5,123
Views
34
CrossRef citations to date
0
Altmetric
Research Article

Stimuli-responsive drug delivery systems for head and neck cancer therapy

, , , , &
Pages 272-284 | Received 12 Nov 2020, Accepted 11 Jan 2021, Published online: 27 Jan 2021

References

  • Agostinis P, Berg K, Cengel KA, et al. (2011). Photodynamic therapy of cancer: an update. CA Cancer J Clin 61:250–81.
  • Akbarzadeh F, Khoshgard K, Arkan E, et al. (2018). Evaluating the photodynamic therapy efficacy using 5-aminolevulinic acid and folic acid-conjugated bismuth oxide nanoparticles on human nasopharyngeal carcinoma cell line. Artif Cells Nanomed Biotechnol 46:S514–S23.
  • Bali A, Bali D, Sharma A. (2013). An overview of gene therapy in head and neck cancer. Indian J Hum Genet 19:282–90.
  • Bansal A, Simon MC. (2018). Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol 217:2291–8.
  • Bhana S, Lin G, Wang L, et al. (2015). Near-infrared-absorbing gold nanopopcorns with iron oxide cluster core for magnetically amplified photothermal and photodynamic cancer therapy. ACS Appl Mater Interfaces 7:11637–47.
  • Bhatnagar S, Venuganti VVK. (2015). Cancer targeting: responsive polymers for stimuli-sensitive drug delivery. J Nanosci Nanotechnol 15:1925–45.
  • Burgoyne JR, Oka S, Ale-Agha N, et al. (2013). Hydrogen peroxide sensing and signaling by protein kinases in the cardiovascular system. Antioxid Redox Signal 18:1042–52.
  • Cardoso VF, Francesko A, Ribeiro C, et al. (2018). Advances in magnetic nanoparticles for biomedical applications. Adv Healthcare Mater 7:1700845.
  • Chen M, He X, Wang K, et al. (2014). A pH-responsive polymer/mesoporous silica nano-container linked through an acid cleavable linker for intracellular controlled release and tumor therapy in vivo. J Mater Chem B 2:428–36.
  • Chi AC, Day TA, Neville BW. (2015). Oral cavity and oropharyngeal squamous cell carcinoma – an update. CA Cancer J Clin 65:401–21.
  • Chien M-H, Lin C-W, Cheng C-W, et al. (2013). Matrix metalloproteinase-2 as a target for head and neck cancer therapy. Expert Opin Ther Targets 17:203–16.
  • Chu C-K, Tu Y-C, Hsiao J-H, et al. (2016). Combination of photothermal and photodynamic inactivation of cancer cells through surface plasmon resonance of a gold nanoring. Nanotechnology 27:115102.
  • Civantos FJ, Karakullukcu B, Biel M, et al. (2018). A review of photodynamic therapy for neoplasms of the head and neck. Adv Ther 35:324–40.
  • Coussens LM, Fingleton B, Matrisian LM. (2002). Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science 295:2387–92.
  • Curry T, Kopelman R, Shilo M, et al. (2014). Multifunctional theranostic gold nanoparticles for targeted CT imaging and photothermal therapy: theranostic au nanoparticles for targeted CT and PTT. Contrast Media Mol Imaging 9:53–61.
  • Damiani V, Falvo E, Fracasso G, et al. (2017). Therapeutic efficacy of the novel stimuli-sensitive nano-ferritins containing doxorubicin in a head and neck cancer model. Int J Mol Sci 18:1555.
  • Datz S, Argyo C, Gattner M, et al. (2016). Genetically designed biomolecular capping system for mesoporous silica nanoparticles enables receptor-mediated cell uptake and controlled drug release. Nanoscale 8:8101–10.
  • Davoodi P, Lee LY, Xu Q, et al. (2018). Drug delivery systems for programmed and on-demand release. Adv Drug Deliv Rev 132:104–38.
  • Epstein JB, Thariat J, Bensadoun R-J, et al. (2012). Oral complications of cancer and cancer therapy: from cancer treatment to survivorship. CA Cancer J Clin 62:400–22.
  • Fan H, Zhu Z, Zhang W, et al. (2020). Light stimulus responsive nanomedicine in the treatment of oral squamous cell carcinoma. Eur J Med Chem 199:112394.
  • Fan L, Wang J, Xia C, et al. (2020). Glutathione-sensitive and folate-targeted nanoparticles loaded with paclitaxel to enhance oral squamous cell carcinoma therapy. J Mater Chem B 8:3113–22.
  • Feller L, Lemmer J. (2012). Oral squamous cell carcinoma: epidemiology, clinical presentation and treatment. J Cancer Ther 03:263–8.
  • Fitzmaurice C. (2018). Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 29 cancer groups, 2006 to 2016: a systematic analysis for the Global Burden of Disease study. J Clin Oncol 36:1568.
  • Gabriel D, Zuluaga MF, Lange N. (2011). On the cutting edge: protease-sensitive prodrugs for the delivery of photoactive compounds. Photochem Photobiol Sci 10:689–703.
  • Gao S, Zheng M, Ren X, et al. (2016). Local hyperthermia in head and neck cancer: mechanism, application and advance. Oncotarget 7:57367–78.
  • Gillies RJ, Raghunand N, Garcia-Martin ML, et al. (2004). pH imaging. A review of pH measurement methods and applications in cancers. IEEE Eng Med Biol Mag 23:57–64.
  • Godin B, Tasciotti E, Liu X, et al. (2011). Multistage nanovectors: from concept to novel imaging contrast agents and therapeutics. Acc Chem Res 44:979–89.
  • Gong J, Chen M, Zheng Y, et al. (2012). Polymeric micelles drug delivery system in oncology. J Control Release 159:312–23.
  • Greish K. (2010). Enhanced permeability and retention (EPR) effect for anticancer nanomedicine drug targeting. In: Grobmyer SR, Moudgil BM, eds. Cancer nanotechnology. Totowa, NJ: Humana Press, 25–37.
  • Guo X, Cheng Y, Zhao X, et al. (2018). Advances in redox-responsive drug delivery systems of tumor microenvironment. J Nanobiotechnol 16:74.
  • Haedicke K, Kozlova D, Gräfe S, et al. (2015). Multifunctional calcium phosphate nanoparticles for combining near-infrared fluorescence imaging and photodynamic therapy. Acta Biomater 14:197–207.
  • Hajebi S, Rabiee N, Bagherzadeh M, et al. (2019). Stimulus-responsive polymeric nanogels as smart drug delivery systems. Acta Biomater 92:1–18.
  • He C, Liu D, Lin W. (2015). Self-assembled core–shell nanoparticles for combined chemotherapy and photodynamic therapy of resistant head and neck cancers. ACS Nano 9:991–1003.
  • Hong Z, Liu H, Zhao M, et al. (2016). Photodynamic therapy of tumors with pyropheophorbide-a-loaded polyethylene glycol-poly(lactic-co-glycolic acid) nanoparticles. Int J Nanomedicine 11:4905–18.
  • Hossen S, Hossain MK, Basher MK, et al. (2019). Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: a review. J Adv Res 15:1–18.
  • Huang J, Huang W, Zhang Z, et al. (2019). Highly uniform synthesis of selenium nanoparticles with EGFR targeting and tumor microenvironment-responsive ability for simultaneous diagnosis and therapy of nasopharyngeal carcinoma. ACS Appl Mater Interfaces 11:11177–93.
  • Huang X, Deng G, Han Y, et al. (2019). Right Cu2-x S@MnS core–shell nanoparticles as a photo/H2O2-responsive platform for effective cancer theranostics. Adv Sci 6:1901461.
  • Huo M, Yuan J, Tao L, et al. (2014). Redox-responsive polymers for drug delivery: from molecular design to applications. Polym Chem 5:1519–28.
  • Jin R, Liu Z, Bai Y, et al. (2018). Core–satellite mesoporous silica–gold nanotheranostics for biological stimuli triggered multimodal cancer therapy. Adv Funct Mater 28:1801961.
  • Kalaydina R-V, Bajwa K, Qorri B, et al. (2018). Recent advances in “smart” delivery systems for extended drug release in cancer therapy. Int J Nanomed 13:4727–45.
  • Kanamala M, Wilson WR, Yang M, et al. (2016). Mechanisms and biomaterials in pH-responsive tumour targeted drug delivery: a review. Biomaterials 85:152–67.
  • Kaomongkolgit R. (2013). Alpha-mangostin suppresses MMP-2 and MMP-9 expression in head and neck squamous carcinoma cells. Odontology 101:227–32.
  • Karimi M, Ghasemi A, Sahandi Zangabad P, et al. (2016). Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chem Soc Rev 45:1457–501.
  • Ketabat F, Pundir M, Mohabatpour F, et al. (2019). Controlled drug delivery systems for oral cancer treatment—current status and future perspectives. Pharmaceutics 11:302.
  • Kim D-H, Vitol EA, Liu J, et al. (2013). Stimuli-responsive magnetic nanomicelles as multifunctional heat and cargo delivery vehicles. Langmuir 29:7425–32.
  • Kim JO, Tran TH, Ramasamy T, et al. (2015). Tumor-targeting, pH-sensitive nanoparticles for docetaxel delivery to drug-resistant cancer cells. Int J Nanomed 10: 5249–62.
  • Kim S, Lee DJ, Kwag DS, et al. (2014). Acid pH-activated glycol chitosan/fullerene nanogels for efficient tumor therapy. Carbohydr Polym 101:692–8.
  • Kim SK, Youn YS, Oh KT, et al. (2017). Development of pH-responsive starch–glycol chitosan nanogels for proapoptotic (KLAKLAK)2 peptide delivery. J Bioactive Compat Polym 32:345–54.
  • Koo OM, Rubinstein I, Onyuksel H. (2005). Role of nanotechnology in targeted drug delivery and imaging: a concise review. Nanomedicine 1:193–212.
  • Koontongkaew S. (2013). The tumor microenvironment contribution to development, growth, invasion and metastasis of head and neck squamous cell carcinomas. J Cancer 4:66–83.
  • LeBrun A, Zhu L. (2018). Magnetic nanoparticle hyperthermia in cancer treatment: history, mechanism, imaging-assisted protocol design, and challenges. In: Shrivastava D, ed. Theory and applications of heat transfer in humans. Chichester, UK: John Wiley & Sons Ltd, 631–67.
  • Lee JO, Lee MJ, Kim D, et al. (2014). A molecular zipping/unzipping nano-vehicles sensitive to tumor extracellular pH. J Bioactive Compat Polym 29:368–81.
  • Lee JO, Oh KT, Kim D, et al. (2014). pH-sensitive short worm-like micelles targeting tumors based on the extracellular pH. J Mater Chem B 2:6363–70.
  • Lee SY, Lee H, In I, et al. (2014). pH/redox/photo responsive polymeric micelle via boronate ester and disulfide bonds with spiropyran-based photochromic polymer for cell imaging and anticancer drug delivery. Eur Polym J 57:1–10.
  • Li L, Yang W-W, Xu D-G. (2019). Stimuli-responsive nanoscale drug delivery systems for cancer therapy. J Drug Target 27:423–33.
  • Li Q, Wen Y, Wen J, et al. (2016). A new biosafe reactive oxygen species (ROS)-responsive nanoplatform for drug delivery. RSC Adv 6:38984–9.
  • Li Q, Wen Y, You X, et al. (2016). Development of a reactive oxygen species (ROS)-responsive nanoplatform for targeted oral cancer therapy. J Mater Chem B 4:4675–82.
  • Li W, Tao C, Wang J, et al. (2019). MMP-responsive in situ forming hydrogel loaded with doxorubicin-encapsulated biodegradable micelles for local chemotherapy of oral squamous cell carcinoma. RSC Adv 9:31264–73.
  • Li X, Li L, Huang Y, et al. (2017). Synergistic therapy of chemotherapeutic drugs and MTH1 inhibitors using a pH-sensitive polymeric delivery system for oral squamous cell carcinoma. Biomater Sci 5:2068–78.
  • Li X, Lovell JF, Yoon J, et al. (2020). Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat Rev Clin Oncol 17:657–18.
  • Lin Q, Yang Y, Hu Q, et al. (2017). Injectable supramolecular hydrogel formed from α-cyclodextrin and PEGylated arginine-functionalized poly(l-lysine) dendron for sustained MMP-9 shRNA plasmid delivery. Acta Biomater 49:456–71.
  • Link S, El-Sayed MA. (2000). Shape and size dependence of radiative, non-radiative and photothermal properties of gold nanocrystals. Int Rev Phys Chem 19:409–53.
  • Liu T, Chen S, Wu X, et al. (2019). Folate-targeted pH and redox dual stimulation-responsive nanocarrier for codelivering of docetaxel and TFPI-2 for nasopharyngeal carcinoma therapy. ACS Appl Bio Mater 2:1830–41.
  • Liu T, Xue W, Ke B, et al. (2014). Star-shaped cyclodextrin-poly(l-lysine) derivative co-delivering docetaxel and MMP-9 siRNA plasmid in cancer therapy. Biomaterials 35:3865–72.
  • Liu Y, Gao F-P, Zhang D, et al. (2014). Molecular structural transformation regulated dynamic disordering of supramolecular vesicles as pH-responsive drug release systems. J Control Release 173:140–7.
  • Liu Z, Shi J, Zhu B, et al. (2020). Development of a multifunctional gold nanoplatform for combined chemo-photothermal therapy against oral cancer. Nanomedicine 15:661–76.
  • Ma C, Shi L, Huang Y, et al. (2017). Nanoparticle delivery of Wnt-1 siRNA enhances photodynamic therapy by inhibiting epithelial–mesenchymal transition for oral cancer. Biomater Sci 5:494–501.
  • Ma D, Lin Q-M, Zhang L-M, et al. (2014). A star-shaped porphyrin–arginine functionalized poly(l-lysine) copolymer for photo-enhanced drug and gene co-delivery. Biomaterials 35:4357–67.
  • Marur S, Forastiere AA. (2016). Head and neck squamous cell carcinoma: update on epidemiology, diagnosis, and treatment. Mayo Clin Proc 91:386–96.
  • Matsumura Y, Maeda H. (1986). A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent Smancs. Cancer Res 46:6387–92.
  • Mehanna H, Paleri V, West CML, et al. (2011). Head and neck cancer – part 1: epidemiology, presentation, and preservation. Clin Otolaryngol 36:65–8.
  • Meulemans J, Delaere P, Vander Poorten V. (2019). Photodynamic therapy in head and neck cancer: indications, outcomes, and future prospects. Curr Opin Otolaryngol Head Neck Surg 27:136–41.
  • Miao L, Liu C, Ge J, et al. (2014). Antitumor effect of TRAIL on oral squamous cell carcinoma using magnetic nanoparticle-mediated gene expression. Cell Biochem Biophys 69:663–72.
  • Nam J, Son S, Ochyl LJ, et al. (2018). Chemo-photothermal therapy combination elicits anti-tumor immunity against advanced metastatic cancer. Nat Commun 9:1074.
  • Narmani A, Rezvani M, Farhood B, et al. (2019). Folic acid functionalized nanoparticles as pharmaceutical carriers in drug delivery systems. Drug Dev Res 80:404–24.
  • Olusanya T, Haj Ahmad R, Ibegbu D, et al. (2018). Liposomal Drug Delivery Systems and Anticancer Drugs. Molecules 23:907.
  • Pérez-Sayáns M, Somoza-Martín JM, Barros-Angueira F, et al. (2010). Multidrug resistance in oral squamous cell carcinoma: the role of vacuolar ATPases. Cancer Lett 295:135–43.
  • Qian X, Nie X, Yao W, et al. (2018). Reactive oxygen species in cancer stem cells of head and neck squamous cancer. Semin Cancer Biol 53:248–57.
  • Qiao Y, Wan J, Zhou L, et al. (2019). Stimuli-responsive nanotherapeutics for precision drug delivery and cancer therapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol 11:e1527.
  • Rao L, Bu L-L, Ma L, et al. (2018). Platelet-facilitated photothermal therapy of head and neck squamous cell carcinoma. Angew Chem 130:998–1003.
  • Rofstad EK, Mathiesen B, Kindem K, et al. (2006). Acidic extracellular pH promotes experimental metastasis of human melanoma cells in athymic nude mice. Cancer Res 66:6699–707.
  • Rosenthal EL, Matrisian LM. (2006). Matrix metalloproteases in head and neck cancer. Head Neck 28:639–48.
  • Saiyin W, Wang D, Li L, et al. (2014). Sequential release of autophagy inhibitor and chemotherapeutic drug with polymeric delivery system for oral squamous cell carcinoma therapy. Mol Pharm 11:1662–75.
  • Salahpour Anarjan F. (2019). Active targeting drug delivery nanocarriers: ligands. Nano-Struct Nano-Objects 19:100370.
  • Saravanakumar G, Kim J, Kim WJ. (2017). Reactive-oxygen-species-responsive drug delivery systems: promises and challenges. Adv Sci 4:1600124.
  • Sasikala ARK, GhavamiNejad A, Unnithan AR, et al. (2015). A smart magnetic nanoplatform for synergistic anticancer therapy: manoeuvring mussel-inspired functional magnetic nanoparticles for pH responsive anticancer drug delivery and hyperthermia. Nanoscale 7:18119–28.
  • Shi S, Zhang L, Zhu M, et al. (2018). Reactive oxygen species-responsive nanoparticles based on PEGlated prodrug for targeted treatment of oral tongue squamous cell carcinoma by combining photodynamic therapy and chemotherapy. ACS Appl Mater Interfaces 10:29260–72.
  • Shim MS, Xia Y. (2013). A reactive oxygen species (ROS)-responsive polymer for safe, efficient, and targeted gene delivery in cancer cells. Angew Chem 125:7064–7.
  • Son J, Yi G, Yoo J, et al. (2019). Light-responsive nanomedicine for biophotonic imaging and targeted therapy. Adv Drug Deliv Rev 138:133–47.
  • Song C, Tang C, Xu W, et al. (2020). Hypoxia-targeting multifunctional nanoparticles for sensitized chemotherapy and phototherapy in head and neck squamous cell carcinoma. Int J Nanomedicine 15:347–61.
  • Song R, Peng S, Lin Q, et al. (2019). pH-responsive oxygen nanobubbles for spontaneous oxygen delivery in hypoxic tumors. Langmuir 35:10166–72.
  • Su Z, Liu D, Chen L, et al. (2019). CD44-targeted magnetic nanoparticles kill head and neck squamous cell carcinoma stem cells in an alternating magnetic field. Int J Nanomed 14:7549–60.
  • Sun C, Li X, Du X, et al. (2018). Redox-responsive micelles for triggered drug delivery and effective laryngopharyngeal cancer therapy. Int J Biol Macromol 112:65–73.
  • Taghizadeh B, Taranejoo S, Monemian SA, et al. (2015). Classification of stimuli-responsive polymers as anticancer drug delivery systems. Drug Deliv 22:145–55.
  • Tan G, Zhong Y, Yang L, et al. (2020). A multifunctional MOF-based nanohybrid as injectable implant platform for drug synergistic oral cancer therapy. Chem Eng J 390:124446.
  • Tarassoli SP, de Pinillos Bayona AM, Pye H, et al. (2017). Cathepsin B-degradable, NIR-responsive nanoparticulate platform for target-specific cancer therapy. Nanotechnology 28:055101.
  • Tawfik SM, Sharipov M, Huy BT, et al. (2018). Naturally modified nonionic alginate functionalized upconversion nanoparticles for the highly efficient targeted pH-responsive drug delivery and enhancement of NIR-imaging. J Ind Eng Chem 57:424–35.
  • Trachootham D, Alexandre J, Huang P. (2009). Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat Rev Drug Discov 8:579–91.
  • Virós D, Camacho M, Zarraonandia I, et al. (2013). Prognostic role of MMP-9 expression in head and neck carcinoma patients treated with radiotherapy or chemoradiotherapy. Oral Oncol 49:322–5.
  • Wang B, Wang J-H, Liu Q, et al. (2014). Rose-Bengal-conjugated gold nanorods for in vivo photodynamic and photothermal oral cancer therapies. Biomaterials 35:1954–66.
  • Wang B-K, Yu X-F, Wang J-H, et al. (2016). Gold-nanorods-siRNA nanoplex for improved photothermal therapy by gene silencing. Biomaterials 78:27–39.
  • Wang D, Fei B, Halig LV, et al. (2014). Targeted iron-oxide nanoparticle for photodynamic therapy and imaging of head and neck cancer. ACS Nano 8:6620–32.
  • Wang F, Wang M, Zhao L, et al. (2019). A new biosafe reactive oxygen species responsive nanoplatform for targeted oral squamous cell carcinoma therapy. Mat Expr 9:1076–81.
  • Wang H, Fu Z, Li W, et al. (2019). The synthesis and application of nano doxorubicin–indocyanine green matrix metalloproteinase-responsive hydrogel in chemophototherapy for head and neck squamous cell carcinoma. Int J Nanomedicine 14:623–38.
  • Wang M, Xiao Y, Li Y, et al. (2019). Reactive oxygen species and near-infrared light dual-responsive indocyanine green-loaded nanohybrids for overcoming tumour multidrug resistance. Eur J Pharm Sci 134:185–93.
  • Wang M, Zhai Y, Ye H, et al. (2019). High co-loading capacity and stimuli-responsive release based on cascade reaction of self-destructive polymer for improved chemo-photodynamic therapy. ACS Nano 13:7010–23.
  • Wang X, Gao S, Qin Z, et al. (2018). Evans Blue derivative-functionalized gold nanorods for photothermal therapy-enhanced tumor chemotherapy. ACS Appl Mater Interfaces 10:15140–9.
  • Wang Y, Wan G, Li Z, et al. (2017). PEGylated doxorubicin nanoparticles mediated by HN-1 peptide for targeted treatment of oral squamous cell carcinoma. Int J Pharm 525:21–31.
  • Wang Y, Xie D, Pan J, et al. (2019). A near infrared light-triggered human serum albumin drug delivery system with coordination bonding of indocyanine green and cisplatin for targeting photochemistry therapy against oral squamous cell cancer. Biomater Sci 7:5270–82.
  • Wang Y, Xie Y, Li J, et al. (2017). Tumor-penetrating nanoparticles for enhanced anticancer activity of combined photodynamic and hypoxia-activated therapy. ACS Nano 11:2227–38.
  • Wang Z, Meng Q, Li S. (2020). The role of NIR fluorescence in MDR cancer treatment: from targeted imaging to phototherapy. CMC 27:5510–29.
  • Wei Z, Yin X, Cai Y, et al. (2018). Antitumor effect of a Pt-loaded nanocomposite based on graphene quantum dots combats hypoxia-induced chemoresistance of oral squamous cell carcinoma. Int J Nanomed 13:1505–24.
  • Weinberg F, Ramnath N, Nagrath D. (2019). Reactive oxygen species in the tumor microenvironment: an overview. Cancers 11:1191.
  • Wen S, Liu H, Cai H, et al. (2013). Targeted and pH-responsive delivery of doxorubicin to cancer cells using multifunctional dendrimer-modified multi-walled carbon nanotubes. Adv Healthc Mater 2:1267–76.
  • Wu T-T, Zhou S-H. (2015). Nanoparticle-based targeted therapeutics in head-and-neck cancer. Int J Med Sci 12:187–200.
  • Xing Q, Li N, Jiao Y, et al. (2015). Near-infrared light-controlled drug release and cancer therapy with polymer-caged upconversion nanoparticles. RSC Adv 5:5269–76.
  • Xiong J, Gao H. (2017). Matrix metalloproteases-responsive nanomaterials for tumor targeting diagnosis and treatment. J Microencapsul 34:440–53.
  • Yan T, He J, Liu R, et al. (2020). Chitosan capped pH-responsive hollow mesoporous silica nanoparticles for targeted chemo-photo combination therapy. Carbohydr Polym 231:115706.
  • Yao Q, Kou L, Tu Y, et al. (2018). MMP-responsive ‘smart’ drug delivery and tumor targeting. Trends Pharmacol Sci 39:766–81.
  • Zhang K, Guan X, Qiu Y, et al. (2016). A pH/glutathione double responsive drug delivery system using molecular imprint technique for drug loading. Appl Surf Sci 389:1208–13.
  • Zhang W. (2015). Supramolecular hydrogels co-loaded with camptothecin and doxorubicin for sustainedly synergistic tumor therapy. J Mater Chem B 3:2127–36.
  • Zhang Z, Zhuang L, Lin Y, et al. (2020). Novel drug delivery system based on hollow mesoporous magnetic nanoparticles for head and neck cancers – targeted therapy in vitro and in vivo. Am J Cancer Res 10:350–64.
  • Zhao Y, Zhou Y, Wang D, et al. (2015). pH-responsive polymeric micelles based on poly(2-ethyl-2-oxazoline)-poly(d,l-lactide) for tumor-targeting and controlled delivery of doxorubicin and P-glycoprotein inhibitor. Acta Biomater 17:182–92.
  • Zhou L, Wang H, Li Y. (2018). Stimuli-responsive nanomedicines for overcoming cancer multidrug resistance. Theranostics 8:1059–74.
  • Zhou Q, Zhang L, Yang T, et al. (2018). Stimuli-responsive polymeric micelles for drug delivery and cancer therapy. Int J Nanomed 13:2921–42.