1
|
Wu W, Li Y, Liu Q, Liu T, Zhao Y, Shao H, Ren P, Tang Y, Feng J, Wang Y, Sun G, Liu H, Bai Y, Chen F. Dual-Targeted Drug Delivery to Myeloid Leukemia Cells via Complement- and Transferrin-Based Protein Corona. NANO LETTERS 2025; 25:147-156. [PMID: 39694635 DOI: 10.1021/acs.nanolett.4c04429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Although traditionally regarded as an impediment, the protein corona can facilitate the advancement of targeted drug delivery systems. This study presents an innovative approach for targeting acute myeloid leukemia (AML) using nanoparticles with agglutinated protein (NAPs). Agglutinated transferrin and C3b in NAPs selectively bind to CD71 and CD11b, receptors that are overexpressed on myeloid leukemic cells compared to nonmalignant cells. In vitro, NAPs achieved a 73.9% doxorubicin (DOX) uptake in leukemic cells, compared to 6.19% for the free drug, while significantly reducing off-target accumulation in normal cells from 42.9% to 5.76%. In vivo, the distribution of NAPs correlated to the organ infiltration pattern of leukemic cells. NAPs demonstrated antileukemic activity in both in vitro and in vivo NSG mouse models, inducing cell death via apoptosis and ferroptosis. In conclusion, NAP-mediated targeted drug delivery represents a promising therapeutic strategy for AML, enhancing treatment efficacy and minimizing off-target effects.
Collapse
Affiliation(s)
- Wen Wu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Yuanyuan Li
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Qihui Liu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Tao Liu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Yanan Zhao
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Hui Shao
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Ping Ren
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130033, P. R. China
| | - Yueyang Tang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Jiayi Feng
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Yihan Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Guodong Sun
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, P. R. China
| | - Haiyan Liu
- Key Laboratory of Pathobiology Ministry of Education, Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun 130021, P. R. China
| | - Yuansong Bai
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Fangfang Chen
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| |
Collapse
|
2
|
Li Y, Saba L, Scheinman RI, Banda NK, Holers M, Monte A, Dylla L, Moghimi SM, Simberg D. Nanoparticle-Binding Immunoglobulins Predict Variable Complement Responses in Healthy and Diseased Cohorts. ACS NANO 2024; 18:28649-28658. [PMID: 39395006 DOI: 10.1021/acsnano.4c05087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Systemic administration of nanomedicines results in the activation of the complement cascade, promoting phagocytic uptake and triggering proinflammatory responses. Identifying the biomarkers that can predict the "risk" of abnormally high complement responders can improve the safety and efficacy of nanomedicines. Polyethylene glycol (PEG) and dextran are two types of clinically approved polymer coatings that trigger complement activation. We performed a multifaceted analysis of the factors affecting the complement activation by PEGylated liposomal doxorubicin (PLD) and dextran-coated superparamagnetic iron oxide nanoworms (SPIO NWs) in plasma from patients with different inflammatory disease conditions and healthy donors. The complement activation (measured as deposition of the complement protein C3) varied greatly, with 29-fold and 26-fold differences for PLD and SPIO NWs, respectively. Chronic inflammation, acute infection, use of steroids, and sex had minor effects on the variable complement activation, whereas age inversely correlated with the complement activation. C-reactive protein level was not predictive of high (top 20th percentile) complement responses. Plasma concentrations of the main complement factors, as well as total IgG and IgM, showed no correlation with the activation by either nanoparticle. On the other hand, plasma concentrations of anti-PEG IgG and IgM showed a strong positive correlation with the activation by PLD. Particularly, titers of anti-PEG IgM showed the best predictive value for the "risk" of high complement activation by PLD. Titers of antidextran IgG and IgM showed a lower correlation with the activation by SPIO NWs and poor predictive value of the top 20% complement responses. Nanoparticle-bound immunoglobulins showed the best correlation with complement activation and a strong predictive value, supporting the critical role of immunoglobulins in inciting complement. The opsonization of PLD with C3 in plasma with high anti-PEG antibodies was predominantly via the alternative pathway. Characterizing the nature of nanoparticle-binding antibodies has important implications in mitigating and stratifying nanomedicine safety.
Collapse
Affiliation(s)
- Yue Li
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Laura Saba
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Robert I Scheinman
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Nirmal K Banda
- Division of Rheumatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Michael Holers
- Division of Rheumatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Andrew Monte
- Department of Emergency Medicine, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Layne Dylla
- Department of Emergency Medicine, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - S Moein Moghimi
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, U.K
- Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, U.K
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
3
|
Li Y, Monte A, Dylla L, Moghimi SM, Simberg D. Validation of dot blot immunoassay for measurement of complement opsonization of nanoparticles. J Immunol Methods 2024; 528:113668. [PMID: 38574804 PMCID: PMC11023749 DOI: 10.1016/j.jim.2024.113668] [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] [Received: 02/07/2024] [Revised: 03/28/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Complement plays a critical role in the immune response toward nanomaterials. The complement attack on a foreign surface results in the deposition of C3, assembly of C3 convertases, the release of anaphylatoxins C3a and C5a, and finally, the formation of membrane attack complex C5b-9. Various technologies can measure complement activation markers in the fluid phase, but measurements of surface C3 deposition are less common. Previously, we developed an ultracentrifugation-based dot blot immunoassay (DBI) to measure the deposition of C3 and other protein corona components on nanoparticles. Here, we validate the repeatability of the DBI and its correlation with pathway-specific and common fluid phase markers. Moreover, we discuss the advantages of DBI, such as cost-effectiveness and versatility, while addressing potential limitations. This study provides insights into complement activation at the nanosurface level, offering a valuable tool for nanomedicine researchers in the field.
Collapse
Affiliation(s)
- Yue Li
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew Monte
- Department of Emergency Medicine, the University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Layne Dylla
- Department of Emergency Medicine, the University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - S Moein Moghimi
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dmitri Simberg
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
4
|
Li M, Yao H, Yi K, Lao YH, Shao D, Tao Y. Emerging nanoparticle platforms for CpG oligonucleotide delivery. Biomater Sci 2024; 12:2203-2228. [PMID: 38293828 DOI: 10.1039/d3bm01970e] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Unmethylated cytosine-phosphate-guanine (CpG) oligodeoxynucleotides (ODNs), which were therapeutic DNA with high immunostimulatory activity, have been applied in widespread applications from basic research to clinics as therapeutic agents for cancer immunotherapy, viral infection, allergic diseases and asthma since their discovery in 1995. The major factors to consider for clinical translation using CpG motifs are the protection of CpG ODNs from DNase degradation and the delivery of CpG ODNs to the Toll-like receptor-9 expressed human B-cells and plasmacytoid dendritic cells. Therefore, great efforts have been devoted to the advances of efficient delivery systems for CpG ODNs. In this review, we outline new horizons and recent developments in this field, providing a comprehensive summary of the nanoparticle-based CpG delivery systems developed to improve the efficacy of CpG-mediated immune responses, including DNA nanostructures, inorganic nanoparticles, polymer nanoparticles, metal-organic-frameworks, lipid-based nanosystems, proteins and peptides, as well as exosomes and cell membrane nanoparticles. Moreover, future challenges in the establishment of CpG delivery systems for immunotherapeutic applications are discussed. We expect that the continuously growing interest in the development of CpG-based immunotherapy will certainly fuel the excitement and stimulation in medicine research.
Collapse
Affiliation(s)
- Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Haochen Yao
- Hepatobiliary and Pancreatic Surgery Department, General Surgery Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Dan Shao
- Institutes of Life Sciences, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
5
|
Li Y, Moein Moghimi S, Simberg D. Complement-dependent uptake of nanoparticles by blood phagocytes: brief overview and perspective. Curr Opin Biotechnol 2024; 85:103044. [PMID: 38091875 PMCID: PMC11214757 DOI: 10.1016/j.copbio.2023.103044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/22/2023] [Indexed: 02/09/2024]
Abstract
Immune recognition and uptake of nanoparticles remain the hot topic in nanomedicine research. Complement is the central player in the immune recognition of engineered nanoparticles. Here, we summarize the accumulated knowledge on the role of complement in the interactions of nanomaterials with blood phagocytes. We describe the interplay between surface properties, complement opsonization, and immune uptake, primarily of iron oxide nanoparticles. We discuss the rigor of the published research and further identify the following knowledge gaps: 1) the role of complement in the variability of uptake of nanomaterials in healthy and diseased subjects, and 2) modulation of complement interactions to improve the performance of nanomaterials. Addressing these gaps is critical to improving translational chances of nanomaterials for drug delivery and imaging applications.
Collapse
Affiliation(s)
- Yue Li
- Translational Bio-Nanosciences Laboratory, USA; Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Seyed Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, USA; Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
6
|
Li Y, Jacques S, Gaikwad H, Wang G, Banda NK, Holers VM, Scheinman RI, Tomlinson S, Moghimi SM, Simberg D. Inhibition of acute complement responses towards bolus-injected nanoparticles using targeted short-circulating regulatory proteins. NATURE NANOTECHNOLOGY 2024; 19:246-254. [PMID: 37798566 PMCID: PMC11034866 DOI: 10.1038/s41565-023-01514-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/25/2023] [Indexed: 10/07/2023]
Abstract
Effective inhibition of the complement system is needed to prevent the accelerated clearance of nanomaterials by complement cascade and inflammatory responses. Here we show that a fusion construct consisting of human complement receptor 2 (CR2) (which recognizes nanosurface-deposited complement 3 (C3)) and complement receptor 1 (CR1) (which blocks C3 convertases) inhibits complement activation with picomolar to low nanomolar efficacy on many types of nanomaterial. We demonstrate that only a small percentage of nanoparticles are randomly opsonized with C3 both in vitro and in vivo, and CR2-CR1 immediately homes in on this subpopulation. Despite rapid in vivo clearance, the co-injection of CR2-CR1 in rats, or its mouse orthologue CR2-Crry in mice, with superparamagnetic iron oxide nanoparticles nearly completely blocks complement opsonization and unwanted granulocyte/monocyte uptake. Furthermore, the inhibitor completely prevents lethargy caused by bolus-injected nanoparticles, without inducing long-lasting complement suppression. These findings suggest the potential of the targeted complement regulators for clinical evaluation.
Collapse
Affiliation(s)
- Yue Li
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sarah Jacques
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hanmant Gaikwad
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Guankui Wang
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nirmal K Banda
- Division of Rheumatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - V Michael Holers
- Division of Rheumatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert I Scheinman
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephen Tomlinson
- Medical University of South Carolina Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Ralph Johnson Veterans Affairs Medical Center, Charleston, SC, USA
| | - S Moein Moghimi
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
7
|
Saafane A, Girard D. Interaction between iron oxide nanoparticles (IONs) and primary human immune cells: An up-to-date review of the literature. Toxicol In Vitro 2023:105635. [PMID: 37356554 DOI: 10.1016/j.tiv.2023.105635] [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: 12/02/2022] [Revised: 04/19/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
Nanotechnology has been gaining more and more momentum lately and the potential use of nanomaterials such as nanoparticles (NPs) continues to grow in a variety of activity sectors. Among the NPs, iron oxide nanoparticles (IONs) have retained an increasing interest from the scientific community and industrials due to their superparamagnetic properties allowing their use in many fields, including medicine. However, some undesired effects of IONs and potential risk for human health are becoming increasingly reported in several studies. Although many in vivo studies reported that IONs induce immunotoxicity in different animal models, it is not clear how IONs can alter the biology of primary human immune cells. In this article, we will review the works that have been done regarding the interaction between IONs and primary immune cells. This review also outlines the importance of using primary immune cells in risk assessment of NPs as a reliable strategy for encouraging non-animal studies approaches, to determine risks that might affect the human immune system following different exposure scenarios. Taken all together, the reported observations help to get a more global picture on how IONs alter the human immune system especially the fact that inflammation, known to involve several immune cell types, is frequently reported as an undesired effect of IONs.
Collapse
Affiliation(s)
- Abdelaziz Saafane
- Laboratoire de Recherche en Inflammation et Physiologie des Granulocytes, Université du Québec, Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
| | - Denis Girard
- Laboratoire de Recherche en Inflammation et Physiologie des Granulocytes, Université du Québec, Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada.
| |
Collapse
|
8
|
Boinapalli Y, Shankar Pandey R, Singh Chauhan A, Sudheesh MS. Physiological relevance of in-vitro cell-nanoparticle interaction studies as a predictive tool in cancer nanomedicine research. Int J Pharm 2023; 632:122579. [PMID: 36603671 DOI: 10.1016/j.ijpharm.2022.122579] [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: 10/23/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Cell uptake study is a routine experiment used as a surrogate to predict in vivo response in cancer nanomedicine research. Cell culture conditions should be designed in such a way that it emulates 'real' physiological conditions and avoid artefacts. It is critical to dissect the steps involved in cellular uptake to understand the physical, chemical, and biological factors responsible for particle internalization. The two-dimensional model (2D) of cell culture is overly simplistic to mimic the complexity of cancer tissues that exist in vivo. It cannot simulate the critical tissue-specific properties like cell-cell interaction and cell-extracellular matrix (ECM) interaction and its influences on the temporal and spatial distribution of nanoparticles (NPs). The three dimensional model organization of heterogenous cancer and normal cells with the ECM acts as a formidable barrier to NP penetration and cellular uptake. The three dimensional cell culture (3D) technology is a breakthrough in this direction that can mimic the barrier properties of the tumor microenvironment (TME). Herein, we discuss the physiological factors that should be considered to bridge the translational gap between in and vitro cell culture studies and in-vivo studies in cancer nanomedicine.
Collapse
Affiliation(s)
- Yamini Boinapalli
- Dept. of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - Ravi Shankar Pandey
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, C.G. 495009, India
| | - Abhay Singh Chauhan
- Department of Biopharmaceutical Sciences, School of Pharmacy, Medical College of Wisconsin, Milwaukee, WI 53226, United States.
| | - M S Sudheesh
- Dept. of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India.
| |
Collapse
|
9
|
Pershina AG, Demin AM, Perekucha NA, Brikunova OY, Efimova LV, Nevskaya KV, Vakhrushev AV, Zgoda VG, Uimin MA, Minin AS, Malkeyeva D, Kiseleva E, Zima AP, Krasnov VP, Ogorodova LM. Peptide ligands on the PEGylated nanoparticle surface and human serum composition are key factors for the interaction between immune cells and nanoparticles. Colloids Surf B Biointerfaces 2023; 221:112981. [DOI: 10.1016/j.colsurfb.2022.112981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
|
10
|
Domingues C, Santos A, Alvarez-Lorenzo C, Concheiro A, Jarak I, Veiga F, Barbosa I, Dourado M, Figueiras A. Where Is Nano Today and Where Is It Headed? A Review of Nanomedicine and the Dilemma of Nanotoxicology. ACS NANO 2022; 16:9994-10041. [PMID: 35729778 DOI: 10.1021/acsnano.2c00128] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Worldwide nanotechnology development and application have fueled many scientific advances, but technophilic expectations and technophobic demands must be counterbalanced in parallel. Some of the burning issues today are the following: (1) Where is nano today? (2) How good are the communication and investment networks between academia/research and governments? (3) Is there any spotlight application for nanotechnology? Nanomedicine is a particular arm of nanotechnology within the healthcare landscape, focused on diagnosis, treatment, and monitoring of emerging (such as coronavirus disease 2019, COVID-19) and contemporary (including diabetes, cardiovascular diseases, neurodegenerative disorders, and cancer) diseases. However, it may only represent the bright side of the coin. In fact, in the recent past, the concept of nanotoxicology has emerged to address the dark shadows of nanomedicine. The nanomedicine field requires more nanotoxicological studies to identify undesirable effects and guarantee safety. Here, we provide an overall perspective on nanomedicine and nanotoxicology as central pieces of the giant puzzle of nanotechnology. First, the impact of nanotechnology on education and research is highlighted, followed by market trends and scientific output tendencies. In the next section, the nanomedicine and nanotoxicology dilemma is addressed through the interplay of in silico, in vitro, and in vivo models with the support of omics and microfluidic approaches. Lastly, a reflection on the regulatory issues and clinical trials is provided. Finally, some conclusions and future perspectives are proposed for a clearer and safer translation of nanomedicines from the bench to the bedside.
Collapse
Affiliation(s)
- Cátia Domingues
- Univ. Coimbra, Faculty of Pharmacy, Galenic and Pharmaceutical Technology Laboratory, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Galenic and Pharmaceutical Technology Laboratory, Faculty of Pharmacy, Univ. Coimbra, 3000-548 Coimbra, Portugal
- Univ. Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, 3000-548 Coimbra, Portugal
| | - Ana Santos
- Univ. Coimbra, Faculty of Pharmacy, Galenic and Pharmaceutical Technology Laboratory, 3000-548 Coimbra, Portugal
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, iMATUS, and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, iMATUS, and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ivana Jarak
- Univ. Coimbra, Faculty of Pharmacy, Galenic and Pharmaceutical Technology Laboratory, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Univ. Coimbra, Faculty of Pharmacy, Galenic and Pharmaceutical Technology Laboratory, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Galenic and Pharmaceutical Technology Laboratory, Faculty of Pharmacy, Univ. Coimbra, 3000-548 Coimbra, Portugal
| | - Isabel Barbosa
- Univ. Coimbra, Faculty of Pharmacy, Phamaceutical Chemistry Laboratory, 3000-548 Coimbra, Portugal
| | - Marília Dourado
- Univ. Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, 3000-548 Coimbra, Portugal
- Univ. Coimbra, Center for Health Studies and Research of the University of Coimbra (CEISUC), Faculty of Medicine, 3000-548 Coimbra, Portugal
- Univ. Coimbra, Center for Studies and Development of Continuous and Palliative Care (CEDCCP), Faculty of Medicine, 3000-548 Coimbra, Portugal
| | - Ana Figueiras
- Univ. Coimbra, Faculty of Pharmacy, Galenic and Pharmaceutical Technology Laboratory, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Galenic and Pharmaceutical Technology Laboratory, Faculty of Pharmacy, Univ. Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
11
|
Haroon H, Hunter A, Farhangrazi Z, Moghimi S. A brief history of long circulating nanoparticles. Adv Drug Deliv Rev 2022; 188:114396. [DOI: 10.1016/j.addr.2022.114396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 12/21/2022]
|
12
|
Sudheesh MS, Pavithran K, M S. Revisiting the outstanding questions in cancer nanomedicine with a future outlook. NANOSCALE ADVANCES 2022; 4:634-653. [PMID: 36131837 PMCID: PMC9418065 DOI: 10.1039/d1na00810b] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/22/2021] [Indexed: 06/01/2023]
Abstract
The field of cancer nanomedicine has been fueled by the expectation of mitigating the inefficiencies and life-threatening side effects of conventional chemotherapy. Nanomedicine proposes to utilize the unique nanoscale properties of nanoparticles to address the most pressing questions in cancer treatment and diagnosis. The approval of nano-based products in the 1990s inspired scientific explorations in this direction. However, despite significant progress in the understanding of nanoscale properties, there are only very few success stories in terms of substantial increase in clinical efficacy and overall patient survival. All existing paradigms such as the concept of enhanced permeability and retention (EPR), the stealth effect and immunocompatibility of nanomedicine have been questioned in recent times. In this review we critically examine impediments posed by biological factors to the clinical success of nanomedicine. We put forth current observations on critical outstanding questions in nanomedicine. We also provide the promising side of cancer nanomedicine as we move forward in nanomedicine research. This would provide a future direction for research in nanomedicine and inspire ongoing investigations.
Collapse
Affiliation(s)
- M S Sudheesh
- Dept. of Pharmaceutics, Amrita School of Pharmacy Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara Kochi - 682041 India +91-9669372019
| | - K Pavithran
- Department of Medical Oncology, Amrita Institute of Medial Sciences and Research Centre Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara Kochi - 682041 India
| | - Sabitha M
- Dept. of Pharmaceutics, Amrita School of Pharmacy Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara Kochi - 682041 India +91-9669372019
| |
Collapse
|
13
|
Rubey KM, Mukhitov AR, Nong J, Wu J, Krymskaya VP, Myerson JW, Worthen GS, Brenner JS. Nanoparticle-Induced Augmentation of Neutrophils' Phagocytosis of Bacteria. Front Pharmacol 2022; 13:923814. [PMID: 35860017 PMCID: PMC9289463 DOI: 10.3389/fphar.2022.923814] [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: 04/19/2022] [Accepted: 05/31/2022] [Indexed: 11/21/2022] Open
Abstract
Despite the power of antibiotics, bacterial infections remain a major killer, due to antibiotic resistance and hosts with dysregulated immune systems. We and others have been developing drug-loaded nanoparticles that home to the sites of infection and inflammation via engineered tropism for neutrophils, the first-responder leukocytes in bacterial infections. Here, we examined how a member of a broad class of neutrophil-tropic nanoparticles affects neutrophil behavior, specifically questioning whether the nanoparticles attenuate an important function, bacterial phagocytosis. We found these nanoparticles actually augment phagocytosis of non-opsonized bacteria, increasing it by ∼50%. We showed this augmentation of phagocytosis is likely co-opting an evolved response, as opsonized bacteria also augment phagocytosis of non-opsonized bacteria. Enhancing phagocytosis of non-opsonized bacteria may prove particularly beneficial in two clinical situations: in hypocomplementemic patients (meaning low levels of the main bacterial opsonins, complement proteins, seen in conditions such as neonatal sepsis and liver failure) or for bacteria that are largely resistant to complement opsonization (e.g., Neisseria). Additionally, we observe that; 1) prior treatment with bacteria augments neutrophil uptake of neutrophil-tropic nanoparticles; 2) neutrophil-tropic nanoparticles colocalize with bacteria inside of neutrophils. The observation that neutrophil-tropic nanoparticles enhance neutrophil phagocytosis and localize with bacteria inside neutrophils suggests that these nanoparticles will serve as useful carriers for drugs to ameliorate bacterial diseases.
Collapse
Affiliation(s)
- Kathryn M Rubey
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Alexander R Mukhitov
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jia Nong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| | - Jichuan Wu
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| | - Vera P Krymskaya
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jacob W Myerson
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| | - G Scott Worthen
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jacob S Brenner
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
14
|
Zhang W, Gaikwad H, Groman EV, Purev E, Simberg D, Wang G. Highly aminated iron oxide nanoworms for simultaneous manufacturing and labeling of chimeric antigen receptor T cells. JOURNAL OF MAGNETISM AND MAGNETIC MATERIALS 2022; 541:168480. [PMID: 34720339 PMCID: PMC8553019 DOI: 10.1016/j.jmmm.2021.168480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Cell based therapies including chimeric antigen receptor (CAR) T cells are promising for treating leukemias and solid cancers. At the same time, there is interest in enhancing the functionality of these cells via surface decoration with nanoparticles (backpacking). Magnetic nanoparticle cell labeling is of particular interest due to opportunities for magnetic separation, in vivo manipulation, drug delivery and magnetic resonance imaging (MRI). While modification of T cells with magnetic nanoparticles (MNPs) was explored before, we questioned whether MNPs are compatible with CAR-T cells when introduced during the manufacturing process. We chose highly aminated 120 nm crosslinked iron oxide nanoworms (CLIO NWs, ~36,000 amines per NW) that could efficiently label different adherent cell lines and we used CD123 CAR-T cells as the labeling model. The CD123 CAR-T cells were produced in the presence of CLIO NWs, CLIO NWs plus protamine sulfate (PS), or PS only. The transduction efficiency of lentiviral CD123 CAR with only NWs was ~23% lower than NW+PS and PS groups (~33% and 35%, respectively). The cell viability from these three transduction conditions was not reduced within CAR-T cell groups, though lower compared to non-transduced T cells (mock T). Use of CLIO NWs instead of, or together with cationic protamine sulfate for enhancement of lentiviral transduction resulted in comparable levels of CAR expression and viability but decreased the proportion of CD8+ cells and increased the proportion of CD4+ cells. CD123 CAR-T transduced in the presence of CLIO NWs, CLIO NWs plus PS, or PS only, showed similar level of cytotoxicity against leukemic cell lines. Furthermore, fluorescence microscopy imaging demonstrated that CD123 CAR-T cells labeled with CLIO NW formed rosettes with CD123+ leukemic cells as the non-labeled CAR-T cells, indicating that the CAR-T targeting to tumor cells has maintained after CLIO NW labeling. The in vivo trafficking of the NW labeled CAR-T cells showed the accumulation of CAR-T labeled with NWs primarily in the bone marrow and spleen. CAR-T cells can be magnetically labeled during their production while maintaining functionality using the positively charged iron oxide NWs, which enable the in vivo biodistribution and tracking of CAR-T cells.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Hanmant Gaikwad
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ernest V. Groman
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Enkhtsetseg Purev
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Corresponding Authors: (Dmitri Simberg), (Guankui Wang)
| | - Guankui Wang
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Corresponding Authors: (Dmitri Simberg), (Guankui Wang)
| |
Collapse
|
15
|
Stater EP, Sonay AY, Hart C, Grimm J. The ancillary effects of nanoparticles and their implications for nanomedicine. NATURE NANOTECHNOLOGY 2021; 16:1180-1194. [PMID: 34759355 PMCID: PMC9031277 DOI: 10.1038/s41565-021-01017-9] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/22/2021] [Indexed: 05/12/2023]
Abstract
Nanoparticles are often engineered as a scaffolding system to combine targeting, imaging and/or therapeutic moieties into a unitary agent. However, mostly overlooked, the nanomaterial itself interacts with biological systems exclusive of application-specific particle functionalization. This nanoparticle biointerface has been found to elicit specific biological effects, which we term 'ancillary effects'. In this Review, we describe the current state of knowledge of nanobiology gleaned from existing studies of ancillary effects with the objectives to describe the potential of nanoparticles to modulate biological effects independently of any engineered function; evaluate how these effects might be relevant for nanomedicine design and functional considerations, particularly how they might be useful to inform clinical decision-making; identify potential clinical harm that arises from adverse nanoparticle interactions with biology; and, finally, highlight the current lack of knowledge in this area as both a barrier and an incentive to the further development of nanomedicine.
Collapse
Affiliation(s)
- Evan P Stater
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Ali Y Sonay
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cassidy Hart
- Department of General Surgery, Lankenau Medical Center, Wynnewood, PA, USA
| | - Jan Grimm
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
16
|
Maisha N, Naik N, Okesola M, Coombs T, Zilberberg R, Pandala N, Lavik E. Engineering PEGylated Polyester Nanoparticles to Reduce Complement-Mediated Infusion Reaction. Bioconjug Chem 2021; 32:2154-2166. [PMID: 34499487 DOI: 10.1021/acs.bioconjchem.1c00339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Translation of intravenously administered nanomaterials to the clinic is limited due to adverse infusion reactions. While these reactions are infrequent, with up to 10% prone to experiencing infusion reactions, the reactions can be severe and life-threatening. One of the innate immune pathways, the complement activation pathway, plays a significant role in mediating this response. Nanoparticle surface properties are a relevant design feature, as they control the blood proteins the nanoparticles interact with and allow the nanoparticles to evade the immune reaction. PEGylation of nanosurfaces is critical in improving the blood circulation of nanoparticles and reducing opsonization. Our goal was to understand whether modifying the surface architecture by varying the PEG density and architecture can impact the complement response in vitro. We utilized block copolymers of poly(lactic acid)-b-poly(ethylene glycol) prepared with poly(ethylene glycol) macroinitiators of molecular weights 3400 and 5000 Da. Tracking the complement biomarker C5a, we monitored the impact of changing PEGylation of the nanoparticles. We also investigated how the changing PEG length on the nanoparticle surface impacts further strengthening the stealth properties. Lastly, we determined which cytokines change upon blood incubation with nanoparticles in vitro to understand the extent to which inflammation may occur and the crosstalk between the complement and immune responses. Increasing PEGylation reduced the generation of complement-mediated anaphylatoxin C5a in vitro, with 5000 Da PEG more effectively reducing levels of C5a generated compared to 3400 Da PEG. The insights gathered regarding the impact of PEG density and PEG chain length would be critical in developing stealth nanoparticles that do not lead to infusion reactions upon intravenous administration.
Collapse
Affiliation(s)
- Nuzhat Maisha
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Nidhi Naik
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Mawuyon Okesola
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Tobias Coombs
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Rose Zilberberg
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Narendra Pandala
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Erin Lavik
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| |
Collapse
|
17
|
Li Y, Wang G, Griffin L, Banda NK, Saba LM, Groman EV, Scheinman R, Moghimi SM, Simberg D. Complement opsonization of nanoparticles: Differences between humans and preclinical species. J Control Release 2021; 338:548-556. [PMID: 34481928 DOI: 10.1016/j.jconrel.2021.08.048] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 12/19/2022]
Abstract
The complement system plays a key role in opsonization and immune clearance of engineered nanoparticles. Understanding the efficiency, inter-subject, and inter-strain differences of complement opsonization in preclinical species can help with translational nanomedicine development and improve our ability to model complement response in humans. Dextran-coated superparamagnetic iron oxide (SPIO) nanoparticles and a wide range of non-magnetic iron oxide nanoparticle formulations are widely used in magnetic resonance imaging and as clinically approved iron supplements. Previously we found that opsonization of SPIO nanoworms (NW) with the third complement protein (C3) proceeds mostly via the alternative pathway in humans, and via the lectin pathway in mice. Here, we studied the pathway and efficiency of opsonization of 106 nm SPIO NW with C3 in different preclinical species and commonly used laboratory strains. In sera of healthy human donors (n = 6), C3 opsonization proceeded exclusively through the alternative pathway. On the other hand, the C3 opsonization in dogs (6 breeds), rats (4 strains) and mice (5 strains) sera was either partially or completely dependent on the complement Ca2+-sensitive pathways (lectin and/or classical). Specifically, C3 opsonization in sera of Long Evans rat strain, and mouse strains widely used in nanomedicine research (BALB/c, C57BL/6 J, and A/J) was only through the Ca2+-dependent pathways. Dogs and humans had the highest between-subject variability in C3 opsonization levels, while rat and mouse sera showed the lowest between-strain variability. Furthermore, using a panel of SPIO nanoparticles of different sizes and dextran coatings, we found that the level of C3 opsonization (C3 molecules per milligram Fe) in human sera was lower than in animal sera. At the same time, there was a strong predictive value of complement opsonization in dog and rat sera; nanoparticles with higher C3 deposition in animals showed higher deposition in humans, and vice versa. Notably, the opsonization decreased with decreasing size in all sera. The studies highlight the importance of the consideration of species and strains for predicting human complement responses (opsonization) towards nanomedicines.
Collapse
Affiliation(s)
- Yue Li
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Guankui Wang
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lynn Griffin
- Department of Environmental and Radiological Health Sciences, Veterinary Teaching Hospital, Colorado State University, Fort Collins, CO, USA
| | - Nirmal K Banda
- Division of Rheumatology, School of Medicine, University of Colorado Anschutz Medical Campus, 1775 Aurora Court, Aurora, CO, USA
| | - Laura M Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ernest V Groman
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert Scheinman
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - S Moein Moghimi
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; School of Pharmacy, King George VI Building, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Translational and Clinical Research Institute, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
18
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxon CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Physiological and Pathological Factors Affecting Drug Delivery to the Brain by Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002085. [PMID: 34105297 PMCID: PMC8188209 DOI: 10.1002/advs.202002085] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/06/2021] [Indexed: 05/04/2023]
Abstract
The prevalence of neurological/neurodegenerative diseases, such as Alzheimer's disease is known to be increasing due to an aging population and is anticipated to further grow in the decades ahead. The treatment of brain diseases is challenging partly due to the inaccessibility of therapeutic agents to the brain. An increasingly important observation is that the physiology of the brain alters during many brain diseases, and aging adds even more to the complexity of the disease. There is a notion that the permeability of the blood-brain barrier (BBB) increases with aging or disease, however, the body has a defense mechanism that still retains the separation of the brain from harmful chemicals in the blood. This makes drug delivery to the diseased brain, even more challenging and complex task. Here, the physiological changes to the diseased brain and aged brain are covered in the context of drug delivery to the brain using nanoparticles. Also, recent and novel approaches are discussed for the delivery of therapeutic agents to the diseased brain using nanoparticle based or magnetic resonance imaging guided systems. Furthermore, the complement activation, toxicity, and immunogenicity of brain targeting nanoparticles as well as novel in vitro BBB models are discussed.
Collapse
Affiliation(s)
- Yamir Islam
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Andrew G. Leach
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- Division of Pharmacy and OptometryThe University of ManchesterStopford Building, Oxford RoadManchesterM13 9PTUK
| | - Jayden Smith
- Cambridge Innovation Technologies Consulting (CITC) LimitedSt. John's Innovation CentreCowley RoadCambridgeCB4 0WSUK
| | - Stefano Pluchino
- Department of Clinical NeurosciencesClifford Allbutt Building – Cambridge Biosciences Campus and NIHR Biomedical Research CentreUniversity of CambridgeHills RoadCambridgeCB2 0HAUK
| | - Christopher R. Coxon
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- School of Engineering and Physical SciencesHeriot‐Watt UniversityWilliam Perkin BuildingEdinburghEH14 4ASUK
| | - Muttuswamy Sivakumaran
- Department of HaematologyPeterborough City HospitalEdith Cavell CampusBretton Gate PeterboroughPeterboroughPE3 9GZUK
| | - James Downing
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Amos A. Fatokun
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Meritxell Teixidò
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 10Barcelona08028Spain
| | - Touraj Ehtezazi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| |
Collapse
|
19
|
Mechanistic investigation of thermosensitive liposome immunogenicity and understanding the drivers for circulation half-life: A polyethylene glycol versus 1,2-dipalmitoyl-sn-glycero-3-phosphodiglycerol study. J Control Release 2021; 333:1-15. [DOI: 10.1016/j.jconrel.2021.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
|
20
|
Viana IMDO, Roussel S, Defrêne J, Lima EM, Barabé F, Bertrand N. Innate and adaptive immune responses toward nanomedicines. Acta Pharm Sin B 2021; 11:852-870. [PMID: 33747756 PMCID: PMC7955583 DOI: 10.1016/j.apsb.2021.02.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/04/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
Since the commercialization of the first liposomes used for drug delivery, Doxil/Caelyx® and Myocet®, tremendous progress has been made in understanding interactions between nanomedicines and biological systems. Fundamental work at the interface of engineering and medicine has allowed nanomedicines to deliver therapeutic small molecules and nucleic acids more efficiently. While nanomedicines are used in oncology for immunotherapy or to deliver combinations of cytotoxics, the clinical successes of gene silencing approaches like patisiran lipid complexes (Onpattro®) have paved the way for a variety of therapies beyond cancer. In parallel, the global severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has highlighted the potential of mRNA vaccines to develop immunization strategies at unprecedented speed. To rationally design therapeutic and vaccines, chemists, materials scientists, and drug delivery experts need to better understand how nanotechnologies interact with the immune system. This review presents a comprehensive overview of the innate and adaptative immune systems and emphasizes the intricate mechanisms through which nanomedicines interact with these biological functions.
Collapse
|
21
|
Zhang Y, Xia M, Zhou Z, Hu X, Wang J, Zhang M, Li Y, Sun L, Chen F, Yu H. p53 Promoted Ferroptosis in Ovarian Cancer Cells Treated with Human Serum Incubated-Superparamagnetic Iron Oxides. Int J Nanomedicine 2021; 16:283-296. [PMID: 33469287 PMCID: PMC7811475 DOI: 10.2147/ijn.s282489] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/25/2020] [Indexed: 12/25/2022] Open
Abstract
Methods In this study, we used MTT assays to demonstrate that a combination of SPIO-Serum and wild-type p53 overexpression can reduce ovarian cancer cell viability in vitro. Prussian blue staining and iron assays were used to determine changes in intracellular iron concentration following SPIO-Serum treatment. TEM was used to evaluate any mitochondrial damage induced by SPIO-Serum treatment, and Western blot was used to evaluate the expression of the iron transporter and lipid peroxidation regulator proteins. JC-1 was used to measure mitochondrial membrane potential, and ROS levels were estimated by flow cytometry. Finally, xCT protein expression and mitochondrial ROS levels were confirmed using fluorescence microscopy. Results SPIO-Serum effectively induced lipid peroxidation and generated abundant toxic ROS. It also facilitated the downregulation of GPX4 and xCT, ultimately resulting in iron-dependent oxidative death. These effects could be reversed by iron chelator DFO and lipid peroxidation inhibitor Fer-1. SPIO-Serum treatment disrupted intracellular iron homeostasis by regulating iron uptake and the cells presented with missing mitochondrial cristae and ruptured outer mitochondrial membranes. Moreover, we were able to show that p53 contributed to SPIO-Serum-induced ferroptosis in ovarian cancer cells. Conclusion SPIO-Serum induced ferroptosis and overexpressed p53 contributed to ferroptosis in ovarian cancer cells. Our data provide a theoretical basis for ferroptosis as a novel cell death phenotype induced by nanomaterials.
Collapse
Affiliation(s)
- Yunhan Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Meihui Xia
- Department of Obstetrics & Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
| | - Zizhen Zhou
- Clinical Medical College, Jilin University, Changchun 130021, People's Republic of China
| | - Xiaoqing Hu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Jiabin Wang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Meiyu Zhang
- Clinical Medical College, Jilin University, Changchun 130021, People's Republic of China
| | - Yi Li
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun, Jilin 130012, People's Republic of China
| | - Liankun Sun
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Fangfang Chen
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun, Jilin 130012, People's Republic of China.,Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Huimei Yu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, People's Republic of China.,Animal Experiment Center, College of Basic Medical Sciences, Jilin University, Changchun 130021, People's Republic of China
| |
Collapse
|
22
|
Brownian motion-based nanoparticle sizing-A powerful approach for in situ analysis of nanoparticle-protein interactions. Biointerphases 2020; 15:061201. [PMID: 33356335 DOI: 10.1116/6.0000438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A key hurdle toward effective application of nanoparticles (NPs) in biomedicine is still the incomplete understanding of the biomolecular adsorption layer, the so-called protein corona, which inevitably forms around NPs when they are immersed in a biofluid. NP sizing techniques via the analysis of Brownian motions offer a powerful way to measure the thickness of the protein corona in situ. Here, the fundamentals of three techniques, dynamic light scattering, fluorescence correlation spectroscopy, and nanoparticle tracking analysis are briefly summarized. Then, experimental procedures for the determination of binding curves are presented in a tutorial fashion. Nanoparticle sizing experiments are illustrated with a selection of recent results on the interactions of transferrin with hydrophilic and hydrophobic polystyrene nanoparticles, and key insights gained from this work are discussed.
Collapse
|
23
|
Papini E, Tavano R, Mancin F. Opsonins and Dysopsonins of Nanoparticles: Facts, Concepts, and Methodological Guidelines. Front Immunol 2020; 11:567365. [PMID: 33154748 PMCID: PMC7587406 DOI: 10.3389/fimmu.2020.567365] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/25/2020] [Indexed: 11/13/2022] Open
Abstract
Understanding the effects mediated by a set of nanoparticle (NP)-bound host biomolecules, often indicated with the umbrella term of NP corona, is essential in nanomedicine, nanopharmacology, and nanotoxicology. Among the NP-adsorbed proteome, some factors mediate cell binding, endocytosis, and clearing by macrophages and other phagocytes (opsonins), while some others display few affinities for the cell surface (dysopsonins). The functional mapping of opsonins and dysopsonins is instrumental to design long-circulating and nanotoxicologically safe next-generation nanotheranostics. In this review, we critically analyze functional data identifying specific proteins with opsonin or dysopsonin properties. Special attention is dedicated to the following: (1) the simplicity or complexity of the NP proteome and its modulation, (2) the role of specific host proteins in mediating the stealth properties of uncoated or polymer-coated NPs, and (3) the ability of the innate immune system, and, in particular, of the complement proteins, to mediate NP clearance by phagocytes. Emerging species-specific peculiarities, differentiating humans from preclinical animal models (the murine especially), are highlighted throughout this overview. The operative definition of opsonin and dysopsonin and the measurement schemes to assess their in vitro efficacy is critically re-examined. This provides a shared and unbiased approach useful for NP opsonin and dysopsonin systematic identification.
Collapse
Affiliation(s)
- Emanuele Papini
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Centre for Innovative Biotechnological Research, University of Padua, Padua, Italy
| | - Regina Tavano
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Centre for Innovative Biotechnological Research, University of Padua, Padua, Italy
| | - Fabrizio Mancin
- Department of Chemical Sciences, University of Padua, Padua, Italy
| |
Collapse
|
24
|
Polymer-coated nanoparticle protein corona formation potentiates phagocytosis of bacteria by innate immune cells and inhibits coagulation in human plasma. Biointerphases 2020; 15:051003. [DOI: 10.1116/6.0000385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
25
|
A structurally diverse library of glycerol monooleate/oleic acid non-lamellar liquid crystalline nanodispersions stabilized with nonionic methoxypoly(ethylene glycol) (mPEG)-lipids showing variable complement activation properties. J Colloid Interface Sci 2020; 582:906-917. [PMID: 32919118 DOI: 10.1016/j.jcis.2020.08.085] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022]
Abstract
Pluronic F127-stabilized non-lamellar liquid crystalline aqueous nanodispersions are promising injectable platforms for drug and contrast agent delivery. These nanodispersions, however, trigger complement activation in the human blood, where the extent of complement activation and opsonization processes may compromise their biological performance and safety. Here, we introduce a broad family of nanodispersions from glycerol monooleate (GMO) and oleic acid (OA) in different weight ratios, and stabilized with a plethora of nonionic methoxypoly(ethylene glycol) (mPEG)-lipids of different PEG chain length and variable lipid moiety (monounsaturated or saturated diglycerides or D-α-tocopheryl succinate). Through an integrated biophysical approach involving dynamic light scattering, synchrotron small-angle scattering, and cryo-transmission electron microscopy, we examine the impact of nonionic mPEG-lipid stabilization on size, internal self-assembled architecture, and gross morphological characteristics of nanodispersions. The results show how the nonionic mPEG-lipid type and concentration, and dependent on GMO/OA weight ratio, can variably modulate the internal architectures of nanoparticles. Assessment of complement profiling from selected nanodispersions with diverse structural heterogeneity further suggests a variable modulatory role for the lipid type of the nonionic mPEG-lipid in the extent of complement activation, which span from no activation to moderate to high levels. We comment on plausible mechanisms driving the observed complement activation variability and discuss the potential utility of these nanodispersions for future development of injectable nanopharmaceuticals.
Collapse
|
26
|
Yang C, Chen F, Ren P, Lofchy L, Wan C, Shen J, Wang G, Gaikwad H, Ponder J, Jordan CT, Scheinman R, Simberg D. Delivery of a model lipophilic membrane cargo to bone marrow via cell-derived microparticles. J Control Release 2020; 326:324-334. [PMID: 32682903 DOI: 10.1016/j.jconrel.2020.07.019] [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] [Received: 05/05/2020] [Revised: 06/26/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022]
Abstract
Bone marrow (BM) is the central immunological organ and the origin of hematological diseases. Efficient and specific drug delivery to the BM is an unmet need. We tested delivery of fluorescent indocarbocyanine lipids (ICLs, DiR, DiD, DiI) as a model lipophilic cargo, via different carriers. Systemically injected T-lymphocyte cell line Jurkat delivered ICLs to the BM more efficiently than erythrocytes, and more selectively than PEGylated liposomes. Near infrared imaging showed that the delivery was restricted to the BM, lungs, liver and spleen, with no accumulation in the kidneys, brain, heart, intestines, fat tissue and pancreas. Following systemic injection of ICL-labeled cells in immunodeficient or immunocompetent mice, few cells arrived in the BM intact. However, between 5 and 10% of BM cells were ICL-positive. Confocal microscopy of intact BM confirmed that ICLs are delivered independently of the injected cells. Flow cytometry analysis showed that the lipid accumulated in both CD11b + and CD11b- cells, and in hematopoietic progenitors. In a xenograft model of acute myeloid leukemia, a single injection of 10 million Jurkat cells delivered DiD to ~15% of the tumor cells. ICL-labeled cells disappeared from blood almost immediately post-intravenous injection, but numerous cell-derived microparticles continued to circulate in blood. The microparticle particle formation was not due to the ICL labeling or complement attack and was observed after injection of both syngeneic and xenogeneic cells. Injection of microparticles produced in vitro from Jurkat cells resulted in a similar ICL delivery as the injection of intact Jurkat cells. Our results demonstrate a novel delivery paradigm wherein systemically injected cells release microparticles that accumulate in the BM. In addition, the results have important implications for studies involving systemically administered cell therapies.
Collapse
Affiliation(s)
- Chunyan Yang
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, China; The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Fangfang Chen
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, China; The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Ping Ren
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Laren Lofchy
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, CO 80309, USA
| | - Jingshi Shen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, CO 80309, USA
| | - Guankui Wang
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hanmant Gaikwad
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jessica Ponder
- Division of Hematology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Craig T Jordan
- Division of Hematology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Robert Scheinman
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dmitri Simberg
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
27
|
Gaikwad H, Li Y, Gifford G, Groman E, Banda NK, Saba L, Scheinman R, Wang G, Simberg D. Complement Inhibitors Block Complement C3 Opsonization and Improve Targeting Selectivity of Nanoparticles in Blood. Bioconjug Chem 2020; 31:1844-1856. [PMID: 32598839 DOI: 10.1021/acs.bioconjchem.0c00342] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Complement is one of the critical branches of innate immunity that determines the recognition of engineered nanoparticles by immune cells. Antibody-targeted iron oxide nanoparticles are a popular platform for magnetic separations, in vitro diagnostics, and molecular imaging. We used 60 nm cross-linked iron oxide nanoworms (CLIO NWs) modified with antibodies against Her2/neu and EpCAM, which are common markers of blood-borne cancer cells, to understand the role of complement in the selectivity of targeting of tumor cells in whole blood. CLIO NWs showed highly efficient targeting and magnetic isolation of tumor cells spiked in lepirudin-anticoagulated blood, but specificity was low due to high uptake by neutrophils, monocytes, and lymphocytes. Complement C3 opsonization in plasma was predominantly via the alternative pathway regardless of the presence of antibody, PEG, or fluorescent tag, but was higher for antibody-conjugated CLIO NWs. Addition of various soluble inhibitors of complement convertase (compstatin, soluble CD35, and soluble CD55) to whole human blood blocked up to 99% of the uptake of targeted CLIO NWs by leukocytes, which resulted in a more selective magnetic isolation of tumor cells. Using well-characterized nanomaterials, we demonstrate here that complement therapeutics can be used to improve targeting selectivity.
Collapse
Affiliation(s)
| | | | | | | | - Nirmal K Banda
- Division of Rheumatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | | | | | | | | |
Collapse
|
28
|
Moghimi SM, Simberg D, Papini E, Farhangrazi ZS. Complement activation by drug carriers and particulate pharmaceuticals: Principles, challenges and opportunities. Adv Drug Deliv Rev 2020; 157:83-95. [PMID: 32389761 DOI: 10.1016/j.addr.2020.04.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/16/2022]
Abstract
Considering the multifaceted protective and homeostatic roles of the complement system, many consequences arise when drug carriers, and particulate pharmaceutical formulations clash with complement proteins, and trigger complement cascade. Complement activation may induce formulation destabilization, promote opsonization, and affect biological and therapeutic performance of pharmaceutical nano- and micro-particles. In some cases, complement activation is beneficial, where complement may play a role in prophylactic protection, whereas uncontrolled complement activation is deleterious, and contributes to disease progression. Accordingly, design initiatives with particulate medicines should consider complement activation properties of the end formulation within the context of administration route, dosing, systems biology, and therapeutic perspective. Here we examine current progress in mechanistic processes underlying complement activation by pre-clinical and clinical particles, identify opportunities and challenges ahead, and suggest future directions in nanomedicine-complement interface research.
Collapse
Affiliation(s)
- S Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Colorado Center for Nanomedicine and Nanosafety, Skagg's School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Dmitri Simberg
- Colorado Center for Nanomedicine and Nanosafety, Skagg's School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emanuele Papini
- Department of Biomedical Sciences, University of Padua, Padua 35121, Italy; CRIBI Biotechnology Center, University of Padua, Padua 35121, Italy
| | - Z Shadi Farhangrazi
- S. M. Discovery Group Inc., Denver, CO, USA; S. M. Discovery Group Ltd., Durham, UK
| |
Collapse
|
29
|
Liu Y, Zhao J, Jiang J, Chen F, Fang X. Doxorubicin Delivered Using Nanoparticles Camouflaged with Mesenchymal Stem Cell Membranes to Treat Colon Cancer. Int J Nanomedicine 2020; 15:2873-2884. [PMID: 32368059 PMCID: PMC7185325 DOI: 10.2147/ijn.s242787] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/06/2020] [Indexed: 12/18/2022] Open
Abstract
PURPOSE The primary goal of the present study was to design doxorubicin (DOX)-loaded superparamagnetic iron oxide (SPIO) nanoparticles (NPs) coated with mesenchymal stem cell (MSC) membranes and explore their effect on colon cancer in vitro and in vivo. METHODS DOX-SPIO NPs were coated with MSC membranes using an extruder, and the morphological characteristics of MSC membrane-camouflaged nanodrug (DOX-SPIO@MSCs) evaluated by transmission electron microscopy (TEM) and NP-tracking analysis. Drug loading and pH response were assessed by UV spectrophotometry. Intracellular colocalization was analyzed using NP-treated MC38 cells stained with 3,3'-dioctadecyloxacarbocyanine perchlorate and Hoechst 33342. Cellular uptake was analyzed using an inverted fluorescence microscope and flow cytometry and cytotoxicity evaluated by cell counting kit-8 assay. Biological compatibility was assessed by hemolysis analysis, immunoactivation test and leukocyte uptake experiments. Furthermore, intravenous injection of chemotherapy drugs into MC38 tumor-bearing C57BL/6 mice was used to study anti-tumor effects. RESULTS Typical core-shell NP structures were observed by TEM. Particle size remained stable in fetal bovine serum and phosphate-buffered saline (PBS). Compared with DOX-SPIO, DOX-SPIO@MSCs improved cellular uptake efficiency, enhanced anti-tumor effects, and reduced the immune system response. Animal experiments demonstrated that DOX-SPIO@MSCs enhanced tumor treatment efficacy while reducing systemic side effects. CONCLUSION Our experimental results demonstrate that DOX-SPIO@MSCs are a promising targeted nanocarrier for application in treatment of colon cancer.
Collapse
Affiliation(s)
- Yi Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin130033, People’s Republic of China
| | - Jingtong Zhao
- Department of Central Laboratory, China-Japan Union Hospital, Jilin University, Changchun, Jilin130033, People’s Republic of China
| | - Jinlan Jiang
- Department of Central Laboratory, China-Japan Union Hospital, Jilin University, Changchun, Jilin130033, People’s Republic of China
| | - Fangfang Chen
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin130033, People’s Republic of China
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun, Jilin130012, People’s Republic of China
- Key Laboratory of Zoonoses Research, Ministry of Education, Jilin University, Changchun, Jilin130062, People’s Republic of China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin130033, People’s Republic of China
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun, Jilin130012, People’s Republic of China
| |
Collapse
|
30
|
Neutrophil-mediated transport is crucial for delivery of short-circulating magnetic nanoparticles to tumors. Acta Biomater 2020; 104:176-187. [PMID: 31945505 DOI: 10.1016/j.actbio.2020.01.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/20/2019] [Accepted: 01/09/2020] [Indexed: 12/31/2022]
Abstract
Recently neutrophil-based nanoparticles (NPs) drug delivery systems have gained considerable attention in cancer therapy. Numerous studies have been conducted to identify optimal NPs parameters for passive tumor targeting, while there is a fundamental dearth of knowledge about the factors governing cell-mediated delivery. Here, by using intravital microscopy and magnetic resonance imaging, we describe accumulation dynamics of 140 nm magnetic cubes and clusters in murine breast cancer (4T1) and colon cancer (CT26) models. Notwithstanding rapid clearance from the blood flow, NPs readily accumulated in tumors at later time points. Both NPs types were captured mostly by intravascular neutrophils immediately after injection, and transmigration of NPs-bound neutrophils through the vessel wall was first shown in real-time. A dramatic drop in NPs accumulation upon Ly6G and Gr1 depletion further confirmed the role of neutrophils as a biocarrier for targeting tumors. Of note, for shorter circulating NPs, a cell-dependent delivery route was more impactful, while the accumulation of longer circulating counterpart was less compromised by neutrophil depletion. Neutrophil-mediated transport was also shown to depend on tumor type, with more efficiency noted in neutrophil-rich tumors. Revealing NPs characteristics and host factors influencing the neutrophil-based tumor targeting will help to rationally design drug delivery systems for improved cancer treatment. STATEMENT OF SIGNIFICANCE: Utilizing host cells as trojan horses for delivery nanodrugs to tumor site is a promising approach for cancer therapy. However, it is not clear yet how nanoparticles characteristics and tumor properties affect the efficiency of cell-based nanoparticles transport. Here, we compare neutrophil-based delivery of different-shaped magnetic nanoparticles (cubes and clusters) in two tumor models. The results suggest that neutrophil-mediated route is more impactful for rapidly cleared cubes, than for longer circulating clusters. The efficiency of cell-based accumulation also correlated with the level of neutrophils recruitment to different tumor types. These finding are important for rationale design of nanocarriers and predicting the efficiency of neutrophil-mediated drug delivery between patients and tumor types.
Collapse
|
31
|
Liu T, Bai R, Zhou H, Wang R, Liu J, Zhao Y, Chen C. The effect of size and surface ligands of iron oxide nanoparticles on blood compatibility. RSC Adv 2020; 10:7559-7569. [PMID: 35492144 PMCID: PMC9049842 DOI: 10.1039/c9ra10969b] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/11/2020] [Indexed: 11/21/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been widely used and have attracted increased attention for their unique physicochemical properties, especially in biomedical sciences as contrast agents following intravenous administration. However, only few studies have systematically reported the blood compatibility of iron oxide nanoparticles with different physicochemical properties such as different sizes and surface ligands. Therefore, we selected three widely used organic ligands (polyacrylic acid, hyaluronic acid, and chitosan) with modified SPIONs at the same size of 5-6 nm, and polyacrylic acid-modified SPIONs with different sizes (5, 10, and 30 nm) at different concentrations to evaluate their haemocompatibility. Our results revealed that SPIONs modified with polyacrylic acid demonstrated size-dependent destruction of red blood cells and complement activation. Interestingly, 5 nm SPIONs prolonged blood clotting time as compared with 10 nm and 30 nm SPIONs in vitro. Compared with polyacrylic acid-modified SPIONs, hyaluronic acid- and chitosan-modified SPIONs least affected red blood cells, platelets, coagulation, and complement activation. Hence, hyaluronic acid- and chitosan-coated SPIONs are more suitable for nanomedicine applications than polyacrylic acid-coated SPIONs. Furthermore, the interaction between SPIONs and blood components strongly correlated with the administered concentration of nanoparticles. These results will provide some experimental information for safe-by-design SPIONs.
Collapse
Affiliation(s)
- Tao Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Ru Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
| | - Huige Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
| | - Rongqi Wang
- Department of Clinical Laboratory, Beijing Haidian Hospital, Haidian Section of Peking University Third Hospital Beijing 100080 P. R. China
| | - Jing Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
- Faculty of Life Sciences & Medicine, Northwest University (NWU) Xi'an 710069 P. R. China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| |
Collapse
|
32
|
Methods of Granulocyte Isolation from Human Blood and Labeling with Multimodal Superparamagnetic Iron Oxide Nanoparticles. Molecules 2020; 25:molecules25040765. [PMID: 32053865 PMCID: PMC7070653 DOI: 10.3390/molecules25040765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/17/2020] [Accepted: 02/02/2020] [Indexed: 02/08/2023] Open
Abstract
This in vitro study aimed to find the best method of granulocyte isolation for subsequent labeling with multimodal nanoparticles (magnetic and fluorescent properties) to enable detection by optical and magnetic resonance imaging (MRI) techniques. The granulocytes were obtained from venous blood samples from 12 healthy volunteers. To achieve high purity and yield, four different methods of granulocyte isolation were evaluated. The isolated granulocytes were labeled with multimodal superparamagnetic iron oxide nanoparticles (M-SPIONs) coated with dextran, and the iron load was evaluated qualitatively and quantitatively by MRI, near-infrared fluorescence (NIRF) and inductively coupled plasma mass spectrometry (ICP-MS). The best method of granulocyte isolation was Percoll with Ficoll, which showed 95.92% purity and 94% viability. After labeling with M-SPIONs, the granulocytes showed 98.0% purity with a yield of 3.5 × 106 cells/mL and more than 98.6% viability. The iron-loading value in the labeled granulocytes, as obtained by MRI, was 6.40 ± 0.18 pg/cell. Similar values were found with the ICP-MS and NIRF imaging techniques. Therefore, our study shows that it is possible to isolate granulocytes with high purity and yield and labeling with M-SPIONs provides a high internalized iron load and low toxicity to cells. Therefore, these M-SPION-labeled granulocytes could be a promising candidate for future use in inflammation/infection detection by optical and MRI techniques.
Collapse
|
33
|
Wang G, Serkova NJ, Groman EV, Scheinman RI, Simberg D. Feraheme (Ferumoxytol) Is Recognized by Proinflammatory and Anti-inflammatory Macrophages via Scavenger Receptor Type AI/II. Mol Pharm 2019; 16:4274-4281. [PMID: 31556296 PMCID: PMC7513579 DOI: 10.1021/acs.molpharmaceut.9b00632] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Feraheme (ferumoxytol), a negatively charged, carboxymethyl dextran-coated ultrasmall superparamagnetic iron oxide nanoparticle (USPIO, 30 nm, -16 mV), is clinically approved as an iron supplement and is used off-label for magnetic resonance imaging (MRI) of macrophage-rich lesions, but the mechanism of recognition is not known. We investigated mechanisms of uptake of Feraheme by various types of macrophages in vitro and in vivo. The uptake by mouse peritoneal macrophages was not inhibited in complement-deficient serum. In contrast, the uptake of larger and less charged SPIO nanoworms (60 nm, -5 mV; 120 nm, -5 mV, respectively) was completely inhibited in complement deficient serum, which could be attributed to more C3 molecules bound per nanoparticle than Feraheme. The uptake of Feraheme in vitro was blocked by scavenger receptor (SR) inhibitor polyinosinic acid (PIA) and by antibody against scavenger receptor type A I/II (SR-AI/II). Antibodies against other SRs including MARCO, CD14, SR-BI, and CD11b had no effect on Feraheme uptake. Intraperitoneally administered PIA inhibited the peritoneal macrophage uptake of Feraheme in vivo. Nonmacrophage cells transfected with SR-AI plasmid efficiently internalized Feraheme but not noncharged ultrasmall SPIO of the same size (26 nm, -6 mV), suggesting that the anionic carboxymethyl groups of Feraheme are responsible for the SR-AI recognition. The uptake by nondifferentiated bone marrow derived macrophages (BMDM) and by BMDM differentiated into M1 (proinflammatory) and M2 (anti-inflammatory) types was efficiently inhibited by PIA and anti-SR-AI/II antibody. Interestingly, all BMDM types expressed similar levels of SR-AI/II. In conclusion, Feraheme is efficiently recognized via SR-AI/II but not via complement by different macrophage types. The recognition by the common phagocytic receptor has implications for specificity of imaging of macrophage subtypes.
Collapse
Affiliation(s)
- Guankui Wang
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Natalie J. Serkova
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Departments of Radiology, Radiation Oncology, and Medicine/Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Ernest V. Groman
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Robert I. Scheinman
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Corresponding Author:
| |
Collapse
|
34
|
Hannon G, Lysaght J, Liptrott NJ, Prina‐Mello A. Immunotoxicity Considerations for Next Generation Cancer Nanomedicines. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900133. [PMID: 31592123 PMCID: PMC6774033 DOI: 10.1002/advs.201900133] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/02/2019] [Indexed: 05/12/2023]
Abstract
Although interest and funding in nanotechnology for oncological applications is thriving, translating these novel therapeutics through the earliest stages of preclinical assessment remains challenging. Upon intravenous administration, nanomaterials interact with constituents of the blood inducing a wide range of associated immunotoxic effects. The literature on the immunological interactions of nanomaterials is vast and complicated. A small change in a particular characteristic of a nanomaterial (e.g., size, shape, or charge) can have a significant effect on its immunological profile in vivo, and poor selection of specific assays for establishing these undesirable effects can overlook this issue until the latest stages of preclinical assessment. This work describes the current literature on unintentional immunological effects associated with promising cancer nanomaterials (liposomes, dendrimers, mesoporous silica, iron oxide, gold, and quantum dots) and puts focus on what is missing in current preclinical evaluations. Opportunities for avoiding or limiting immunotoxicity through efficient preclinical assessment are discussed, with an emphasis placed on current regulatory views and requirements. Careful consideration of these issues will ensure a more efficient preclinical assessment of cancer nanomedicines, enabling a smoother clinical translation with less failures in the future.
Collapse
Affiliation(s)
- Gary Hannon
- Nanomedicine and Molecular Imaging GroupTrinity Translational Medicine Institute (TTMI)Trinity College DublinDublin 8Ireland
| | - Joanne Lysaght
- Department of SurgeryTTMITrinity College DublinDublin 8Ireland
| | - Neill J. Liptrott
- Department of Molecular and Clinical PharmacologyInstitute of Translational MedicineThe University of LiverpoolLiverpoolL69 3GFUK
| | - Adriele Prina‐Mello
- Nanomedicine and Molecular Imaging GroupTrinity Translational Medicine Institute (TTMI)Trinity College DublinDublin 8Ireland
- Laboratory for Biological Characterisation of Advanced Materials (LBCAM)TTMITrinity College DublinDublin 8Ireland
- Advanced Materials and Bioengineering Research (AMBER) CentreCRANN InstituteTrinity College DublinDublin 2Ireland
| |
Collapse
|
35
|
Moghimi SM, Simberg D, Skotland T, Yaghmur A, Hunter AC. The Interplay Between Blood Proteins, Complement, and Macrophages on Nanomedicine Performance and Responses. J Pharmacol Exp Ther 2019; 370:581-592. [PMID: 30940695 PMCID: PMC11047092 DOI: 10.1124/jpet.119.258012] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 03/28/2019] [Indexed: 12/17/2022] Open
Abstract
In the blood, depending on their physicochemical characteristics, nanoparticles attract a wide range of plasma biomolecules. The majority of blood biomolecules bind nonspecifically to nanoparticles. On the other hand, biomolecules such as pattern-recognition complement-sensing proteins may recognize some structural determinants of the pristine surface, causing complement activation. Adsorption of nonspecific blood proteins could also recruit natural antibodies and initiate complement activation, and this seems to be a global process with many preclinical and clinical nanomedicines. We discuss these issues, since complement activation has ramifications in nanomedicine stability and pharmacokinetics, as well as in inflammation and disease progression. Some studies have also predicted a role for complement systems in infusion-related reactions, whereas others show a direct role for macrophages and other immune cells independent of complement activation. We comment on these discrepancies and suggest directions for exploring the underlying mechanisms.
Collapse
Affiliation(s)
- S Moein Moghimi
- School of Pharmacy and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (S.M.M.); Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus (S.M.M., D.S.), and Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences (D.S.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway (T.S.); Department of Pharmacy, University of Copenhagen, Copenhagen Ø, Denmark (A.Y.); and Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, United Kingdom (A.C.H.)
| | - Dmitri Simberg
- School of Pharmacy and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (S.M.M.); Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus (S.M.M., D.S.), and Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences (D.S.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway (T.S.); Department of Pharmacy, University of Copenhagen, Copenhagen Ø, Denmark (A.Y.); and Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, United Kingdom (A.C.H.)
| | - Tore Skotland
- School of Pharmacy and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (S.M.M.); Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus (S.M.M., D.S.), and Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences (D.S.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway (T.S.); Department of Pharmacy, University of Copenhagen, Copenhagen Ø, Denmark (A.Y.); and Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, United Kingdom (A.C.H.)
| | - Anan Yaghmur
- School of Pharmacy and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (S.M.M.); Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus (S.M.M., D.S.), and Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences (D.S.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway (T.S.); Department of Pharmacy, University of Copenhagen, Copenhagen Ø, Denmark (A.Y.); and Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, United Kingdom (A.C.H.)
| | - A Christy Hunter
- School of Pharmacy and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (S.M.M.); Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus (S.M.M., D.S.), and Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences (D.S.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway (T.S.); Department of Pharmacy, University of Copenhagen, Copenhagen Ø, Denmark (A.Y.); and Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, United Kingdom (A.C.H.)
| |
Collapse
|
36
|
Ren J, Cai R, Wang J, Daniyal M, Baimanov D, Liu Y, Yin D, Liu Y, Miao Q, Zhao Y, Chen C. Precision Nanomedicine Development Based on Specific Opsonization of Human Cancer Patient-Personalized Protein Coronas. NANO LETTERS 2019; 19:4692-4701. [PMID: 31244235 DOI: 10.1021/acs.nanolett.9b01774] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
When a nanomedicine is administrated into the human body, biomolecules in biological fluids, particularly proteins, form a layer on the surface of the nanoparticle known as a "personalized protein corona". An understanding of the formation and behavior of the personalized protein corona not only benefits the nanotherapy treatment efficacy but also can aid in disease diagnosis. Here we used Gd@C82(OH)22 nanoparticles, a nanomedicine effective against several types of cancer, as a model nanomedicine to investigate the natural protein fingerprint of the personalized protein corona formed in 10 human lung squamous cell carcinoma patients. Our analysis revealed a specific biomarker, complement component C1q, in lung cancer personalized protein coronas, abundantly bound to Gd@C82(OH)22 NPs. This binding altered the secondary structure of C1q protein and led to the activation of an innate immune response, which could be exploited for cancer immune therapy. On the basis of this finding, we provide a new strategy for the development of precision nanomedicine derived from opsonization of a unique protein fingerprint within patients. This approach overcomes the common pitfall of protein corona formation and exploits the corona proteins to generate a precision nanomedicine and diagnostic tool.
Collapse
Affiliation(s)
- Jiayu Ren
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Rong Cai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
| | - Muhammad Daniyal
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Didar Baimanov
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
| | - Dongtao Yin
- Department of Thoracic Surgery , Chinese PLA General Hospital , Beijing 100853 , China
| | - Yang Liu
- Department of Thoracic Surgery , Chinese PLA General Hospital , Beijing 100853 , China
| | - Qing Miao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
37
|
Nienhaus K, Nienhaus GU. Towards a molecular-level understanding of the protein corona around nanoparticles – Recent advances and persisting challenges. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019. [DOI: 10.1016/j.cobme.2019.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
38
|
Gifford G, Vu VP, Banda NK, Holers VM, Wang G, Groman EV, Backos D, Scheinman R, Moghimi SM, Simberg D. Complement therapeutics meets nanomedicine: overcoming human complement activation and leukocyte uptake of nanomedicines with soluble domains of CD55. J Control Release 2019; 302:181-189. [PMID: 30974134 PMCID: PMC6684249 DOI: 10.1016/j.jconrel.2019.04.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/22/2019] [Accepted: 04/07/2019] [Indexed: 01/04/2023]
Abstract
Complement activation plays an important role in pharmacokinetic and performance of intravenously administered nanomedicines. Significant efforts have been directed toward engineering of nanosurfaces with low complement activation, but due to promiscuity of complement factors and redundancy of pathways, it is still a major challenge. Cell membrane-anchored Decay Accelerating Factor (DAF, a.k.a. CD55) is an efficient membrane bound complement regulator that inhibits both classical and alternative C3 convertases by accelerating their spontaneous decay. Here we tested the effect of various short consensus repeats (SCRs, "sushi" domains) of human CD55 on nanoparticle-mediated complement activation in human sera and plasma. Structural modeling suggested that SCR-2, SCR-3 and SCR-4 are critical for binding to the alternative pathway C3bBb convertase, whereas SCR-1 is dispensable. Various domains were expressed in E.coli and purified by an affinity column. SCRs were added to lepirudin plasma or sera from different healthy subjects, to monitor nanoparticle-mediated complement activation as well as C3 opsonization. Using superparamagnetic iron oxide nanoworms (SPIO NWs), we found that SCR-2-3-4 was the most effective inhibitor (IC50 ~0.24 μM for C3 opsonization in sera), followed by SCR-1-2-3-4 (IC50 ~0.6 μM), whereas shorter domains (SCR-3, SCR-2-3, SCR-3-4) were ineffective. SCR-2-3-4 also inhibited C5a generation (IC50 ~0.16 μM in sera). In addition to SPIO NWs, SCR-2-3-4 effectively inhibited C3 opsonisation and C5a production by clinically approved nanoparticles (Feraheme, LipoDox and Onivyde). SCR-2-3-4 inhibited both lectin and alternative pathway activation by nanoparticles. When added to lepirudin-anticoagulated blood from healthy donors, it significantly reduced the uptake of SPIO NWs by neutrophils and monocytes. These results suggest that soluble domains of membrane-bound complement inhibitors are potential candidates for preventing nanomedicine-mediated complement activation in human subjects.
Collapse
Affiliation(s)
- Geoffrey Gifford
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vivian P Vu
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nirmal K Banda
- Division of Rheumatology, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Court, Aurora, CO 80045, USA
| | - V Michael Holers
- Division of Rheumatology, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Court, Aurora, CO 80045, USA
| | - Guankui Wang
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Division of Rheumatology, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Court, Aurora, CO 80045, USA
| | - Ernest V Groman
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Donald Backos
- Computational Chemistry and Biology Core Facility, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E. Montview Blvd., Aurora, CO 80045, USA
| | - Robert Scheinman
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - S Moein Moghimi
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
39
|
Lofchy LA, Vu VP, Banda NK, Ramirez JR, Smith WJ, Gifford G, Gaikwad H, Scheinman RI, Simberg D. Evaluation of Targeting Efficiency of Joints with Anticollagen II Antibodies. Mol Pharm 2019; 16:2445-2451. [PMID: 31091104 DOI: 10.1021/acs.molpharmaceut.9b00059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diseases of the joints affect over 10% of the world's population, resulting in significant morbidity. There is an unmet need in strategies for specific delivery of therapeutics to the joints. Collagen type II is synthesized by chondrocytes and is mainly restricted to the cartilage and tendons. Arthrogen-CIA is a commercially available anticollagen II antibody cocktail that reacts with 5 different epitopes on human, bovine, and mouse collagen II. Arthrogen has been used for induction of experimental rheumatoid arthritis (RA) in mice because of high complement activation on the cartilage surface. Native collagen II might serve as a useful target for potential delivery of therapeutics to the joint. To evaluate the efficiency and specificity of targeting collagen II, Arthrogen was labeled with near-infrared (NIR) dye IRDye 800 or IRDye 680. Using ex vivo NIR imaging, we demonstrate that Arthrogen efficiently and specifically accumulated in the limb joints regardless of the label dye or injection route (intravenous and subcutaneous). After subcutaneous injection, the mean fluorescence of the hind limb joints was 19 times higher than that of the heart, 8.7 times higher than that of the liver, and 3.7 times higher than that of the kidney. Control mouse IgG did not show appreciable accumulation. Microscopically, the antibody accumulated on the cartilage surface of joints and on endosteal surfaces. A monoclonal antibody against a single epitope of collagen II showed similar binding affinity and elimination half-life, but about three times lower targeting efficiency than Arthrogen in vitro and ex vivo, and about two times lower targeting efficiency in vivo. We suggest that an antibody against multiple epitopes of collagen II could be developed into a highly effective and specific targeting strategy for diseases of the joints or spine.
Collapse
Affiliation(s)
| | | | - Nirmal K Banda
- Division of Rheumatology, School of Medicine , University of Colorado Denver , Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Joseline Ramos Ramirez
- Division of Rheumatology, School of Medicine , University of Colorado Denver , Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | | | | | | | | | | |
Collapse
|
40
|
Escamilla-Rivera V, Solorio-Rodríguez A, Uribe-Ramírez M, Lozano O, Lucas S, Chagolla-López A, Winkler R, De Vizcaya-Ruiz A. Plasma protein adsorption on Fe 3O 4-PEG nanoparticles activates the complement system and induces an inflammatory response. Int J Nanomedicine 2019; 14:2055-2067. [PMID: 30988608 PMCID: PMC6438142 DOI: 10.2147/ijn.s192214] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Understanding of iron oxide nanoparticles (IONP) interaction with the body milieu is crucial to guarantee their efficiency and biocompatibility in nanomedicine. Polymer coating to IONP, with polyethyleneglycol (PEG) and polyvinylpyrrolidone (PVP), is an accepted strategy to prevent toxicity and excessive protein binding. AIM The aim of this study was to investigate the feature of IONP adsorption of complement proteins, their activation and consequent inflammatory response as a strategy to further elucidate their biocompatibility. METHODS Three types of IONP with different surface characteristics were used: bare (IONP-bare), coated with PVP (IONP-PVP) and PEG-coated (IONP-PEG). IONPs were incubated with human plasma and adsorbed proteins were identified. BALB/c mice were intravenously exposed to IONP to evaluate complement activation and proinflammatory response. RESULTS Protein corona fingerprinting showed that PEG surface around IONP promoted a selective adsorption of complement recognition molecules which would be responsible for the complement system activation. Furthermore, IONP-PEG activated in vitro, the complement system and induced a substantial increment of C3a and C4a anaphylatoxins while IONP-bare and IONP-PVP did not. In vivo IONP-PEG induced an increment in complement activation markers (C5a and C5b-9), and proinflammatory cytokines (IL-1β, IL-6, TNF-α). CONCLUSION The engineering of nanoparticles must incorporate the association between complement proteins and nanomedicines, which will regulate the immunostimulatory effects through a selective adsorption of plasma proteins and will enable a safer application of IONP in human therapy.
Collapse
Affiliation(s)
- V Escamilla-Rivera
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, México,
| | - A Solorio-Rodríguez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, México,
| | - M Uribe-Ramírez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, México,
| | - O Lozano
- Namur Nanosafety Centre, Namur Research Institute for Life Sciences, University of Namur, Namur, Belgium
- Research Centre for the Physics of Matter and Radiation, University of Namur, Namur, Belgium
- Cátedra de Cardiología y Medicina Vascular, Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, México
| | - S Lucas
- Namur Nanosafety Centre, Namur Research Institute for Life Sciences, University of Namur, Namur, Belgium
- Research Centre for the Physics of Matter and Radiation, University of Namur, Namur, Belgium
| | - A Chagolla-López
- Departmento de Biotecnología y Bioquímica, CINVESTAV-IPN, Unidad Irapuato, Irapuato, México
| | - R Winkler
- Departmento de Biotecnología y Bioquímica, CINVESTAV-IPN, Unidad Irapuato, Irapuato, México
- Max Planck Institute for Chemical Ecology, Mass Spectrometry Group, Beutenberg Campus, Jena, Germany
| | - A De Vizcaya-Ruiz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, México,
| |
Collapse
|
41
|
Saito E, Kuo R, Pearson RM, Gohel N, Cheung B, King NJC, Miller SD, Shea LD. Designing drug-free biodegradable nanoparticles to modulate inflammatory monocytes and neutrophils for ameliorating inflammation. J Control Release 2019; 300:185-196. [PMID: 30822435 DOI: 10.1016/j.jconrel.2019.02.025] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 12/12/2022]
Abstract
Inflammation associated with autoimmune diseases and chronic injury is an initiating event that leads to tissue degeneration and dysfunction. Inflammatory monocytes and neutrophils systemically circulate and enter inflamed tissue, and pharmaceutical based targeting of these cells has not substantially improved outcomes and has had side effects. Herein, we investigated the design of drug-free biodegradable nanoparticles, notably without any active pharmaceutical ingredient or targeting ligand, that target circulating inflammatory monocytes and neutrophils in the vasculature to inhibit them from migrating into inflamed tissue. Nanoparticles were formed from 50:50 poly(DL-lactide-co-glycolide) (PLG) with two molecular weights (Low, High) and poly(DL-lactide) (PLA) (termed PLG-L, PLG-H, and PDLA, respectively) and were analyzed for their association with monocytes and neutrophils and their impact on disease course along with immune cell trafficking. For particles injected intravenously for 6 consecutive days to mice with experimental autoimmune encephalomyelitis (EAE), PLG-H particles had significantly lower EAE clinical scores than PBS control, while PLG-L and PDLA particles had modest or negligible effect on EAE onset. In vivo and in vitro data suggests that PLG-H particles had high association with immune cells, with preferential association with blood neutrophils relative to other particles. PLG-H particles restrained immune cells from the central nervous system (CNS), with increased accumulation in the spleen, which was not observed for mice receiving PDLA or control treatments. These results demonstrate that the particle composition influences the association with inflammatory monocytes and neutrophils in the vasculature, with the potential to redirect trafficking and ameliorate inflammation.
Collapse
Affiliation(s)
- Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Robert Kuo
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21201, USA
| | - Nishant Gohel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brandon Cheung
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas J C King
- The Discipline of Pathology, School of Medical Science, Bosch Institute, Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL 60208, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
42
|
Wang X, Zheng K, Si Y, Guo X, Xu Y. Protein⁻Polyelectrolyte Interaction: Thermodynamic Analysis Based on the Titration Method †. Polymers (Basel) 2019; 11:E82. [PMID: 30960066 PMCID: PMC6402006 DOI: 10.3390/polym11010082] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/26/2018] [Accepted: 01/02/2019] [Indexed: 01/05/2023] Open
Abstract
This review discussed the mechanisms including theories and binding stages concerning the protein⁻polyelectrolyte (PE) interaction, as well as the applications for both complexation and coacervation states of protein⁻PE pairs. In particular, this review focused on the applications of titration techniques, that is, turbidimetric titration and isothermal titration calorimetry (ITC), in understanding the protein⁻PE binding process. To be specific, by providing thermodynamic information such as pHc, pHφ, binding constant, entropy, and enthalpy change, titration techniques could shed light on the binding affinity, binding stoichiometry, and driving force of the protein⁻PE interaction, which significantly guide the applications by utilization of these interactions. Recent reports concerning interactions between proteins and different types of polyelectrolytes, that is, linear polyelectrolytes and polyelectrolyte modified nanoparticles, are summarized with their binding differences systematically discussed and compared based on the two major titration techniques. We believe this short review could provide valuable insight in the understanding of the structure⁻property relationship and the design of applied biomedical PE-based systems with optimal performance.
Collapse
Affiliation(s)
- Xiaohan Wang
- State-Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Kai Zheng
- State-Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Yi Si
- Institute of Vascular Surgery, Fudan University, 180 Fenglin road, Shanghai 200032, China.
| | - Xuhong Guo
- State-Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
- International Joint Research Center of Green Energy Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
- Engineering Research Center of Xinjiang Bingtuan of Materials Chemical Engineering, Shihezi University, Xinjiang 832000, China.
| | - Yisheng Xu
- State-Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
- International Joint Research Center of Green Energy Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
- Engineering Research Center of Xinjiang Bingtuan of Materials Chemical Engineering, Shihezi University, Xinjiang 832000, China.
| |
Collapse
|
43
|
Betker JL, Jones D, Childs CR, Helm KM, Terrell K, Nagel MA, Anchordoquy TJ. Nanoparticle uptake by circulating leukocytes: A major barrier to tumor delivery. J Control Release 2018; 286:85-93. [PMID: 30030182 PMCID: PMC6936323 DOI: 10.1016/j.jconrel.2018.07.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/05/2018] [Accepted: 07/16/2018] [Indexed: 12/18/2022]
Abstract
Decades of research into improving drug delivery to tumors has documented uptake of particulate delivery systems by resident macrophages in the lung, liver, and spleen, and correlated short circulation times with reduced tumor accumulation. An implicit assumption in these studies is that nanoparticles present in the blood are available for distribution to the tumor. This study documents significant levels of lipoplex uptake by circulating leukocytes, and its effect on distribution to the tumor and other organs. In agreement with previous studies, PEGylation dramatically extends circulation times and enhances tumor delivery. However, our studies suggest that this relationship is not straightforward, and that particle sequestration by leukocytes can significantly alter biodistribution, especially with non-PEGylated nanoparticle formulations. We conclude that leukocyte uptake should be considered in biodistribution studies, and that delivery to these circulating cells may present opportunities for treating viral infections and leukemia.
Collapse
Affiliation(s)
- Jamie L Betker
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Dallas Jones
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Christine R Childs
- Flow Cytometry Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Karen M Helm
- Flow Cytometry Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kristina Terrell
- Flow Cytometry Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Maria A Nagel
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Thomas J Anchordoquy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
44
|
Quach QH, Ang SK, Chu JHJ, Kah JCY. Size-dependent neutralizing activity of gold nanoparticle-based subunit vaccine against dengue virus. Acta Biomater 2018; 78:224-235. [PMID: 30099200 DOI: 10.1016/j.actbio.2018.08.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 12/17/2022]
Abstract
Dengue results in substantial human morbidity and significant socio-economic impacts, but a specific dengue therapeutic is not available. The currently available dengue vaccine has low efficacy and high rate of adverse effects, necessitating different strategies for the development of a safer and more efficient vaccine against dengue virus. We describe here a hybrid combination of different-sized gold nanoparticles (AuNPs) and domain III of envelope glycoprotein derived from serotype 2 of dengue virus (EDIII) as dengue subunit vaccine. The efficacy of the EDIII-functionalized AuNPs (AuNP-E) to induce neutralizing antibody in BALB/c mice is evaluated. Obtained results show that AuNP-E induced a high level of antibody which mediates serotype-specific neutralization of dengue virus. More importantly, the level of antibody is dependent on both the size of AuNPs and the concentration of AuNP-E, implicating the possibility to modulate it through adjusting these parameters. These results represent an important step towards the development of tetravalent AuNP-based subunit dengue vaccine. STATEMENT OF SIGNIFICANCE This research presents a novel subunit vaccine against dengue virus using a hybrid comprising gold nanoparticles (AuNPs) and domain III of envelop protein (EDIII). We proved the neutralizing activity of anti-EDIII antibody induced in immunized mice on Dengue virus serotype 2 in an AuNP core size and concentration dependent manner. The hybrid concept behind this work could also be adopted for the development of a tetravalent vaccine against four serotypes of Dengue virus.
Collapse
|
45
|
Deshantri AK, Varela Moreira A, Ecker V, Mandhane SN, Schiffelers RM, Buchner M, Fens MHAM. Nanomedicines for the treatment of hematological malignancies. J Control Release 2018; 287:194-215. [PMID: 30165140 DOI: 10.1016/j.jconrel.2018.08.034] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/24/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022]
Abstract
Hematological malignancies (HM) are a collection of malignant transformations originating from cells in the primary or secondary lymphoid organs. Leukemia, lymphoma, and multiple myeloma comprise the three major types of HM. Current treatment consists of bone marrow transplantation, radiotherapy, immunotherapy and chemotherapy. Although, many chemotherapeutic drugs are clinically available for the treatment of HM, the use of these agents is limited due to dose-related toxicity and lack of specificity to tumor tissue. Moreover, the poor pharmacokinetic profile of most of the chemotherapeutics requires high dosage and frequent administration to maintain therapeutic levels at the target site, both increasing adverse effects. This underlines an urgent need for a suitable drug delivery system to improve efficacy, safety, and pharmacokinetic properties of conventional therapeutics. Nanomedicines have proven to enhance these properties for anticancer therapeutics. The most extensively studied nanomedicine systems are lipid-based nanoparticles and polymeric nanoparticles. Typically, nanomedicines are small sub-micron sized particles in the size range of 20-200 nm. The biocompatible and biodegradable nature of nanomedicines makes them attractive vehicles to improve drug delivery. Their small size allows them to extravasate and accumulate at malignant sites passively by means of the enhanced permeability and retention (EPR) effect, resulting from rapid angiogenesis and inflammation. Moreover, the specificity to the target tissue can be further enhanced by surface modification of nanoparticles. This review describes currently available therapies as well as limitations and potential advantages of nanomedicine formulations for treatment of various types of HM. Additionally, recent investigational and approved nanomedicine formulations and their limited applications in HM are discussed.
Collapse
Affiliation(s)
- Anil K Deshantri
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Biological Research Pharmacology Department, Sun Pharma Advanced Research Company Ltd, India
| | - Aida Varela Moreira
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Veronika Ecker
- Institute for Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Sanjay N Mandhane
- Biological Research Pharmacology Department, Sun Pharma Advanced Research Company Ltd, India
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maike Buchner
- Institute for Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Marcel H A M Fens
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
46
|
Yang P, Xu H, Zhang Z, Yang L, Kuang H, Aguilar ZP. Surface modification affect the biodistribution and toxicity characteristics of iron oxide magnetic nanoparticles in rats. IET Nanobiotechnol 2018; 12:562-568. [PMID: 30095413 PMCID: PMC8676196 DOI: 10.1049/iet-nbt.2017.0152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/12/2017] [Accepted: 01/09/2018] [Indexed: 07/20/2023] Open
Abstract
Various surface modifications of iron oxide magnetic nanoparticles (IOMNs) can improve their stability and long-term retention time in vivo, expanding applications of biomedical fields. However, whether the long-term retention of IOMNs coated with different surface modifications has toxic effects remains poorly understood. Here, the toxicity of IOMNs modified with polyethylene glycol (PEG), bovine serum albumin (BSA), and carboxyl group (COOH), forming PEG-IOMNs, BSA-IOMNs, and COOH-IOMNs, respectively, were evaluated in the rats. The high accumulation of PEG-IOMNs and COOH-IOMNs both in the liver and lung, and the high accumulation BSA-IOMNs in blood after 24 day recovery were observed by elemental content analysis. Except individual neutrophils in the local portal area, PEG-IOMNs can also induce cytoplasmic vacuolisation in partial liver cells by histopathological examination. Furthermore, the results of RT-qPCR showed that PEG-IOMNs, BSA-IOMNs, and COOH-IOMNs can change the transcript levels of most genes related to iron homeostasis, mitochondria apoptosis, inflammatory response, but <2-fold alteration. COOH-IOMNs seemed to induce normal cell apoptosis more easily than BSA-IOMNs and PEG-IOMNs. In conclusion, BSA-IOMNs had longer-term retention time in blood. IOMNs coated with PEG and BSA can still induce side effects on the liver.
Collapse
Affiliation(s)
- Pengfei Yang
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, People's Republic of China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, People's Republic of China.
| | - Zhihong Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, People's Republic of China
| | - Lin Yang
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, People's Republic of China
| | - Huijuan Kuang
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, People's Republic of China
| | | |
Collapse
|
47
|
Experimental challenges regarding the in vitro investigation of the nanoparticle-biocorona in disease states. Toxicol In Vitro 2018; 51:40-49. [PMID: 29738787 DOI: 10.1016/j.tiv.2018.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/11/2018] [Accepted: 05/03/2018] [Indexed: 11/20/2022]
Abstract
Toxicological evaluation of nanoparticles (NPs) requires the utilization of in vitro techniques due to their number and diverse properties. Cell culture systems are often lacking in their ability to perform comparative toxicity assessment due to dosimetry issues and capacity to simulate in vivo environments. Upon encountering a physiological environment, NPs become coated with biomolecules forming a biocorona (BC), influencing function, biodistribution, and toxicity. Disease-induced alterations in the biological milieu can alter BC formation. This study evaluates the role of low-density lipoprotein (LDL) in altering macrophage responses to iron oxide (Fe3O4) NPs. BCs were formed by incubating Fe3O4 NPs in serum-free media, or 10% fetal bovine serum with or without LDL present. Following exposures to a normalized dose (25 μg/mL), macrophage association of Fe3O4 NPs with a LDL-BC was enhanced. TNF-α mRNA expression and protein levels were differentially induced due to BCs. Cell surface expression of SR-B1 was reduced following all Fe3O4 NPs exposures, while only NPs with an LDL-BC enhanced mitochondrial membrane potential. These findings suggest that elevations in LDL may contribute to distinct BC formation thereby influencing NP-cellular interactions and response. Further, our study highlights challenges that may arise during the in vitro evaluation of disease-related variations in the NP-BC.
Collapse
|
48
|
Szebeni J, Bedőcs P, Dézsi L, Urbanics R. A porcine model of complement activation-related pseudoallergy to nano-pharmaceuticals: Pros and cons of translation to a preclinical safety test. PRECISION NANOMEDICINE 2018. [DOI: 10.29016/180427.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pigs provide a sensitive and quantitative animal model of non-IgE-mediated(pseudoallergic) hypersensitivity reactions (HSRs) caused by liposomes and many other nanoparticulate drugs or drug-carrier nanosystems (nanomedicines). The rapidly arising symptoms, including cardiopulmonary, hemodynamic, hematological, blood chemistry and skin changes, resemble the clinical picture in man undergoing infusion reactions toreactogenic nanoparticles. In addition to summarizing the basic features of the pig CARPA model, thereviewconsiderssome of the advantages and disadvantages of using the modelforpreclinical evaluation of nanomedicine safety.
Collapse
Affiliation(s)
- János Szebeni
- Nanomedicine Research and Education Center, Semmelweis University, Budapest, Hungary
| | - Péter Bedőcs
- Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - László Dézsi
- Nanomedicine Research and Education Center, Semmelweis University, Budapest, Hungary
| | | |
Collapse
|
49
|
Abdollah MRA, Carter TJ, Jones C, Kalber TL, Rajkumar V, Tolner B, Gruettner C, Zaw-Thin M, Baguña Torres J, Ellis M, Robson M, Pedley RB, Mulholland P, T M de Rosales R, Chester KA. Fucoidan Prolongs the Circulation Time of Dextran-Coated Iron Oxide Nanoparticles. ACS NANO 2018; 12:1156-1169. [PMID: 29341587 DOI: 10.1021/acsnano.7b06734] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The magnetic properties and safety of dextran-coated superparamagnetic iron oxide nanoparticles (SPIONs) have facilitated their clinical use as MRI contrast agents and stimulated research on applications for SPIONs in particle imaging and magnetic hyperthermia. The wider clinical potential of SPIONs, however, has been limited by their rapid removal from circulation via the reticuloendothelial system (RES). We explored the possibility of extending SPION circulatory time using fucoidan, a seaweed-derived food supplement, to inhibit RES uptake. The effects of fucoidan on SPION biodistribution were evaluated using ferucarbotran, which in its pharmaceutical formulation (Resovist) targets the RES. Ferucarbotran was radiolabeled at the iron oxide core with technetium-99m (99mTc; t1/2 = 6 h) or zirconium-89 (89Zr; t1/2 = 3.3 days). Results obtained with 99mTc-ferucarbotran demonstrated that administration of fucoidan led to a 4-fold increase in the circulatory half-life (t1/2 slow) from 37.4 to 150 min (n = 4; P < 0.0001). To investigate whether a longer circulatory half-life could lead to concomitant increased tumor uptake, the effects of fucoidan were tested with 89Zr-ferucarbotran in mice bearing syngeneic subcutaneous (GL261) tumors. In this model, the longer circulatory half-life achieved with fucoidan was associated with a doubling in tumor SPION uptake (n = 5; P < 0.001). Fucoidan was also effective in significantly increasing the circulatory half-life of perimag-COOH, a commercially available SPION with a larger hydrodynamic size (130 nm) than ferucarbotran (65 nm). These findings indicate successful diversion of SPIONs away from the hepatic RES and show realistic potential for future clinical applications.
Collapse
Affiliation(s)
- Maha R A Abdollah
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE) , El Shorouk City, Misr- Ismalia Desert Road, Cairo 11837, Egypt
| | - Thomas J Carter
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Clare Jones
- School of Biomedical Engineering & Imaging Sciences, King's College London (KCL) , St Thomas' Hospital, London SE1 7EH, U.K
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London , London WC1E 6DD, U.K
| | - Vineeth Rajkumar
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Berend Tolner
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Cordula Gruettner
- Micromod Partikeltechnologie GmbH , Friedrich-Barnewitz-Str. 4, D-18119 Rostock, Germany
| | - May Zaw-Thin
- Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London , London WC1E 6DD, U.K
| | - Julia Baguña Torres
- School of Biomedical Engineering & Imaging Sciences, King's College London (KCL) , St Thomas' Hospital, London SE1 7EH, U.K
| | - Matthew Ellis
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Institute of Neurology (ION), University College London (UCL) , Queen Square, London WC1N 3BG, U.K
| | - Mathew Robson
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - R Barbara Pedley
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Paul Mulholland
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London (KCL) , St Thomas' Hospital, London SE1 7EH, U.K
| | - Kerry Ann Chester
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| |
Collapse
|
50
|
Chen X, Gao C. Influences of surface coating of PLGA nanoparticles on immune activation of macrophages. J Mater Chem B 2018; 6:2065-2077. [DOI: 10.1039/c7tb03080k] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Uptake of BSA-coated PLGA NPs induces a stronger inflammatory response which is represented by the up-expression of TNF-α.
Collapse
Affiliation(s)
- Xinyi Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization
- Department of Polymer Science and Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization
- Department of Polymer Science and Engineering
- Zhejiang University
- Hangzhou 310027
- China
| |
Collapse
|