1,113
Views
2
CrossRef citations to date
0
Altmetric
Review

Bioresponsive functional nanogels as an emerging platform for cancer therapy

, , , , &
Pages 703-716 | Received 12 Apr 2018, Accepted 02 Jul 2018, Published online: 16 Jul 2018

References

  • Kabanov AV, Vinogradov SV. Nanogels as pharmaceutical carriers: finite networks of infinite capabilities, Angew. Chem Int Ed. 2009;48:5418–5429.
  • Eckmann DM, Composto RJ, Tsourkas A, et al. Nanogel carrier design for targeted drug delivery. J Mater Chem B. 2014;2:8085–8097.
  • Li D, Van Nostrum CF, Mastrobattista E, et al. Nanogels for intracellular delivery of biotherapeutics. J Control. Release. 2017;259:16–28.
  • Chacko RT, Ventura J, Zhuang J, et al. Polymer nanogels: A versatile nanoscopic drug delivery platform. Adv Drug Deliv Rev. 2012;64:836–851.
  • Raemdonck K, Demeester J, De Smedt S. Advanced nanogel engineering for drug delivery. Soft Matter. 2009;5:707–715.
  • Zhang H, Zhai Y, Wang J, et al. New progress and prospects: the application of nanogel in drug delivery. Sci Eng C Mater Biol Appl. 2016;60:560–568.
  • Deng C, Jiang Y, Cheng R, et al. Biodegradable polymeric micelles for targeted and controlled anticancer drug delivery: promises, progress and prospects. Nano Today. 2012;7:467–480.
  • Sun T, Zhang YS, Pang B, et al. Engineered nanoparticles for drug delivery in cancer therapy. Angew Chem Int Ed. 2014;53:12320–12364.
  • Torchilin VP. Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery. Nat Rev Drug Discov. 2014;13:813.
  • Cheng R, Meng F, Deng C, et al. Bioresponsive polymeric nanotherapeutics for targeted cancer chemotherapy. Nano Today. 2015;10:656–670.
  • Lu Y, Aimetti AA, Langer R, et al. Bioresponsive materials. Nat Rev Mater. 2017;2:16075.
  • Zha L, Banik B, Alexis F. Stimulus responsive nanogels for drug delivery. Soft Matter. 2011;7:5908–5916.
  • Smith MH, Lyon LA. Multifunctional nanogels for siRNA delivery. Acc Chem Res. 2012;45:985–993.
  • Jhaveri A, Deshpande P, Torchilin V. Stimuli-sensitive nanopreparations for combination cancer therapy. J Control. Release. 2014;190:352–370.
  • Li Y, Maciel D, Rodrigues J, et al. Biodegradable polymer nanogels for drug/nucleic acid delivery. Chem Rev. 2015;115:8564–8608.
  • Oh JK, Drumright R, Siegwart DJ, et al. The development of microgels/nanogels for drug delivery applications. Prog Polym Sci. 2008;33:448–477.
  • Oh JK, Lee DI, Park JM. Biopolymer-based microgels/nanogels for drug delivery application. Prog Polym Sci. 2009;34:1261–1282.
  • Liu D, Zhang H, Fontana F. et al. Microfluidic-assisted fabrication of carriers for controlled drug delivery. Lab on a Chip. 2017;17:1856–1883.
  • Bazban-Shotorbani S, Dashtimoghadam E, Karkhaneh A, et al. Microfluidic directed synthesis of alginate nanogels with tunable pore size for efficient protein delivery. Langmuir. 2016;32:4996–5003.
  • Ko DY, Shinde UP, Yeon B, et al. Recent progress of in situ formed gels for biomedical applications. Prog Polym Sci. 2013;38:672–701.
  • Zhang X, Malhotra S, Molina M, et al. Micro- and nanogels with labile crosslinks-from synthesis to biomedical applications. Chem Soc Rev. 2015;44:1948–1973.
  • Jiang Y, Chen J, Deng C, et al. Click hydrogels, microgels and nanogels: emerging platforms for drug delivery and tissue engineering. Biomaterials. 2014;35:4969–4985.
  • Steinhilber D, Rossow T, Wedepohl S, et al. A microgel construction kit for bioorthogonal encapsulation and pH-controlled release of living cells. Angew Chem Int Ed. 2013;52:13538–13543.
  • Steinhilber D, Witting M, Zhang X, et al. Surfactant free preparation of biodegradable dendritic polyglycerol nanogels by inverse nanoprecipitation for encapsulation and release of pharmaceutical biomacromolecules, J. Control. Release. 2013;169:289–295.
  • Chen J, Zou Y, Deng C, et al. Multifunctional click hyaluronic acid nanogels for targeted protein delivery and effective cancer treatment in vivo. Chem Mater. 2016;28:8792–8799.
  • Chen J, Ouyang J, Chen Q, et al. EGFR and CD44 dual-targeted multifunctional hyaluronic acid nanogels boost protein delivery to ovarian and breast cancers in vitro and in vivo. ACS Appl Mater Interface. 2017;9:24140–24147.
  • Soni KS, Desale SS, Bronich TK. Nanogels: an overview of properties, biomedical applications and obstacles to clinical translation. J Control. Release. 2016;240:109–126.
  • Neamtu I, Rusu AG, Diaconu A, et al. Basic concepts and recent advances in nanogels as carriers for medical applications. Drug Deliv. 2017;24:539–557.
  • Sivaram AJ, Rajitha P, Maya S, et al. Nanogels for delivery, imaging and therapy, Wiley Interdiscip. Rev Nanomed Nanobiotechnol. 2015;7:509–533.
  • Wu H-Q, Wang -C-C. Biodegradable smart nanogels: A new platform for targeting drug delivery and biomedical diagnostics. Langmuir. 2016;32:6211–6225.
  • Ding L, Jiang Y, Zhang J, et al. pH-sensitive coiled-coil peptide-cross-linked hyaluronic acid nanogels: synthesis and targeted intracellular protein delivery to CD44 positive cancer cells. Biomacromolecules. 2018;19:555–562.
  • Binauld S, Stenzel MH. Acid-degradable polymers for drug delivery: A decade of innovation. Chem Commun. 2013;49:2082–2102.
  • Zhu X, Sun Y, Chen D, et al. Mastocarcinoma therapy synergistically promoted by lysosome dependent apoptosis specifically evoked by 5-Fu@nanogel system with passive targeting and pH activatable dual function. J Control. Release. 2017;254:107–118.
  • Arunraj TR, Sanoj Rejinold N, Ashwin Kumar N, et al. Doxorubicin-chitin-poly(caprolactone) composite nanogel for drug delivery. Inter J Biolog Macromol. 2013;62:35–43.
  • Khaled SZ, Cevenini A, Yazdi IK, et al. One-pot synthesis of pH-responsive hybrid nanogel particles for the intracellular delivery of small interfering RNA. Biomaterials. 2016;87:57–68.
  • Lee ES, Kim D, Youn YS, et al. A virus-mimetic nanogel vehicle. Angew Chem. 2008;120:2452–2455.
  • Ju C, Mo R, Xue J, et al. Sequential intra-intercellular nanoparticle delivery system for deep tumor penetration. Angew Chem Int Ed. 2014;53:6253–6258.
  • Song Q, Yin Y, Shang L, et al. Tumor microenvironment responsive nanogel for the combinatorial antitumor effect of chemotherapy and immunotherapy. Nano Lett. 2017;17:6366–6375.
  • Mavuso S, Marimuthu T, Choonara YE, et al. A review of polymeric colloidal nanogels in transdermal drug delivery. Curr Pharm Des. 2015;21:2801–2813.
  • Mangalathillam S, Rejinold NS, Nair A, et al. Curcumin loaded chitin nanogels for skin cancer treatment via the transdermal route. Nanoscale. 2012;4:239–250.
  • Divya G, Panonnummal R, Gupta S, et al. Acitretin and aloe-emodin loaded chitin nanogel for the treatment of psoriasis. Eur J Pharm Biopharm. 2016;107:97–109.
  • Gao C, Lin Z, Jurado-Sánchez B, et al. Stem cell membrane-coated nanogels for highly efficient in vivo tumor targeted drug delivery. Small. 2016;12:4056–4062.
  • Yahia-Ammar A, Sierra D, Mérola F, et al. Self-assembled gold nanoclusters for bright fluorescence imaging and enhanced drug delivery. ACS Nano. 2016;10:2591–2599.
  • Li X, Du P, Liu P. Layer-by-layer polyelectrolyte complex coated poly(methacrylic acid) nanogels as a drug delivery system for controlled release: structural effects. RSC Adv. 2014;4:56323–56331.
  • Lin L, Xu W, Liang H, et al. Construction of pH-sensitive lysozyme/pectin nanogel for tumor methotrexate delivery. Colloids Surf B Biointerfaces. 2015;126:459–466.
  • Su S, Wang H, Liu X, et al. iRGD-coupled responsive fluorescent nanogel for targeted drug delivery. Biomaterials. 2013;34:3523–3533.
  • Wu W, Aiello M, Zhou T, et al. In-situ immobilization of quantum dots in polysaccharide-based nanogels for integration of optical pH-sensing, tumor cell imaging, and drug delivery. Biomaterials. 2010;31:3023–3031.
  • Wu W, Shen J, Banerjee P, et al. Chitosan-based responsive hybrid nanogels for integration of optical pH-sensing, tumor cell imaging and controlled drug delivery. Biomaterials. 2010;31:8371–8381.
  • Du J-Z, Sun T-M, Song W-J, et al. A tumor-acidity-activated charge-conversional nanogel as an intelligent vehicle for promoted tumoral-cell uptake and drug delivery. Angew Chem. 2010;122:3703–3708.
  • Chen W, Hou Y, Tu Z, et al. pH-degradable PVA-based nanogels via photo-crosslinking of thermo-preinduced nanoaggregates for controlled drug delivery, J. Control. Release. 2017;259:160–167.
  • Li Y, Bui QN, Duy LTM. et al. One-step preparation of pH-responsive polymeric nanogels as intelligent drug delivery systems for tumor therapy, Biomacromolecules. 2018;19:2062–2070.
  • Dimde M, Neumann F, Reisbeck F, et al. Defined pH-sensitive nanogels as gene delivery platform for siRNA mediated in vitro gene silencing. Biomater Sci. 2017;5:2328–2336.
  • Ulijn RV. Enzyme-responsive materials: A new class of smart biomaterials. J Mater Chem. 2006;16:2217–2225.
  • De La Rica R, Aili D, Stevens MM. Enzyme-responsive nanoparticles for drug release and diagnostics. Adv Drug Deliv Rev. 2012;64:967–978.
  • Hu J, Zhang G, Liu S. Enzyme-responsive polymeric assemblies, nanoparticles and hydrogels. Chem Soc Rev. 2012;41:5933–5949.
  • Yang C, Wang X, Yao X, et al. Hyaluronic acid nanogels with enzyme-sensitive cross-linking group for drug delivery. J Control. Release. 2015;205:206–217.
  • Jiang T, Mo R, Bellotti A, et al. Gel-liposome-mediated co-delivery of anticancer membrane-associated proteins and small-molecule drugs for enhanced therapeutic efficacy. Adv Funct Mater. 2014;24:2295–2304.
  • Qiuwen Z, Xiaojie C, Xiao X, et al. Tumor-specific self-degradable nanogels as potential carriers for systemic delivery of anticancer proteins. Adv Funct Mater. 2018;28:1707371.
  • Wang Y, Luo Y, Zhao Q, et al. An enzyme-responsive nanogel carrier based on PAMAM dendrimers for drug delivery. ACS Appl Mater Interface. 2016;8:19899–19906.
  • Xiong M-H, Bao Y, Du X-J, et al. Differential anticancer drug delivery with a nanogel sensitive to bacteria-accumulated tumor artificial environment. ACS Nano. 2013;7:10636–10645.
  • Wang Y, Qiao S-L, Wang H. Facile synthesis of peptide cross-linked nanogels for tumor metastasis inhibition. ACS Appl Nano Mater. 2018;1:785–792.
  • Wang Y, Xu H, Wang J, et al. Development of a thermally responsive nanogel based on chitosan-poly(N-isopropylacrylamide-co-acrylamide) for paclitaxel delivery. J Pharm Sci. 2014;103:2012–2021.
  • Molina M, Wedepohl S, Miceli E, et al. Overcoming drug resistance with on-demand charged thermoresponsive dendritic nanogels. Nanomedicine. 2017;12:117–129.
  • Shirakura T, Kelson TJ, Ray A, et al. Hydrogel nanoparticles with thermally controlled drug release. ACS Macro Lett. 2014;3:602–606.
  • Meng F, Hennink WE, Zhong Z. Reduction-sensitive polymers and bioconjugates for biomedical applications. Biomaterials. 2009;30:2180–2198.
  • Cheng R, Feng F, Meng F, et al. Glutathione-responsive nano-vehicles as a promising platform for targeted intracellular drug and gene delivery. J Control Release. 2011;152:2–12.
  • Arunachalam B, Phan UT, Geuze HJ, et al. Enzymatic reduction of disulfide bonds in lysosomes: characterization of a Gamma-interferon-inducible lysosomal thiol reductase (GILT). Proc Natl Acad Sci USA. 2000;97:745–750.
  • Li Y-L, Zhu L, Liu Z, et al. Reversibly stabilized multifunctional dextran nanoparticles efficiently deliver doxorubicin into the nuclei of cancer cells. Angew Chem Int Ed. 2009;48:9914–9918.
  • Chen W, Zhong P, Meng F, et al. Redox and pH-responsive degradable micelles for dually activated intracellular anticancer drug release. J Control Release. 2013;169:171–179.
  • Chen W, Zou Y, Jia J, et al. Functional poly(ε-caprolactone)s via copolymerization of ε-caprolactone and pyridyl disulfide-containing cyclic carbonate: controlled synthesis and facile access to reduction-sensitive biodegradable graft copolymer micelles. Macromolecules. 2013;46:699–707.
  • Ryu J-H, Jiwpanich S, Chacko R, et al. Surface-functionalizable polymer nanogels with facile hydrophobic guest encapsulation capabilities . J Am Chem Soc. 2010;132:8246–8247.
  • ParK H, Choi Y, Jeena M, et al. Reduction-triggered self-cross-linked hyperbranched polyglycerol nanogels for intracellular delivery of drugs and proteins. Macromol Biosci. 2018;18:1700356.
  • Zhong Y, Zhang J, Cheng R, et al. Reversibly crosslinked hyaluronic acid nanoparticles for active targeting and intelligent delivery of doxorubicin to drug resistant CD44+ human breast tumor xenograft. J Control Release. 2015;205:144–154.
  • Zhong Y, Meng F, Deng C, et al. Targeted inhibition of human hematological cancers in vivo by doxorubicin encapsulated in smart lipoic acid-crosslinked hyaluronic acid nanoparticles. Drug Deliv. 2017;24:1482–1490.
  • Sun B, Deng C, Meng F. et al. Robust, active tumor-targeting and fast bioresponsive anticancer nanotherapeutics based on natural endogenous materials. Acta Biomater. 2016;45:223–233.
  • Pedrosa SS, Gonçalves C, David L, et al. A novel crosslinked hyaluronic acid nanogel for drug delivery. Macromol Biosci. 2014;14:1556–1568.
  • Wu W, Yao W, Wang X, et al. Bioreducible heparin-based nanogel drug delivery system. Biomaterials. 2015;39:260–268.
  • Zhang F, Gong S, Wu J, et al. CXCR4-targeted and redox responsive dextrin nanogel for metastatic breast cancer therapy. Biomacromolecules. 2017;18:1793–1802.
  • Zhu Y, Wang X, Chen J, et al. Bioresponsive and fluorescent hyaluronic acid-iodixanol nanogels for targeted X-ray computed tomography imaging and chemotherapy of breast tumors. J Control. Release. 2016;244:229–239.
  • Huang K, Shi B, Xu W. et al. Reduction-responsive polypeptide nanogel delivers antitumor drug for improved efficacy and safety. Acta Biomater. 2015;27:179–193.
  • Guo H, Xu W, Chen J, et al. Positively charged polypeptide nanogel enhances mucoadhesion and penetrability of 10-hydroxycamptothecin in orthotopic bladder carcinom. J Control. Release. 2017;259:136–148.
  • Chen J, Ding J, Xu W, et al.. Receptor and microenvironment dual-recognizable nanogel for targeted chemotherapy of highly metastatic malignancy. Nano Lett. 2017;17:4526–4533.
  • Chen W, Zheng M, Meng F, et al. In situ forming reduction-sensitive degradable nanogels for facile loading and triggered intracellular release of proteins. Biomacromolecules. 2013;14:1214–1222.
  • Li D, Kordalivand N, Fransen MF, et al. Reduction-sensitive dextran nanogels aimed for intracellular delivery of antigens. Adv Funct Mater. 2015;25:2993–3003.
  • Li S, Zhang J, Deng C, et al. Redox-sensitive and intrinsically fluorescent photoclick hyaluronic acid nanogels for traceable and targeted delivery of cytochrome c to breast tumor in mice. ACS Appl Mater Interface. 2016;8:21155–21162.
  • Gouda N, Miyata K, Christie RJ, et al. Silica nanogelling of environment-responsive PEGylated polyplexes for enhanced stability and intracellular delivery of siRNA. Biomaterials. 2013;34:562–570.
  • Averick SE, Paredes E, Irastorza A, et al. Preparation of cationic nanogels for nucleic acid delivery. Biomacromolecules. 2012;13:3445–3449.
  • Hong CA, Kim JS, Lee SH, et al. Reductively dissociable siRNA-polymer hybrid nanogels for efficient targeted gene silencing. Adv Funct Mater. 2013;23:316–322.
  • Dunn SS, Tian S, Blake S, et al. Reductively responsive siRNA-conjugated hydrogel nanoparticles for gene silencing. J Am Chem Soc. 2012;134:7423–7430.
  • Li J, Zheng C, Cansiz S, et al. Self-assembly of DNA nanohydrogels with controllable size and stimuli-responsive property for targeted gene regulation therapy. J Am Chem Soc. 2015;137:1412–1415.
  • Nuhn L, Braun L, Overhoff I, et al. Degradable cationic nanohydrogel particles for stimuli-responsive release of siRNA. Macromol Rapid Commun. 2014;35:2057–2064.
  • Li H, Yang X, Gao F. et al. Bioreduction-ruptured nanogel for switch on/off release of Bcl2 siRNA in breast tumor therapy. J ControlRelease. 2018. doi:10.1016/j.jconrel.2018.02.036
  • Shim MS, Xia Y. A reactive oxygen species (ROS)-responsive polymer for safe, efficient, and targeted gene delivery in cancer cells. Angew Chem Int Ed. 2013;52:6926–6929.
  • Deepagan VG, Kwon S, You DG, et al. In situ diselenide-crosslinked polymeric micelles for ROS-mediated anticancer drug delivery. Biomaterials. 2016;103:56–66.
  • Tian Y, Zheng J, Tang X, et al. Near‐Infrared Light‐Responsive Nanogels with Diselenide-Cross-Linkers for On-Demand Degradation and Triggered Drug Release. Part Syst Char. 2015;32:547–551.
  • Cheng R, Meng F, Deng C, et al. Dual and multi-stimuli responsive polymeric nanoparticles for programmed site-specific drug delivery. Biomaterials. 2013;34:3647–3657.
  • Chen W, Meng F, Cheng R, et al. Facile construction of dual-bioresponsive biodegradable micelles with superior extracellular stability and activated intracellular drug release. J Control. Release. 2015;210:125–133.
  • Ding J, Xu W, Zhang Y, et al. Self-reinforced endocytoses of smart polypeptide nanogels for “on-demand” drug delivery. J Control. Release. 2013;172:444–455.
  • Li M, Tang Z, Sun H, et al. pH and reduction dual-responsive nanogel cross-linked by quaternization reaction for enhanced cellular internalization and intracellular drug delivery. Polym Chem. 2013;4:1199–1207.
  • Bahadur K. C R, Xu P, Multicompartment intracellular self-expanding nanogel for targeted delivery of drug cocktail. Adv Mater. 2012;24:6479–6483.
  • Wang H, Dai T, Zhou S, et al. Self-assembly assisted fabrication of dextran-based nanohydrogels with reduction-cleavable junctions for applications as efficient drug delivery systems. Sci Rep. 2017;7:40011.
  • Wu H, Jin H, Wang C, et al. Synergistic cisplatin/doxorubicin combination chemotherapy for multidrug-resistant cancer via polymeric nanogels targeting delivery. ACS Appl Mater Interface. 2017;9:9426–9436.
  • Li L, Raghupathi K, Yuan C, et al. Surface charge generation in nanogels for activated cellular uptake at tumor-relevant pH. Chem Sci. 2013;4:3654–3660.
  • Zhang X, Achazi K, Steinhilber D, et al. A facile approach for dual-responsive prodrug nanogels based on dendritic polyglycerols with minimal leaching. J Control. Release. 2014;174:209–216.
  • Chen W, Achazi K, Schade B, et al. Charge-conversional and reduction-sensitive poly(vinyl alcohol) nanogels for enhanced cell uptake and efficient intracellular doxorubicin release . J Control. Release. 2015;205:15–24.
  • Chen W, Zou Y, Zhong Z, et al. Cyclo(RGD)-decorated reduction-responsive nanogels mediate targeted chemotherapy of integrin overexpressing human glioblastoma in vivo. Small. 2017;13:1601997–1602005.
  • Mo R, Jiang T, DiSanto R, et al. ATP-triggered anticancer drug delivery. Nat Commun. 2014;5:3364.
  • Zhang X, Achazi K, Haag R. Boronate cross-linked ATP- and pH-responsive nanogels for intracellular delivery of anticancer drugs. Adv Healthc. 2015;4:585–592.
  • Zhang X, Zhang K, Haag R. Multi-stage, charge conversional, stimuli-responsive nanogels for therapeutic protein delivery. Biomater Sci. 2015;3:1487–1496.
  • Rimondino GN, Miceli E, Molina M, et al. Rational design of dendritic thermoresponsive nanogels that undergo phase transition under endolysosomal conditions. J Mater Chem B. 2017;5:866–874.
  • Hu Q, Sun W, Lu Y. et al. Tumor microenvironment-mediated construction and deconstruction of extracellular drug-delivery depots. Nano Lett. 2016;16(1118–1126).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.