arguing that intelligent nanoparticle design can improve efficacy in general delivery applications while enabling tailored designs for precision applications, thereby ultimately improving patient outcome overall. Advances in nanoparticle design could make substantial contributions to personalized and non-personalized medicine. In this Review, Langer, Mitchell, Peppas and colleagues discuss advances in nanoparticle design that overcome heterogeneous barriers to delivery, as well as the challenges in translating these design improvements into personalized medicine approaches.
Collapse
Affiliation(s)
- Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA. .,Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. .,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. .,Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. .,Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | | | - Rebecca M Haley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Marissa E Wechsler
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA. .,Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA. .,Department of Pediatrics, The University of Texas at Austin, Austin, TX, USA. .,Department of Surgery and Perioperative Care, The University of Texas at Austin, Austin, TX, USA. .,Department of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, USA.
| | - Robert Langer
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
18
|
Shurbaji S, G. Anlar G, A. Hussein E, Elzatahry A, C. Yalcin H. Effect of Flow-Induced Shear Stress in Nanomaterial Uptake by Cells: Focus on Targeted Anti-Cancer Therapy. Cancers (Basel) 2020; 12:E1916. [PMID: 32708521 PMCID: PMC7409087 DOI: 10.3390/cancers12071916] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 02/02/2023] Open
Abstract
Recently, nanomedicines have gained a great deal of attention in diverse biomedical applications, including anti-cancer therapy. Being different from normal tissue, the biophysical microenvironment of tumor cells and cancer cell mechanics should be considered for the development of nanostructures as anti-cancer agents. Throughout the last decades, many efforts devoted to investigating the distinct cancer environment and understanding the interactions between tumor cells and have been applied bio-nanomaterials. This review highlights the microenvironment of cancer cells and how it is different from that of healthy tissue. We gave special emphasis to the physiological shear stresses existing in the cancerous surroundings, since these stresses have a profound effect on cancer cell/nanoparticle interaction. Finally, this study reviews relevant examples of investigations aimed at clarifying the cellular nanoparticle uptake behavior under both static and dynamic conditions.
Collapse
Affiliation(s)
- Samar Shurbaji
- Materials Science and Technology Department, College of Arts and Sciences, Qatar University, Doha 2713, Qatar; (S.S.); (E.A.H.)
| | - Gulsen G. Anlar
- College of Medicine, Department of Medical Sciences, Qatar University, Doha 2713, Qatar;
| | - Essraa A. Hussein
- Materials Science and Technology Department, College of Arts and Sciences, Qatar University, Doha 2713, Qatar; (S.S.); (E.A.H.)
| | - Ahmed Elzatahry
- Materials Science and Technology Department, College of Arts and Sciences, Qatar University, Doha 2713, Qatar; (S.S.); (E.A.H.)
| | - Huseyin C. Yalcin
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
- Department of Biomedical Sciences, College of Health Science-QU Health, Qatar University, Doha 2713, Qatar
| |
Collapse
|
19
|
Bourn MD, Batchelor DVB, Ingram N, McLaughlan JR, Coletta PL, Evans SD, Peyman SA. High-throughput microfluidics for evaluating microbubble enhanced delivery of cancer therapeutics in spheroid cultures. J Control Release 2020; 326:13-24. [PMID: 32562855 DOI: 10.1016/j.jconrel.2020.06.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/12/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023]
Abstract
Drug penetration into solid tumours remains a major challenge in the effective treatment of cancer. Microbubble (MB) mediated sonoporation offers a potential solution to this by enhancing the uptake of drugs into cells. Additionally, in using an ultrasound (US) trigger, drug delivery can be localised to the tumour, thus reducing the off-site toxicity associated with systemic delivery. The majority of in vitro studies involving the observation of MB-enhanced drug efficacy have been conducted on 2D monolayer cell cultures, which are known to be poor models for in vivo tumours. 3D spheroid cultures allow for the production of multicellular cultures complete with extracellular matrix (ECM) components. These cultures effectively recreate many of the physiological features of the tumour microenvironment and have been shown to be far superior to previous 2D monolayer models. However, spheroids are typically handled in well-plates in which the fluid environment is static, limiting the physiological relevance of the model. The combination of 3D cultures and microfluidics would allow for the production of a dynamic system in which spheroids are subjected to in vivo like fluid flow and shear stresses. This study presents a microfluidic device containing an array of spheroid traps, into which multiple pre-grown colorectal cancer (CRC) spheroids were loaded. Reservoirs interfaced with the chip use hydrostatic pressure to passively drive flow through the system and subject spheroids to capillary like flow velocities. The use of reservoirs also enabled multiple chips to be run in parallel, allowing for the screening of multiple therapeutic treatments (n = 690 total spheroids analysed). This microfluidic platform was used to investigate MB enhanced drug delivery and showed that co-delivery of 3 μM doxorubicin (DOX) + MB + US reduced spheroid viability to 48 ± 2%, compared to 75 ± 5% observed with 3 μM DOX alone. Delivery of drug loaded MBs (DLMBs), in which DOX-loaded liposomes (DOX-LS) were conjugated to MBs, reduced spheroid viability to 62 ± 3%, a decrease compared to the 75 ± 3% viability observed with DOX-LS in the absence of MBs + US.
Collapse
Affiliation(s)
- Matthew D Bourn
- School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom; Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds LS9 7TF, United Kingdom
| | - Damien V B Batchelor
- School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Nicola Ingram
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds LS9 7TF, United Kingdom
| | - James R McLaughlan
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds LS9 7TF, United Kingdom; School of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - P Louise Coletta
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds LS9 7TF, United Kingdom
| | - Stephen D Evans
- School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Sally A Peyman
- School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom; Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds LS9 7TF, United Kingdom.
| |
Collapse
|
20
|
Shear Stress-Dependent Targeting Efficiency Using Self-Assembled Gelatin-Oleic Nanoparticles in a Biomimetic Microfluidic System. Pharmaceutics 2020; 12:pharmaceutics12060555. [PMID: 32560107 PMCID: PMC7356760 DOI: 10.3390/pharmaceutics12060555] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 11/16/2022] Open
Abstract
Cellular properties and microenvironments, as well as the characteristics of nanoparticles (NPs), affect the cellular uptake and cytotoxic effects of drug-loaded NPs. Since there is fluid flow in the human blood system, fluid flow also affects the drug delivery efficiency of NPs. This study aimed to evaluate the cellular behaviors of drug-loaded soft NPs on A549 cancer cells under different levels of shear stress (0.5, 5, and 50 dynes/cm2) in the biomimetic microfluidic system. The soft self-assembled NPs were formed by the gelatin-oleic conjugate (GOC). The poorly water-soluble coumarin-6 or paclitaxel (PTX) were used as model markers for encapsulation within self-assembled NPs (C-GONs or PTX-GONs, respectively). The cellular uptake of C-GONs was found to be improved with shear-stress dependence. The inhibitory concentration (IC50) of PTX-GONs at 0.5, 5, and 50 dynes/cm2 was 0.106 µg/mL, 0.108 µg/mL, and 0.091 µg/mL, respectively, as compared to 0.138 µg/mL in a static condition. The cell killing efficiency of PTX-GONs was increased in the highest shear stress of 50 dynes/cm2 in the static condition, and other levels of shear stress in dynamic conditions.
Collapse
|
21
|
Del Favero G, Kraegeloh A. Integrating Biophysics in Toxicology. Cells 2020; 9:E1282. [PMID: 32455794 PMCID: PMC7290780 DOI: 10.3390/cells9051282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/10/2020] [Accepted: 05/15/2020] [Indexed: 12/20/2022] Open
Abstract
Integration of biophysical stimulation in test systems is established in diverse branches of biomedical sciences including toxicology. This is largely motivated by the need to create novel experimental setups capable of reproducing more closely in vivo physiological conditions. Indeed, we face the need to increase predictive power and experimental output, albeit reducing the use of animals in toxicity testing. In vivo, mechanical stimulation is essential for cellular homeostasis. In vitro, diverse strategies can be used to model this crucial component. The compliance of the extracellular matrix can be tuned by modifying the stiffness or through the deformation of substrates hosting the cells via static or dynamic strain. Moreover, cells can be cultivated under shear stress deriving from the movement of the extracellular fluids. In turn, introduction of physical cues in the cell culture environment modulates differentiation, functional properties, and metabolic competence, thus influencing cellular capability to cope with toxic insults. This review summarizes the state of the art of integration of biophysical stimuli in model systems for toxicity testing, discusses future challenges, and provides perspectives for the further advancement of in vitro cytotoxicity studies.
Collapse
Affiliation(s)
- Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38-40, 1090 Vienna, Austria
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna Währinger Straße 38-40, 1090 Vienna, Austria
| | - Annette Kraegeloh
- INM—Leibniz-Institut für Neue Materialien GmbH, Campus D2 2, 66123 Saarbrücken, Germany;
| |
Collapse
|
22
|
Liu X, Jansman MMT, Thulstrup PW, Mendes AC, Chronakis IS, Hosta‐Rigau L. Low‐Fouling Electrosprayed Hemoglobin Nanoparticles with Antioxidant Protection as Promising Oxygen Carriers. Macromol Biosci 2019; 20:e1900293. [DOI: 10.1002/mabi.201900293] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/31/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Xiaoli Liu
- DTU Health TechCenter for Nanomedicine and TheranosticsTechnical University of DenmarkNils Koppels Allé B423 2800 Kgs Lyngby Denmark
| | - Michelle M. T. Jansman
- DTU Health TechCenter for Nanomedicine and TheranosticsTechnical University of DenmarkNils Koppels Allé B423 2800 Kgs Lyngby Denmark
| | - Peter W. Thulstrup
- Department of ChemistryUniversity of Copenhagen Universitetsparken 5 2100 Copenhagen Denmark
| | - Ana C. Mendes
- DTU FoodTechnical University of Denmark Kemitorvet, B202 2800 Kgs Lyngby Denmark
| | - Ioannis S. Chronakis
- DTU FoodTechnical University of Denmark Kemitorvet, B202 2800 Kgs Lyngby Denmark
| | - Leticia Hosta‐Rigau
- DTU Health TechCenter for Nanomedicine and TheranosticsTechnical University of DenmarkNils Koppels Allé B423 2800 Kgs Lyngby Denmark
| |
Collapse
|
23
|
York-Duran MJ, Godoy-Gallardo M, Jansman MMT, Hosta-Rigau L. A dual-component carrier with both non-enzymatic and enzymatic antioxidant activity towards ROS depletion. Biomater Sci 2019; 7:4813-4826. [PMID: 31535107 DOI: 10.1039/c9bm00913b] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
While ROS display crucial functions in many physiological processes, elevated ROS levels are also related to the initiation and progression of many severe diseases such as cancer, cardiovascular conditions or neurologic disorders. Research approaches to diminish ROS levels during disease progression are currently being focused on the therapeutic administration of antioxidant enzymes. However, enzyme administration suffers from several limitations including their fast elimination from blood upon administration, thus making crucial the development of enzyme encapsulating platforms. We have recently reported a multicompartment architecture constituted by two inherently different types of materials, i.e., polymeric microgels and liposomes. Poly(N-isopropylacrylamide-co-acrylic acid) microgels decorated with liposomes and subsequently coated by a protective poly(dopamine) shell (PDA) combine the benefits of both systems while minimizing some of their drawbacks. Herein, we exploit this dual-component platform as a microreactor for ROS depletion. We combine the intrinsic PDA's antioxidant properties with the encapsulation of the catalase enzyme within the liposomal compartments. The surface of the carrier is further functionalised with a poly(ethylene glycol) layer and the low fouling properties are demonstrated in terms of reduction of protein adsorption and cellular uptake. The potential of the carrier as an antioxidant microreactor is shown by its ability to deplete superoxide radicals and hydrogen peroxide, which can also take place in the presence of the two relevant cell lines.
Collapse
Affiliation(s)
- Maria Jose York-Duran
- Department of Health Technology, Centre for Nanomedicine and Theranostics, DTU Health Tech, Technical University of Denmark, Building 423, 2800, Lyngby, Denmark.
| | - Maria Godoy-Gallardo
- Department of Health Technology, Centre for Nanomedicine and Theranostics, DTU Health Tech, Technical University of Denmark, Building 423, 2800, Lyngby, Denmark.
| | - Michelle Maria Theresia Jansman
- Department of Health Technology, Centre for Nanomedicine and Theranostics, DTU Health Tech, Technical University of Denmark, Building 423, 2800, Lyngby, Denmark.
| | - Leticia Hosta-Rigau
- Department of Health Technology, Centre for Nanomedicine and Theranostics, DTU Health Tech, Technical University of Denmark, Building 423, 2800, Lyngby, Denmark.
| |
Collapse
|
24
|
Gal N, Charwat V, Städler B, Reimhult E. Poly(ethylene glycol) Grafting of Nanoparticles Prevents Uptake by Cells and Transport Through Cell Barrier Layers Regardless of Shear Flow and Particle Size. ACS Biomater Sci Eng 2019; 5:4355-4365. [PMID: 33438401 DOI: 10.1021/acsbiomaterials.9b00611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
It has long been a central tenet of biomedical research that coating of nanoparticles with hydrated polymers can improve their performance in biomedical applications. However, the efficacy of the approach in vivo is still debated. In vitro model systems to test the performance of engineered nanoparticles for in vivo applications often use nonrepresentative cell lines and conditions for uptake and toxicity tests. We use our platform of monodisperse iron oxide nanoparticles densely grafted with nitrodopamide-poly(ethylene glycol) (PEG) to probe cell interactions with a set of cell types and culture conditions that are relevant for applications in which nanoparticles are injected into the bloodstream. In the past, these particles have proved to have excellent stability and negligible interaction with proteins and membranes under physiological conditions. We test the influence of flow on the uptake of nanoparticles. We also investigate the transport through endothelial barrier cell layers, as well as the effect that PEG-grafted iron oxide nanoparticles have on cell layers relevant for nanoparticles injected into the bloodstream. Our results show that the dense PEG brush and resulting lack of nonspecific protein and membrane interaction lead to negligible cell uptake, toxicity, and transport across barrier layers. These results contrast with far less well-defined polymer-coated nanoparticles that tend to aggregate and consequently strongly interact with cells, for example, by endocytosis.
Collapse
Affiliation(s)
- Noga Gal
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark
| | | | - Brigitte Städler
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark
| | | |
Collapse
|
25
|
Ruzycka M, Cimpan MR, Rios-Mondragon I, Grudzinski IP. Microfluidics for studying metastatic patterns of lung cancer. J Nanobiotechnology 2019; 17:71. [PMID: 31133019 PMCID: PMC6537392 DOI: 10.1186/s12951-019-0492-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/04/2019] [Indexed: 01/09/2023] Open
Abstract
The incidence of lung cancer continues to rise worldwide. Because the aggressive metastasis of lung cancer cells is the major drawback of successful therapies, the crucial challenge of modern nanomedicine is to develop diagnostic tools to map the molecular mechanisms of metastasis in lung cancer patients. In recent years, microfluidic platforms have been given much attention as tools for novel point-of-care diagnostic, an important aspect being the reconstruction of the body organs and tissues mimicking the in vivo conditions in one simple microdevice. Herein, we present the first comprehensive overview of the microfluidic systems used as innovative tools in the studies of lung cancer metastasis including single cancer cell analysis, endothelial transmigration, distant niches migration and finally neoangiogenesis. The application of the microfluidic systems to study the intercellular crosstalk between lung cancer cells and surrounding tumor microenvironment and the connection with multiple molecular signals coming from the external cellular matrix are discussed. We also focus on recent breakthrough technologies regarding lab-on-chip devices that serve as tools for detecting circulating lung cancer cells. The superiority of microfluidic systems over traditional in vitro cell-based assays with regard to modern nanosafety studies and new cancer drug design and discovery is also addressed. Finally, the current progress and future challenges regarding printable and paper-based microfluidic devices for personalized nanomedicine are summarized.
Collapse
Affiliation(s)
- Monika Ruzycka
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097, Warsaw, Poland
| | - Mihaela R Cimpan
- Biomaterials - Department for Clinical Dentistry, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Ivan Rios-Mondragon
- Biomaterials - Department for Clinical Dentistry, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Ireneusz P Grudzinski
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097, Warsaw, Poland.
| |
Collapse
|
26
|
Godoy-Gallardo M, Labay C, Hosta-Rigau L. Tyrosinase-Loaded Multicompartment Microreactor toward Melanoma Depletion. ACS APPLIED MATERIALS & INTERFACES 2019; 11:5862-5876. [PMID: 30605301 DOI: 10.1021/acsami.8b20275] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Melanoma is malignant skin cancer occurring with increasing prevalence with no effective treatment. A unique feature of melanoma cells is that they require higher concentrations of ltyrosine (l-tyr) for expansion than normal cells. As such, it has been demonstrated that dietary l-tyr restriction lowers systemic l-tyr and suppresses melanoma advancement in mice. Unfortunately, this diet is not well tolerated by humans. An alternative approach to impede melanoma progression will be to administer the enzyme tyrosinase (TYR), which converts l-tyr into melanin. Herein, a multicompartment carrier consisting of a polymer shell entrapping thousands of liposomes is employed to act as a microreactor depleting l-tyr in the presence of melanoma cells. It is shown that the TYR enzyme can be incorporated within the liposomal subunits with preserved catalytic activity. Aiming to mimic the dynamic environment at the tumor site, l-tyr conversion is conducted by co-culturing melanoma cells and microreactors in a microfluidic setup with applied intratumor shear stress. It is demonstrated that the microreactors are concurrently depleting l-tyr, which translates into inhibited melanoma cell growth. Thus, the first microreactor where the depletion of a substrate translates into antitumor properties in vitro is reported.
Collapse
Affiliation(s)
- Maria Godoy-Gallardo
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech , Technical University of Denmark , Building 423 , 2800 Lyngby , Denmark
| | - Cédric Labay
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech , Technical University of Denmark , Building 423 , 2800 Lyngby , Denmark
| | - Leticia Hosta-Rigau
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech , Technical University of Denmark , Building 423 , 2800 Lyngby , Denmark
| |
Collapse
|
27
|
York-Duran MJ, Ek PK, Godoy-Gallardo M, Hosta-Rigau L. Shear stress regulated uptake of liposome-decorated microgels coated with a poly(dopamine) shell. Colloids Surf B Biointerfaces 2018; 171:427-436. [PMID: 30075418 DOI: 10.1016/j.colsurfb.2018.07.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/12/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022]
Abstract
Advanced multicompartment drug delivery platforms ensure the co-localization of several drugs within the same carrier, thus making it possible to achieve a more effective and safe therapeutic outcome. Herein, we report a novel multicompartment architecture by combining two intrinsically different systems, i.e., polymeric microgels and liposomes, with the aim to achieve different release kinetics for model compounds. We assemble poly(N-isopropylacrylamide-co-acrylic acid) microgels decorated with liposomes which are subsequently coated with a protective poly(dopamine) shell and a poly(ethylene glycol) (PEG) layer. Since any intravenous administered drug delivery vehicle will get in contact with the dynamics of the blood flow, we evaluate the stealth properties of this novel multicompartment carrier towards protein adsorption and cellular uptake by three relevant cell lines (macrophages, endothelial and cancer cells) under physiological shear stress conditions. Our results demonstrate less protein adsorption for the PEGylated carriers and differences in the extent of internalized carriers depending on the presence of a PEG coating, the studied cell line and the intensity of the applied shear stress. Additionally, we demonstrate that, for all three tested cell lines, shear stress results in the activation of different cell entry pathways as compared to static conditions. All in all, we report a thorough study about the effect of shear stress on the cell association/uptake with a novel multicompartment carrier.
Collapse
Affiliation(s)
- Maria Jose York-Duran
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, 2800, Lyngby, Denmark
| | - Pramod Kumar Ek
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, 2800, Lyngby, Denmark
| | - Maria Godoy-Gallardo
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, 2800, Lyngby, Denmark
| | - Leticia Hosta-Rigau
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, 2800, Lyngby, Denmark.
| |
Collapse
|
28
|
Moore TL, Hauser D, Gruber T, Rothen-Rutishauser B, Lattuada M, Petri-Fink A, Lyck R. Cellular Shuttles: Monocytes/Macrophages Exhibit Transendothelial Transport of Nanoparticles under Physiological Flow. ACS APPLIED MATERIALS & INTERFACES 2017; 9:18501-18511. [PMID: 28517937 DOI: 10.1021/acsami.7b03479] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A major hurdle in the development of biomedical nanoparticles (NP) is understanding how they interact with complex biological systems and navigate biological barriers to arrive at pathological targets. It is becoming increasingly evident that merely controlling particle physicochemical properties may not be sufficient to mediate particle biodistribution in dynamic environments. Thus, researchers are increasingly turning toward more complex but likewise more physiological in vitro systems to study particle--cell/particle-system interactions. An emerging paradigm is to utilize naturally migratory cells to act as so-called "Trojan horses" or cellular shuttles. We report here the use of monocytes/macrophages to transport NP across a confluent endothelial cell layer using a microfluidic in vitro model. With a custom-built flow chamber, we showed that physiological shear stress, when compared to low flow or static conditions, increased NP uptake by macrophages. We further provided a mathematical explanation for the effect of flow on NP uptake, namely that the physical exposure times of NP to cells is dictated by shear stress (i.e., flow rate) and results in increased particle uptake under flow. This study was extended to a multicellular, hydrodynamic in vitro model. Because monocytes are cells that naturally translocate across biological barriers, we utilized a monocyte/macrophage cell line as cellular NP transporters across an endothelial layer. In this exploratory study, we showed that monocyte/macrophage cells adhere to an endothelial layer and dynamically interact with the endothelial cells. The monocytes/macrophages took up NP and diapedesed across the endothelial layer with NP accumulating within the cellular uropod. These data illustrate that monocytes/macrophages may therefore act as active shuttles to deliver particles across endothelial barriers.
Collapse
Affiliation(s)
| | | | - Thomas Gruber
- Theodor Kocher Institute, Universität Bern , 3000 Bern, Switzerland
| | | | | | | | - Ruth Lyck
- Theodor Kocher Institute, Universität Bern , 3000 Bern, Switzerland
| |
Collapse
|
29
|
Kang T, Tran TTT, Park C, Lee BJ. Biomimetic shear stress and nanoparticulate drug delivery. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0313-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
30
|
Affiliation(s)
- Yuqi Zhang
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Center
for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics,
UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department
of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Jicheng Yu
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Center
for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics,
UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Hunter N. Bomba
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Yong Zhu
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Department
of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Zhen Gu
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Center
for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics,
UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department
of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
31
|
Kang T, Park C, Choi JS, Cui JH, Lee BJ. Effects of shear stress on the cellular distribution of polystyrene nanoparticles in a biomimetic microfluidic system. J Drug Deliv Sci Technol 2016. [DOI: 10.1016/j.jddst.2015.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
32
|
Godoy-Gallardo M, Ek PK, Jansman MMT, Wohl BM, Hosta-Rigau L. Interaction between drug delivery vehicles and cells under the effect of shear stress. BIOMICROFLUIDICS 2015; 9:052605. [PMID: 26180575 PMCID: PMC4491015 DOI: 10.1063/1.4923324] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/18/2015] [Indexed: 05/06/2023]
Abstract
Over the last decades, researchers have developed an ever greater and more ingenious variety of drug delivery vehicles (DDVs). This has made it possible to encapsulate a wide selection of therapeutic agents, ranging from proteins, enzymes, and peptides to hydrophilic and hydrophobic small drugs while, at the same time, allowing for drug release to be triggered through a diverse range of physical and chemical cues. While these advances are impressive, the field has been lacking behind in translating these systems into the clinic, mainly due to low predictability of in vitro and rodent in vivo models. An important factor within the complex and dynamic human in vivo environment is the shear flow observed within our circulatory system and many other tissues. Within this review, recent advances to leverage microfluidic devices to better mimic these conditions through novel in vitro assays are summarized. By grouping the discussion in three prominent classes of DDVs (lipidic and polymeric particles as well as inorganic nanoparticles), we hope to guide researchers within drug delivery into this exciting field and advance a further implementation of these assay systems within the development of DDVs.
Collapse
Affiliation(s)
- M Godoy-Gallardo
- Department of Micro-and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
| | - P K Ek
- Department of Micro-and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
| | - M M T Jansman
- Department of Micro-and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
| | - B M Wohl
- Department of Micro-and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
| | - L Hosta-Rigau
- Department of Micro-and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
| |
Collapse
|
33
|
Panneerselvam K, Lynge ME, Riber CF, Mena-Hernando S, Smith AAA, Goldie KN, Zelikin AN, Städler B. Phospholipid-polymer amphiphile hybrid assemblies and their interaction with macrophages. BIOMICROFLUIDICS 2015; 9:052610. [PMID: 26339330 PMCID: PMC4552693 DOI: 10.1063/1.4929405] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/11/2015] [Indexed: 05/06/2023]
Abstract
Recently, the combination of lipids and block copolymers has become an alternative to liposomes and polymersomes as nano-sized drug carriers. We synthesize novel block copolymers consisting of poly(cholesteryl acrylate) as the hydrophobic core and poly(N-isopropylacrylamide) (PNIPAAm) as the hydrophilic extensions. Their successful phospholipid-assisted assembly into vesicles is demonstrated using the evaporation-hydration method. The preserved thermo-responsive property of the lipid-polymer hybrids is shown by a temperature dependent adsorption behaviour of the vesicles to poly(l lysine) coated surfaces. As expected, the vesicle adsorption is found to be higher at elevated temperatures. The cellular uptake efficiency of hybrids is assessed using macrophages with applied shear stress. The amount of adhering macrophages is affected by the time and level of applied shear stress. Further, it is found that shorter PNIPAAm extensions lead to higher uptake of the assemblies by the macrophages with applied shear stress. No inherent cytotoxicity is observed at the tested conditions. Taken together, this first example of responsive lipid-polymer hybrids, and their positive biological evaluation makes them promising nano-sized drug carrier candidates.
Collapse
Affiliation(s)
| | - Martin E Lynge
- Aarhus University , Interdisciplinary Nanoscience Center (iNANO), Aarhus, Denmark
| | | | - Sofia Mena-Hernando
- Aarhus University , Interdisciplinary Nanoscience Center (iNANO), Aarhus, Denmark
| | - Anton A A Smith
- Department of Chemistry, Aarhus University , Aarhus, Denmark
| | - Kenneth N Goldie
- Center for Cellular Imaging and Nano Analytics, Biozentrum, University of Basel , Basel, Switzerland
| | | | - Brigitte Städler
- Aarhus University , Interdisciplinary Nanoscience Center (iNANO), Aarhus, Denmark
| |
Collapse
|
34
|
Lynge ME, Fernandez-Medina M, Postma A, Städler B. Liposomal Drug Deposits in Poly(Dopamine) Coatings: Effect of Their Composition, Cell Type, Uptake Pathway Considerations, and Shear Stress. Macromol Biosci 2014; 14:1677-87. [DOI: 10.1002/mabi.201400350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 08/15/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Martin E. Lynge
- iNANO, Aarhus University; Gustav Wieds Vej 14 Aarhus 8000 Denmark
| | | | - Almar Postma
- CSIRO Materials Science and Engineering; Ian Wark Laboratory; Bayview Ave, Clayton Victoria 3168 Australia
| | - Brigitte Städler
- iNANO, Aarhus University; Gustav Wieds Vej 14 Aarhus 8000 Denmark
| |
Collapse
|
35
|
Panneerselvam K, Mena-Hernando S, Teo BM, Goldie KN, Städler B. Liposomes equipped with poly(N-isopropyl acryl amide)-containing coatings as potential drug carriers. RSC Adv 2014. [DOI: 10.1039/c4ra07720b] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
36
|
Björnmalm M, Yan Y, Caruso F. Engineering and evaluating drug delivery particles in microfluidic devices. J Control Release 2014; 190:139-49. [DOI: 10.1016/j.jconrel.2014.04.030] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 03/14/2014] [Accepted: 03/21/2014] [Indexed: 02/03/2023]
|
37
|
Fejerskov B, Jensen NBS, Teo BM, Städler B, Zelikin AN. Biocatalytic polymer coatings: on-demand drug synthesis and localized therapeutic effect under dynamic cell culture conditions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:1314-1324. [PMID: 24376172 DOI: 10.1002/smll.201303101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/05/2013] [Indexed: 06/03/2023]
Abstract
Biocatalytic surface coatings are prepared herein for localized synthesis of drugs and their on-demand, site-specific delivery to adhering cells. This novel approach is based on the incorporation of an enzyme into multilayered polymer coatings to accomplish enzyme-prodrug therapy (EPT). The build-up of enzyme-containing multilayered coatings is characterized and correlations are drawn between the multilayer film assembly conditions and the enzymatic activity of the resulting coatings. Therapeutic effect elicited by the substrate mediated EPT (SMEPT) strategy is investigated using a prodrug for an anticancer agent, SN-38. The performance of biocatalytic coatings under flow conditions is investigated and it is demonstrated that EPT allows synthesizing the drugs on-demand, at the time desired and in a controllable amount to suit particular applications. Finally, using cells cultured in sequentially connected flow chambers, it is demonstrated that SMEPT affords a site-specific drug delivery, that is, exerts a higher therapeutic effect in cells adhering directly to the biocatalytic coatings than in the cells cultured "downstream". Taken together, these data illustrate biomedical opportunities made possible by engineering tools of EPT into multilayered polymer coatings and present a novel, highly versatile tool for surface mediated drug delivery.
Collapse
Affiliation(s)
- Betina Fejerskov
- Department of Chemistry, Aarhus University, Aarhus, 8000, Denmark
| | | | | | | | | |
Collapse
|
38
|
Teo BM, van der Westen R, Hosta-Rigau L, Städler B. Cell response to PEGylated poly(dopamine) coated liposomes considering shear stress. Biochim Biophys Acta Gen Subj 2013; 1830:4838-47. [DOI: 10.1016/j.bbagen.2013.06.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 06/06/2013] [Accepted: 06/17/2013] [Indexed: 12/13/2022]
|