1
|
Zimmermann S, Weißenfels M, Krümmer N, Härtig W, Weise G, Branzan D, Michalski D, Pelz JO. Elevated serum levels of anti-collagen type I antibodies in patients with spontaneous cervical artery dissection and ischemic stroke: a prospective multicenter study. Front Immunol 2024; 15:1348430. [PMID: 38840911 PMCID: PMC11150572 DOI: 10.3389/fimmu.2024.1348430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction Spontaneous cervical artery dissection (sCAD) is a rare vasculopathy whose trigger is still unknown. We hypothesized that autoimmunity against components of the vascular wall might play a critical role in sCAD and examined anti-collagen type I antibodies in patients with sCAD, acute ischemic stroke, patients with thromboendarterectomy, and controls. Methods Fifty-seven patients with sCAD (age 45.7 ± 10.2 years, female 18 (31.6%)) were prospectively enrolled in four German stroke centers. Blood samples were collected at baseline, at day 10 ± 3, and after 6 ± 1 months. Patients with ischemic stroke not related to CAD (n=54, age 56.7 ± 13.7 years, female 15 (27.8%)), healthy probands (n=80, age 57.4 ± 12.9 years, female 56 (70%)), and patients undergoing thromboendarterectomy of the carotid artery (n=9, age 70.7 ± 9.3 years, female 2 (22.2%)) served as controls. Anti-collagen type I antibodies were determined by enzyme-linked immunosorbent assays (ELISAs). Results Patients with acute sCAD had higher serum levels of anti-collagen type I antibodies (33.9 ± 24.6 µg/ml) than probands (18.5 ± 11.0 µg/ml; p <0.001) but lower levels than patients with ischemic stroke not related to sCAD (47.8 ± 28.4 µg/ml; p=0.003). In patients with sCAD, serum levels of anti-collagen type I antibodies were similar in the acute, subacute, and chronic phase. Levels of anti-collagen type I antibodies significantly correlated with circulating collagen type I (rho=0.207, p=0.003). Conclusion Anti-collagen type I antibodies seem not to represent a trigger for acute sCAD or ischemic stroke but may rather be linked to the metabolism and turnover of collagen type I.
Collapse
Affiliation(s)
- Silke Zimmermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | | | - Norma Krümmer
- Department of Neurology, Klinikum Altenburger Land, Altenburg, Germany
| | - Wolfgang Härtig
- Paul Flechsig Institute – Center of Neuropathology and Brain Research, University of Leipzig, Leipzig, Germany
| | - Gesa Weise
- Department of Neurology, Sana Kliniken Leipziger Land, Borna, Germany
| | - Daniela Branzan
- Department of Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Dominik Michalski
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Johann Otto Pelz
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
2
|
Berikkhanova K, Taigulov E, Bokebaev Z, Kusainov A, Tanysheva G, Yedrissov A, Seredin G, Baltabayeva T, Zhumadilov Z. Drug-loaded erythrocytes: Modern approaches for advanced drug delivery for clinical use. Heliyon 2024; 10:e23451. [PMID: 38192824 PMCID: PMC10772586 DOI: 10.1016/j.heliyon.2023.e23451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
Scientific organizations worldwide are striving to create drug delivery systems that provide a high local concentration of a drug in pathological tissue without side effects on healthy organs in the body. Important physiological properties of red blood cells (RBCs), such as frequent renewal ability, good oxygen carrying ability, unique shape and membrane flexibility, allow them to be used as natural carriers of drugs in the body. Erythrocyte carriers derived from autologous blood are even more promising drug delivery systems due to their immunogenic compatibility, safety, natural uniqueness, simple preparation, biodegradability and convenience of use in clinical practice. This review is focused on the achievements in the clinical application of targeted drug delivery systems based on osmotic methods of loading RBCs, with an emphasis on advancements in their industrial production. This article describes the basic methods used for encapsulating drugs into erythrocytes, key strategic approaches to the clinical use of drug-loaded erythrocytes obtained by hypotonic hemolysis. Moreover, clinical trials of erythrocyte carriers for the targeted delivery are discussed. This article explores the recent advancements and engineering approaches employed in the encapsulation of erythrocytes through hypotonic hemolysis methods, as well as the most promising inventions in this field. There is currently a shortage of reviews focused on the automation of drug loading into RBCs; therefore, our work fills this gap. Finally, further prospects for the development of engineering and technological solutions for the automatic production of drug-loaded RBCs were studied. Automated devices have the potential to provide the widespread production of RBC-encapsulated therapeutic drugs and optimize the process of targeted drug delivery in the body. Furthermore, they can expedite the widespread introduction of this innovative treatment method into clinical practice, thereby significantly expanding the effectiveness of treatment in both surgery and all areas of medicine.
Collapse
Affiliation(s)
- Kulzhan Berikkhanova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| | - Erlan Taigulov
- University Medical Center, Nazarbayev University, Astana, 010000, Kazakhstan
- Astana Medical University, Astana, 010000, Kazakhstan
| | - Zhanybek Bokebaev
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
- Astana Medical University, Astana, 010000, Kazakhstan
| | - Aidar Kusainov
- Semey State Medical University, Semey, 071400, Kazakhstan
| | | | - Azamat Yedrissov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| | - German Seredin
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| | - Tolkyn Baltabayeva
- Scientific-Production Center of Transfusiology, Astana, 010000, Kazakhstan
| | - Zhaxybay Zhumadilov
- Departament of Surgery, School of Medicine, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| |
Collapse
|
3
|
Wang X, Meng X, Mao K, Chen H, Cong X, Liu F, Wang J, Liu S, Xin Y, Zhu G, Tan H, Yang YG, Sun T. Maleimide as the PEG end-group promotes macrophage-targeted drug delivery of PEGylated nanoparticles in vivo by enhancing interaction with circulating erythrocytes. Biomaterials 2023; 300:122187. [PMID: 37302279 DOI: 10.1016/j.biomaterials.2023.122187] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/13/2023]
Abstract
Radiotherapy (IR) is capable of enhancing antitumor immune responses. However, IR treatment also aggravates the infiltration of peripheral macrophages into the tumor, resulting in reversing the therapeutic effects of antitumor immunity. Thus, a strategy to effectively prevent tumor infiltration by macrophages may further improved the therapeutic efficacy of radiotherapy. Herein, we found that PEGylated solid lipid nanoparticles with maleimide as PEG end-group (SLN-PEG-Mal) show significantly enhanced adsorption onto RBCs through reacting with reactive sulfhydryl groups on RBCs' surface both in vitro and in vivo, and caused significant changes in the surface properties and morphology of RBCs. These RBCs adsorbed by SLN-PEG-Mal were rapidly removed from circulation due to efficient engulfment by reticuloendothelial macrophages, supporting the usefulness of SLN-PEG-Mal for macrophage-targeted drug delivery. While lacking the use of radioisotope tracing (considered the gold standard for PK/BD studies), our data align with the expected pathway of host defense activation through surface-loaded RBCs. Importantly, injection of paclitaxel-loaded SLN-PEG-Mal effectively inhibited the tumor-infiltration by macrophages, and significantly improved the antitumor immune responses in tumor-bearing mice treated with low-dose irradiation. This study provides insights into the effects of maleimide as PEG end-group on enhancing the interaction between PEGylated nanoparticles and RBCs and offers an effective strategy to inhibit tumor infiltration by circulating macrophages.
Collapse
Affiliation(s)
- Xin Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; Medical Laboratory Center, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Hongmei Chen
- Department of Oncology Chemotherapy, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Feiqi Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yanbao Xin
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Ge Zhu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Huizhu Tan
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
4
|
Avsievich T, Zhu R, Popov AP, Yatskovskiy A, Popov AA, Tikhonowsky G, Pastukhov AI, Klimentov S, Bykov A, Kabashin A, Meglinski I. Impact of Plasmonic Nanoparticles on Poikilocytosis and Microrheological Properties of Erythrocytes. Pharmaceutics 2023; 15:1046. [PMID: 37111532 PMCID: PMC10143243 DOI: 10.3390/pharmaceutics15041046] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Plasmonic nanoparticles (NP) possess great potential in photothermal therapy and diagnostics. However, novel NP require a detailed examination for potential toxicity and peculiarities of interaction with cells. Red blood cells (RBC) are important for NP distribution and the development of hybrid RBC-NP delivery systems. This research explored RBC alterations induced by noble (Au and Ag) and nitride-based (TiN and ZrN) laser-synthesized plasmonic NP. Optical tweezers and conventional microscopy modalities indicated the effects arising at non-hemolytic levels, such as RBC poikilocytosis, and alterations in RBC microrheological parameters, elasticity and intercellular interactions. Aggregation and deformability significantly decreased for echinocytes independently of NP type, while for intact RBC, all NP except Ag NP increased the interaction forces but had no effect on RBC deformability. RBC poikilocytosis promoted by NP at concentration 50 μg mL-1 was more pronounced for Au and Ag NP, compared to TiN and ZrN NP. Nitride-based NP demonstrated better biocompatibility towards RBC and higher photothermal efficiency than their noble metal counterparts.
Collapse
Affiliation(s)
- Tatiana Avsievich
- Optoelectronics and Measurement Techniques, University of Oulu, 90570 Oulu, Finland
| | - Ruixue Zhu
- Optoelectronics and Measurement Techniques, University of Oulu, 90570 Oulu, Finland
| | - Alexey P. Popov
- VTT Technical Research Centre of Finland, Kaitovayla 1, 90590 Oulu, Finland
| | - Alexander Yatskovskiy
- Department of Histology, Cytology and Embryology, Institute of Clinical Medicine N.V. Sklifosovsky, I.M. Sechenov First Moscow State Medical University, Trubetskaya Street 8, 119991 Moscow, Russia
| | - Anton A. Popov
- Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University (MEPhI), Kashirskoe Shosse, 31, 115409 Moscow, Russia
| | - Gleb Tikhonowsky
- Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University (MEPhI), Kashirskoe Shosse, 31, 115409 Moscow, Russia
| | - Andrei I. Pastukhov
- CNRS, LP3, Aix-Marseille University, 163 Av. de Luminy, 13009 Marseille, France
| | - Sergei Klimentov
- Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University (MEPhI), Kashirskoe Shosse, 31, 115409 Moscow, Russia
| | - Alexander Bykov
- Optoelectronics and Measurement Techniques, University of Oulu, 90570 Oulu, Finland
| | - Andrei Kabashin
- Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University (MEPhI), Kashirskoe Shosse, 31, 115409 Moscow, Russia
- CNRS, LP3, Aix-Marseille University, 163 Av. de Luminy, 13009 Marseille, France
| | - Igor Meglinski
- Optoelectronics and Measurement Techniques, University of Oulu, 90570 Oulu, Finland
- College of Engineering and Physical Sciences, Aston University, Birmingham B4 7ET, UK
| |
Collapse
|
5
|
Supramolecular erythrocytes-hitchhiking drug delivery system for specific therapy of acute pneumonia. J Control Release 2022; 350:777-786. [PMID: 35995300 DOI: 10.1016/j.jconrel.2022.08.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/26/2022] [Accepted: 08/14/2022] [Indexed: 12/15/2022]
Abstract
Acute pneumonia is an inflammatory syndrome often associated with severe multi-organ dysfunction and high mortality. The therapeutic efficacy of current anti-inflammatory medicines is greatly limited due to the short systemic circulation and poor specificity in the lungs. New drug delivery systems (DDS) are urgently needed to efficiently transport anti-inflammatory drugs to the lungs. Here, we report an inflammation-responsive supramolecular erythrocytes-hitchhiking DDS to extend systemic circulation of the nanomedicine via hitchhiking red blood cells (RBCs) and specifically "drop off" the payloads in the inflammatory lungs. β-cyclodextrin (β-CD) modified RBCs and ferrocene (Fc) modified liposomes (NP) were prepared and co-incubated to attach NP to RBCs via β-CD/Fc host-guest interactions. RBCs extended the systemic circulation of the attached NP, meanwhile, the NP may get detached from RBCs due to the high ROS level in the inflammatory lungs. In acute pneumonia mice, this strategy delivered curcumin specifically to the lungs and effectively alleviated the inflammatory syndrome.
Collapse
|
6
|
Glassman PM, Villa CH, Marcos-Contreras OA, Hood ED, Walsh LR, Greineder CF, Myerson JW, Shuvaeva T, Puentes L, Brenner JS, Siegel DL, Muzykantov VR. Targeted In Vivo Loading of Red Blood Cells Markedly Prolongs Nanocarrier Circulation. Bioconjug Chem 2022; 33:1286-1294. [PMID: 35710322 DOI: 10.1021/acs.bioconjchem.2c00196] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Engineering drug delivery systems for prolonged pharmacokinetics (PK) has been an ongoing pursuit for nearly 50 years. The gold standard for PK enhancement is the coating of nanoparticles with polymers, namely polyethylene glycol (PEGylation), which has been applied in several clinically used products. In the present work, we utilize the longest circulating and most abundant component of blood─the erythrocyte─to improve the PK behavior of liposomes. Antibody-mediated coupling of liposomes to erythrocytes was tested in vitro to identify a loading dose that did not adversely impact the carrier cells. Injection of erythrocyte targeting liposomes into mice resulted in a ∼2-fold improvement in the area under the blood concentration versus time profile versus PEGylated liposomes and a redistribution from the plasma into the cellular fraction of blood. These results suggest that in vivo targeting of erythrocytes is a viable strategy to improve liposome PK relative to current, clinically viable strategies.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Carlos H Villa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Landis R Walsh
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Colin F Greineder
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Laura Puentes
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Don L Siegel
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
7
|
|
8
|
Glassman PM, Hood ED, Ferguson LT, Zhao Z, Siegel DL, Mitragotri S, Brenner JS, Muzykantov VR. Red blood cells: The metamorphosis of a neglected carrier into the natural mothership for artificial nanocarriers. Adv Drug Deliv Rev 2021; 178:113992. [PMID: 34597748 PMCID: PMC8556370 DOI: 10.1016/j.addr.2021.113992] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/26/2021] [Accepted: 09/24/2021] [Indexed: 12/18/2022]
Abstract
Drug delivery research pursues many types of carriers including proteins and other macromolecules, natural and synthetic polymeric structures, nanocarriers of diverse compositions and cells. In particular, liposomes and lipid nanoparticles represent arguably the most advanced and popular human-made nanocarriers, already in multiple clinical applications. On the other hand, red blood cells (RBCs) represent attractive natural carriers for the vascular route, featuring at least two distinct compartments for loading pharmacological cargoes, namely inner space enclosed by the plasma membrane and the outer surface of this membrane. Historically, studies of liposomal drug delivery systems (DDS) astronomically outnumbered and surpassed the RBC-based DDS. Nevertheless, these two types of carriers have different profile of advantages and disadvantages. Recent studies showed that RBC-based drug carriers indeed may feature unique pharmacokinetic and biodistribution characteristics favorably changing benefit/risk ratio of some cargo agents. Furthermore, RBC carriage cardinally alters behavior and effect of nanocarriers in the bloodstream, so called RBC hitchhiking (RBC-HH). This article represents an attempt for the comparative analysis of liposomal vs RBC drug delivery, culminating with design of hybrid DDSs enabling mutual collaborative advantages such as RBC-HH and camouflaging nanoparticles by RBC membrane. Finally, we discuss the key current challenges faced by these and other RBC-based DDSs including the issue of potential unintended and adverse effect and contingency measures to ameliorate this and other concerns.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Laura T Ferguson
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Don L Siegel
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02138, United States
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
9
|
Della Pelle G, Kostevšek N. Nucleic Acid Delivery with Red-Blood-Cell-Based Carriers. Int J Mol Sci 2021; 22:5264. [PMID: 34067699 PMCID: PMC8156122 DOI: 10.3390/ijms22105264] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/27/2022] Open
Abstract
Gene therapy has the potential to become a staple of 21st-century medicine. However, to overcome the limitations of existing gene-delivery therapies, that is, poor stability and inefficient and delivery and accumulation of nucleic acids (NAs), safe drug-delivery systems (DDSs) allowing the prolonged circulation and expression of the administered genes in vivo are needed. In this review article, the development of DDSs over the past 70 years is briefly described. Since synthetic DDSs can be recognized and eliminated as foreign substances by the immune system, new approaches must be found. Using the body's own cells as DDSs is a unique and exciting strategy and can be used in a completely new way to overcome the critical limitations of existing drug-delivery approaches. Among the different circulatory cells, red blood cells (RBCs) are the most abundant and thus can be isolated in sufficiently large quantities to decrease the complexity and cost of the treatment compared to other cell-based carriers. Therefore, in the second part, this article describes 70 years of research on the development of RBCs as DDSs, covering the most important RBC properties and loading methods. In the third part, it focuses on RBCs as the NA delivery system with advantages and drawbacks discussed to decide whether they are suitable for NA delivery in vivo.
Collapse
Affiliation(s)
- Giulia Della Pelle
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova Cesta 39, 1000 Ljubljana, Slovenia;
- Jožef Stefan International Postgraduate School, Jamova Cesta 39, 1000 Ljubljana, Slovenia
| | - Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova Cesta 39, 1000 Ljubljana, Slovenia;
| |
Collapse
|
10
|
Yin M, Li C, Jiang J, Le J, Luo B, Yang F, Fang Y, Yang M, Deng Z, Ni W, Shao J. Cell adhesion molecule-mediated therapeutic strategies in atherosclerosis: From a biological basis and molecular mechanism to drug delivery nanosystems. Biochem Pharmacol 2021; 186:114471. [PMID: 33587918 DOI: 10.1016/j.bcp.2021.114471] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/30/2021] [Accepted: 02/08/2021] [Indexed: 01/13/2023]
Abstract
Atherosclerosis (AS), characterized by pathological constriction of blood vessels due to chronic low-grade inflammation and lipid deposition, is a leading cause of human morbidity and mortality worldwide. Cell adhesion molecules (CAMs) have the ability to regulate the inflammatory response and endothelial function, as well as potentially driving plaque rupture, which all contribute to the progression of AS. Moreover, recent advances in the development of clinical agents in the cardiovascular field are based on CAMs, which show promising results in the fight against AS. Here, we review the current literature on mechanisms by which CAMs regulate atherosclerotic progression from the earliest induction of inflammation to plaques formation. In particular, we focused on therapeutic strategies based on CAMs inhibitors that prevent leukocyte from migrating to endothelium, including high-affinity antibodies and antagonists, nonspecific traditional medicinal formulas and lipid lowering drugs. The CAMs-based drug delivery nanosystem and the available data on the more reasonable and effective clinical application of CAMs inhibitors have been emphasized, raising hope for further progress in the field of AS therapy.
Collapse
Affiliation(s)
- Mengdie Yin
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Chao Li
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jiali Jiang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jingqing Le
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Bangyue Luo
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Fang Yang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Yifan Fang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Mingyue Yang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Zhenhua Deng
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Wenxin Ni
- Ocean College, Minjiang University, Fuzhou 350108, China
| | - Jingwei Shao
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China.
| |
Collapse
|
11
|
Ye H, Shen Z, Wei M, Li Y. Red blood cell hitchhiking enhances the accumulation of nano- and micro-particles in the constriction of a stenosed microvessel. SOFT MATTER 2021; 17:40-56. [PMID: 33285555 DOI: 10.1039/d0sm01637c] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
We investigate the circulation of nano- and micro-particles, including spherical particles and filamentous nanoworms, with red blood cells (RBCs) suspension in a constricted channel that mimics a stenosed microvessel. Through three-dimensional simulations using the immersed boundary-based Lattice Boltzmann method, the influence of channel geometries, such as the length and ratio of the constriction, on the accumulation of particles is systematically studied. Firstly, we find that the accumulation of spherical particles with 1 μm diameter in the constriction increases with the increases of both the length and ratio of the constriction. This is attributed to the interaction between spheres and RBCs. The RBCs "carry" the spheres and they accumulate inside the constriction together, due to the altered local hydrodynamics induced by the existence of the constriction. Secondly, nanoworms demonstrate higher accumulation than that of spheres inside the constriction, which is associated with the escape of nanoworms from RBC clusters and their accumulation near the wall of main channel. The accumulated near-wall nanoworms will eventually enter the constriction, thus enhancing their concentration inside the constriction. However, an exceptional case occurs in the case of constrictions with large ratio and long length. In such circumstances, the RBCs aggregate together tightly and concentrate at the center of the channel, which makes the nanoworms hardly able to escape from RBC clusters, leading to a similar accumulation of nanoworms and spheres inside the constriction. This study may provide theoretical guidance for the design of nano- and micro-particles for biomedical engineering applications, such as drug delivery systems for patients with stenosed microvessels.
Collapse
Affiliation(s)
- Huilin Ye
- Department of Mechanical Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, Connecticut 06269, USA.
| | | | | | | |
Collapse
|
12
|
Combes F, Meyer E, Sanders NN. Immune cells as tumor drug delivery vehicles. J Control Release 2020; 327:70-87. [PMID: 32735878 DOI: 10.1016/j.jconrel.2020.07.043] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022]
Abstract
This review article describes the use of immune cells as potential candidates to deliver anti-cancer drugs deep within the tumor microenvironment. First, the rationale of using drug carriers to target tumors and potentially decrease drug-related side effects is discussed. We further explain some of the current limitations when using nanoparticles for this purpose. Next, a comprehensive step-by-step description of the migration cascade of immune cells is provided as well as arguments on why immune cells can be used to address some of the limitations associated with nanoparticle-mediated drug delivery. We then describe the benefits and drawbacks of using red blood cells, platelets, granulocytes, monocytes, macrophages, myeloid-derived suppressor cells, T cells and NK cells for tumor-targeted drug delivery. An additional section discusses the versatility of nanoparticles to load anti-cancer drugs into immune cells. Lastly, we propose increasing the circulatory half-life and development of conditional release strategies as the two main future pillars to improve the efficacy of immune cell-mediated drug delivery to tumors.
Collapse
Affiliation(s)
- Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Evelyne Meyer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| |
Collapse
|
13
|
Glycoprotein VI (GPVI)-functionalized nanoparticles targeting arterial injury sites under physiological flow. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102274. [PMID: 32712174 DOI: 10.1016/j.nano.2020.102274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/31/2022]
Abstract
Thrombus formation at athero-thrombotic sites is initiated by the exposure of collagen followed by platelet adhesion mediated by the platelet-specific collagen receptor glycoprotein VI (GPVI). Here, dimeric GPVI was used as a targeting motif to functionalize polymeric nanoparticle-based drug carriers and to show that with proper design, such GPVI-coated nanoparticles (GPNs) can efficiently and specifically target arterial injury sites while withstanding physiological flow. In a microfluidic model, under physiological shear levels (1-40 dyne/cm2), 200 nm and 2 μm GPNs exhibited a >60 and >10-fold increase in binding to collagen compared to control particles, respectively. In vitro experiments in an arterial stenosis injury model, subjected to physiological pulsatile flow, showed shear-enhanced adhesion of 200 nm GPNs at the stenosis region which was confirmed in vivo in a mice ligation carotid injury model using intravital microscopy. Altogether, our results illustrate how engineering tools can be harnessed to design nano-carriers that efficiently target cardiovascular disease sites.
Collapse
|
14
|
Glassman PM, Villa CH, Ukidve A, Zhao Z, Smith P, Mitragotri S, Russell AJ, Brenner JS, Muzykantov VR. Vascular Drug Delivery Using Carrier Red Blood Cells: Focus on RBC Surface Loading and Pharmacokinetics. Pharmaceutics 2020; 12:E440. [PMID: 32397513 PMCID: PMC7284780 DOI: 10.3390/pharmaceutics12050440] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 04/30/2020] [Accepted: 05/08/2020] [Indexed: 01/26/2023] Open
Abstract
Red blood cells (RBC) have great potential as drug delivery systems, capable of producing unprecedented changes in pharmacokinetics, pharmacodynamics, and immunogenicity. Despite this great potential and nearly 50 years of research, it is only recently that RBC-mediated drug delivery has begun to move out of the academic lab and into industrial drug development. RBC loading with drugs can be performed in several ways-either via encapsulation within the RBC or surface coupling, and either ex vivo or in vivo-depending on the intended application. In this review, we briefly summarize currently used technologies for RBC loading/coupling with an eye on how pharmacokinetics is impacted. Additionally, we provide a detailed description of key ADME (absorption, distribution, metabolism, elimination) changes that would be expected for RBC-associated drugs and address unique features of RBC pharmacokinetics. As thorough understanding of pharmacokinetics is critical in successful translation to the clinic, we expect that this review will provide a jumping off point for further investigations into this area.
Collapse
Affiliation(s)
- Patrick M. Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA; (C.H.V.); (J.S.B.)
| | - Carlos H. Villa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA; (C.H.V.); (J.S.B.)
| | - Anvay Ukidve
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (A.U.); (Z.Z.); (S.M.)
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
| | - Zongmin Zhao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (A.U.); (Z.Z.); (S.M.)
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
| | - Paige Smith
- Disruptive Health Technology Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (P.S.); (A.J.R.)
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (A.U.); (Z.Z.); (S.M.)
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
| | - Alan J. Russell
- Disruptive Health Technology Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (P.S.); (A.J.R.)
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jacob S. Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA; (C.H.V.); (J.S.B.)
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir R. Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA; (C.H.V.); (J.S.B.)
| |
Collapse
|
15
|
Pan DC, Myerson JW, Brenner JS, Patel PN, Anselmo AC, Mitragotri S, Muzykantov V. Nanoparticle Properties Modulate Their Attachment and Effect on Carrier Red Blood Cells. Sci Rep 2018; 8:1615. [PMID: 29371620 PMCID: PMC5785499 DOI: 10.1038/s41598-018-19897-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/04/2018] [Indexed: 01/01/2023] Open
Abstract
Attachment of nanoparticles (NPs) to the surface of carrier red blood cells (RBCs) profoundly alters their interactions with the host organism, decelerating NP clearance from the bloodstream while enabling NP transfer from the RBC surface to the vascular cells. These changes in pharmacokinetics of NPs imposed by carrier RBCs are favorable for many drug delivery purposes. On the other hand, understanding effects of NPs on the carrier RBCs is vital for successful translation of this novel drug delivery paradigm. Here, using two types of distinct nanoparticles (polystyrene (PSNP) and lysozyme-dextran nanogels (LDNG)) we assessed potential adverse and sensitizing effects of surface adsorption of NPs on mouse and human RBCs. At similar NP loadings (approx. 50 particles per RBC), adsorption of PSNPs, but not LDNGs, induces RBCs agglutination and sensitizes RBCs to damage by osmotic, mechanical and oxidative stress. PSNPs, but not LDNGs, increase RBC stiffening and surface exposure of phosphatidylserine, both known to accelerate RBC clearance in vivo. Therefore, NP properties and loading amounts have a profound impact on RBCs. Furthermore, LDNGs appear conducive to nanoparticle drug delivery using carrier RBCs.
Collapse
Affiliation(s)
- Daniel C Pan
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States
| | - Jacob W Myerson
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States
| | - Jacob S Brenner
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States
- Pulmonary and Critical Care Division, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States
| | - Priyal N Patel
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States
| | - Aaron C Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, United States
| | - Samir Mitragotri
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, 02138, United States
| | - Vladimir Muzykantov
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States.
| |
Collapse
|
16
|
Liposomal-delivery of phosphodiesterase 5 inhibitors augments UT-15C-stimulated ATP release from human erythrocytes. Biochem Biophys Rep 2017; 12:114-119. [PMID: 28955799 PMCID: PMC5613235 DOI: 10.1016/j.bbrep.2017.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/09/2017] [Accepted: 09/09/2017] [Indexed: 11/21/2022] Open
Abstract
The use of liposomes to affect targeted delivery of pharmaceutical agents to specific sites may result in the reduction of side effects and an increase in drug efficacy. Since liposomes are delivered intravascularly, erythrocytes, which constitute almost half of the volume of blood, are ideal targets for liposomal drug delivery. In vivo, erythrocytes serve not only in the role of oxygen transport but also as participants in the regulation of vascular diameter through the regulated release of the potent vasodilator, adenosine triphosphate (ATP). Unfortunately, erythrocytes of humans with pulmonary arterial hypertension (PAH) do not release ATP in response to the physiological stimulus of exposure to increases in mechanical deformation as would occur when these cells traverse the pulmonary circulation. This defect in erythrocyte physiology has been suggested to contribute to pulmonary hypertension in these individuals. In contrast to deformation, both healthy human and PAH erythrocytes do release ATP in response to incubation with prostacyclin analogs via a well-characterized signaling pathway. Importantly, inhibitors of phosphodiesterase 5 (PDE5) have been shown to significantly increase prostacyclin analog-induced ATP release from human erythrocytes. Here we investigate the hypothesis that targeted delivery of PDE5 inhibitors to human erythrocytes, using a liposomal delivery system, potentiates prostacyclin analog- induced ATP release. The findings are consistent with the hypothesis that directed delivery of this class of drugs to erythrocytes could be a new and important method to augment prostacyclin analog-induced ATP release from these cells. Such an approach could significantly limit side effects of both classes of drugs without compromising their therapeutic effectiveness in diseases such as PAH. PDE5 inhibitors can be successfully delivered to human erythrocytes via liposomes. This results in augmented PGI2 analog-mediated ATP release. Liposomal binding to erythrocytes is rapid without affecting erythrocyte rheology. This is a novel method to augment PGI2 analog-induced ATP release from erythrocytes.
Collapse
Key Words
- ATP, (adenosine triphosphate)
- DMPC, (1,2-Dimyristoyl-sn-glycero-3-phosphocholine)
- FSC, (forward scatter)
- Liposomes
- PAH, (pulmonary arterial hypertension)
- PDE, (phosphodiesterase)
- PGI2, (prostacyclin)
- PSS, (physiological salt solution)
- Red blood cell
- SSC, (side scatter)
- TAD, (tadalafil)
- Tadalafil
- Tadalafil (PubChem CID: 110635)
- Treprostinil
- UT-15C
- UT-15C (PubChem CID: 691840)
- ZAP, (zaprinast),
- Zaprinast
- Zaprinast (PubChem CID: 5722)
- cAMP, (cyclic adenosine monophosphate)
- cGMP, (cyclic guanosine monophosphate)
- sGC, (soluble guanylyl cyclase)
Collapse
|
17
|
Red blood cells: Supercarriers for drugs, biologicals, and nanoparticles and inspiration for advanced delivery systems. Adv Drug Deliv Rev 2016; 106:88-103. [PMID: 26941164 DOI: 10.1016/j.addr.2016.02.007] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/17/2016] [Accepted: 02/19/2016] [Indexed: 12/19/2022]
Abstract
Red blood cells (RBCs) constitute a unique drug delivery system as a biologic or hybrid carrier capable of greatly enhancing pharmacokinetics, altering pharmacodynamics (for example, by changing margination within the intravascular space), and modulating immune responses to appended cargoes. Strategies for RBC drug delivery systems include internal and surface loading, and the latter can be performed both ex vivo and in vivo. A relatively new avenue for RBC drug delivery is their application as a carrier for nanoparticles. Efforts are also being made to incorporate features of RBCs in nanocarriers to mimic their most useful aspects, such as long circulation and stealth features. RBCs have also recently been explored as carriers for the delivery of antigens for modulation of immune response. Therefore, RBC-based drug delivery systems represent supercarriers for a diverse array of biomedical interventions, and this is reflected by several industrial and academic efforts that are poised to enter the clinical realm.
Collapse
|
18
|
Villa CH, Cines DB, Siegel DL, Muzykantov V. Erythrocytes as Carriers for Drug Delivery in Blood Transfusion and Beyond. Transfus Med Rev 2016; 31:26-35. [PMID: 27707522 DOI: 10.1016/j.tmrv.2016.08.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/01/2016] [Accepted: 08/12/2016] [Indexed: 10/20/2022]
Abstract
Red blood cells (RBCs) are innate carriers that can also be engineered to improve the pharmacokinetics and pharmacodynamics of many drugs, particularly biotherapeutics. Successful loading of drugs, both internally and on the external surface of RBCs, has been demonstrated for many drugs including anti-inflammatory, antimicrobial, and antithrombotic agents. Methods for internal loading of drugs within RBCs are now entering clinical use. Although internal loading can result in membrane disruption that may compromise biocompatibility, surface loading using either affinity or chemical ligands offers a diverse set of approaches for the production of RBC drug carriers. A wide range of surface determinants is potentially available for this approach, although there remains a need to characterize the effects of coupling agents to these surface proteins. Somewhat surprisingly, recent data also suggest that red cell-mediated delivery may confer tolerogenic immune effects. Questions remaining before widespread application of these technologies include determining the optimal loading protocol, source of RBCs, and production logistics, as well as addressing regulatory hurdles. Red blood cell drug carriers, after many decades of progress, are now poised to enter the clinic and broaden the potential application of RBCs in blood transfusion.
Collapse
Affiliation(s)
- Carlos H Villa
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA.
| | - Douglas B Cines
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Don L Siegel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
19
|
Myerson JW, Anselmo AC, Liu Y, Mitragotri S, Eckmann DM, Muzykantov VR. Non-affinity factors modulating vascular targeting of nano- and microcarriers. Adv Drug Deliv Rev 2016; 99:97-112. [PMID: 26596696 PMCID: PMC4798918 DOI: 10.1016/j.addr.2015.10.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/29/2015] [Accepted: 10/09/2015] [Indexed: 12/22/2022]
Abstract
Particles capable of homing and adhering to specific vascular biomarkers have potential as fundamental tools in drug delivery for mediation of a wide variety of pathologies, including inflammation, thrombosis, and pulmonary disorders. The presentation of affinity ligands on the surface of a particle provides a means of targeting the particle to sites of therapeutic interest, but a host of other factors come into play in determining the targeting capacity of the particle. This review presents a summary of several key considerations in nano- and microparticle design that modulate targeted delivery without directly altering epitope-specific affinity. Namely, we describe the effect of factors in definition of the base carrier (including shape, size, and flexibility) on the capacity of carriers to access, adhere to, and integrate in target biological milieus. Furthermore, we present a summary of fundamental dynamics of carrier behavior in circulation, taking into account interactions with cells in circulation and the role of hemodynamics in mediating the direction of carriers to target sites. Finally, we note non-affinity aspects to uptake and intracellular trafficking of carriers in target cells. In total, recent findings presented here may offer an opportunity to capitalize on mitigating factors in the behavior of ligand-targeted carriers in order to optimize targeting.
Collapse
|
20
|
Villa CH, Pan DC, Zaitsev S, Cines DB, Siegel DL, Muzykantov VR. Delivery of drugs bound to erythrocytes: new avenues for an old intravascular carrier. Ther Deliv 2015; 6:795-826. [PMID: 26228773 PMCID: PMC4712023 DOI: 10.4155/tde.15.34] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
For several decades, researchers have used erythrocytes for drug delivery of a wide variety of therapeutics in order to improve their pharmacokinetics, biodistribution, controlled release and pharmacodynamics. Approaches include encapsulation of drugs within erythrocytes, as well as coupling of drugs onto the red cell surface. This review focuses on the latter approach, and examines the delivery of red blood cell (RBC)-surface-bound anti-inflammatory, anti-thrombotic and anti-microbial agents, as well as RBC carriage of nanoparticles. Herein, we discuss the progress that has been made in surface loading approaches, and address in depth the issues relevant to surface loading of RBC, including intrinsic features of erythrocyte membranes, immune considerations, potential surface targets and techniques for the production of affinity ligands.
Collapse
Affiliation(s)
- Carlos H Villa
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel C Pan
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sergei Zaitsev
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas B Cines
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Donald L Siegel
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
Parseghian MH, Mechetner E, Osidak MS, Domogatskii SP. Application of monoclonal antibodies for the diagnostic and therapeutic targeting of human tumors with a necrotic component. RUSS J GEN CHEM+ 2014. [DOI: 10.1134/s1070363214020364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
22
|
Kona S, Dong JF, Liu Y, Tan J, Nguyen KT. Biodegradable nanoparticles mimicking platelet binding as a targeted and controlled drug delivery system. Int J Pharm 2011; 423:516-24. [PMID: 22172292 DOI: 10.1016/j.ijpharm.2011.11.043] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/09/2011] [Accepted: 11/28/2011] [Indexed: 11/15/2022]
Abstract
This research aims to develop targeted nanoparticles as drug carriers to the injured arterial wall under fluid shear stress by mimicking the natural binding ability of platelets via interactions of glycoprotein Ib-alpha (GPIbα) of platelets with P-selectin of damaged endothelial cells (ECs) and/or with von Willebrand factor (vWF) of the subendothelium. Drug-loaded poly(d,l-lactic-co-glycolic acid) (PLGA) nanoparticles were formulated using a standard emulsion method and conjugated with glycocalicin, the external fraction of platelet GPIbα, via carbodiimide chemistry. Surface-coated and cellular uptake studies in ECs showed that conjugation of PLGA nanoparticles, with GPIb, significantly increased nanoparticle adhesion to P-selectin- and vWF-coated surfaces as well as nanoparticle uptake by activated ECs under fluid shear stresses. In addition, effects of nanoparticle size and shear stress on adhesion efficiency were characterized through parallel flow chamber studies. The observed decrease in bound nanoparticle density with increased particle sizes and shear stresses is also explained through a computational model. Our results demonstrate that the GPIb-conjugated PLGA nanoparticles can be used as a targeted and controlled drug delivery system under flow conditions at the site of vascular injury.
Collapse
Affiliation(s)
- Soujanya Kona
- Department of Bioengineering, University of Texas at Arlington, TX, United States
| | | | | | | | | |
Collapse
|
23
|
Lewis DR, Kamisoglu K, York AW, Moghe PV. Polymer-based therapeutics: nanoassemblies and nanoparticles for management of atherosclerosis. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2011; 3:400-20. [PMID: 21523920 DOI: 10.1002/wnan.145] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Coronary arterial disease, one of the leading causes of adult mortality, is triggered by atherosclerosis. A disease with complex etiology, atherosclerosis results from the progressive long-term combination of atherogenesis, the accumulation of modified lipoproteins within blood vessel walls, along with vascular and systemic inflammatory processes. The management of atherosclerosis is challenged by the localized flare-up of several multipronged signaling interactions between activated monocytes, atherogenic macrophages and inflamed or dysfunctional endothelial cells. A new generation of approaches is now emerging founded on multifocal, targeted therapies that seek to reverse or ameliorate the atheroinflammatory cascade within the vascular intima. This article reviews the various classes and primary examples of bioactive configurations of nanoscale assemblies. Of specific interest are polymer-based or polymer-lipid micellar assemblies designed as multimodal receptor-targeted blockers or drug carriers whose activity can be tuned by variations in polymer hydrophobicity, charge, and architecture. Also reviewed are emerging reports on multifunctional nanoassemblies and nanoparticles for improved circulation and enhanced targeting to atheroinflammatory lesions and atherosclerotic plaques.
Collapse
Affiliation(s)
- Daniel R Lewis
- Department of Chemical & Biochemical Engineering, Rutgers University, Piscataway, NJ, USA
| | | | | | | |
Collapse
|
24
|
Tiwari B, Agarwal A, Kharya AK, Lariya N, Saraogi G, Agrawal H, Agrawal GP. Immunoglobulin immobilized liposomal constructs for transmucosal vaccination through nasal route. J Liposome Res 2010; 21:181-93. [PMID: 20626315 DOI: 10.3109/08982104.2010.498003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The aim of the present investigation was to evaluate the prospective of surface-engineered vesicular carriers for mucosal immunization via the nasal route. IgG antibody was immobilized on the surface of hepatitis B surface antigen (HBsAg) antigen-loaded liposomes. The developed formulations were characterized on the basis of physicochemical parameters, such as morphology, particle size, polydispersity index, entrapment efficiency, and zeta potential. Liposomal formulations were then evaluated for in-process antigen stability and storage stability. In vivo studies were conducted to visualize targeting potential, localization pattern, and immunogenicity. In addition, immune response was compared with alum-HBsAg vaccine injected intramuscularly. The serum anti-HBsAg titer, obtained from the postnasal administration of IgG-coupled liposomes, was significantly higher than plain liposomes. Moreover, IgG-coupled liposomes generated both humoral (i.e., systemic and mucosal) and cellular immune responses upon nasal administration, while the alum-adsorbed antigen displayed neither cellular (cytokine level) nor mucosal (IgA) response. The formulation also displayed enhanced transmucosal transport, improved in vitro stability, and effective immunoadjuvant property. To conclude, IgG antibody-coupled liposomes may serve as novel carriers to augment the secretory immune response of antigen encapsulated in the liposomes, apparently by escalating liposome uptake via M cells, thereby rationalizing their use as a carrier adjuvant for nasal subunit vaccines.
Collapse
Affiliation(s)
- Brajesh Tiwari
- Department of Pharmaceutical Sciences, Pharmaceutics Research Laboratory, Dr. H.S. Gour University, Sagar, India
| | | | | | | | | | | | | |
Collapse
|
25
|
Muzykantov VR. Drug delivery by red blood cells: vascular carriers designed by mother nature. Expert Opin Drug Deliv 2010; 7:403-27. [PMID: 20192900 DOI: 10.1517/17425241003610633] [Citation(s) in RCA: 281] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Vascular delivery of several classes of therapeutic agents may benefit from carriage by red blood cells (RBC), for example, drugs that require delivery into phagocytic cells and those that must act within the vascular lumen. The fact that several protocols of infusion of RBC-encapsulated drugs are now being explored in patients illustrates a high biomedical importance for the field. AREAS COVERED BY THIS REVIEW: Two strategies for RBC drug delivery are discussed: encapsulation into isolated RBC ex vivo followed by infusion in compatible recipients and coupling therapeutics to the surface of RBC. Studies of pharmacokinetics and effects in animal models and in human studies of diverse therapeutic enzymes, antibiotics and other drugs encapsulated in RBC are described and critically analyzed. Coupling to RBC surface of compounds regulating immune response and complement, affinity ligands, polyethylene glycol alleviating immune response to donor RBC and fibrinolytic plasminogen activators are described. Also described is a new, translation-prone approach for RBC drug delivery by injection of therapeutics conjugated with fragments of antibodies providing safe anchoring of cargoes to circulating RBC, without need for ex vivo modification and infusion of RBC. WHAT THE READER WILL GAIN Readers will gain historical perspective, current status, challenges and perspectives of medical applications of RBC for drug delivery. TAKE HOME MESSAGE RBC represent naturally designed carriers for intravascular drug delivery, characterized by unique longevity in the bloodstream, biocompatibility and safe physiological mechanisms for metabolism. New approaches for encapsulating drugs into RBC and coupling to RBC surface provide promising avenues for safe and widely useful improvement of drug delivery in the vascular system.
Collapse
Affiliation(s)
- Vladimir R Muzykantov
- University of Pennsylvania Medical Center, Department of Pharmacology and Program in Targeted Therapeutics of Institute of Translational Medicine and Therapeutics, IFEM, One John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104-6068, USA.
| |
Collapse
|
26
|
|
27
|
Angiogenic endothelium shows lactadherin-dependent phagocytosis of aged erythrocytes and apoptotic cells. Blood 2008; 111:4542-50. [PMID: 18292292 DOI: 10.1182/blood-2007-06-094763] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Angiogenic endothelium plays a crucial role in tumor growth. During angiogenesis, complex alterations in the microenvironment occur. In response, the endothelium undergoes phenotypic changes, for example overexpression of alpha(v)-integrins. Here, we show that the overexpression of alpha(v)-integrins on angiogenic endothelial cells is engaged in phagocytic actions involving binding ("tethering") and uptake ("tickling") of lactadherin (also termed MFG-E8)--opsonized particles. Phosphatidylserine (PS)--exposing multilamellar vesicles, "aged" erythrocytes, and apoptotic melanoma cells incubated with lactadherin were all phagocytosed by angiogenic endothelial cells in vitro. Furthermore, we demonstrated lactadherin expression in and around tumor blood vessels making opsonization in situ plausible. By engineering the surface of erythrocytes with covalently coupled cyclic Arg-Gly-Asp (RGD) peptides--mimicking lactadherin opsonization--we could induce phagocytosis by angiogenic endothelial cells both in vitro and in vivo. In vitro, this was confirmed by cytochalasin D preincubation. When RGD-erythrocytes were administered intravenously in tumor-bearing mice, blood vessel congestion followed by tumor core necrosis was seen. Moreover, RGD-erythrocytes could delay tumor growth in a murine melanoma model, possibly through induction of tumor infarctions. These results reveal that angiogenic endothelial cells have phagocytic properties for lactadherin-opsonized large particles and apoptotic cells. Implications of our findings for diagnostic and therapy of angiogenesis-driven diseases are discussed.
Collapse
|
28
|
Abstract
Drug targeting to selected subcellular compartments of the pulmonary endothelium may optimise treatment of many diseases. This paper describes endothelial determinants that are potentially useful for such targeting, including endothelial ectopeptidases, cell adhesion molecules and novel candidates identified by high-throughput methods, as well as the means to achieve optimal subcellular targeting of drugs in the endothelium that have been explored in cell culture and animal studies. Criteria for determining the applicability for targeting include accessibility, specificity, safety and subcellular precision. The effects of endothelial delivery of therapeutic agents, including the effects mediated by the intervention in the function of the target determinants, must be characterised in the context of given pathological conditions.
Collapse
Affiliation(s)
- Vladimir R Muzykantov
- University of Pennsylvania, Institute for Environmental Medicine and Department of Pharmacology, Philadelphia, 19104-6068, USA.
| |
Collapse
|
29
|
Murciano JC, Medinilla S, Eslin D, Atochina E, Cines DB, Muzykantov VR. Prophylactic fibrinolysis through selective dissolution of nascent clots by tPA-carrying erythrocytes. Nat Biotechnol 2003; 21:891-6. [PMID: 12845330 DOI: 10.1038/nbt846] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2003] [Accepted: 05/23/2003] [Indexed: 11/09/2022]
Abstract
A fibrinolytic agent consisting of a tissue-type plasminogen activator (tPA) coupled to the surface of red blood cells (RBCs) can dissolve nascent clots from within the clot, in a Trojan horse-like strategy, while having minimal effects on preexisting hemostatic clots or extravascular tissue. After intravenous injection, the fibrinolytic activity of RBC-tPA persisted in the bloodstream at least tenfold longer than did that of free tPA. In a model of venous thrombosis induced by intravenously injected fibrin microemboli aggregating in pulmonary vasculature, soluble tPA lysed pulmonary clots lodged before but not after tPA injection, whereas the converse was true for RBC-tPA. Free tPA failed to lyse occlusive carotid thrombosis whether injected before or after vascular trauma, whereas RBC-tPA circulating before, but not injected after, thrombus formation restored blood flow. This RBC-based drug delivery strategy alters the fibrinolytic profile of tPA, permitting prophylactic fibrinolysis.
Collapse
Affiliation(s)
- Juan-Carlos Murciano
- Institute for Environmental Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | |
Collapse
|
30
|
Manodori AB. Sickle erythrocytes adhere to fibronectin-thrombospondin-integrin complexes exposed by thrombin-induced endothelial cell contraction. Microvasc Res 2001; 61:263-74. [PMID: 11336537 DOI: 10.1006/mvre.2000.2317] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Vascular damage appears to be associated with sickle erythrocyte (SS RBC) adherence to the endothelium. Thrombin, which has been found in abnormal levels in many sickle patients, causes endothelial cell (EC) retraction and increased SS RBC adherence, and SS RBC adhere in the gaps opened between the EC. Our objective was to elucidate the mechanism of adherence to activated EC monolayers and to determine whether the matrix proteins thrombospondin (TSP) and fibronectin (FN) are mediators of this adherence. Thrombin activation elicited the same 2.5-fold increase in adherence whether 10 or 35% of the matrix was exposed, and the majority of the RBC adhered at the edges of the EC regardless of the extent of matrix exposed. Using static adherence assays we investigated whether TSP, FN, or the integrins alpha(v)beta(3) and alpha(5)beta(1) mediated adherence. Blocking antibodies to any of these four had no effect on adherence to untreated monolayers. However, all the increased adherence elicited by thrombin was abrogated by each one, whereas control antibodies had no effect. Immunofluorescent microscopy demonstrated that both integrins were present on the luminal surface of confluent EC. Neither TSP nor FN was exposed in confluent cultures but they both became available as receptors after EC retraction. These data suggest that SS RBC adhere to a complex of matrix TSP and FN maintained in an adhesive conformation by interactions with both integrins.
Collapse
Affiliation(s)
- A B Manodori
- Children's Hospital Oakland Research Institute, Oakland, California 94609, USA
| |
Collapse
|
31
|
Abstract
The main problems currently associated with systemic drug administration are: even biodistribution of pharmaceuticals throughout the body; the lack of drug specific affinity toward a pathological site; the necessity of a large total dose of a drug to achieve high local concentration; non-specific toxicity and other adverse side-effects due to high drug doses. Drug targeting, i.e. predominant drug accumulation in the target zone independently on the method and route of drug administration, may resolve many of these problems. Currently, the principal schemes of drug targeting include direct application of a drug into the affected zone, passive drug targeting (spontaneous drug accumulation in the areas with leaky vasculature, or Enhanced Permeability and Retention-EPR-effect), 'physical' targeting (based on abnormal pH value and/or temperature in the pathological zone), magnetic targeting (or targeting of a drug immobilized on paramagnetic materials under the action of an external magnetic field), and targeting using a specific 'vector' molecules (ligands having an increased affinity toward the area of interest). The last approach provides the widest opportunities. Such pharmaceutical carriers as soluble polymers, microcapsules, microparticles, cells, cell ghosts, liposomes, and micelles have been successfully used for targeted drug delivery in vivo. Though the direct conjugation of a drug molecule with a targeted moiety is also possible (immunotoxin), the use of microreservoir-type systems provides clear advantages, such as high loading capacity, possibility to control size and permeability of drug carrier systems and use relatively small number of vector molecules to deliver substantial quantities of a drug to the target. The practical use of the listed systems and approaches for the delivery of therapeutic and diagnostic agents will be considered.
Collapse
Affiliation(s)
- V P Torchilin
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Abstract
Red blood cells have unique properties that offer attractive therapeutic possibilities. It is proposed that by covalently linking drug pharmacophores to red cell surface proteins, significant enhancement of pharmacokinetics can be achieved. Specific anchoring to erythrocytes can be accomplished in principle, using unique, affinity labeling, combinatorial libraries targeting the surface protein, glycophorin A. The anticipated advantages of red cell-anchored drugs over free drugs, include extended half-lives, controlled volumes of distribution, and multivalent interactions.
Collapse
Affiliation(s)
- A Krantz
- Red Cell Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
33
|
Abstract
To make universal and efficient liposome-based drug carriers, liposomes should be able to recognize and bind other targets beyond their natural targets, the cells of the reticuloendothial system. To make liposomes targeted, numerous methods to couple active substances, primarily, monoclonal antibodies, to the liposome surface have been developed. Resulting immunoliposomes (or affinity liposomes) demonstrate good targeting to cells and organs both in vitro and in vivo. However, the short circulation time of immunoliposomes prevented them from accumulating in targets with diminished blood flow or low antigen concentration. Long-circulating liposomes were prepared by coupling soluble and flexible polymers, such as polyethylene glycol, to the liposome surface. The mechanism of liposome steric protection with flexible polymers is based on the formation of dense "conformational cloud' by a grafted polymer over the liposome surface, and might be analyzed in terms of a statistical model of polymer solutions. By co-immobilization of specific antibodies and protecting polymers on the liposome surface, liposomes can be prepared combining both targetability and prolonged circulation in vivo. A biological model (experimental myocardial infarction in rabbit) was used to estimate the relative importance of different factors (liposome size and coating with protective polymer and/or specific antibody) for effective accumulation of liposomes in the target. Statistical analysis demonstrated that different types of liposomes have to be used in order to reach maximum absolute delivery of liposomes to the target, or maximum target-to-non-target ratio (relative delivery). Therefore, different liposomes should be used as carriers of diagnostic and therapeutic agents.
Collapse
Affiliation(s)
- V P Torchilin
- Department of Radiology, Massachusetts General Hospital, Charlestown, USA
| |
Collapse
|
34
|
Muzykantov VR, Atochina EN, Ischiropoulos H, Danilov SM, Fisher AB. Immunotargeting of antioxidant enzyme to the pulmonary endothelium. Proc Natl Acad Sci U S A 1996; 93:5213-8. [PMID: 8643555 PMCID: PMC39224 DOI: 10.1073/pnas.93.11.5213] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Oxidative injury to the pulmonary endothelium has pathological significance for a spectrum of diseases. Administration of antioxidant enzymes, superoxide dismutase (SOD) and catalase (Cat), has been proposed as a method to protect endothelium. However, neither these enzymes nor their derivatives possess specific affinity to endothelium and do not accumulate in the lung. Previously we have described a monoclonal antibody to angiotensin-converting enzyme (ACE) that accumulates selectively in the lung after systemic injection in rats, hamsters, cats, monkeys, and humans. In the present work we describe a system for selective intrapulmonary delivery of CuZn-SOD and Cat conjugated with biotinylated anti-ACE antibody mAb 9B9 (b-mAb 9B9) by a streptavidin (SA)-biotin bridge. Both enzymes biotinylated with biotin ester at biotin/enzyme ratio 20 retain enzymatic activity and bind SA without loss of activity. We have constructed tri-molecular heteropolymer complexes consisting of b-mAb 9B9, SA, and biotinylated SOD or biotinylated Cat and have studied biodistribution and pulmonary uptake of these complexes in the rat after i.v. injection. Biodistribution of biotinylated enzymes was similar to that of nonmodified enzymes. Binding of SA markedly prolonged lifetime of biotinylated enzymes in the circulation. In contrast to enzymes conjugated with nonspecific IgG, other enzyme derivatives, and nonmodified enzymes, biotinylated enzymes conjugated with b-mAb 9B9 accumulated specifically in the rat lung (9% of injected SOD/g of lung tissue and 7.5% of injected Cat/g of lung tissue). Pulmonary uptake of nonmodified enzymes or derivatives with nonspecific IgG did not exceed 0.5% of injected dose/g. Both SOD and Cat conjugated with b-mAb 9B9 were retained in the rat lung for at least several hours. Trichloracetic acid-precipitable radiolabeled Cat was associated with microsomal and plasma membrane fractions of the lung tissue homogenate. Thus, modification of antioxidant enzymes with biotin and SA-mediated conjugation with b-mAb 9B9 prolongs the circulation of enzymes resulting in selective accumulation in the lung and intracellular delivery of enzymes to the pulmonary endothelium. These results provide the background for an approach to provide protection of pulmonary endothelium against oxidative insults.
Collapse
Affiliation(s)
- V R Muzykantov
- Insitute for Environmental Medicine, University of Pennsylvania Medical Center, Philadelphia, 19104, USA
| | | | | | | | | |
Collapse
|
35
|
Avidin acylation prevents the complement-dependent lysis of avidin-carrying erythrocytes. Biochem J 1991; 273(Pt 2):393-7. [PMID: 1991038 PMCID: PMC1149858 DOI: 10.1042/bj2730393] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Non-covalent binding of avidin to biotinylated erythrocytes results in complement-dependent haemolysis. Biotinylated erythrocytes, as well as native cells, are not lysed by complement. Complement activation requires a tight contact between avidin and the erythrocyte membrane, since avidin does not in itself activate complement and does not inhibit lysis of sensitized sheep erythrocytes. The efficiency of haemolysis depends on avidin's surface density. When the avidin concentration in the reaction mixture is less than 15 micrograms/ml, erythrocyte lysis is not induced. However, the attachment of biotinylated antibodies to avidin-carrying erythrocytes decreases dramatically. Acylation of avidin with succinic anhydride strongly decreases its ability to induce complement-dependent haemolysis. However, the ability of avidin to cross-link the biotin-containing structures decreases after acylation. A 50% modification of avidin by succinic anhydride (pI about 7.0) allows preparation of 'immunoerythrocytes', which retain their affinity to antigen and stability in the presence of complement.
Collapse
|
36
|
Idelson GL, Muzykantov VR, Chekneva EE, Shnyra AA, Shekhonin BV, Domogatsky SP. In vivo administration of antibodies against type I collagen in rat: the specific accumulation in spleen. COLLAGEN AND RELATED RESEARCH 1987; 7:383-97. [PMID: 3328669 DOI: 10.1016/s0174-173x(87)80037-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
[125I]-labelled rabbit antibodies against rat type I collagen and non-immune IgG were injected into rat circulation. The kinetics of their clearance and the biodistribution in different organs were studied. Both preparations showed very similar clearance rate, the kinetics fitting bi-exponential approximation with characteristic parameters t1,1/2 = 201 +/- 20 min before 320 min and t2,1/2 = 1350 +/- 450 min at times over 320 min for antibodies and 258 +/- 45 min and 890 +/- 140 min for IgG. The specific affinity of the circulating antibodies did not decrease within 24 hours. The antibodies were specifically accumulated in spleen, where their accumulation was 5-fold higher than that of non-immune IgG. Accumulation of antibodies was maximal 3 hours after the injection. The localization ratio (i.e. the ratio of the amount of the antibodies per g of tissue to that per g of blood) reached a maximum 24 hours after the injection and remained stable for 120 hours. Immunofluorescent staining of spleen sections resulted in a bright fluorescence of dense collagenous structures in the trabeculae and in the wall of the central follicular arterium, bright spot fluorescence in the marginal zone of the follicle, and diffuse fluorescence in the red pulp. These findings suggest an unusually high accessibility of collagen type I in spleen to circulating blood plasma components.
Collapse
Affiliation(s)
- G L Idelson
- Institute of Experimental Cardiology, National Cardiology Center, Moscow, USSR
| | | | | | | | | | | |
Collapse
|
37
|
Affiliation(s)
- J E O'Mullane
- Advanced Drug Delivery Research, Ciba-Geigy Pharmaceuticals, Horsham, West Sussex, United Kingdom
| | | | | |
Collapse
|