AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1,014.3 KB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review | Open Access

Stimuli-responsive nanocarriers for therapeutic applications in cancer

Xubo Zhao1 ( )Jie Bai1Wenjing Yang2 ( )
Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
Show Author Information

Abstract

Cancer has become a very serious challenge with aging of the human population. Advances in nanotechnology have provided new perspectives in the treatment of cancer. Through the combination of nanotechnology and therapeutics, nanomedicine has been successfully used to treat cancer in recent years. In terms of nanomedicine, nanocarriers play a key role in delivering therapeutic agents, reducing severe side effects, simplifying the administration scheme, and improving therapeutic efficacies. Modulations of the structure and function of nanocarriers for improved therapeutic efficacy in cancer have attracted increasing attention in recent years. Stimuli-responsive nanocarriers penetrate deeply into tissues and respond to external or internal stimuli by releasing the therapeutic agent for cancer therapy. Notably, stimuli-responsive nanocarriers reduce the severe side effects of therapeutic agents, when compared with systemic chemotherapy, and achieve controlled drug release at tumor sites. Therefore, the development of stimuli-responsive nanocarriers plays a crucial role in drug delivery for cancer therapy. This article focuses on the development of nanomaterials with stimuli-responsive properties for use as nanocarriers, in the last few decades. These nanocarriers are more effective at delivering the therapeutic agent under the control of external or internal stimuli. Furthermore, nanocarriers with theranostic features have been designed and fabricated to confirm their great potential in achieving effective treatment of cancer, which will provide us with better choices for cancer therapy.

References

1

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA-Cancer J Clin. 2018; 68: 394-424.

2

Senapati S, Mahanta AK, Kumar S, Maiti P. Controlled drug delivery vehicles for cancer treatment and their performance. Signal Transduct Tar. 2018; 3: 7.

3

Miao Y, Qiu Y, Zhang M, Yan K, Zhang P, Lu S, et al. Aqueous self-assembly of block copolymers to form manganese oxide-based polymeric vesicles for tumor microenvironment-activated drug delivery. Nano-Micro Lett. 2020; 12: 124.

4

Qiu Y, Zhu Z, Miao Y, Zhang P, Jia X, Liu Z, et al. Polymerization of dopamine accompanying its coupling to induce self-assembly of block copolymer and application in drug delivery. Polym Chem. 2020; 11: 2811-21.

5

Liu Y, Luo J, Chen X, Liu W, Chen T. Cell membrane coating technology: a promising strategy for biomedical applications. Nano-Micro Lett. 2019; 11: 100.

6

Wang J, Fang L, Li P, Ma L, Na W, Cheng C, et al. Inorganic Nanozyme with combined selfoxygenation/degradable capabilities for sensitized cancer immunochemotherapy. Nano-Micro Lett. 2019; 11: 74.

7

Kakkar A, Traverso G, Farokhzad OC, Weissleder R, Langer R. Evolution of macromolecular complexity in drug delivery systems. Nat Rev Chem. 2017; 1: 0063.

8

Zhao X, Qiu Y, Miao Y, Liu Z, Yang W, Hou H. Unconventional preparation of polymer/amorphous manganese oxide-based biodegradable nanohybrids for low premature release and acid/glutathione-activated magnetic resonance imaging. ACS Appl Nano Mater. 2018; 1: 2621-31.

9

Yang W, Zhao X. Glutathione-induced structural transform of double-cross-linked PEGylated nanogel for efficient intracellular anticancer drug delivery. Mol Pharm. 2019; 16: 2826-37.

10

Chen H, Gu Z, An H, Chen C, Chen J, Cui R, et al. Precise nanomedicine for intelligent therapy of cancer. Sci China Chem. 2018; 61: 1503-52.

11

Li Y, Lu H, Liang S, Xu S. Dual stable nanomedicines prepared by cisplatin-crosslinked camptothecin prodrug micelles for effective drug delivery. ACS Appl Mater Inter. 2019; 11: 20649-59.

12

Gaio E, Conte C, Esposito D, Miotto G, Quaglia F, Moret F, et al. Co-delivery of docetaxel and disulfonate tetraphenyl chlorin in one nanoparticle produces strong synergism between chemo- and photodynamic therapy in drug-sensitive and -resistant cancer cells. Mol Pharm. 2018; 15: 4599-611.

13

Jin Z, Chen Z, Wu K, Shen Y, Guo S. Investigation of migration-preventing tracheal stent with high dose of 5-fluorouracil or paclitaxel for local drug delivery. ACS Appl Bio Mater. 2018; 1: 1328-36.

14

Hu Z, Qiao C, Xia Z, Li F, Han J, Wei Q, et al. A luminescent Mg-Metal-Organic Framework for sustained release of 5-fluorouracil: appropriate host-guest interaction and satisfied acid-base resistance. ACS Appl Mater Inter. 2020; 12: 14914-23.

15

Guissi NEI, Li H, Xu Y, Semcheddine F, Chen M, Su Z, et al. Mitoxantrone- and Folate-TPGS2k conjugate hybrid micellar aggregates to circumvent toxicity and enhance efficiency for breast cancer therapy. Mol Pharm. 2017; 14: 1082-94.

16

Zheng K, Chen R, Sun Y, Tan Z, Liu Y, Cheng X, et al. Cantharidin-loaded functional mesoporous titanium peroxide nanoparticles for non-small cell lung cancer targeted chemotherapy combined with high effective photodynamic therapy. Thorac Cancer. 2020; 11: 1476-86.

17

Liu X, Jiang J, Chan R, Ji Y, Lu J, Liao Y, et al. Improved efficacy and reduced toxicity using a custom-designed irinotecan-delivering silicasome for orthotopic colon cancer. ACS Nano. 2019; 13: 38-53.

18

Folkman J, Long D. The use of silicone rubber as a carrier for prolonged drug therapy. J Surg Res. 1964; 4: 139-42.

19

Batz HG, Franzmann G, Ringsdorf H. Model reactions for synthesis of pharmacologically active polymers by way of monomeric and polymeric reactive esters. Angew Chem Int Ed Engl. 1972; 11: 1103-4.

20

Langer R, Folkman J. Polymers for the sustained release of proteins and other macromolecules. Nature. 1976; 263: 797-800.

21

Peppas NA, Merrill EW. Development of semicrystalline poly(vinyl alcohol) hydrogels for biomedical applications. J Biomed Mater Res. 1977; 11: 423-34.

22

Zhao X, Wei Z, Zhao Z, Miao Y, Qiu Y, Yang W, et al. Design and development of graphene oxide nanoparticle/chitosan hybrids showing pH-sensitive surface charge-reversible ability for efficient intracellular doxorubicin delivery. ACS Appl Mater Inter. 2018; 10: 6608-17.

23

Sun L, Wei H, Zhang X, Meng C, Kang G, Ma W, et al. Synthesis of polymeric micelles with dual-functional sheddable PEG stealth for enhanced tumor-targeted drug delivery. Polym Chem. 2020; 11: 4469-76.

24

Liao J, Song Y, Liu C, Li D, Zheng H, Lu B. Dual-drug delivery based charge-conversional polymeric micelles for enhanced cellular uptake and combination therapy. Polym Chem. 2019; 10: 5879-93.

25

Miao Y, Zhao X, Qiu Y, Liu Z, Yang W, Jia X. Metal-organic framework-assisted nanoplatform with hydrogen peroxide/glutathione dual-sensitive on-demand drug release for targeting tumors and their microenvironment. ACS Appl Bio Mater. 2019; 2: 895-905.

26

Zhang X, Zhang X, Sun L, Liu F, Wang M, Peng J, et al. Fabrication of biocleavable shell cross-linked hybrid micelles for controlled drug release using a reducible silica monomer. Chem Commun. 2018; 54: 13495-8.

27

Heuberger R, Sukhorukov G, Vörös J, Textor M, Möhwald H. Biofunctional polyelectrolyte multilayers and microcapsules: control of non-specific and bio-specific protein adsorption. Adv Funct Mater. 2005; 15: 357-66.

28

Prencipe G, Tabakman SM, Welsher K, Liu Z, Goodwin AP, Zhang L, et al. PEG branched polymer for functionalization of nanomaterials with ultralong blood circulation. J Am Chem Soc. 2009; 131: 4783-7.

29

Mosqueira VCF, Legrand P, Gulik A, Bourdon O, Gref R, Labarre D, et al. Relationship between complement activation, cellular uptake and surface physicochemical aspects of novel PEG-modified nanocapsules. Biomaterials. 2001; 22: 2967-79.

30

Cui M, Liu S, Song B, Guo D, Wang J, Hu G, et al. Fluorescent silicon nanorods-based nanotheranostic agents for multimodal imaging-guided photothermal therapy. Nano-Micro Lett. 2019; 11: 73.

31

Liu Q, Luo Q, Ju Y, Song G. Role of the mechanical microenvironment in cancer development and progression. Cancer Biol Med. 2020; 17: 282-92.

32

Sun Z, Song C, Wang C, Hu Y, Wu J. Hydrogel-based controlled drug delivery for cancer treatment: a review. Mol Pharm. 2020; 17: 373-91.

33

Dong Y, Tu Y, Wang K, Xu C, Yuan Y, Wang J. A general strategy for macrotheranostic prodrug activation: synergy by tumor acidic microenvironment and bioorthogonal chemistry. Angew Chem Int Ed. 2020; 132: 7235-9.

34

Miao Y, Qiu Y, Yang W, Guo Y, Hou H, Liu Z, et al. Charge reversible and biodegradable nanocarriers showing dual pH-/reduction-sensitive disintegration for rapid site-specific drug delivery. Colloids Surf B: Biointerfaces. 2018; 169: 313-20.

35

Qu X, Yang Z. Benzoic-imine based physiological pH responsive materials for biomedical applications. Chem Asian J. 2013; 11: 2633-41.

36

Puglisi A, Bayir E, Timur S, Yagci Y. pH-Responsive polymersome microparticles as smart cyclodextrin-releasing agents. Biomacromolecules. 2019; 20: 4001-7.

37

Wang H, Liu G, Gao H, Wang Y. A pH-responsive drug delivery system with an aggregation-induced emission feature for cell imaging and intracellular drug delivery. Polym Chem. 2015; 6: 4715-8.

38

Jia X, Zhao X, Tian K, Zhou T, Li J, Zhang R, et al. Fluorescent copolymer-based prodrug for pH-triggered intracellular release of DOX. Biomacromolecules. 2015; 16: 3624-31.

39

Zhao X, Liu P. pH-sensitive fluorescent hepatocyte-targeting multilayer polyelectrolyte hollow microspheres as a smart drug delivery system. Mol Pharm. 2014; 11: 1599-610.

40

Zhao X, Yao Y, Tian K, Zhou T, Jia X, Li J, et al. Leakage-free DOX/PEGylated chitosan micelles fabricated via facile one-step assembly for tumor intracellular pH-triggered release. Eur J Pharm Biopharm. 2016; 108: 91-9.

41

Sobolev AS, Aliev RA, Kalmykov SN. Radionuclides emitting short-range particles and modular nanotransporters for their delivery to target cancer cells. Russ Chem Rev. 2016; 85: 1011-32.

42

Osminkina LA, Nikolaev AL, Sviridov AP, Andronova NV, Tamarov KP, Gongalsky MB, et al. Porous silicon nanoparticles as efficient sensitizers for sonodynamic therapy of cancer. Micropor Mesopor Mat. 2015; 210: 169-75.

43

Kuppusamy P, Li H, Ilangovan G, Cardounel AJ, Zweier JL, Yamada K, et al. Noninvasive imaging of tumor redox status and its modification by tissue glutathione levels. Cancer Res. 2002; 62: 307.

44

Schafer FQ, Buettner GR. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med. 2001; 30: 1191-212.

45

Li T, Pan S, Zhuang H, Gao S, Xu H. Selenium-containing carrier-free assemblies with aggregation-induced emission property combine cancer radiotherapy with chemotherapy. ACS Appl Bio Mater. 2020; 3: 1283.

46

Deng Z, Yuan S, Xu RX, Liang H, Liu S. Reduction-triggered transformation of crosslinking modules of disulfide-containing micelles with chemically tunable rates. Angew Chem Int Ed. 2018; 57: 8896-900.

47

Zhao X, Liu P. Reduction-responsive core-shell-corona micelles based on triblock copolymers: novel synthetic strategy, characterization, and application as a tumor microenvironment-responsive drug delivery system. ACS Appl Mater Inter. 2015; 7: 166-74.

48

Wang L, Zhu K, Cao W, Sun C, Lu C, Xu H. ROS-triggered degradation of selenide-containing polymers based on selenoxide elimination. Polym Chem. 2019; 10: 2039-46.

49

Gao S, Li T, Guo Y, Sun C, Xianyu B, Xu H. Selenium-containing nanoparticles combine the NK cells mediated immunotherapy with radiotherapy and chemotherapy. Adv Mater. 2020; 32: 1907568.

50

Chen Y, Ye D, Wu M, Chen H, Zhang L, Shi J, et al. Break-up of two-dimensional MnO2 nanosheets promotes ultrasensitive pH-triggered theranostics of cancer. Adv Mater. 2014; 26: 7019-26.

51

Lu Y, Aimetti AA, Langer R, Gu Z. Bioresponsive materials. Nat Rev Mater. 2017; 2: 16075.

52

Kamaly N, Yameen B, Wu J, Farokhzad OC. Degradable controlled-release polymers and polymeric nanoparticles: mechanisms of controlling drug release. Chem Rev. 2016; 116: 2602-63.

53

Harnoy AJ, Rosenbaum I, Tirosh E, Ebenstein Y, Shaharabani R, Beck R, et al. Enzyme-responsive amphiphilic PEG-dendron hybrids and their assembly into smart micellar nanocarriers. Chem Soc. 2014; 136: 7531-4.

54

Zelzer M, Todd SJ, Hirst AR, Mcdonald TO, Ulijn RV. Enzyme responsive materials: design strategies and future developments. Biomater Sci. 2013; 1: 11-39.

55

Chen D, Liu W, Shen Y, Mu H, Zhang Y, Liang R, et al. Effects of a novel pH-sensitive liposomewith cleavable esterase-catalyzed and pH-responsive double smart mPEGlipid derivative on ABC phenomenon. Int J Nanomed. 2011; 6: 2053-61.

56

Guarnieri D, Biondi M, Yu H, Belli V, Falanga AP, Cantisani M, et al. Tumor-activated prodrug (TAP)-conjugated nanoparticles with cleavable domains for safe doxorubicin delivery. Biotechnol Bioeng. 2015; 112: 601-11.

57

Zhu L, Kate P, Torchilin VP. Matrix metalloprotease 2-responsive multifunctional liposomal nanocarrier for enhanced tumor targeting. ACS Nano. 2012; 6: 3491-8.

58

Wang N, Chen X, Ding R, Yang X, Li J, Yu X, et al. Synthesis of high drug loading, reactive oxygen species and esterase dual-responsive polymeric micelles for drug delivery. RSC Adv. 2019; 9: 2371-8.

59

Wu C, Hu W, Wei Q, Qiao L, Gao Y, Lv Y, et al. Controllable growth of core-shell nanogels via esterase-induced self-assembly of peptides for drug delivery. J Biomed Nanotechnol. 2018; 14: 354-61.

60

Stern R, Jedrzejas MJ. Hyaluronidases: their genomics, structures, and mechanisms of action. Chem Rev. 2006; 106: 818-39.

61

Chen J, Huang K, Chen Q, Deng C, Zhang J, Zhong Z. Tailor-making fluorescent hyaluronic acid microgels via combining microfluidics and photoclick chemistry for sustained and localized delivery of herceptin in tumors. ACS Appl Mater Inter. 2018; 10: 3929-37.

62

Bai Y, Liu C, Chen D, Liu C, Zhuo L, Li H, et al. β-Cyclodextrinmodified hyaluronic acid-based supramolecular selfassemblies for pH- and esterase-dual-responsive drug delivery. Carbohydr Polym. 2020; 246: 116654.

63

Zhao W, Zhao Y, Wang Q, Liu T, Sun J, Zhang R. Remote light-responsive nanocarriers for controlled drug delivery: advances and perspectives. Small. 2019; 15: 1903060.

64

He C, Wang S, Yu Y, Shen H, Zhao Y, Gao H, et al. Advances in biodegradable nanomaterials for photothermal therapy of cancer. Cancer Biol Med. 2016; 13: 299-312.

65

Zhao X, Qi M, Liang S, Tian K, Zhou T, Jia X, et al. Synthesis of photo- and pH dual-sensitive amphiphilic copolymer PEG43-b-P(AA76-co-NBA35-co-tBA9) and its micellization as leakage-free drug delivery system for UV-triggered intracellular delivery of doxorubicin. ACS Appl Mater Inter. 2016; 8: 22127-34.

66

Hu D, Li Y, Niu Y, Li L, He J, Liu X, et al. Photo-responsive reversible micelles based on azobenzene-modified poly(carbonate)s via azide-alkyne click chemistry. RSC Adv. 2014; 4: 47929-36.

67

Bai Y, Liu C, Song X, Zhuo L, Bu H, Tian W. Photo- and pH-dual-responsive β-cyclodextrin-based supramolecular prodrug complex self-assemblies for programmed drug delivery. Chem Asian J. 2018; 13: 3903-11.

68

Kozlovskaya V, Liu F, Yang Y, Ingle KA, Qian S, Halade GV, et al. Temperature-responsive polymersomes of poly(3-methyl-N-vinylcaprolactam)-block-poly(N-vinylpyrrolidone) to decrease doxorubicin-induced cardiotoxicity. Biomacromolecules. 2019; 20: 3989-4000.

69

Luo G, Chen W, Zhang X. 100th anniversary of macromolecular science viewpoint: poly(N-isopropylacrylamide)-based thermally responsive micelles. ACS Macro Lett. 2020; 9: 872-81.

70

Scarpa JS, Mueller DD, Klotz, IM. Slow hydrogen/deuterium exchange in a non-alpha-helical polyamide. J Am Chem Soc. 1967; 89: 6024-30.

71

An X, Zhu A, Luo H, Ke H, Chen H, Zhao Y. Rational design of multi-stimuli-responsive nanoparticles for precise cancer therapy. ACS Nano. 2016; 10: 5947-58.

72

Maeda Y, Nakamura T, Ikeda I. Hydration and phase behavior of poly(N-vinylcaprolactam) and poly(N-vinylpyrrolidone) in water. Macromolecules. 2002; 35: 217-22.

73

Manouras T, Vamvakaki M. Field responsive materials: photo-, electro-, magnetic- and ultrasound-sensitive polymers. Polym Chem. 2017; 8: 74-96.

74

Gao Z, Fain HD, Rapoport N. Controlled and targeted tumor chemotherapy by micellar-encapsulated drug and ultrasound. J Control Release. 2005; 102: 203-22.

75

Chen W, Du J. Ultrasound and pH dually responsive polymer vesicles for anticancer drug delivery. Sci Rep. 2013; 3: 2162.

76

Wang Y, Yin T, Su Z, Qiu C, Wang Y, Zheng R, et al. Highly uniform ultrasound-sensitive nanospheres produced by a pH-induced micelle-to-vesicle transition for tumor-targeted drug delivery. Nano Res. 2018; 11: 3710-21.

77

Yin T, Wang P, Li J, Zheng R, Zheng B, Cheng D, et al. Ultrasound-sensitive siRNA-loaded nanobubbles formed by hetero-assembly of polymeric micelles and liposomes and their therapeutic effect in gliomas. Biomaterials. 2013; 34: 4532-43.

78

Nikolaev AL, Gopin AV, Bozhevol’nov VE, Treshalina HM, Andronova NV, Melikhov IV, et al. Combined method of ultrasound therapy of oncological diseases. Russ J Gen Chem. 2015; 85: 303-20.

79

Plummer R, Wilson RH, Calvert H, Boddy AV, Griffin M, Sludden J, et al. A phase Ⅰ clinical study of cisplatin-incorporated polymeric micelles (NC-6004) in patients with solid tumours. Br J Cancer. 2011; 104: 593-8.

80

Cabral H, Kataoka K. Progress of drug-loaded polymeric micelles into clinical studies. J Control Release. 2014; 190: 465-76.

81

Zhou Q, Sun X, Zeng L, Liu J, Zhang Z. A randomized multicenter phase Ⅱ clinical trial of mitoxantrone-loaded nanoparticles in the treatment of 108 patients with unresected hepatocellular carcinoma. Nanomed Nanotechnol Biol Med. 2009; 5: 419-23.

82

Harada M, Bobe I, Saito H, Shibata N, Tanaka R, Hayashi T, et al. Improved anti-tumor activity of stabilized anthracycline polymeric micelle formulation, NC-6300. Cancer Sci. 2011; 102: 192-9.

83

Dobrovolskaia MA, McNeil SE. Immunological properties of engineered nanomaterials. Nat Nanotechnol. 2007; 2: 469-78.

84

Wicki A, Witzigmann D, Balasubramanian V, Huwyler J. Nanomedicine in cancer therapy: challenges, opportunities, and clinical applications. J Controll Release. 2015; 200: 138-57.

85

Yan S, Zhao P, Yu T, Gu N. Current applications and future prospects of nanotechnology in cancer immunotherapy. Cancer Biol Med. 2019; 16: 487-97.

86

Kibria G, Ramos EK, Wan Y, Gius DR, Liu H. Exosomes as a drug delivery system in cancer therapy: potential and challenges. Mol Pharm. 2018; 15: 3625-33.

87

Li Z, Xiao C, Yong T, Li Z, Gan L, Yang X. Influence of nanomedicine mechanical properties on tumor targeting delivery. Chem Soc Rev. 2020; 49: 2273-90.

Cancer Biology & Medicine
Pages 319-335
Cite this article:
Zhao X, Bai J, Yang W. Stimuli-responsive nanocarriers for therapeutic applications in cancer. Cancer Biology & Medicine, 2021, 18(2): 319-335. https://doi.org/10.20892/j.issn.2095-3941.2020.0496

75

Views

1

Downloads

44

Crossref

39

Web of Science

42

Scopus

Altmetrics

Received: 25 August 2020
Accepted: 16 November 2020
Published: 01 May 2021
©2021 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return