1
|
Schorr K, Beck S, Zimmer O, Baumann F, Keller M, Witzgall R, Goepferich A. The quantity of ligand-receptor interactions between nanoparticles and target cells. NANOSCALE HORIZONS 2025; 10:803-823. [PMID: 39951050 DOI: 10.1039/d4nh00645c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Achieving high target cell avidity in combination with cell selectivity are fundamental, but largely unachieved goals in the development of biomedical nanoparticle systems, which are intricately linked to the quantity of targeting functionalities on their surface. Viruses, regarded as almost ideal role models for nanoparticle design, are evolutionary optimized, so that they cope with this challenge bearing an extremely low number of spikes, and thus binding domains, on their surface. In comparison, nanoparticles are usually equipped with more than an order of magnitude more ligands. It is therefore obvious that one key factor for increasing nanoparticle efficiency in terms of avidity and selectivity lies in optimizing their ligand number. A first step along this way is to know how many ligands per nanoparticle are involved in specific binding with target cell receptors. This question is addressed experimentally for a block copolymer nanoparticle model system. The data confirm that only a fraction of the nanoparticle ligands is involved in the binding processes: with a total ligand valency of 29 ligands/100 nm2 surface area a maximum 5.3 ligands/100 nm2 are involved in specific receptor binding. This corresponds to an average number of 251 binding ligands per nanoparticle, a number that can be rationalized within the biological context of the model system.
Collapse
Affiliation(s)
- Kathrin Schorr
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Sebastian Beck
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Oliver Zimmer
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Felix Baumann
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, D-93040 Regensburg, Germany.
| | - Ralph Witzgall
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| |
Collapse
|
2
|
ten Hove M, Smyris A, Booijink R, Wachsmuth L, Hansen U, Alic L, Faber C, Hӧltke C, Bansal R. Engineered SPIONs functionalized with endothelin a receptor antagonist ameliorate liver fibrosis by inhibiting hepatic stellate cell activation. Bioact Mater 2024; 39:406-426. [PMID: 38855059 PMCID: PMC11157122 DOI: 10.1016/j.bioactmat.2024.05.034] [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: 11/16/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/11/2024] Open
Abstract
Endothelin-1/endothelin A receptor (ET-1/ETAR) pathway plays an important role in the progression of liver fibrosis by activating hepatic stellate cells (HSCs) - a key cell type involved in the pathogenesis of liver fibrosis. Inactivating HSCs by blocking the ET-1/ETAR pathway using a selective ETAR antagonist (ERA) represents a promising therapeutic approach for liver fibrosis. Unfortunately, small-molecule ERAs possess limited clinical potential due to poor bioavailability, short half-life, and rapid renal clearance. To improve the clinical applicability, we conjugated ERA to superparamagnetic iron-oxide nanoparticles (SPIONs) and investigated the therapeutic efficacy of ERA and ERA-SPIONs in vitro and in vivo and analyzed liver uptake by in vivo and ex vivo magnetic resonance imaging (MRI), HSCs-specific localization, and ET-1/ETAR-pathway antagonism in vivo. In murine and human liver fibrosis/cirrhosis, we observed overexpression of ET-1 and ETAR that correlated with HSC activation, and HSC-specific localization of ETAR. ERA and successfully synthesized ERA-SPIONs demonstrated significant attenuation in TGFβ-induced HSC activation, ECM production, migration, and contractility. In an acute CCl4-induced liver fibrosis mouse model, ERA-SPIONs exhibited higher liver uptake, HSC-specific localization, and ET-1/ETAR pathway antagonism. This resulted in significantly reduced liver-to-body weight ratio, plasma ALT levels, and α-SMA and collagen-I expression, indicating attenuation of liver fibrosis. In conclusion, our study demonstrates that the delivery of ERA using SPIONs enhances the therapeutic efficacy of ERA in vivo. This approach holds promise as a theranostic strategy for the MRI-based diagnosis and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Marit ten Hove
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Andreas Smyris
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Richell Booijink
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Lydia Wachsmuth
- Clinic of Radiology, University Hospital Muenster, Muenster, Germany
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University Hospital Muenster, Muenster, Germany
| | - Lejla Alic
- Department of Magnetic Detection and Imaging, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Cornelius Faber
- Clinic of Radiology, University Hospital Muenster, Muenster, Germany
| | - Carsten Hӧltke
- Clinic of Radiology, University Hospital Muenster, Muenster, Germany
| | - Ruchi Bansal
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| |
Collapse
|
3
|
Garnaik UC, Chandra A, Goel VK, Gulyás B, Padmanabhan P, Agarwal S. Development of SERS Active Nanoprobe for Selective Adsorption and Detection of Alzheimer's Disease Biomarkers Based on Molecular Docking. Int J Nanomedicine 2024; 19:8271-8284. [PMID: 39161360 PMCID: PMC11330857 DOI: 10.2147/ijn.s446212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/08/2024] [Indexed: 08/21/2024] Open
Abstract
Purpose Development of SERS-based Raman nanoprobes can detect the misfolding of Amyloid beta (Aβ) 42 peptides, making them a viable diagnostic technique for Alzheimer's disease (AD). The detection and imaging of amyloid peptides and fibrils are expected to help in the early identification of AD. Methods Here, we propose a fast, easy-to-use, and simple scheme based on the selective adsorption of Aβ42 molecules on SERS active gold nanoprobe (RB-AuNPs) of diameter 29 ± 3 nm for Detection of Alzheimer's Disease Biomarkers. Binding with the peptides results in a spectrum shift, which correlates with the target peptide. We also demonstrated the possibility of using silver nanoparticles (AgNPs) as precursors for the preparation of a SERS active nanoprobe with carbocyanine (CC) dye and AgNPs known as silver nanoprobe (CC-AgNPs) of diameter 25 ± 4 nm. Results RB-AuNPs probe binding with the peptides results in a spectrum shift, which correlates with the target peptide. Arginine peak appears after the conjugation confirms the binding of Aβ 42 with the nanoprobe. Tyrosine peaks appear after conjugated Aβ42 with CC-AgNPs providing binding of the peptide with the probe. The nanoprobe produced a strong, stable SERS signal. Further molecular docking was utilized to analyse the interaction and propose a structural hypothesis for the process of binding the nanoprobe to Aβ42 and Tau protein. Conclusion This peptide-probe interaction provides a general enhancement factor and the molecular structure of the misfolded peptides. Secondary structural information may be obtained at the molecular level for specific residues owing to isotope shifts in the Raman spectra. Conjugation of the nanoprobe with Aβ42 selectively detected AD in bodily fluids. The proposed nanoprobes can be easily applied to the detection of Aβ plaques in blood, saliva, and sweat samples.
Collapse
Affiliation(s)
| | - Anshuman Chandra
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Vijay Kumar Goel
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Balázs Gulyás
- Cognitive Neuroimaging Centre, Nanyang Technological University (NTU), Singapore, Singapore
| | | | - Shilpi Agarwal
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
4
|
Mohammad-Rafiei F, Khojini JY, Ghazvinian F, Alimardan S, Norioun H, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Cell membrane biomimetic nanoparticles in drug delivery. Biotechnol Appl Biochem 2023; 70:1843-1859. [PMID: 37387120 DOI: 10.1002/bab.2487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/05/2023] [Indexed: 07/01/2023]
Abstract
Despite the efficiency of nanoparticle (NP) therapy, in vivo investigations have shown that it does not perform as well as in vitro. In this case, NP confronts many defensive hurdles once they enter the body. The delivery of NP to sick tissue is inhibited by these immune-mediated clearance mechanisms. Hence, using a cell membrane to hide NP for active distribution offers up a new path for focused treatment. These NPs are better able to reach the disease's target location, leading to enhanced therapeutic efficacy. In this emerging class of drug delivery vehicles, the inherent relation between the NPs and the biological components obtained from the human body was utilized, which mimic the properties and activities of native cells. This new technology has shown the viability of using biomimicry to evade immune system-provided biological barriers, with an emphasis on restricting clearance from the body before reaching its intended target. Furthermore, by providing signaling cues and transplanted biological components that favorably change the intrinsic immune response at the disease site, the NPs would be capable interacting with immune cells regarding the biomimetic method. Thus, we aimed to provide a current landscape and future trends of biomimetic NPs in drug delivery.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Ghazvinian
- Department of Life science and biotechnology, Faculty of Natural Sciences, University of Shahid Beheshti, Tehran, Iran
| | - Sajad Alimardan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Norioun
- Medical Genetics Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Munich, Germany
| |
Collapse
|
5
|
Clemente B, Denis M, Silveira CP, Schiavetti F, Brazzoli M, Stranges D. Straight to the point: targeted mRNA-delivery to immune cells for improved vaccine design. Front Immunol 2023; 14:1294929. [PMID: 38090568 PMCID: PMC10711611 DOI: 10.3389/fimmu.2023.1294929] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
With the deepening of our understanding of adaptive immunity at the cellular and molecular level, targeting antigens directly to immune cells has proven to be a successful strategy to develop innovative and potent vaccines. Indeed, it offers the potential to increase vaccine potency and/or modulate immune response quality while reducing off-target effects. With mRNA-vaccines establishing themselves as a versatile technology for future applications, in the last years several approaches have been explored to target nanoparticles-enabled mRNA-delivery systems to immune cells, with a focus on dendritic cells. Dendritic cells (DCs) are the most potent antigen presenting cells and key mediators of B- and T-cell immunity, and therefore considered as an ideal target for cell-specific antigen delivery. Indeed, improved potency of DC-targeted vaccines has been proved in vitro and in vivo. This review discusses the potential specific targets for immune system-directed mRNA delivery, as well as the different targeting ligand classes and delivery systems used for this purpose.
Collapse
|
6
|
Walter M, Baumann F, Schorr K, Goepferich A. Ectoenzymes as promising cell identification structures for the high avidity targeting of polymeric nanoparticles. Int J Pharm 2023; 647:123453. [PMID: 37783283 DOI: 10.1016/j.ijpharm.2023.123453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/04/2023]
Abstract
Pharmacotherapy is often limited by undesired side effects while insufficient drug reaches the site of action. Active-targeted nanotherapy should provide a solution for this problem, by using ligands in the nanoparticle corona for the identification of receptors on the target-cell surface. However, since receptor binding is directly associated with pharmacological responses, today's targeting concepts must be critically evaluated. We hypothesized that addressing ectoenzymes would help to overcome this problem, but it was not clear if particles would show sufficiently high avidity to provide us with a viable alternative to classical ligand-receptor concepts. We scrutinized this aspect by immobilizing the highly selective angiotensin-converting enzyme 2 (ACE2) inhibitor MLN-4760 in the corona of block-copolymer nanoparticles and investigated enzyme binding via microscale thermophoresis and flow cytometry. Excellent avidities with Kd values as low as 243 pM for soluble ACE2 and 306 pM for ACE2-positive cells were obtained. In addition, the inhibitory activity had an IC50 value of 2.88 nM. Reliable target cell identification could be proven in coculture experiments. High avidity is the basis for minimizing material loss to off-target sites and paves the way for a paradigm shift in nanoparticle targeting which does not trigger unintended side effects following target cell identification.
Collapse
Affiliation(s)
- Melanie Walter
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany
| | - Felix Baumann
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany
| | - Kathrin Schorr
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany.
| |
Collapse
|
7
|
Gu S, Luo Q, Wen C, Zhang Y, Liu L, Liu L, Liu S, Chen C, Lei Q, Zeng S. Application of Advanced Technologies-Nanotechnology, Genomics Technology, and 3D Printing Technology-In Precision Anesthesia: A Comprehensive Narrative Review. Pharmaceutics 2023; 15:2289. [PMID: 37765258 PMCID: PMC10535504 DOI: 10.3390/pharmaceutics15092289] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/10/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
There has been increasing interest and rapid developments in precision medicine, which is a new medical concept and model based on individualized medicine with the joint application of genomics, bioinformatics engineering, and big data science. By applying numerous emerging medical frontier technologies, precision medicine could allow individualized and precise treatment for specific diseases and patients. This article reviews the application and progress of advanced technologies in the anesthesiology field, in which nanotechnology and genomics can provide more personalized anesthesia protocols, while 3D printing can yield more patient-friendly anesthesia supplies and technical training materials to improve the accuracy and efficiency of decision-making in anesthesiology. The objective of this manuscript is to analyze the recent scientific evidence on the application of nanotechnology in anesthesiology. It specifically focuses on nanomedicine, precision medicine, and clinical anesthesia. In addition, it also includes genomics and 3D printing. By studying the current research and advancements in these advanced technologies, this review aims to provide a deeper understanding of the potential impact of these advanced technologies on improving anesthesia techniques, personalized pain management, and advancing precision medicine in the field of anesthesia.
Collapse
Affiliation(s)
- Shiyao Gu
- Department of Anesthesiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Qingyong Luo
- Department of Anesthesiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Cen Wen
- Department of Anesthesiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yu Zhang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Li Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Liu Liu
- Department of Anesthesiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Su Liu
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Chunhua Chen
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Qian Lei
- Department of Anesthesiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Si Zeng
- Department of Anesthesiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| |
Collapse
|
8
|
Vanbilloen WJF, Rechberger JS, Anderson JB, Nonnenbroich LF, Zhang L, Daniels DJ. Nanoparticle Strategies to Improve the Delivery of Anticancer Drugs across the Blood-Brain Barrier to Treat Brain Tumors. Pharmaceutics 2023; 15:1804. [PMID: 37513992 PMCID: PMC10383584 DOI: 10.3390/pharmaceutics15071804] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Primary brain and central nervous system (CNS) tumors are a diverse group of neoplasms that occur within the brain and spinal cord. Although significant advances in our understanding of the intricate biological underpinnings of CNS neoplasm tumorigenesis and progression have been made, the translation of these discoveries into effective therapies has been stymied by the unique challenges presented by these tumors' exquisitely sensitive location and the body's own defense mechanisms (e.g., the brain-CSF barrier and blood-brain barrier), which normally protect the CNS from toxic insult. These barriers effectively prevent the delivery of therapeutics to the site of disease. To overcome these obstacles, new methods for therapeutic delivery are being developed, with one such approach being the utilization of nanoparticles. Here, we will cover the current state of the field with a particular focus on the challenges posed by the BBB, the different nanoparticle classes which are under development for targeted CNS tumor therapeutics delivery, and strategies which have been developed to bypass the BBB and enable effective therapeutics delivery to the site of disease.
Collapse
Affiliation(s)
- Wouter J. F. Vanbilloen
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Neurology, Elisabeth-Tweesteden Hospital, 5022 GC Tilburg, The Netherlands
| | - Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jacob B. Anderson
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
- Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Leo F. Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Hopp Children’s Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - Liang Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
9
|
Li Z, Zhu Y, Zeng H, Wang C, Xu C, Wang Q, Wang H, Li S, Chen J, Xiao C, Yang X, Li Z. Mechano-boosting nanomedicine antitumour efficacy by blocking the reticuloendothelial system with stiff nanogels. Nat Commun 2023; 14:1437. [PMID: 36918575 PMCID: PMC10015032 DOI: 10.1038/s41467-023-37150-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
Nanomedicine has been developed for cancer therapy over several decades, while rapid clearance from blood circulation by reticuloendothelial system (RES) severely limits nanomedicine antitumour efficacy. We design a series of nanogels with distinctive stiffness and investigate how nanogel mechanical properties could be leveraged to overcome RES. Stiff nanogels are injected preferentially to abrogate uptake capacity of macrophages and temporarily block RES, relying on inhibition of clathrin and prolonged liver retention. Afterwards, soft nanogels deliver doxorubicin (DOX) with excellent efficiency, reflected in high tumour accumulation, deep tumour penetration and outstanding antitumour efficacy. In this work, we combine the advantage of stiff nanogels in RES-blockade with the superiority of soft nanogels in drug delivery leads to the optimum tumour inhibition effect, which is defined as mechano-boosting antitumour strategy. Clinical implications of stiffness-dependent RES-blockade are also confirmed by promoting antitumour efficacy of commercialized nanomedicines, such as Doxil and Abraxane.
Collapse
Affiliation(s)
- Zheng Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Yabo Zhu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Haowen Zeng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Chong Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Chen Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Qiang Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Huimin Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Shiyou Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Jitang Chen
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Chen Xiao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
- GBA Research Innovation Institute for Nanotechnology, 510530, Guangzhou, Guangdong, P. R. China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China.
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China.
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China.
| |
Collapse
|
10
|
Zhang S, Ouyang T, Reinhard BM. Multivalent Ligand-Nanoparticle Conjugates Amplify Reactive Oxygen Species Second Messenger Generation and Enhance Epidermal Growth Factor Receptor Phosphorylation. Bioconjug Chem 2022; 33:1716-1728. [PMID: 35993676 PMCID: PMC9815836 DOI: 10.1021/acs.bioconjchem.2c00335] [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] [Indexed: 01/11/2023]
Abstract
The epidermal growth factor (EGF) receptor (EGFR) is heterogeneously distributed on the cellular surface and enriched in clusters with diameters of tens of nanometers. Multivalent presentation of EGF ligand on nanoparticles (NPs) provides an approach for controlling and amplifying the local activation of EGFR in these clusters. Reactive oxygen species (ROS) have been indicated to play a role in the regulation of EGFR activation as second messengers, but the effect of nanoconjugation on EGF-mediated ROS formation and ROS-induced EGFR activation is not well established. The goal of this manuscript is to characterize the multivalent enhancement of EGF-induced ROS formation and to test its effect on EGFR phosphorylation in breast cancer cell models using gold (Au) NPs with a diameter of 81 ± 1 nm functionalized with two different EGF ligand densities (12 ± 7 EGF/NP (NP-EGF12) and 87 ± 6 EGF/NP (NP-EGF87)). In the EGFR overexpressing cell lines MDA-MB-231 and MDA-MB-468, NP-EGF87 achieved a measurable multivalent enhancement of ROS that peaked at concentrations c ROSmax ≤ 25 pM and that were EGFR and nicotinamide adenine dinucleotide phosphate oxidase (NOX) dependent. NP-EGF12 failed to generate comparable ROS levels as NP-EGF87 in the investigated NP input concentration range (0-100 pM). In cells with nearly identical numbers of bound NP-EGF87 and NP-EGF12, the ROS levels for NP-EGF87 were systematically higher, indicating that the multivalent enhancement is exclusively related not only to avidity but also to a stronger stimulation per NP. Importantly, the increase in EGF-induced ROS formation associated with EGF nanoconjugation at c ROSmax resulted in a measurable gain in EGFR phosphorylation, confirming that ROS generation contributes to the multivalent enhancement of EGFR activation in response to NP-EGF87.
Collapse
Affiliation(s)
- Sandy Zhang
- Department of Chemistry and The Photonics Center, Boston University, Boston, MA 02215
| | - Tianhong Ouyang
- Department of Chemistry and The Photonics Center, Boston University, Boston, MA 02215
| | - Björn M. Reinhard
- Department of Chemistry and The Photonics Center, Boston University, Boston, MA 02215
| |
Collapse
|
11
|
Zhang S, Li R, Zheng Y, Zhou Y, Fan X. Erythrocyte Membrane-Enveloped Salvianolic Acid B Nanoparticles Attenuate Cerebral Ischemia-Reperfusion Injury. Int J Nanomedicine 2022; 17:3561-3577. [PMID: 35974873 PMCID: PMC9376004 DOI: 10.2147/ijn.s375908] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Ischemic stroke is the second leading cause of death and the third leading cause of disability worldwide. Salvianolic acid B (SAB), a water-soluble phenolic acid derived from the traditional Chinese medicine Salvia miltiorrhiza, exerted protective effects on cerebral ischemia-reperfusion injury. However, the efficacy of SAB is seriously hindered by poor blood brain barrier (BBB) permeability and short biological half-life in plasma. Brain targeted biomimetic nanoparticle delivery systems offer much promise in overcoming these limitations. Methods A brain targeted biomimetic nanomedicine (RR@SABNPs) was developed, which comprised of SAB loaded bovine serum albumin nanoparticles and functionalized red blood cell membrane (RBCM) with Arg-Gly-Asp (RGD). The characterization parameters, including particle size, zeta potential, morphology, Encapsulation Efficiency (EE), Drug Loading (DL), release behavior, stability, and biocompatibility, were investigated. Moreover, the middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model was used to assess the therapeutic efficacy of RR@SABNPs on ischemic stroke. Finally, the reactive oxygen species (ROS) levels and mitochondrial membrane potential (MMP) were detected by DHE and JC‑1 staining in oxygen-glucose deprivation/reperfusion (OGD/R) and H2O2 injured PC12 cells. Results RR@SABNPs exhibited spheric morphology with core-shell structures and good stability and biocompatibility. Meanwhile, RR@SABNPs can significantly prolong SAB circulation time by overcoming the reticuloendothelial system (RES) and actively targeting ischemic BBB. Moreover, RR@SABNPs had comprehensive protective effects on MCAO/R model mice, manifested as a reduced infarct volume and improved neurological and sensorimotor functions, and significantly scavenged excess ROS and maintained MMP. Conclusion The designed brain targeted biomimetic nanomedicine RR@SABNPs can significantly prolong the half-time of SAB, deliver SAB into the ischemic brain and exhibit good therapeutic effects on MCAO/R model mice.
Collapse
Affiliation(s)
- Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Ruoqi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Yingyi Zheng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China.,Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| |
Collapse
|
12
|
A Nanoparticle's Journey to the Tumor: Strategies to Overcome First-Pass Metabolism and Their Limitations. Cancers (Basel) 2022; 14:cancers14071741. [PMID: 35406513 PMCID: PMC8996837 DOI: 10.3390/cancers14071741] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Traditional cancer therapeutics suffer from off-target toxicity, limiting their effective dose and preventing patients’ tumors from being sufficiently treated by chemotherapeutics alone. Nanomedicine is an emerging class of therapeutics in which a drug is packaged into a nanoparticle that promotes uptake of the drug at a tumor site, shielding it from uptake by peripheral organs and enabling the safe delivery of chemotherapeutics that have poor aqueous solubility, short plasma half-life, narrow therapeutic window, and toxic side effects. Despite the advantages of nanomedicines for cancer, there remains significant challenges to improve uptake at the tumor and prevent premature clearance from the body. In this review, we summarize the effects of first-pass metabolism on a nanoparticle’s journey to a tumor and outline future steps that we believe will improve the efficacy of cancer nanomedicines. Abstract Nanomedicines represent the cutting edge of today’s cancer therapeutics. Seminal research decades ago has begun to pay dividends in the clinic, allowing for the delivery of cancer drugs with enhanced systemic circulation while also minimizing off-target toxicity. Despite the advantages of delivering cancer drugs using nanoparticles, micelles, or other nanostructures, only a small fraction of the injected dose reaches the tumor, creating a narrow therapeutic window for an otherwise potent drug. First-pass metabolism of nanoparticles by the reticuloendothelial system (RES) has been identified as a major culprit for the depletion of nanoparticles in circulation before they reach the tumor site. To overcome this, new strategies, materials, and functionalization with stealth polymers have been developed to improve nanoparticle circulation and uptake at the tumor site. This review summarizes the strategies undertaken to evade RES uptake of nanomedicines and improve the passive and active targeting of nanoparticle drugs to solid tumors. We also outline the limitations of current strategies and the future directions we believe will be explored to yield significant benefits to patients and make nanomedicine a promising treatment modality for cancer.
Collapse
|
13
|
Andrian T, Pujals S, Albertazzi L. Quantifying the effect of PEG architecture on nanoparticle ligand availability using DNA-PAINT. NANOSCALE ADVANCES 2021; 3:6876-6881. [PMID: 34977461 PMCID: PMC8650147 DOI: 10.1039/d1na00696g] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/01/2021] [Indexed: 06/14/2023]
Abstract
The importance of PEG architecture on nanoparticle (NP) functionality is known but still difficult to investigate, especially at a single particle level. Here, we apply DNA Point Accumulation for Imaging in Nanoscale Topography (DNA-PAINT), a super-resolution microscopy (SRM) technique, to study the surface functionality in poly(lactide-co-glycolide)-poly(ethylene glycol) (PLGA-PEG) NPs with different PEG structures. We demonstrated how the length of the PEG spacer can influence the accessibility of surface chemical functionality, highlighting the importance of SRM techniques to support the rational design of functionalized NPs.
Collapse
Affiliation(s)
- Teodora Andrian
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology Baldiri Reixac 15-21 08028 Barcelona Spain
| | - Silvia Pujals
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology Baldiri Reixac 15-21 08028 Barcelona Spain
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology Baldiri Reixac 15-21 08028 Barcelona Spain
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology 5612AZ Eindhoven The Netherlands
| |
Collapse
|
14
|
Advances in the Application of Nanomaterials as Treatments for Bacterial Infectious Diseases. Pharmaceutics 2021; 13:pharmaceutics13111913. [PMID: 34834328 PMCID: PMC8618949 DOI: 10.3390/pharmaceutics13111913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/01/2022] Open
Abstract
Bacteria-targeting nanomaterials have been widely used in the diagnosis and treatment of bacterial infectious diseases. These nanomaterials show great potential as antimicrobial agents due to their broad-spectrum antibacterial capacity and relatively low toxicity. Recently, nanomaterials have improved the accurate detection of pathogens, provided therapeutic strategies against nosocomial infections and facilitated the delivery of antigenic protein vaccines that induce humoral and cellular immunity. Biomaterial implants, which have traditionally been hindered by bacterial colonization, benefit from their ability to prevent bacteria from forming biofilms and spreading into adjacent tissues. Wound repair is improving in terms of both the function and prevention of bacterial infection, as we tailor nanomaterials to their needs, select encapsulation methods and materials, incorporate activation systems and add immune-activating adjuvants. Recent years have produced numerous advances in their antibacterial applications, but even further expansion in the diagnosis and treatment of infectious diseases is expected in the future.
Collapse
|
15
|
Zhang M, Gao S, Yang D, Fang Y, Lin X, Jin X, Liu Y, Liu X, Su K, Shi K. Influencing factors and strategies of enhancing nanoparticles into tumors in vivo. Acta Pharm Sin B 2021; 11:2265-2285. [PMID: 34522587 PMCID: PMC8424218 DOI: 10.1016/j.apsb.2021.03.033] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
The administration of nanoparticles (NPs) first faces the challenges of evading renal filtration and clearance of reticuloendothelial system (RES). After that, NPs infiltrate through the expanded endothelial space and penetrated the dense stroma of tumor microenvironment to tumor cells. As long as possible to prolong the time of NPs remaining in tumor tissue, NPs release active agent and induce pharmacological action. This review provides a comprehensive summary of the physical and chemical properties of NPs and the influence of various biological factors in tumor microenvironment, and discusses how to improve the final efficacy through adjusting the characteristics and structure of NPs. Perspectives and future directions are also provided.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kai Shi
- Corresponding author. Tel./fax: +86 24 43520557.
| |
Collapse
|
16
|
Andrian T, Delcanale P, Pujals S, Albertazzi L. Correlating Super-Resolution Microscopy and Transmission Electron Microscopy Reveals Multiparametric Heterogeneity in Nanoparticles. NANO LETTERS 2021; 21:5360-5368. [PMID: 34125548 PMCID: PMC8227466 DOI: 10.1021/acs.nanolett.1c01666] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/03/2021] [Indexed: 05/22/2023]
Abstract
The functionalization of nanoparticles with functional moieties is a key strategy to achieve cell targeting in nanomedicine. The interplay between size and ligand number is crucial for the formulation performance and needs to be properly characterized to understand nanoparticle structure-activity relations. However, there is a lack of methods able to measure both size and ligand number at the same time and at the single particle level. Here, we address this issue by introducing a correlative light and electron microscopy (CLEM) method combining super-resolution microscopy (SRM) and transmission electron microscopy (TEM) imaging. We apply our super-resCLEM method to characterize the relationship between size and ligand number and density in PLGA-PEG nanoparticles. We highlight how heterogeneity found in size can impact ligand distribution and how a significant part of the nanoparticle population goes completely undetected in the single-technique analysis. Super-resCLEM holds great promise for the multiparametric analysis of other parameters and nanomaterials.
Collapse
Affiliation(s)
- Teodora Andrian
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028 Barcelona, Spain
| | - Pietro Delcanale
- Dipartimento
di Scienze Matematiche, Fisiche e Informatiche, Università di Parma, Parco area delle Scienze 7/A, 43124 Parma, Italy
| | - Silvia Pujals
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Department
of Electronics and Biomedical Engineering, Faculty of Physics, Universitat de Barcelona, Avenido Diagonal 647, 08028, Barcelona, Spain
| | - Lorenzo Albertazzi
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Department
of Biomedical Engineering, Institute for Complex Molecular Systems
(ICMS), Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
- ;
| |
Collapse
|
17
|
Nguyen A, Böttger R, Li SD. Recent trends in bioresponsive linker technologies of Prodrug-Based Self-Assembling nanomaterials. Biomaterials 2021; 275:120955. [PMID: 34130143 DOI: 10.1016/j.biomaterials.2021.120955] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/19/2021] [Accepted: 05/29/2021] [Indexed: 12/15/2022]
Abstract
Prodrugs are designed to improve pharmaceutical properties of potent compounds and represent a central approach in drug development. The success of the prodrug strategy relies on incorporation of a reversible linkage facilitating controlled release of the parent drug. While prodrug approaches enhance pharmacokinetic properties over their parent drug, they still face challenges in absorption, distribution, metabolism, elimination, and toxicity (ADMET). Conjugating a drug to a carrier molecule such as a polymer can create an amphiphile that self-assembles into nanoparticles. These nanoparticles display prolonged blood circulation and passive targeting ability. Furthermore, the drug release can be tailored using a variety of linkers between the parent drug and the carrier molecule. In this review, we introduce the concept of self-assembling prodrugs and summarize different approaches for controlling the drug release with a focus on the linker technology. We also summarize recent clinical trials, discuss the emerging challenges, and provide our perspective on the utility and future potential of this technology.
Collapse
Affiliation(s)
- Anne Nguyen
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Roland Böttger
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|
18
|
Wang W, Gil-Garcia M, Ventura S. Dual Antibody-Conjugated Amyloid Nanorods to Promote Selective Cell-Cell Interactions. ACS APPLIED MATERIALS & INTERFACES 2021; 13:14875-14884. [PMID: 33759489 PMCID: PMC9262253 DOI: 10.1021/acsami.0c21996] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Grafting biomolecules on nanostructures' surfaces is an increasingly used strategy to target pathogenic cells, with both diagnostic and therapeutic applications. However, nanomaterials monofunctionalized by conjugating a single type of ligand find limited uses in pathologies/therapies that require two or more targets/receptors to be targeted and/or activated with a single molecular entity simultaneously. Therefore, multivalent nanomaterials for dual- or multitargeting are attracting significant interest. This study provides a proof of concept of such nanostructures. We have recently developed a modular methodology that allows obtaining amyloid-based materials decorated with active globular domains. Here, this approach is exploited to generate functional amyloid fibrils displaying antibody capture moieties. A high antibody binding affinity and capacity for the resulting nanofibrils, whose size can be manipulated to obtain homogeneous nanorods with high biocompatibility, are demonstrated. These nanorods are then used for specific antibody-mediated targeting of different cell types. Simultaneous conjugation of these nanorods with different antibodies allows obtaining a mimic of a bispecific antibody that redirects T lymphocytes to tumoral cells, holding high potential for immunotherapy. Overall, the work illustrates a modular and straightforward strategy to obtain preparative quantities of multivalent antibody-functionalized nanomaterials with multitargeting properties without the need for covalent modification.
Collapse
|
19
|
Baghdadi NE, Burke BP, Alresheedi T, Nigam S, Saeed A, Almutairi F, Domarkas J, Khan A, Archibald SJ. Multivalency in CXCR4 chemokine receptor targeted iron oxide nanoparticles. Dalton Trans 2021; 50:1599-1603. [PMID: 33502425 DOI: 10.1039/d0dt02626c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The CXCR4 chemokine receptor is an important biomolecular target in cancer diagnostics and therapeutics. In a new multivalent approach, iron oxide nanoparticles were conjugated with multiple binding units of a low affinity azamacrocylic CXCR4 antagonist. The silica coated nanostructure has good suspension stability, a mode size of 72 nm and high affinity for CXCR4, showing >98% inhibition of anti-CXCR4 mAb binding in a receptor binding competition assay on Jurkat cells.
Collapse
Affiliation(s)
- Neazar E Baghdadi
- Centre of Nanotechnology, King Abdul-Aziz University, Jeddah, Saudi Arabia and Department of Chemistry, University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - Benjamin P Burke
- Department of Biomedical Sciences and PET Research Centre, University of Hull Cottingham Road, Hull, HU6 7RX, UK
| | - Tahani Alresheedi
- Department of Chemistry, University of Hull, Cottingham Road, Hull, HU6 7RX, UK. and Department of Biomedical Sciences and PET Research Centre, University of Hull Cottingham Road, Hull, HU6 7RX, UK and Department of Chemistry, College of Science and Art, Qassim University, Qassim, Saudi Arabia
| | - Shubhanchi Nigam
- Department of Chemistry, University of Hull, Cottingham Road, Hull, HU6 7RX, UK. and Department of Biomedical Sciences and PET Research Centre, University of Hull Cottingham Road, Hull, HU6 7RX, UK
| | - Abdu Saeed
- Department of Physics, Faculty of Science, King Abdul-Aziz University, Jeddah 21589, Saudi Arabia
| | - Farooq Almutairi
- Department of Chemistry, University of Hull, Cottingham Road, Hull, HU6 7RX, UK. and College of Applied Medical Sciences, University of Hafar Al-Batin, Hafar Al-Batin, Saudi Arabia
| | - Juozas Domarkas
- Department of Chemistry, University of Hull, Cottingham Road, Hull, HU6 7RX, UK. and Department of Biomedical Sciences and PET Research Centre, University of Hull Cottingham Road, Hull, HU6 7RX, UK
| | - Abid Khan
- Department of Biomedical Sciences and PET Research Centre, University of Hull Cottingham Road, Hull, HU6 7RX, UK and Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Stephen J Archibald
- Department of Chemistry, University of Hull, Cottingham Road, Hull, HU6 7RX, UK. and Department of Biomedical Sciences and PET Research Centre, University of Hull Cottingham Road, Hull, HU6 7RX, UK
| |
Collapse
|
20
|
Biomedical nanoparticle design: What we can learn from viruses. J Control Release 2021; 329:552-569. [PMID: 33007365 PMCID: PMC7525328 DOI: 10.1016/j.jconrel.2020.09.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 01/02/2023]
Abstract
Viruses are nanomaterials with a number of properties that surpass those of many synthetic nanoparticles (NPs) for biomedical applications. They possess a rigorously ordered structure, come in a variety of shapes, and present unique surface elements, such as spikes. These attributes facilitate propitious biodistribution, the crossing of complex biological barriers and a minutely coordinated interaction with cells. Due to the orchestrated sequence of interactions of their stringently arranged particle corona with cellular surface receptors they effectively identify and infect their host cells with utmost specificity, while evading the immune system at the same time. Furthermore, their efficacy is enhanced by their response to stimuli and the ability to spread from cell to cell. Over the years, great efforts have been made to mimic distinct viral traits to improve biomedical nanomaterial performance. However, a closer look at the literature reveals that no comprehensive evaluation of the benefit of virus-mimetic material design on the targeting efficiency of nanomaterials exists. In this review we, therefore, elucidate the impact that viral properties had on fundamental advances in outfitting nanomaterials with the ability to interact specifically with their target cells. We give a comprehensive overview of the diverse design strategies and identify critical steps on the way to reducing them to practice. More so, we discuss the advantages and future perspectives of a virus-mimetic nanomaterial design and try to elucidate if viral mimicry holds the key for better NP targeting.
Collapse
|
21
|
Zhang X, Zhao M, Cao N, Qin W, Zhao M, Wu J, Lin D. Construction of a tumor microenvironment pH-responsive cleavable PEGylated hyaluronic acid nano-drug delivery system for colorectal cancer treatment. Biomater Sci 2020; 8:1885-1896. [PMID: 32022813 DOI: 10.1039/c9bm01927h] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In order to improve active tumor targeting, tumor cell uptake efficiency and circulation time of doxorubicin (DOX) in vivo, we constructed a cleavable PEGylated hyaluronic acid nano-drug delivery system (HA-mPEG2k-DOX) based on a tumor microenvironment pH-responsive imine bond. In this study, HA-mPEG2k-DOX can self-assemble into stable nanoparticles (HA-mPEG2k-DOX NPs) with a particle size of 50 nm. And the NPs can efficiently target CD44 positive CT26 cells and the pH-responsive cleavable PEG shell can be detached under weakly acidic environments and effectively promote the cellular uptake of HA-DOX NPs. Compared with DOX·HCl, the HA-mPEG2k-DOX NPs can significantly increase the DOX circulation time by 12.5 times, efficiently target the tumor tissues of CT26 tumor-bearing mice and remain for 72 hours. Therefore, the antitumor results in vivo indicated that the HA-mPEG2k-DOX NPs have the best anti-tumor effect while reducing the toxicity of the DOX. Overall, the cleavable PEGylated HA-mPEG2k-DOX NPs responding to pH-sensitive imine bonds, while actively targeting CD44-positive tumor cells, improve the dilemma of cellular uptake and delivery by the PEGylated nano delivery system.
Collapse
Affiliation(s)
- Xinyu Zhang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Minyi Zhao
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Nan Cao
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Wei Qin
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510006, China
| | - Meng Zhao
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Jun Wu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China. and Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, China and Research Institute of Sun Yat-sen University in Shenzhen, Shenzhen, 518057, China
| | - Dongjun Lin
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
22
|
Fleischmann D, Maslanka Figueroa S, Beck S, Abstiens K, Witzgall R, Schweda F, Tauber P, Goepferich A. Adenovirus-Mimetic Nanoparticles: Sequential Ligand-Receptor Interplay as a Universal Tool for Enhanced In Vitro/ In Vivo Cell Identification. ACS APPLIED MATERIALS & INTERFACES 2020; 12:34689-34702. [PMID: 32639709 DOI: 10.1021/acsami.0c10057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Viral infection patterns often rely on precisely coordinated sequences of distinct ligand-receptor interactions, leading in many cases to an outstanding target cell specificity. A successful mimicry of viral targeting strategies to create more site-specific nanoparticles (NPs) would therefore require particle-cell interactions to also be adequately controllable. In the present study, hetero-multivalent block-copolymer NPs present their attached ligands in a sterically controlled manner to create a sequential NP-cell interaction similar to the cell infiltration strategy of human adenovirus type 2. Targeting renal mesangial cells, particles therefore initially bind angiotensin II receptor type 1 (AT1r) on the cell surface via a structurally flexible AT1r antagonist. After a mandatory spatial approach, particle endocytosis is realized via binding of immobile αVβ3 integrins with a previously concealed secondary ligand, thereby creating a stepwise particle-cell interplay of primary NP attachment and subsequent uptake. Manufactured adenovirus-mimetic NPs show great avidity for both target motifs in vitro, leading to a substantial binding as well as subsequent cell uptake into target mesangial cells. Additionally, steric shielding of secondary ligand visibility leads to a highly controllable, sequential ligand-receptor interaction, whereby hetero-functional NPs activate mesangial cell surface integrins only after a successful prior binding to the AT1r. This stepwise cell identification significantly enhances mesangial cell specificity in co-culture assays with different off-target cells. Additionally, described NPs display excellent in vivo robustness by efficiently accumulating in the mesangium upon injection, thereby opening new paths for possible drug delivery applications.
Collapse
Affiliation(s)
- Daniel Fleischmann
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Sara Maslanka Figueroa
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Sebastian Beck
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Kathrin Abstiens
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Ralph Witzgall
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany
| | - Frank Schweda
- Department of Physiology II, Institute for Physiology, University of Regensburg, 93053 Regensburg, Germany
| | - Philipp Tauber
- Department of Physiology II, Institute for Physiology, University of Regensburg, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| |
Collapse
|
23
|
Maslanka Figueroa S, Fleischmann D, Beck S, Tauber P, Witzgall R, Schweda F, Goepferich A. Nanoparticles Mimicking Viral Cell Recognition Strategies Are Superior Transporters into Mesangial Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903204. [PMID: 32537398 PMCID: PMC7284201 DOI: 10.1002/advs.201903204] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 05/22/2023]
Abstract
Poor drug availability in the tissue of interest is a frequent cause of therapy failure. While nanotechnology has developed a plethora of nanocarriers for drug transport, their ability to unequivocally identify cells of interest remains moderate. Viruses are the ideal nanosized carriers as they are able to address their embedded nucleic acids with high specificity to their host cells. Here, it is reported that particles endowed with a virus-like ability to identify cells by three consecutive checks have a superior ability to recognize mesangial cells (MCs) in vivo compared to conventional nanoparticles. Mimicking the initial viral attachment followed by a stepwise target cell recognition process leads to a 5- to 15-fold higher accumulation in the kidney mesangium and extensive cell uptake compared to particles lacking one or both of the viral traits. These results highlight the relevance that the viral cell identification process has on specificity and its application on the targeting strategies of nanomaterials. More so, these findings pave the way for transporting drugs into the mesangium, a tissue that is pivotal in the development of diabetic nephropathy and for which currently no efficient pharmacotherapy exists.
Collapse
Affiliation(s)
| | - Daniel Fleischmann
- Department of Pharmaceutical TechnologyUniversity of RegensburgRegensburg93053Germany
| | - Sebastian Beck
- Department of Pharmaceutical TechnologyUniversity of RegensburgRegensburg93053Germany
| | - Philipp Tauber
- Department of Physiology IIUniversity of RegensburgRegensburg93053Germany
| | - Ralph Witzgall
- Department of Molecular and Cellular AnatomyUniversity of RegensburgRegensburg93053Germany
| | - Frank Schweda
- Department of Physiology IIUniversity of RegensburgRegensburg93053Germany
| | - Achim Goepferich
- Department of Pharmaceutical TechnologyUniversity of RegensburgRegensburg93053Germany
| |
Collapse
|
24
|
Maslanka Figueroa S, Fleischmann D, Beck S, Goepferich A. The Effect of Ligand Mobility on the Cellular Interaction of Multivalent Nanoparticles. Macromol Biosci 2020; 20:e1900427. [PMID: 32077622 DOI: 10.1002/mabi.201900427] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/27/2020] [Indexed: 12/23/2022]
Abstract
Multivalent nanoparticle binding to cells can be of picomolar avidity making such interactions almost as intense as those seen with antibodies. However, reducing nanoparticle design exclusively to avidity optimization by the choice of ligand and its surface density does not sufficiently account for controlling and understanding cell-particle interactions. Cell uptake, for example, is of paramount significance for a plethora of biomedical applications and does not exclusively depend on the intensity of multivalency. In this study, it is shown that the mobility of ligands tethered to particle surfaces has a substantial impact on particle fate upon binding. Nanoparticles carrying angiotensin-II tethered to highly mobile 5 kDa long poly(ethylene glycol) (PEG) chains separated by ligand-free 2 kDa short PEG chains show a superior accumulation in angiotensin-II receptor type 1 positive cells. In contrast, when ligand mobility is constrained by densely packing the nanoparticle surface with 5 kDa PEG chains only, cell uptake decreases by 50%. Remarkably, irrespective of ligand mobility and density both particle types have similar EC50 values in the 1-3 × 10-9 m range. These findings demonstrate that ligand mobility on the nanoparticle corona is an indispensable attribute to be considered in particle design to achieve optimal cell uptake via multivalent interactions.
Collapse
Affiliation(s)
- Sara Maslanka Figueroa
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Universitaetsstrasse 31, 93053, Germany
| | - Daniel Fleischmann
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Universitaetsstrasse 31, 93053, Germany
| | - Sebastian Beck
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Universitaetsstrasse 31, 93053, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Universitaetsstrasse 31, 93053, Germany
| |
Collapse
|
25
|
Multivalent HER2-binding polymer conjugates facilitate rapid endocytosis and enhance intracellular drug delivery. J Control Release 2019; 319:285-299. [PMID: 31899273 DOI: 10.1016/j.jconrel.2019.12.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/18/2019] [Accepted: 12/28/2019] [Indexed: 01/26/2023]
Abstract
Incorporating targeting moieties that recognize cancer-specific cellular markers can enhance specificity of anticancer nanomedicines. The HER2 receptor is overexpressed on numerous cancers, making it an attractive target. However, unlike many receptors that trigger endocytosis upon ligand binding, HER2 is an internalization-resistant receptor. As most chemotherapeutics act on intracellular targets, this presents a significant challenge for exploiting HER2 overexpression for improved tumor killing. However, hyper-crosslinking of HER2 has been shown to override the receptor's native behavior and trigger internalization. This research co-opts this crosslinking-mediated internalization for efficient intracellular delivery of an anticancer nanomedicine - specifically a HPMA copolymer-based drug delivery system. This polymeric carrier was conjugated with a small (7 kDa) HER2-binding affibody peptide to produce a panel of polymer-affibody conjugates with valences from 2 to 10 peptides per polymer chain. The effect of valence on surface binding and uptake was evaluated separately. All conjugates demonstrated similar (nanomolar) binding affinity towards HER2-positive ovarian carcinoma cells, but higher-valence conjugates induced more rapid endocytosis, with over 90% of the surface-bound conjugate internalized within 4 h. Furthermore, this enhancement was sensitive to crowding - high surface loading reduced conjugates' ability to crosslink receptors. Collectively, this evidence strongly supports a crosslinking-mediated endocytosis mechanism. Lead candidates from this panel achieved high intracellular delivery even at picomolar treatment concentrations; untargeted HPMA copolymers required 1000-fold higher treatment concentrations to achieve similar levels of intracellular accumulation. This increased intracellular delivery also translated to a more potent nanomedicine against HER2-positive cells; incorporation of the chemotherapeutic paclitaxel into this targeted carrier enhanced cytotoxicity over untargeted polymer-drug conjugate.
Collapse
|
26
|
Critical design criteria for engineering a nanoparticulate HIV-1 vaccine. J Control Release 2019; 317:322-335. [PMID: 31786187 DOI: 10.1016/j.jconrel.2019.11.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022]
Abstract
Inducing a long-lasting as well as broad and potent immune response by generating broadly neutralizing antibodies is a major goal and at the same time the main challenge of preventive HIV-1 vaccine design. Immunization with soluble, stabilized and native-like envelope (Env) glycoprotein so far only led to low neutralization breadth and displayed low immunogenicity. A promising approach to generate a potent immune response is the presentation of Env on the surface of nanoparticles. In this review, we will focus on two key processes essential for the induction of immune response that can be addressed by specific features of nanoparticulate carriers: first, the trafficking to and within distinct compartments of the lymph node, and second, the use of multivalent Env display allowing for high avidity interactions. To optimize these pivotal steps critical design criteria should be considered for the presentation of Env on nanoparticles. These include an optimal particle size below 100 nm, distances between two adjacent Env antigens of approximately 10-15 nm, an appropriate orientation of Env, and finally, the stability of both the Env attachment and the nanoparticle platform. Hence, an interdisciplinary approach that combines a suitable delivery system and a straightforward presentation of the Env antigen may have the potential to drive the immune response towards increased breadth and potency.
Collapse
|
27
|
Tang Y, Wang X, Li J, Nie Y, Liao G, Yu Y, Li C. Overcoming the Reticuloendothelial System Barrier to Drug Delivery with a "Don't-Eat-Us" Strategy. ACS NANO 2019; 13:13015-13026. [PMID: 31689086 DOI: 10.1021/acsnano.9b05679] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Overcoming the reticuloendothelial system (RES) has long been a vital challenge to nanoparticles as drug carriers. Modification of nanoparticles with polyethylene glycol helps them avoid clearance by macrophages but also suppresses their internalization by target cells. To overcome this paradox, we developed an RES-specific blocking system utilizing a "don't-eat-us" strategy. First, a CD47-derived, enzyme-resistant peptide ligand was designed and placed on liposomes (d-self-peptide-labeled liposome, DSL). After mainline administration, DSL was quickly adsorbed onto hepatic phagocyte membranes (including those of Kupffer cells and liver sinusoidal endothelial cells), forming a long-lasting mask that enclosed the cell membranes and thus reducing interactions between phagocytes and subsequently injected nanoparticles. Compared with blank conventional liposomes (CL), DSL blocked the RES at a much lower dose, and the effect was sustained for a much longer time, highly prolonging the elimination half-life of the subsequently injected nanoparticles. This "don't-eat-us" strategy by DSL was further verified on the brain-targeted delivery against a cryptococcal meningitis model, providing dramatically enhanced brain accumulation of the targeted delivery system and superior therapeutic outcome of model drug Amphotericin B compared with CL. Our study demonstrates a strategy that blocks the RES by masking phagocyte surfaces to prolong nanoparticle circulation time without excess modification and illustrates its utility in enhancing nanoparticle delivery.
Collapse
Affiliation(s)
- Yixuan Tang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing 400715 , P.R. China
| | - Xiaoyou Wang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing 400715 , P.R. China
| | - Jie Li
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing 400715 , P.R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials , Sichuan University , Sichuan 610065 , P.R. China
| | - Guojian Liao
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing 400715 , P.R. China
| | - Yang Yu
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing 400715 , P.R. China
| | - Chong Li
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing 400715 , P.R. China
| |
Collapse
|
28
|
Abstract
Poor target cell specificity is currently a major shortcoming of nanoparticles (NPs) used for biomedical applications. It causes significant material loss to off-target sites and poor availability at the intended delivery site. To overcome this limitation, we designed particles that identify cells in a virus-like manner. As a blueprint, we chose a mechanism typical of influenza A virus particles in which ectoenzymatic hemagglutinin activation by target cells is a mandatory prerequisite for binding to a secondary target structure that finally confirms cell identity and allows for uptake of the virus. We developed NPs that probe mesangial cells for the presence of angiotensin-converting enzyme on their surface using angiotensin I (Ang-I) as a proligand. This initial interaction enzymatically transforms Ang-I to a secondary ligand angiotensin II (Ang-II) that has the potential to bind in a second stage to Ang-II type-1 receptor (AT1R). The presence of the receptor confirms the target cell identity and triggers NP uptake via endocytosis. Our virus-mimetic NPs showed outstanding target-cell affinity with picomolar avidities and were able to selectively identify these cells in the presence of 90% off-target cells that carried only the AT1R. Our results demonstrate that the design of virus-mimetic cell interactive NPs is a valuable strategy to enhance NP specificity for therapeutic and diagnostic applications. Our set of primary and secondary targets is particularly suited for the identification of mesangial cells that play a pivotal role in diabetic nephropathy, one of the leading causes of renal failure, for which currently no treatment exists.
Collapse
|
29
|
Weldon C, Ji T, Nguyen MT, Rwei A, Wang W, Hao Y, Zhao C, Mehta M, Wang BY, Tsui J, Marini RP, Kohane DS. Nanoscale Bupivacaine Formulations To Enhance the Duration and Safety of Intravenous Regional Anesthesia. ACS NANO 2019; 13:18-25. [PMID: 30351910 DOI: 10.1021/acsnano.8b05408] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Intravenous regional anesthesia (IVRA; Bier block) is commonly used to anesthetize an extremity for surgery. Limitations of the procedure include pain from the required tourniquet, the toxicity that can occur from systemic release of local anesthetics, and the lack of postoperative pain relief. We hypothesized that the nanoencapsulation of the local anesthetic would prolong local anesthesia and enhance safety. Here, we developed an ∼15 nm micellar bupivacaine formulation (M-Bup) and tested it in a rat tail vein IVRA model, in which active agents were restricted in the tail by a tourniquet for 15 min. After tourniquet removal, M-Bup provided local anesthesia for 4.5 h, which was two times longer than that from a larger dose of free bupivacaine. Approximately 100 nm liposomal bupivacaine (L-Bup) with the same drug dose as M-Bup did not cause anesthesia. Blood levels of bupivacaine after tourniquet removal were lower in animals receiving M-Bup than L-Bup or free bupivacaine, demonstrating enhanced safety. Tissue reaction to M-Bup was benign.
Collapse
Affiliation(s)
- Christopher Weldon
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Surgery , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Tianjiao Ji
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Minh-Thuy Nguyen
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Alina Rwei
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Weiping Wang
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Yi Hao
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Chao Zhao
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Manisha Mehta
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Bruce Y Wang
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Jonathan Tsui
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Robert P Marini
- Division of Comparative Medicine , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| |
Collapse
|
30
|
Abstiens K, Gregoritza M, Goepferich AM. Ligand Density and Linker Length are Critical Factors for Multivalent Nanoparticle-Receptor Interactions. ACS APPLIED MATERIALS & INTERFACES 2019; 11:1311-1320. [PMID: 30521749 DOI: 10.1021/acsami.8b18843] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Although there are a large number of studies available for the evaluation of the therapeutic efficacy of targeted polymeric nanoparticles, little is known about the critical attributes that can further influence their uptake into target cells. In this study, varying cRGD ligand densities (0-100% surface functionalization) were combined with different poly(ethylene glycol) (PEG) spacer lengths (2/3.5/5 kDa), and the specific receptor binding of targeted core-shell structured poly(lactic- co-glycolic acid)/poly(lactic acid)-PEG nanoparticles was evaluated using αvβ3 integrin-overexpressing U87MG glioblastoma cells. Nanoparticles with 100% surface functionalization and short PEG2k linkers displayed a high propensity to form colloidal clusters, allowing for the cooperative binding to integrin receptors on the cellular membrane. In contrast, the high flexibility of longer PEG chains enhanced the chance of ligand entanglement and shrouding, decreasing the number of ligand-receptor binding events. As a result, the combination of short PEG2k linkers and a high cRGD surface modification synergistically increased the uptake of nanoparticles into target cells. Even though to date, the nanoparticle size and its degree of functionalization are considered to be the major determinants for controlling the uptake efficiency of targeted colloids, these results strongly suggest that the role of the linker length should be carefully taken into consideration for the design of targeted drug delivery formulations to maximize the therapeutic efficacy and minimize adverse side effects.
Collapse
Affiliation(s)
- Kathrin Abstiens
- Department of Pharmaceutical Technology, Faculty of Chemistry and Pharmacy , University of Regensburg , 93040 Regensburg , Germany
| | - Manuel Gregoritza
- Department of Pharmaceutical Technology, Faculty of Chemistry and Pharmacy , University of Regensburg , 93040 Regensburg , Germany
| | - Achim M Goepferich
- Department of Pharmaceutical Technology, Faculty of Chemistry and Pharmacy , University of Regensburg , 93040 Regensburg , Germany
| |
Collapse
|
31
|
Rabanel JM, Adibnia V, Tehrani SF, Sanche S, Hildgen P, Banquy X, Ramassamy C. Nanoparticle heterogeneity: an emerging structural parameter influencing particle fate in biological media? NANOSCALE 2019; 11:383-406. [PMID: 30560970 DOI: 10.1039/c8nr04916e] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Drug nanocarriers' surface chemistry is often presumed to be uniform. For instance, the polymer surface coverage and distribution of ligands on nanoparticles are described with averaged values obtained from quantification techniques based on particle populations. However, these averaged values may conceal heterogeneities at different levels, either because of the presence of particle sub-populations or because of surface inhomogeneities, such as patchy surfaces on individual particles. The characterization and quantification of chemical surface heterogeneities are tedious tasks, which are rather limited by the currently available instruments and research protocols. However, heterogeneities may contribute to some non-linear effects observed during the nanoformulation optimization process, cause problems related to nanocarrier production scale-up and correlate with unexpected biological outcomes. On the other hand, heterogeneities, while usually unintended and detrimental to nanocarrier performance, may, in some cases, be sought as adjustable properties that provide NPs with unique functionality. In this review, results and processes related to this issue are compiled, and perspectives and possible analytical developments are discussed.
Collapse
Affiliation(s)
- Jean-Michel Rabanel
- Centre INRS Institut Armand-Frappier, 531, boul. des Prairies, Laval, QC H7V 1B7, Canada.
| | - Vahid Adibnia
- Faculté de Pharmacie, Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, Québec H3C 3J7, Canada.
| | - Soudeh F Tehrani
- Faculté de Pharmacie, Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, Québec H3C 3J7, Canada.
| | - Steven Sanche
- Faculté de Pharmacie, Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, Québec H3C 3J7, Canada.
| | - Patrice Hildgen
- Faculté de Pharmacie, Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, Québec H3C 3J7, Canada.
| | - Xavier Banquy
- Faculté de Pharmacie, Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, Québec H3C 3J7, Canada.
| | - Charles Ramassamy
- Centre INRS Institut Armand-Frappier, 531, boul. des Prairies, Laval, QC H7V 1B7, Canada.
| |
Collapse
|
32
|
Zhang Q, Reinhard BM. Ligand Density and Nanoparticle Clustering Cooperate in the Multivalent Amplification of Epidermal Growth Factor Receptor Activation. ACS NANO 2018; 12:10473-10485. [PMID: 30289688 PMCID: PMC6252274 DOI: 10.1021/acsnano.8b06141] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Multivalent presentation of ligands on nanoparticles (NPs) is considered a general strategy for enhancing receptor binding and activation through amplification of ligand-receptor interactions within the footprint of the individual NPs. The spatial clustering of ligand-functionalized NPs represents an additional, less well understood mechanism for increasing local ligand-receptor interactions, especially for receptors that form higher-order assemblies, such as the epidermal growth factor (EGF) receptor (EGFR). To shed light on the interplay between ligand density ( i.e., multivalency) and NP clustering in signal amplification, we apply EGF-functionalized 72 ± 1 nm gold nanoparticles (NP-EGF) with known ligand loading (10-200 EGF/NP) as quantifiable and experimentally tractable units of EGFR activation and characterize the NP-mediated amplification of EGFR phosphorylation as a function of both EGF surface density and NP-EGF clustering for two cancer cell lines (HeLa and MDA-MB-468). The measurements confirm a strong multivalent amplification of EGFR phosphorylation through NP-EGF on the cellular level that results in EGF-loading-dependent maximum EGFR phosphorylation levels. A microscopic analysis of NP-EGF-induced EGFR phosphorylation reveals a heterogeneous spatial distribution of EGFR activation across the cell surface. Clustering of multivalent NP-EGF on sub-diffraction-limited length scales is found to result in a local enhancement of EGFR phosphorylation in signaling "hot spots" from where the signal can spread laterally in an EGF-independent fashion. Increasing EGF loadings of the NP enhances NP-EGF clustering and intensifies EGFR phosphorylation. These observations suggest that NP-EGF clustering and the associated local enhancement of ligand-receptor interactions are intrinsic components of the multivalent amplification of phosphorylation for the heterogeneously distributed EGFR through NP-EGF.
Collapse
Affiliation(s)
- Qianyun Zhang
- Department of Chemistry and The Photonics Center , Boston University , Boston , Massachusetts 02215 , United States
| | - Björn M Reinhard
- Department of Chemistry and The Photonics Center , Boston University , Boston , Massachusetts 02215 , United States
| |
Collapse
|
33
|
Roointan A, Kianpour S, Memari F, Gandomani M, Gheibi Hayat SM, Mohammadi-Samani S. Poly(lactic-co-glycolic acid): The most ardent and flexible candidate in biomedicine! INT J POLYM MATER PO 2018. [DOI: 10.1080/00914037.2017.1405350] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Amir Roointan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sedigheh Kianpour
- Department of Pharmaceutical Biotechnology, Pharmaceutical Sciences Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Memari
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Molood Gandomani
- Department of Bioengineering, Biotechnology Research Center, Cyprus international University, Nicosia, Cyprus
| | - Seyed Mohammad Gheibi Hayat
- Student Research Committee, Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soliman Mohammadi-Samani
- Department of Pharmaceutics, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
34
|
Shaw SK, Liu W, Brennan SP, de Lourdes Betancourt-Mendiola M, Smith BD. Non-Covalent Assembly Method that Simultaneously Endows a Liposome Surface with Targeting Ligands, Protective PEG Chains, and Deep-Red Fluorescence Reporter Groups. Chemistry 2017; 23:12646-12654. [PMID: 28736857 DOI: 10.1002/chem.201702649] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Indexed: 12/28/2022]
Abstract
A new self-assembly method is used to rapidly functionalize the surface of liposomes without perturbing the membrane integrity or causing leakage of the aqueous contents. The key molecule is a cholesterol-squaraine-PEG conjugate with three important structural elements: a cholesterol membrane anchor, a fluorescent squaraine docking station that allows rapid and high-affinity macrocycle threading, and a long PEG-2000 chain to provide steric shielding of the decorated liposome. The two-step method involves spontaneous insertion of the conjugate into the outer leaflet of pre-formed liposomes followed by squaraine threading with a tetralactam macrocycle that has appended targeting ligands. A macrocycle with six carboxylates permitted immobilization of intact fluorescent liposomes on the surface of cationic polymer beads, whereas a macrocycle with six zinc(II)-dipicolylamine units enabled selective targeting of anionic membranes, including agglutination of bacteria in the presence of human cells.
Collapse
Affiliation(s)
- Scott K Shaw
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN., 46545, USA
| | - Wenqi Liu
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN., 46545, USA
| | - Seamus P Brennan
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN., 46545, USA
| | | | - Bradley D Smith
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN., 46545, USA
| |
Collapse
|
35
|
Kalia P, Jain A, Radha Krishnan R, Demuth DR, Steinbach-Rankins JM. Peptide-modified nanoparticles inhibit formation of Porphyromonas gingivalis biofilms with Streptococcus gordonii. Int J Nanomedicine 2017; 12:4553-4562. [PMID: 28790818 PMCID: PMC5488760 DOI: 10.2147/ijn.s139178] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
PURPOSE The interaction of Porphyromonas gingivalis with commensal streptococci promotes P. gingivalis colonization of the oral cavity. We previously showed that a synthetic peptide (BAR) derived from Streptococcus gordonii potently inhibited the formation of P. gingivalis/S. gordonii biofilms (IC50 =1.3 µM) and reduced P. gingivalis virulence in a mouse model of periodontitis. Thus, BAR represents a novel therapeutic to control periodontitis by limiting P. gingivalis colonization of the oral cavity. Here, we sought to develop drug-delivery vehicles for potential use in the oral cavity that comprise BAR-modified poly(lactic-co-glycolic)acid (PLGA) nanoparticles (NPs). METHODS PLGA-NPs were initially modified with palmitylated avidin and subsequently conjugated with biotinylated BAR. The extent of BAR modification was quantified using a fluorescent-labeled peptide. Inhibition of P. gingivalis adherence to S. gordonii by BAR-modified NPs was compared with free peptide using a two-species biofilm model. RESULTS BAR-modified NPs exhibited an average size of 99±29 nm and a more positive surface charge than unmodified NPs (zeta potentials of -7 mV and -25 mV, respectively). Binding saturation occurred when 37 nmol BAR/mg of avidin-NPs was used, which resulted in a payload of 7.42 nmol BAR/mg NPs. BAR-modified NPs bound to P. gingivalis in a dose-dependent manner and more potently inhibited P. gingivalis/S. gordonii adherence and biofilm formation relative to an equimolar amount of free peptide (IC50 of 0.2 µM versus 1.3 µM). BAR-modified NPs also disrupted the preformed P. gingivalis/S. gordonii biofilms more effectively than free peptide. Finally, we demonstrate that BAR-modified NPs promoted multivalent association with P. gingivalis, providing an explanation for the increased effectiveness of NPs. CONCLUSION These results indicate that BAR-modified NPs deliver a higher local dose of peptide and may represent a more effective therapeutic approach to limit P. gingivalis colonization of the oral cavity compared to treatment with formulations of free peptide.
Collapse
Affiliation(s)
- Paridhi Kalia
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry
| | - Ankita Jain
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry
| | - Ranjith Radha Krishnan
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry
| | - Donald R Demuth
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry.,Department of Microbiology and Immunology, University of Louisville School of Medicine
| | - Jill M Steinbach-Rankins
- Department of Microbiology and Immunology, University of Louisville School of Medicine.,Department of Bioengineering, University of Louisville Speed School of Engineering.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine.,Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
36
|
Lo Giudice MC, Meder F, Polo E, Thomas SS, Alnahdi K, Lara S, Dawson KA. Constructing bifunctional nanoparticles for dual targeting: improved grafting and surface recognition assessment of multiple ligand nanoparticles. NANOSCALE 2016; 8:16969-16975. [PMID: 27714073 DOI: 10.1039/c6nr05478a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Nanoparticles (NPs) functionalized with two active targeting ligands have been proposed in drug delivery for their promising capability to stimulate different pathways with one object. Due to the multivalency, the construction and analysis of the effective surface of such bifunctional nanoparticles, however, is significantly more complex than for nanoparticles bearing only one ligand. Here, we optimize construction and analysis of bifunctional NPs containing recognizable combinations of human serum albumin (HSA), transferrin (Tf), and epidermal growth factor (EGF) on fluorescent silica NPs grafted via common polyethylene glycol (PEG) linkers as a model system. Combined with an overall protein quantification, a mapping of exposed recognizable sequences using monoclonal antibodies conjugated to gold nanoparticles (AuNPs) or quantum dots (QDs) for enhanced spectroscopic and microscopic detection revealed that active protein sequences can be one to two orders of magnitude lower than overall conjugated proteins while possessing specific cellular recognition. In addition, we found that common conjugation strategies lead to a large excess of non-specifically compared to covalently bound ligands and instabilities that may impact targeting. These can be avoided by certain synthetic conditions presented here for effective exploitation of multivalent surfaces in nanomedicine.
Collapse
Affiliation(s)
- Maria Cristina Lo Giudice
- Centre for BioNano Interactions, University College Dublin, School of Chemistry and Chemical Biology, Belfield 4, Dublin, Ireland.
| | - Fabian Meder
- Centre for BioNano Interactions, University College Dublin, School of Chemistry and Chemical Biology, Belfield 4, Dublin, Ireland.
| | - Ester Polo
- Centre for BioNano Interactions, University College Dublin, School of Chemistry and Chemical Biology, Belfield 4, Dublin, Ireland.
| | - Steffi S Thomas
- Centre for BioNano Interactions, University College Dublin, School of Chemistry and Chemical Biology, Belfield 4, Dublin, Ireland.
| | - Kholoud Alnahdi
- Centre for BioNano Interactions, University College Dublin, School of Chemistry and Chemical Biology, Belfield 4, Dublin, Ireland.
| | - Sandra Lara
- Centre for BioNano Interactions, University College Dublin, School of Chemistry and Chemical Biology, Belfield 4, Dublin, Ireland.
| | - Kenneth A Dawson
- Centre for BioNano Interactions, University College Dublin, School of Chemistry and Chemical Biology, Belfield 4, Dublin, Ireland.
| |
Collapse
|
37
|
Abd Ellah NH, Abouelmagd SA. Surface functionalization of polymeric nanoparticles for tumor drug delivery: approaches and challenges. Expert Opin Drug Deliv 2016; 14:201-214. [DOI: 10.1080/17425247.2016.1213238] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Noura H. Abd Ellah
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Sara A. Abouelmagd
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
38
|
Peck EM, Battles PM, Rice DR, Roland FM, Norquest KA, Smith BD. Pre-Assembly of Near-Infrared Fluorescent Multivalent Molecular Probes for Biological Imaging. Bioconjug Chem 2016; 27:1400-10. [PMID: 27088305 DOI: 10.1021/acs.bioconjchem.6b00173] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A programmable pre-assembly method is described and shown to produce near-infrared fluorescent molecular probes with tunable multivalent binding properties. The modular assembly process threads one or two copies of a tetralactam macrocycle onto a fluorescent PEGylated squaraine scaffold containing a complementary number of docking stations. Appended to the macrocycle periphery are multiple copies of a ligand that is known to target a biomarker. The structure and high purity of each threaded complex was determined by independent spectrometric methods and also by gel electrophoresis. Especially helpful were diagnostic red-shift and energy transfer features in the absorption and fluorescence spectra. The threaded complexes were found to be effective multivalent molecular probes for fluorescence microscopy and in vivo fluorescence imaging of living subjects. Two multivalent probes were prepared and tested for targeting of bone in mice. A pre-assembled probe with 12 bone-targeting iminodiacetate ligands produced more bone accumulation than an analogous pre-assembled probe with six iminodiacetate ligands. Notably, there was no loss in probe fluorescence at the bone target site after 24 h in the living animal, indicating that the pre-assembled fluorescent probe maintained very high mechanical and chemical stability on the skeletal surface. The study shows how this versatile pre-assembly method can be used in a parallel combinatorial manner to produce libraries of near-infrared fluorescent multivalent molecular probes for different types of imaging and diagnostic applications, with incremental structural changes in the number of targeting groups, linker lengths, linker flexibility, and degree of PEGylation.
Collapse
Affiliation(s)
- Evan M Peck
- Department of Chemistry and Biochemistry, 236 Nieuwland Science Hall, University of Notre Dame , Notre Dame, Indiana 46556, United States
| | - Paul M Battles
- Department of Chemistry and Biochemistry, 236 Nieuwland Science Hall, University of Notre Dame , Notre Dame, Indiana 46556, United States
| | - Douglas R Rice
- Department of Chemistry and Biochemistry, 236 Nieuwland Science Hall, University of Notre Dame , Notre Dame, Indiana 46556, United States
| | - Felicia M Roland
- Department of Chemistry and Biochemistry, 236 Nieuwland Science Hall, University of Notre Dame , Notre Dame, Indiana 46556, United States
| | - Kathryn A Norquest
- Department of Chemistry and Biochemistry, 236 Nieuwland Science Hall, University of Notre Dame , Notre Dame, Indiana 46556, United States
| | - Bradley D Smith
- Department of Chemistry and Biochemistry, 236 Nieuwland Science Hall, University of Notre Dame , Notre Dame, Indiana 46556, United States
| |
Collapse
|
39
|
Ieranò C, Portella L, Lusa S, Salzano G, D'Alterio C, Napolitano M, Buoncervello M, Macchia D, Spada M, Barbieri A, Luciano A, Barone MV, Gabriele L, Caraglia M, Arra C, De Rosa G, Scala S. CXCR4-antagonist Peptide R-liposomes for combined therapy against lung metastasis. NANOSCALE 2016; 8:7562-7571. [PMID: 26983756 DOI: 10.1039/c5nr06335c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The chemokine CXCL12 activates CXCR4, initiating multiple pathways that control immune cell trafficking, angiogenesis and embryogenesis; CXCR4 is also overexpressed in multiple tumors affecting metastatic dissemination. While there has been great enthusiasm for exploiting the CXCR4-CXCL12 axis as a target in cancer therapy, to date the promise has yet to be fulfilled. A new class of CXCR4-antagonist cyclic peptides was recently developed and the compound named Peptide R was identified as the most active. With the intent to improve the efficacy and biodistribution of Peptide R, stealth liposomes decorated with Peptide R were developed (PL-Peptide R). In vitro PL-Peptide R efficiently inhibited CXCR4-dependent migration and in vivo it significantly reduced lung metastases and increased overall survival in B16-CXCR4 injected C57BL/6 mice. To evaluate if PL-Peptide R could also be a drug delivery system for CXCR4 expressing tumors, the PL-Peptide R was loaded with doxorubicin (DOX) (PL-Peptide R-DOX). PL-Peptide R-DOX efficiently delivered DOX to CXCR4 expressing cell lines with a consequent decrease in the DOX IC50 efficient dose. In vivo, B16-CXCR4 injected C57BL/6 mice treated with PL-Peptide R-DOX developed fewer lung metastases compared to PL-DOX treated mice. This work provides the proof-of-concept to prevent metastasis by using combined nanomedicine.
Collapse
Affiliation(s)
- Caterina Ieranò
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Luigi Portella
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Sara Lusa
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
| | - Giuseppina Salzano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy. and Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Ave, Boston, MA, USA
| | - Crescenzo D'Alterio
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Maria Napolitano
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Maria Buoncervello
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Daniele Macchia
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Massimo Spada
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy
| | - Antonio Luciano
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy
| | - Maria Vittoria Barone
- Department of Translational Medical Science and European Laboratory for the Investigation of Food Induced Disease (ELFID), University of Naples, Federico II, Via S. Pansini 5, 80131, Naples, Italy
| | - Lucia Gabriele
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via L. De Crecchio 7, 80138, Naples, Italy
| | - Claudio Arra
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
| | - Stefania Scala
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| |
Collapse
|
40
|
Peng Q, Mu H. The potential of protein-nanomaterial interaction for advanced drug delivery. J Control Release 2016; 225:121-32. [PMID: 26812004 DOI: 10.1016/j.jconrel.2016.01.041] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 01/22/2016] [Accepted: 01/22/2016] [Indexed: 02/05/2023]
Abstract
Nanomaterials, like nanoparticles, micelles, nano-sheets, nanotubes and quantum dots, have great potentials in biomedical fields. However, their delivery is highly limited by the formation of protein corona upon interaction with endogenous proteins. This new identity, instead of nanomaterial itself, would be the real substance the organs and cells firstly encounter. Consequently, the behavior of nanomaterials in vivo is uncontrollable and some undesired effects may occur, like rapid clearance from blood stream; risk of capillary blockage; loss of targeting capacity; and potential toxicity. Therefore, protein-nanomaterial interaction is a great challenge for nanomaterial systems and should be inhibited. However, this interaction can also be used to functionalize nanomaterials by forming a selected protein corona. Unlike other decoration using exogenous molecules, nanomaterials functionalized by selected protein corona using endogenous proteins would have greater promise for clinical use. In this review, we aim to provide a comprehensive understanding of protein-nanomaterial interaction. Importantly, a discussion about how to use such interaction is launched and some possible applications of such interaction for advanced drug delivery are presented.
Collapse
Affiliation(s)
- Qiang Peng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark.
| | - Huiling Mu
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| |
Collapse
|
41
|
Hennig R, Ohlmann A, Staffel J, Pollinger K, Haunberger A, Breunig M, Schweda F, Tamm ER, Goepferich A. Multivalent nanoparticles bind the retinal and choroidal vasculature. J Control Release 2015; 220:265-274. [DOI: 10.1016/j.jconrel.2015.10.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 10/22/2022]
|
42
|
Hennig R, Goepferich A. Nanoparticles for the treatment of ocular neovascularizations. Eur J Pharm Biopharm 2015; 95:294-306. [DOI: 10.1016/j.ejpb.2015.02.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/13/2015] [Accepted: 02/27/2015] [Indexed: 12/27/2022]
|
43
|
Lane LA, Qian X, Nie S. SERS Nanoparticles in Medicine: From Label-Free Detection to Spectroscopic Tagging. Chem Rev 2015; 115:10489-529. [DOI: 10.1021/acs.chemrev.5b00265] [Citation(s) in RCA: 607] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Lucas A. Lane
- Departments
of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, Health Sciences Research Building,
Room E116, 1760 Haygood Drive, Atlanta, Georgia 30322, United States
| | - Ximei Qian
- Departments
of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, Health Sciences Research Building,
Room E116, 1760 Haygood Drive, Atlanta, Georgia 30322, United States
| | - Shuming Nie
- Departments
of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, Health Sciences Research Building,
Room E116, 1760 Haygood Drive, Atlanta, Georgia 30322, United States
- College
of Engineering and Applied Sciences, Nanjing University, 22 Hankou
Road, Nanjing, Jiangsu Province 210093, China
| |
Collapse
|
44
|
Hennig R, Veser A, Kirchhof S, Goepferich A. Branched Polymer-Drug Conjugates for Multivalent Blockade of Angiotensin II Receptors. Mol Pharm 2015; 12:3292-302. [PMID: 26252154 DOI: 10.1021/acs.molpharmaceut.5b00301] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The use of angiotensin receptor blockers (ARBs) for treatment of ocular diseases associated with neovascularizations, such as proliferative diabetic retinopathy, shows tremendous promise but is presently limited due to short intravitreal half-life. Conjugation of ARB molecules to branched polymers could vastly augment their therapeutic efficacy. EXP3174, a potent non-peptide ARB, was conjugated to branched poly(ethylene glycol) (PEG) and poly(amido amine) (PAMAM) dendrimers: 7.8 ligand molecules were tethered to each 40 kDa PEG molecule whereas 16.7 ligand molecules were linked to each PAMAM generation 5 dendrimer. The multivalent PEG and PAMAM conjugates blocked AT1R signaling with an IC50 of 224 and 36.3 nM, respectively. The 6-fold higher affinity of the multivalent ligand-conjugated PAMAM dendrimers was due to their unique microarchitecture and ability to suppress polymer-drug interactions. Remarkably, both polymer-drug conjugates exhibited no cytotoxicity, in stark contrast to plain PAMAM dendrimers. With sufficiently long vitreous half-lives, both synthesized polymer-ARB conjugates have the potential to pave a new path for the therapy of ocular diseases accompanied by retinal neovascularizations.
Collapse
Affiliation(s)
- Robert Hennig
- Department of Pharmaceutical Technology, University of Regensburg , Regensburg, Germany
| | - Anika Veser
- Department of Pharmaceutical Technology, University of Regensburg , Regensburg, Germany
| | - Susanne Kirchhof
- Department of Pharmaceutical Technology, University of Regensburg , Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg , Regensburg, Germany
| |
Collapse
|
45
|
Sobczynski DJ, Fish MB, Fromen CA, Carasco-Teja M, Coleman RM, Eniola-Adefeso O. Drug carrier interaction with blood: a critical aspect for high-efficient vascular-targeted drug delivery systems. Ther Deliv 2015; 6:915-34. [PMID: 26272334 PMCID: PMC4618056 DOI: 10.4155/tde.15.38] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Vascular wall endothelial cells control several physiological processes and are implicated in many diseases, making them an attractive candidate for drug targeting. Vascular-targeted drug carriers (VTCs) offer potential for reduced side effects and improved therapeutic efficacy, however, only limited therapeutic success has been achieved to date. This is perhaps due to complex interactions of VTCs with blood components, which dictate VTC transport and adhesion to endothelial cells. This review focuses on VTC interaction with blood as well as novel 'bio-inspired' designs to mimic and exploit features of blood in VTC development. Advanced approaches for enhancing VTCs are discussed along with applications in regenerative medicine, an area of massive potential growth and expansion of VTC utility in the near future.
Collapse
Affiliation(s)
- Daniel J Sobczynski
- Department of Chemical Engineering, University of Michigan, Ann Arbor MI, USA 48109
| | - Margaret B Fish
- Department of Chemical Engineering, University of Michigan, Ann Arbor MI, USA 48109
| | - Catherine A Fromen
- Department of Chemical Engineering, University of Michigan, Ann Arbor MI, USA 48109
| | - Mariana Carasco-Teja
- Department of Chemical Engineering, University of Michigan, Ann Arbor MI, USA 48109
| | - Rhima M Coleman
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA 48109
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor MI, USA 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA 48109
| |
Collapse
|
46
|
Wang X, Song Y, Su Y, Tian Q, Li B, Quan J, Deng Y. Are PEGylated liposomes better than conventional liposomes? A special case for vincristine. Drug Deliv 2015; 23:1092-100. [PMID: 26024386 DOI: 10.3109/10717544.2015.1027015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cancer poses a significant threat to human health worldwide, and many therapies have been used for its palliative and curative treatments. Vincristine has been extensively used in chemotherapy. However, there are two major challenges concerning its applications in various tumors: (1) Vincristine's antitumor mechanism is cell-cycle-specific, and the duration of its exposure to tumor cells can significantly affect its antitumor activity and (2) Vincristine is widely bio-distributed and can be rapidly eliminated. One solution to these challenges is the encapsulation of vincristine into liposomes. Vincristine can be loaded into conventional liposomes, but it quickly leak out owing to its high membrane permeability. Numerous approaches have been attempted to overcome this problem. Vincristine has been loaded into PEGylated liposomes to prolong circulation time and improve tumor accumulation. These liposomes indeed prolong circulation time, but the payout characteristic of vincristine is severer, resulting in a compromised outcome rather than a better efficacy compared to conventional sphingomyelin (SM)/cholesterol (Chol) liposomes. In 2012, the USA Food and Drug Administration (FDA) approved SM/Chol liposomal vincristine (Marqibo®) for commercial use. In this review, we mainly focus on the drug's rapid leakage problem and the potentially relevant solutions that can be applied during the development of liposomal vincristine and the reason for conventional liposomal vincristine rather than PEGylated liposomes has access to the market.
Collapse
Affiliation(s)
- Xuling Wang
- a School of Pharmacy, Shenyang Pharmaceutical University , Shenyang , P.R. China
| | - Yanzhi Song
- a School of Pharmacy, Shenyang Pharmaceutical University , Shenyang , P.R. China
| | - Yuqing Su
- a School of Pharmacy, Shenyang Pharmaceutical University , Shenyang , P.R. China
| | - Qingjing Tian
- a School of Pharmacy, Shenyang Pharmaceutical University , Shenyang , P.R. China
| | - Boqun Li
- a School of Pharmacy, Shenyang Pharmaceutical University , Shenyang , P.R. China
| | - Jingjing Quan
- a School of Pharmacy, Shenyang Pharmaceutical University , Shenyang , P.R. China
| | - Yihui Deng
- a School of Pharmacy, Shenyang Pharmaceutical University , Shenyang , P.R. China
| |
Collapse
|
47
|
Hennig R, Pollinger K, Tessmar J, Goepferich A. Multivalent targeting of AT1 receptors with angiotensin II-functionalized nanoparticles. J Drug Target 2015; 23:681-9. [PMID: 25950599 DOI: 10.3109/1061186x.2015.1035276] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The angiotensin II receptor type 1 (AT1R) is a G protein-coupled receptor of paramount significance since it is overexpressed in a number of diseased tissues that are highly attractive for nanoparticle targeting. However, it is also expressed at physiological levels in healthy tissue. Multivalent interactions mediated by multiple AT1R-binding moieties per nanoparticle could promote a high binding avidity to AT1R overexpressing cells and concomitantly spare off-target tissue. To investigate the feasibility of this approach, angiotensin II was thiolated and conjugated to PEGylated quantum dots. Nanoparticle binding, uptake and affinity to several cell lines was investigated in detail. The colloids were rapidly taken up by clathrin-mediated endocytosis into AT1R-expressing cells and showed no interaction with receptor negative cells. The EC50 of the thiolated angiotensin II was determined to be 261 nM, whereas the ligand-conjugated Qdots activated the receptor with an EC50 of 8.9 nM. This 30-fold higher affinity of the nanoparticles compared to the unconjugated peptide clearly demonstrated the presence of multivalent effects when using agonist-targeted nanoparticles. Our study provides compelling evidence that, despite being immediately endocytosed, Ang II-coupled nanoparticles exert potent multivalent ligand-receptor interactions that can be used to establish high affinities to an AT1R overexpressing cell and tissue.
Collapse
Affiliation(s)
- Robert Hennig
- a Department of Pharmaceutical Technology , University of Regensburg , Regensburg , Germany and
| | - Klaus Pollinger
- a Department of Pharmaceutical Technology , University of Regensburg , Regensburg , Germany and
| | - Joerg Tessmar
- b Department for Functional Materials in Medicine and Dentistry , University Hospital of Wuerzburg , Wuerzburg , Germany
| | - Achim Goepferich
- a Department of Pharmaceutical Technology , University of Regensburg , Regensburg , Germany and
| |
Collapse
|
48
|
Fourmy D, Carrey J, Gigoux V. Targeted nanoscale magnetic hyperthermia: challenges and potentials of peptide-based targeting. Nanomedicine (Lond) 2015; 10:893-6. [DOI: 10.2217/nnm.14.236] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Daniel Fourmy
- University of Toulouse 3, EA4552 Receptor & Therapeutic Targeting of Cancers, Toulouse, France
| | - Julian Carrey
- Université de Toulouse 3, INSA, CNRS UMR5215, Laboratoire de Physique et Chimie des Nano-Objets (LPCNO), Toulouse, France
| | - Véronique Gigoux
- University of Toulouse 3, EA4552 Receptor & Therapeutic Targeting of Cancers, Toulouse, France
| |
Collapse
|
49
|
|