Recent advances and perspectives of multifunctional nanogels in biomedical applications

Bicheng Han , Zideng Dai , Hangrong Chen

MEDCOMM - Biomaterials and Applications ›› 2024, Vol. 3 ›› Issue (4) : e104

PDF
MEDCOMM - Biomaterials and Applications ›› 2024, Vol. 3 ›› Issue (4) : e104 DOI: 10.1002/mba2.104
REVIEW ARTICLE

Recent advances and perspectives of multifunctional nanogels in biomedical applications

Author information +
History +
PDF

Abstract

Nanogels (NGs) are considered as a kind of nanoscale hydrogels (<200 nm) endowing with the functions of both nanomaterials and hydrogels. In the last 20 years, NGs have garnered significant attention due to their versatility and adaptability. Herein, a comprehensive overview of the latest advancements and current research status of NGs is provided, with a particular focus on the synthesis strategies involving physical and chemical cross-linking methods, as well as the advantages of NGs in drug loading and responsive release. Based on the diverse design strategies of NGs, four key biomedical applications, including inflammation therapy, regenerative medicine, bioimaging and tumor therapy are further summarized and discussed. Moreover, the existed inherent challenges facing NGs are proposed, while highlighting their potential to revolutionize therapeutic and diagnostic approaches. Finally, we look forward to the further development and promising potentials of NGs in biomedical applications. This review aims to serve as a valuable reference for researchers, providing some insights into the evolving landscape of NGs and their potential in advanced biomedical applications.

Keywords

biomedical applications / multifunctional nanogels / therapy / tumor

Cite this article

Download citation ▾
Bicheng Han, Zideng Dai, Hangrong Chen. Recent advances and perspectives of multifunctional nanogels in biomedical applications. MEDCOMM - Biomaterials and Applications, 2024, 3(4): e104 DOI:10.1002/mba2.104

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

AkiyoshiK, Deguchi S, MoriguchiN, YamaguchiS, Sunamoto J. Self-aggregates of hydrophobized polysaccharides in water -formation and characteristics of nanoparticles. Macromolecules. 1993;26(12):3062-3068.

[2]

SivaramAJ, Rajitha P, MayaS, JayakumarR, Sabitha M. Nanogels for delivery, imaging and therapy. WIREs Nanomed Nanobiotechnol. 2015;7(4):509-533.

[3]

SuhailM, Rosenholm JM, MinhasMU, et al. Nanogels as drug-delivery systems: a comprehensive overview. Ther Delivery. 2019;10(11):697-717.

[4]

SoniG, YadavKS. Nanogels as potential nanomedicine carrier for treatment of cancer: a mini review of the state of the art. Saudi Pharm J. 2016;24(2):133-139.

[5]

ZhangC, ShiX. Hybrid nanogels with unique designs for improved tumor theranostics. Nanomedicine. 2020;15(15):1455-1458.

[6]

PengHS, ChiuDT. Soft fluorescent nanomaterials for biological and biomedical imaging. Chem Soc Rev. 2015;44(14):4699-4722.

[7]

RyuJH, Jiwpanich S, ChackoR, BickertonS, Thayumanavan S. Surface-functionalizable polymer nanogels with facile hydrophobic guest encapsulation capabilities. J Am Chem Soc. 2010;132(24):8246-8247.

[8]

HuiY, YiX, WibowoD, et al. Nanoparticle elasticity regulates phagocytosis and cancer cell uptake. Sci Adv. 2020;6(16):eaaz4316.

[9]

GuoP, LiuD, SubramanyamK, et al. Nanoparticle elasticity directs tumor uptake. Nat Commun. 2018;9(130):130.

[10]

DesaiP, RimalR, FloreaA, et al. Tuning the elasticity of nanogels improves their circulation time by evading immune cells. Angew Chem Int Ed. 2022;61(20):e202116653.

[11]

ZhangL, WangZ, ZhangR, et al. Multi-stimuli-responsive and cell membrane camouflaged aggregation-induced emission nanogels for precise chemo-photothermal synergistic therapy of tumors. ACS Nano. 2023;17(24):25205-25221.

[12]

HuppertsbergA, KapsL, ZhongZ, et al. Squaric ester-based, pH-degradable nanogels: modular nanocarriers for safe, systemic administration of toll-like receptor 7/8 agonistic immune modulators. J Am Chem Soc. 2021;143(26):9872-9883.

[13]

DengP, SunJ, ChenJ, Zou X, LiaoL. Fast responsive photo-switchable dual-color fluorescent cyclodextrin nanogels for cancer cell imaging. Carbohydr Polymers. 2019;210:379-388.

[14]

TheuneLE, Buchmann J, WedepohlS, MolinaM, LauferJ, CalderónM. NIR-and thermo-responsive semi-interpenetrated polypyrrole nanogels for imaging guided combinational photothermal and chemotherapy. J Controlled Release. 2019;311-312:147-161.

[15]

ChackoRT, Ventura J, ZhuangJ, ThayumanavanS. Polymer nanogels: a versatile nanoscopic drug delivery platform. Adv Drug Deliv Rev. 2012;64(9):836-851.

[16]

WangH, GaoL, FanT, et al. Strategic design of intelligent-responsive nanogel carriers for cancer therapy. ACS Appl Mater Interfaces. 2021;13(46):54621-54647.

[17]

RamosJ, Forcada J, Hidalgo-AlvarezR. Cationic polymer nanoparticles and nanogels: from synthesis to biotechnological applications. Chem Rev. 2014;114(1):367-428.

[18]

MolinaM, Asadian-Birjand M, BalachJ, BergueiroJ, MiceliE, CalderónM. Stimuli-responsive nanogel composites and their application in nanomedicine. Chem Soc Rev. 2015;44(17):6161-6186.

[19]

DuanQY, ZhuYX, JiaHR, Wang SH, WuFG. Nanogels: synthesis, properties, and recent biomedical applications. Prog Mater Sci. 2023;139:101167.

[20]

MaricontiM, Dechamboux L, HeckmannM, et al. Intracellular delivery of functional proteins with DNA-protein nanogels-lipids complex. J Am Chem Soc. 2024;146(8):5118-5127.

[21]

JiangT, MaY, XuX, et al. Enzyme-instructed hybrid nanogel/nanofiber oligopeptide hydrogel for localized protein delivery. Acta Pharm Sin B. 2021;11(7):2070-2079.

[22]

ZhaoH, ZhaoY, XuJ, et al. Programmable co-assembly of various drugs with temperature sensitive nanogels for optimizing combination chemotherapy. Chem Eng J. 2020;398:125614.

[23]

SandorfyC. Hydrogen bonding. Science. 1965;147(3660):910-911.

[24]

BawnCEH. The hydrogen bond. Nature. 1940;145(3683):846-848.

[25]

FanW-L, HuangS-Y, YangX-J, Bintang IlhamiF, ChenJ-K, ChengC-C. Hydrogen-bonded cytosine-endowed supramolecular polymeric nanogels: highly efficient cancer cell targeting and enhanced therapeutic efficacy. J Colloid Interface Sci. 2024;665:329-344.

[26]

LeiX, HuQ, GeH, et al. A redox-reactive delivery system via neural stem cell nanoencapsulation enhances white matter regeneration in intracerebral hemorrhage mice. Bioeng Transl Med. 2023;8(2):e10451.

[27]

WeiH, ZhaoZ, WangY, Zou J, LinQ, DuanY. One-step self-assembly of multifunctional DNA nanohydrogels: an enhanced and harmless strategy for guiding combined antitumor therapy. ACS Appl Mater Interfaces. 2019;11(50):46479-46489.

[28]

BaiX, SunQ, CuiH, et al. Controlled covalent self-assembly of a homopolymer for multiscale materials engineering. Adv Mater. 2022;34(39):2109701.

[29]

AbediF, Davaran S, HekmatiM, AkbarzadehA, Baradaran B, MoghaddamSV. An improved method in fabrication of smart dual-responsive nanogels for controlled release of doxorubicin and curcumin in HT-29 colon cancer cells. J Nanobiotechnology. 2021;19(1):18.

[30]

ZhaoY, ChengX, LiJ, et al. Pulmonary delivery of immune checkpoint inhibitors using a responsive polyethylene glycol nanogel for treating lung metastasis. Nano Today. 2023;52:101988.

[31]

ZhuX, HeQ, HeS, et al. Facile manganese ion-coordination assembly of nanogels for synergistic cancer chemo-chemodynamic-immunotherapy. Chem Eng J. 2024;485:149752.

[32]

FroimchukE, CareyST, EdwardsC, Jewell CM. Self-assembly as a molecular strategy to improve immunotherapy. Acc Chem Res. 2020;53(11):2534-2545.

[33]

OmenettoFG, KaplanDL. New opportunities for an ancient material. Science. 2010;329(5991):528-531.

[34]

MasonAF, Buddingh’ BC, WilliamsDS, van HestJCM. Hierarchical self-assembly of a copolymer-stabilized coacervate protocell. J Am Chem Soc. 2017;139(48):17309-17312.

[35]

CaiJ, LiC, KongN, et al. Tailored multifunctional micellar brushes via crystallization-driven growth from a surface. Science. 2019;366(6469):1095-1098.

[36]

FreemanR, HanM, ÁlvarezZ, et al. Reversible self-assembly of superstructured networks. Science. 2018;362(6416):808-813.

[37]

TaoK, MakamP, AizenR, Gazit E. Self-assembling peptide semiconductors. Science. 2017;358(6365):eaam9756.

[38]

DaiZ, LiX, ChenQ, et al. Injectable responsive hydrogel delivery platform: enabling high tissue penetration and sonogenetic-like potentiating anti-tumor immunotherapy. Adv Funct Mater. 2024;34(19):2313723.

[39]

ZhuY, DengX, DaiZ, et al. A “Ferroptosis-Amplifier” hydrogel for eliminating refractory cancer stem cells post-lumpectomy. Nano Lett. 2024;24(26):8179-8188.

[40]

WangX, ChenS, WuD, et al. Oxidoreductase-initiated radical polymerizations to design hydrogels and micro/nanogels: mechanism, molding, and applications. Adv Mater. 2018;30(17):1705668.

[41]

RostovtsevVV, GreenLG, FokinVV, Sharpless KB. A stepwise huisgen cycloaddition process: copper(I)-catalyzed regioselective “ligation” of azides and terminal alkynes. Angew Chem Int Ed. 2002;41(14):2596-2599.

[42]

BoyerC, Corrigan NA, JungK, et al. Copper-mediated living radical polymerization (atom transfer radical polymerization and copper(0) mediated polymerization):from fundamentals to bioapplications. Chem Rev. 2016;116(4):1803-1949.

[43]

ElnaggarYSR, El-Refaie WM, El-MassikMA, AbdallahOY. Lecithin-based nanostructured gels for skin delivery: an update on state of art and recent applications. J Controlled Release. 2014;180:10-24.

[44]

WuY, ChangX, YangG, et al. A physiologically responsive nanocomposite hydrogel for treatment of head and neck squamous cell carcinoma via proteolysis-targeting chimeras enhanced immunotherapy. Adv Mater. 2023;35(12):2210787.

[45]

WuD, WangL, LiW, XuX, JiangW. DNA nanostructure-based drug delivery nanosystems in cancer therapy. Int J Pharm. 2017;533(533):169-178.

[46]

ZetterlundPB, Thickett SC, PerrierS, Bourgeat-LamiE, Lansalot M. Controlled/living radical polymerization in dispersed systems: an update. Chem Rev. 2015;115(18):9745-9800.

[47]

GurnaniP, Perrier S. Controlled radical polymerization in dispersed systems for biological applications. Prog Polym Sci. 2020;102:101209.

[48]

CorriganN, JungK, MoadG, Hawker CJ, MatyjaszewskiK, BoyerC. Reversible-deactivation radical polymerization (Controlled/living radical polymerization):from discovery to materials design and applications. Prog Polym Sci. 2020;111:101311.

[49]

QiM, PanH, ShenH, et al. Nanogel multienzyme mimics synthesized by biocatalytic ATRP and metal coordination for bioresponsive fluorescence imaging. Angew Chem Int Ed. 2020;59(29):11748-11753.

[50]

JayeoyeTJ, Cheewasedtham W, PutsonC, RujiralaiT. Colorimetric determination of sialic acid based on boronic acid-mediated aggregation of gold nanoparticles. Microchim Acta. 2018;185(9):409.

[51]

JayeoyeTJ, Rujiralai T. Green, fabrication of silver/poly(3-aminophenyl boronic acid)/sodium alginate nanogel and hydrogen peroxide sensing capacity. Carbohyd Polym. 2020;246:116657.

[52]

KolbHC, FinnMG, SharplessKB. Click chemistry: diverse chemical function from a few good reactions. Angew Chem Int Ed. 2001;40(11):2004-2021.

[53]

ThirumuruganP, Matosiuk D, JozwiakK. Click chemistry for drug development and diverse chemical-biology applications. Chem Rev. 2013;113(7):4905-4979.

[54]

JiangY, ChenJ, DengC, Suuronen EJ, ZhongZ. Click hydrogels, microgels and nanogels: emerging platforms for drug delivery and tissue engineering. Biomaterials. 2014;35(18):4969-4985.

[55]

NagelG, Sousa-Herves A, WedepohlS, CalderónM. Matrix metalloproteinase-sensitive multistage nanogels promote drug transport in 3D tumor model. Theranostics. 2020;10(1):91-108.

[56]

FanY, Lüchow M, ZhangY, et al. Nanogel encapsulated hydrogels as advanced wound dressings for the controlled delivery of antibiotics. Adv Funct Mater. 2021;31(7):2006453.

[57]

GaoWX, FengHJ, GuoBB, Lu Y, JinGX. Coordination-directed construction of molecular links. Chem Rev. 2020;120(13):6288-6325.

[58]

XuJ, WangJ, YeJ, et al. Metal-coordinated supramolecular self-assemblies for cancer theranostics. Adv Sci. 2021;8(16):e2101101.

[59]

LiuP, QinR, FuG, ZhengN. Surface coordination chemistry of metal nanomaterials. J Am Chem Soc. 2017;139(6):2122-2131.

[60]

KothawadeS, ShendeP. Coordination bonded stimuli-responsive drug delivery system of chemical actives with metal in pharmaceutical applications. Coord Chem Rev. 2024;510:215851.

[61]

CooleyM, SarodeA, HooreM, Fedosov DA, MitragotriS, Sen GuptaA. Influence of particle size and shape on their margination and wall-adhesion: implications in drug delivery vehicle design across nano-to-micro scale. Nanoscale. 2018;10(32):15350-15364.

[62]

XuanL, JuZ, SkoniecznaM, Zhou PK, HuangR. Nanoparticles-induced potential toxicity on human health: applications, toxicity mechanisms, and evaluation models. MedComm. 2023;4(4):e327.

[63]

ZhongX, WeiG, LiuB, et al. Polyhedral oligomeric silsesquioxane-based nanoparticles for efficient chemotherapy of glioblastoma. Small. 2023;19(18):2207248.

[64]

TanQ, ZhaoS, XuT, et al. Inorganic nano-drug delivery systems for crossing the blood–brain barrier: advances and challenges. Coord Chem Rev. 2023;494:215344.

[65]

ZhangM, XuF, CaoJ, et al. Research advances of nanomaterials for the acceleration of fracture healing. Bioactive Mater. 2024;31:368-394.

[66]

van der KoogL, GandekTB, NagelkerkeA. Liposomes and extracellular vesicles as drug delivery systems: a comparison of composition, pharmacokinetics, and functionalization. Adv Healthcare Mater. 2022;11(5):2100639.

[67]

LiY, YeZ, YangH, Xu Q. Tailoring combinatorial lipid nanoparticles for intracellular delivery of nucleic acids, proteins, and drugs. Acta Pharm Sin B. 2022;12(6):2624-2639.

[68]

ZhengM, DuQ, WangX, et al. Tuning the elasticity of polymersomes for brain tumor targeting. Adv Sci. 2021;8(20):2102001.

[69]

JörgensenAM, Steinbring C, StengelD, ToD, SchmidP, Bernkop-SchnürchA. Self-emulsifying drug delivery systems (SEDDS) containing reverse micelles: advanced oral formulations for therapeutic peptides. Adv Healthcare Mater. 2023;12(31):2302034.

[70]

WangJ, LiB, HuangD, et al. Nano-in-Nano dendrimer gel particles for efficient topical delivery of antiglaucoma drugs into the eye. Chem Eng J. 2021;425:130498.

[71]

QinQ, WangM, ZouY, et al. Development of nanoparticle-based drug delivery system for inflammation treatment and diagnosis. MedComm – Biomater Appl. 2023;2(4):e65.

[72]

ShahS, Rangaraj N, LaxmikeshavK, SampathiS. Nanogels as drug carriers – introduction, chemical aspects, release mechanisms and potential applications. Int J Pharm. 2020;581:119268.

[73]

AhmedS, Alhareth K, MignetN. Advancement in nanogel formulations provides controlled drug release. Int J Pharm. 2020;584:119435.

[74]

YallapuMM, ReddyMK, LabhasetwarV. Nanogels: chemistry to drug delivery. In: Labhasetwar V, Leslie-Pelecky DL, eds. Biomedical Applications of Nanotechnology. Wiley; 2007:131-171.

[75]

KhizarS, Alrushaid N, Alam KhanF, et al. Nanocarriers based novel and effective drug delivery system. Int J Pharm. 2023;632:122570.

[76]

SharmaD, SharmaN, PathakM, Agrawala PK, BasuM, OjhaH. Chapter 2 -Nanotechnology-based drug delivery systems: Challenges and opportunities. In: Grumezescu AM, ed., Drug Targeting and Stimuli Sensitive Drug Delivery Systems. William Andrew Publishing; 2018:39-79.

[77]

PremanNK, BarkiRR, VijayanA, Sanjeeva SG, JohnsonRP. Recent developments in stimuli-responsive polymer nanogels for drug delivery and diagnostics: a review. Eur J Pharmaceut Biopharmaceut. 2020;157:121-153.

[78]

KeskinD, ZuG, ForsonAM, Tromp L, SjollemaJ, van RijnP. Nanogels: a novel approach in antimicrobial delivery systems and antimicrobial coatings. Bioactive Materials. 2021;6(10):3634-3657.

[79]

ZhuH, WuJ, ZhaoJ, et al. Dual-functional DNA nanogels for anticancer drug delivery. Acta Biomater. 2024;175:240-249.

[80]

ChenY-B, ZhangY-B, WangY-L, et al. A novel inhalable quercetin-alginate nanogel as a promising therapy for acute lung injury. J Nanobiotechnology. 2022;20(1):272.

[81]

WangY, ZuM, MaX, et al. Glutathione-responsive multifunctional “trojan horse” nanogel as a nanotheranostic for combined chemotherapy and photodynamic anticancer therapy. ACS Appl Mater Interfaces. 2020;12(45):50896-50908.

[82]

YaoJ, LiT, ShiX, WangY, FangS, Wang H. A general prodrug nanohydrogel platform for reduction-triggered drug activation and treatment of taxane-resistant malignancies. Acta Biomater. 2021;130:409-422.

[83]

Rodriguez-AbetxukoA, Romero-Ben E, OntoriaA, et al. Engineered metal-loaded biohybrids to promote the attachment and electron-shuttling between enzymes and carbon electrodes. Adv Funct Mater. 2024;34(18):2400479.

[84]

LiuX, ZhengD, LongY, Wang L. Highly robust nanogels from thermal-responsive nanoparticles with controlled swelling for engineering deployments. ACS Appl Mater Interfaces. 2023;15(8):11175-11184.

[85]

FengZ, LiQ, WangW, et al. Superhydrophilic fluorinated polymer and nanogel for high-performance 19F magnetic resonance imaging. Biomaterials. 2020;256:120184.

[86]

WangH, TiwariN, OrellanoMS, et al. Polyglycerol-functionalized β-cyclodextrins as crosslinkers in thermoresponsive nanogels for the enhanced dermal penetration of hydrophobic drugs. Small. 2024;20(32):2311166.

[87]

DingP, HuangJ, WeiC, et al. Efficient and generic preparation of diverse polyelectrolyte nanogels by electrostatic assembly directed polymerization. CCS Chemistry. 2020;2(6):1016-1025.

[88]

LiZ, ZhuY, ZengH, et al. Mechano-boosting nanomedicine antitumour efficacy by blocking the reticuloendothelial system with stiff nanogels. Nat Commun. 2023;14(1):1437.

[89]

LiX, LiX, XiaT, ChenW, SheaKJ, Lu X. Remarkable sol–gel transition of PNIPAm-based nanogels via large steric hindrance of side-chains. Materials Horizons. 2023;10(10):4452-4462.

[90]

ChoiES, KimS, KimD, ChoiE, RyuJ-H. Drug-loaded nanogel for efficient orchestration of cell death pathways by intramitochondrial disulfide polymerization. Small. 2024;20(15):2308872.

[91]

ChenS-X, ZhangJ, XueF, et al. In situ forming oxygen/ROS-responsive niche-like hydrogel enabling gelation-triggered chemotherapy and inhibition of metastasis. Bioactive Materials. 2023;21:86-96.

[92]

XiaT, LiX, WuY, LuX. Synthesis and thermally-induced gelation of interpenetrating nanogels. J Colloid Interface Sci. 2024;669:754-765.

[93]

QiuY, BaiJ, FengY, Shi X, ZhaoX. Use of pH-active catechol-bearing polymeric nanogels with glutathione-responsive dissociation to codeliver bortezomib and doxorubicin for the synergistic therapy of cancer. ACS Appl Mater Interfaces. 2021;13(31):36926-36937.

[94]

SunX, LiY, LiuX, CuiD, ShiY, HuangG. Tumor-specific enhanced NIR-II photoacoustic imaging via photothermal and low-pH coactivated AuNR@PNIPAM-VAA nanogel. J Nanobiotechnology. 2024;22(1):326.

[95]

CzyschC, Medina-Montano C, ZhongZ, et al. Transient lymph node immune activation by hydrolysable polycarbonate nanogels. Adv Funct Mater. 2022;32(35):2203490.

[96]

ZhangY, DostaP, CondeJ, Oliva N, WangM, ArtziN. Prolonged local in vivo delivery of stimuli-responsive nanogels that rapidly release doxorubicin in triple-negative breast cancer cells. Adv Healthcare Mater. 2020;9(4):e1901101.

[97]

DuanQ-Y, ZhuY-X, JiaH-R, et al. Platinum-coordinated dual-responsive nanogels for universal drug delivery and combination cancer therapy. Small. 2022;18(46):2203260.

[98]

ZhangY, ZouZ, LiuS, et al. Edaravone-loaded poly(amino acid) nanogel inhibits ferroptosis for neuroprotection in cerebral ischemia injury. Asian J Pharm Sci. 2024;19(2):100886.

[99]

XiaoT, HeM, XuF, et al. Macrophage membrane-camouflaged responsive polymer nanogels enable magnetic resonance imaging-guided chemotherapy/chemodynamic therapy of orthotopic glioma. ACS Nano. 2021;15(12):20377-20390.

[100]

FanX, WangK, LuQ, et al. Surface-anchored tumor microenvironment-responsive protein nanogel-platelet system for cytosolic delivery of therapeutic protein in the post-surgical cancer treatment. Acta Biomater. 2022;154:412-423.

[101]

KovacevicB, WagleSR, IonescuCM, et al. Biotechnological effects of advanced smart-bile acid cyclodextrin-based nanogels for ear delivery and treatment of hearing loss. Adv Healthcare Mater. 2024;13(16):2303149.

[102]

PengS, WangH, ZhaoW, et al. Zwitterionic polysulfamide drug nanogels with microwave augmented tumor accumulation and on-demand drug release for enhanced cancer therapy. Adv Funct Mater. 2020;30(23):2001832.

[103]

XuJ, QiuW, LiangM, et al. Dual-stimulus phototherapeutic nanogel for triggering pyroptosis to promote cancer immunotherapy. J Controlled Release. 2023;358:219-231.

[104]

LeeCG, LeeC, LeeJ, NamJS, KimB-S, Kwon T-H. Dual-Modulated release of a cytotoxic photosensitizer using photogenerated reactive oxygen species and glutathione. Angew Chem Int Ed. 2022;61(42):e202210623.

[105]

XiangY, QiuZ, DingY, et al. Dexamethasone-loaded ROS stimuli-responsive nanogels for topical ocular therapy of corneal neovascularization. J Controlled Release. 2024;372:874-884.

[106]

HuY, ZhaoM, WangH, et al. Exosome-sheathed ROS-responsive nanogel to improve targeted therapy in perimenopausal depression. J Nanobiotechnology. 2023;21(1):261.

[107]

MoradiMR, Salahinejad E, SharifiE, TayebiL. Controlled drug delivery from chitosan-coated heparin-loaded nanopores anodically grown on nitinol shape-memory alloy. Carbohydr Polymers. 2023;314:120961.

[108]

LuT, ten Hagen TLM. A novel kinetic model to describe the ultra-fast triggered release of thermosensitive liposomal drug delivery systems. J Controlled Release. 2020;324:669-678.

[109]

RavindranR, MitraK, ArumugamSK, Doble M. Preparation of curdlan sulphate -chitosan nanoparticles as a drug carrier to target Mycobacterium smegmatis infected macrophages. Carbohydr Polymers. 2021;258:117686.

[110]

CiroY, RojasJ, Di VirgilioAL, AlhajjMJ, Carabali GA, SalamancaCH. Production, physicochemical characterization, and anticancer activity of methotrexate-loaded phytic acid-chitosan nanoparticles on HT-29 human colon adenocarcinoma cells. Carbohydr Polymers. 2020;243:116436.

[111]

BaoH, DingHH, CharlesAPR, et al. Application of yellow mustard mucilage in encapsulation of essential oils and polyphenols using spray drying. Food Hydrocolloids. 2023;143:108815.

[112]

GilroyDW. Resolving inflammation. Nat Rev Immunol. 2021;21(10):620-621.

[113]

XiongMH, LiYJ, BaoY, YangXZ, HuB, WangJ. Bacteria-responsive multifunctional nanogel for targeted antibiotic delivery. Adv Mater. 2012;24(46):6175-6180.

[114]

LuoW, ZengY, SongQ, et al. Strengthening the combinational immunotherapy from modulating the tumor inflammatory environment via hypoxia-responsive nanogels. Adv Healthcare Mater. 2024;13(8):2302865.

[115]

MaY, YangH, ZongX, et al. Artificial M2 macrophages for disease-modifying osteoarthritis therapeutics. Biomaterials. 2021;274:120865.

[116]

FuYJ, ZhaoX, WangLY, et al. A gas therapy strategy for intestinal flora regulation and colitis treatment by nanogel-based multistage NO delivery microcapsules. Adv Mater. 2024;36(19):2309972.

[117]

ZhuH, KongB, CheJ, ZhaoY, SunL. Bioinspired nanogels as cell-free DNA trapping and scavenging organelles for rheumatoid arthritis treatment. Proc Natl Acad Sci USA. 2023;120(33):e2303385120.

[118]

ZhouZ, LiK, GuoY, et al. ROS/electro dual-reactive nanogel for targeting epileptic foci to remodel aberrant circuits and inflammatory microenvironment. ACS Nano. 2023;17(8):7847-7864.

[119]

SunY, DingSL, ZhaoX, et al. Self-reinforced MOF-based nanogel alleviates osteoarthritis by long-acting drug release. Adv Mater. 2024;1:2401094.

[120]

DaiB, ZhuY, LiX, et al. Blockage of osteopontin-integrin β3 signaling in infrapatellar fat pad attenuates osteoarthritis in mice. Adv Sci. 2023;10(22):2300897.

[121]

CossuG, Birchall M, BrownT, et al. Lancet commission: stem cells and regenerative Medicine. The Lancet. 2018;391(10123):883-910.

[122]

EichholzKF, Gonçalves I, BarcelóX, FedericiAS, HoeyDA, KellyDJ. How to design, develop and build a fully-integrated melt electrowriting 3D printer. Additive Manufacturing. 2022;58:102998.

[123]

BehCW, YewDS, ChaiRJ, Chin SY, SeowY, HoonSS. A fluid-supported 3D hydrogel bioprinting method. Biomaterials. 2021;276:121034.

[124]

MoroniL, Burdick JA, HighleyC, et al. Biofabrication strategies for 3D in vitro models and regenerative medicine. Nature Reviews Materials. 2018;3(5):21-37.

[125]

TheLancet. Stem cells, regenerative medicine, and prometheus. The Lancet. 2018;391(10123):814.

[126]

BrodyH. Regenerative medicine. Nature. 2016;540(7632):S49.

[127]

DirckxN, MoorerMC, ClemensTL, Riddle RC. The role of osteoblasts in energy homeostasis. Nat Rev Endocrinol. 2019;15(11):651-665.

[128]

FulzeleK, RiddleRC, DiGirolamoDJ, et al. Insulin receptor signaling in osteoblasts regulates postnatal bone acquisition and body composition. Cell. 2010;142(2):309-319.

[129]

MostovK, WerbZ. Journey across the osteoclast. Science. 1997;276(5310):219-220.

[130]

KoonsGL, DibaM, MikosAG. Materials design for bone-tissue engineering. Nat Rev Mater. 2020;5(8):584-603.

[131]

TangY, WangJ, CaoQ, et al. Dopamine/DOPAC-assisted immobilization of bone morphogenetic protein-2 loaded Heparin/PEI nanogels onto three-dimentional printed calcium phosphate ceramics for enhanced osteoinductivity and osteogenicity. Biomaterials Advances. 2022;140:213030.

[132]

FanM, YanJ, TanH, MiaoY, HuX. Magnetic biopolymer nanogels via biological assembly for vectoring delivery of biopharmaceuticals. J Mater Chem B. 2014;2(47):8399-8405.

[133]

ChenK, LiuY, LiuX, et al. Hyaluronic acid-modified and verteporfin-loaded polylactic acid nanogels promote scarless wound healing by accelerating wound re-epithelialization and controlling scar formation. J Nanobiotechnology. 2023;21(1):241.

[134]

ZhangS, LiY, QiuX, et al. Incorporating redox-sensitive nanogels into bioabsorbable nanofibrous membrane to acquire ROS-balance capacity for skin regeneration. Bioactive Materials. 2021;6(10):3461-3472.

[135]

XinP, HanS, HuangJ, You X, WuJ. Natural soybean milk-derived bioactive coatings for enhanced wound healing. ACS Appl Mater Interfaces. 2022;14(30):34480-34487.

[136]

DongR, GuoB. Smart wound dressings for wound healing. Nano Today. 2021;41:101290.

[137]

CzajkaC. Capturing chemokines in chronic wounds. Science. 2017;356(6335):280.2-280.

[138]

KoettingMC, PetersJT, SteichenSD, Peppas NA. Stimulus-responsive hydrogels: theory, modern advances, and applications. Materi Sci Eng: R: Rep. 2015;93:1-49.

[139]

GuoB, DongR, LiangY, Li M. Haemostatic materials for wound healing applications. Nat Rev Chem. 2021;5(11):773-791.

[140]

FalangaV, Isseroff RR, SoulikaAM, et al. Chronic wounds. Nat Rev Dis Primers. 2022;8(1):50.

[141]

YuanJ, WangY, YangW, et al. Biomimetic peptide dynamic hydrogel inspired by humanized defensin nanonets as the wound-healing gel coating. Chem Eng J. 2023;470:144266.

[142]

NiethammerP, Grabher C, LookAT, MitchisonTJ. A tissue-scale gradient of hydrogen peroxide mediates rapid wound detection in zebrafish. Nature. 2009;459(7249):996-999.

[143]

RatherHA, Thakore R, SinghR, JhalaD, SinghS, VasitaR. Antioxidative study of cerium oxide nanoparticle functionalised PCL-Gelatin electrospun fibers for wound healing application. Bioactive Materials. 2018;3(2):201-211.

[144]

WangH, ChenQ, ZhouS. Carbon-based hybrid nanogels: a synergistic nanoplatform for combined biosensing, bioimaging, and responsive drug delivery. Chem Soc Rev. 2018;47(11):4198-4232.

[145]

ChanM, Almutairi A. Nanogels as imaging agents for modalities spanning the electromagnetic spectrum. Materials Horizons. 2016;3(1):21-40.

[146]

FuW, ZhangX, MeiL, et al. Stimuli-responsive small-on-large nanoradiosensitizer for enhanced tumor penetration and radiotherapy sensitization. ACS Nano. 2020;14(8):10001-10017.

[147]

HuangH, HeL, ZhouW, et al. Stable black phosphorus/Bi2O3 heterostructures for synergistic cancer radiotherapy. Biomaterials. 2018;171:12-22.

[148]

ZhangC, TuW, ChenX, et al. Intelligent design of polymer nanogels for full-process sensitized radiotherapy and dual-mode computed tomography/magnetic resonance imaging of tumors. Theranostics. 2022;12(7):3420-3437.

[149]

CarniatoF, RicciM, TeiL, et al. Novel nanogels loaded with Mn(II) chelates as effective and biologically stable MRI probes. Small. 2023;19(42):2302868.

[150]

ZhangW, DuB, GaoM, TungCH. A hybrid nanogel to preserve lysosome integrity for fluorescence imaging. ACS Nano. 2021;15(10):16442-16451.

[151]

LuxJ, WhiteAG, ChanM, Anderson CJ, AlmutairiA. Nanogels from metal-chelating crosslinkers as versatile platforms applied to Copper-64 PET imaging of tumors and metastases. Theranostics. 2015;5(3):277-288.

[152]

MaX, ZhangT, QiuW, et al. Bioresponsive prodrug nanogel-based polycondensate strategy deepens tumor penetration and potentiates oxidative stress. Chem Eng J. 2021;420:127657.

[153]

HeM, XiaoT, WangY, et al. Multifunctional PVCL nanogels enable magnetic resonance imaging and immunostimulated radiotherapy of orthotopic glioblastoma. Chem Eng J. 2023;453:139634.

[154]

WangS, RenWX, HouJT, et al. Fluorescence imaging of pathophysiological microenvironments. Chem Soc Rev. 2021;50(16):8887-8902.

[155]

ZhaoZ, FanH, ZhouG, et al. Activatable fluorescence/MRI bimodal platform for tumor cell imaging via MnO2 nanosheet-aptamer nanoprobe. J Am Chem Soc. 2014;136(32):11220-11223.

[156]

XinQ, MaH, WangH, Zhang X-D. Tracking tumor heterogeneity and progression with near-infrared II fluorophores. Exploration. 2023;3(2):20220011.

[157]

ChenX, ZhangH, ZhangM, et al. Amorphous Fe-based nanoagents for Self-Enhanced chemodynamic therapy by re-establishing tumor acidosis. Adv Funct Mater. 2020;30(6):1908365.

[158]

MiaoS, Yueqiang Z, JunbinC, et al. Microfluidic synthesis of manganese-alginate nanogels with self-supplying H2O2 capability for synergistic chemo/chemodynamic therapy and boosting anticancer immunity. Chem Eng J. 2022;435:134926.

[159]

ZiyingL, Huifang S, HuangzhangX, et al. Tri-component programmable nanoregulator with three-pronged penetration boosts immunotherapy of Triple-Negative breast cancer. Chem Eng J. 2022;439:135712.

[160]

JiP, SunW, ZhangS, et al. Modular hydrogel vaccine for programmable and coordinate elicitation of cancer immunotherapy. Adv Sci. 2023;10:2301789.

[161]

SuM, ZhuY, ChenJ, et al. Microfluidic synthesis of manganese-alginate nanogels with self-supplying H2O2 capability for synergistic chemo/chemodynamic therapy and boosting anticancer immunity. Chem Eng J. 2022;435:134926.

[162]

QinX, WuC, NiuD, et al. Peroxisome inspired hybrid enzyme nanogels for chemodynamic and photodynamic therapy. Nat Commun. 2021;12(1):5243.

[163]

ZhangF, DongJ, HuangK, et al. Dominolike” barriers elimination with an intratumoral adenosine-triphosphate-supersensitive nanogel to enhance cancer chemoimmunotherapy. ACS Nano. 2023;17(19):18805-18817.

[164]

LiZ, ShiH, XieH, et al. Tri-component programmable nanoregulator with three-pronged penetration boosts immunotherapy of Triple-Negative breast cancer. Chem Eng J. 2022;439:135712.

[165]

TianH, LiW, WangG, et al. Self-degradable nanogels reshape immunosuppressive tumor microenvironment via drug repurposing strategy to reactivate cytotoxic CD8+ T cells. Adv Sci. 2023;10(21):2301661.

[166]

MuHY, TaYNN, ThamMJR, et al. A chemoimmunotherapy nanogel enables efficient delivery of interleukin-2 and induction of immunogenic cell death for effective cancer therapy. Adv Funct Mater. 2024;34(1):2303033.

[167]

CorreiaJH, Rodrigues JA, PimentaS, DongT, YangZ. Photodynamic therapy review: principles, photosensitizers, applications, and future directions. Pharmaceutics. 2021;13(9):1332.

[168]

DaiZ, ZhangQ, LiX, et al. In situ forming pH/ROS-responsive niche-like hydrogel for ultrasound-mediated multiple therapy in synergy with potentiating anti-tumor immunity. Mater Today. 2023;65:62-77.

[169]

WangC, TianY, WuB, ChengW. Recent progress toward imaging application of multifunction sonosensitizers in sonodynamic therapy. Int J Nanomedicine. 2022;17:3511-3529.

[170]

RuiR, ZhouL, HeS. Cancer immunotherapies: advances and bottlenecks. Front Immunol. 2023;14:1212476.

[171]

FangY, YuA, YeL, ZhaiG. Research progress in tumor targeted immunotherapy. Expert Opin Drug Delivery. 2021;18(8):1067-1090.

[172]

ZhengY, HanY, SunQ, LiZ. Harnessing anti-tumor and tumor-tropism functions of macrophages via nanotechnology for tumor immunotherapy. Exploration. 2022;2(3):20210166.

[173]

BinnewiesM, Roberts EW, KerstenK, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nature Med. 2018;24(5):541-550.

[174]

HuangJ, YangB, PengY, et al. Nanomedicine-boosting tumor immunogenicity for enhanced immunotherapy. Adv Funct Mater. 2021;31(21):2011171.

RIGHTS & PERMISSIONS

2024 The Author(s). MedComm - Biomaterials and Applications published by John Wiley & Sons Australia, Ltd on behalf of Sichuan International Medical Exchange & Promotion Association (SCIMEA).

AI Summary AI Mindmap
PDF

147

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/