1
|
Xi W, Wu W, Zhou L, Zhang Q, Yang S, Huang L, Lu Y, Wang J, Chi X, Kang Y. Multifunctional nanoparticles confers both multiple inflammatory mediators scavenging and macrophage polarization for sepsis therapy. Mater Today Bio 2025; 30:101421. [PMID: 39811612 PMCID: PMC11732566 DOI: 10.1016/j.mtbio.2024.101421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/29/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Sepsis is a serious and life-threatening condition, which can lead to organ failure and death clinically. Abnormally increased cell-free DNA (cfDNA) and inflammatory cytokines are involved in the development and progression of sepsis. Thus, cfDNA clearance and down-regulation of inflammatory factors are essential for the effective treatment of sepsis. Here we designed and constructed a polydopamine-based multifunctional nanoparticle for the treatment of sepsis. These nanoparticles (NPs) are composed of polydopamine (PDA) grafted with cationic polyethyleneimine (PEI). On the one hand, the NPs can utilize the electrostatic interaction to effectively adsorb cfDNA in blood, then effectively inhibiting the activation of toll like receptors (TLRs) and nuclear factor kappa B (NF-κB) pathways induced by cfDNA. On the other hand, the NPs have an immunomodulatory function, which can effectively convert pro-inflammatory macrophage (M1) into anti-inflammatory macrophage (M2), thus reduce the release of inflammatory cytokines and slow down the inflammatory storm of sepsis. In addition, the NPs possess good reactive oxygen species (ROS) scavenging ability. Briefly, the effective treatment of sepsis can be achieved by multiple strategies of effectively capturing the inflammatory triggering factor cfDNA, modulating the polarization of M1 macrophage to M2 macrophage and scavenging ROS, which has a promising clinical application.
Collapse
Affiliation(s)
- Wenjie Xi
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Weijie Wu
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lili Zhou
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Qi Zhang
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shushu Yang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lihong Huang
- Department of Orthopaedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, China
| | - Yijun Lu
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jing Wang
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinjin Chi
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yang Kang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
2
|
Yadav P, Wairkar S. Tofacitinib in focus: Fascinating voyage from conventional formulations to novel delivery systems. Int J Pharm 2025; 671:125253. [PMID: 39842741 DOI: 10.1016/j.ijpharm.2025.125253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/11/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Tofacitinib, a Janus kinase (JAK) inhibitor, has emerged as a primary therapeutic agent for managing autoimmune diseases such as rheumatoid arthritis, psoriatic arthritis, dermatitis and ulcerative colitis. By inhibiting the phosphorylation of JAK enzymes, tofacitinib prevents their activation within the JAK-STAT signaling pathway, which is vital for inflammatory responses. However, the tofacitinib delivery presents significant challenges, including pH-dependent solubility, poor permeability and susceptibility to oral degradation. This review provides an in-depth analysis of current and emerging formulations to enhance the delivery and efficiency of tofacitinib. This review highlights the physicochemical, pharmacodynamic and pharmacokinetic properties of tofacitinib. Additionally, it discusses various strategies, including oral modified release formulations, topical/transdermal delivery utilizing lipid-based and polymeric systems, and parenteral delivery systems. Recent advancements in nanotechnology, such as liposomes, micelles, keratinocyte exosomes, proposomes, proglycosomes, transethosomes, squalenyl nanoparticles and lyotropic liquid crystalline nanoparticles, are explored as potential nanocarriers to existing delivery constraints. The development of advanced tofacitinib delivery systems can address the challenges in its delivery and improve therapeutic outcomes and patient compliance, paving the way for enhanced treatment strategies in autoimmune and inflammatory conditions.
Collapse
Affiliation(s)
- Priti Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India.
| |
Collapse
|
3
|
Wang W, Wang Y, Huang X, Wu P, Li L, Zhang Y, Chen Y, Chen Z, Li C, Zhou Y, Zhang J. Pathophysiology-Directed Engineering of a Combination Nanoanalgesic for Neuropathic Pain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2405483. [PMID: 39716944 DOI: 10.1002/advs.202405483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 12/08/2024] [Indexed: 12/25/2024]
Abstract
Neuropathic pain, one of the most refractory pain diseases, remains a formidable medical challenge. There is still an unmet demand for effective and safe therapies to address this condition. Herein, a rat model of nerve injury-induced neuropathic pain is first established to explore its pathophysiological characteristics. Recognizing the role of neuroinflammation, an inflammation-resolving amphiphilic conjugate PPT is designed and synthesized by simultaneously conjugating polyethylene glycol, phenylboronic acid pinacol ester, and Tempol onto a cyclic scaffold. PPT can self-assemble into nanomicelles (termed PPTN). Following intravenous injection, PPTN preferentially accumulates in the injured nerve, ameliorates the neuroinflammatory milieu, and promotes nerve regeneration, thereby shortening neuropathic pain duration in rats. Moreover, the Ca2+ channel α2δ1 subunit is identified as a therapeutic target by RNA-sequencing analysis of the injured nerve. Based on this target, a mimicking peptide (AD peptide) is screened as an analgesic. By packaging AD peptide into PPTN, a combination nano-analgesic APTN is developed. Besides potentiated anti-hyperalgesic effects due to site-specific delivery and on-demand release of AD peptide at target sites, APTN simultaneously inhibits neuroinflammation and promotes nerve regeneration by reprogramming macrophages via regulating MAPK/NF-kB signaling pathways and NLRP3 inflammasome activation, thus affording synergistic efficacies in treating nerve injury-induced neuropathic pain.
Collapse
Affiliation(s)
- Wenkai Wang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, P. R. China
- Department of Orthopedics, General Hospital of PLA Xizang Military Area Command, Lhasa, 850007, P. R. China
| | - Yan Wang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
- War Trauma Medical Center, State key Laboratory of Trauma, Burns and Combined injury, Army Medical Center, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Xinle Huang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, P. R. China
- Department of Orthopedics, The Second Naval Hospital of Southern Theater Command, Sanya, 572000, P. R. China
| | - Peng Wu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
- School of Pharmacy, Hanzhong Vocational and Technical College, Hanzhong, 723002, P. R. China
| | - Lanlan Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Yang Zhang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, P. R. China
| | - Yihui Chen
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, P. R. China
| | - Zhiyu Chen
- Department of Orthopedics, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, P. R. China
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, P. R. China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
- Yu-Yue Pathology Scientific Research Center, 313 Gaoteng Avenue, Jiulongpo District, Chongqing, 400039, P. R. China
| |
Collapse
|
4
|
Kim M, Shin M, Zhao Y, Ghosh M, Son Y. Transformative Impact of Nanocarrier‐Mediated Drug Delivery: Overcoming Biological Barriers and Expanding Therapeutic Horizons. SMALL SCIENCE 2024; 4. [DOI: 10.1002/smsc.202400280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Advancing therapeutic progress is centered on developing drug delivery systems (DDS) that control therapeutic molecule release, ensuring precise targeting and optimal concentrations. Targeted DDS enhances treatment efficacy and minimizes off‐target effects, but struggles with drug degradation. Over the last three decades, nanopharmaceuticals have evolved from laboratory concepts into clinical products, highlighting the profound impact of nanotechnology in medicine. Despite advancements, the effective delivery of therapeutics remains challenging because of biological barriers. Nanocarriers offer a solution with a small size, high surface‐to‐volume ratios, and customizable properties. These systems address physiological and biological challenges, such as shear stress, protein adsorption, and quick clearance. They allow targeted delivery to specific tissues, improve treatment outcomes, and reduce adverse effects. Nanocarriers exhibit controlled release, decreased degradation, and enhanced efficacy. Their size facilitates cell membrane penetration and intracellular delivery. Surface modifications increase affinity for specific cell types, allowing precise treatment delivery. This study also elucidates the potential integration of artificial intelligence with nanoscience to innovate future nanocarrier systems.
Collapse
Affiliation(s)
- Minhye Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science Jeju National University Jeju‐si Jeju Special Self‐Governing Province 63243 Republic of Korea
| | - Myeongyeon Shin
- Department of Animal Biotechnology Faculty of Biotechnology College of Applied Life Sciences Jeju National University Jeju‐si Jeju Special Self‐Governing Province 63243 Republic of Korea
| | - Yaping Zhao
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Mrinmoy Ghosh
- Department of Animal Biotechnology Faculty of Biotechnology College of Applied Life Sciences Jeju National University Jeju‐si Jeju Special Self‐Governing Province 63243 Republic of Korea
| | - Young‐Ok Son
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science Jeju National University Jeju‐si Jeju Special Self‐Governing Province 63243 Republic of Korea
- Department of Animal Biotechnology Faculty of Biotechnology College of Applied Life Sciences Jeju National University Jeju‐si Jeju Special Self‐Governing Province 63243 Republic of Korea
- Bio‐Health Materials Core‐Facility Center Jeju National University Jeju‐si 63243 Republic of Korea
- Practical Translational Research Center Jeju National University Jeju‐si 63243 Republic of Korea
| |
Collapse
|
5
|
Calvert ND, Baxter J, Torrens AA, Thompson J, Kirby A, Walia J, Ntais S, Hemmer E, Berini P, Hibbert B, Ramunno L, Shuhendler AJ. NIR-II scattering gold superclusters for intravascular optical coherence tomography molecular imaging. NATURE NANOTECHNOLOGY 2024:10.1038/s41565-024-01802-2. [PMID: 39468361 DOI: 10.1038/s41565-024-01802-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/12/2024] [Indexed: 10/30/2024]
Abstract
Currently, intravascular optical coherence tomography (IV-OCT) is limited to anatomical imaging, providing structural information about atherosclerotic plaque morphology, thrombus and dissection. Earlier detection and risk stratification would be possible through molecular characterization of endothelium but necessitates a purpose-engineered IV-OCT contrast agent. Here we developed gold superclusters (AuSCs) tailored to clinical instrumentation and integrated into clinically relevant workflows. AuSCs are aqueously dispersible clusters of closely packed small gold nanoparticles, affording plasmon hybridization to maximize light scattering at the IV-OCT laser line (~1,350 nm). A polymer coating fosters AuSC uniformity and provides a functionalizable handle, which we targeted to intravascular P-selectin, an early vascular endothelial marker of inflammation. In a rat model of intravascular inflammation, P-selectin-targeted AuSC facilitated IV-OCT molecular imaging, where the strength of the signal correlates with the severity of vascular inflammation.
Collapse
Affiliation(s)
- Nicholas D Calvert
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Joshua Baxter
- Department of Physics, University of Ottawa, Ottawa, Ontario, Canada
| | - Aidan A Torrens
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Jesse Thompson
- Department of Physics, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexia Kirby
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jaspreet Walia
- School of Electrical Engineering and Computer Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Spyridon Ntais
- School of Electrical Engineering and Computer Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Eva Hemmer
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Pierre Berini
- Department of Physics, University of Ottawa, Ottawa, Ontario, Canada
- School of Electrical Engineering and Computer Science, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Lora Ramunno
- Department of Physics, University of Ottawa, Ottawa, Ontario, Canada
| | - Adam J Shuhendler
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada.
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada.
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
6
|
Arenhoevel J, Kuppe A, Addante A, Wei LF, Boback N, Butnarasu C, Zhong Y, Wong C, Graeber SY, Duerr J, Gradzielski M, Lauster D, Mall MA, Haag R. Thiolated polyglycerol sulfate as potential mucolytic for muco-obstructive lung diseases. Biomater Sci 2024; 12:4376-4385. [PMID: 39028033 DOI: 10.1039/d4bm00381k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Increased disulfide crosslinking of secreted mucins causes elevated viscoelasticity of mucus and is a key determinant of mucus dysfunction in patients with cystic fibrosis (CF) and other muco-obstructive lung diseases. In this study, we describe the synthesis of a novel thiol-containing, sulfated dendritic polyglycerol (dPGS-SH), designed to chemically reduce these abnormal crosslinks, which we demonstrate with mucolytic activity assays in sputum from patients with CF. This mucolytic polymer, which is based on a reportedly anti-inflammatory polysulfate scaffold, additionally carries multiple thiol groups for mucolytic activity and can be produced on a gram-scale. After a physicochemical compound characterization, we compare the mucolytic activity of dPGS-SH to the clinically approved N-acetylcysteine (NAC) using western blot studies and investigate the effect of dPGS-SH on the viscoelastic properties of sputum samples from CF patients by oscillatory rheology. We show that dPGS-SH is more effective than NAC in reducing multimer intensity of the secreted mucins MUC5B and MUC5AC and demonstrate significant mucolytic activity by rheology. In addition, we provide data for dPGS-SH demonstrating a high compound stability, low cytotoxicity, and superior reaction kinetics over NAC at different pH levels. Our data support further development of the novel reducing polymer system dPGS-SH as a potential mucolytic to improve mucus function and clearance in patients with CF as well as other muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Justin Arenhoevel
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, SupraFAB, Altensteinstr. 23a, 14195 Berlin, Germany.
| | - Aditi Kuppe
- Charité - Universitätsmedizin Berlin, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Augustenburger Platz 1, 13353 Berlin, Germany.
- German Center for Lung Research (DZL), Associated Partner Site, 13353 Berlin, Germany
| | - Annalisa Addante
- Charité - Universitätsmedizin Berlin, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Augustenburger Platz 1, 13353 Berlin, Germany.
- German Center for Lung Research (DZL), Associated Partner Site, 13353 Berlin, Germany
| | - Ling-Fang Wei
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, SupraFAB, Altensteinstr. 23a, 14195 Berlin, Germany.
- Freie Universität Berlin, Institute of Pharmacy, Biopharmaceuticals, Kelchstraße 31, 12169 Berlin, Germany
| | - Nico Boback
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, SupraFAB, Altensteinstr. 23a, 14195 Berlin, Germany.
- Freie Universität Berlin, Institute of Pharmacy, Biopharmaceuticals, Kelchstraße 31, 12169 Berlin, Germany
| | - Cosmin Butnarasu
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, SupraFAB, Altensteinstr. 23a, 14195 Berlin, Germany.
- Freie Universität Berlin, Institute of Pharmacy, Biopharmaceuticals, Kelchstraße 31, 12169 Berlin, Germany
| | - Yinan Zhong
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, SupraFAB, Altensteinstr. 23a, 14195 Berlin, Germany.
| | - Christine Wong
- Charité - Universitätsmedizin Berlin, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Augustenburger Platz 1, 13353 Berlin, Germany.
- German Center for Lung Research (DZL), Associated Partner Site, 13353 Berlin, Germany
| | - Simon Y Graeber
- Charité - Universitätsmedizin Berlin, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Augustenburger Platz 1, 13353 Berlin, Germany.
- German Center for Lung Research (DZL), Associated Partner Site, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Julia Duerr
- Charité - Universitätsmedizin Berlin, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Augustenburger Platz 1, 13353 Berlin, Germany.
- German Center for Lung Research (DZL), Associated Partner Site, 13353 Berlin, Germany
| | - Michael Gradzielski
- Technische Universität Berlin, Institute of Chemistry, Straße des 17. Juni 124, 10623 Berlin, Germany
| | - Daniel Lauster
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, SupraFAB, Altensteinstr. 23a, 14195 Berlin, Germany.
- Freie Universität Berlin, Institute of Pharmacy, Biopharmaceuticals, Kelchstraße 31, 12169 Berlin, Germany
| | - Marcus A Mall
- Charité - Universitätsmedizin Berlin, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Augustenburger Platz 1, 13353 Berlin, Germany.
- German Center for Lung Research (DZL), Associated Partner Site, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Rainer Haag
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, SupraFAB, Altensteinstr. 23a, 14195 Berlin, Germany.
| |
Collapse
|
7
|
Chen Y, Luo X, Kang R, Cui K, Ou J, Zhang X, Liang P. Current therapies for osteoarthritis and prospects of CRISPR-based genome, epigenome, and RNA editing in osteoarthritis treatment. J Genet Genomics 2024; 51:159-183. [PMID: 37516348 DOI: 10.1016/j.jgg.2023.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/31/2023]
Abstract
Osteoarthritis (OA) is one of the most common degenerative joint diseases worldwide, causing pain, disability, and decreased quality of life. The balance between regeneration and inflammation-induced degradation results in multiple etiologies and complex pathogenesis of OA. Currently, there is a lack of effective therapeutic strategies for OA treatment. With the development of CRISPR-based genome, epigenome, and RNA editing tools, OA treatment has been improved by targeting genetic risk factors, activating chondrogenic elements, and modulating inflammatory regulators. Supported by cell therapy and in vivo delivery vectors, genome, epigenome, and RNA editing tools may provide a promising approach for personalized OA therapy. This review summarizes CRISPR-based genome, epigenome, and RNA editing tools that can be applied to the treatment of OA and provides insights into the development of CRISPR-based therapeutics for OA treatment. Moreover, in-depth evaluations of the efficacy and safety of these tools in human OA treatment are needed.
Collapse
Affiliation(s)
- Yuxi Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Xiao Luo
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Rui Kang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Kaixin Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Jianping Ou
- Center for Reproductive Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Xiya Zhang
- Center for Reproductive Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, Guangdong 510630, China.
| | - Puping Liang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China.
| |
Collapse
|
8
|
Koide H, Yamaguchi K, Sato K, Aoshima M, Kanata S, Yonezawa S, Asai T. Engineering Temperature-Responsive Polymer Nanoparticles that Load and Release Paclitaxel, a Low-Molecular-Weight Anticancer Drug. ACS OMEGA 2024; 9:1011-1019. [PMID: 38222561 PMCID: PMC10785788 DOI: 10.1021/acsomega.3c07226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 01/16/2024]
Abstract
Poly(N-isopropylacrylamide) (pNIPAm) undergoes a hydrophilicity/hydrophobicity change around its lower critical solution temperature (LCST). Therefore, pNIPAm-based polymer nanoparticles (NPs) shrink above their LCST and swell below their LCST. Although temperature responsiveness is an important characteristic of synthetic polymers in drug and gene delivery, few studies have investigated the temperature-responsive catch and release of low-molecular-weight drugs (LMWDs) as their affinity to the target changes. Since LMWDs have only a few functional groups, preparation of NPs with high affinity for LMWDs is hard compared with that for peptides and proteins. However, LMWDs such as anticancer drugs often have a stronger effect than peptides and proteins. Therefore, the development of NPs that can load and release LMWDs is needed for drug delivery. Here, we engineered pNIPAm-based NPs that capture paclitaxel (PTX), an anticancer LMWD that inhibits microtubules, above their LCST and release it below their LCST. The swelling transition of the NPs depended on their hydrophobic monomer structure. NPs with swelling ratios (=NP size at 25 °C/NP size at 37 °C) exceeding 1.90 released captured PTX when cooled to below their LCST by changing the affinity for PTX. On the other hand, NPs with a swelling ratio of only 1.14 released melittin. Therefore, optimizing the functional monomers of temperature-responsive NPs is essential for the catch and release of the target in a temperature-dependent manner. These results can guide the design of stimuli-responsive polymers that catch and release their target molecules.
Collapse
Affiliation(s)
- Hiroyuki Koide
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Kazuma Yamaguchi
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Keijiro Sato
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Maki Aoshima
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Shoko Kanata
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Sei Yonezawa
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| |
Collapse
|
9
|
Feng J, Nie C, Xie E, Thongrom B, Reiter-Scherer V, Block S, Herrmann A, Quaas E, Sieben C, Haag R. Sulfated Polyglycerol-Modified Hydrogels for Binding HSV-1 and RSV. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37903283 DOI: 10.1021/acsami.3c09553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Heparan sulfate (HS) is a highly sulfated polysaccharide on the surface of mammalian cells and in the extracellular matrix and has been found to be important for virus binding and infection. In this work, we designed synthetic hydrogels with viral binding and deactivation activities through the postfunctionalization of an HS-mimicking polyelectrolyte and alkyl chains. Three polyglycerol-based hydrogels were prepared as substrates and postfunctionalized by sulfated linear polyglycerol (lPGS) via thiol-ene click reaction. The viral binding properties were studied using herpes simplex virus type 1 (HSV-1) and respiratory syncytial virus (RSV). The effect of hydrogel types and molecular weight (Mw) of conjugated lPGS on viral binding properties was also assessed, and promising binding activities were observed in all lPGS-functionalized samples. Further coupling of 11 carbons long alkyl chains to the hydrogel revealed virucidal properties caused by destruction of the viral envelope, as shown by atomic force microscopy (AFM) imaging.
Collapse
Affiliation(s)
- Jun Feng
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Chuanxiong Nie
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Enyu Xie
- Nanoscale Infection Biology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
| | - Boonya Thongrom
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Valentin Reiter-Scherer
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Stephan Block
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Andreas Herrmann
- Institute of Chemistry and Biochemistry, SupraFAB, Freie Universität Berlin, Altensteinstr. 23a,14195 Berlin, Germany
| | - Elisa Quaas
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Christian Sieben
- Nanoscale Infection Biology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
- Institute of Genetics, Technische Universität Braunschweig, Spielmannstr. 7, Braunschweig 38106, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| |
Collapse
|
10
|
Pejchinovski I, Turkkan S, Pejchinovski M. Recent Advances of Proteomics in Management of Acute Kidney Injury. Diagnostics (Basel) 2023; 13:2648. [PMID: 37627907 PMCID: PMC10453063 DOI: 10.3390/diagnostics13162648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Acute Kidney Injury (AKI) is currently recognized as a life-threatening disease, leading to an exponential increase in morbidity and mortality worldwide. At present, AKI is characterized by a significant increase in serum creatinine (SCr) levels, typically followed by a sudden drop in glomerulus filtration rate (GFR). Changes in urine output are usually associated with the renal inability to excrete urea and other nitrogenous waste products, causing extracellular volume and electrolyte imbalances. Several molecular mechanisms were proposed to be affiliated with AKI development and progression, ultimately involving renal epithelium tubular cell-cycle arrest, inflammation, mitochondrial dysfunction, the inability to recover and regenerate proximal tubules, and impaired endothelial function. Diagnosis and prognosis using state-of-the-art clinical markers are often late and provide poor outcomes at disease onset. Inappropriate clinical assessment is a strong disease contributor, actively driving progression towards end stage renal disease (ESRD). Proteins, as the main functional and structural unit of the cell, provide the opportunity to monitor the disease on a molecular level. Changes in the proteomic profiles are pivotal for the expression of molecular pathways and disease pathogenesis. Introduction of highly-sensitive and innovative technology enabled the discovery of novel biomarkers for improved risk stratification, better and more cost-effective medical care for the ill patients and advanced personalized medicine. In line with those strategies, this review provides and discusses the latest findings of proteomic-based biomarkers and their prospective clinical application for AKI management.
Collapse
Affiliation(s)
- Ilinka Pejchinovski
- Department of Quality Assurance, Nikkiso Europe GmbH, 30885 Langenhagen, Germany; (I.P.); (S.T.)
| | - Sibel Turkkan
- Department of Quality Assurance, Nikkiso Europe GmbH, 30885 Langenhagen, Germany; (I.P.); (S.T.)
| | - Martin Pejchinovski
- Department of Analytical Instruments Group, Thermo Fisher Scientific, 82110 Germering, Germany
| |
Collapse
|
11
|
Fitzgerald DM, Zhang H, Bordeianu C, Colson YL, Grinstaff MW. Synthesis of Polyethylene Glycol-Poly(glycerol carbonate) Block Copolymeric Micelles as Surfactant-Free Drug Delivery Systems. ACS Macro Lett 2023; 12:974-979. [PMID: 37390500 PMCID: PMC11331582 DOI: 10.1021/acsmacrolett.3c00275] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2023]
Abstract
We report the synthesis of block copolymers of monomethoxylated polyethylene glycol and poly(glycerol carbonate) (mPEG-b-PGC) via the ring-opening polymerization of benzyl glycidyl ether, monomethoxylated polyethylene glycol, and carbon dioxide using a cobalt salen catalyst. The resulting block copolymers display high polymer/cyclic carbonate selectivity (>99%) and, if two oxirane monomers are used, random incorporation into the polymer feed. The resulting diblock mPEG-b-PGC polymer shows promise as a nanocarrier for surfactant-free, sustained chemotherapeutic delivery. mPEG-b-PGC, with paclitaxel conjugated to the pendant primary alcohol of the glycerol polymer backbone, readily forms 175 nm diameter particles in solution and contains 4.6 wt % paclitaxel (PTX), which is released over 42 days. The mPEG-b-PGC polymer itself is noncytotoxic, whereas the PTX-loaded nanoparticles are cytotoxic to lung, breast, and ovarian cancer cell lines.
Collapse
Affiliation(s)
- Danielle M. Fitzgerald
- Departments of Chemistry and Biomedical Engineering Boston University, 590 Commonwealth Ave, Boston, MA, 02115
| | - Heng Zhang
- Departments of Chemistry and Biomedical Engineering Boston University, 590 Commonwealth Ave, Boston, MA, 02115
| | - Catalina Bordeianu
- Departments of Chemistry and Biomedical Engineering Boston University, 590 Commonwealth Ave, Boston, MA, 02115
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA 02214
| | - Mark W. Grinstaff
- Departments of Chemistry and Biomedical Engineering Boston University, 590 Commonwealth Ave, Boston, MA, 02115
| |
Collapse
|
12
|
Hu X, Wang T, Li F, Mao X. Surface modifications of biomaterials in different applied fields. RSC Adv 2023; 13:20495-20511. [PMID: 37435384 PMCID: PMC10331796 DOI: 10.1039/d3ra02248j] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/26/2023] [Indexed: 07/13/2023] Open
Abstract
Biomaterial implantation into the human body plays a key role in the medical field and biological applications. Increasing the life expectancy of biomaterial implants, reducing the rejection reaction inside the human body and reducing the risk of infection are the problems in this field that need to be solved urgently. The surface modification of biomaterials can change the original physical, chemical and biological properties and improve the function of materials. This review focuses on the application of surface modification techniques in various fields of biomaterials reported in the past few years. The surface modification techniques include film and coating synthesis, covalent grafting, self-assembled monolayers (SAMs), plasma surface modification and other strategies. First, a brief introduction to these surface modification techniques for biomaterials is given. Subsequently, the review focuses on how these techniques change the properties of biomaterials, and evaluates the effects of modification on the cytocompatibility, antibacterial, antifouling and surface hydrophobic properties of biomaterials. In addition, the implications for the design of biomaterials with different functions are discussed. Finally, based on this review, it is expected that the biomaterials have development prospects in the medical field.
Collapse
Affiliation(s)
- Xi Hu
- State Key Laboratory of Ultrasound in Medicine and Engineering College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
| | - Teng Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
| | - Faqi Li
- State Key Laboratory of Ultrasound in Medicine and Engineering College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
| | - Xiang Mao
- State Key Laboratory of Ultrasound in Medicine and Engineering College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
| |
Collapse
|
13
|
Koeppe H, Horn D, Scholz J, Quaas E, Schötz S, Reisbeck F, Achazi K, Mohammadifar E, Dernedde J, Haag R. Shell-Sheddable Dendritic Polyglycerol Sulfates Loaded with Sunitinib for Inhibition of Tumor Angiogenesis. Int J Pharm 2023:123158. [PMID: 37336299 DOI: 10.1016/j.ijpharm.2023.123158] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/05/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Induced angiogenesis, a specific hallmark of cancer, plays a vital role in tumor progression and can be targeted by inhibitors like sunitinib. Sunitinib is a small hydrophobic molecule suffering from low bioavailability and a short half-life in the bloodstream. To overcome these drawbacks, suitable drug delivery systems need to be developed. In this work dendritic polyglycerol (dPG), a well-known polymer, was functionalized with a sheddable shell. Therefore, aliphatic chains of different lengths (C5, C9, C11) were coupled to dPG through a cleavable ester bond. To restore water solubility and improve tumor targeting, the surface was decorated with sulfate groups. The resulting shell-sheddable dPG sulfates were characterized and evaluated regarding their loading capacity and biocompatibility in cell culture. The nine-carbon chain derivative (dPG-TNS) was selected as the best candidate for further experiments due to its high drug loading capacity (20wt%), and a sustained release in vitro. The cellular biocompatibility of the blank carrier up to 1mg/mL was confirmed after 24h incubation on HeLa cells. Furthermore, the shell-cleavability of dPG-TNS under different physiological conditions was shown in a degradation study over four weeks. The activity of sunitinib-loaded dPG-TNS was demonstrated in a tube formation assay on Human umbilical vein endothelial cells (HUVECs). Our results suggest that the drug-loaded nanocarrier is a promising candidate to be further investigated in tumor treatments, as it shows similar efficacy to free sunitinib while overcoming its limitations.
Collapse
Affiliation(s)
- Hanna Koeppe
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Daniel Horn
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Johanna Scholz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Elisa Quaas
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Sebastian Schötz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Felix Reisbeck
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Katharina Achazi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Ehsan Mohammadifar
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.
| | - Jens Dernedde
- Institute for Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.
| |
Collapse
|
14
|
Parshad B, Schlecht MN, Baumgardt M, Ludwig K, Nie C, Rimondi A, Hönzke K, Angioletti-Uberti S, Khatri V, Schneider P, Herrmann A, Haag R, Hocke AC, Wolff T, Bhatia S. Dual-Action Heteromultivalent Glycopolymers Stringently Block and Arrest Influenza A Virus Infection In Vitro and Ex Vivo. NANO LETTERS 2023; 23:4844-4853. [PMID: 37220024 DOI: 10.1021/acs.nanolett.3c00408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Here, we demonstrate the concerted inhibition of different influenza A virus (IAV) strains using a low-molecular-weight dual-action linear polymer. The 6'-sialyllactose and zanamivir conjugates of linear polyglycerol are optimized for simultaneous targeting of hemagglutinin and neuraminidase on the IAV surface. Independent of IAV subtypes, hemagglutination inhibition data suggest better adsorption of the heteromultivalent polymer than homomultivalent analogs onto the virus surface. Cryo-TEM images imply heteromultivalent compound-mediated virus aggregation. The optimized polymeric nanomaterial inhibits >99.9% propagation of various IAV strains 24 h postinfection in vitro at low nM concentrations and is up to 10000× more effective than the commercial zanamivir drug. In a human lung ex vivo multicyclic infection setup, the heteromultivalent polymer outperforms the commercial drug zanamivir and homomultivalent analogs or their physical mixtures. This study authenticates the translational potential of the dual-action targeting approach using small polymers for broad and high antiviral efficacy.
Collapse
Affiliation(s)
- Badri Parshad
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Marlena N Schlecht
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestraße 10, 13353 Berlin, Germany
- Medical Clinic III, Division of Nephrology, Medizinische Fakultät Carl Gustav Carus an der TU Dresden, Fiedlerstr. 40, 01307 Dresden, Germany
| | - Morris Baumgardt
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Kai Ludwig
- Forschungszentrum für Elektronenmikroskopie and Core Facility BioSupraMol, Institut für Chemie und Biochemie, Freie Universität Berlin, Fabeckstr. 36a, 14195 Berlin, Germany
| | - Chuanxiong Nie
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Agustina Rimondi
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestraße 10, 13353 Berlin, Germany
| | - Katja Hönzke
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | | | - Vinod Khatri
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Paul Schneider
- Department for Thoracic Surgery, DRK Clinics, 13359 Berlin, Germany
| | - Andreas Herrmann
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Rainer Haag
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Andreas C Hocke
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Thorsten Wolff
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestraße 10, 13353 Berlin, Germany
| | - Sumati Bhatia
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| |
Collapse
|
15
|
Witt M, Cherri M, Ferraro M, Yapto C, Vogel K, Schmidt L, Haag R, Danker K, Dommisch H. Anti-inflammatory IL-8 Regulation via an Advanced Drug Delivery System at the Oral Mucosa. ACS APPLIED BIO MATERIALS 2023. [PMID: 37216981 DOI: 10.1021/acsabm.3c00024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Oral inflammatory diseases are highly prevalent in the worldwide population. Topical treatment of inflammation is challenging due to dilution effects of saliva and crevicular fluid. Thus, there is a great medical need to develop smart anti-inflammatory drug delivery systems for mucosa treatment. We compared two promising anti-inflammatory dendritic poly(glycerol-caprolactone) sulfate (dPGS-PCL) polymers for their applicability to the oral mucosa. Using an ex vivo porcine tissue model, cell monolayers, and full-thickness 3D oral mucosal organoids, the polymers were evaluated for muco-adhesion, penetration, and anti-inflammatory properties. The biodegradable dPGS-PCL97 polymers adhered to and penetrated the masticatory mucosa within seconds. No effects on metabolic activity and cell proliferation were found. dPGS-PCL97 revealed a significant downregulation of pro-inflammatory cytokines with a clear preference for IL-8 in cell monolayers and mucosal organoids. Thus, dPGS-PCL97 exhibits excellent properties for topical anti-inflammatory therapy, suggesting new therapeutic avenues in the treatment of oral inflammatory diseases.
Collapse
Affiliation(s)
- Maren Witt
- Department of Periodontology, Oral Medicine and Oral Surgery, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin 14197 , Germany
| | - Mariam Cherri
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Magda Ferraro
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Cynthia Yapto
- Institute of Biochemistry, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Katrin Vogel
- Department of Periodontology, Oral Medicine and Oral Surgery, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin 14197 , Germany
| | - Lena Schmidt
- Institute of Biochemistry, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Kerstin Danker
- Institute of Biochemistry, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Henrik Dommisch
- Department of Periodontology, Oral Medicine and Oral Surgery, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin 14197 , Germany
| |
Collapse
|
16
|
Moreira DA, Santos SD, Leiro V, Pêgo AP. Dendrimers and Derivatives as Multifunctional Nanotherapeutics for Alzheimer's Disease. Pharmaceutics 2023; 15:pharmaceutics15041054. [PMID: 37111540 PMCID: PMC10140951 DOI: 10.3390/pharmaceutics15041054] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/15/2023] [Accepted: 03/18/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. It affects more than 30 million people worldwide and costs over US$ 1.3 trillion annually. AD is characterized by the brain accumulation of amyloid β peptide in fibrillar structures and the accumulation of hyperphosphorylated tau aggregates in neurons, both leading to toxicity and neuronal death. At present, there are only seven drugs approved for the treatment of AD, of which only two can slow down cognitive decline. Moreover, their use is only recommended for the early stages of AD, meaning that the major portion of AD patients still have no disease-modifying treatment options. Therefore, there is an urgent need to develop efficient therapies for AD. In this context, nanobiomaterials, and dendrimers in particular, offer the possibility of developing multifunctional and multitargeted therapies. Due to their intrinsic characteristics, dendrimers are first-in-class macromolecules for drug delivery. They have a globular, well-defined, and hyperbranched structure, controllable nanosize and multivalency, which allows them to act as efficient and versatile nanocarriers of different therapeutic molecules. In addition, different types of dendrimers display antioxidant, anti-inflammatory, anti-bacterial, anti-viral, anti-prion, and most importantly for the AD field, anti-amyloidogenic properties. Therefore, dendrimers can not only be excellent nanocarriers, but also be used as drugs per se. Here, the outstanding properties of dendrimers and derivatives that make them excellent AD nanotherapeutics are reviewed and critically discussed. The biological properties of several dendritic structures (dendrimers, derivatives, and dendrimer-like polymers) that enable them to be used as drugs for AD treatment will be pointed out and the chemical and structural characteristics behind those properties will be analysed. The reported use of these nanomaterials as nanocarriers in AD preclinical research is also presented. Finally, future perspectives and challenges that need to be overcome to make their use in the clinic a reality are discussed.
Collapse
Affiliation(s)
- Débora A Moreira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP-Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sofia D Santos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Victoria Leiro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Ana P Pêgo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
17
|
Koide H, Saito K, Yoshimatsu K, Chou B, Hoshino Y, Yonezawa S, Oku N, Asai T, Shea KJ. Cooling-induced, localized release of cytotoxic peptides from engineered polymer nanoparticles in living mice for cancer therapy. J Control Release 2023; 355:745-759. [PMID: 36804558 DOI: 10.1016/j.jconrel.2023.02.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/31/2023] [Accepted: 02/13/2023] [Indexed: 02/23/2023]
Abstract
Temperature-responsive polymers are often characterized by an abrupt change in the degree of swelling brought about by small changes in temperature. Polymers with a lower critical solution temperature (LCST) in particular, are important as drug and gene delivery vehicles. Drug molecules are taken up by the polymer in their solvent swollen state below their LCST. Increasing the temperature above the LCST, typically physiological temperatures, results in desolvation of polymer chains and microstructure collapse. The trapped drug is released slowly by passive diffusion through the collapsed polymer network. Since diffusion is dependent on many variables, localizing and control of the drug delivery rate can be challenging. Here, we report a fundamentally different approach for the rapid (seconds) tumor-specific delivery of a biomacromolecular drug. A copolymer nanoparticle (NP) was engineered with affinity for melittin, a peptide with potent anti-cancer activity, at physiological temperature. Intravenous injection of the NP-melittin complex results in its accumulation in organs and at the tumor. We demonstrate that by local cooling of the tumor the melittin is rapidly released from the NP-melittin complex. The release occurs only at the cooled tumor site. Importantly, tumor growth was significantly suppressed using this technique demonstrating therapeutically useful quantities of the drug can be delivered. This work reports the first example of an in vivo site-specific release of a macromolecular drug by local cooling for cancer therapy. In view of the increasing number of cryotherapeutic devices for in vivo applications, this work has the potential to stimulate cryotherapy for in vivo drug delivery.
Collapse
Affiliation(s)
- Hiroyuki Koide
- Department of Medical Biochemistry, Graduate school of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan.
| | - Kazuhiro Saito
- Department of Medical Biochemistry, Graduate school of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Keiichi Yoshimatsu
- Department of Chemistry, Missouri State University, 901 South National Avenue, Springfield, MO 65897, USA; Department of Chemistry, University of California Irvine, Irvine, CA 92697, USA.
| | - Beverly Chou
- Department of Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Yu Hoshino
- Department of Applied Chemistry, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Sei Yonezawa
- Department of Medical Biochemistry, Graduate school of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Naoto Oku
- Department of Medical Biochemistry, Graduate school of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan; Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, Graduate school of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Kenneth J Shea
- Department of Chemistry, University of California Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
18
|
Maysinger D, Zhang I, Wu PY, Kagelmacher M, Luo HD, Kizhakkedathu JN, Dernedde J, Ballauff M, Haag R, Shobo A, Multhaup G, McKinney RA. Sulfated Hyperbranched and Linear Polyglycerols Modulate HMGB1 and Morphological Plasticity in Neural Cells. ACS Chem Neurosci 2023; 14:677-688. [PMID: 36717083 DOI: 10.1021/acschemneuro.2c00558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The objective of this study was to establish if polyglycerols with sulfate or sialic acid functional groups interact with high mobility group box 1 (HMGB1), and if so, which polyglycerol could prevent loss of morphological plasticity in excitatory neurons in the hippocampus. Considering that HMGB1 binds to heparan sulfate and that heparan sulfate has structural similarities with dendritic polyglycerol sulfates (dPGS), we performed the experiments to show if polyglycerols can mimic heparin functions by addressing the following questions: (1) do dendritic and linear polyglycerols interact with the alarmin molecule HMGB1? (2) Does dPGS interaction with HMGB1 influence the redox status of HMGB1? (3) Can dPGS prevent the loss of dendritic spines in organotypic cultures challenged with lipopolysaccharide (LPS)? LPS plays a critical role in infections with Gram-negative bacteria and is commonly used to test candidate therapeutic agents for inflammation and endotoxemia. Pathologically high LPS concentrations and other stressful stimuli cause HMGB1 release and post-translational modifications. We hypothesized that (i) electrostatic interactions of hyperbranched and linear polysulfated polyglycerols with HMGB1 will likely involve sites similar to those of heparan sulfate. (ii) dPGS can normalize HMGB1 compartmentalization in microglia exposed to LPS and prevent dendritic spine loss in the excitatory hippocampal neurons. We performed immunocytochemistry and biochemical analyses combined with confocal microscopy to determine cellular and extracellular locations of HMGB1 and morphological plasticity. Our results suggest that dPGS interacts with HMGB1 similarly to heparan sulfate. Hyperbranched dPGS and linear sulfated polymers prevent dendritic spine loss in hippocampal excitatory neurons. MS/MS analyses reveal that dPGS-HMGB1 interactions result in fully oxidized HMGB1 at critical cysteine residues (Cys23, Cys45, and Cys106). Triply oxidized HMGB1 leads to the loss of its pro-inflammatory action and could participate in dPGS-mediated spine loss prevention. LPG-Sia exposure to HMGB1 results in the oxidation of Cys23 and Cys106 but does not normalize spine density.
Collapse
Affiliation(s)
- Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| | - Issan Zhang
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| | - Pei You Wu
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| | - Marten Kagelmacher
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin14195, Germany
| | - Haiming Daniel Luo
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, Life Science Institute, Department of Chemistry, School of Biomedical Engineering, University of British Columbia, VancouverV6T 1Z3, Canada
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, Life Science Institute, Department of Chemistry, School of Biomedical Engineering, University of British Columbia, VancouverV6T 1Z3, Canada
| | - Jens Dernedde
- Institute of Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin13353, Germany
| | - Matthias Ballauff
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin14195, Germany
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin14195, Germany
| | - Adeola Shobo
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| | - Gerhard Multhaup
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| | - R Anne McKinney
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| |
Collapse
|
19
|
Gaitsch H, Hersh AM, Alomari S, Tyler BM. Dendrimer Technology in Glioma: Functional Design and Potential Applications. Cancers (Basel) 2023; 15:1075. [PMID: 36831418 PMCID: PMC9954563 DOI: 10.3390/cancers15041075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/02/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Novel therapeutic and diagnostic methods are sorely needed for gliomas, which contribute yearly to hundreds of thousands of cancer deaths worldwide. Despite the outpouring of research efforts and funding aimed at improving clinical outcomes for patients with glioma, the prognosis for high-grade glioma, and especially glioblastoma, remains dire. One of the greatest obstacles to improving treatment efficacy and destroying cancer cells is the safe delivery of chemotherapeutic drugs and biologics to the tumor site at a high enough dose to be effective. Over the past few decades, a burst of research has leveraged nanotechnology to overcome this obstacle. There has been a renewed interest in adapting previously understudied dendrimer nanocarriers for this task. Dendrimers are small, highly modifiable, branched structures featuring binding sites for a variety of drugs and ligands. Recent studies have demonstrated the potential for dendrimers and dendrimer conjugates to effectively shuttle therapeutic cargo to the correct tumor location, permeate the tumor, and promote apoptosis of tumor cells while minimizing systemic toxicity and damage to surrounding healthy brain tissue. This review provides a primer on the properties of dendrimers; outlines the mechanisms by which they can target delivery of substances to the site of brain pathology; and delves into current trends in the application of dendrimers to drug and gene delivery, and diagnostic imaging, in glioma. Finally, future directions for translating these in vitro and in vivo findings to the clinic are discussed.
Collapse
Affiliation(s)
- Hallie Gaitsch
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- NIH Oxford-Cambridge Scholars Program, Wellcome—MRC Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Andrew M. Hersh
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Safwan Alomari
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Betty M. Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
20
|
Hoffmann M, Snyder NL, Hartmann L. Polymers Inspired by Heparin and Heparan Sulfate for Viral Targeting. Macromolecules 2022; 55:7957-7973. [PMID: 36186574 PMCID: PMC9520969 DOI: 10.1021/acs.macromol.2c00675] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/12/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Miriam Hoffmann
- Department of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Nicole L. Snyder
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Laura Hartmann
- Department of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
21
|
Caminade AM, Turrin CO, Poupot R. Curing inflammatory diseases using phosphorous dendrimers. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1783. [PMID: 35194953 DOI: 10.1002/wnan.1783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Different types of water-soluble phosphorous dendrimers have been synthesized and display many different biological properties. It has been shown in particular that phosphorous dendrimers of first generation functionalized with azabisphosphonate terminal functions are able to stimulate the human immune system ex vivo. These dendrimers are internalized by monocytes within a few seconds, and induce their anti-inflammatory activation. The presence of the dendrimers induces also the inhibition of the differentiation of monocytes into osteoclasts, the maturation of dendritic cells, and inhibits the proliferation of the proinflammatory CD4+ T lymphocytes. Finally, after 2-3 weeks of culture of peripheral blood mononuclear cells, amplifications by several tens of natural killer cells is observed. In view of all these properties, the influence of these azabisphosphonate-dendrimers has been tested in vivo with several animal models, against different chronic or acute inflammatory diseases, such as multiple sclerosis, rheumatoid arthritis, uveitis, and psoriasis, but also against myeloid leukemia, a hematological cancer. The hematological safety has been demonstrated in mice, as there is no platelet aggregation, no hemolysis, and no disturbance in the hematological formula. The safety of the azabisphosphonate-dendrimer has been assessed also with non-human primates (cynomolgus monkeys) which received repeated injections, as a de-risking pre-clinical test. Biochemical, hematological, and all immunological parameters in peripheral blood remained within a normal physiological range throughout the study, and all survived well. Other phosphorous dendrimers also display anti-inflammatory properties in vivo, in particular dendrimers functionalized with mannose derivatives, which prevent acute lung diseases when given orally (per os) to mice. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Anne-Marie Caminade
- Laboratoire de Chimie de Coordination (LCC), CNRS UPR8241, Toulouse Cedex 4, France
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Cédric-Olivier Turrin
- Laboratoire de Chimie de Coordination (LCC), CNRS UPR8241, Toulouse Cedex 4, France
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
- IMD-Pharma, Toulouse Cedex 4, France
| | - Rémy Poupot
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, CHU Purpan, Toulouse Cedex 3, France
- Infinity, Université Toulouse, CNRS, INSERM, UPS, Toulouse, France
| |
Collapse
|
22
|
Pouyan P, Cherri M, Haag R. Polyglycerols as Multi-Functional Platforms: Synthesis and Biomedical Applications. Polymers (Basel) 2022; 14:polym14132684. [PMID: 35808728 PMCID: PMC9269438 DOI: 10.3390/polym14132684] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 02/07/2023] Open
Abstract
The remarkable and unique characteristics of polyglycerols (PG) have made them an attractive candidate for many applications in the biomedical and pharmaceutical fields. The presence of multiple hydroxy groups on the flexible polyether backbone not only enables the further modification of the PG structure but also makes the polymer highly water-soluble and results in excellent biocompatibility. In this review, the polymerization routes leading to PG with different architectures are discussed. Moreover, we discuss the role of these polymers in different biomedical applications such as drug delivery systems, protein conjugation, and surface modification.
Collapse
|
23
|
Milošević N, Rütter M, David A. Endothelial Cell Adhesion Molecules- (un)Attainable Targets for Nanomedicines. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:846065. [PMID: 35463298 PMCID: PMC9021548 DOI: 10.3389/fmedt.2022.846065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/15/2022] [Indexed: 01/21/2023] Open
Abstract
Endothelial cell adhesion molecules have long been proposed as promising targets in many pathologies. Despite promising preclinical data, several efforts to develop small molecule inhibitors or monoclonal antibodies (mAbs) against cell adhesion molecules (CAMs) ended in clinical-stage failure. In parallel, many well-validated approaches for targeting CAMs with nanomedicine (NM) were reported over the years. A wide range of potential applications has been demonstrated in various preclinical studies, from drug delivery to the tumor vasculature, imaging of the inflamed endothelium, or blocking immune cells infiltration. However, no NM drug candidate emerged further into clinical development. In this review, we will summarize the most advanced examples of CAM-targeted NMs and juxtapose them with known traditional drugs against CAMs, in an attempt to identify important translational hurdles. Most importantly, we will summarize the proposed strategies to enhance endothelial CAM targeting by NMs, in an attempt to offer a catalog of tools for further development.
Collapse
|
24
|
Tu Z, Zhong Y, Hu H, Shao D, Haag R, Schirner M, Lee J, Sullenger B, Leong KW. Design of therapeutic biomaterials to control inflammation. NATURE REVIEWS. MATERIALS 2022; 7:557-574. [PMID: 35251702 PMCID: PMC8884103 DOI: 10.1038/s41578-022-00426-z] [Citation(s) in RCA: 215] [Impact Index Per Article: 71.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 05/03/2023]
Abstract
Inflammation plays an important role in the response to danger signals arising from damage to our body and in restoring homeostasis. Dysregulated inflammatory responses occur in many diseases, including cancer, sepsis and autoimmunity. The efficacy of anti-inflammatory drugs, developed for the treatment of dysregulated inflammation, can be potentiated using biomaterials, by improving the bioavailability of drugs and by reducing side effects. In this Review, we first outline key elements and stages of the inflammatory environment and then discuss the design of biomaterials for different anti-inflammatory therapeutic strategies. Biomaterials can be engineered to scavenge danger signals, such as reactive oxygen and nitrogen species and cell-free DNA, in the early stages of inflammation. Materials can also be designed to prevent adhesive interactions of leukocytes and endothelial cells that initiate inflammatory responses. Furthermore, nanoscale platforms can deliver anti-inflammatory agents to inflammation sites. We conclude by discussing the challenges and opportunities for biomaterial innovations in addressing inflammation.
Collapse
Affiliation(s)
- Zhaoxu Tu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Department of Biomedical Engineering, Columbia University, New York, NY USA
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Yiling Zhong
- Department of Biomedical Engineering, Columbia University, New York, NY USA
- School of Chemistry, University of New South Wales, Sydney, New South Wales Australia
| | - Hanze Hu
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Dan Shao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin, Germany
| | - Michael Schirner
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin, Germany
| | - Jaewoo Lee
- School of Medicine, Duke University, Durham, NC USA
| | | | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY USA
- Department of Systems Biology, Columbia University, New York, NY USA
| |
Collapse
|
25
|
Critical parameters for design and development of multivalent nanoconstructs: recent trends. Drug Deliv Transl Res 2022; 12:2335-2358. [PMID: 35013982 PMCID: PMC8747862 DOI: 10.1007/s13346-021-01103-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2021] [Indexed: 12/16/2022]
Abstract
A century ago, the groundbreaking concept of the magic bullet was given by Paul Ehrlich. Since then, this concept has been extensively explored in various forms to date. The concept of multivalency is among such advancements of the magic bullet concept. Biologically, the concept of multivalency plays a critical role in significantly huge numbers of biochemical interactions. This concept is the sole reason behind the higher affinity of biological molecules like viruses to more selectively target the host cell surface receptors. Multivalent nanoconstructs are a promising approach for drug delivery by the active targeting principle. Designing and developing effective and target-specific multivalent drug delivery nanoconstructs, on the other hand, remain a challenge. The underlying reason for this is a lack of understanding of the crucial interactions between ligands and cell surface receptors, as well as the design of nanoconstructs. This review highlights the need for a better theoretical understanding of the multivalent effect of what happens to the receptor-ligand complex after it has been established. Furthermore, the critical parameters for designing and developing robust multivalent systems have been emphasized. We have also discussed current advances in the design and development of multivalent nanoconstructs for drug delivery. We believe that a thorough knowledge of theoretical concepts and experimental methodologies may transform a brilliant idea into clinical translation.
Collapse
|
26
|
Sun L, Liu Y, Liu X, Wang R, Gong J, Saferali A, Gao W, Ma A, Ma H, Turvey SE, Fung S, Yang H. Nano-Enabled Reposition of Proton Pump Inhibitors for TLR Inhibition: Toward A New Targeted Nanotherapy for Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104051. [PMID: 34816630 PMCID: PMC8787384 DOI: 10.1002/advs.202104051] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/25/2021] [Indexed: 05/30/2023]
Abstract
Toll-like receptor (TLR) activation in macrophages plays a critical role in the pathogenesis of acute lung injury (ALI). While TLR inhibition is a promising strategy to control the overwhelming inflammation in ALI, there still lacks effective TLR inhibitors for clinical uses to date. A unique class of peptide-coated gold nanoparticles (GNPs) is previously discovered, which effectively inhibited TLR signaling and protected mice from lipopolysaccharide (LPS)-induced ALI. To fast translate such a discovery into potential clinical applicable nanotherapeutics, herein an elegant strategy of "nano-enabled drug repurposing" with "nano-targeting" is introduced to empower the existing drugs for new uses. Combining transcriptome sequencing with Connectivity Map analysis, it is identified that the proton pump inhibitors (PPIs) share similar mechanisms of action to the discovered GNP-based TLR inhibitor. It is confirmed that PPIs (including omeprazole) do inhibit endosomal TLR signaling and inflammatory responses in macrophages and human peripheral blood mononuclear cells, and exhibits anti-inflammatory activity in an LPS-induced ALI mouse model. The omeprazole is then formulated into a nanoform with liposomes to enhance its macrophage targeting ability and the therapeutic efficacy in vivo. This research provides a new translational strategy of nano-enabled drug repurposing to translate bioactive nanoparticles into clinically used drugs and targeted nano-therapeutics for ALI.
Collapse
Affiliation(s)
- Liya Sun
- School of Biomedical EngineeringThe Province and Ministry Co‐Sponsored Collaborative Innovation Center for Medical EpigeneticsIntensive Care Unit of the Second HospitalTianjin Medical UniversityNo. 22 Qixiangtai Road, Heping DistrictTianjin300070China
| | - Yuan Liu
- School of Biomedical EngineeringThe Province and Ministry Co‐Sponsored Collaborative Innovation Center for Medical EpigeneticsIntensive Care Unit of the Second HospitalTianjin Medical UniversityNo. 22 Qixiangtai Road, Heping DistrictTianjin300070China
| | - Xiali Liu
- Department of Pulmonary and Critical Care MedicineShanghai General HospitalShanghai Jiao Tong University School of MedicineNo. 650 Xinsongjiang RoadShanghai201620China
| | - Rui Wang
- School of Biomedical EngineeringThe Province and Ministry Co‐Sponsored Collaborative Innovation Center for Medical EpigeneticsIntensive Care Unit of the Second HospitalTianjin Medical UniversityNo. 22 Qixiangtai Road, Heping DistrictTianjin300070China
| | - Jiameng Gong
- School of Biomedical EngineeringThe Province and Ministry Co‐Sponsored Collaborative Innovation Center for Medical EpigeneticsIntensive Care Unit of the Second HospitalTianjin Medical UniversityNo. 22 Qixiangtai Road, Heping DistrictTianjin300070China
| | - Aabida Saferali
- Channing Division of Network MedicineBrigham and Women's HospitalHarvard Medical School181 Longwood AvenueBostonMA02115USA
| | - Wei Gao
- Department of Pulmonary and Critical Care MedicineShanghai General HospitalShanghai Jiao Tong University School of MedicineNo. 650 Xinsongjiang RoadShanghai201620China
| | - Aying Ma
- Department of Pulmonary and Critical Care MedicineShanghai General HospitalShanghai Jiao Tong University School of MedicineNo. 650 Xinsongjiang RoadShanghai201620China
| | - Huiqiang Ma
- School of Biomedical EngineeringThe Province and Ministry Co‐Sponsored Collaborative Innovation Center for Medical EpigeneticsIntensive Care Unit of the Second HospitalTianjin Medical UniversityNo. 22 Qixiangtai Road, Heping DistrictTianjin300070China
| | - Stuart E. Turvey
- BC Children's Research InstituteUniversity of British Columbia950 West 28th AvenueVancouverBC V5Z 4H4Canada
| | - Shan‐Yu Fung
- Department of ImmunologyKey Laboratory of Immune Microenvironment and Disease (Ministry of Education)The Province and Ministry Co‐Sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical ScienceTianjin Medical UniversityNo. 22 Qixiangtai Road, Heping DistrictTianjin300070China
| | - Hong Yang
- School of Biomedical EngineeringThe Province and Ministry Co‐Sponsored Collaborative Innovation Center for Medical EpigeneticsIntensive Care Unit of the Second HospitalTianjin Medical UniversityNo. 22 Qixiangtai Road, Heping DistrictTianjin300070China
| |
Collapse
|
27
|
Xu X, Jia X, Zhang Y. Dendritic polyelectrolytes with monovalent and divalent counterions: the charge regulation effect and counterion release. SOFT MATTER 2021; 17:10862-10872. [PMID: 34806740 DOI: 10.1039/d1sm01392k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The charge regulation and the release of counterions are extremely important and substantial in determining the charge state of polyelectrolytes and the interaction between polyelectrolytes and proteins. Going beyond monovalent to multivalent cations, it is well-known that the effects of ions are qualitatively different. Therefore, the well-accepted descriptions of the charge regulation and the counterion release based on monovalent ions do not immediately apply to systems with multivalent ions. Here, we study the key structural and electrostatic features of charged dendrimers at hand of the pharmaceutically important dendritic polyglycerol sulfate (dPGS) macromolecule equilibrated with monovalent and divalent salts by molecular dynamics (MD) simulations. Following a simple but accurate scheme to determine its effective radius, the counterion condensed layer of the dPGS is determined with high accuracy and we observe the sequential replacement of condensed monovalent cations (MCs) to divalent cations (DCs) rendering a smaller dPGS effective charge versus the DC concentration. We resolve and track the release of counterions on the dPGS along its binding pathway with the plasma protein Human Serum Albumin (HSA). We find that the release of MCs remains favorable for the complexation leading to a considerable amount of release entropy as the driving force for complexation. The release of DCs only occurs above a certain DC concentration with a comparably smaller number of released ions than MCs. Its contribution to the binding free energy is small indicating a subtle cancellation between the entropy gain in releasing DCs and the enthalpy penalty from dissociating DCs from the dendrimer.
Collapse
Affiliation(s)
- Xiao Xu
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, P. R. China.
| | - Xu Jia
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, P. R. China.
| | - Yuejun Zhang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, P. R. China.
| |
Collapse
|
28
|
Ahmadi V, Nie C, Mohammadifar E, Achazi K, Wedepohl S, Kerkhoff Y, Block S, Osterrieder K, Haag R. One-pot gram-scale synthesis of virucidal heparin-mimicking polymers as HSV-1 inhibitors. Chem Commun (Camb) 2021; 57:11948-11951. [PMID: 34671786 DOI: 10.1039/d1cc04703e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A straightforward and gram-scale synthesis method was developed to engineer highly sulfated hyperbranched polyglycerol bearing sulfated alkyl chains. The compounds with shorter alkyl chains showed multivalent virustatic inhibition against herpes simplex virus type 1 (HSV-1), similar to heparin. In contrast, the compound with the longest alkyl chains irreversibly inhibited the virus.
Collapse
Affiliation(s)
- Vahid Ahmadi
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany.
| | - Chuanxiong Nie
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany.
| | - Ehsan Mohammadifar
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany.
| | - Katharina Achazi
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany.
| | - Stefanie Wedepohl
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany.
| | - Yannic Kerkhoff
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany.
| | - Stephan Block
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany.
| | - Klaus Osterrieder
- Institut für Virologie, Robert von Ostertag-Haus, Zentrum für Infektionsmedizin Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163 Berlin, Germany
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany.
| |
Collapse
|
29
|
Koide H. [Design of Synthetic Polymer Nanoparticles That Capture and Neutralize Target Molecules]. YAKUGAKU ZASSHI 2021; 141:1079-1086. [PMID: 34471009 DOI: 10.1248/yakushi.21-00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Protein affinity reagents that specifically and strongly bind to target molecules are widely used in disease detection, diagnosis, and therapy. Although antibodies and their fragments are the gold standard in protein-protein inhibitors (PPIs), synthetic polymers such as linear polymers, dendrimers, and nanoparticles as cost-effective PPIs have attracted great attention as alternatives to antibodies. These polymers exhibit high affinity to the target by imitating natural protein-protein interactions. However, only a few in vivo applications have been reported. Here, our recent advances in the development of synthetic polymers for in vivo application are reviewed. Poly(N-isopropylacrylamide) (pNIPAm) was used as a model of synthetic affinity reagents. Incorporation of both sulfated carbohydrate and hydrophobic monomers into lightly crosslinked pNIPAm nanoparticles (NPs) captured and neutralized vascular endothelial growth factor (VEGF) and inhibited tumor growth upon intravenous injection into tumor-bearing mice. Modification of a liposome with the pNIPAm-based linear polymer increased the polymer circulation time after intravenous injection and improved the affinity for the target. The pNIPAm-based NPs delivered by oral administration captured the target small molecules and inhibited their absorption from the intestine. Our recent findings provide useful information for the design of synthetic polymers that capture target molecules in vivo.
Collapse
Affiliation(s)
- Hiroyuki Koide
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
30
|
Yasuno G, Koide H, Oku N, Asai T. Influence of Purification Process on the Function of Synthetic Polymer Nanoparticles. Chem Pharm Bull (Tokyo) 2021; 69:773-780. [PMID: 34334521 DOI: 10.1248/cpb.c21-00273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Multifunctional synthetic polymers can bind to target molecules and are therefore widely investigated in diagnostics, drug delivery carriers, and separation carriers. Because these polymers are synthesized from nonbiological components, purification processes (e.g., chromatography, dialysis, extraction, and centrifugation) must be conducted after the synthesis. Although several purification methods are used for polymer purification, few reports have revealed the influence of purification process on the functions of polymer. In this study, we demonstrated that the characteristics, function, and stability of synthetic polymer depend on the purification process. N-Isopropylacrylamide-based polymer nanoparticles (NPs) and melittin (i.e., honey bee venom) were used as a model of synthetic polymer and target toxic peptide, respectively. Synthesized NPs were purified by dialysis in methanol, acetone precipitation, or centrifugation. NPs purified by dialysis in ultrapure water were used as control NPs. Then, NP size, surface charge, toxin neutralization effect, and stability were determined. NP size did not considerably change by purification with centrifugation; however, it decreased by purification using dialysis in methanol and acetone precipitation compared with that of control NPs. The ζ-potential of NPs changed after each purification process compared with that of control NPs. The melittin neutralization efficiency of NPs depended on the purification process; i.e., it decreased by acetone precipitation and increased by dialysis in methanol and centrifugation compared with that of control NPs. Of note, the addition of methanol and acetone decreased NP stability. These studies implied the importance of considering the effect of the purification method on synthetic polymer function.
Collapse
Affiliation(s)
- Go Yasuno
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences
| | - Hiroyuki Koide
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences
| | - Naoto Oku
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences.,Faculty of Pharma-Science, Teikyo University
| | - Tomohiro Asai
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences
| |
Collapse
|
31
|
Nakamoto M, Escalante T, Gutiérrez JM, Shea KJ. A Biomimetic of Endogenous Tissue Inhibitors of Metalloproteinases: Inhibition Mechanism and Contribution of Composition, Polymer Size, and Shape to the Inhibitory Effect. NANO LETTERS 2021; 21:5663-5670. [PMID: 34181420 DOI: 10.1021/acs.nanolett.1c01357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
A biomimetic of endogenous tissue inhibitors of metalloproteinases (TIMPs) was engineered by introducing three binding elements to a synthetic tetrapolymer. We evaluated the contribution of composition, size, and shape of the TIMP-mimicking polymers to the inhibition of BaP1, a P-I class snake venom metalloproteinase (SVMP). Inhibition was achieved when the size of the linear polymer (LP) was comparable to or greater than that of the enzyme, indicating the efficacy requires binding to a significant portion of the enzyme surface in the vicinity of the active site. The efficacy of a low cross-linked polymer hydrogel nanoparticle (NP) of substantially greater molecular weight was comparable to that of the LPs despite differences in size and shape, an important finding for in vivo applications. The abiotic TIMP was effective against two classes of SVMPs in whole snake venom. The results can serve as a design principle for biomimetic polymer inhibitors of enzymes.
Collapse
Affiliation(s)
- Masahiko Nakamoto
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Teresa Escalante
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José 11501, Costa Rica
| | - José M Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José 11501, Costa Rica
| | - Kenneth J Shea
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| |
Collapse
|
32
|
Nie C, Pouyan P, Lauster D, Trimpert J, Kerkhoff Y, Szekeres GP, Wallert M, Block S, Sahoo AK, Dernedde J, Pagel K, Kaufer BB, Netz RR, Ballauff M, Haag R. Polysulfate hemmen durch elektrostatische Wechselwirkungen die SARS‐CoV‐2‐Infektion**. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Chuanxiong Nie
- Institut für Chemie und Biochemie Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
- Institut für Virologie Freie Universität Berlin Robert-von-Ostertag-Straße 7–13 14163 Berlin Deutschland
| | - Paria Pouyan
- Institut für Chemie und Biochemie Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
| | - Daniel Lauster
- Institut für Chemie und Biochemie Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
| | - Jakob Trimpert
- Institut für Virologie Freie Universität Berlin Robert-von-Ostertag-Straße 7–13 14163 Berlin Deutschland
| | - Yannic Kerkhoff
- Department of Chemistry and Biochemistry Emmy-Noether Group “Bionanointerfaces” Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
| | - Gergo Peter Szekeres
- Institut für Chemie und Biochemie Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
- Department of Molecular Physics Fritz Haber Institute of the Max Planck Society Faradayweg 4–6 14195 Berlin Deutschland
| | - Matthias Wallert
- Department of Chemistry and Biochemistry Emmy-Noether Group “Bionanointerfaces” Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
| | - Stephan Block
- Department of Chemistry and Biochemistry Emmy-Noether Group “Bionanointerfaces” Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
| | - Anil Kumar Sahoo
- Fachbereich Physik Freie Universität Berlin Arnimallee 14 14195 Berlin Deutschland
- Max Planck Institute of Colloids and Interfaces Am Mühlenberg 1 14476 Potsdam Deutschland
| | - Jens Dernedde
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie Charité-Universitätsmedizin Berlin Augustenburgerplatz 1 13353 Berlin Deutschland
| | - Kevin Pagel
- Institut für Chemie und Biochemie Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
- Department of Molecular Physics Fritz Haber Institute of the Max Planck Society Faradayweg 4–6 14195 Berlin Deutschland
| | - Benedikt B. Kaufer
- Institut für Virologie Freie Universität Berlin Robert-von-Ostertag-Straße 7–13 14163 Berlin Deutschland
| | - Roland R. Netz
- Fachbereich Physik Freie Universität Berlin Arnimallee 14 14195 Berlin Deutschland
| | - Matthias Ballauff
- Institut für Chemie und Biochemie Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
| | - Rainer Haag
- Institut für Chemie und Biochemie Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
| |
Collapse
|
33
|
Nie C, Pouyan P, Lauster D, Trimpert J, Kerkhoff Y, Szekeres GP, Wallert M, Block S, Sahoo AK, Dernedde J, Pagel K, Kaufer BB, Netz RR, Ballauff M, Haag R. Polysulfates Block SARS-CoV-2 Uptake through Electrostatic Interactions*. Angew Chem Int Ed Engl 2021; 60:15870-15878. [PMID: 33860605 PMCID: PMC8250366 DOI: 10.1002/anie.202102717] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/29/2021] [Indexed: 12/20/2022]
Abstract
Here we report that negatively charged polysulfates can bind to the spike protein of SARS-CoV-2 via electrostatic interactions. Using a plaque reduction assay, we compare inhibition of SARS-CoV-2 by heparin, pentosan sulfate, linear polyglycerol sulfate (LPGS) and hyperbranched polyglycerol sulfate (HPGS). Highly sulfated LPGS is the optimal inhibitor, with an IC50 of 67 μg mL-1 (approx. 1.6 μm). This synthetic polysulfate exhibits more than 60-fold higher virus inhibitory activity than heparin (IC50 : 4084 μg mL-1 ), along with much lower anticoagulant activity. Furthermore, in molecular dynamics simulations, we verified that LPGS can bind more strongly to the spike protein than heparin, and that LPGS can interact even more with the spike protein of the new N501Y and E484K variants. Our study demonstrates that the entry of SARS-CoV-2 into host cells can be blocked via electrostatic interactions, therefore LPGS can serve as a blueprint for the design of novel viral inhibitors of SARS-CoV-2.
Collapse
Affiliation(s)
- Chuanxiong Nie
- Institut für Chemie und BiochemieFreie Universität BerlinArnimallee 2214195BerlinGermany
- Institut für VirologieFreie Universität BerlinRobert-von-Ostertag-Strasse 7–1314163BerlinGermany
| | - Paria Pouyan
- Institut für Chemie und BiochemieFreie Universität BerlinArnimallee 2214195BerlinGermany
| | - Daniel Lauster
- Institut für Chemie und BiochemieFreie Universität BerlinArnimallee 2214195BerlinGermany
| | - Jakob Trimpert
- Institut für VirologieFreie Universität BerlinRobert-von-Ostertag-Strasse 7–1314163BerlinGermany
| | - Yannic Kerkhoff
- Department of Chemistry and BiochemistryEmmy-Noether Group “Bionanointerfaces”Freie Universität BerlinArnimallee 2214195BerlinGermany
| | - Gergo Peter Szekeres
- Institut für Chemie und BiochemieFreie Universität BerlinArnimallee 2214195BerlinGermany
- Department of Molecular PhysicsFritz Haber Institute of the Max Planck SocietyFaradayweg 4–614195BerlinGermany
| | - Matthias Wallert
- Department of Chemistry and BiochemistryEmmy-Noether Group “Bionanointerfaces”Freie Universität BerlinArnimallee 2214195BerlinGermany
| | - Stephan Block
- Department of Chemistry and BiochemistryEmmy-Noether Group “Bionanointerfaces”Freie Universität BerlinArnimallee 2214195BerlinGermany
| | - Anil Kumar Sahoo
- Fachbereich PhysikFreie Universität BerlinArnimallee 1414195BerlinGermany
- Max Planck Institute of Colloids and InterfacesAm Mühlenberg 114476PotsdamGermany
| | - Jens Dernedde
- Institut für Laboratoriumsmedizin, Klinische Chemie und PathobiochemieCharité-Universitätsmedizin BerlinAugustenburgerplatz 113353BerlinGermany
| | - Kevin Pagel
- Institut für Chemie und BiochemieFreie Universität BerlinArnimallee 2214195BerlinGermany
- Department of Molecular PhysicsFritz Haber Institute of the Max Planck SocietyFaradayweg 4–614195BerlinGermany
| | - Benedikt B. Kaufer
- Institut für VirologieFreie Universität BerlinRobert-von-Ostertag-Strasse 7–1314163BerlinGermany
| | - Roland R. Netz
- Fachbereich PhysikFreie Universität BerlinArnimallee 1414195BerlinGermany
| | - Matthias Ballauff
- Institut für Chemie und BiochemieFreie Universität BerlinArnimallee 2214195BerlinGermany
| | - Rainer Haag
- Institut für Chemie und BiochemieFreie Universität BerlinArnimallee 2214195BerlinGermany
| |
Collapse
|
34
|
Koide H, Yamauchi I, Hoshino Y, Yasuno G, Okamoto T, Akashi S, Saito K, Oku N, Asai T. Design of abiotic polymer ligand-decorated lipid nanoparticles for effective neutralization of target toxins in the blood. Biomater Sci 2021; 9:5588-5598. [PMID: 34241600 DOI: 10.1039/d1bm00515d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Macromolecular toxins often induce inflammatory cytokine production, multiple-organ dysfunction, and cell death. Synthetic polymer ligands (PLs) prepared with several functional monomers have the potential of neutralizing target toxins after binding to them; therefore, they are of significant interest as abiotic antidotes. Although PLs show little toxin neutralization effect in the bloodstream because of immediate elimination from there, the toxin neutralization effect is significantly improved by the direct decoration of PLs onto lipid nanoparticles (PL-LNPs). However, this direct decoration decreases PL mobility, induces LNP aggregation after capturing the target, and decreases LNP blood circulation time. We designed novel PL-LNPs to improve PL mobility, inhibit the aggregation tendency after capturing the target, and increase LNP blood circulation time in order to achieve highly effective toxin neutralization in vivo. Specifically, LNPs were modified with PLs-conjugated polyethylene glycol (PEG), and additional PEG was used to modify the PL-decorated LNPs (PL-PEG-LNPs). Histones were used as target toxins, and N-isopropylacrylamide-based PLs were used for histone capture. PEGylation increased the plasma LNP level 24 h after intravenous injection by ∼90 times and inhibited LNP aggregation after histone capture. The dissociation constant (Kd) of PL-PEG-LNPs against histone was two times smaller compared to that of PL-LNPs. Although PL-LNPs inhibited histone-platelet interaction in the bloodstream, a large amount of histone-PL-LNP complexes accumulated in the lungs because of aggregation. However, PL-PEG-LNPs inhibited both histone-platelet interaction and histone accumulation in the lungs. Importantly, PL-PEG-LNP treatment increased the survival rate of histone-treated mice compared to PL-LNPs. These results provide a platform for the development of abiotic antidote nanoparticles in vivo.
Collapse
Affiliation(s)
- Hiroyuki Koide
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan.
| | - Ikumi Yamauchi
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan.
| | - Yu Hoshino
- Department of Chemical Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Go Yasuno
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan.
| | - Takumi Okamoto
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan.
| | - Sotaro Akashi
- Department of Chemical Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Kazuhiro Saito
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan.
| | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan. and Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan.
| |
Collapse
|
35
|
Braatz D, Dimde M, Ma G, Zhong Y, Tully M, Grötzinger C, Zhang Y, Mavroskoufis A, Schirner M, Zhong Z, Ballauff M, Haag R. Toolbox of Biodegradable Dendritic (Poly glycerol sulfate)-SS-poly(ester) Micelles for Cancer Treatment: Stability, Drug Release, and Tumor Targeting. Biomacromolecules 2021; 22:2625-2640. [PMID: 34076415 DOI: 10.1021/acs.biomac.1c00333] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this paper, we present well-defined dPGS-SS-PCL/PLGA/PLA micellar systems demonstrating excellent capabilities as a drug delivery platform in light of high stability and precise in vitro and in vivo drug release combined with active targetability to tumors. These six amphiphilic block copolymers were each targeted in two different molecular weights (8 or 16 kDa) and characterized using 1H NMR, gel permeation chromatography (GPC), and elemental analysis. The block copolymer micelles showed monodispersed size distributions of 81-187 nm, strong negative charges between -52 and -41 mV, and low critical micelle concentrations (CMCs) of up to 1.13-3.58 mg/L (134-527 nM). The serum stability was determined as 94% after 24 h. The drug-loading efficiency for Sunitinib ranges from 38 to 83% (8-17 wt %). The release was selectively triggered by glutathione (GSH) and lipase, reaching 85% after 5 days, while only 20% leaching was observed under physiological conditions. Both the in vitro and in vivo studies showed sustained release of Sunitinib over 1 week. CCK-8 assays on HeLa lines demonstrated the high cell compatibility (1 mg/mL, 94% cell viability, 48 h) and the high cancer cell toxicity of Sunitinib-loaded micelles (IC50 2.5 μg/mL). By in vivo fluorescence imaging studies on HT-29 tumor-bearing mice, the targetability of dPGS7.8-SS-PCL7.8 enabled substantial accumulation in tumor tissue compared to nonsulfated dPG3.9-SS-PCL7.8. As a proof of concept, Sunitinib-loaded dPGS-SS-poly(ester) micelles improved the antitumor efficacy of the chemotherapeutic. A tenfold lower dosage of loaded Sunitinib led to an even higher tumor growth inhibition compared to the free drug, as demonstrated in a HeLa human cervical tumor-bearing mice model. No toxicity for the organism was observed, confirming the good biocompatibility of the system.
Collapse
Affiliation(s)
- Daniel Braatz
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Mathias Dimde
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Guoxin Ma
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Yinan Zhong
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Michael Tully
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Carsten Grötzinger
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, 13353 Berlin, Germany
| | - Yuanyuan Zhang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Alexandros Mavroskoufis
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Michael Schirner
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Matthias Ballauff
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| |
Collapse
|
36
|
Cherri M, Ferraro M, Mohammadifar E, Quaas E, Achazi K, Ludwig K, Grötzinger C, Schirner M, Haag R. Biodegradable Dendritic Polyglycerol Sulfate for the Delivery and Tumor Accumulation of Cytostatic Anticancer Drugs. ACS Biomater Sci Eng 2021; 7:2569-2579. [PMID: 34061498 DOI: 10.1021/acsbiomaterials.1c00439] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Targeted delivery and extended blood circulation of anticancer drugs have been the challenges for decreasing the adverse side effects and improving the therapeutic efficiency in cancer chemotherapy. Herein, we present a drug delivery system (DDS) based on biodegradable dendritic polyglycerol sulfate-bearing poly(caprolactone) (dPGS-PCL) chains, which has been synthesized on 20 g scale using a straightforward two-step protocol. In vivo fluorescence imaging demonstrated a significant accumulation of the DDS in the tumor environment. Sunitinib, an anticancer drug, was loaded into the DDS and the drug-induced toxicity was investigated in vitro and in vivo. The drug encapsulated in dPGS-PCL and the free drug showed similar toxicities in A431 and HT-29 cells, and the cellular uptake was comparable. The straightforward and large-scale synthesis, the organic solvent-free drug-loading approach, together with the tumor targetability of the biodegradable dendritic polyglycerols, render this copolymer a promising candidate for targeted cancer nanomedicine drug delivery systems.
Collapse
Affiliation(s)
- Mariam Cherri
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Magda Ferraro
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Ehsan Mohammadifar
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Elisa Quaas
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Katharina Achazi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Kai Ludwig
- Research Center for Electron Microscopy and Core Facility BioSupraMol, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 36a, 14195 Berlin, Germany
| | - Carsten Grötzinger
- Department of Hepatology and Gastroenterology, and Molecular Cancer Research Center (MKFZ), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CVK Augustenburger Platz 1, 13353 Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, 13353 Berlin, Germany
| | - Michael Schirner
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| |
Collapse
|
37
|
Koide H, Suzuki H, Ochiai H, Egami H, Hamashima Y, Oku N, Asai T. Enhancement of target toxin neutralization effect in vivo by PEGylation of multifunctionalized lipid nanoparticles. Biochem Biophys Res Commun 2021; 555:32-39. [PMID: 33812056 DOI: 10.1016/j.bbrc.2021.03.073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/15/2021] [Indexed: 01/02/2023]
Abstract
Protein-protein (e.g., antibody-antigen) interactions comprise multiple weak interactions. We have previously reported that lipid nanoparticles (LNPs) bind to and neutralize target toxic peptides after multifunctionalization of the LNP surface (MF-LNPs) with amino acid derivatives that induce weak interactions; however, the MF-LNPs aggregated after target capture and showed short blood circulation times. Here we optimized polyethylene glycol (PEG)-modified MF-LNPs (PEG-MF-LNPs) to inhibit the aggregation and increase the blood circulation time. Melittin was used as a target toxin, and MF-LNPs were prepared with negatively charged, hydrophobic, and neutral amino-acid-derivative-conjugated functional lipids. In this study, MF-LNPs modified with only PEG5k (PEG5k-MF-LNPs) and with both PEG5k and PEG2k (PEGmix-MF-LNPs) were prepared, where PEG5k and PEG2k represent PEG with a molecular weight of 5000 and 2000, respectively. PEGylation of the MF-LNPs did not decrease the melittin neutralization ability of nonPEGylated MF-LNPs, as tested by hemolysis assay. The PEGmix-MF-LNPs showed better blood circulation characteristics than the PEG5k-MF-LNPs. Although the nonPEGylated MF-LNPs immediately aggregated when mixed with melittin, the PEGmix-MF-LNPs did not aggregate. The PEGmix-MF-LNPs dramatically increased the survival rate of melittin-treated mice, whereas the nonPEGylated MF-LNPs increased slightly. These results provide a fundamental strategy to improve the in vivo toxin neutralization ability of MF-LNPs.
Collapse
Affiliation(s)
- Hiroyuki Koide
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan.
| | - Hikaru Suzuki
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Hiroki Ochiai
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Hiromichi Egami
- Department of Synthetic Organic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka, 422-8526, Japan
| | - Yoshitaka Hamashima
- Department of Synthetic Organic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka, 422-8526, Japan
| | - Naoto Oku
- Faculty of Pharma-Science, Teikyo University, 2-11-1 kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| |
Collapse
|
38
|
Zhu FD, Hu YJ, Yu L, Zhou XG, Wu JM, Tang Y, Qin DL, Fan QZ, Wu AG. Nanoparticles: A Hope for the Treatment of Inflammation in CNS. Front Pharmacol 2021; 12:683935. [PMID: 34122112 PMCID: PMC8187807 DOI: 10.3389/fphar.2021.683935] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation, an inflammatory response within the central nervous system (CNS), is a main hallmark of common neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), among others. The over-activated microglia release pro-inflammatory cytokines, which induces neuronal death and accelerates neurodegeneration. Therefore, inhibition of microglia over-activation and microglia-mediated neuroinflammation has been a promising strategy for the treatment of neurodegenerative diseases. Many drugs have shown promising therapeutic effects on microglia and inflammation. However, the blood–brain barrier (BBB)—a natural barrier preventing brain tissue from contact with harmful plasma components—seriously hinders drug delivery to the microglial cells in CNS. As an emerging useful therapeutic tool in CNS-related diseases, nanoparticles (NPs) have been widely applied in biomedical fields for use in diagnosis, biosensing and drug delivery. Recently, many NPs have been reported to be useful vehicles for anti-inflammatory drugs across the BBB to inhibit the over-activation of microglia and neuroinflammation. Therefore, NPs with good biodegradability and biocompatibility have the potential to be developed as an effective and minimally invasive carrier to help other drugs cross the BBB or as a therapeutic agent for the treatment of neuroinflammation-mediated neurodegenerative diseases. In this review, we summarized various nanoparticles applied in CNS, and their mechanisms and effects in the modulation of inflammation responses in neurodegenerative diseases, providing insights and suggestions for the use of NPs in the treatment of neuroinflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Feng-Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yu-Jiao Hu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Anesthesia, Southwest Medical University, Luzhou, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qing-Ze Fan
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
39
|
Liu F, Sheng S, Shao D, Xiao Y, Zhong Y, Zhou J, Quek CH, Wang Y, Hu Z, Liu H, Li Y, Tian H, Leong KW, Chen X. A Cationic Metal-Organic Framework to Scavenge Cell-Free DNA for Severe Sepsis Management. NANO LETTERS 2021; 21:2461-2469. [PMID: 33686851 PMCID: PMC8320336 DOI: 10.1021/acs.nanolett.0c04759] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Circulating cell-free DNA (cfDNA) released by damaged cells causes inflammation and has been associated with the progression of sepsis. One proposed strategy to treat sepsis is to scavenge this inflammatory circulating cfDNA. Here, we develop a cfDNA-scavenging nanoparticle (NP) that consists of cationic polyethylenimine (PEI) of different molecular weight grafted to zeolitic imidazolate framework-8 (PEI-g-ZIF) in a simple one-pot process. PEI-g-ZIF NPs fabricated using PEI 1800 and PEI 25k but not PEI 600 suppressed cfDNA-induced TLR activation and subsequent nuclear factor kappa B pathway activity. PEI 1800-g-ZIF NPs showed greater inhibition of cfDNA-associated inflammation and multiple organ injury than naked PEI 1800 (lacking ZIF), and had greater therapeutic efficacy in treating sepsis. These results indicate that PEI-g-ZIF NPs acts as a "nanotrap" that improves upon naked PEI in scavenging circulating cfDNA, reducing inflammation, and reversing the progression of sepsis, thus providing a novel strategy for sepsis treatment.
Collapse
Affiliation(s)
- Feng Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shu Sheng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dan Shao
- Institutes for Life Sciences, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 510006, China
| | - Yongqiang Xiao
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Yiling Zhong
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Jie Zhou
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Chai Hoon Quek
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Yanbing Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- University of Science and Technology of China, Hefei 230026, China
| | - Zhiming Hu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Heshi Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yanhui Li
- School of Materials Science and Engineering, Changchun University of Science and Technology, Changchun 130022, China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- University of Science and Technology of China, Hefei 230026, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
40
|
Reisbeck F, Ozimkovski A, Cherri M, Dimde M, Quaas E, Mohammadifar E, Achazi K, Haag R. Gram Scale Synthesis of Dual-Responsive Dendritic Polyglycerol Sulfate as Drug Delivery System. Polymers (Basel) 2021; 13:982. [PMID: 33806866 PMCID: PMC8004855 DOI: 10.3390/polym13060982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 11/18/2022] Open
Abstract
Biocompatible polymers with the ability to load and release a cargo at the site of action in a smart response to stimuli have attracted great attention in the field of drug delivery and cancer therapy. In this work, we synthesize a dual-responsive dendritic polyglycerol sulfate (DR-dPGS) drug delivery system by copolymerization of glycidol, ε-caprolactone and an epoxide monomer bearing a disulfide bond (SSG), followed by sulfation of terminal hydroxyl groups of the copolymer. The effect of different catalysts, including Lewis acids and organic bases, on the molecular weight, monomer content and polymer structure was investigated. The degradation of the polymer backbone was proven in presence of reducing agents and candida antarctica Lipase B (CALB) enzyme, which results in the cleavage of the disulfides and ester bonds, respectively. The hydrophobic anticancer drug Doxorubicin (DOX) was loaded in the polymer and the kinetic assessment showed an enhanced drug release with glutathione (GSH) or CALB as compared to controls and a synergistic effect of a combination of both stimuli. Cell uptake was studied by using confocal laser scanning microscopy with HeLa cells and showed the uptake of the Dox-loaded carriers and the release of the drug into the nucleus. Cytotoxicity tests with three different cancer cell lines showed good tolerability of the polymers of as high concentrations as 1 mg mL-1, while cancer cell growth was efficiently inhibited by DR-dPGS@Dox.
Collapse
Affiliation(s)
| | | | | | | | | | - Ehsan Mohammadifar
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany; (F.R.); (A.O.); (M.C.); (M.D.); (E.Q.)
| | - Katharina Achazi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany; (F.R.); (A.O.); (M.C.); (M.D.); (E.Q.)
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany; (F.R.); (A.O.); (M.C.); (M.D.); (E.Q.)
| |
Collapse
|
41
|
Pouyan P, Nie C, Bhatia S, Wedepohl S, Achazi K, Osterrieder N, Haag R. Inhibition of Herpes Simplex Virus Type 1 Attachment and Infection by Sulfated Polyglycerols with Different Architectures. Biomacromolecules 2021; 22:1545-1554. [PMID: 33706509 DOI: 10.1021/acs.biomac.0c01789] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inhibition of herpes simplex virus type 1 (HSV-1) binding to the host cell surface by highly sulfated architectures is among the promising strategies to prevent virus entry and infection. However, the structural flexibility of multivalent inhibitors plays a major role in effective blockage and inhibition of virus receptors. In this study, we demonstrate the inhibitory effect of a polymer scaffold on the HSV-1 infection by using highly sulfated polyglycerols with different architectures (linear, dendronized, and hyperbranched). IC50 values for all synthesized sulfated polyglycerols and the natural sulfated polymer heparin were determined using plaque reduction infection assays. Interestingly, an increase in the IC50 value from 0.03 to 374 nM from highly flexible linear polyglycerol sulfate (LPGS) to less flexible scaffolds, namely, dendronized polyglycerol sulfate and hyperbranched polyglycerol sulfate was observed. The most potent LPGS inhibits HSV-1 infection 295 times more efficiently than heparin, and we show that LPGS has a much reduced anticoagulant capacity when compared to heparin as evidenced by measuring the activated partial thromboplastin time. Furthermore, prevention of infection by LPGS and the commercially available drug acyclovir were compared. All tested sulfated polymers do not show any cytotoxicity at concentrations of up to 1 mg/mL in different cell lines. We conclude from our results that more flexible polyglycerol sulfates are superior to less flexible sulfated polymers with respect to inhibition of HSV-1 infection and may constitute an alternative to the current antiviral treatments of this ubiquitous pathogen.
Collapse
Affiliation(s)
- Paria Pouyan
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Chuanxiong Nie
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany.,Institut für Virologie, Robert von Ostertag-Haus, Zentrum für Infektionsmedizin, Freie Universität Berlin, Robert-von-Ostertag-Street 7-13, Berlin 14163, Germany
| | - Sumati Bhatia
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Stefanie Wedepohl
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee. 22, Berlin 14195, Germany
| | - Katharina Achazi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee. 22, Berlin 14195, Germany
| | - Nikolaus Osterrieder
- Institut für Virologie, Robert von Ostertag-Haus, Zentrum für Infektionsmedizin, Freie Universität Berlin, Robert-von-Ostertag-Street 7-13, Berlin 14163, Germany.,Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon Tong TYB-1B-507, Hong Kong
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| |
Collapse
|
42
|
Achazi K, Haag R, Ballauff M, Dernedde J, Kizhakkedathu JN, Maysinger D, Multhaup G. Understanding the Interaction of Polyelectrolyte Architectures with Proteins and Biosystems. Angew Chem Int Ed Engl 2021; 60:3882-3904. [PMID: 32589355 PMCID: PMC7894192 DOI: 10.1002/anie.202006457] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Indexed: 02/06/2023]
Abstract
The counterions neutralizing the charges on polyelectrolytes such as DNA or heparin may dissociate in water and greatly influence the interaction of such polyelectrolytes with biomolecules, particularly proteins. In this Review we give an overview of studies on the interaction of proteins with polyelectrolytes and how this knowledge can be used for medical applications. Counterion release was identified as the main driving force for the binding of proteins to polyelectrolytes: Patches of positive charge become multivalent counterions of the polyelectrolyte and lead to the release of counterions from the polyelectrolyte and a concomitant increase in entropy. This is shown from investigations on the interaction of proteins with natural and synthetic polyelectrolytes. Special emphasis is paid to sulfated dendritic polyglycerols (dPGS). The Review demonstrates that we are moving to a better understanding of charge-charge interactions in systems of biological relevance. Research along these lines will aid and promote the design of synthetic polyelectrolytes for medical applications.
Collapse
Affiliation(s)
- Katharina Achazi
- Institut für Chemie und BiochemieFreie Universität BerlinTakustrasse 314195BerlinGermany
| | - Rainer Haag
- Institut für Chemie und BiochemieFreie Universität BerlinTakustrasse 314195BerlinGermany
| | - Matthias Ballauff
- Institut für Chemie und BiochemieFreie Universität BerlinTakustrasse 314195BerlinGermany
- IRIS AdlershofHumboldt Universität zu BerlinZum Grossen Windkanal 612489BerlinGermany
| | - Jens Dernedde
- Charité-Universitätsmedizin BerlinInstitute of Laboratory MedicineClinical Chemistry, and PathobiochemistryCVK Augustenburger Platz 113353BerlinGermany
| | - Jayachandran N. Kizhakkedathu
- Centre for Blood ResearchDepartment of Pathology and Laboratory MedicineLife Science InstituteDepartment of ChemistrySchool of Biomedical EngineeringUniversity of British ColumbiaVancouverV6T 1Z3Canada
| | - Dusica Maysinger
- Department of Pharmacology and TherapeuticsMcGill UniversityMontrealH3G 1Y6Canada
| | - Gerd Multhaup
- Department of Pharmacology and TherapeuticsMcGill UniversityMontrealH3G 1Y6Canada
| |
Collapse
|
43
|
Straßburger D, Herziger S, Huth K, Urschbach M, Haag R, Besenius P. Supramolecular polymerization of sulfated dendritic peptide amphiphiles into multivalent L-selectin binders. Beilstein J Org Chem 2021; 17:97-104. [PMID: 33519996 PMCID: PMC7814183 DOI: 10.3762/bjoc.17.10] [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: 10/13/2020] [Accepted: 12/14/2020] [Indexed: 11/23/2022] Open
Abstract
The synthesis of a sulfate-modified dendritic peptide amphiphile and its self-assembly into one-dimensional rod-like architectures in aqueous medium is reported. The influence of the ionic strength on the supramolecular polymerization was probed via circular dichroism spectroscopy and cryogenic transmission electron microscopy. Physiological salt concentrations efficiently screen the charges of the dendritic building block equipped with eight sulfate groups and trigger the formation of rigid supramolecular polymers. Since multivalent sulfated supramolecular structures mimic naturally occurring L-selectin ligands, the corresponding affinity was evaluated using a competitive SPR binding assay and benchmarked to an ethylene glycol-decorated supramolecular polymer.
Collapse
Affiliation(s)
- David Straßburger
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Svenja Herziger
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany.,Research Center of Electron Microscopy, Freie Universität Berlin, Fabeckstr. 34a, 14195 Berlin
| | - Katharina Huth
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany
| | - Moritz Urschbach
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany
| | - Pol Besenius
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| |
Collapse
|
44
|
Kumari M, Prasad S, Fruk L, Parshad B. Polyglycerol-based hydrogels and nanogels: from synthesis to applications. Future Med Chem 2021; 13:419-438. [PMID: 33403867 DOI: 10.4155/fmc-2020-0205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hydrogels and nanogels have emerged as promising materials for biomedical applications owing to their large surface area and tunable mechanical and chemical properties. Their large surface area is well suited for bioconjugation, whilst the interior porous network can be utilized for the transport of valuable biomolecules. The use of biocompatible hydrophilic building blocks/linkers for the preparation of hydrogels and nanogels not only avoids undesired side effects within the biological system, but also retains high water content, thereby creating an environment which is very similar to extracellular matrix. Their tunable multivalency and hydrophilicity and excellent biocompatibility, together with ease of functionalization, makes polyglycerol macromonomers well suited for synthesizing cross-linked networks that can be used as extracellular matrix mimics. Here we provide an overview of the synthesis of polyglycerol-based hydrogels and nanogels for various biomedical applications.
Collapse
Affiliation(s)
- Meena Kumari
- Department of Chemistry, Government College for Women, Badhra, Ch. Dadri, Haryana 127308, India
| | - Suchita Prasad
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Ljiljana Fruk
- Department of Chemical Engineering & Biotechnology, University of Cambridge, Cambridge, CB3 0AS, UK
| | - Badri Parshad
- Department of Chemical Engineering & Biotechnology, University of Cambridge, Cambridge, CB3 0AS, UK
| |
Collapse
|
45
|
Abstract
Protein affinity reagents are widely used for basic research, diagnostics, and disease therapy. Antibodies and their fragments are known as the most common protein affinity reagents. They specifically and strongly bind to target molecules and inhibit their functions. Thus, antibody drugs have increased in the recent two decades for disease therapy, such as cancer. These strong protein-protein interactions are composed of a nexus of multiple weak interactions. Synthetic polymers that bind to target molecules have been developed by the imitation of protein-protein interactions. These polymers show nanomolar affinity for the target and neutralize their functions; thus, they are of significant interest as a cost-effective protein affinity reagent. We have been developing synthetic polymer nanoparticles (NPs) that bind to target peptides and proteins by the inclusion of several functional monomers, such as charged and hydrophobic monomers. In this review, the focus is on the design of synthetic polymer NPs that bind to target molecules for disease therapy. We succeeded in neutralization of toxic peptides and signaling proteins both in vitro and in vivo. Additionally, linear polymers were modified on a lipid nanoparticle surface to improve polymer biodistribution. Our recent findings should provide useful information for the development of abiotic protein affinity reagents.
Collapse
Affiliation(s)
- Hiroyuki Koide
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
46
|
Salta J, Arp FF, Kühne C, Reissig H. Multivalent 1,2,3‐Triazole‐Linked Carbohydrate Mimetics by Huisgen–Meldal‐Sharpless Cycloadditions of an Azidopyran. European J Org Chem 2020. [DOI: 10.1002/ejoc.202001389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Joana Salta
- Institut für Chemie und Biochemie Freie Universität Berlin Takustrasse 3 14195 Berlin Germany
| | - Fabian F. Arp
- Institut für Chemie und Biochemie Freie Universität Berlin Takustrasse 3 14195 Berlin Germany
| | - Christian Kühne
- Institut für Laboratoriumsmedizin Klinische Chemie und Pathobiochemie Charité‐Universitätsmedizin Berlin Augustenburger Platz 1 13353 Berlin Germany
| | - Hans‐Ulrich Reissig
- Institut für Chemie und Biochemie Freie Universität Berlin Takustrasse 3 14195 Berlin Germany
| |
Collapse
|
47
|
Rütter M, Milošević N, David A. Say no to drugs: Bioactive macromolecular therapeutics without conventional drugs. J Control Release 2020; 330:1191-1207. [PMID: 33207257 DOI: 10.1016/j.jconrel.2020.11.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/17/2022]
Abstract
The vast majority of nanomedicines (NM) investigated today consists of a macromolecular carrier and a drug payload (conjugated or encapsulated), with a purpose of preferential delivery of the drug to the desired site of action, either through passive accumulation, or by active targeting via ligand-receptor interaction. Several drug delivery systems (DDS) have already been approved for clinical use. However, recent reports are corroborating the notion that NM do not necessarily need to include a drug payload, but can exert biological effects through specific binding/blocking of important target proteins at the site of action. The seminal work of Kopeček et al. on N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers containing biorecognition motifs (peptides or oligonucleotides) for crosslinking cell surface non-internalizing receptors of malignant cells and inducing their apoptosis, without containing any low molecular weight drug, led to the definition of a special group of NM, termed Drug-Free Macromolecular Therapeutics (DFMT). Systems utilizing this approach are typically designed to employ pendant targeting-ligands on the same macromolecule to facilitate multivalent interactions with receptors. The lack of conventional small molecule drugs reduces toxicity and adverse effects at off-target sites. In this review, we describe different types of DFMT that possess biological activity without attached low molecular weight drugs. We classified the relevant research into several groups by their mechanisms of action, and compare the advantages and disadvantages of these different approaches. We show that identification of target sites, specificity of attached targeting ligands, binding affinity and the synthesis of carriers of defined size and ligand spacing are crucial aspects of DFMT development. We further discuss how knowledge in the field of NM accumulated in the past few decades can help in the design of a successful DFMT to speed up the translation into clinical practice.
Collapse
Affiliation(s)
- Marie Rütter
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Nenad Milošević
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Ayelet David
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel.
| |
Collapse
|
48
|
Lenders V, Koutsoumpou X, Sargsian A, Manshian BB. Biomedical nanomaterials for immunological applications: ongoing research and clinical trials. NANOSCALE ADVANCES 2020; 2:5046-5089. [PMID: 36132021 PMCID: PMC9418019 DOI: 10.1039/d0na00478b] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/22/2020] [Indexed: 05/04/2023]
Abstract
Research efforts on nanomaterial-based therapies for the treatment of autoimmune diseases and cancer have spiked and have made rapid progress over the past years. Nanomedicine has been shown to contribute significantly to overcome current therapeutic limitations, exhibiting advantages compared to conventional therapeutics, such as sustained drug release, delayed drug degradation and site-specific drug delivery. Multiple nanodrugs have reached the clinic, but translation is often hampered by either low targeting efficiency or undesired side effects. Nanomaterials, and especially inorganic nanoparticles, have gained criticism due to their potential toxic effects, including immunological alterations. However, many strategies have been attempted to improve the therapeutic efficacy of nanoparticles and exploit their unique properties for the treatment of inflammation and associated diseases. In this review, we elaborate on the immunomodulatory effects of nanomaterials, with a strong focus on the underlying mechanisms that lead to these specific immune responses. Nanomaterials to be discussed include inorganic nanoparticles such as gold, silica and silver, as well as organic nanomaterials such as polymer-, dendrimer-, liposomal- and protein-based nanoparticles. Furthermore, various approaches for tuning nanomaterials in order to enhance their efficacy and attenuate their immune stimulation or suppression, with respect to the therapeutic application, are described. Additionally, we illustrate how the acquired insights have been used to design immunotherapeutic strategies for a variety of diseases. The potential of nanomedicine-based therapeutic strategies in immunotherapy is further illustrated by an up to date overview of current clinical trials. Finally, recent efforts into enhancing immunogenic cell death through the use of nanoparticles are discussed.
Collapse
Affiliation(s)
- Vincent Lenders
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| | - Xanthippi Koutsoumpou
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| | - Ara Sargsian
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| | - Bella B Manshian
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| |
Collapse
|
49
|
Zhang I, Lépine P, Han C, Lacalle-Aurioles M, Chen CXQ, Haag R, Durcan TM, Maysinger D. Nanotherapeutic Modulation of Human Neural Cells and Glioblastoma in Organoids and Monocultures. Cells 2020; 9:cells9112434. [PMID: 33171886 PMCID: PMC7695149 DOI: 10.3390/cells9112434] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammatory processes in the brain are orchestrated by microglia and astrocytes in response to activators such as pathogen-associated molecular patterns, danger-associated molecular patterns and some nanostructures. Microglia are the primary immune responders in the brain and initiate responses amplified by astrocytes through intercellular signaling. Intercellular communication between neural cells can be studied in cerebral organoids, co-cultures or in vivo. We used human cerebral organoids and glioblastoma co-cultures to study glia modulation by dendritic polyglycerol sulfate (dPGS). dPGS is an extensively studied nanostructure with inherent anti-inflammatory properties. Under inflammatory conditions, lipocalin-2 levels in astrocytes are markedly increased and indirectly enhanced by soluble factors released from hyperactive microglia. dPGS is an effective anti-inflammatory modulator of these markers. Our results show that dPGS can enter neural cells in cerebral organoids and glial cells in monocultures in a time-dependent manner. dPGS markedly reduces lipocalin-2 abundance in the neural cells. Glioblastoma tumoroids of astrocytic origin respond to activated microglia with enhanced invasiveness, whereas conditioned media from dPGS-treated microglia reduce tumoroid invasiveness. Considering that many nanostructures have only been tested in cancer cells and rodent models, experiments in human 3D cerebral organoids and co-cultures are complementary in vitro models to evaluate nanotherapeutics in the pre-clinical setting. Thoroughly characterized organoids and standardized procedures for their preparation are prerequisites to gain information of translational value in nanomedicine. This study provides data for a well-characterized dendrimer (dPGS) that modulates the activation state of human microglia implicated in brain tumor invasiveness.
Collapse
Affiliation(s)
- Issan Zhang
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada;
| | - Paula Lépine
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Chanshuai Han
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - María Lacalle-Aurioles
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Carol X.-Q. Chen
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany;
| | - Thomas M. Durcan
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada;
- Correspondence: ; Tel.: +1-514-398-1264
| |
Collapse
|
50
|
Achazi K, Haag R, Ballauff M, Dernedde J, Kizhakkedathu JN, Maysinger D, Multhaup G. Wechselwirkung von Polyelektrolyt‐Architekturen mit Proteinen und Biosystemen. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202006457] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Katharina Achazi
- Institut für Chemie und Biochemie Freie Universität Berlin Takustraße 3 14195 Berlin Deutschland
| | - Rainer Haag
- Institut für Chemie und Biochemie Freie Universität Berlin Takustraße 3 14195 Berlin Deutschland
| | - Matthias Ballauff
- Institut für Chemie und Biochemie Freie Universität Berlin Takustraße 3 14195 Berlin Deutschland
- IRIS Adlershof Humboldt-Universität zu Berlin Zum Großen Windkanal 6 12489 Berlin Deutschland
| | - Jens Dernedde
- Charité-Universitätsmedizin Berlin Institut für Laboratoriumsmedizin Klinische Chemie und Pathobiochemie CVK Augustenburger Platz 1 13353 Berlin Deutschland
| | - Jayachandran N. Kizhakkedathu
- Centre for Blood Research Department of Pathology and Laboratory Medicine Life Science Institute Department of Chemistry School of Biomedical Engineering University of British Columbia Vancouver V6T 1Z3 Kanada
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics McGill University Montreal H3G 1Y6 Kanada
| | - Gerd Multhaup
- Department of Pharmacology and Therapeutics McGill University Montreal H3G 1Y6 Kanada
| |
Collapse
|