1
|
Dress RJ, Ho WW, Ho V, Lam JH, Décaillot FM, Sinsinbar G, Soo J, Rengasamy G, Khan AK, Cornell TA, Chia TW, Venkataraman S, Nallani M, Ginhoux F. A Novel Polymersome Nanocarrier Promotes Anti-Tumour Immunity by Improved Priming of CD8 + T Cells. Immunology 2025; 175:21-35. [PMID: 39873184 PMCID: PMC11982605 DOI: 10.1111/imm.13903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/30/2025] Open
Abstract
Cancer is one of the leading causes of death worldwide. In recent years, immune checkpoint inhibitor therapies, in addition to standard immuno- or chemotherapy and surgical approaches, have massively improved the outcome for cancer patients. However, these therapies have their limitations and improved strategies, including access to reliable cancer vaccines, are needed. Here, we describe the use of self-assembling artificial cell membrane (ACM) polymersomes to deliver tumour-specific peptides to trigger sustainable and efficient anti-tumour immune responses. We found that ACM polymersomes were highly efficient in targeting and activating mononuclear phagocytes (MNP) including dendritic cells (DC), while providing long-term reservoirs of antigens for continued immune cell priming. Subcutaneous injection of ACM-encapsulated tumour-antigen-peptides into tumour-bearing mice resulted in improved priming of CD8+ T cells and increased generation of tumour-antigen-peptide specific CD8+ effector T cells. Prophylactic and therapeutic immunisation with ACM-encapsulated peptides resulted in changes to the MNP composition in the tumour microenvironment, tumour regression and improved survival of immunised mice. Combining anti-PD-1 immune checkpoint inhibitor therapy with ACM polymersome peptide delivery further boosted anti-tumour immunity. Our results show that ACM polymersome nanocarriers efficiently instruct anti-tumour immune responses offering a promising new approach for vaccination and cancer immunotherapy. Trial Registration: NCT05385991.
Collapse
Affiliation(s)
- Regine J. Dress
- Singapore Immunology Network (SIgN), A*STARSingaporeSingapore
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI)University Medical Center Hamburg‐EppendorfHamburgGermany
| | - William W. Ho
- Singapore Immunology Network (SIgN), A*STARSingaporeSingapore
| | - Victor Ho
- Singapore Immunology Network (SIgN), A*STARSingaporeSingapore
| | | | - Fabien M. Décaillot
- ACM Biolabs Pte LtdSingaporeSingapore
- Sapreme Development B.VBilthovenThe Netherlands
| | | | - Jenetta Soo
- Singapore Immunology Network (SIgN), A*STARSingaporeSingapore
| | | | | | | | | | | | | | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A*STARSingaporeSingapore
- Gustave Roussy Cancer CampusVillejuifFrance
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015Equipe Labellisée—Ligue Nationale Contre le CancerVillejuifFrance
- Shanghai Institute of ImmunologyShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
2
|
Panda S, Eaton EJ, Muralikrishnan P, Stelljes EM, Seelig D, Leyden MC, Gilkey AK, Barnes JT, Morrissey DV, Sarupria S, Moriarity BS, Reineke TM. Machine Learning Reveals Amine Type in Polymer Micelles Determines mRNA Binding, In Vitro, and In Vivo Performance for Lung-Selective Delivery. JACS AU 2025; 5:1845-1861. [PMID: 40313817 PMCID: PMC12041957 DOI: 10.1021/jacsau.5c00084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 05/03/2025]
Abstract
Cationic micelles, composed of amphiphilic block copolymers with polycationic coronas, offer a customizable platform for mRNA delivery. Here, we present a library of 30 cationic micelle nanoparticles (MNPs) formulated from diblock copolymers with reactive poly(pentafluorophenol acrylate) backbones modified with diverse amines. This library systematically varies in nitrogen-based cationic functionalities, exhibiting a spectrum of properties that encompass varied degrees of alkyl substitution (A1-A5), piperazine (A6), oligoamine (A7), guanidinium (A8), and hydroxylation (A9-A10) that vary in side-chain volume, substitution pattern, hydrophilicity, and pK a to assess parameter impact on mRNA delivery. In vitro delivery assays using GFP+ mRNA across multiple cell lines reveal that amine side-chain bulk and chemical structure critically affect performance. Using machine learning analysis via SHapley Additive exPlanations (SHAP) on 180 formulations (3780 experimental measurements), we mapped key relationships between amine chemistry and performance metrics, finding that amine-specific binding efficiency was a major determinant of mRNA delivery efficacy, cell viability, and GFP intensity. Micelles with stronger mRNA binding capabilities (A1 and A7) have higher cellular delivery performance, whereas those with intermediate binding tendencies deliver a higher amount of functional mRNA per cell (A2, A10). This indicates that balancing the binding strength is crucial for performance. Micelles with hydrophobic and bulky pendant groups (A3-A5) tend to induce necrosis during cellular delivery, highlighting the significance of chemical optimization. A7 amphiphile, displaying primary and secondary amine, consistently demonstrates the highest GFP expression across various cell types and in vivo achieves high delivery specificity to lung tissue upon intravenous administration. Moreover, we established a strong correlation between in vitro and in vivo performance using Multitask Gaussian Process models, underscoring the predictive power of in vitro models for anticipating in vivo outcomes. Overall, this innovative study integrates advanced data science with experimental design, demonstrating the pivotal role of chemical amine-dependent optimization for advancing targeted mRNA delivery to the lungs.
Collapse
Affiliation(s)
- Sidharth Panda
- Department
of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ella J. Eaton
- Department
of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department
of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Masonic
Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Center
for Genome Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Praveen Muralikrishnan
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Erin M. Stelljes
- Department
of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Davis Seelig
- Department
of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, Minnesota 55108, United States
| | - Michael C. Leyden
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Alexandria K. Gilkey
- Department
of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jackson T. Barnes
- Department
of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - David V. Morrissey
- Pfizer
Research
and Development, 610
Main Street, Cambridge, Massachusetts 02139, United States
| | - Sapna Sarupria
- Department
of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Branden S. Moriarity
- Department
of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Masonic
Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Center
for Genome Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M. Reineke
- Department
of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
3
|
Vu C, Abu Amara N, Alaboalirat M, Nativ-Roth E, Zalk R, Leite W, Carrillo JM, Bitton R, Matson JB. Aqueous Self-Assembly of Cylindrical and Tapered Bottlebrush Block Copolymers. Angew Chem Int Ed Engl 2025:e202500771. [PMID: 40277335 DOI: 10.1002/anie.202500771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/06/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
The self-assembly of amphiphilic bottlebrush block copolymers (BCPs), featuring backbones densely grafted with two types of side chains, is less well understood compared to linear BCPs. In particular, the solution self-assembly of tapered bottlebrush BCPs-cone-shaped BCPs with hydrophilic or hydrophobic tips-remains unexplored. This study investigates eight tapered and four cylindrical bottlebrush BCPs with varied ratios of hydrophobic polystyrene (PS) and hydrophilic poly(acrylic acid) (PAA) side chains, synthesized via sequential addition of macromonomers using ring-opening metathesis polymerization (SAM-ROMP). Self-assembled nanostructures formed in water were analyzed using cryogenic transmission electron microscopy, small-angle neutron scattering, and dynamic light scattering. Most BCPs generated multiple nanostructures with surface protrusions, including spherical micelles, cylindrical micelles, and vesicles, alongside transitional forms like ellipsoids and semi-vesicles. Coarse-grained molecular dynamics simulations supported the experimental findings, which revealed two distinct self-assembly pathways. The first involved micelle fusion, producing elliptical and cylindrical aggregates, sometimes forming Y-junctions. The second pathway featured micelle maturation into semivesicles, which developed into vesicles or large compound vesicles. This work provides the first experimental evidence of vesicle formation via semivesicles in bottlebrush BCPs and demonstrates the significant influence of cone directionality on self-assembly behavior in these cone-shaped polymeric amphiphiles.
Collapse
Affiliation(s)
- Clark Vu
- Department of Chemistry, Virginia Tech, Blacksburg, VA, 24061, USA
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Narjess Abu Amara
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Einat Nativ-Roth
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ran Zalk
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Wellington Leite
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
| | - Jan-Michael Carrillo
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
| | - Ronit Bitton
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - John B Matson
- Department of Chemistry, Virginia Tech, Blacksburg, VA, 24061, USA
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| |
Collapse
|
4
|
Kenchegowda M, Angolkar M, Hani U, Al Fatease A, Fatima F, Talath S, Dera AA, Paramshetti S, Gangadharappa HV, Osmani RAM, Kazi HS. Polymeric microneedle advancements in macromolecule drug delivery: current trends, challenges, and future perspectives. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04117-8. [PMID: 40244451 DOI: 10.1007/s00210-025-04117-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Microneedles (MNs) offer a transformative solution for delivering macromolecules, including proteins, RNA, and peptides. These are critical in treating complex diseases but face significant challenges such as immunogenicity, poor stability, high molecular weight, and delivery efficiency. Unlike conventional methods, MNs efficiently bypass biological barriers like the stratum corneum, enabling precise and minimally invasive transdermal drug delivery. This review explores various MN types such as solid, coated, hollow, hydrogel-forming, and dissolving and their therapeutic applications in cancer immunotherapy, diabetes management, and osteoporosis treatment. For instance, dissolving MNs have been employed for transdermal insulin delivery, enhancing patient compliance and therapeutic outcomes. Similarly, hydrogel MNs have shown promise in sustained drug release for immunotherapy applications. By addressing cost and scalability issues, polymeric MNs demonstrate significant potential for clinical translation, paving the way for innovations in macromolecule delivery, diagnostics, and personalised medicine. This review underscores the pivotal role of MNs in redefining drug delivery systems, offering improved efficacy, patient comfort, and accessibility.
Collapse
Affiliation(s)
- Madhuchandra Kenchegowda
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, 570015, India
| | - Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, 570015, India
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Guraiger, Abha, 62529, Saudi Arabia
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Guraiger, Abha, 62529, Saudi Arabia
| | - Farhat Fatima
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj, 11942, Saudi Arabia
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, 11172, United Arab Emirates
| | - Ayed A Dera
- Department of Clinical Laboratory Sciences, Central Research Laboratory, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia
| | - Sharanya Paramshetti
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, 570015, India
| | | | - Riyaz Ali M Osmani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Al-Faraa, Abha, 62223, Saudi Arabia.
| | - Heena Shijauddin Kazi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, 570015, India
| |
Collapse
|
5
|
Trumbull K, Fetten S, Arnold N, Marahrens V, Montgomery D, Myers O, Twiss JL, Larsen J. Targeted Polymersomes Enable Enhanced Delivery to Peripheral Nerves Post-Injury. Bioconjug Chem 2025; 36:823-837. [PMID: 40068147 DOI: 10.1021/acs.bioconjchem.5c00072] [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: 04/17/2025]
Abstract
The gold standard therapy for peripheral nerve injuries involves surgical repair, which is invasive and leads to major variations in therapeutic outcomes. Because of this, smaller injuries often go untreated. However, alternative, noninvasive routes of administration are currently unviable due to the presence of the blood-nerve barrier (BNB), which prevents passage of small molecules from the blood into the endoneurium and the nerve. This paper demonstrates that ligands on the surface of nanoparticles, called polymersomes, can enable delivery to the nerve through noninvasive intramuscular injections. Polymersomes made from polyethylene glycol (PEG)-b-polylactic acid (PLA) were conjugated with either apolipoprotein E (ApoE) or rabies virus glycoprotein-based peptide RVG29 (RVG) and loaded with near-infrared dye, AlexaFluor647. ApoE was used to target receptors upregulated in post-injury inflammation, while RVG targets neural-specific receptors. Untagged, ApoE-tagged, and RVG-tagged polymersomes were injected at 100 mM either intranerve (IN) or intramuscular (IM) into Sprague-Dawley rats post sciatic nerve injury. The addition of the ApoE and RVG tags enabled increased AlexaFluor647 fluorescence in the injury site at 1 h post IN injection compared to the untagged polymersome control. However, only the RVG-tagged polymersomes increased the AlexaFluor647 fluorescence after IM injection. Ex vivo analysis of sciatic nerves demonstrated that ApoE-tagged polymersomes enabled the greatest retention of AlexaFluor647 regardless of the injection route. This led us to conclude that using ApoE to target inflammation enabled the greatest retention of polymersome-delivered payloads while using RVG to target neural cells more specifically enabled the penetration of polymersome-delivered payloads. Observations were confirmed by calculating the area under the curve pharmacokinetic parameters and the use of a two-compartment pharmacokinetic model.
Collapse
Affiliation(s)
- Kayleigh Trumbull
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Sophia Fetten
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, United States
| | - Noah Arnold
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Vanessa Marahrens
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Dru Montgomery
- Department of Biochemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Olivia Myers
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Jeffery L Twiss
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Jessica Larsen
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
- Department of Bioengineering, Clemson University, Clemson, South Carolina 29634, United States
| |
Collapse
|
6
|
Wang Y, Ming Y, Yu Z, Xu Z, Zou M, Chen C, Luo F, Huang D, Wang N, Lin Z, Weng Z. ROS-Responsive Cationic Polymers with Intrinsic Anti-Inflammatory Activity for Intracellular Protein Delivery. Biomacromolecules 2025; 26:2268-2281. [PMID: 40114391 DOI: 10.1021/acs.biomac.4c01593] [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: 03/22/2025]
Abstract
The intracellular delivery of protein drugs via nanocarriers offers significant potential for expanding their therapeutic applications. However, the unintended activation of innate immune responses and inflammation triggered by the carriers presents a major challenge, often compromising therapeutic efficacy. Here, we present oligoethylenimine-thioketal (OEI-TK), a reactive oxygen species-responsive cationic polymer with intrinsic anti-inflammatory properties, to overcome this challenge. OEI-TK self-assembles electrostatically with bovine serum albumin (BSA) to form stable nanoparticles (OTB NPs) with excellent encapsulation efficiency. In vitro studies confirmed that OTB NPs retained OEI-TK's antioxidant and anti-inflammatory properties, enhanced biocompatibility, and efficiently delivered BSA into cells. Furthermore, OEI-TK facilitated the intracellular delivery of β-galactosidase while preserving its enzymatic activity, demonstrating its potential for functional protein transport. These findings highlight OEI-TK as a promising platform with dual benefits of inflammation modulation and intracellular protein delivery, holding potential for the synergistic treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Yongming Wang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yangcan Ming
- Department of Pediatrics, Wuhan NO.1 Hospital, Wuhan, Hubei 430022, China
| | - Zhichao Yu
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Zhenjin Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Minglang Zou
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Cuiping Chen
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Fang Luo
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Da Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Na Wang
- Department of Pediatrics, Wuhan NO.1 Hospital, Wuhan, Hubei 430022, China
| | - Zhenyu Lin
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Zuquan Weng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| |
Collapse
|
7
|
Laturski AE, Dulay MT, Perry JL, DeSimone JM. Transfection via RNA-Based Nanoparticles: Comparing Encapsulation vs Adsorption Approaches of RNA Incorporation. Bioconjug Chem 2025; 36:367-376. [PMID: 39999074 DOI: 10.1021/acs.bioconjchem.5c00028] [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: 02/27/2025]
Abstract
Historically, RNA delivery via nanoparticles has primarily relied on encapsulation, as demonstrated by lipid nanoparticles in SARS-CoV-2 vaccines. Concerns about RNA degradation on nanoparticle surfaces initially limited the exploration of adsorption-based approaches. However, recent advancements have renewed interest in adsorption as a viable alternative. This Viewpoint explores the approaches of RNA incorporation in nanoparticles, comparing encapsulation, adsorption, and the combination of encapsulation and adsorption, and presents a framework to guide the selection of the most suitable strategy based on general characteristics.
Collapse
Affiliation(s)
- Amy E Laturski
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Maria T Dulay
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Jillian L Perry
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7575, United States
| | - Joseph M DeSimone
- Department of Chemical Engineering and Department of Radiology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
8
|
Gupta J, Sharma G. Nanogel: A versatile drug delivery system for the treatment of various diseases and their future perspective. Drug Deliv Transl Res 2025; 15:455-482. [PMID: 39103593 DOI: 10.1007/s13346-024-01684-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
Nanogel (NG) drug delivery systems have emerged as promising tools for targeted and controlled drug release, revolutionizing treatment approaches across various diseases. Their unique physicochemical properties, such as nano size, high surface area, biocompatibility, stability, and tunable drug release, make them ideal carriers for a wide range of therapeutic agents. Nanogels (NGs), characterized by their 3D network of crosslinked polymers, offer unique edges like high drug loading capacity, controlled release, and targeted delivery. Additionally, the diverse applications of NGs in medical therapeutics highlight their versatility and potential impact on improving patient outcomes. Their application spans cancer treatment, infectious diseases, and chronic conditions, allowing for precise drug delivery to specific tissues or cells, minimizing side effects, and enhancing therapeutic efficacy. Despite their potential, challenges such as scalability, manufacturing reproducibility, and regulatory hurdles must be addressed. Achieving clinical translation requires overcoming these obstacles to ensure therapeutic payloads' safe and efficient delivery. Strategies such as surface modification and incorporating stimuli-responsive elements enhanced NG performance and addressed specific therapeutic challenges. Advances in nanotechnology, biomaterials, and targeted drug design offer opportunities to improve the performance of NGs and address current limitations. Tailoring NGs for exploring combination therapies and integrating diagnostics for real-time monitoring represent promising avenues for future research. In conclusion, NG drug delivery systems have demonstrated tremendous potential in diverse disease applications. Overcoming challenges and leveraging emerging technologies will pave the way for their widespread clinical implementation, ushering in a new era of precision medicine and improved patient care.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India.
| | - Gaurang Sharma
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India
| |
Collapse
|
9
|
Jony MJ, Joshi A, Dash A, Shukla S. Non-Viral Delivery Systems to Transport Nucleic Acids for Inherited Retinal Disorders. Pharmaceuticals (Basel) 2025; 18:87. [PMID: 39861150 PMCID: PMC11768406 DOI: 10.3390/ph18010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Inherited retinal disorders (IRDs) represent a group of challenging genetic conditions that often lead to severe visual impairment or blindness. The complexity of these disorders, arising from their diverse genetic causes and the unique structural and functional aspects of retinal cells, has made developing effective treatments particularly challenging. Recent advancements in gene therapy, especially non-viral nucleic acid delivery systems like liposomes, solid lipid nanoparticles, dendrimers, and polymersomes, offer promising solutions. These systems provide advantages over viral vectors, including reduced immunogenicity and enhanced targeting capabilities. This review delves into introduction of common IRDs such as Leber congenital amaurosis, retinitis pigmentosa, Usher syndrome, macular dystrophies, and choroideremia and critically assesses current treatments including neuroprotective agents, cellular therapy, and gene therapy along with their limitations. The focus is on the emerging role of non-viral delivery systems, which promise to address the current limitations of specificity, untoward effects, and immunogenicity in existing gene therapies. Additionally, this review covers recent clinical trial developments in gene therapy for retinal disorders.
Collapse
Affiliation(s)
- Md Jobair Jony
- Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University, Omaha, NE 68178, USA
| | - Ameya Joshi
- Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University, Omaha, NE 68178, USA
| | - Alekha Dash
- Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University, Omaha, NE 68178, USA
| | - Surabhi Shukla
- Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University, Omaha, NE 68178, USA
| |
Collapse
|
10
|
Sun H, Zhong Z. Bioresponsive Polymeric Nanoparticles: From Design, Targeted Therapy to Cancer Immunotherapy. Biomacromolecules 2025; 26:33-42. [PMID: 39667037 DOI: 10.1021/acs.biomac.4c01257] [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: 12/14/2024]
Abstract
Bioresponsive polymeric nanoparticles (NPs) that are capable of delivering and releasing therapeutics and biotherapeutics to target sites have attracted vivid interest in cancer therapy and immunotherapy. In contrast to enthusiastic evolution in the academic world, the clinical translation of these smart systems is scarce, partly due to concerns about safety, stability, complexity, and scalability. The moderate targetability, responsivity, and benefits are other concerns. In the past 17 years, we have devoted ourselves to exploring elegant strategies to address the above basic and translational problems by introducing diverse functional groups and/or targeting ligands to safe biomedical materials, such as biodegradable polymers and water-soluble polymers. This minimal modification is critical for further clinical translation. We have tailor-made various bioresponsive NPs including shell-sheddable and/or acid-sensitive biodegradable NPs, disulfide-cross-linked biodegradable micelles and polymersomes, and blood-brain barrier (BBB)-permeable NPs, to target different tumors. This perspective provides an overview of our work path toward targeted nanomedicines and personalized vaccines, which might inspire clinical translation and future research on cancer therapy.
Collapse
Affiliation(s)
- Huanli Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - 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, PR China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, 215222, PR China
| |
Collapse
|
11
|
Sanati M, Figueroa-Espada CG, Han EL, Mitchell MJ, Yavari SA. Bioengineered Nanomaterials for siRNA Therapy of Chemoresistant Cancers. ACS NANO 2024; 18:34425-34463. [PMID: 39666006 DOI: 10.1021/acsnano.4c11259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Chemoresistance remains a long-standing challenge after cancer treatment. Over the last two decades, RNA interference (RNAi) has emerged as a gene therapy modality to sensitize cancer cells to chemotherapy. However, the use of RNAi, specifically small-interfering RNA (siRNA), is hindered by biological barriers that limit its intracellular delivery. Nanoparticles can overcome these barriers by protecting siRNA in physiological environments and facilitating its delivery to cancer cells. In this review, we discuss the development of nanomaterials for siRNA delivery in cancer therapy, current challenges, and future perspectives for their implementation to overcome cancer chemoresistance.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand 97178, Iran
| | - Christian G Figueroa-Espada
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Emily L Han
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
| |
Collapse
|
12
|
Kreofsky NW, Roy P, Reineke TM. pH-Responsive Micelles Containing Quinine Functionalities Enhance Intracellular Gene Delivery and Expression. Bioconjug Chem 2024; 35:1762-1778. [PMID: 39467734 DOI: 10.1021/acs.bioconjchem.4c00326] [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: 10/30/2024]
Abstract
Quinine is a promising building block for creating polymer carriers for intracellular nucleic acid delivery. This is due to its ability to bind to genetic material through intercalation and electrostatic interactions and the balance of hydrophobicity and hydrophilicity dependent on the pH/charge state. Yet, studies utilizing cinchona alkaloid natural products in gene delivery are limited. Herein, we present the incorporation of a quinine functionalized monomer (Q) into block polymer architectures to form self-assembled micelles for highly efficient gene delivery. Q was incorporated into the core and/or the shell of the micelles to introduce the unique advantages of quinine to the system. We found that incorporation of Q into the core of the micelle resulted in acid-induced disassembly of the micelle and a boost in transfection efficiency by promoting endosomal escape. This effect was especially evident in the cancerous cell line, A549, which has a more acidic intracellular environment. Incorporation of Q into the shell of the micelles resulted in intercalative binding to the genetic payload as well as larger micelle-DNA complexes (micelleplexes) from the hydrophobicity of Q in the shell. These factors enable the micelleplexes to be more resistant to serum and have more persistent protein expression post-transfection. Overall, this study is the first to demonstrate the benefits of including quinine functionalities into self-assembled micelles for highly efficient gene delivery and presents a platform for inclusion of other natural products with similar properties into micellar systems.
Collapse
Affiliation(s)
- Nicholas W Kreofsky
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Punarbasu Roy
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
13
|
Ma J, Du Z, Gao S, Zang J. Tea polyphenols-mediated supramolecular architectures: Design and applications. Trends Food Sci Technol 2024; 152:104665. [DOI: 10.1016/j.tifs.2024.104665] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Bouazzaoui A, Abdellatif AA. Vaccine delivery systems and administration routes: Advanced biotechnological techniques to improve the immunization efficacy. Vaccine X 2024; 19:100500. [PMID: 38873639 PMCID: PMC11170481 DOI: 10.1016/j.jvacx.2024.100500] [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: 01/03/2023] [Revised: 03/21/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
Since the first use of vaccine tell the last COVID-19 pandemic caused by spread of SARS-CoV-2 worldwide, the use of advanced biotechnological techniques has accelerated the development of different types and methods for immunization. The last pandemic showed that the nucleic acid-based vaccine, especially mRNA, has an advantage in terms of development time; however, it showed a very critical drawback namely, the higher costs when compared to other strategies, and its inability to protect against new variants. This showed the need of more improvement to reach a better delivery and efficacy. In this review we will describe different vaccine delivery systems including, the most used viral vector, and also variable strategies for delivering of nucleic acid-based vaccines especially lipid-based nanoparticles formulation, polymersomes, electroporation and also the new powerful tools for the delivery of mRNA, which is based on the use of cell-penetrating peptides (CPPs). Additionally, we will also discuss the main challenges associated with each system. Finlay, the efficacy and safety of the vaccines depends not only on the formulations and delivery systems, but also the dosage and route of administration are also important players, therefore we will see the different routes for the vaccine administration including traditionally routes (intramuscular, Transdermal, subcutaneous), oral inhalation or via nasal mucosa, and will describe the advantages and disadvantage of each administration route.
Collapse
Affiliation(s)
- Abdellatif Bouazzaoui
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
- Science and Technology Unit, Umm Al Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
| | - Ahmed A.H. Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, 51452 Qassim, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, 71524 Assiut, Egypt
| |
Collapse
|
15
|
Tang Q, Tan Y, Leng S, Liu Q, Zhu L, Wang C. Cupric-polymeric nanoreactors integrate into copper metabolism to promote chronic diabetic wounds healing. Mater Today Bio 2024; 26:101087. [PMID: 38784443 PMCID: PMC11111831 DOI: 10.1016/j.mtbio.2024.101087] [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: 02/13/2024] [Revised: 04/13/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Given multifunction of copper (Cu) contributing to all stages of the physiology of wound healing, Cu-based compounds have great therapeutic potentials to accelerate the wound healing, but they must be limited to a very low concentration range to avoid detrimental accumulation. Additionally, the cellular mechanism of Cu-based compounds participating the healing process remains elusive. In this study, copper oxide nanoparticles (CuONPs) were synthesized to mimic the multiple natural enzymes and trapped into PEG-b-PCL polymersomes (PS) to construct cupric-polymeric nanoreactors (CuO@PS) via a direct hydration method, thus allowing to compartmentalize Cu-based catalytic reactions in an isolated space to improve the efficiency, selectivity, recyclability as well as biocompatibility. While nanoreactors trafficked to lysosomes following endocytosis, the released Cu-based compounds in lysosomal lumen drove a cytosolic Cu+ influx to mobilize Cu metabolism mostly via Atox1-ATP7a/b-Lox axis, thereby activating the phosphorylation of mitogen-activated protein kinase 1 and 2 (MEK1/2) to initiate downstream signaling events associated with cell proliferation, migration and angiogenesis. Moreover, to facilitate to lay on wounds, cupric-polymeric nanoreactors were finely dispersed into a thermosensitive Pluronic F127 hydrogel to form a composite hydrogel sheet that promoted the healing of chronic wounds in diabetic rat models. Hence, cupric-polymeric nanoreactors represented an attractive translational strategy to harness cellular Cu metabolism for chronic wounds healing.
Collapse
Affiliation(s)
- Qi Tang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yinqiu Tan
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Shaolong Leng
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Qi Liu
- The First Dongguan Affiliated Hospital Guangdong Medical University No. 42, Jiaoping Road Dongguan, Guangdong, 523710, China
| | - Linyu Zhu
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Cuifeng Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Department of Neurosurgery, JiuJiang Hospital of Traditional Chinese Medicine, Jiujiang, China
| |
Collapse
|
16
|
Çalbaş B, Keobounnam AN, Korban C, Doratan AJ, Jean T, Sharma AY, Wright TA. Protein-polymer bioconjugation, immobilization, and encapsulation: a comparative review towards applicability, functionality, activity, and stability. Biomater Sci 2024; 12:2841-2864. [PMID: 38683585 DOI: 10.1039/d3bm01861j] [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: 05/01/2024]
Abstract
Polymer-based biomaterials have received a lot of attention due to their biomedical, agricultural, and industrial potential. Soluble protein-polymer bioconjugates, immobilized proteins, and encapsulated proteins have been shown to tune enzymatic activity, improved pharmacokinetic ability, increased chemical and thermal stability, stimuli responsiveness, and introduced protein recovery. Controlled polymerization techniques, increased protein-polymer attachment techniques, improved polymer surface grafting techniques, controlled polymersome self-assembly, and sophisticated characterization methods have been utilized for the development of well-defined polymer-based biomaterials. In this review we aim to provide a brief account of the field, compare these methods for engineering biomaterials, provide future directions for the field, and highlight impacts of these forms of bioconjugation.
Collapse
Affiliation(s)
- Berke Çalbaş
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Ashley N Keobounnam
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Christopher Korban
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ainsley Jade Doratan
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Tiffany Jean
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Aryan Yashvardhan Sharma
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Thaiesha A Wright
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Li L, Ma T, Wang M. Protein-Integrated Hydrogen-Bonded Organic Frameworks: Chemistry and Biomedical Applications. Angew Chem Int Ed Engl 2024; 63:e202400926. [PMID: 38529812 DOI: 10.1002/anie.202400926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 03/27/2024]
Abstract
Hydrogen-bonded organic frameworks (HOFs) are porous nanomaterials that offer exceptional biocompatibility and versatility for integrating proteins for biomedical applications. This minireview concisely discusses recent advancements in the chemistry and functionality of protein-HOF interfaces. It particularly focuses on strategic methodologies, such as the careful selection of building blocks and the genetic engineering of proteins, to facilitate protein-HOF interactions. We examine the role of enzyme encapsulation within HOFs, highlighting its capability to preserve enzyme function, a crucial aspect for applications in biosensing and disease diagnosis. Moreover, we discuss the emerging utility of nanoscale HOFs for intracellular protein delivery, illustrating their applicability as nanoreactors for intracellular catalysis and neuroprotective biorthogonal catalysis within cellular compartments. We highlight the significant advancement of designing biodegradable HOFs tailored for cytosolic protein delivery, underscoring their promising application in targeted cancer therapies. Finally, we provide a perspective viewpoint on the design of biocompatible protein-HOF assemblies, underlining their promising prospects in drug delivery, disease diagnosis, and broader biomedical applications.
Collapse
Affiliation(s)
- Lijuan Li
- Beijing National Laboratory for Molecular Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianyu Ma
- Beijing National Laboratory for Molecular Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ming Wang
- Beijing National Laboratory for Molecular Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
18
|
Xu Q, Wang Y, Zheng Y, Zhu Y, Li Z, Liu Y, Ding M. Polymersomes in Drug Delivery─From Experiment to Computational Modeling. Biomacromolecules 2024; 25:2114-2135. [PMID: 38011222 DOI: 10.1021/acs.biomac.3c00903] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Polymersomes, composed of amphiphilic block copolymers, are self-assembled vesicles that have gained attention as potential drug delivery systems due to their good biocompatibility, stability, and versatility. Various experimental techniques have been employed to characterize the self-assembly behaviors and properties of polymersomes. However, they have limitations in revealing molecular details and underlying mechanisms. Computational modeling techniques have emerged as powerful tools to complement experimental studies and enabled researchers to examine drug delivery mechanisms at molecular resolution. This review aims to provide a comprehensive overview of the state of the art in the field of polymersome-based drug delivery systems, with an emphasis on insights gained from both experimental and computational studies. Specifically, we focus on polymersome morphologies, self-assembly kinetics, fusion and fission, behaviors in flow, as well as drug encapsulation and release mechanisms. Furthermore, we also identify existing challenges and limitations in this rapidly evolving field and suggest possible directions for future research.
Collapse
Affiliation(s)
- Qianru Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yiwei Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yi Zheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yuling Zhu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Zifen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yang Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Mingming Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| |
Collapse
|
19
|
Adachi T, Tahara Y, Yamamoto K, Yamamoto T, Kanamura N, Akiyoshi K, Mazda O. Cholesterol-Bearing Polysaccharide-Based Nanogels for Development of Novel Immunotherapy and Regenerative Medicine. Gels 2024; 10:206. [PMID: 38534624 DOI: 10.3390/gels10030206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024] Open
Abstract
Novel functional biomaterials are expected to bring about breakthroughs in developing immunotherapy and regenerative medicine through their application as drug delivery systems and scaffolds. Nanogels are defined as nanoparticles with a particle size of 100 nm or less and as having a gel structure. Nanogels have a three-dimensional network structure of cross-linked polymer chains, which have a high water content, a volume phase transition much faster than that of a macrogel, and a quick response to external stimuli. As it is possible to transmit substances according to the three-dimensional mesh size of the gel, a major feature is that relatively large substances, such as proteins and nucleic acids, can be taken into the gel. Furthermore, by organizing nanogels as a building block, they can be applied as a scaffold material for tissue regeneration. This review provides a brief overview of the current developments in nanogels in general, especially drug delivery, therapeutic applications, and tissue engineering. In particular, polysaccharide-based nanogels are interesting because they have excellent complexation properties and are highly biocompatible.
Collapse
Affiliation(s)
- Tetsuya Adachi
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Yoshiro Tahara
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara Miyakodani, Kyoto-fu, Kyotanabe-shi 610-0321, Japan
| | - Kenta Yamamoto
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| |
Collapse
|
20
|
Hua C, Qiu L. Polymersomes for Therapeutic Protein and Peptide Delivery: Towards Better Loading Properties. Int J Nanomedicine 2024; 19:2317-2340. [PMID: 38476284 PMCID: PMC10929215 DOI: 10.2147/ijn.s444910] [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: 10/16/2023] [Accepted: 01/24/2024] [Indexed: 03/14/2024] Open
Abstract
Therapeutics based on proteins and peptides have profoundly transformed the landscape of treatment for diseases, from diabetes mellitus to cancers, yet the short half-life and low bioavailability of therapeutic proteins and peptides hinder their wide applications. To break through this bottleneck, biomolecules-loaded polymersomes with strong adjustability and versatility have attracted more and more attentions recently. Loading proteins or peptides into polymersomes is the first but extremely important step towards developing high-quality formulation products. However, increasing protein and peptide loading content is quite challenging due to the inherent nature of self-assembled vesicle formation mechanism and physiochemical characteristics of biomacromolecules. This review highlights the potential of polymersomes as the next-generation therapeutic proteins and peptides carrier and emphatically introduces novel approaches and recent progress to achieve satisfactory encapsulation capability of polymersomes for proteins and peptides. On the one hand, with the help of intermolecular interactions, such as electrostatic, lipid-protein, and hydrophobic interactions, the drug loading could be significantly improved. On the other hand, loading improvement could be attained through innovation of preparation methods, ranging from modified traditional film hydration techniques to the novel phase-guided assembly method.
Collapse
Affiliation(s)
- Chengxu Hua
- Ministry of Educational (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, People’s Republic of China
| | - Liyan Qiu
- Ministry of Educational (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, People’s Republic of China
| |
Collapse
|
21
|
Slezak A, Chang K, Hossainy S, Mansurov A, Rowan SJ, Hubbell JA, Guler MO. Therapeutic synthetic and natural materials for immunoengineering. Chem Soc Rev 2024; 53:1789-1822. [PMID: 38170619 PMCID: PMC11557218 DOI: 10.1039/d3cs00805c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Immunoengineering is a rapidly evolving field that has been driving innovations in manipulating immune system for new treatment tools and methods. The need for materials for immunoengineering applications has gained significant attention in recent years due to the growing demand for effective therapies that can target and regulate the immune system. Biologics and biomaterials are emerging as promising tools for controlling immune responses, and a wide variety of materials, including proteins, polymers, nanoparticles, and hydrogels, are being developed for this purpose. In this review article, we explore the different types of materials used in immunoengineering applications, their properties and design principles, and highlight the latest therapeutic materials advancements. Recent works in adjuvants, vaccines, immune tolerance, immunotherapy, and tissue models for immunoengineering studies are discussed.
Collapse
Affiliation(s)
- Anna Slezak
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Kevin Chang
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Samir Hossainy
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Aslan Mansurov
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Stuart J Rowan
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Jeffrey A Hubbell
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Mustafa O Guler
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
22
|
Sinsinbar G, Bindra AK, Liu S, Chia TW, Yoong Eng EC, Loo SY, Lam JH, Schultheis K, Nallani M. Amphiphilic Block Copolymer Nanostructures as a Tunable Delivery Platform: Perspective and Framework for the Future Drug Product Development. Biomacromolecules 2024; 25:541-563. [PMID: 38240244 DOI: 10.1021/acs.biomac.3c00858] [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: 02/13/2024]
Abstract
Nanoformulation of active payloads or pharmaceutical ingredients (APIs) has always been an area of interest to achieve targeted, sustained, and efficacious delivery. Various delivery platforms have been explored, but loading and delivery of APIs have been challenging because of the chemical and structural properties of these molecules. Polymersomes made from amphiphilic block copolymers (ABCPs) have shown enormous promise as a tunable API delivery platform and confer multifold advantages over lipid-based systems. For example, a COVID booster vaccine comprising polymersomes encapsulating spike protein (ACM-001) has recently completed a Phase I clinical trial and provides a case for developing safe drug products based on ABCP delivery platforms. However, several limitations need to be resolved before they can reach their full potential. In this Perspective, we would like to highlight such aspects requiring further development for translating an ABCP-based delivery platform from a proof of concept to a viable commercial product.
Collapse
Affiliation(s)
- Gaurav Sinsinbar
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Anivind Kaur Bindra
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Shaoqiong Liu
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Teck Wan Chia
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Eunice Chia Yoong Eng
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Ser Yue Loo
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Jian Hang Lam
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Katherine Schultheis
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Madhavan Nallani
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| |
Collapse
|
23
|
Edmans JG, Harrison S, Hatton PV, Murdoch C, Spain SG, Colley HE. Electrospinning polymersomes into bead-on-string polyethylene oxide fibres for the delivery of biopharmaceuticals to mucosal epithelia. BIOMATERIALS ADVANCES 2024; 157:213734. [PMID: 38109830 DOI: 10.1016/j.bioadv.2023.213734] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023]
Abstract
Fibrous mucoadhesive polymer membranes prepared using electrospinning demonstrate many advantages for mucosal drug delivery compared to other formulations. Previous electrospun membrane formulations have been developed mainly for the delivery of small molecule drugs. There remains great potential to further develop the technology for the delivery of vesicular vectors that allow administration of advanced therapeutic agents. However, there are no previous reports demonstrating the release of intact drug delivery vesicles from electrospun materials. Here, we describe incorporation and release of protein-loaded polymersomes from polyethylene oxide (PEO)-based electrospun membranes. Polymersomes comprising a copolymer of glycerol monomethacrylate (GMA) and hydroxypropyl methacrylate (HPMA) were prepared using polymerization-induced self-assembly and incorporated within PEO membranes using bead-on-string electrospinning at approximately 40 % w/w by polymer mass. Super-resolution fluorescence imaging showed that the vesicles remained intact and retained their encapsulated protein load within the fibre beads. Transmission electron microscopy and dynamic light scattering demonstrated that polymersomes retained their morphology following release from the polymer fibres. F(ab) antibody fragments were encapsulated within polymersomes and then electrospun into membranes. 78 ± 13 % of the F(ab) remained encapsulated within polymersomes during electrospinning and retained functionality when released from electrospun membranes, demonstrating that the formulation is suitable for the delivery of biologics. Membranes were non-irritant to the oral epithelium and fluorescence microscopy detected accumulation of polymersomes within the epithelia following application. This innovative drug delivery approach represents a novel and potentially highly useful method for the administration of large molecular mass therapeutic molecules to diseased mucosal sites.
Collapse
Affiliation(s)
- Jake G Edmans
- School of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield S10 2TA, United Kingdom; Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, United Kingdom
| | - Samuel Harrison
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, United Kingdom
| | - Paul V Hatton
- School of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield S10 2TA, United Kingdom
| | - Craig Murdoch
- School of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield S10 2TA, United Kingdom.
| | - Sebastian G Spain
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, United Kingdom
| | - Helen E Colley
- School of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield S10 2TA, United Kingdom
| |
Collapse
|
24
|
Foster D, Cakley A, Larsen J. Optimizing enzyme-responsive polymersomes for protein-based therapies. Nanomedicine (Lond) 2024; 19:213-229. [PMID: 38271081 DOI: 10.2217/nnm-2023-0300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Abstract
Aims: Stimuli-responsive polymersomes are promising tools for protein-based therapies, but require deeper understanding and optimization of their pathology-responsive behavior. Materials & methods: Hyaluronic acid (HA)-poly(b-lactic acid) (PLA) polymersomes self-assembled from block copolymers of varying molecular weights of HA were compared for their physical properties, degradation and intracellular behavior. Results: Major results showed increasing enzyme-responsivity associated with decreasing molecular weight. The major formulation differences were as follows: the HA(5 kDa)-PLA formulation exhibited the most pronounced release of encapsulated proteins, while the HA(7 kDa)-PLA formulation showed the most different release behavior from neutral. Conclusion: We have discovered design rules for HA-PLA polymersomes for protein delivery, with lower molecular weight leading to higher encapsulation efficiency, greater release and greater intracellular uptake.
Collapse
Affiliation(s)
- Dorian Foster
- Department of Chemical & Biomolecular Engineering, Center for Nanotherapeutic Strategies in the Central Nervous System, Clemson University, Clemson, SC 29631, USA
| | - Alaura Cakley
- Department of Chemical & Biomolecular Engineering, Center for Nanotherapeutic Strategies in the Central Nervous System, Clemson University, Clemson, SC 29631, USA
| | - Jessica Larsen
- Department of Chemical & Biomolecular Engineering, Center for Nanotherapeutic Strategies in the Central Nervous System, Clemson University, Clemson, SC 29631, USA
- Department of Bioengineering, Center for Nanotherapeutic Strategies in the Central Nervous System, Clemson University, Clemson, SC 29631, USA
| |
Collapse
|
25
|
Fonseca M, Jarak I, Victor F, Domingues C, Veiga F, Figueiras A. Polymersomes as the Next Attractive Generation of Drug Delivery Systems: Definition, Synthesis and Applications. MATERIALS (BASEL, SWITZERLAND) 2024; 17:319. [PMID: 38255485 PMCID: PMC10817611 DOI: 10.3390/ma17020319] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/23/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024]
Abstract
Polymersomes are artificial nanoparticles formed by the self-assembly process of amphiphilic block copolymers composed of hydrophobic and hydrophilic blocks. They can encapsulate hydrophilic molecules in the aqueous core and hydrophobic molecules within the membrane. The composition of block copolymers can be tuned, enabling control of characteristics and properties of formed polymersomes and, thus, their application in areas such as drug delivery, diagnostics, or bioimaging. The preparation methods of polymersomes can also impact their characteristics and the preservation of the encapsulated drugs. Many methods have been described, including direct hydration, thin film hydration, electroporation, the pH-switch method, solvent shift method, single and double emulsion method, flash nanoprecipitation, and microfluidic synthesis. Considering polymersome structure and composition, there are several types of polymersomes including theranostic polymersomes, polymersomes decorated with targeting ligands for selective delivery, stimuli-responsive polymersomes, or porous polymersomes with multiple promising applications. Due to the shortcomings related to the stability, efficacy, and safety of some therapeutics in the human body, polymersomes as drug delivery systems have been good candidates to improve the quality of therapies against a wide range of diseases, including cancer. Chemotherapy and immunotherapy can be improved by using polymersomes to deliver the drugs, protecting and directing them to the exact site of action. Moreover, this approach is also promising for targeted delivery of biologics since they represent a class of drugs with poor stability and high susceptibility to in vivo clearance. However, the lack of a well-defined regulatory plan for polymersome formulations has hampered their follow-up to clinical trials and subsequent market entry.
Collapse
Affiliation(s)
- Mariana Fonseca
- Univ. Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (M.F.); (I.J.); (C.D.); (F.V.)
| | - Ivana Jarak
- Univ. Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (M.F.); (I.J.); (C.D.); (F.V.)
- Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Francis Victor
- Department of Pharmacy, University Chenab Gujarat, Punjab 50700, Pakistan;
| | - Cátia Domingues
- Univ. Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (M.F.); (I.J.); (C.D.); (F.V.)
- Univ. Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, 3000-548 Coimbra, Portugal
- Univ. Coimbra, Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Univ. Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (M.F.); (I.J.); (C.D.); (F.V.)
- Univ. Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, 3000-548 Coimbra, Portugal
| | - Ana Figueiras
- Univ. Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (M.F.); (I.J.); (C.D.); (F.V.)
- Univ. Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, 3000-548 Coimbra, Portugal
| |
Collapse
|
26
|
Balafouti A, Forys A, Trzebicka B, Gerardos AM, Pispas S. Anionic Hyperbranched Amphiphilic Polyelectrolytes as Nanocarriers for Antimicrobial Proteins and Peptides. MATERIALS (BASEL, SWITZERLAND) 2023; 16:7702. [PMID: 38138846 PMCID: PMC10745097 DOI: 10.3390/ma16247702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
This manuscript presents the synthesis of hyperbranched amphiphilic poly (lauryl methacrylate-co-tert-butyl methacrylate-co-methacrylic acid), H-P(LMA-co-tBMA-co-MAA) copolymers via reversible addition fragmentation chain transfer (RAFT) copolymerization of tBMA and LMA, and their post-polymerization modification to anionic amphiphilic polyelectrolytes. The focus is on investigating whether the combination of the hydrophobic characters of LMA and tBMA segments, as well as the polyelectrolyte and hydrophilic properties of MAA segments, both distributed within a unique hyperbranched polymer chain topology, would result in intriguing, branched copolymers with the potential to be applied in nanomedicine. Therefore, we studied the self-assembly behavior of these copolymers in aqueous media, as well as their ability to form complexes with cationic proteins, namely lysozyme (LYZ) and polymyxin (PMX). Various physicochemical characterization techniques, including size exclusion chromatography (SEC) and proton nuclear magnetic resonance (1H-NMR), verified the molecular characteristics of these well-defined copolymers, whereas light scattering and fluorescence spectroscopy techniques revealed promising nanoparticle (NP) self- and co-assembly properties of the copolymers in aqueous media.
Collapse
Affiliation(s)
- Anastasia Balafouti
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., 11635 Athens, Greece; (A.B.); (A.M.G.)
- Department of Chemistry, National and Kapodistrian University of Athens (NKUA), 15784 Athens, Greece
| | - Aleksander Forys
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34 ul. M. Curie-Skłodowskiej, 41-819 Zabrze, Poland; (A.F.); (B.T.)
| | - Barbara Trzebicka
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34 ul. M. Curie-Skłodowskiej, 41-819 Zabrze, Poland; (A.F.); (B.T.)
| | - Angelica Maria Gerardos
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., 11635 Athens, Greece; (A.B.); (A.M.G.)
- Department of Chemistry, National and Kapodistrian University of Athens (NKUA), 15784 Athens, Greece
| | - Stergios Pispas
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., 11635 Athens, Greece; (A.B.); (A.M.G.)
| |
Collapse
|
27
|
Klajnert-Maculewicz B, Janaszewska A, Majecka A. Dendrimersomes: Biomedical applications. Chem Commun (Camb) 2023; 59:14611-14625. [PMID: 37999927 DOI: 10.1039/d3cc03182a] [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: 11/25/2023]
Abstract
In recent years, dendrimer-based vesicles, known as dendrimersomes, have garnered significant attention as highly promising alternatives to lipid vesicles in a variety of biomedical applications. Dendrimersomes offer several advantages, including relatively straightforward synthesis, non-immunogenic properties, stability in circulation, and minimal size variability. These vesicles are composed of Janus dendrimers, which are polymers characterized by two dendritic wedges with different terminal groups - hydrophilic and hydrophobic. This dendrimer structure enables the self-assembly of dendrimersomes. The purpose of this highlight is to provide an overview of recent advancements achieved through the utilization of biomimetic dendrimersomes in various biomedical applications such as drug and nucleic acid delivery.
Collapse
Affiliation(s)
- Barbara Klajnert-Maculewicz
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biophysics, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Anna Janaszewska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biophysics, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Agata Majecka
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biophysics, Pomorska 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
28
|
Gao Y, Gao C, Fan Y, Sun H, Du J. Physically and Chemically Compartmentalized Polymersomes for Programmed Delivery and Biological Applications. Biomacromolecules 2023; 24:5511-5538. [PMID: 37933444 DOI: 10.1021/acs.biomac.3c00826] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Multicompartment polymersomes (MCPs) refer to polymersomes that not only contain one single compartment, either in the membrane or in the internal cavity, but also mimic the compartmentalized structure of living cells, attracting much attention in programmed delivery and biological applications. The investigation of MCPs may promote the application of soft nanomaterials in biomedicine. This Review seeks to highlight the recent advances of the design principles, synthetic strategies, and biomedical applications of MCPs. The compartmentalization types including chemical, physical, and hybrid compartmentalization are discussed. Subsequently, the design and controlled synthesis of MCPs by the self-assembly of amphiphilic polymers, double emulsification, coprecipitation, microfluidics and particle assembly, etc. are summarized. Furthermore, the diverse applications of MCPs in programmed delivery of various cargoes and biological applications including cancer therapy, antimicrobials, and regulation of blood glucose levels are highlighted. Finally, future perspectives of MCPs from the aspects of controlled synthesis and applications are proposed.
Collapse
Affiliation(s)
- Yaning Gao
- State Key Laboratory of High-Efficiency Coal Utilization and Green Chemical Engineering, School of Chemistry and Chemical Engineering, Ningxia University, Yinchuan 750021, China
| | - Chenchen Gao
- State Key Laboratory of High-Efficiency Coal Utilization and Green Chemical Engineering, School of Chemistry and Chemical Engineering, Ningxia University, Yinchuan 750021, China
| | - Yirong Fan
- State Key Laboratory of High-Efficiency Coal Utilization and Green Chemical Engineering, School of Chemistry and Chemical Engineering, Ningxia University, Yinchuan 750021, China
| | - Hui Sun
- State Key Laboratory of High-Efficiency Coal Utilization and Green Chemical Engineering, School of Chemistry and Chemical Engineering, Ningxia University, Yinchuan 750021, China
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai 200072, China
| |
Collapse
|
29
|
Xu X, Moreno S, Gentzel M, Zhang K, Wang D, Voit B, Appelhans D. Biomimetic Protocells Featuring Macrophage-Like Capture and Digestion of Protein Pathogens. SMALL METHODS 2023; 7:e2300257. [PMID: 37599260 DOI: 10.1002/smtd.202300257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/11/2023] [Indexed: 08/22/2023]
Abstract
Modern medical research develops interest in sophisticated artificial nano- and microdevices for future treatment of human diseases related to biological dysfunctions. This covers the design of protocells capable of mimicking the structure and functionality of eukaryotic cells. The authors use artificial organelles based on trypsin-loaded pH-sensitive polymeric vesicles to provide macrophage-like digestive functions under physiological conditions. Herein, an artificial cell is established where digestive artificial organelles (nanosize) are integrated into a protocell (microsize). With this method, mimicking crossing of different biological barriers, capture of model protein pathogens, and compartmentalized digestive function are possible. This allows the integration of different components (e.g., dextran as stabilizing block) and the diffusion of pathogens in simulated cytosolic environment under physiological conditions. An integrated characterization approach is carried out, with identifying electrospray ionization mass spectrometry as an excellent detection method for the degradation of a small peptide such as β-amyloid. The degradation of model enzymes is measured by enzyme activity assays. This work is an important contribution to effective biomimicry with the design of cell-like functions having potential for therapeutic action.
Collapse
Affiliation(s)
- Xiaoying Xu
- Leibniz-Institut für Polymerforschung Dresden e.V., D-01069, Dresden, Germany
- Organic Chemistry of Polymers, Technische Universität Dresden, D-01062, Dresden, Germany
| | - Silvia Moreno
- Leibniz-Institut für Polymerforschung Dresden e.V., D-01069, Dresden, Germany
| | - Marc Gentzel
- Center for Molecular and Cellular Bioengineering (CMCB), Core Facility Mass Spectrometry & Proteomics, Technische Universität Dresden, 01307, Dresden, Germany
| | - Kehu Zhang
- Leibniz-Institut für Polymerforschung Dresden e.V., D-01069, Dresden, Germany
- Organic Chemistry of Polymers, Technische Universität Dresden, D-01062, Dresden, Germany
| | - Dishi Wang
- Leibniz-Institut für Polymerforschung Dresden e.V., D-01069, Dresden, Germany
- Organic Chemistry of Polymers, Technische Universität Dresden, D-01062, Dresden, Germany
| | - Brigitte Voit
- Leibniz-Institut für Polymerforschung Dresden e.V., D-01069, Dresden, Germany
- Organic Chemistry of Polymers, Technische Universität Dresden, D-01062, Dresden, Germany
| | - Dietmar Appelhans
- Leibniz-Institut für Polymerforschung Dresden e.V., D-01069, Dresden, Germany
| |
Collapse
|
30
|
Gao R, Yu X, Kumar BVVSP, Tian L. Hierarchical Structuration in Protocellular System. SMALL METHODS 2023; 7:e2300422. [PMID: 37438327 DOI: 10.1002/smtd.202300422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/12/2023] [Indexed: 07/14/2023]
Abstract
Spatial control is one of the ubiquitous features in biological systems and the key to the functional complexity of living cells. The strategies to achieve such precise spatial control in protocellular systems are crucial to constructing complex artificial living systems with functional collective behavior. Herein, the authors review recent advances in the spatial control within a single protocell or between different protocells and discuss how such hierarchical structured protocellular system can be used to understand complex living systems or to advance the development of functional microreactors with the programmable release of various biomacromolecular payloads, or smart protocell-biological cell hybrid system.
Collapse
Affiliation(s)
- Rui Gao
- Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xinran Yu
- Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | | | - Liangfei Tian
- Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
- Department of Ultrasound, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310027, China
- Innovation Center for Smart Medical Technologies & Devices, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| |
Collapse
|
31
|
Kayani A, Raza A, Si J, Dutta D, Zhou Q, Ge Z. Polymersome Membrane Engineering with Active Targeting or Controlled Permeability for Responsive Drug Delivery. Biomacromolecules 2023; 24:4622-4645. [PMID: 37870458 DOI: 10.1021/acs.biomac.3c00839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Polymersomes have been extensively investigated for drug delivery as nanocarriers for two decades due to a series of advantages including high stability under physiological conditions, simultaneous encapsulation of hydrophilic and hydrophobic drugs inside inner cavities and membranes, respectively, and facile adjustment of membrane and surface properties, as well as controlled drug release through incorporation of stimuli-responsive components. Despite these features, polymersome nanocarriers frequently suffer from nontargeting delivery and poor membrane permeability. In recent years, polymersomes have been functionalized for more efficient drug delivery. The surface shells were explored to be modified with diverse active targeting groups to improve disease-targeting delivery. The membrane permeability of the polymersomes was adjusted by incorporation of the stimuli-responsive components for smart controlled transportation of the encapsulated drugs. Therefore, being the polymersome-biointerface, tailorable properties can be introduced by its carefully modulated engineering. This review elaborates on the role of polymersome membranes as a platform to incorporate versatile features. First, we discuss how surface functionalization facilitates the directional journey to the targeting sites toward specific diseases, cells, or intracellular organelles via active targeting. Moreover, recent advances in the past decade related to membrane permeability to control drug release are also summarized. We finally discuss future development to promote polymersomes as in vivo drug delivery nanocarriers.
Collapse
Affiliation(s)
- Anum Kayani
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Arsalan Raza
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Jiale Si
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Debabrata Dutta
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Qinghao Zhou
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Zhishen Ge
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| |
Collapse
|
32
|
Grillo DA, Albano JMR, Valladares T. RE, Mocskos EE, Facelli JC, Pickholz M, Ferraro MB. Molecular dynamics study of the mechanical properties of drug loaded model systems: A comparison of a polymersome with a bilayer. J Chem Phys 2023; 159:174908. [PMID: 37929867 PMCID: PMC10629967 DOI: 10.1063/5.0165478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023] Open
Abstract
In this work we implement a new methodology to study structural and mechanical properties of systems having spherical and planar symmetries throughout Molecular Dynamics simulations. This methodology is applied here to a drug delivery system based in polymersomes, as an example. The chosen model drug was the local anesthetic prilocaine due to previous parameterization within the used coarse grain scheme. In our approach, mass density profiles (MDPs) are used to obtain key structural parameters of the systems, and pressure profiles are used to estimate the curvature elastic parameters. The calculation of pressure profiles and radial MPDs required the development of specific methods, which were implemented in an in-house built version of the GROMACS 2018 code. The methodology presented in this work is applied to characterize poly(ethylene oxide)-poly(butadiene) polymersomes and bilayers loaded with the model drug prilocaine. Our results show that structural properties of the polymersome membrane could be obtained from bilayer simulations, with significantly lower computational cost compared to whole polymersome simulations, but the bilayer simulations are insufficient to get insights on their mechanical aspects, since the elastic parameters are canceled out for the complete bilayer (as consequence of the symmetry). The simulations of entire polymersomes, although more complex, offer a complementary approach to get insights on the mechanical behavior of the systems.
Collapse
Affiliation(s)
| | - Juan M. R. Albano
- CONICET - Universidad de Buenos Aires, Instituto de Física de Buenos Aires (IFIBA), Buenos Aires, Argentina
| | - Rufino E. Valladares T.
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Física, Buenos Aires, Argentina
| | | | - Julio C. Facelli
- Department of Biomedical Informatics, University of Utah, 421 Wakara Way, Suite 140, Salt Lake City, Utah 84108, USA
| | | | | |
Collapse
|
33
|
Stengel D, Jörgensen AM, Polidori I, Kapitza P, Ricci F, Bernkop-Schnürch A. The power of sulfhydryl groups: Thiolated lipid-based nanoparticles enhance cellular uptake of nucleic acids. J Colloid Interface Sci 2023; 654:1136-1145. [PMID: 39491903 DOI: 10.1016/j.jcis.2023.10.039] [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: 07/11/2023] [Revised: 09/29/2023] [Accepted: 10/09/2023] [Indexed: 11/05/2024]
Abstract
AIM The aim of the study was to evaluate the effect of thiolation of lipid-based nanoparticles (LNPs) on cellular uptake of nucleic acids. METHODS A thiolated surfactant was synthesized by binding palmitic acid covalently to cysteine. Green fluorescent protein (GFP) encoding plasmid DNA (pDNA) was used as model nucleic acid and incorporated via hydrophobic ion-pairing with a cationic cholesterol derivate (DcCholesterol) in LNPs that were prepared by solvent injection method using the thiolated surfactant for surface decoration. LNPs were characterized regarding size, polydispersity index, zeta potential and stability in biorelevant media. The endosomal escape properties of LNPs were evaluated by erythrocytes interaction studies. Cell viability and transfection efficiency on HEK293 cells were investigated. RESULTS The structure of the thiolated surfactant was confirmed by 1H NMR and FT-IR. LNPs containing the nucleic acid-DcCholesterol complex with and without the thiolated surfactant were developed and displayed sizes in the range from 173 nm to 233 nm with a narrow size distribution (PDI < 0.3) and a negative zeta potential. LNPs showed no significant increase in size after 4 h of incubation in artificial body fluids. Erythrocytes interaction studies revealed enhanced endosomal escape properties for thiolated LNPs compared to non-thiolated LNPs. LNPs showed in concentrations lower than 0.74 mg/mL cell viability ≥ 80 %. Transfection studies on HEK cells with thiolated LNPs compared to non-thiolated LNP showed a 4.6-fold higher expression of GFP. CONCLUSION Surface thiolation of LNPs represents a promising tool for enhancing intracellular nucleic acid delivery of LNPs.
Collapse
Affiliation(s)
- Daniel Stengel
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Arne M Jörgensen
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Ilaria Polidori
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Paul Kapitza
- Department of Pharmaceutical Chemistry, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Fabrizio Ricci
- Thiomatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, 6020 Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria.
| |
Collapse
|
34
|
Trimaille T, Verrier B. Copolymer Micelles: A Focus on Recent Advances for Stimulus-Responsive Delivery of Proteins and Peptides. Pharmaceutics 2023; 15:2481. [PMID: 37896241 PMCID: PMC10609739 DOI: 10.3390/pharmaceutics15102481] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Historically used for the delivery of hydrophobic drugs through core encapsulation, amphiphilic copolymer micelles have also more recently appeared as potent nano-systems to deliver protein and peptide therapeutics. In addition to ease and reproducibility of preparation, micelles are chemically versatile as hydrophobic/hydrophilic segments can be tuned to afford protein immobilization through different approaches, including non-covalent interactions (e.g., electrostatic, hydrophobic) and covalent conjugation, while generally maintaining protein biological activity. Similar to many other drugs, protein/peptide delivery is increasingly focused on stimuli-responsive nano-systems able to afford triggered and controlled release in time and space, thereby improving therapeutic efficacy and limiting side effects. This short review discusses advances in the design of such micelles over the past decade, with an emphasis on stimuli-responsive properties for optimized protein/peptide delivery.
Collapse
Affiliation(s)
- Thomas Trimaille
- Ingénierie des Matériaux Polymères, Univ Lyon, CNRS, Université Claude Bernard Lyon 1, INSA Lyon, Université Jean Monnet, UMR 5223, CEDEX, 69622 Villeurbanne, France
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et d’Ingénierie Thérapeutique, Univ Lyon, CNRS, Université Claude Bernard Lyon 1, UMR 5305, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France;
| |
Collapse
|
35
|
Rathee S, Ojha A, Upadhyay A, Xiao J, Bajpai VK, Ali S, Shukla S. Biogenic engineered nanomaterials for enhancing bioavailability via developing nano-iron-fortified smart foods: advances, insight, and prospects of nanobionics in fortification of food. Food Funct 2023; 14:9083-9099. [PMID: 37750182 DOI: 10.1039/d3fo02473c] [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: 09/27/2023]
Abstract
Iron deficiency is a significant cause of iron deficiency anemia (IDA). Treatment of IDA is challenging due to several challenges, including low target bioavailability, low palatability, poor pharmacokinetics, and extended therapeutic regimes. Nanotechnology holds the promise of revolutionizing the management and treatment of IDA. Smart biogenic engineered nanomaterials (BENMs) such as lipids, protein, carbohydrates, and complex nanomaterials have been the subject of extensive research and opened new avenues for people and the planet due to their enhanced physicochemical, rheological, optoelectronic, thermomechanical, biological, magnetic, and nutritional properties. Additionally, they show eco-sustainability, low biotoxicity, active targeting, enhanced permeation and retention, and stimuli-responsive characteristics. We examine the opportunities offered by emerging smart BENMs for the treatment of iron deficiency anemia by utilizing iron-fortified smart foods. We review the progress made so far and other future directions to maximize the impact of smart nanofortification on the global population. The toxicity effects are also discussed with commercialization challenges.
Collapse
Affiliation(s)
- Shweta Rathee
- Department of Food Science and Technology, National Institute of Food Science Technology Entrepreneurship and Management, Kundli, Sonipat, India.
| | - Ankur Ojha
- Department of Food Science and Technology, National Institute of Food Science Technology Entrepreneurship and Management, Kundli, Sonipat, India.
| | - Ashutosh Upadhyay
- Department of Food Science and Technology, National Institute of Food Science Technology Entrepreneurship and Management, Kundli, Sonipat, India.
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, E-32004 Ourense, Spain
| | - Vivek K Bajpai
- Department of Energy and Materials Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Sajad Ali
- Department of Biotechnology, Yeungnam University, South Korea.
| | - Shruti Shukla
- Department of Nanotechnology, North Eastern Hill University (NEHU), East Khasi Hills, Shillong, 793022, Meghalaya, India.
| |
Collapse
|
36
|
Yadav K, Sahu KK, Sucheta, Gnanakani SPE, Sure P, Vijayalakshmi R, Sundar VD, Sharma V, Antil R, Jha M, Minz S, Bagchi A, Pradhan M. Biomedical applications of nanomaterials in the advancement of nucleic acid therapy: Mechanistic challenges, delivery strategies, and therapeutic applications. Int J Biol Macromol 2023; 241:124582. [PMID: 37116843 DOI: 10.1016/j.ijbiomac.2023.124582] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023]
Abstract
In the past few decades, substantial advancement has been made in nucleic acid (NA)-based therapies. Promising treatments include mRNA, siRNA, miRNA, and anti-sense DNA for treating various clinical disorders by modifying the expression of DNA or RNA. However, their effectiveness is limited due to their concentrated negative charge, instability, large size, and host barriers, which make widespread application difficult. The effective delivery of these medicines requires safe vectors that are efficient & selective while having non-pathogenic qualities; thus, nanomaterials have become an attractive option with promising possibilities despite some potential setbacks. Nanomaterials possess ideal characteristics, allowing them to be tuned into functional bio-entity capable of targeted delivery. In this review, current breakthroughs in the non-viral strategy of delivering NAs are discussed with the goal of overcoming challenges that would otherwise be experienced by therapeutics. It offers insight into a wide variety of existing NA-based therapeutic modalities and techniques. In addition to this, it provides a rationale for the use of non-viral vectors and a variety of nanomaterials to accomplish efficient gene therapy. Further, it discusses the potential for biomedical application of nanomaterials-based gene therapy in various conditions, such as cancer therapy, tissue engineering, neurological disorders, and infections.
Collapse
Affiliation(s)
- Krishna Yadav
- Raipur Institute of Pharmaceutical Education and Research, Sarona, Raipur, Chhattisgarh 492010, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | | | - Pavani Sure
- Department of Pharmaceutics, Vignan Institute of Pharmaceutical Sciences, Hyderabad, Telangana, India
| | - R Vijayalakshmi
- Department of Pharmaceutical Analysis, GIET School of Pharmacy, Chaitanya Knowledge City, Rajahmundry, AP 533296, India
| | - V D Sundar
- Department of Pharmaceutical Technology, GIET School of Pharmacy, Chaitanya Knowledge City, Rajahmundry, AP 533296, India
| | - Versha Sharma
- Department of Biotechnology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, M.P. 470003, India
| | - Ruchita Antil
- Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, England, United Kingdom of Great Britain and Northern Ireland
| | - Megha Jha
- Department of Biotechnology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, M.P. 470003, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, M.P., 484887, India
| | - Anindya Bagchi
- Tumor Initiation & Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road La Jolla, CA 92037, USA
| | | |
Collapse
|
37
|
Foster D, Larsen J. Polymeric Metal Contrast Agents for T 1-Weighted Magnetic Resonance Imaging of the Brain. ACS Biomater Sci Eng 2023; 9:1224-1242. [PMID: 36753685 DOI: 10.1021/acsbiomaterials.2c01386] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Imaging plays an integral role in diagnostics and treatment monitoring for conditions affecting the brain; enhanced brain imaging capabilities will improve upon both while increasing the general understanding of how the brain works. T1-weighted magnetic resonance imaging is the preferred modality for brain imaging. Commercially available contrast agents, which are often required to render readable brain images, have considerable toxicity concerns. In recent years, much progress has been made in developing new contrast agents based on the magnetic features of gadolinium, iron, or magnesium. Nanotechnological approaches for these systems allow for the protected integration of potentially harmful metals with added benefits like reduced dosage and improved transport. Polymeric enhancement of each design further improves biocompatibility while allowing for specific brain targeting. This review outlines research on polymeric nanomedicine designs for T1-weighted contrast agents that have been evaluated for performance in the brain.
Collapse
|
38
|
Guo L, Xu J, Du B. Self-assembly of ABCBA Linear Pentablock Terpolymers. POLYM REV 2023. [DOI: 10.1080/15583724.2023.2178008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Affiliation(s)
- Lei Guo
- State Key Laboratory of Motor Vehicle Biofuel Technology, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou, China
| | - Junting Xu
- State Key Laboratory of Motor Vehicle Biofuel Technology, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou, China
| | - Binyang Du
- State Key Laboratory of Motor Vehicle Biofuel Technology, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
A Comprehensive Review of mRNA Vaccines. Int J Mol Sci 2023; 24:ijms24032700. [PMID: 36769023 PMCID: PMC9917162 DOI: 10.3390/ijms24032700] [Citation(s) in RCA: 159] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
mRNA vaccines have been demonstrated as a powerful alternative to traditional conventional vaccines because of their high potency, safety and efficacy, capacity for rapid clinical development, and potential for rapid, low-cost manufacturing. These vaccines have progressed from being a mere curiosity to emerging as COVID-19 pandemic vaccine front-runners. The advancements in the field of nanotechnology for developing delivery vehicles for mRNA vaccines are highly significant. In this review we have summarized each and every aspect of the mRNA vaccine. The article describes the mRNA structure, its pharmacological function of immunity induction, lipid nanoparticles (LNPs), and the upstream, downstream, and formulation process of mRNA vaccine manufacturing. Additionally, mRNA vaccines in clinical trials are also described. A deep dive into the future perspectives of mRNA vaccines, such as its freeze-drying, delivery systems, and LNPs targeting antigen-presenting cells and dendritic cells, are also summarized.
Collapse
|
40
|
Gouveia MG, Wesseler JP, Ramaekers J, Weder C, Scholten PBV, Bruns N. Polymersome-based protein drug delivery - quo vadis? Chem Soc Rev 2023; 52:728-778. [PMID: 36537575 PMCID: PMC9890519 DOI: 10.1039/d2cs00106c] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Indexed: 12/24/2022]
Abstract
Protein-based therapeutics are an attractive alternative to established therapeutic approaches and represent one of the fastest growing families of drugs. While many of these proteins can be delivered using established formulations, the intrinsic sensitivity of proteins to denaturation sometimes calls for a protective carrier to allow administration. Historically, lipid-based self-assembled structures, notably liposomes, have performed this function. After the discovery of polymersome-based targeted drug-delivery systems, which offer manifold advantages over lipid-based structures, the scientific community expected that such systems would take the therapeutic world by storm. However, no polymersome formulations have been commercialised. In this review article, we discuss key obstacles for the sluggish translation of polymersome-based protein nanocarriers into approved pharmaceuticals, which include limitations imparted by the use of non-degradable polymers, the intricacies of polymersome production methods, and the complexity of the in vivo journey of polymersomes across various biological barriers. Considering this complex subject from a polymer chemist's point of view, we highlight key areas that are worthy to explore in order to advance polymersomes to a level at which clinical trials become worthwhile and translation into pharmaceutical and nanomedical applications is realistic.
Collapse
Affiliation(s)
- Micael G Gouveia
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Justus P Wesseler
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Jobbe Ramaekers
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Christoph Weder
- Adolphe Merkle Institute, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Philip B V Scholten
- Adolphe Merkle Institute, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Nico Bruns
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
- Department of Chemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany.
| |
Collapse
|
41
|
Zong W, Shao X, Li J, Chai Y, Hu X, Zhang X. Synthetic Intracellular Environments: From Basic Science to Applications. Anal Chem 2023; 95:535-549. [PMID: 36625127 DOI: 10.1021/acs.analchem.2c04199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Wei Zong
- College of Chemistry and Chemical Engineering, Qiqihar University, No. 42 Wenhua Street, Qiqihar161006, China
| | - Xiaotong Shao
- College of Chemistry and Chemical Engineering, Qiqihar University, No. 42 Wenhua Street, Qiqihar161006, China
| | - Jinlong Li
- College of Chemistry and Chemical Engineering, Qiqihar University, No. 42 Wenhua Street, Qiqihar161006, China.,Heilongjiang Provincial Key Laboratory of Catalytic Synthesis for Fine Chemicals, Qiqihar University, Qiqihar161006, China
| | - Yunhe Chai
- College of Chemistry and Chemical Engineering, Qiqihar University, No. 42 Wenhua Street, Qiqihar161006, China
| | - Xinyu Hu
- Key Laboratory of Micro-Nano Optoelectronic Devices (Wenzhou), College of Electrical and Electronic Engineering, Wenzhou University, Wenzhou325035, China
| | - Xunan Zhang
- College of Chemistry and Chemical Engineering, Qiqihar University, No. 42 Wenhua Street, Qiqihar161006, China
| |
Collapse
|
42
|
Non-viral nucleic acid delivery approach: A boon for state-of-the-art gene delivery. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
43
|
Du H, Kalem S, Huin C, Illy N, Tresset G, Giacomelli FC, Guégan P. Engineering of ion permeable planar membranes and polymersomes based on β-cyclodextrin-cored star copolymers. J Colloid Interface Sci 2023; 630:465-476. [DOI: 10.1016/j.jcis.2022.09.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/09/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022]
|
44
|
Phase separation in polymer-based biomimetic structures containing planar membranes. Biointerphases 2022; 17:060802. [PMID: 36575113 DOI: 10.1116/6.0002078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Phase separation in biological membranes is crucial for proper cellular functions, such as signaling and trafficking, as it mediates the interactions of condensates on membrane-bound organelles and transmembrane transport to targeted destination compartments. The separation of a lipid bilayer into phases and the formation of lipid rafts involve the restructuring of molecular localization, their immobilization, and local accumulation. By understanding the processes underlying the formation of lipid rafts in a cellular membrane, it is possible to reconstitute this phenomenon in synthetic biomimetic membranes, such as hybrids of lipids and polymers or membranes composed solely of polymers, which offer an increased physicochemical stability and unlimited possibilities of chemical modification and functionalization. In this article, we relate the main lipid bilayer phase transition phenomenon with respect to hybrid biomimetic membranes, composed of lipids mixed with polymers, and fully synthetic membranes. Following, we review the occurrence of phase separation in biomimetic hybrid membranes based on lipids and/or direct lipid analogs, amphiphilic block copolymers. We further exemplify the phase separation and the resulting properties and applications in planar membranes, free-standing and solid-supported. We briefly list methods leading to the formation of such biomimetic membranes and reflect on their improved overall stability and influence on the separation into different phases within the membranes. Due to the importance of phase separation and compartmentalization in cellular membranes, we are convinced that this compiled overview of this phenomenon will be helpful for any researcher in the biomimicry area.
Collapse
|
45
|
Alsaab HO, Alharbi FD, Alhibs AS, Alanazi NB, Alshehri BY, Saleh MA, Alshehri FS, Algarni MA, Almugaiteeb T, Uddin MN, Alzhrani RM. PLGA-Based Nanomedicine: History of Advancement and Development in Clinical Applications of Multiple Diseases. Pharmaceutics 2022; 14:pharmaceutics14122728. [PMID: 36559223 PMCID: PMC9786338 DOI: 10.3390/pharmaceutics14122728] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
Research on the use of biodegradable polymers for drug delivery has been ongoing since they were first used as bioresorbable surgical devices in the 1980s. For tissue engineering and drug delivery, biodegradable polymer poly-lactic-co-glycolic acid (PLGA) has shown enormous promise among all biomaterials. PLGA are a family of FDA-approved biodegradable polymers that are physically strong and highly biocompatible and have been extensively studied as delivery vehicles of drugs, proteins, and macromolecules such as DNA and RNA. PLGA has a wide range of erosion times and mechanical properties that can be modified. Many innovative platforms have been widely studied and created for the development of methods for the controlled delivery of PLGA. In this paper, the various manufacturing processes and characteristics that impact their breakdown and drug release are explored in depth. Besides different PLGA-based nanoparticles, preclinical and clinical applications for different diseases and the PLGA platform types and their scale-up issues will be discussed.
Collapse
Affiliation(s)
- Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
- Correspondence: ; Tel.: +966-556047523
| | - Fatima D. Alharbi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alanoud S. Alhibs
- Department of Pharmacy, King Fahad Medical City, Riyadh 11564, Saudi Arabia
| | - Nouf B. Alanazi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Bayan Y. Alshehri
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Marwa A. Saleh
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11754, Egypt
| | - Fahad S. Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 24382, Saudi Arabia
| | - Majed A. Algarni
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Turki Almugaiteeb
- Taqnia-Research Products Development Company, Riyadh 13244, Saudi Arabia
| | | | - Rami M. Alzhrani
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
| |
Collapse
|
46
|
Cerofolini L, Parigi G, Ravera E, Fragai M, Luchinat C. Solid-state NMR methods for the characterization of bioconjugations and protein-material interactions. SOLID STATE NUCLEAR MAGNETIC RESONANCE 2022; 122:101828. [PMID: 36240720 DOI: 10.1016/j.ssnmr.2022.101828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 06/16/2023]
Abstract
Protein solid-state NMR has evolved dramatically over the last two decades, with the development of new hardware and sample preparation methodologies. This technique is now ripe for complex applications, among which one can count bioconjugation, protein chemistry and functional biomaterials. In this review, we provide our account on this aspect of protein solid-state NMR.
Collapse
Affiliation(s)
- Linda Cerofolini
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
| | - Giacomo Parigi
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy; Magnetic Resonance Center (CERM), Università degli Studi di Firenze, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy; Department of Chemistry "Ugo Schiff", Università degli Studi di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Enrico Ravera
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy; Magnetic Resonance Center (CERM), Università degli Studi di Firenze, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy; Department of Chemistry "Ugo Schiff", Università degli Studi di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy; Florence Data Science, Università degli Studi di Firenze, Italy.
| | - Marco Fragai
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy; Magnetic Resonance Center (CERM), Università degli Studi di Firenze, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy; Department of Chemistry "Ugo Schiff", Università degli Studi di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy.
| | - Claudio Luchinat
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy; Magnetic Resonance Center (CERM), Università degli Studi di Firenze, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy; Department of Chemistry "Ugo Schiff", Università degli Studi di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy.
| |
Collapse
|
47
|
Liu Y, Hu F, Wang S, Xu M, Yu Q, Wang L. Evaluating the integrity of polymersomes by FRET for optimization of the lyophilization parameters. POLYMER 2022. [DOI: 10.1016/j.polymer.2022.125375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Alhaj-Suliman SO, Wafa EI, Salem AK. Engineering nanosystems to overcome barriers to cancer diagnosis and treatment. Adv Drug Deliv Rev 2022; 189:114482. [PMID: 35944587 DOI: 10.1016/j.addr.2022.114482] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 01/24/2023]
Abstract
Over the past two decades, multidisciplinary investigations into the development of nanoparticles for medical applications have continually increased. However, nanoparticles are still subject to biological barriers and biodistribution challenges, which limit their overall clinical potential. This has motivated the implementation of innovational modifications to a range of nanoparticle formulations designed for cancer imaging and/or cancer treatment to overcome specific barriers and shift the accumulation of payloads toward the diseased tissues. In recent years, novel technological and chemical approaches have been employed to modify or functionalize the surface of nanoparticles or manipulate the characteristics of nanoparticles. Combining these approaches with the identification of critical biomarkers provides new strategies for enhancing nanoparticle specificity for both cancer diagnostic and therapeutic applications. This review discusses the most recent advances in the design and engineering of nanoparticles as well as future directions for developing the next generation of nanomedicines.
Collapse
Affiliation(s)
- Suhaila O Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Emad I Wafa
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States; Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, United States.
| |
Collapse
|
49
|
Singh A, Agarwal A, Chakraborty A, Bhardwaj R, Sutradhar S, Kumar Mittal A, Kumar Rajput S, Gupta M, Ray D, Mukherjee M. Click chemistry tailored benzimidazole functionalized triazole block-co-polymer for emergence of exotic chimaeric nano-crystalsomes. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
50
|
Domingues C, Santos A, Alvarez-Lorenzo C, Concheiro A, Jarak I, Veiga F, Barbosa I, Dourado M, Figueiras A. Where Is Nano Today and Where Is It Headed? A Review of Nanomedicine and the Dilemma of Nanotoxicology. ACS NANO 2022; 16:9994-10041. [PMID: 35729778 DOI: 10.1021/acsnano.2c00128] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Worldwide nanotechnology development and application have fueled many scientific advances, but technophilic expectations and technophobic demands must be counterbalanced in parallel. Some of the burning issues today are the following: (1) Where is nano today? (2) How good are the communication and investment networks between academia/research and governments? (3) Is there any spotlight application for nanotechnology? Nanomedicine is a particular arm of nanotechnology within the healthcare landscape, focused on diagnosis, treatment, and monitoring of emerging (such as coronavirus disease 2019, COVID-19) and contemporary (including diabetes, cardiovascular diseases, neurodegenerative disorders, and cancer) diseases. However, it may only represent the bright side of the coin. In fact, in the recent past, the concept of nanotoxicology has emerged to address the dark shadows of nanomedicine. The nanomedicine field requires more nanotoxicological studies to identify undesirable effects and guarantee safety. Here, we provide an overall perspective on nanomedicine and nanotoxicology as central pieces of the giant puzzle of nanotechnology. First, the impact of nanotechnology on education and research is highlighted, followed by market trends and scientific output tendencies. In the next section, the nanomedicine and nanotoxicology dilemma is addressed through the interplay of in silico, in vitro, and in vivo models with the support of omics and microfluidic approaches. Lastly, a reflection on the regulatory issues and clinical trials is provided. Finally, some conclusions and future perspectives are proposed for a clearer and safer translation of nanomedicines from the bench to the bedside.
Collapse
Affiliation(s)
- Cátia Domingues
- Univ. Coimbra, Faculty of Pharmacy, Galenic and Pharmaceutical Technology Laboratory, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Galenic and Pharmaceutical Technology Laboratory, Faculty of Pharmacy, Univ. Coimbra, 3000-548 Coimbra, Portugal
- Univ. Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, 3000-548 Coimbra, Portugal
| | - Ana Santos
- Univ. Coimbra, Faculty of Pharmacy, Galenic and Pharmaceutical Technology Laboratory, 3000-548 Coimbra, Portugal
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, iMATUS, and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, iMATUS, and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ivana Jarak
- Univ. Coimbra, Faculty of Pharmacy, Galenic and Pharmaceutical Technology Laboratory, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Univ. Coimbra, Faculty of Pharmacy, Galenic and Pharmaceutical Technology Laboratory, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Galenic and Pharmaceutical Technology Laboratory, Faculty of Pharmacy, Univ. Coimbra, 3000-548 Coimbra, Portugal
| | - Isabel Barbosa
- Univ. Coimbra, Faculty of Pharmacy, Phamaceutical Chemistry Laboratory, 3000-548 Coimbra, Portugal
| | - Marília Dourado
- Univ. Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, 3000-548 Coimbra, Portugal
- Univ. Coimbra, Center for Health Studies and Research of the University of Coimbra (CEISUC), Faculty of Medicine, 3000-548 Coimbra, Portugal
- Univ. Coimbra, Center for Studies and Development of Continuous and Palliative Care (CEDCCP), Faculty of Medicine, 3000-548 Coimbra, Portugal
| | - Ana Figueiras
- Univ. Coimbra, Faculty of Pharmacy, Galenic and Pharmaceutical Technology Laboratory, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Galenic and Pharmaceutical Technology Laboratory, Faculty of Pharmacy, Univ. Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|