1
|
Santra S, Molla MR. Small molecule-based core and shell cross-linked nanoassemblies: from self-assembly and programmed disassembly to biological applications. Chem Commun (Camb) 2024; 60:12101-12117. [PMID: 39301871 DOI: 10.1039/d4cc03515a] [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: 09/22/2024]
Abstract
Supramolecular assemblies of stimuli-responsive amphiphilic molecules have been of utmost interest in targeted drug delivery applications, owing to their capability of sequestering drug molecules in one set of conditions and releasing them in another. To minimize undesired disassembly and stabilize noncovalently encapsulated drug molecules, the strategy of core or shell cross-linking has become a fascinating approach to constructing cross-linked polymeric or small molecule-based nanoassemblies. In this article, we discuss the design and synthetic strategies for cross-linked nanoassemblies from small molecule-based amphiphiles, with robust stability and enhanced drug encapsulation capability. We highlight their potential biomedical applications, particularly in drug or gene delivery, and cell imaging. This feature article offers a comprehensive overview of the recent developments in the application of small molecule-based covalently cross-linked nanocarriers for materials and biomedical applications, which may inspire the use of these materials as a potential drug delivery system for future chemotherapeutic applications.
Collapse
Affiliation(s)
- Subrata Santra
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata-700009, India.
| | - Mijanur Rahaman Molla
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata-700009, India.
| |
Collapse
|
2
|
Kitayama Y, Takigawa S, Yuba E, Harada A. Poly(vinyl alcohol)-Incorporated Core-Shell Polymer Nanogels Functionalized by Block Copolymer Installation for Cisplatin Delivery. Biomacromolecules 2024; 25:6465-6473. [PMID: 39230243 DOI: 10.1021/acs.biomac.4c00605] [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: 09/05/2024]
Abstract
The functionalization approach for nanomaterials is of great importance for their application in drug delivery systems. Herein, an approach based on block copolymer installation into polymer nanogels was newly developed. Poly(vinyl alcohol)-incorporated polymer nanogels were prepared by a two-step dispersion/precipitation polymerization. Poly(methacrylic acid)-block-poly(3-fluorophenylboronic acid methacrylamide) (PMAA-b-PFPBMA) prepared by two-step reversible addition-fragmentation chain transfer polymerization was installed into the polymer nanogels via boronate ester formation. Furthermore, cisplatin as a cancer therapeutic drug was successfully loaded on the block copolymer-installed polymer nanogels, and cell death was achieved by using the resulting cisplatin-loaded nanogels. We believe that the functionality of the nanogels can be changed by varying the installed block copolymer, leading to the functionalization approach of polymer nanogels based on block copolymer installation, which will be of great utility in many fields.
Collapse
Affiliation(s)
- Yukiya Kitayama
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Shunsuke Takigawa
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Atsushi Harada
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| |
Collapse
|
3
|
Tang Z, Lan W, Wen K, Li W, Wang T, Zhou D, Su H. Spontaneous assembly of a class of small molecule prodrugs directed by SN38. J Mater Chem B 2024; 12:9921-9929. [PMID: 39252501 DOI: 10.1039/d4tb01429d] [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: 09/11/2024]
Abstract
Small molecule self-assembling prodrugs (SAPDs) are an emerging class of amphiphilic monomers that can aggregate into supramolecular nanostructures with high drug loading identical to that of the individual prodrug. Despite great progress in creating nanodrugs via nanoprecipitation, the direct self-assembly of small molecule SAPDs in aqueous solution remains challenging, as the proper hydrophilic-hydrophobic balance and intermolecular interactions have to be rationally considered. We report a class of small molecule SAPDs by conjugating the anticancer drug SN38 as the structure-directing component with various hydrophilic auxiliaries (i.e., oligo ethylene glycol (OEG) of different lengths, amino, and carboxyl groups) via a self-immolative disulfanyl-ethyl carbonate linker. Driven by π-π interactions between SN38 units, these SAPDs spontaneously assembled into well-defined fibrous nanostructures. Variations in hydrophilic domains can robustly regulate the hydrophobicity of SAPDs, as well as the morphologies and surface features of supramolecular filaments, subsequently influencing cellular internalization behaviors. Furthermore, our study also reveals that the parent drug can be efficiently and controllably released in the presence of glutathione (GSH), exhibiting high in vitro toxicity against colorectal cancer cells. In this work, we present a delicate platform to design small molecule SAPDs that can spontaneously self-assemble into supramolecular filamentous assemblies directed by aromatic interaction of the parent drugs, providing a new strategy to optimize supramolecular drug delivery systems.
Collapse
Affiliation(s)
- Zhenhai Tang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Wenning Lan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Kaiying Wen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Wenting Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Tao Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd Section, South Renmin Road, Chengdu 610041, China.
| | - Dongdong Zhou
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Hao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
4
|
Bera S, Ghosh S. Alternating vs. random amphiphilic polydisulfides: aggregation, enzyme activity inhibition and redox-responsive guest release. NANOSCALE 2024; 16:17886-17892. [PMID: 39248029 DOI: 10.1039/d4nr02494j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Herein, we report the synthesis of an alternating copolymer (ACP) with a bio-reducible amphiphilic polydisulfide backbone and highlight the impact of the alternating monomer connectivity on the self-assembly, morphology, chain-exchange dynamics, drug-release kinetics, and enzyme activity inhibition. Condensation polymerization between hydrophobic 1,10-bis(pyridin-2-yldisulfaneyl)decane and hydrophilic 2,3-mercaptosuccinic acid (1.04 : 1.00 ratio) generated amphiphilic ACP P1 (Mw = 8450 g mol-1, Đ = 1.3), which exhibited self-assembly in water, leading to the formation of an ultra-thin (height <5.0 nm) entangled fibrillar network. In contrast, structurally similar amphiphilic random copolymer P2 exhibited a truncated irregular disc-like morphology under the same conditions. It is postulated that due to the perfect alternating sequence of the hydrophobic and hydrophilic segments in P1, its immiscibility-driven aggregation in water leads to a pleated structure, which further assembles and forms the observed long fibrillar structures, similar to crystallization-driven self-assembly. In fact, wide-angle X-ray diffraction (WXRD) analysis of a lyophilized P1 sample showed sharp peaks, indicating its crystalline nature (approximately 37% crystallinity), and these were completely missing for P2. The effect of such distinct self-assembly on the chain-exchange dynamics was probed by fluorescence resonance energy transfer (FRET) using 3,3'-dioctadecyloxacarbocyanine perchlorate (DiO) and 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI) as the FRET-donor and -acceptor, respectively. For DiI- and DiO-entrapped solutions of P1, when mixed, no prominent FRET appeared even after 24 h. In sharp contrast, for P2, intense FRET emission occurred, and the FRET ratio (approximately 0.9) reached saturation in approximately 15 h, indicating the greatly enhanced kinetic stability of P1 aggregates. Glutathione-induced release of encapsulated Nile red showed much slower kinetics for P1 compared to that of P2, which was corroborated by the observed slow chain-exchange dynamics of the highly stable alternating copolymer assembly. Furthermore, the well-ordered assembly of P1 exhibited an excellent surface-functional group display (zeta potential of -32 mV compared to -14 mV for P2), which resulted in the effective recognition of the α-chymotrypsin (Cht) protein surface by electrostatic interaction. Consequently, P1 significantly (>70%) suppressed the enzymatic activity of Cht, while in the presence of P2, the enzyme was still active with >70% efficacy.
Collapse
Affiliation(s)
- Sukanya Bera
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, India 700032.
| | - Suhrit Ghosh
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, India 700032.
| |
Collapse
|
5
|
Obata M, Yamaguchi S, Yoshimura T. A Stochastic FRET Study on the Core Dimension of Polystyrene- block-Poly(Polyethylene Glycol Monomethyl Ether Acrylate) Micelles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:20596-20603. [PMID: 39292970 DOI: 10.1021/acs.langmuir.4c02374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Polystyrene-b-poly(polyethylene glycol monomethyl ether acrylate) (PSt-b-PPEGA) copolymers featuring pyrene and perylene as the Förster resonance energy transfer (FRET) donor (denoted as D-BCP) and acceptor (denoted as A-BCP), respectively, were synthesized via the reversible addition and fragmentation chain transfer (RAFT) polymerization. These copolymers were then used to form DA-mixed micelles via a dialysis method. The micelles consisted of D-BCP (mole fraction fD = 0.04), A-BCP (fA = 0.04), and label-free PSt-b-PPEGA (fN = 0.92). The decrease in fluorescence intensity of pyrene in the micelles confirmed the occurrence of FRET, with an observed efficiency of 0.32. A Monte Carlo approach was employed to estimate the expected FRET efficiency, assuming the random fractional distribution of D-BCP and A-BCP, along with the random spatial distribution of pyrene and perylene within the micellar core. The observed FRET efficiency suggested a core radius (Rc) of 0.95R0, where R0 was the Förster critical distance. With R0 calculated to be 3.2 nm based on Förster theory, Rc was determined to be approximately 3.0 nm, aligning closely with the dried-out core radius estimated from aggregation number and polystyrene density. This stochastic FRET methodology was further applied to investigate the swelling behavior of the polymer micelles in a mixture of N,N-dimethylformamide (DMF) and water. Dynamic light scattering analysis revealed minimal change in core dimension below 60 vol % DMF content. However, FRET analysis provided a deeper insight, showing an increase in core radius with DMF content up to 20 vol %, followed by saturation up to 50 vol %, offering a more comprehensive understanding of the micelle swelling behavior.
Collapse
Affiliation(s)
- Makoto Obata
- Graduate Faculty of Interdisciplinary Research, University of Yamanashi, 4-4-37 Takeda, Kofu 400-8510, Japan
| | - Shougo Yamaguchi
- Graduate Faculty of Interdisciplinary Research, University of Yamanashi, 4-4-37 Takeda, Kofu 400-8510, Japan
| | - Tomokazu Yoshimura
- Department of Chemistry, Faculty of Science, Nara Women's University, Kitauoyanishi-machi, Nara 630-8506, Japan
| |
Collapse
|
6
|
Yildirim T, Bali A, Koch M, Paul P, Latta L, Schneider-Daum N, Gallei M, Lehr CM. A New Class of Polyion Complex Vesicles (PIC-somes) to Improve Antimicrobial Activity of Tobramycin in Pseudomonas Aeruginosa Biofilms. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401926. [PMID: 38829185 DOI: 10.1002/smll.202401926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Pseudomonas aeruginosa (PA) is a major healthcare concern due to its tolerance to antibiotics when enclosed in biofilms. Tobramycin (Tob), an effective cationic aminoglycoside antibiotic against planktonic PA, loses potency within PA biofilms due to hindered diffusion caused by interactions with anionic biofilm components. Loading Tob into nano-carriers can enhance its biofilm efficacy by shielding its charge. Polyion complex vesicles (PIC-somes) are promising nano-carriers for charged drugs, allowing higher drug loadings than liposomes and polymersomes. In this study, a new class of nano-sized PIC-somes, formed by Tob-diblock copolymer complexation is presented. This approach replaces conventional linear PEG with brush-like poly[ethylene glycol (methyl ether methacrylate)] (PEGMA) in the shell-forming block, distinguishing it from past methods. Tob paired with a block copolymer containing hydrophilic PEGMA induces micelle formation (PIC-micelles), while incorporating hydrophobic pyridyldisulfide ethyl methacrylate (PDSMA) monomer into PEGMA chains reduces shell hydrophilicity, leads to the formation of vesicles (PIC-somes). PDSMA unit incorporation enables unprecedented dynamic disulfide bond-based shell cross-linking, significantly enhancing stability under saline conditions. Neither PIC-somes nor PIC-micelles show any relevant cytotoxicity on A549, Calu-3, and dTHP-1 cells. Tob's antimicrobial efficacy against planktonic PA remains unaffected after encapsulation into PIC-somes and PIC-micelles, but its potency within PA biofilms significantly increases.
Collapse
Affiliation(s)
- Turgay Yildirim
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
| | - Aghiad Bali
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Pascal Paul
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
| | - Lorenz Latta
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
| | - Nicole Schneider-Daum
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
| | - Markus Gallei
- Polymer Chemistry, Saarland University, Campus C4 2, 66123, Saarbrücken, Germany
- Saarene - Saarland Center for Energy Materials and Sustainability, Campus C4 2, 66123, Saarbrücken, Germany
| | - Claus-Michael Lehr
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| |
Collapse
|
7
|
Boase NRB, Gillies ER, Goh R, Kieltyka RE, Matson JB, Meng F, Sanyal A, Sedláček O. Stimuli-Responsive Polymers at the Interface with Biology. Biomacromolecules 2024; 25:5417-5436. [PMID: 39197109 DOI: 10.1021/acs.biomac.4c00690] [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: 08/30/2024]
Abstract
There has been growing interest in polymeric systems that break down or undergo property changes in response to stimuli. Such polymers can play important roles in biological systems, where they can be used to control the release of therapeutics, modulate imaging signals, actuate movement, or direct the growth of cells. In this Perspective, after discussing the most important stimuli relevant to biological applications, we will present a selection of recent exciting developments. The growing importance of stimuli-responsive polysaccharides will be discussed, followed by a variety of stimuli-responsive polymeric systems for the delivery of small molecule drugs and nucleic acids. Switchable polymers for the emerging area of therapeutic response measurement in theranostics will be described. Then, the diverse functions that can be achieved using hydrogels cross-linked covalently, as well as by various dynamic approaches will be presented. Finally, we will discuss some of the challenges and future perspectives for the field.
Collapse
Affiliation(s)
- Nathan R B Boase
- Centre for Materials Science and School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Elizabeth R Gillies
- Department of Chemistry; Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Rubayn Goh
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Singapore 138634, Singapore
| | - Roxanne E Kieltyka
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, PO Box 9502, Leiden 2300 RA, The Netherlands
| | - John B Matson
- Department of Chemistry and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Amitav Sanyal
- Department of Chemistry and Center for Life Sciences and Technologies, Bogazici University, Bebek, 34342 Istanbul, Türkiye
| | - Ondřej Sedláček
- Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, 128 00 Prague 2, Czech Republic
| |
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 2024:10.1007/s13346-024-01684-w. [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] [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
|
Altinbasak I, Alp Y, Sanyal R, Sanyal A. Theranostic nanogels: multifunctional agents for simultaneous therapeutic delivery and diagnostic imaging. NANOSCALE 2024; 16:14033-14056. [PMID: 38990143 DOI: 10.1039/d4nr01423e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
In recent years, there has been a growing interest in multifunctional theranostic agents capable of delivering therapeutic payloads while facilitating simultaneous diagnostic imaging of diseased sites. This approach offers a comprehensive strategy particularly valuable in dynamically evolving diseases like cancer, where combining therapy and diagnostics provides crucial insights for treatment planning. Nanoscale platforms, specifically nanogels, have emerged as promising candidates due to their stability, tunability, and multifunctionality as carriers. As a well-studied subgroup of soft polymeric nanoparticles, nanogels exhibit inherent advantages due to their size and chemical compositions, allowing for passive and active targeting of diseased tissues. Moreover, nanogels loaded with therapeutic and diagnostic agents can be designed to respond to specific stimuli at the disease site, enhancing their efficacy and specificity. This capability enables fine-tuning of theranostic platforms, garnering significant clinical interest as they can be tailored for personalized treatments. The ability to monitor tumor progression in response to treatment facilitates the adaptation of therapies according to individual patient responses, highlighting the importance of designing theranostic platforms to guide clinicians in making informed treatment decisions. Consequently, the integration of therapy and diagnostics using theranostic platforms continues to advance, offering intelligent solutions to address the challenges of complex diseases such as cancer. In this context, nanogels capable of delivering therapeutic payloads and simultaneously armed with diagnostic modalities have emerged as an attractive theranostic platform. This review focuses on advances made toward the fabrication and utilization of theranostic nanogels by highlighting examples from recent literature where their performances through a combination of therapeutic agents and imaging methods have been evaluated.
Collapse
Affiliation(s)
- Ismail Altinbasak
- Department of Chemistry, Bogazici University, Bebek, Istanbul 34342, Türkiye.
| | - Yasin Alp
- Department of Chemistry, Bogazici University, Bebek, Istanbul 34342, Türkiye.
| | - Rana Sanyal
- Department of Chemistry, Bogazici University, Bebek, Istanbul 34342, Türkiye.
- Center for Life Sciences and Technologies, Bogazici University, Bebek, Istanbul 34342, Türkiye
| | - Amitav Sanyal
- Department of Chemistry, Bogazici University, Bebek, Istanbul 34342, Türkiye.
- Center for Life Sciences and Technologies, Bogazici University, Bebek, Istanbul 34342, Türkiye
| |
Collapse
|
10
|
Calik F, Degirmenci A, Maouati H, Sanyal R, Sanyal A. Redox-Responsive "Catch and Release" Cryogels: A Versatile Platform for Capture and Release of Proteins and Cells. ACS Biomater Sci Eng 2024; 10:3017-3028. [PMID: 38655791 DOI: 10.1021/acsbiomaterials.4c00239] [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] [Indexed: 04/26/2024]
Abstract
Macroporous cryogels are attractive scaffolds for biomedical applications, such as biomolecular immobilization, diagnostic sensing, and tissue engineering. In this study, thiol-reactive redox-responsive cryogels with a porous structure are prepared using photopolymerization of a pyridyl disulfide poly(ethylene glycol) methacrylate (PDS-PEG-MA) monomer. Reactive cryogels are produced using PDS-PEG-MA and hydrophilic poly(ethylene glycol) methyl ether methacrylate (PEGMEMA) monomers, along with a PEG-based cross-linker and photoinitiator. Functionalization of cryogels using a fluorescent dye via the disulfide-thiol exchange reactions is demonstrated, followed by release under reducing conditions. For ligand-mediated protein immobilization, first, thiol-containing biotin or mannose is conjugated onto the cryogels. Subsequently, fluorescent dye-labeled proteins streptavidin and concanavalin A (ConA) are immobilized via ligand-mediated conjugation. Furthermore, we demonstrate that the mannose-decorated cryogel could capture ConA selectively from a mixture of lectins. The efficiency of protein immobilization could be easily tuned by changing the ratio of the thiol-sensitive moiety in the scaffold. Finally, an integrin-binding cell adhesive peptide is attached to cryogels to achieve successful attachment, and the on-demand detachment of integrin-receptor-rich fibroblast cells is demonstrated. Redox-responsive cryogels can serve as potential scaffolds for a variety of biomedical applications because of their facile synthesis and modification.
Collapse
Affiliation(s)
- Filiz Calik
- Department of Chemistry, Bogazici University, Istanbul 34342, Türkiye
| | - Aysun Degirmenci
- Department of Chemistry, Bogazici University, Istanbul 34342, Türkiye
| | - Hamida Maouati
- Department of Chemistry, Bogazici University, Istanbul 34342, Türkiye
| | - Rana Sanyal
- Department of Chemistry, Bogazici University, Istanbul 34342, Türkiye
- Center for Life Sciences and Technologies, Bogazici University, Istanbul 34342, Türkiye
| | - Amitav Sanyal
- Department of Chemistry, Bogazici University, Istanbul 34342, Türkiye
- Center for Life Sciences and Technologies, Bogazici University, Istanbul 34342, Türkiye
| |
Collapse
|
11
|
Pal J, Sharma M, Tiwari A, Tiwari V, Kumar M, Sharma A, Hassan Almalki W, Alzarea SI, Kazmi I, Gupta G, Kumarasamy V, Subramaniyan V. Oxidative Coupling and Self-Assembly of Polyphenols for the Development of Novel Biomaterials. ACS OMEGA 2024; 9:19741-19755. [PMID: 38737049 PMCID: PMC11080037 DOI: 10.1021/acsomega.3c08528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 05/14/2024]
Abstract
In recent years, the development of biomaterials from green organic sources with nontoxicity and hyposensitivity has been explored for a wide array of biotherapeutic applications. Polyphenolic compounds have unique structural features, and self-assembly by oxidative coupling allows molecular species to rearrange into complex biomaterial that can be used for multiple applications. Self-assembled polyphenolic structures, such as hollow spheres, can be designed to respond to various chemical and physical stimuli that can release therapeutic drugs smartly. The self-assembled metallic-phenol network (MPN) has been used for modulating interfacial properties and designing biomaterials, and there are several advantages and challenges associated with such biomaterials. This review comprehensively summarizes current challenges and prospects of self-assembled polyphenolic hollow spheres and MPN coatings and self-assembly for biomedical applications.
Collapse
Affiliation(s)
- Jyoti Pal
- Department
of Chemistry and Toxicology, National Forensic
Sciences University, Sector 3 Rohini, Delhi 110085 India
| | - Manu Sharma
- Department
of Chemistry and Toxicology, National Forensic
Sciences University, Sector 3 Rohini, Delhi 110085 India
| | - Abhishek Tiwari
- Pharmacy
Academy, IFTM University, Lodhipur-Rajput, Moradabad, U.P. 244102, India
| | - Varsha Tiwari
- Pharmacy
Academy, IFTM University, Lodhipur-Rajput, Moradabad, U.P. 244102, India
| | - Manish Kumar
- Department
of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab 142001, India
| | - Ajay Sharma
- School of
Pharmaceutical Sciences, Delhi Pharmaceutical
Sciences and Research University, Pushp Vihar, New Delhi 110017, India
| | - Waleed Hassan Almalki
- Department
of Pharmacology, College of Pharmacy, Umm
Al-Qura University, Makkah 21421, Saudi Arabia
| | - Sami I. Alzarea
- Department
of Pharmacology, College of Pharmacy, Jouf
University, Al-Jouf, Sakaka, 72388, Saudi Arabia
| | - Imran Kazmi
- Department
of Biochemistry, Faculty of Science, King
Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Gaurav Gupta
- Centre for
Global Health Research, Saveetha Medical College, Saveetha Institute
of Medical and Technical Sciences, Saveetha
University, Chennai, Tamil Nadu 602105, India
- School of
Pharmacy, Graphic Era Hill University, Dehradun 248007, India
- School
of Pharmacy, Suresh Gyan Vihar University, Jagatpura, 302017 Jaipur, India
| | - Vinoth Kumarasamy
- Department
of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology
Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
| |
Collapse
|
12
|
Basak S, Mukherjee I, Das TK. Injectable biocompatible RAFT mediated nitroxide nanogels: A robust ROS-reduction antioxidant approach. Colloids Surf B Biointerfaces 2024; 236:113790. [PMID: 38367288 DOI: 10.1016/j.colsurfb.2024.113790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/20/2024] [Accepted: 02/05/2024] [Indexed: 02/19/2024]
Abstract
This work introduces novel nitroxide-based nanogels (NGs) crafted through controlled RAFT (Reversible Addition Fragmentation chain Transfer) polymerization, showcasing over 85% improved shelf-life compared to native superoxide dismutase (SOD) enzymes. These 30-40 nm NGs hold great promise for injectable delivery, effectively reducing foam cell formation and displaying potent antioxidant behavior against various reactive oxygen species (ROS), revolutionizing antioxidant therapy. Featuring a meticulously designed core-shell structure via precise RAFT polymerization, these NGs mimic SOD enzymatic activity with nitroxide-based antioxidants, providing unprecedented defense against ROS. Combining methacrylated 2,2,6,6-Tetramethyl-4-piperidyl methacrylate (PMA) and Glycidyl methacrylate (GMA) monomers with precisely synthesized nitroxyl radicals results in exceptional properties. Validated through comprehensive analytical methods, these NGs exhibit remarkable stability, halting foam cell formation even at high concentrations, and demonstrate notable biocompatibility. Their ability to protect low density lipoprotein (LDL) from oxidation for up to a month positions them at the forefront of combating cardiovascular diseases, especially atherosclerosis. This study pioneers injectable antioxidant therapy, offering an innovative approach to cardiovascular ailments. Targeting narrow plaques signifies a promising intervention, reshaping cardiovascular disease treatments. It highlights the potential of advanced drug delivery in biomedicine, promising more effective cardiovascular disease treatments.
Collapse
Affiliation(s)
- Suman Basak
- Department of Health Technology, DTU Health Tech, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Department of Chemistry, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Ishita Mukherjee
- Department of Inorganic and Physical Chemistry (IPC), Indian Institute of Science (IISc), Bangalore 560012, India
| | - Tushar Kanti Das
- Institute of Physics - Center for Science and Education, Silesian University of Technology, Krasińskiego 8, 40-019 Katowice, Poland.
| |
Collapse
|
13
|
Zhang Y, Zhong A, Min J, Tu H, Cao Y, Fu J, Li Y, Liu X, Yang Y, Wang J, Liu J, Wu M. Biomimetic Responsive Nanoconverters with Immune Checkpoint Blockade Plus Antiangiogenesis for Advanced Hepatocellular Carcinoma Treatment. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6894-6907. [PMID: 38306190 DOI: 10.1021/acsami.3c18140] [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: 02/04/2024]
Abstract
The first-line treatment for advanced hepatocellular carcinoma (HCC) combines immune checkpoint inhibitors and antiangiogenesis agents to prolong patient survival. Nonetheless, this approach has several limitations, including stringent inclusion criteria and suboptimal response rates that stem from the severe off-tumor side effects and the unfavorable pharmacodynamics and pharmacokinetics of different drugs delivered systemically. Herein, we propose a single-agent smart nanomedicine-based approach that mimics the therapeutic schedule in a targeted and biocompatible manner to elicit robust antitumor immunity in advanced HCC. Our strategy employed pH-responsive carriers, poly(ethylene glycol)-poly(β-amino esters) amphiphilic block copolymer (PEG-PAEs), for delivering apatinib (an angiogenesis inhibitor), that were surface-coated with plasma membrane derived from engineered cells overexpressing PD-1 proteins (an immune checkpoint inhibitor to block PD-L1). In an advanced HCC mouse model with metastasis, these biomimetic responsive nanoconverters induced significant tumor regression (5/9), liver function recovery, and complete suppression of lung metastasis. Examination of the tumor microenvironment revealed an increased infiltration of immune effector cells (CD8+ and CD4+ T cells) and reduced immunosuppressive cells (myeloid-derived suppressor cells and T regulatory cells) in treated tumors. Importantly, our nanomedicine selectively accumulated in both small and large HCC occupying >50% of the liver volume to exert therapeutic effects with minimal systemic side effects. Overall, these findings highlight the potential of such multifunctional nanoconverters to effectively reshape the tumor microenvironment for advanced HCC treatment.
Collapse
Affiliation(s)
- Yuting Zhang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Aoxue Zhong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Juan Min
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P. R. China
| | - Haibin Tu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Yanbing Cao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Jinghao Fu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Yonghao Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Yong Yang
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
| | - Jianmin Wang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
| | - Jingfeng Liu
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| |
Collapse
|
14
|
Liu H, Lu HH, Alp Y, Wu R, Thayumanavan S. Structural Determinants of Stimuli-Responsiveness in Amphiphilic Macromolecular Nano-assemblies. Prog Polym Sci 2024; 148:101765. [PMID: 38476148 PMCID: PMC10927256 DOI: 10.1016/j.progpolymsci.2023.101765] [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] [Indexed: 03/14/2024]
Abstract
Stimuli-responsive nano-assemblies from amphiphilic macromolecules could undergo controlled structural transformations and generate diverse macroscopic phenomenon under stimuli. Due to the controllable responsiveness, they have been applied for broad material and biomedical applications, such as biologics delivery, sensing, imaging, and catalysis. Understanding the mechanisms of the assembly-disassembly processes and structural determinants behind the responsive properties is fundamentally important for designing the next generation of nano-assemblies with programmable responsiveness. In this review, we focus on structural determinants of assemblies from amphiphilic macromolecules and their macromolecular level alterations under stimuli, such as the disruption of hydrophilic-lipophilic balance (HLB), depolymerization, decrosslinking, and changes of molecular packing in assemblies, which eventually lead to a series of macroscopic phenomenon for practical purposes. Applications of stimuli-responsive nano-assemblies in delivery, sensing and imaging were also summarized based on their structural features. We expect this review could provide readers an overview of the structural considerations in the design and applications of nanoassemblies and incentivize more explorations in stimuli-responsive soft matters.
Collapse
Affiliation(s)
- Hongxu Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065 P. R. China
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Hung-Hsun Lu
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Yasin Alp
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Ruiling Wu
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
15
|
López-Iglesias C, Klinger D. Rational Design and Development of Polymeric Nanogels as Protein Carriers. Macromol Biosci 2023; 23:e2300256. [PMID: 37551821 DOI: 10.1002/mabi.202300256] [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: 06/02/2023] [Revised: 07/26/2023] [Indexed: 08/09/2023]
Abstract
Proteins have gained significant attention as potential therapeutic agents owing to their high specificity and reduced toxicity. Nevertheless, their clinical utility is hindered by inherent challenges associated with stability during storage and after in vivo administration. To overcome these limitations, polymeric nanogels (NGs) have emerged as promising carriers. These colloidal systems are capable of efficient encapsulation and stabilization of protein cargoes while improving their bioavailability and targeted delivery. The design of such delivery systems requires a comprehensive understanding of how the synthesis and formulation processes affect the final performance of the protein. This review highlights critical aspects involved in the development of NGs for protein delivery, with specific emphasis on loading strategies and evaluation techniques. For example, factors influencing loading efficiency and release kinetics are discussed, along with strategies to optimize protein encapsulation through protein-carrier interactions to achieve the desired therapeutic outcomes. The discussion is based on recent literature examples and aims to provide valuable insights for researchers working toward the advancement of protein-based therapeutics.
Collapse
Affiliation(s)
- Clara López-Iglesias
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise Straße 2-4, 14195, Berlin, Germany
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, I+D Farma group (GI-1645), Faculty of Pharmacy, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Campus Vida s/n, Santiago de Compostela, 15782, Spain
| | - Daniel Klinger
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise Straße 2-4, 14195, Berlin, Germany
| |
Collapse
|
16
|
Davis MA, Cho E, Teplensky MH. Harnessing biomaterial architecture to drive anticancer innate immunity. J Mater Chem B 2023; 11:10982-11005. [PMID: 37955201 DOI: 10.1039/d3tb01677c] [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: 11/14/2023]
Abstract
Immunomodulation is a powerful therapeutic approach that harnesses the body's own immune system and reprograms it to treat diseases, such as cancer. Innate immunity is key in mobilizing the rest of the immune system to respond to disease and is thus an attractive target for immunomodulation. Biomaterials have widely been employed as vehicles to deliver immunomodulatory therapeutic cargo to immune cells and raise robust antitumor immunity. However, it is key to consider the design of biomaterial chemical and physical structure, as it has direct impacts on innate immune activation and antigen presentation to stimulate downstream adaptive immunity. Herein, we highlight the widespread importance of structure-driven biomaterial design for the delivery of immunomodulatory cargo to innate immune cells. The incorporation of precise structural elements can be harnessed to improve delivery kinetics, uptake, and the targeting of biomaterials into innate immune cells, and enhance immune activation against cancer through temporal and spatial processing of cargo to overcome the immunosuppressive tumor microenvironment. Structural design of immunomodulatory biomaterials will profoundly improve the efficacy of current cancer immunotherapies by maximizing the impact of the innate immune system and thus has far-reaching translational potential against other diseases.
Collapse
Affiliation(s)
- Meredith A Davis
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
| | - Ezra Cho
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
| | - Michelle H Teplensky
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
- Department of Materials Science and Engineering, Boston University, Boston, Massachusetts, 02215, USA
| |
Collapse
|
17
|
Ashwani PV, Gopika G, Arun Krishna KV, Jose J, John F, George J. Stimuli-Responsive and Multifunctional Nanogels in Drug Delivery. Chem Biodivers 2023; 20:e202301009. [PMID: 37718283 DOI: 10.1002/cbdv.202301009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/19/2023]
Abstract
Nanogels represent promising drug delivery systems in the biomedical field, designed to overcome challenges associated with standard treatment approaches. Stimuli-responsive nanogels, often referred to as intelligent materials, have garnered significant attention for their potential to enhance control over properties such as drug release and targeting. Furthermore, researchers have recently explored the application of nanogels in diverse sectors beyond biomedicine including sensing materials, catalysts, or adsorbents for environmental applications. However, to fully harness their potential as practical delivery systems, further research is required to better understand their pharmacokinetic behaviour, interactions between nanogels and bio distributions, as well as toxicities. One promising future application of stimuli-responsive multifunctional nanogels is their use as delivery agents in cancer treatment, offering an alternative to overcome the challenges with conventional approaches. This review discusses various synthetic methods employed in developing nanogels as efficient carriers for drug delivery in cancer treatment. The investigations explore, the key aspects of nanogels, including their multifunctionality and stimuli-responsive properties, as well as associated toxicity concerns. The discussions presented herein aim to provide the readers a comprehensive understanding of the potential of nanogels as smart drug delivery systems in the context of cancer therapy.
Collapse
Affiliation(s)
- P V Ashwani
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - G Gopika
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - K V Arun Krishna
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Josena Jose
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Franklin John
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Jinu George
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| |
Collapse
|
18
|
Princen K, Marien N, Guedens W, Graulus GJ, Adriaensens P. Hydrogels with Reversible Crosslinks for Improved Localised Stem Cell Retention: A Review. Chembiochem 2023; 24:e202300149. [PMID: 37220343 DOI: 10.1002/cbic.202300149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 05/25/2023]
Abstract
Successful stem cell applications could have a significant impact on the medical field, where many lives are at stake. However, the translation of stem cells to the clinic could be improved by overcoming challenges in stem cell transplantation and in vivo retention at the site of tissue damage. This review aims to showcase the most recent insights into developing hydrogels that can deliver, retain, and accommodate stem cells for tissue repair. Hydrogels can be used for tissue engineering, as their flexibility and water content makes them excellent substitutes for the native extracellular matrix. Moreover, the mechanical properties of hydrogels are highly tuneable, and recognition moieties to control cell behaviour and fate can quickly be introduced. This review covers the parameters necessary for the physicochemical design of adaptable hydrogels, the variety of (bio)materials that can be used in such hydrogels, their application in stem cell delivery and some recently developed chemistries for reversible crosslinking. Implementing physical and dynamic covalent chemistry has resulted in adaptable hydrogels that can mimic the dynamic nature of the extracellular matrix.
Collapse
Affiliation(s)
- Ken Princen
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Neeve Marien
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Wanda Guedens
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Geert-Jan Graulus
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Peter Adriaensens
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| |
Collapse
|
19
|
Santra S, Das S, Sengupta A, Molla MR. Tumor acidity-induced surface charge modulation in covalent nanonetworks for activated cellular uptake: targeted delivery of anticancer drugs and selective cancer cell death. Biomater Sci 2023; 11:5549-5559. [PMID: 37401615 DOI: 10.1039/d3bm00491k] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
A β-thioester and tertiary amine based covalently cross-linked nanoassembly coined as a nanonetwork (NN) endowed with dual pH responsive features (tumor acidity induced surface charge modulation and endosomal pH triggered controlled degradation) has been designed and synthesized for stable sequestration and sustained release of drug molecules in response to endosomal pH. An amphiphile integrated with tertiary amine and acrylate (ATA) functionalities was synthesized to fabricate the nanonetwork. This amphiphile showed entropically driven self-assembly and micellar nanostructures (nanoassemblies), which can sequester hydrophobic drug molecules at neutral pH. To further stabilize the nanoassemblies and the sequestered drug molecules even below its critical aggregation concentration (CAC), the micellar core was cross-linked via the thiol-acrylate Michael addition click reaction to generate multiple copies of acid labile β-thioester functionalities in the core, which undergo slow hydrolysis at endosomal pH (∼5.0), thus enabling sustained release of the anti-cancer drug doxorubicin at endosomal pH. The nanonetworks showed a significant decrease in drug leakage compared to the nanoassemblies (NAs), which was also justified by a low leakage coefficient calculated from the fluorescence resonance energy transfer experiment. The NN also exhibited dilution insensitivity and high serum stability, whereas the NA disassembled upon dilution and during serum treatment. The biological evaluation revealed tumor extracellular matrix pH (∼6.4-6.8) induced surface charge modulation and cancer cell (HeLa) selective activated cellular uptake of the doxorubicin loaded nanonetwork (NN-DOX). In contrast, the benign nature of NN-DOX towards normal cells (H9c2) suggests excellent cell specificity. Thus, we believe that the ease of synthesis, nanonetwork fabrication reproducibility, robust stability, smart nature of tumor microenvironment sensitive surface charge modulation, boosted tumoral-cell uptake, and triggered drug release will make this system a potential nanomedicine for chemotherapeutic treatments.
Collapse
Affiliation(s)
- Subrata Santra
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata-700009, India.
| | - Shreya Das
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata-700032, India
| | - Arunima Sengupta
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata-700032, India
| | - Mijanur Rahaman Molla
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata-700009, India.
| |
Collapse
|
20
|
Wu R, Prachyathipsakul T, Zhuang J, Liu H, Han Y, Liu B, Gong S, Qiu J, Wong S, Ribbe A, Medeiros J, Bhagabati J, Gao J, Wu P, Dutta R, Herrera R, Faraci S, Xiao H, Thayumanavan S. Conferring liver selectivity to a thyromimetic using a novel nanoparticle increases therapeutic efficacy in a diet-induced obesity animal model. PNAS NEXUS 2023; 2:pgad252. [PMID: 37649581 PMCID: PMC10465086 DOI: 10.1093/pnasnexus/pgad252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/21/2023] [Indexed: 09/01/2023]
Abstract
Optimization of metabolic regulation is a promising solution for many pathologies, including obesity, dyslipidemia, type 2 diabetes, and inflammatory liver disease. Synthetic thyroid hormone mimics-based regulation of metabolic balance in the liver showed promise but was hampered by the low biocompatibility and harmful effects on the extrahepatic axis. In this work, we show that specifically directing the thyromimetic to the liver utilizing a nanogel-based carrier substantially increased therapeutic efficacy in a diet-induced obesity mouse model, evidenced by the near-complete reversal of body weight gain, liver weight and inflammation, and cholesterol levels with no alteration in the thyroxine (T4) / thyroid stimulating hormone (TSH) axis. Mechanistically, the drug acts by binding to thyroid hormone receptor β (TRβ), a ligand-inducible transcription factor that interacts with thyroid hormone response elements and modulates target gene expression. The reverse cholesterol transport (RCT) pathway is specifically implicated in the observed therapeutic effect. Overall, the study demonstrates a unique approach to restoring metabolic regulation impacting obesity and related metabolic dysfunctions.
Collapse
Affiliation(s)
- Ruiling Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Theeraphop Prachyathipsakul
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jiaming Zhuang
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Hongxu Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Yanhui Han
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Bin Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Shuai Gong
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jingyi Qiu
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Siu Wong
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Alexander Ribbe
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jayashree Bhagabati
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jingjing Gao
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Peidong Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Ranit Dutta
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | | | | | - Hang Xiao
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
21
|
Mondal A, Das S, Ali SM, Kolay S, Sengupta A, Molla MR. Bioderived Lipoic Acid-Based Dynamic Covalent Nanonetworks of Poly(disulfide)s: Enhanced Encapsulation Stability and Cancer Cell-Selective Delivery of Drugs. Bioconjug Chem 2023; 34:489-500. [PMID: 36693213 DOI: 10.1021/acs.bioconjchem.2c00493] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Dynamic covalent poly(disulfide)-based cross-linked nanoaggregates, termed nanonetworks (NNs), endowed with pH- and redox-responsive degradation features have been fabricated for stable noncovalent encapsulation and triggered cargo release in a controlled fashion. A bioderived lipoic acid-based Gemini surfactant-like amphiphilic molecule was synthesized for the preparation of nanoaggregates. It self-assembles by a entropy-driven self-assembly process in aqueous milieu. To further stabilize the self-assembled nanostructure, the core was cross-linked by ring-opening disulfide exchange polymerization (RODEP) of 1,2-dithiolane rings situated inside the core of the nanoaggregates. The cross-linked nanoaggregates, i.e., nanonetwork, are found to be stable in the presence of blood serum, and also, they maintain the self-assembled structure even below the critical aggregation concentration (CAC) as probed by dynamic light scattering (DLS) experiments. The nanonetwork showed almost 50% reduction in guest leakage compared to that of the nanoaggregates as shown by the release profile in the absence of stimuli, suggesting high encapsulation stability as evidenced by the fluorescence resonance energy transfer (FRET) experiment. The decross-linking of the nanonetwork occurs in response to redox and pH stimuli due to disulfide reduction and β-thioester hydrolysis, respectively, thus empowering disassembly-mediated controlled cargo release up to ∼87% for 55 h of incubation. The biological evaluation of the doxorubicin (DOX)-loaded nanonetwork revealed environment-specific surface charge modulation-mediated cancer cell-selective cellular uptake and cytotoxicity. The benign nature of the nanonetwork toward normal cells makes the system very promising in targeted drug delivery applications. Thus, the ease of synthesis, nanonetwork fabrication reproducibility, robust stability, triggered drug release in a controlled fashion, and cell-selective cytotoxicity behavior, we believe, will make the system a potential candidate in the development of robust materials for chemotherapeutic applications.
Collapse
Affiliation(s)
- Arun Mondal
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Shreya Das
- Department of Life Science & Biotechnology, Jadavpur University, 188 R. S. C. M. Road, Jadavpur, Kolkata 700032, India
| | - Sk Mursed Ali
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Soumya Kolay
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Arunima Sengupta
- Department of Life Science & Biotechnology, Jadavpur University, 188 R. S. C. M. Road, Jadavpur, Kolkata 700032, India
| | - Mijanur Rahaman Molla
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| |
Collapse
|
22
|
Liu X, Zheng D, Long Y, Wang L. Highly Robust Nanogels from Thermal-Responsive Nanoparticles with Controlled Swelling for Engineering Deployments. ACS APPLIED MATERIALS & INTERFACES 2023; 15:11175-11184. [PMID: 36799692 DOI: 10.1021/acsami.3c00166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Regular nanogels have been demonstrated their inefficiency for subterranean oil recovery due to their intrinsic drawbacks of fast swelling within minutes, thermal instability, and salinity vulnerability. Prior deployment of swelling delayed nanogels mainly depended on the reservoirs at a relatively higher temperature. To address the issues encountered during engineering deployment, hereinwe devised an integrative approach to in situ form swelling delayed robust nanogels by introducing radically active monomers with thermally sensitive moieties. The nanoparticles with hydrophobic cores in brine in response to thermal input in situ generated well-dispersed hydrophilic nanogels, which showed a pronounced delayed swelling of a week compared to traditional nanogels showing swelling kinetics within minutes. Furthermore, the formation of swelling-delayed nanogels could occur at ambient temperature. This behavior was radically different from that of temperature-controlled labile cross-linkers containing nanogels, requiring temperatures greater than 50 °C for volume increase thanks to ester hydrolysis. In addition, the in-situ formed nanogels displayed long-term thermal stability and salinity tolerance under hostile media at temperatures up to 130 °C. The release of an acidic proton under aqueous conditions has been demonstrated to control the microenvironment for various scenarios. The nanotechnology of converting hydrophobic nanoparticles to hydrophilic nanogels could be applied in a wide range of practical applications such as plugging materials and foaming stabilizers for in-depth conformance control during water and CO2 flooding.
Collapse
Affiliation(s)
- Xing Liu
- Department of Petroleum Engineering, School of Earth Resources, China University of Geosciences, Wuhan 430074, China
| | - Da Zheng
- PetroChina Oil, Gas & New Energies Company, Beijing 100007, China
| | - Yifu Long
- CNPC Research Institute of Engineering Technology, Beijing 102206, China
| | - Lizhu Wang
- Department of Petroleum Engineering, School of Earth Resources, China University of Geosciences, Wuhan 430074, China
| |
Collapse
|
23
|
Mondal B, Padhy A, Maji S, Gupta A, Sen Gupta S. Dual stimuli-responsive cross-linked nanoassemblies from an amphiphilic mannose-6-phosphate based tri-block copolymer for lysosomal membrane permeabilization. Biomater Sci 2023; 11:1810-1827. [PMID: 36655818 DOI: 10.1039/d2bm02110b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Stimuli-responsive cross-linked nanocarriers that can induce lysosomal cell death (LCD) via lysosomal membrane permeabilization (LMP) represent a new class of delivery platforms and have attracted the attention of researchers in the biomedical field. The advantages of such cross-linked nanocarriers are as follows (i) they remain intact during blood circulation; and (ii) they reach the target site via specific receptor-mediated endocytosis leading to the enhancement of therapeutic efficacy and reduction of side effects. Herein, we have synthesized a mannose-6-phosphate (M6P) based amphiphilic ABC type tri-block copolymer having two chains of FDA-approved poly(ε-caprolactone) (PCL) as the hydrophobic block, and poly(S-(o-nitrobenzyl)-L-cysteine) (NBC) acts as the photoresponsive crosslinker block. Two different tri-block copolymers, [(PCL35)2-b-NBC20-b-M6PGP20] and [(PCL35)2-b-NBC15-b-M6PGP20], were synthesized which upon successful self-assembly initially formed spherical uncross-linked "micellar-type" aggregates (UCL-M) and vesicles (UCL-V), respectively. The uncross-linked nanocarriers upon UV treatment for thirty minutes were covalently crosslinked in the middle PNBC block giving rise to the di-sulfide bonds and forming interface cross-linked "micellar-type" aggregates (ICL-M) and vesicles (ICL-V). DLS, TEM, and AFM techniques were used to successfully characterize the morphology of these nanocarriers. The dual stimuli (redox and enzyme) responsiveness of the cross-linked nanocarriers and their trafficking to the lysosome in mammalian cells via receptor-mediated endocytosis was probed using confocal microscopy images. Furthermore, the addition of a chloroquine (CQ, a known lysosomotropic agent) encapsulated cross-linked nanocarrier (CQ@ICL-V) to non-cancerous (HEK-293T) cells and liver (HepG2), and breast cancer cells (MDA-MB-231) was found to initiate lysosomal membrane permeabilization (LMP) followed by lysosomal destabilization which eventually led to lysosomal cell death (LCD). Due to the targeted delivery of CQ to the lysosomes of cancerous cells, almost a 90% smaller amount of CQ was able to achieve similar cell death to CQ alone.
Collapse
Affiliation(s)
- Basudeb Mondal
- Indian Institute of Science Education and Research Kolkata, Department of Chemical Sciences, Mohanpur Campus, Nadia-741246, India.
| | - Abinash Padhy
- Indian Institute of Science Education and Research Kolkata, Department of Chemical Sciences, Mohanpur Campus, Nadia-741246, India.
| | - Saptarshi Maji
- Indian Institute of Science Education and Research Kolkata, Department of Biological Sciences, Mohanpur Campus, Nadia-741246, India
| | - Arnab Gupta
- Indian Institute of Science Education and Research Kolkata, Department of Biological Sciences, Mohanpur Campus, Nadia-741246, India
| | - Sayam Sen Gupta
- Indian Institute of Science Education and Research Kolkata, Department of Chemical Sciences, Mohanpur Campus, Nadia-741246, India.
| |
Collapse
|
24
|
Yang H, Duan Z, Liu F, Zhao Z, Liu S. Cucurbit[7]uril-Based Supramolecular DNA Nanogel for Targeted Codelivery of Chemo/Photodynamic Drugs. ACS Macro Lett 2023; 12:295-301. [PMID: 36779651 DOI: 10.1021/acsmacrolett.2c00763] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Nanodrug delivery systems for the delivery of combination therapeutics have shown their exceptionally potential clinical application by facilitating better synergistic anticancer effects. Herein, we developed a universal strategy to fabricate supramolecular DNA nanogels from DNA tetrahedron skeleton and cucurbit[7]uril-based host-guest interaction for codelivery the chemo and photodynamic therapy drugs. The constructed supramolecular DNA nanogels showed the size tunability, host-guest competition and DNA enzyme responsibility. The cell uptake and MTT experiments demonstrated that the nanogel has excellent biocompatibility and specificity, and achieved the enrichment and slow release of drug in cells. Finally, the combined chemo/photodynamic therapy was realized by coloading doxorubicin hydrochloride and methylene blue. It was proven to be a better stragety to promote apoptosis of cancer cells compared to single chemotherapy or photodynamic therapy. These results suggest that our proposed supramolecular nanogels have provided an effective nanoplatform for drug delivery in the combinational therapy for cancer.
Collapse
Affiliation(s)
- Hai Yang
- Key Laboratory of Hubei Province for Coal Conversion and New Carbon Materials, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Zongze Duan
- Key Laboratory of Hubei Province for Coal Conversion and New Carbon Materials, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Fengbo Liu
- Key Laboratory of Hubei Province for Coal Conversion and New Carbon Materials, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Zhiyong Zhao
- Key Laboratory of Hubei Province for Coal Conversion and New Carbon Materials, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China.,Institute of Advanced Materials and Nanotechnology, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Simin Liu
- Key Laboratory of Hubei Province for Coal Conversion and New Carbon Materials, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China.,Institute of Advanced Materials and Nanotechnology, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
25
|
Huynh U, Wu P, Qiu J, Prachyathipsakul T, Singh K, Jerry DJ, Gao J, Thayumanavan S. Targeted Drug Delivery Using a Plug-to-Direct Antibody-Nanogel Conjugate. Biomacromolecules 2023; 24:849-857. [PMID: 36639133 PMCID: PMC9928872 DOI: 10.1021/acs.biomac.2c01269] [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: 01/15/2023]
Abstract
Targeted drug delivery using antibody-drug conjugates has attracted great attention due to its enhanced therapeutic efficacy compared to traditional chemotherapy. However, the development has been limited due to a low drug-to-antibody ratio and laborious linker-payload optimization. Herein, we present a simple and efficient strategy to combine the favorable features of polymeric nanocarriers with antibodies to generate an antibody-nanogel conjugate (ANC) platform for targeted delivery of cytotoxic agents. Our nanogels stably encapsulate several chemotherapeutic agents with a wide range of mechanisms of action and solubility. We showcase the targetability of ANCs and their selective killing of cancer cells over-expressing disease-relevant antigens such as human epidermal growth factor receptor 2, epidermal growth factor receptor, and tumor-specific mucin 1, which cover a broad range of breast cancer cell types while maintaining low to no toxicity to non-targeted cells. Overall, our system represents a versatile approach that could impact next-generation nanomedicine in antibody-targeted therapeutics.
Collapse
Affiliation(s)
- Uyen Huynh
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Peidong Wu
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jingyi Qiu
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | | | - Khushboo Singh
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - D. Joseph Jerry
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jingjing Gao
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
26
|
Balas M, Iconaru SL, Dinischiotu A, Buton N, Predoi D. Response of the Endogenous Antioxidant Defense System Induced in RAW 264.7 Macrophages upon Exposure to Dextran-Coated Iron Oxide Nanoparticles. Pharmaceutics 2023; 15:552. [PMID: 36839874 PMCID: PMC9967892 DOI: 10.3390/pharmaceutics15020552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
Presently, iron oxide nanoparticles are the only ones approved for clinical use as contrast agents in magnetic resonance imaging (MRI). Even though there is a high demand for these types of nanoparticles both for clinical use as well as for research, there are difficulties in obtaining stable nanoparticles with reproducible properties. In this context, in this study, we report the obtaining by an adapted coprecipitation method of dextran-coated maghemite nanoparticles (ɤ-Fe2O3 NPs). The morphology and structure of the dextran-coated maghemite nanoparticles (ɤ-Fe2O3 NPs) were determined using scanning electron microscopy (SEM) and transmission electron microscopy (TEM). The TEM and SEM micrographs highlighted the obtaining of particles of nanometric size and spherical shape morphology. Furthermore, the high-resolution transmission electron microscopy (HRTEM), as well as selected area diffraction (SAED), revealed that the obtained samples presented the structure of cubic maghemite. In this study, we also explored the effects of the co-precipitation synthesized dextran-coated maghemite nanoparticles (ɤ-Fe2O3 NPs) on the redox status of macrophages. For cytotoxicity evaluation of these NPs, murine macrophages (RAW 264.7 cell line) were exposed to different concentrations of dextran-coated maghemite nanoparticles (ɤ-Fe2O3 NPs) corresponding to 0-500 μg Fe3+/mL and incubated for 24, 48, and 72 h. Intracellular iron uptake, changes in the oxidative stress parameters (reactive oxygen species production and malondialdehyde level), and the activity of antioxidant enzymes, as well as GSH concentration in cells, were evaluated after incubation with a lower (50 μg Fe3+/mL) and higher (500 μg Fe3+/mL) dose of NPs. The results indicated a significant decrease in RAW 264.7 cell viability after 72 h in the presence of NPs at concentrations above 25 μg Fe3+/mL. An important accumulation of NPs, dependent on dose and exposure time, was detected in macrophages, but it induced only a limited raise in the oxidative status. We showed here that the antioxidant capacity of RAW 264.7 macrophages was efficient in counteracting dextran-coated maghemite nanoparticles (ɤ-Fe2O3 NPs) toxicity even at higher doses.
Collapse
Affiliation(s)
- Mihaela Balas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania
| | | | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania
| | - Nicolas Buton
- National Institute of Materials Physics, 405A Atomistilor Street, 077125 Magurele, Romania
| | - Daniela Predoi
- HORIBA Jobin Yvon S.A.S., 6-18, Rue du Canal, CEDEX, 91165 Longjumeau, France
| |
Collapse
|
27
|
New Insights into the Biological Response Triggered by Dextran-Coated Maghemite Nanoparticles in Pancreatic Cancer Cells and Their Potential for Theranostic Applications. Int J Mol Sci 2023; 24:ijms24043307. [PMID: 36834718 PMCID: PMC9965009 DOI: 10.3390/ijms24043307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Iron oxide nanoparticles are one of the most promising tools for theranostic applications of pancreatic cancer due to their unique physicochemical and magnetic properties making them suitable for both diagnosis and therapy. Thus, our study aimed to characterize the properties of dextran-coated iron oxide nanoparticles (DIO-NPs) of maghemite (γ-Fe2O3) type synthesized by co-precipitation and to investigate their effects (low-dose versus high-dose) on pancreatic cancer cells focusing on NP cellular uptake, MR contrast, and toxicological profile. This paper also addressed the modulation of heat shock proteins (HSPs) and p53 protein expression as well as the potential of DIO-NPs for theranostic purposes. DIO-NPs were characterized by X-ray diffraction (XRD), transmission electron microscopy (TEM), dynamic light scattering analyses (DLS), and zeta potential. Pancreatic cancer cells (PANC-1 cell line) were exposed to different doses of dextran-coated ɣ-Fe2O3 NPs (14, 28, 42, 56 μg/mL) for up to 72 h. The results revealed that DIO-NPs with a hydrodynamic diameter of 16.3 nm produce a significant negative contrast using a 7 T MRI scanner correlated with dose-dependent cellular iron uptake and toxicity levels. We showed that DIO-NPs are biocompatible up to a concentration of 28 μg/mL (low-dose), while exposure to a concentration of 56 μg/mL (high-dose) caused a reduction in PANC-1 cell viability to 50% after 72 h by inducing reactive oxygen species (ROS) production, reduced glutathione (GSH) depletion, lipid peroxidation, enhancement of caspase-1 activity, and LDH release. An alteration in Hsp70 and Hsp90 protein expression was also observed. At low doses, these findings provide evidence that DIO-NPs could act as safe platforms in drug delivery, as well as antitumoral and imaging agents for theranostic uses in pancreatic cancer.
Collapse
|
28
|
Makhathini SS, Mdanda S, Kondiah PJ, Kharodia ME, Rumbold K, Alagidede I, Pathak Y, Bulbulia Z, Rants’o TA, Kondiah PPD. Biomedicine Innovations and Its Nanohydrogel Classifications. Pharmaceutics 2022; 14:2839. [PMID: 36559335 PMCID: PMC9787506 DOI: 10.3390/pharmaceutics14122839] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
As one of the most cutting-edge and promising polymer crosslinked network nanoparticle systems. Polymer nano-sized hydrogels (nanogels) have been a hot topic in the biomedical field over the last few decades. Due to their unique characteristics, which include their relatively high drug encapsulation efficiency, ease of preparation, high tunability, low toxicity, high stability in serum and responsive behavior to a range of stimuli to facilitate drug release. Nanogels are thought to be the next generation of drug delivery systems that can completely change the way that drug delivery systems have an impact on patients' lives. Nanogels have demonstrated significant potential in a variety of fields, including chemotherapy, diagnosis, organ targeting, and delivery of bioactive molecules of different dimensions. However, the lack of substantial clinical data from nanogels becomes one of the major barriers to translating the nanogel concept into a practical therapeutic application for many disease conditions. In addition, nanogel safety profiles have been the major concern that hinders it advancement to the clinical trial phase. This review aims to emphasize the unique properties of nanogels as delivery systems for a variety of bioactive molecules over other nano-delivery systems. Also, this review attempts to give insight into the recent progress in nanogels as a carrier in the field of nanomedicine to overcome complex biological barriers. Relevant scientific data and clinical rationale for the development and the potential use of nanogel as a carrier for targeted therapeutic interventions are discussed. Finally, the concluding points of this review highlight the importance of understanding the long-term toxicity profile of nanogel within the biological system to fully understand their biocompatibility.
Collapse
Affiliation(s)
- Sifiso S. Makhathini
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Sipho Mdanda
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Pariksha J. Kondiah
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Moosa E. Kharodia
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Karl Rumbold
- FH Campus Wien, University of Applied Sciences, Vienna, Höchstädtpl. 6, 1200 Wien, Austria
| | - Imhotep Alagidede
- Simon Diedong Dombo University of Business and Integrated Development Studies, Bamahu Box WA64 Wa, Upper West Region, Ghana
- Wits Business School, University of the Witwatersrand, 2 St Davids Pl &, St Andrew Rd, Parktown, Johannesburg 2193, South Africa
| | - Yashwant Pathak
- USF Health Taneja College of Pharmacy, University of South Florida, 12901 Bruce B Downs Blvd, MDC 030, Tampa, FL 33612-4749, USA
- Faculty of Pharmacy, Airlangga University, Surabaya 60115, East Java, Indonesia
| | - Zain Bulbulia
- Policy Research & Advisory Services Branch, Gauteng Office of Premier, 1 Central Place 30 Rahima Moosa Street Newtown, Johannesburg 2113, South Africa
| | - Thankhoe A. Rants’o
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Pierre P. D. Kondiah
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown, Johannesburg 2193, South Africa
- USF Health Taneja College of Pharmacy, University of South Florida, 12901 Bruce B Downs Blvd, MDC 030, Tampa, FL 33612-4749, USA
- Pearson College London Alumni (Pearson plc), London WC1V 7BH, UK
| |
Collapse
|
29
|
Kolay S, Mondal A, Ali SM, Santra S, Molla MR. Photoswitchable polyurethane based nanoaggregates for on-command release of noncovalent guest molecules. JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2022. [DOI: 10.1080/10601325.2022.2132168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Soumya Kolay
- Department of Chemistry, University of Calcutta, Kolkata, India
| | - Arun Mondal
- Department of Chemistry, University of Calcutta, Kolkata, India
| | - Sk. Mursed Ali
- Department of Chemistry, University of Calcutta, Kolkata, India
| | - Subrata Santra
- Department of Chemistry, University of Calcutta, Kolkata, India
| | | |
Collapse
|
30
|
Cyclodextrin-containing redox-responsive nanogels: Fabrication of a modular targeted drug delivery system. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
31
|
Precise Design Strategies of Nanotechnologies for Controlled Drug Delivery. J Funct Biomater 2022; 13:jfb13040188. [PMID: 36278656 PMCID: PMC9590086 DOI: 10.3390/jfb13040188] [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: 09/19/2022] [Revised: 10/03/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
Rapid advances in nanotechnologies are driving the revolution in controlled drug delivery. However, heterogeneous barriers, such as blood circulation and cellular barriers, prevent the drug from reaching the cellular target in complex physiologic environments. In this review, we discuss the precise design of nanotechnologies to enhance the efficacy, quality, and durability of drug delivery. For drug delivery in vivo, drugs loaded in nanoplatforms target particular sites in a spatial- and temporal-dependent manner. Advances in stimuli-responsive nanoparticles and carbon-based drug delivery platforms are summarized. For transdermal drug delivery systems, specific strategies including microneedles and hydrogel lead to a sustained release efficacy. Moreover, we highlight the current limitations of clinical translation and an incentive for the future development of nanotechnology-based drug delivery.
Collapse
|
32
|
Kusmus DNM, van Veldhuisen TW, Khan A, Cornelissen JJLM, Paulusse JMJ. Uniquely sized nanogels via crosslinking polymerization. RSC Adv 2022; 12:29423-29432. [PMID: 36320766 PMCID: PMC9562763 DOI: 10.1039/d2ra04123e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/21/2022] [Indexed: 12/31/2022] Open
Abstract
Nanogels are very promising carriers for nanomedicine, as they can be prepared in the favorable nanometer size regime, can be functionalized with targeting agents and are responsive to stimuli, i.e. temperature and pH. This induces shrinking or swelling, resulting in controlled release of a therapeutic cargo. Our interest lies in the controlled synthesis of functional nanogels, such as those containing epoxide moieties, that can be subsequently functionalized. Co-polymerization of glycidyl methacrylate and a bifunctional methacrylate crosslinker under dilute conditions gives rise to well-defined epoxide-functional nanogels, of which the sizes are controlled by the degree of polymerization. Nanogels with well-defined sizes (polydispersity of 0.2) ranging from 38 nm to 95 nm were prepared by means of controlled radical polymerization. The nanogels were characterized in detail by FT-IR, DLS, size exclusion chromatography, NMR spectroscopy, AFM and TEM. Nucleophilic attack with functional thiols or amines on the least hindered carbon of the epoxide provides water-soluble nanogels, without altering the backbone structure, while reaction with sodium azide provides handles for further functionalization via click chemistry.
Collapse
Affiliation(s)
- Disraëli N. M. Kusmus
- MESA+ Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Department of Biomolecular Nanotechnology, University of TwenteDrienerlolaan 57522EnschedeNBNetherlands
| | - Thijs W. van Veldhuisen
- MESA+ Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Department of Biomolecular Nanotechnology, University of TwenteDrienerlolaan 57522EnschedeNBNetherlands
| | - Anzar Khan
- Korea University145 Anam-ro, Anam-dongSeoulSeongbuk-guKorea
| | - Jeroen J. L. M. Cornelissen
- MESA+ Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Department of Biomolecular Nanotechnology, University of TwenteDrienerlolaan 57522EnschedeNBNetherlands
| | - Jos M. J. Paulusse
- MESA+ Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Department of Biomolecular Nanotechnology, University of TwenteDrienerlolaan 57522EnschedeNBNetherlands
| |
Collapse
|
33
|
Lee CG, Lee C, Lee J, Nam JS, Kim B, Kwon T. Dual‐Modulated Release of a Cytotoxic Photosensitizer Using Photogenerated Reactive Oxygen Species and Glutathione. Angew Chem Int Ed Engl 2022; 61:e202210623. [DOI: 10.1002/anie.202210623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Chae Gyu Lee
- Department of Chemistry Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Center for Wave Energy Materials Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| | - Chaiheon Lee
- Department of Chemistry Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Center for Wave Energy Materials Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| | - Joonhee Lee
- Department of Chemistry Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Department of Chemistry Yonsei University Seoul 03722 Republic of Korea
| | - Jung Seung Nam
- Department of Chemistry Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Center for Wave Energy Materials Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Institute for Cancer Genetics Department of Genetics and Development Columbia University Irving Medical Center New York NY 10032 USA
- Herbert Irving Comprehensive Cancer Center Columbia University Irving Medical Center New York NY 10032 USA
| | - Byeong‐Su Kim
- Department of Chemistry Yonsei University Seoul 03722 Republic of Korea
| | - Tae‐Hyuk Kwon
- Department of Chemistry Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Center for Wave Energy Materials Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| |
Collapse
|
34
|
Kanjilal P, Dutta K, Thayumanavan S. Thiol-Disulfide Exchange as a Route for Endosomal Escape of Polymeric Nanoparticles. Angew Chem Int Ed Engl 2022; 61:e202209227. [PMID: 35866880 PMCID: PMC9452476 DOI: 10.1002/anie.202209227] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Indexed: 11/08/2022]
Abstract
Endosomal entrapment has remained the major bottleneck for cytosolic delivery of nanoparticle-based delivery systems. Uncovering fundamentally new pathways for endosomal escape is therefore highly sought. Herein, we report that disulfide bonds can enhance endosomal escape through contacts with cellular exofacial thiols, in addition to facilitating cellular uptake. Our results are supported through comparative analysis of polymeric nanogels with variable accessibility to disulfide bonds by placing these functionalities at the core or the shell of the nanogels. The findings here inform future chemical design of delivery vehicles.
Collapse
Affiliation(s)
- Pintu Kanjilal
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, United States
| | - Kingshuk Dutta
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, United States
| | - S Thayumanavan
- Department of Chemistry, Department of Biomedical Engineering, Molecular and Cellular Biology Program, and Centre for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA 01003, United States
| |
Collapse
|
35
|
Bai X, Sun Q, Cui H, Guerzoni LPB, Wuttke S, Kiessling F, De Laporte L, Lammers T, Shi Y. Controlled Covalent Self-Assembly of a Homopolymer for Multiscale Materials Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109701. [PMID: 35906820 DOI: 10.1002/adma.202109701] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 07/04/2022] [Indexed: 06/15/2023]
Abstract
Polymer self-assembly is a crucial process in materials engineering. Currently, almost all polymer self-assembly is limited to non-covalent bonding methods, even though these methods have drawbacks as they require complicated synthesis techniques and produce relatively unstable structures. Here, a novel mechanism of covalent polymer self-assembly is discovered and employed to address drawbacks of non-covalent polymer self-assembly. A simple ketone homopolymer is found to self-assemble into nano- to macroscale hydrogels during covalent crosslinking. In contrast to non-covalent self-assembly, the covalent self-assembly is independent of and unaffected by solvent conditions (e.g., polarity and ionic strength) and does not require additional agents, e.g., organic solvents and surfactants. The covalent polymer self-assembly is subjected to a new mechanism of control by tuning the covalent crosslinking rate. This leads to nanogels with an unprecedented and tightly controlled range of dimensions from less than 10 nm to above 100 nm. Moreover, the crosslinking rate also regulates the assembly behavior of microgels fabricated by microfluidics. The microgels self-assemble into granular fibers, which is 3D printed into stable porous scaffolds. The novel covalent polymer assembly method has enormous potential to revolutionize multiscale materials fabrication for applications in drug delivery, tissue engineering, and many other fields.
Collapse
Affiliation(s)
- Xiangyang Bai
- Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
| | - Qingxue Sun
- Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
| | - Hao Cui
- Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
| | - Luis P B Guerzoni
- DWI-Leibniz Institute for Interactive Materials, 52074, Aachen, Germany
| | - Stefan Wuttke
- BCMaterials, Basque Center for Materials, UPV/EHU Science Park, Leioa, 48940, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, 48013, Spain
| | - Fabian Kiessling
- Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
| | - Laura De Laporte
- DWI-Leibniz Institute for Interactive Materials, 52074, Aachen, Germany
- Institute of Applied Medical Engineering, Department of Advanced Materials for Biomedicine, RWTH Aachen University, 52074, Aachen, Germany
- Institute for Technical and Macromolecular Chemistry, RWTH Aachen University, 52074, Aachen, Germany
| | - Twan Lammers
- Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
- Department of Pharmaceutics, Utrecht University, Utrecht, 3584 CG, The Netherlands
- Department of Targeted Therapeutics, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Yang Shi
- Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
| |
Collapse
|
36
|
Lee CG, Lee C, Lee J, Nam JS, Kim BS, Kwon TH. Dual‐Modulated Release of a Cytotoxic Photosensitizer Using Photogenerated Reactive Oxygen Species and Glutathione. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202210623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Chae Gyu Lee
- Ulsan National Institute of Science and Technology Department of Chemistry KOREA, REPUBLIC OF
| | - Chaiheon Lee
- Ulsan National Institute of Science and Technology Department of Chemistry KOREA, REPUBLIC OF
| | - Joonhee Lee
- Ulsan National Institute of Science and Technology Department of Chemistry KOREA, REPUBLIC OF
| | - Jung Seung Nam
- Ulsan National Institute of Science and Technology Department of Chemistry KOREA, REPUBLIC OF
| | - Byeong-Su Kim
- Yonsei University Department of Chemistry KOREA, REPUBLIC OF
| | - Tae-Hyuk Kwon
- Ulsan National Institute of Science and Technology Department of Chemistry KOREA, REPUBLIC OF
| |
Collapse
|
37
|
Kanjilal P, Dutta K, Thayumanavan S. Thiol‐Disulfide Exchange as a Route for Endosomal Escape of Polymeric Nanoparticles. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202209227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Pintu Kanjilal
- University of Massachusetts Amherst chemistry UNITED STATES
| | - Kingshuk Dutta
- University of Massachusetts Amherst Chemistry UNITED STATES
| | - Sankaran Thayumanavan
- University of Massachusetts Amherst Department of Chemistry 710 N. Pleasant Street 01003 Amherst UNITED STATES
| |
Collapse
|
38
|
Valdez S, Robertson M, Qiang Z. Fluorescence Resonance Energy Transfer Measurements in Polymer Science: A Review. Macromol Rapid Commun 2022; 43:e2200421. [PMID: 35689335 DOI: 10.1002/marc.202200421] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/06/2022] [Indexed: 12/27/2022]
Abstract
Fluorescence resonance energy transfer (FRET) is a non-invasive characterization method for studying molecular structures and dynamics, providing high spatial resolution at nanometer scale. Over the past decades, FRET-based measurements are developed and widely implemented in synthetic polymer systems for understanding and detecting a variety of nanoscale phenomena, enabling significant advances in polymer science. In this review, the basic principles of fluorescence and FRET are briefly discussed. Several representative research areas are highlighted, where FRET spectroscopy and imaging can be employed to reveal polymer morphology and kinetics. These examples include understanding polymer micelle formation and stability, detecting guest molecule release from polymer host, characterizing supramolecular assembly, imaging composite interfaces, and determining polymer chain conformations and their diffusion kinetics. Finally, a perspective on the opportunities of FRET-based measurements is provided for further allowing their greater contributions in this exciting area.
Collapse
Affiliation(s)
- Sara Valdez
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Mark Robertson
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Zhe Qiang
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| |
Collapse
|
39
|
Nguyen NGT, Nguyen XT, Nguyen NH, Luu TXT, Dao XT. Ground solid permanganate oxidative coupling of thiols into symmetrical/unsymmetrical disulfides: selective and improved process. J Sulphur Chem 2022. [DOI: 10.1080/17415993.2022.2083914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
| | - Xuan-Triet Nguyen
- Faculty of Chemistry, University of Science, Ho Chi Minh City, Vietnam
| | - Ngoc-Huy Nguyen
- Faculty of Chemistry, University of Science, Ho Chi Minh City, Vietnam
| | - Thi Xuan Thi Luu
- Faculty of Chemistry, University of Science, Ho Chi Minh City, Vietnam
- Department of Chemistry, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Xuan-Tien Dao
- Faculty of Chemistry, University of Science, Ho Chi Minh City, Vietnam
| |
Collapse
|
40
|
Agrohia DK, Wu P, Huynh U, Thayumanavan S, Vachet RW. Multiplexed Analysis of the Cellular Uptake of Polymeric Nanocarriers. Anal Chem 2022; 94:7901-7908. [PMID: 35612963 DOI: 10.1021/acs.analchem.2c00648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Polymeric nanocarriers (PNCs) are versatile drug delivery vehicles capable of delivering a variety of therapeutics. Quantitatively monitoring their uptake in biological systems is essential for realizing their potential as next-generation delivery systems; however, existing quantification strategies are limited due to the challenges of detecting polymeric materials in complex biological samples. Here, we describe a metal-coded mass tagging approach that enables the multiplexed quantification of the PNC uptake in cells using mass spectrometry (MS). In this approach, PNCs are conjugated with ligands that bind strongly to lanthanide ions, allowing the PNCs to be sensitively quantitated by inductively coupled plasma-MS. The metal-coded tags have little effect on the properties or toxicity of the PNCs, making them biocompatible. We demonstrate that the conjugation of different metals to the PNCs enables the multiplexed analysis of cellular uptake of multiple distinct PNCs at the same time. This multiplexing capability should improve the design and optimization of PNCs by minimizing biological variability and reducing analysis time, effort, and cost.
Collapse
Affiliation(s)
- Dheeraj K Agrohia
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Peidong Wu
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Uyen Huynh
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States.,Center for Bioactive Delivery─Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Richard W Vachet
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States.,Center for Bioactive Delivery─Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| |
Collapse
|
41
|
Jiang Y, Zhou H, Zhao W, Zhang S. ATP-Triggered Drug Release of Self-Assembled 3D DNA Nanostructures for Fluorescence Imaging and Tumor Therapy. Anal Chem 2022; 94:6771-6780. [PMID: 35471011 DOI: 10.1021/acs.analchem.2c00409] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stimulus-responsive materials are ideal carriers for precisely controlled drug delivery due to their high selectivity. However, the complex physiological environment hinders its development in clinical medicine. Here, we aim to design a self-assembled three-dimensional (3D) DNA nanostructure drug delivery system with adenosine-5'-triphosphate (ATP)-triggered drug release for tumor fluorescence imaging analysis and targeted drug delivery. Dox@3D DNA nanostructures are self-assembled by a simple one-pot annealing reaction and embedded with drugs, which are structurally stable but can be induced using high concentrations of ATP in tumor cells to cleave and release drugs rapidly, facilitating the rapid accumulation of drugs in tumors and exerting therapeutic effects, thus effectively avoiding damage to normal tissues. This work demonstrates that 3D DNA nanostructures can be used as efficient drug nanocarriers with promising applications in tumor therapy.
Collapse
Affiliation(s)
- Yao Jiang
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, P. R. China.,Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| | - Huimin Zhou
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| | - Wenjing Zhao
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| | - Shusheng Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| |
Collapse
|
42
|
Kilic Boz R, Aydin D, Kocak S, Golba B, Sanyal R, Sanyal A. Redox-Responsive Hydrogels for Tunable and "On-Demand" Release of Biomacromolecules. Bioconjug Chem 2022; 33:839-847. [PMID: 35446015 PMCID: PMC9121344 DOI: 10.1021/acs.bioconjchem.2c00094] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
In
recent years, stimuli-responsive degradation has emerged as
a desirable design criterion for functional hydrogels to tune the
release of encapsulated payload as well as ensure degradation of the
gel upon completion of its function. Herein, redox-responsive hydrogels
with a well-defined network structure were obtained using a highly
efficient thiol-disulfide exchange reaction. In particular, gelation
occurred upon combining thiol-terminated tetra-arm polyethylene glycol
(PEG) polymers with linear telechelic PEG-based polymers containing
pyridyl disulfide units at their chain ends. Rapid gelation proceeds
with good conversions (>85%) to yield macroporous hydrogels possessing
high water uptake. Furthermore, due to the presence of the disulfide
linkages, the thus-obtained hydrogels can self-heal. The obtained
hydrogels undergo complete degradation when exposed to environments
rich in thiol-containing agents such as dithiothreitol (DTT) and L-glutathione
(GSH). Also, the release profile of encapsulated protein, namely,
bovine serum albumin, can be tuned by varying the molecular weight
of the polymeric precursors. Additionally, it was demonstrated that
complete dissolution of the hydrogel to rapidly release the encapsulated
protein occurs upon treating these hydrogels with DTT. Cytotoxicity
evaluation of the hydrogels and their degradation products indicated
the benign nature of these hydrogels. Additionally, the cytocompatible
nature of these materials was also evident from a live/dead cell viability
assay. One can envision that the facile fabrication and their ability
to degrade on-demand and release their payload will make these benign
polymeric scaffolds attractive for various biomedical applications.
Collapse
Affiliation(s)
- Ruveyda Kilic Boz
- Department of Chemistry, Bogazici University, Istanbul 34342, Turkey
| | - Duygu Aydin
- Department of Chemistry, Bogazici University, Istanbul 34342, Turkey
| | - Salli Kocak
- Department of Chemistry, Bogazici University, Istanbul 34342, Turkey
| | - Bianka Golba
- Department of Chemistry, Bogazici University, Istanbul 34342, Turkey
| | - Rana Sanyal
- Department of Chemistry, Bogazici University, Istanbul 34342, Turkey.,Center for Life Sciences and Technologies, Bogazici University, Istanbul 34342, Turkey
| | - Amitav Sanyal
- Department of Chemistry, Bogazici University, Istanbul 34342, Turkey.,Center for Life Sciences and Technologies, Bogazici University, Istanbul 34342, Turkey
| |
Collapse
|
43
|
Xu J, Abetz V. Synthesis of a Degradable Hydrogel Based on a Graft Copolymer with Unexpected Thermoresponsiveness. MACROMOL CHEM PHYS 2022. [DOI: 10.1002/macp.202200058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Jingcong Xu
- Institute of Physical Chemistry Universität Hamburg Grindelallee 117 Hamburg 20146 Germany
| | - Volker Abetz
- Institute of Physical Chemistry Universität Hamburg Grindelallee 117 Hamburg 20146 Germany
- Institute of Membrane Research Helmholtz‐Zentrum Hereon Max‐Planck‐Straße 1 Geesthacht 21502 Germany
| |
Collapse
|
44
|
Krishnan A, Roy S, Menon S. Amphiphilic Block Copolymers: From Synthesis Including Living Polymerization Methods to Applications in Drug Delivery. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
45
|
Singh K, Canakci M, Kanjilal P, Williams N, Shanthalingam S, Osborne BA, Thayumanavan S. Evaluation of Cellular Targeting by Fab' vs Full-Length Antibodies in Antibody-Nanoparticle Conjugates (ANCs) Using CD4 T-cells. Bioconjug Chem 2022; 33:486-495. [PMID: 35139308 PMCID: PMC9254259 DOI: 10.1021/acs.bioconjchem.2c00024] [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: 11/28/2022]
Abstract
Targeted delivery of chemotherapeutic drugs can improve their therapeutic efficiency by localizing their toxic effects at the diseased site. This is often achieved either by direct conjugation of drugs to antibodies targeting overexpressed receptors on cancer cells (antibody-drug conjugates/ADCs) or by conjugating antibodies to nanoparticles bearing drugs (antibody-nanoparticle conjugates/ANCs). Here, we report a platform for utilizing hinge cysteines on antigen-binding fragment (Fab') of an anti-CD4 antibody for site-specific conjugation to nanoparticles giving rise to anti-CD4 Fab'-nanoparticle conjugates (Fab'-NCs). We demonstrate a convenient route for obtaining functional anti-CD4 Fab' from full-length antibody and examine the targeted delivery efficiencies of anti-CD4 Fab'-NCs vs ANCs for selective delivery to CD4high mT-ALL cells. Our results indicate that higher avidity of full-length anti-CD4 antibody, i.e., protein alone translated to higher binding ability to CD4high mT-ALL cells in comparison with anti-CD4 Fab' alone. However, the targeted delivery efficiency of anti-CD4 Fab'-NCs was comparable to ANCs indicating that the avidity of Fab' is restored in a nanoparticle-conjugate format. Fab'-NCs are equally capable of achieving targeted drug delivery to CD4high T-cells as ANCs and are a versatile alternative to ANCs by offering site-selective modification strategy while retaining their advantages.
Collapse
Affiliation(s)
- Khushboo Singh
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences University of Massachusetts, Amherst, Amherst, Massachusetts 01003, United States
| | - Mine Canakci
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Pintu Kanjilal
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences University of Massachusetts, Amherst, Amherst, Massachusetts 01003, United States
| | - Natalie Williams
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Sudarvili Shanthalingam
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Barbara A Osborne
- Center for Bioactive Delivery, Institute for Applied Life Sciences University of Massachusetts, Amherst, Amherst, Massachusetts 01003, United States
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences University of Massachusetts, Amherst, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| |
Collapse
|
46
|
Shahi S, Roghani-Mamaqani H, Talebi S, Mardani H. Chemical stimuli-induced reversible bond cleavage in covalently crosslinked hydrogels. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214368] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
47
|
Zhu M, Lu D, Milani AH, Mahmoudi N, King SM, Saunders BR. Comparing pH-responsive nanogel swelling in dispersion and inside a polyacrylamide gel using photoluminescence spectroscopy and small-angle neutron scattering. J Colloid Interface Sci 2022; 608:378-385. [PMID: 34626983 DOI: 10.1016/j.jcis.2021.09.163] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/18/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
Nanosized probes that report their changes in dimensions within networks in response to environmental stimuli are potentially important for applications such as drug delivery, load-supporting hydrogels and soft robotics. Recently, we developed a fluorescent pH-responsive nanogel (NG) that used Förster-resonance energy transfer (FRET) to report changes in the probe separation and NG swelling within hydrogels using photoluminescence (PL) spectroscopy. However, FRET cannot measure nanoparticle dimensions and is subject to artefacts. Here, we report the use of small-angle neutron scattering (SANS) to study both the NGs in dispersion and in polyacrylamide (PAAm) gels as a function of pH. We compare the PL and SANS data for both systems and as a function of pH. The SANS data for the dispersed NGs indicate that they have a core-shell structure with a swollen mesh size of ∼1.0 nm. We hypothesized that the NGs inside the PAAm gel would show the same general changes in scattering as the pH is increased, as observed for the dispersed NGs, and this is confirmed by the data. In summary, the data confirm that PL is a suitable (accessible) method for reporting internal environmental changes within gels using NG probes.
Collapse
Affiliation(s)
- Mingning Zhu
- School of Materials, University of Manchester, MSS Tower, Manchester M13 9PL, UK
| | - Dongdong Lu
- School of Materials, University of Manchester, MSS Tower, Manchester M13 9PL, UK
| | - Amir H Milani
- School of Materials, University of Manchester, MSS Tower, Manchester M13 9PL, UK
| | - Najet Mahmoudi
- ISIS Facility, STFC, Rutherford Appleton Laboratory, Harwell Campus, Didcot OX11 0QX, UK
| | - Stephen M King
- ISIS Facility, STFC, Rutherford Appleton Laboratory, Harwell Campus, Didcot OX11 0QX, UK
| | - Brian R Saunders
- School of Materials, University of Manchester, MSS Tower, Manchester M13 9PL, UK
| |
Collapse
|
48
|
Nanohydrogels: Advanced Polymeric Nanomaterials in the Era of Nanotechnology for Robust Functionalization and Cumulative Applications. Int J Mol Sci 2022; 23:ijms23041943. [PMID: 35216058 PMCID: PMC8875080 DOI: 10.3390/ijms23041943] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 12/17/2022] Open
Abstract
In the era of nanotechnology, the synthesis of nanomaterials for advanced applications has grown enormously. Effective therapeutics and functionalization of effective drugs using nano-vehicles are considered highly productive and selectively necessary. Polymeric nanomaterials have shown their impact and influential role in this process. Polymeric nanomaterials in molecular science are well facilitated due to their low cytotoxic behavior, robust functionalization, and practical approach towards in vitro and in vivo therapeutics. This review highlights a brief discussion on recent techniques used in nanohydrogel designs, biomedical applications, and the applied role of nanohydrogels in the construction of advanced therapeutics. We reviewed recent studies on nanohydrogels for their wide applications in building strategies for advantageously controlled biological applications. The classification of polymers is based on their sources of origin. Nanohydrogel studies are based on their polymeric types and their endorsed utilization for reported applications. Nanotechnology has developed significantly in the past decades. The novel and active role of nano biomaterials with amplified aspects are consistently being studied to minimize the deleterious practices and side effects. Here, we put forth challenges and discuss the outlook regarding the role of nanohydrogels, with future perspectives on delivering constructive strategies and overcoming the critical objectives in nanotherapeutic systems.
Collapse
|
49
|
Wu P, Gao J, Prasad P, Dutta K, Kanjilal P, Thayumanavan S. Influence of Polymer Structure and Architecture on Drug Loading and Redox-Triggered Release. Biomacromolecules 2022; 23:339-348. [PMID: 34890192 PMCID: PMC8757658 DOI: 10.1021/acs.biomac.1c01295] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Disulfide cross-linked nanoassemblies have attracted considerable attention as a drug delivery vehicle due to their responsiveness to the natural redox gradient in biology. Fundamentally understanding the factors that influence the drug loading capacity, encapsulation stability, and precise control of the liberation of encapsulated cargo would be profoundly beneficial to redox-responsive materials. Reported herein are block copolymer (BCP)-based self-cross-linked nanogels, which exhibit high drug loading capacity, high encapsulation stability, and controllable release kinetics. BCP nanogels show considerably higher loading capacity and better encapsulation stability than the random copolymer nanogels at micromolar glutathione concentrations. By partially substituting thiol-reactive pyridyl disulfide into the unreactive benzyl or butyl group, we observed opposite effects on the cross-linking process of BCP nanogels. We further studied the redox-responsive cytotoxicity of our drug-encapsulated nanogels in various cancer cell lines.
Collapse
Affiliation(s)
- Peidong Wu
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Jingjing Gao
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Current address: Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA 02115
| | - Priyaa Prasad
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Kingshuk Dutta
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Pintu Kanjilal
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts, USA
- Center for Bioactive Delivery, The Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| |
Collapse
|
50
|
Biocatalytic self-assembled synthetic vesicles and coacervates: From single compartment to artificial cells. Adv Colloid Interface Sci 2022; 299:102566. [PMID: 34864354 DOI: 10.1016/j.cis.2021.102566] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 12/18/2022]
Abstract
Compartmentalization is an intrinsic feature of living cells that allows spatiotemporal control over the biochemical pathways expressed in them. Over the years, a library of compartmentalized systems has been generated, which includes nano to micrometer sized biomimetic vesicles derived from lipids, amphiphilic block copolymers, peptides, and nanoparticles. Biocatalytic vesicles have been developed using a simple bag containing enzyme design of liposomes to multienzymes immobilized multi-vesicular compartments for artificial cell generation. Additionally, enzymes were also entrapped in membrane-less coacervate droplets to mimic the cytoplasmic macromolecular crowding mechanisms. Here, we have discussed different types of single and multicompartment systems, emphasizing their recent developments as biocatalytic self-assembled structures using recent examples. Importantly, we have summarized the strategies in the development of the self-assembled structure to improvise their adaptivity and flexibility for enzyme immobilization. Finally, we have presented the use of biocatalytic assemblies in mimicking different aspects of living cells, which further carves the path for the engineering of a minimal cell.
Collapse
|