Smart Nanomaterials for Biomedical Applications: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor:

Recent advances in nanotechnology have forced the obtaining of new materials with multiple functionalities. Due to their reduced dimensions, nanomaterials exhibit outstanding physio-chemical functionalities: increased absorption and reactivity, higher surface area, molar extinction coefficients, tunable plasmonic properties, quantum effects, and magnetic and photo properties. However, in the biomedical field, it is still difficult to use tools made of nanomaterials for better therapeutics due to their limitations (including non-biocompatible, poor photostabilities, low targeting capacity, rapid renal clearance, side effects on other organs, insufficient cellular uptake, and small blood retention), so other types with controlled abilities must be developed, called “smart” nanomaterials.

  • smart nanomaterials
  • stimuli-responsive polymers
  • biomedical applications

1. Introduction

From the oldest times, humanity has tried to mimic nature in the way that living organisms adapt their behavior to environmental conditions to improve survival. It is well known that living systems from nature can dynamically change their properties in a smart way for adapting to the surrounding environment. Some examples are given by the Mimosa pudica plant which responds to stimuli such as temperature and light by undergoing a change in leaf direction [1]; by pinecones [2], wheat awn [3], and orchid tree seedpods [4] which adopt different shapes, responding to the changing environmental humidity; and by the Venus flytrap [5] which is able to capture insects by rapidly closing its leaves, among many others [6,7]. The powerful abilities of the biological systems abovementioned in converting energy and executing multiple tasks inspired the researchers to develop “stimuli-responsive” materials with biomimetic behavior and a high potential of use in smart or intelligent devices.

To our knowledge, the first complete report of “intelligent materials” defined as “the materials that respond to environmental changes at the most optimum condition and manifest their own functions according to these changes” was made by Toshinori Takagi in April 1990 [8]. At that time, the coverage and the achievability of this concept was not comprehensible, but it was anticipated to open an unused field in science and innovation [9]. Nowadays, the term “intelligent material” is synonymous with “stimuli-responsive material” or “smart material” and has gained a growing interest in researchers’ concerns due to the development of advanced technologies and the increased need for new materials that meet the new requirements.

Richard Feynman, laureate of the Nobel Prize, was the first to introduce, in 1959, the nanotechnology concept. He had a revolutionary vision in a lecture entitled: “Why can’t we write the entire 24 volumes of the Encyclopedia Britannica on the head of a pin?” [10]. The term “nanotechnology” was used and defined in 1974, by Norio Taniguchi, as “nanotechnology mainly consists of the processing of separation, consolidation, and deformation of materials by one atom or one molecule” [11]. In 1997–1998, the perception of nanotechnology was more a “science fiction” vision, still being far from the practical applications [12], but in the early 2000s, nanomaterials were intensively studied and finally utilized in practice. As the field of biomedical engineering is developing new insights, the demand for highly functionalized biomaterials is increasing. Despite the astonishing diversity and complexity of living systems, they all share the possibility to react to environmental changes, crucial for maintaining normal functions. This need for adaptation has led to the development of smart nanomaterials, defined as materials that can react to a large variety of stimuli by adapting their own properties such as shape, surface area, size, permeability, solubility, mechanical properties, and others. Depending on the capacity of the nanomaterial to restore its initial state, the response can be reversible or not. In this specific situation, polymer-based materials have substantiated themselves as sharp choices in creating upgraded responsive frameworks on the grounds that their structure permits regulating their properties. A large variety of polymers have been obtained to respond to physical stimuli (temperature, light, ultrasound, electrical, magnetic, mechanical), chemical stimuli (pH, solvent, electrochemical), or biological stimuli (enzymes). A special type of polymer is dual or multi-stimuli-responsive, in light of the fact that it simultaneously reacts to multiple stimuli. As a rule, the polymer responsivity is directed by the science of the monomers and their distribution/concentration in the polymer chains [13].

2. Types of Stimuli

Smart nanomaterials are categorized in different groups by means of the applied stimuli. The properties of smart nanomaterials are modified by external triggers in a controlled way [15,16,17,18,19,20,21,22,23]. By considering their various properties, many kinds of smart nanomaterials are known.

2.1. Physico-Responsive Nanomaterials

Examples of physico-sensitive nanomaterials and their applications are listed in Table 1 [24-40]. Recent works [41-76] describe the design of phisico-responsive smart nanomaterials and their applications in the biomedical field.

Table 1. Examples of physico-responsive nanomaterials and their biomedical applications.

Nr. Crt. Stimuli Nanomaterial Application Reference
1. Temperature Poly(ethylene oxide)a-poly(propylene oxide)b-
poly(ethylene oxide)a
PEO-PPO-PEO
Oral drug delivery, wound healing [24]
2. Temperature Gold nanoparticles—Pluronic®F127-Hydroxypropyl methylcellulose
AuNPs-PF127-HPMC
Drug delivery, photothermal platform, skin wound healing [25]
3. Temperature Poly(oligo(ethylene glycol) methacrylate –co-poly(glycidal methacrylate) copolymers/poly(lactic
acid-co-glycolic acid)
P(OEGMA-co-PGMA) copolymers/PLGA
Tissue engineering [26]
4. Temperature Collagen- or chitosan-based Drug delivery [27]
5. Temperature Poly(N-isopropylacrylamide)- poly(N,N-dimethylacrylamide)- poly(acrylic acid)
PNIPAM-PDMA-PAA
Drug delivery [28]
6. Temperature Poly(Nisopropylacrylamide-co-sulfobetaine methacrylate) nanogel
PNS nanogels
Diagnosis/chemotherapy [29]
7. Electrical Poly(3,4-ethylenedioxythiophene)-coated poly(lactic acid-co-glycolic acid) nanofiber
PEDOT-coated PLGA nanofiber
Drug delivery [30]
8. Electrical Fe3O4/Polyaniline
Fe3O4/PANI
Antimicrobial, drug delivery [31]
9. Electrical Polyaniline/gold nanocomposite
PANI/AuNCs
Immunosensor detection of chronic kidney disease [32]
10. Electrical Polyaniline, poly(3,4-ethylenedioxythiophene)
PANIP, PEDOT
Neural prostheses [33,34]
11. Electrochemical Biosynthesized gold nanoparticles/ poly(catechol)/graphene sheets/glassy carbon electrode
Bio AuNP/Pol/Gr/GCE
Biosensor, DNA mutation and acute lymphoblastic leukemia detection [35]
12. Light poly(ethylene glycol)
PEG
Switchable fluorescent probes [36]
13. Light Ruthenium-containing block copolymer
Poly-Ru nanoparticles
In vivo photodynamic therapy and photochemotherapy [37]
14. Magnetic Fe3O4/methoxy poly(ethylene glycol)-poly- (lactide) composite nanocapsules
Fe3O4/MePEG-PLA composite nanocapsules
MRI [38]
15. Magnetic Trastuzumab (Tra, a humanized monoclonal antibody that specifically recognizes HER2)- doxorubicin
poly(vinyl alcohol)/ single-component thiol-functionalized poly (methacrylic acid T-DOX
PVA/PMASH magnetic nanocapsules
Tumor therapy [39]
16. Magnetic 3D collagen hydrogel Directed neuronal regeneration [40]

2.2. Chemical-Responsive Nanomaterials

Examples of chemical-sensitive nanomaterials and their applications are listed in Table 2 [77-88]. Recent works [89-102] describe the design of chemical-responsive smart nanomaterials and their applications in the biomedical field.

Table 2. Examples of chemical-responsive nanomaterials and their biomedical applications.

Nr. Crt. Stimuli Nanomaterial Application Reference.
1. pH Ppoly (ethylene glycol)-Ag nanoparticle
PEG-Ag NPs
Antibacterial, wound healing [77]
2. pH Hybrid ultra-pH-sensitive (HyUPS) nanotransistor
HyUPS nanotransistors
Receptor-mediated endocytosis in tumor cells [78]
3. pH Layered double hydroxides-zinc (II) phthalocyanine containing octasulfonate nanohybrid
LDH-ZnPcS8 nanohybrid
Theranostics [79]
4. pH Melanin-like nanoparticles Photoacoustic imaging of tumors [80]
5. pH polylactic acid-Resveratrol
PLA-RSV
Drug delivery [81]
6. pH Poly(carboxybetaine methacrylate)-nanodiamonds
PCBSA-@-NDs
Theranostics [82]
7. Redox Poly (ethylene glycol)-Pluronic F68-nanoscale covalent organic frameworks
F68@SS-COFs
Cancer therapy [83]
8. Redox Hyaluronic acid–chitosan–lipoic acid nanoparticles
(HACSLA-NPs)
Breast cancer therapy [84]
9. Redox Folate redox-responsive chitosan nanoparticles
FTC-NPs
Anticancer drug delivery [85]
10. Redox Poly (ethylene glycol) conjugated to paclitaxel via disulfide linkage
PEG2000-S-S-PTX
Prodrug for breast cancer cells [86]
11. Redox Prodrug/AgNPs hybrid nanoparticles Drug delivery [87]
12. Redox P[(2-((2- ((camptothecin)-oxy)ethyl)disulfanyl)ethylmethacrylate) -co- (2-(D-galactose)methylmethacryl-ate)] and silver nanoparticles
P(MACPTS-co-MAGP)@AgNPs nanoparticles
Drug release [88]

2.3. Biological-Responsive Nanomaterials

Examples of biological-sensitive nanomaterials and their applications are listed in Table 3 [103-113]. Recent works [114-127] describe the design of biological-responsive smart nanomaterials and their applications in the biomedical field.

Table 3. Examples of biological-responsive nanomaterials and their biomedical applications.

Nr. Crt. Stimuli Nanomaterial Application Reference.
1. Glucose Acetalated dextran nanoparticles
Ac-Dex Nps
Glycemic control [103]
2. Glucose Boronic acid-derived polymers Drug delivery [104]
3. Glucose Glycidyl methacrylated dextran/Concanavalin A
Dex-GMA/Con A
ConA micro/nanospheres
Insulin treatment [105]
4. Glucose Chitosan-g-polyethylene glycol monomethyl ether nanocomplex
CS-g-(mPEG) NP
Oral insulin delivery [106]
5. Glucose Hyaluronic Acid (HA)-coated calcium carbonate NPs Oral insulin delivery [107]
6. Glucose Chitosan/poly(gamma-glutamic acid) nanoparticles Oral insulin delivery [108]
7. Glucose Carboxymethyl chitosan-phenylboronic acid-Lvaline nanoparticles
(CMCS-PBA-LV) NPs
Oral administration of insulin [109]
8. Enzyme Nanoplatform formed from Ti substrates modified with
layer-by layer mesoporous silica nanoparticles-silver nanoparticles
LBL@MSN-Ag nanoparticles
Tissue growth in vivo and, simultaneously, treat implant-associated bacterial infection [110]
9. Enzyme Adenosine triphosphate coated with silver nanoparticles
ATP-Ag nanoparticles
Participate in signal transduction and protein activity [111]
10. Enzyme Activatable low-molecular weight protamine—poly(ethylene glycol) poly(ε-caprolactone) nanoparticles—loaded with paclitaxel
ALMWP-NP-PTX
Glioblastoma therapy [112]
11. Enzyme Layer-by-layer assembly of poly(2-oxazoline)-based materials Therapeutic delivery [113]

2.4. Dual and Multi-Responsive Nanomaterials

A step forward for biomedical applications is attained when the smart nanomaterials are simultaneously sensitive to more stimuli. The nanomaterials which are sensitive to a few sorts of stimuli are the key for expanding the efficacy of drug delivery and for supporting the diagnosis by monitoring a few physiological changes at once.

The development of nanomaterials with both diagnostic and therapeutic properties is the most powerful technological frontier for moving forward to nanotheranostics. The demand for these technologies is based on the advantage of multiple functions such as multimodal imaging, synergistic therapies, and targeting. The working system of nanotheranostics depends on biological, chemical, and physical triggers considering the activation of the diagnostic and/or the therapeutic properties only at the infected site. In this era of the “war on cancer”, the dual and multi-stimuli-responsive methodology is undeniably appropriate for theranostics as some properties can provide diagnostics, while others could initiate therapy and curing. Consequently, multi-stimuli-sensitive polymers are drawing in expanding consideration for their advantages in the biomedical field.

Multi-stimuli-sensitive polymeric nanoparticles were developed as emerging targeting drug delivery systems. External stimuli such as temperature and pH facilitate the emergence of nanoparticles, while stimuli such as light, the magnetic field, the temperature, and ultrasonic are intended to control drug delivery. Examples of multi-sensitive nanomaterials and their applications are listed in Table 4 [128-142].Recent works [143-150] describe the design of multi-responsive smart nanomaterials and their applications in the biomedical field.

Nr. Crt. Stimuli Nanomaterial Application Ref.
1. pH/redox/temperature N,N0 -bis(acryloyl)cystamine, Poly(N-isopropylacrylamide), 2-hydroxyethylmethacrylate, Methacrylic acid, a disulfide bond contained cross-linker, and doxorubicin
SS-NPs@DOX
Drug delivery [128]
2. Ultrasound/pH Poly(ethylene oxide, 2-(diethylamino)ethyl methacrylate, (2-tetrahydrofuranyloxy)ethyl methacrylate
PEO43-b-P(DEA33-stat-TMA
Drug release [129]
3. Temperature/magnetic field Poly(N-isopropylacrylamide)- Magnetic nanoparticles
b-PNIPAM-mNPs
The isolation of diagnostic targets that can be used in point-of-care devices [130]
4. Light/pH rGO-PDA nanosheets Drug delivery, phototherapy [131]
5. pH/magnetic field Magnetic nanoparticles
MFNPs
Targeting, drug delivery, MRI [132]
6. Temperature/pH Poly(N-isopropylacrylamide)
pNIPAM
Drug release [133]
7. pH/light/enzyme Copper sulfide nanoparticles
CuS NPs
Theranostics [134]
8. pH/redox Thiol-modified polylysine- indocyanine green/ poly(ethylene glycol) nanoparticles
PLL-ICG/DPEG Nps
Photothermal and photodynamic therapy [135]
9. pH/redox Poly (ethylene glycol) –polylacticacid-thioketal groups-Paclitaxel-(Maleimide thioether) Chlorin e6
mPEG-PLA-TKI-PTX nanoparticles and Ce6-(SS-mal-)-Ce6 (PNPCe6)
Chemotherapy, drug release [136]
10. pH/redox Histidine -4 polyamidoamine dendrimer -Disulfide bonds- (poly (ethylene glycol)- Transferrin
(His-PAMAM-ss-PEG-Tf, HP-ss-PEG-Tf) nanocarrier
Anticancer drug delivery [137]
11. pH/redox Lipoic acid ethylenediamine- Polyethylene glycol diglycidyl ether- Llysine
poly(LAE-co-PGDE-co-Lys) core-crosslinked nano aggregate
Anticancer drug delivery [138]
12. pH/redox Paclitaxel- poly(6-O-methacryloyl-d-galactopyranose)- gemcitabine/ N-acetyl-d-glucosamine(NAG)-poly(styrene-alt-maleic anhydride)-b-polystyrene
PTXL-ss-PMAGP-GEM/NAG NLCs
Anticancer drug delivery [139]
13. UV light/redox/pH Six-arm star-shaped amphiphilic copolymer with poly (caprolactone) -bpoly (acrylic acid) -b-poly (poly (ethylene glycol) methyl ether methacrylate) Anticancer drug delivery [140]
14. pH/temperature Poly(NIPAM)nanogel @ Fe3O4 NPs/poly(acrylic acid) -graft—κ—carrageenan Drug delivery [141]
15. Redox/pH/temperature Nanogels based on alginate and cystamine Anticancer drug delivery [142]

3. Advances in Plasmonic Nanomaterials

A special class of smart nanomaterials is derived from plasmonic nanoparticles used in innovative sensitive tools for diagnostics and therapeutics. The collective electronic (plasmon) resonances of noble/coinage metal nanoparticles enable a strong optical response essential in applications such as photocatalysis, sensing, photothermal heating, and enhanced fluorescence. Biomedical applications rely on plasmonic nanoparticles’ properties to absorb or scatter light at near-infrared wavelengths, transmissive in the human body [151]. A large number of applications misuse the extraordinary properties of metals to support electromagnetic waves at their surfaces, through the oscillation of their conduction electrons known as surface plasmons. The local dielectric environment, size, structure, shape, and composition determine the surface plasmon polariton modes enabling nanostructures to focus and direct light down to the nanoscale. The ability of plasmonic nanostructures to strongly interact with light at wavelengths that significantly exceed their dimensions led to the appearance of the nanoplasmoic field [152]. Consequently, most recent strategies for the design and manufacture of plasmonic nanostructures for accurately controlling light have opened new entryways for the applications that were recently perceived as impossible.

Recent studies have shown that by targeting gold nanoparticles to the cell nucleus region, the nuclear stiffness is enhanced, slowing down the migration and invasion speed of cancer cells and suppressing metastasis [153]. Further, gold nanoparticles exhibit high contrast in photothermal therapeutic treatments, as well as photoacoustic, optical coherence, and X-ray CT imaging. Conjugates of gold nanoparticles present augmented binding affinity, long circulatory half-life, size-enhanced tumor uptake, increased targeting selectivity, high biocompatibility, and rapid transport kinetics. If all those properties are put together in a highly multifunctional platform, one can obtain an increasingly selective and potent oncologic treatment [154].

As diagnosis is the key in the screening and treatment of human diseases, modern-day researchers developed sensitive tools for real-time and accurate tracking of the treatment effect. In a recent paper, the authors obtained a core–shell structure MPs@ SiO2@Pd–Au with a crystalline magnetic core, amorphous silica interaction layer, and Pd–Au shell for medulloblastoma diagnosis and radiotherapy evaluation. Owing to the plasmonic and alloying effects, MPs@SiO2@Pd–Au may contribute to efficient electron transfer and high surface stability under laser irradiation during the laser desorption/ionization process [155]. Other authors described the design of a plasmonic gold nano-island (pGold) chip assay for enhanced diagnosis and monitoring of myocardial infarction [156]. A multifunctional platinum nanoreactor intended for point-of-care metabolic analysis, visual detection, and mass spectrometry fingerprinting for in vitro pancreatic cancer diagnostics was designed using controlled core–shell structured Fe3O4@SiO2@Pt particles [157]. Another work describing the application of laser desorption/ionization mass spectrometry in large-scale clinical in vitro cervical cancer diagnosis utilized a plasmonic chip with Au nanoparticles deposited on a dopamine bubble layer [158].

An increasing interest was paid to the field of thermoplasmonics, defined as plasmonic nanoparticles remotely controlled by light to release heat on the nanoscale volumes. The capability of using plasmonic materials as photothermal agents is based on a combination of properties such as the high density of free electrons, the absence of thermobleaching, resonances that enhance light–matter interaction, and low losses for noble metals. Those materials are best choices in applications requiring spatially confined heating, such as in nanosurgery and photoacoustic and photothermal imaging [159]. Recent works described the capability of gold nanorods to convert NIR radiation into heat for antibacterial application without affecting cells’ viability and proliferation [160] and of keratin-coated gold nanoparticles to kill the brain cancer cells by photothermal therapy [161]. The photothermal therapy induced by the presence of gold nanoparticles in a system capable to develop immunotherapy represents a major breakthrough in the fight against malignant solid tumors. This synergistic new approach was comprehensively described moving from in vivo studies to clinical trial applications in patients suffering from solid tumors. Although those systems hold great promise in nanomedicine, there are still risks involved, such as the wrong cells being targeted, unknown long-term side effects, and unwanted immune reaction systems. For this reason, the combination of hyperthermia with chemotherapeutic activity or cancer immunotherapy demonstrated improved care of oncological patients [162].

The properties of Au and Ag nanoparticles have inspired the field of plasmonic nanoparticles in the last two decades, but recently, non-noble metals have been the subject of quickly expanding interest as less expensive, more practical alternatives. Colloidal nanocrystals functionalized with silica have been utilized for plasmon-driven photocatalysis and surface-enhanced Raman spectroscopy at visible and near-infrared wavelengths due to their enhanced stability in water and efficient broadband photothermal heating [163].

References

  1. Amador-Vargas, S.; Dominguez, M.; León, G.; Maldonado, B.; Murillo, J.; Vides, G.L. Leaf-folding response of a sensitive plant shows context-dependent behavioral plasticity. Plant Ecol. 2014215, 1445–1454. [Google Scholar] [CrossRef]
  2. Reyssat, E.; Mahadevan, L. Hygromorphs: From pine cones to biomimetic bilayers. J. R. Soc. Interface 20096, 951–957. [Google Scholar] [CrossRef] [PubMed]
  3. Elbaum, R.; Zaltzman, L.; Burgert, I.; Fratzl, P. The role of wheat awns in the seed dispersal unit. Science 2007316, 884–886. [Google Scholar] [CrossRef] [PubMed]
  4. Erb, R.M.; Sander, J.S.; Grisch, R.; Studart, A.R. Self-shaping composites with programmable bioinspired microstructures. Nat. Commun. 20134, 1712. [Google Scholar] [CrossRef] [PubMed]
  5. Forterre, Y.; Skotheim, J.M.; Dumais, J.; Mahadevan, L. How the Venus flytrap snaps. Nature 2005433, 421–425. [Google Scholar] [CrossRef]
  6. Fu, Q.; Li, C. Robotic modelling of snake traversing large, smooth obstacles reveals stability benefits of body compliance. R. Soc. Open Sci. 20207, 191192. [Google Scholar] [CrossRef]
  7. Teyssier, J.; Saenko, S.V.; Van Der Marel, D.; Milinkovitch, M.C. Photonic crystals cause active colour change in chameleons. Nat. Commun. 20156, 6368. [Google Scholar] [CrossRef] [PubMed]
  8. Totakagi, T. A Concept of intelligent materials. J. Intell. Mater. Syst. Struct. 19901, 149–156. [Google Scholar] [CrossRef]
  9. Totakagi, T. Proceedings of the International Workshop on Intelligent Materials, Tsukuba Science City; The Society of Non-Traditional Technology: Tokyo, Japan, 1989. [Google Scholar]
  10. Feynman, R. There’s plenty of room at the bottom. Eng. Sci. 196023, 22–36. [Google Scholar]
  11. Taniguchi, N. On the basic concept of ‘nano-technology’. In International Conference on Production Engineering, Part II; Japan Society of Precision Engineering: Tokyo, Japan, 1974. [Google Scholar]
  12. Roco, M.C. International strategy for nanotechnology research and development. J. Nanoparticle Res. 20013, 353–360. [Google Scholar] [CrossRef]
  13. Aflori, M.; Butnaru, M.; Tihauan, B.-M.; Doroftei, F. Eco-friendly method for tailoring biocompatible and antimicrobial surfaces of poly-L-lactic acid. Nanomaterials 20199, 428. [Google Scholar] [CrossRef]
  14. Jing, L.; Yang, C.; Zhang, P.; Zeng, J.; Li, Z.; Gao, M. Nanoparticles weaponized with built-in functions for imaging-guided cancer therapy. View 20201, e19. [Google Scholar] [CrossRef]
  15. Genchi, G.G.; Marino, A.; Tapeinos, C.; Ciofani, G. Smart materials meet multifunctional biomedical devices: Current and prospective implications for nanomedicine. Front. Bioeng. Biotechnol. 20175, 80. [Google Scholar] [CrossRef] [PubMed]
  16. Cabane, E.; Zhang, X.; Langowska, K.; Palivan, C.G.; Meier, W. Stimuli-responsive polymers and their applications in nano-medicine. Biointerphases 20127, 9. [Google Scholar] [CrossRef]
  17. Thangudu, S. Next generation nanomaterials: Smart nanomaterials, significance, and biomedical applications. In Applications of Nanomaterials in Human Health; Khan, F.A., Ed.; Springer: Singapore, 2020; pp. 287–312. [Google Scholar]
  18. Pinteala, M.; Abadie, M.J.M.; Rusu, R.D. Smart supra-and macro-molecular tools for biomedical applications. Materials 202013, 3343. [Google Scholar] [CrossRef] [PubMed]
  19. Das, S.S.; Bharadwaj, P.; Bilal, M.; Barani, M.; Rahdar, A.; Taboada, P.; Bungau, S.; Kyzas, G.Z. Stimuli-responsive polymeric nanocarriers for drug delivery, imaging, and theragnosis. Polymers 202012, 1397. [Google Scholar] [CrossRef] [PubMed]
  20. Aflori, M. (Ed.) Intelligent Polymers for Nanomedicine and Biotechnologies; CRC Press: Boca Raton, FL, USA, 2017. [Google Scholar]
  21. Flory, P.J. Thermodynamics of high polymer solutions. J. Chem. Phys. 19419, 660. [Google Scholar] [CrossRef]
  22. Huggins, M.L. Some properties of solutions of long-chain compounds. J. Phys. Chem. 194246, 151–158. [Google Scholar] [CrossRef]
  23. Aguilar, M.R.; San Roman, J. Introduction to smart polymers and their applications. In Smart Polymers and their Applications; Woodhead Publish: Cambridge, UK, 2014; Volume 1, pp. 1–11. [Google Scholar]
  24. Liu, Y.; Yang, F.; Feng, L.; Yang, L.; Chen, L.; Wei, G.; Lu, W. In vivo retention of poloxamer based in situ hydrogels for vaginal application in mouse and rat models. Acta Pharm. Sin. B 20177, 502–509. [Google Scholar] [CrossRef]
  25. Arafa, M.G.; El-Kased, R.F.; Elmazar, M.M. Thermoresponsive gels containing gold nanoparticles as smart antibacterial and wound healing agents. Sci. Rep. 20188, 1–16. [Google Scholar] [CrossRef]
  26. Debashish, R.W.L.; Brooks, A.; Sumerlin, B.S. New directions in thermoresponsive polymers. Chem. Soc. Rev. 201342, 7214–7243. [Google Scholar]
  27. Hoffman, A.S. Stimuli-responsive polymers: Biomedical applications and challenges for clinical translation. Adv. Drug Deliv. Rev. 201365, 10–16. [Google Scholar] [CrossRef]
  28. Chen, Y.; Gao, Y.; Da Silva, L.P.; Pirraco, R.P.; Ma, M.; Yang, L.; Reis, R.L.; Chen, J. A thermo-/pH-responsive hydrogel (PNIPAM-PDMA-PAA) with diverse nanostructures and gel behaviors as a general drug carrier for drug release. Polym. Chem. 20189, 4063–4072. [Google Scholar] [CrossRef]
  29. Zheng, A.; Wu, D.; Fan, M.; Wang, H.; Liao, Y.; Wang, Q.; Yang, Y. Injectable zwitterionic thermosensitive hydrogels with low-protein adsorption and combined effect of photothermal-chemotherapy. J. Mater. Chem. B 20208, 10637–10649. [Google Scholar] [CrossRef]
  30. Abidian, M.R.; Kim, D.-H.; Martin, D.C. Conducting-polymer nanotubes for controlled drug. Adv. Mater. 200618, 405–409. [Google Scholar] [CrossRef] [PubMed]
  31. Hossain, M.K.; Minami, H.; Hoque, S.M.; Rahman, M.M.; Sharafat, M.K.; Begum, M.F.; Islam, M.E.; Ahmad, H. Mesoporous electromagnetic composite particles: Electric current responsive release of biologically active molecules and antibacterial properties. Colloids Surf. B 2019181, 85–93. [Google Scholar] [CrossRef] [PubMed]
  32. Shaikh, M.O.; Srikanth, B.; Zhu, P.-Y.; Chuang, C.-H. Impedimetric immunosensor utilizing polyaniline/Gold nanocomposite-modified screen-printed electrodes for early detection of chronic kidney disease. Sensors 201919, 3990. [Google Scholar] [CrossRef] [PubMed]
  33. Otto, K.J.; Schmidt, C.E. Neuron-targeted electrical modulation. Science 2020367, 1303–1304. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, J.; Kim, Y.S.; Joubert, L.-M.; Jiang, Y.; Wang, H.; Fenno, L.E.; Tok, J.B.-H.; Paşca, S.P.; Shen, K.; Bao, Z.; et al. Genetically targeted chemical assembly of functional materials in living cells, tissues, and animals. Science 2020367, 1372–1376. [Google Scholar] [CrossRef] [PubMed]
  35. Mazloum-Ardakani, M.; Barazesh, B.; Khoshroo, A.; Moshtaghiun, M.; Sheikhha, M.H. A new composite consisting of electro-synthesized conducting polymers, graphene sheets and biosynthesized gold nanoparticles for biosensing acute lymphoblastic leukemia. Bioelectrochemistry 2018121, 38–45. [Google Scholar] [CrossRef]
  36. Klajn, R. Spiropyran-based dynamic materials. Chem. Soc. Rev. 201443, 148–184. [Google Scholar] [CrossRef] [PubMed]
  37. Sun, W.; Li, S.; Haupler, B.; Liu, J.; Jin, S.; Steffen, W.; Schubert, U.S.; Butt, H.-J.; Liang, X.-J.; Wu, S. An amphiphilic ruthenium polymetallodrug for combined photodynamic therapy and potochemotherapy in vivoAdv. Mater. 201729, 1603702. [Google Scholar] [CrossRef] [PubMed]
  38. Xu, B.; Dou, H.; Tao, K.; Sun, K.; Ding, J.; Shi, W.; Guo, X.; Li, J.; Zhang, D.; Sun, K. Two-in-one fabrication of Fe3O4/MePEG-PLA composite nanocapsules as a potential ultrasonic/MRI dual contrast agent. Langmuir 201127, 12134–12142. [Google Scholar] [CrossRef]
  39. Chiang, C.; Shen, Y.-S.; Liu, J.-J.; Shyu, W.-C.; Chen, S.-Y. Synergistic combination of multistage magnetic guidance and optimized ligand density in targeting a nanoplatform for enhanced cancer therapy. Adv. Health Mater. 20165, 2131–2141. [Google Scholar] [CrossRef]
  40. Antman-Passig, M.; Shefi, O. Remote magnetic orientation of 3D collagen hydrogels for directed neuronal regeneration. Nano Lett. 201616, 2567–2573. [Google Scholar] [CrossRef] [PubMed]
  41. Ogoina, D. Fever, fever patterns and diseases called ‘fever’—A review. J. Infect. Public Health 20114, 108–124. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, R.; Fraylich, M.; Saunders, B.R. Thermoresponsive copolymers: From fundamental studies to applications. Colloid Polym. Sci. 2009287, 627–643. [Google Scholar] [CrossRef]
  43. Zhang, Q.; Weber, C.; Schubert, U.S.; Hoogenboom, R. Thermoresponsive polymers with lower critical solution temperature: From fundamental aspects and measuring techniques to recommended turbidimetry conditions. Mater. Horizons 20174, 109–116. [Google Scholar] [CrossRef]
  44. Niebuur, B.-J.; Chiappisi, L.; Jung, F.; Zhang, X.; Schulte, A.; Papadakis, C.M. Kinetics of mesoglobule formation and growth in aqueous poly(N-isopropylacrylamide) Solutions: Pressure jumps at low and at high pressure. Macromolecules 201952, 6416–6427. [Google Scholar] [CrossRef]
  45. Sahle, F.F.; Gulfam, M.; Lowe, T.L. Design strategies for physical-stimuli-responsive programmable nanotherapeutics. Drug Discov. Today 201823, 992–1006. [Google Scholar] [CrossRef]
  46. Marcelo, G.; Areias, L.R.; Viciosa, M.T.; Gaspar-Martinho, J.; Farinha, J.P.S. PNIPAm-based microgels with a UCST response. Polymer 2017116, 261–267. [Google Scholar] [CrossRef]
  47. Bischofberger, I.; Calzolari, D.C.E.; Rios, P.D.L.; Jelezarov, I.; Trappe, V. Hydrophobic hydration of poly-N-isopropyl acrylamide: A matter of the mean energetic state of water. Sci. Rep. 20144, 4377. [Google Scholar] [CrossRef] [PubMed]
  48. Kotsuchibashi, Y. Recent advances in multi-temperature-responsive polymeric materials. Polym. J. 202052, 681–689. [Google Scholar] [CrossRef]
  49. Alejo, T.; Uson, L.; Arruebo, M. Reversible stimuli-responsive nanomaterials with on-off switching ability for biomedical ap-plications. J. Control. Release 2019314, 162–176. [Google Scholar] [CrossRef] [PubMed]
  50. Tamaki, M.; Kojima, C. pH-Switchable LCST/UCST-type thermosensitive behaviors of phenylalanine-modified zwitterionic dendrimers. RSC Adv. 202010, 10452–10460. [Google Scholar] [CrossRef]
  51. Qiao, S.; Wang, H. Temperature-responsive polymers: Synthesis, properties, and biomedical applications. Nano Res. 201811, 5400–5423. [Google Scholar] [CrossRef]
  52. Bellotti, E.; Schilling, A.L.; Little, S.R.; Decuzzi, P. Injectable thermoresponsive hydrogels as drug delivery system for the treatment of central nervous system disorders: A review. J. Control. Release 2021329, 16–35. [Google Scholar] [CrossRef]
  53. Hogan, K.J.; Mikos, A.G. Biodegradable thermoresponsive polymers: Applications in drug delivery and tissue engineering. Polymer 2020211, 123063. [Google Scholar] [CrossRef]
  54. Lavrador, P.; Esteves, M.R.; Gaspar, V.M.; Mano, J.F. Stimuli-responsive nanocomposite hydrogels for biomedical applications. Adv. Funct. Mater. 2020, 2005941. [Google Scholar] [CrossRef]
  55. Custodio, C.A.; Reis, R.L.; Mano, J.F.; Del Campo, A. Smart instructive polymer substrates for tissue engineering. In Smart Polymers and Their Applications, 2nd ed.; Aguilar, M.R., Roman, J.S., Eds.; Woodhead Publishing in Materials: Cambridge, UK, 2014; Volume 10, pp. 10301–10326. [Google Scholar]
  56. Sun, H.; Feng, M.; Chen, S.; Wang, R.; Luo, Y.; Yin, B.; Li, J.; Wang, X.J. Near-infrared photothermal liposomal nanoantagonists for amplified cancer photodynamic therapy. Mater. Chem. B 20208, 7149–7159. [Google Scholar] [CrossRef]
  57. Ha, J.H.; Shin, H.H.; Choi, H.W.; Lim, J.H.; Mo, S.J.; Ahrberg, C.D.; Lee, J.M.; Chung, B.G. Electro-responsive hydrogel-based microfluidic actuator platform for photothermal therapy. Lab Chip 202020. [Google Scholar] [CrossRef]
  58. Hosseini-Nassab, N.; Samanta, D.; Abdolazimi, Y.; Annes, J.P.; Zare, R.N. Electrically controlled release of insulin using polypyrrole nanoparticles. Nanoscale 20179, 143–149. [Google Scholar] [CrossRef]
  59. Groenendaal, B.L.; Jonas, F.; Freitag, D.; Pielartzik, H.; Reynolds, J.R. Poly(3,4-ethylenedioxythiophene) and its derivatives: Past, present, and future. Adv. Mater. 200012, 481–494. [Google Scholar] [CrossRef]
  60. Murdan, S. Electro-responsive drug delivery from hydrogels. J. Control. Release 200392, 1–17. [Google Scholar] [CrossRef]
  61. Svirskis, D.; Travas-Sejdic, J.; Rodgers, A.; Garg, S. Electrochemically controlled drug delivery based on intrinsically conducting polymers. J. Control. Release 2010146, 6–15. [Google Scholar] [CrossRef] [PubMed]
  62. Delcea, M.; Möhwald, H.; Skirtach, A.G. Stimuli-responsive LbL capsules and nanoshells for drug delivery. Adv. Drug Deliv. Rev. 201163, 730–747. [Google Scholar] [CrossRef] [PubMed]
  63. Kotov, N.A.; Winter, J.O.; Clements, I.P.; Jan, E.; Timko, B.P.; Campidelli, S.; Pathak, S.; Mazzatenta, A.; Lieber, C.M.; Prato, M.; et al. Nanomaterials for Neural Interfaces. Adv. Mater. 200921, 3970–4004. [Google Scholar] [CrossRef]
  64. Sun, S.; Liang, S.; Xu, W.-C.; Xu, G.; Wu, S. Photoresponsive polymers with multi-azobenzene groups. Polym. Chem. 201910, 4389–4401. [Google Scholar] [CrossRef]
  65. Bertrand, O.; Gohy, J. Photo-responsive polymers: Synthesis and applications. Polym. Chem. 20168, 52–73. [Google Scholar] [CrossRef]
  66. Pierini, F.; Nakielski, P.; Urbanek, O.; Pawłowska, S.; Lanzi, M.; De Sio, L.; Kowalewski, T.A. Polymer-based nanomaterials for photothermal therapy: From light-responsive to multifunctional nanoplatforms for synergistically combined technologies. Biomacromolecules 201819, 4147–4167. [Google Scholar] [CrossRef]
  67. Thangudu, S.; Kalluru, P.; Vankayala, R. Preparation, cytotoxicity, and in vitro bioimaging of water soluble and highly fluorescent palladium nanoclusters. Bioengineering 20207, 20. [Google Scholar] [CrossRef]
  68. Deng, G.; Li, S.; Sun, Z.; Li, W.; Zhou, L.; Zhang, J.; Gong, P.; Cai, L. Near-infrared fluorescence imaging in the largely unexplored window of 900–1000 nm. Theranostics 20188, 4116–4128. [Google Scholar] [CrossRef]
  69. Molla, M.R.; Rangadurai, P.; Antony, L.; Swaminathan, S.; De Pablo, J.J.; Thayumanavan, S. Dynamic actuation of glassy polymerases through isomerization of a single azobenzene unit at the block copolymer interface. Nat. Chem. 201810, 659–666. [Google Scholar] [CrossRef] [PubMed]
  70. Mena-Giraldo, P.; Perez-Buitrago, S.; Londono-Berrío, M.; Ortiz-Trujillo, I.C.; Hoyos-Palacio, L.M.; Orozco, J. Photosensitive nanocarriers for specific delivery of cargo into cells. Sci. Rep. 202010, 2110. [Google Scholar] [CrossRef]
  71. Pei, C.; Liu, C.; Wang, Y.; Cheng, D.; Li, R.; Shu, W.; Zhang, C.; Hu, W.; Jin, A.; Yang, Y.; et al. FeOOH@metal–organic framework core–satellite nanocomposites for the serum metabolic fingerprinting of gynecological cancers. Angew. Chem. Int. Ed. 202059, 10831–10835. [Google Scholar] [CrossRef] [PubMed]
  72. Habash, R.W.Y.; Bansal, R.; Krewski, D.; Alhafid, H.T. Thermal therapy, part III: Ablation techniques. Crit. Rev. Biomed. Eng. 200735, 37–121. [Google Scholar] [CrossRef] [PubMed]
  73. Zborowski, M.; Dutz, S.; Häfeli, U.; Schütt, W. JMMM special issue “scientific and clinical applications of magnetic carriers”. J. Magn. Magn. Mater. 2020, 167667. [Google Scholar] [CrossRef]
  74. Kostevsek, N. A review on the optimal design of magnetic nanoparticle-based T2 MRI contrast agents. Magnetochemistry 20206, 11. [Google Scholar] [CrossRef]
  75. Shen, Y.; Goerner, F.L.; Snyder, C.; Morelli, J.; Hao, D.; Hu, D.; Li, X.; Runge, V.M. T1 relativities of gadolinium-based magnetic resonance contrast agents in human whole blood at 1.5, 3, and 7 t. Investig. Radiol. 201550, 330–338. [Google Scholar] [CrossRef]
  76. Perlman, O.; Azhari, H. MRI and Ultrasound Imaging of Nanoparticles for Medical Diagnosis. In Nanotechnology Characterization Tools for Biosensing and Medical Diagnosis; Springer Nature: Berlin/Heidelberg, Germany, 2018; pp. 333–365. [Google Scholar]
  77. Xie, X.; Sun, T.C.; Xue, J.; Miao, Z.; Yan, X.; Fang, W.; Li, Q.; Tang, R.; Lu, Y.; Tang, L.; et al. AG nanoparticles cluster with PH-triggered reassembly in targeting antimicrobial applications. Adv. Funct. Mater. 202030, 2000511. [Google Scholar] [CrossRef]
  78. Wang, Y.; Wang, C.; Li, Y.; Huang, G.; Zhao, T.; Ma, X.; Wang, Z.; Sumer, B.D.; White, M.A.; Gao, J. Digitization of endocytic PH by hybrid ultra-PH-sensitive nanoprobes at single-organelle resolution. Adv. Mater. 201729. [Google Scholar] [CrossRef]
  79. Li, X.; Zheng, B.-Y.; Ke, M.-R.; Zhang, Y.; Huang, J.-D.; Yoon, J. A tumor-pH-responsive supramolecular photosensitizer for Activatable photodynamic therapy with minimal in vivo skin Phototoxicity. Theranostics 20177, 2746–2756. [Google Scholar] [CrossRef]
  80. Ju, K.-Y.; Kang, J.; Pyo, J.; Lim, J.; Chang, J.H.; Lee, J.-K. pH-induced aggregated melanin nanoparticles for photoacoustic signal amplification. Nanoscale 20168, 14448–14456. [Google Scholar] [CrossRef]
  81. Bonadies, I.; Di Cristo, F.; Peluso, A.V.G.; Calarco, A.; Salle, A.D. Resveratrol-loaded electrospun membranes for the prevention of implant-associated infections. Nanomaterials 202010, 1175. [Google Scholar] [CrossRef] [PubMed]
  82. Zhou, B.; Li, J.; Lu, B.; Wu, W.; Zhang, L.; Liang, J.; Yi, J.; Li, X. Novel polyzwitterion shell with adaptable surface chemistry engineered to enhance anti-fouling and intracellular imaging of detonation nanodiamonds under tumor pHe. Front. Mater. Sci. 202014, 402–412. [Google Scholar] [CrossRef]
  83. Liu, S.; Yang, J.; Guo, R.; Deng, L.; Dong, A.; Zhang, J. Facile fabrication of redox-responsive covalent organic framework nanocarriers for efficiently loading and delivering doxorubicin. Macromol. Rapid Commun. 202041, e1900570. [Google Scholar] [CrossRef] [PubMed]
  84. Rezaei, S.; Kashanian, S.; Bahrami, Y.; Cruz, L.J.; Motiei, M. Redox-sensitive and hyaluronic acid-functionalized nanoparticles for improving breast cancer treatment by cytoplasmic 17α-methyltestosterone delivery. Molecules 202025, 1181. [Google Scholar] [CrossRef]
  85. Mazzotta, E.; De Benedittis, S.; Qualtieri, A.; Muzzalupo, R. Actively targeted and redox responsive delivery of anticancer drug by chitosan nanoparticles. Pharmaceutics 201912, 26. [Google Scholar] [CrossRef]
  86. Mutlu-Agardan, N.B.; Sarisozen, C.; Torchilin, V.P. Cytotoxicity of novel redox sensitive PEG2000-S-S-PTX micelles against drug-resistant ovarian and breast cancer cells. Pharm. Res. 202037, 1–8. [Google Scholar] [CrossRef]
  87. Mollazadeh, S.; Mackiewicz, M.; Yazdimamaghani, M. Recent advances in the redox-responsive drug delivery nanoplatforms: A chemical structure and physical property perspective. Mater. Sci. Eng. C 2021118, 111536. [Google Scholar] [CrossRef]
  88. Qiu, L.; Zhao, L.; Xing, C.; Zhan, Y. Redox-responsive polymer prodrug/AgNPs hybrid nanoparticles for drug delivery. Chin. Chem. Lett. 201829, 301–304. [Google Scholar] [CrossRef]
  89. Abhijit, A.; Hanes, J.; Ensign, L.M. Nanoparticles for oral delivery: Design, evaluation and state-of the-art. J. Control. Release 2016240, 504–526. [Google Scholar]
  90. Lund, P.A.; De Biase, D.; O’Byrne, C.; Liran, O.; Scheler, O.; Mira, N.P.; Cetecioglu, Z.; Fernández, E.N.; Bover-Cid, S.; Hall, R.; et al. Understanding how microorganisms respond to acid PH is central to their control and successful exploitation. Front. Microbiol. 202011, 556140. [Google Scholar] [CrossRef] [PubMed]
  91. Garcia-Fernandez, L.; Mora-Boza, A.; Reyes-Ortega, F. pH-responsive polymers: Properties, synthesis and applications. In Smart Polymers and their Applications, 2nd ed.; Aguilar, M.R., Roman, J.S., Eds.; Woodhead Publishing in Materials: Cambridge, UK, 2019; Volume 3, pp. 45–86. [Google Scholar]
  92. Constantin, M.; Bucatariu, S.; Stoica, I.; Fundueanu, G. Smart nanoparticles based onpullulan-g-poly(N-isopropylacrylamide) for controlled delivery of indomethacin. Int. J. Biol. Macromol. 201794, 698–708. [Google Scholar] [CrossRef] [PubMed]
  93. Sultankulov, B.; Berillo, D.; Sultankulova, K.; Tokay, T.; Saparov, A. Progress in the development of chitosan-based biomaterials for tissue engineering and regenerative medicine. Biomolecules 20199, 470. [Google Scholar] [CrossRef] [PubMed]
  94. Shenoy, D.; Little, S.; Langer, R.; Amiji, M. Poly(ethylene oxide)-modified poly(beta-amino ester) nanoparticles as a pH-sensitive system for tumor-targeted delivery of hydrophobic drugs: Part 2. In vivo distribution and tumor localization studies. Pharm Res. 200522, 2107–2114. [Google Scholar] [CrossRef] [PubMed]
  95. Cerritelli, S.; Velluto, D.; Hubbell, J.A. PEG-SS-PPS: Reduction-Sensitive Disulfide Block Copolymer Vesicles for Intracellular Drug Delivery. Biomacromolecules 20078, 1966–1972. [Google Scholar] [CrossRef]
  96. Zhang, L.; Ding, Y.; Wen, Q.; Ni, C. Synthesis of core-crosslinked zwitterionic polymer nano aggregates and pH/Redox responsiveness in drug controlled release. Mater. Sci. Eng. C 2020106, 110288. [Google Scholar] [CrossRef]
  97. Ling, X.; Tu, J.; Aljaeid, B.M.; Shi, B.; Tao, W.; Farokhzad, O.C.; Wang, J.; Shajii, A.; Kong, N.; Feng, C.; et al. Glutathione-responsive prodrug nanoparticles for effective drug delivery and cancer therapy. ACS Nano 201913, 357–370. [Google Scholar] [CrossRef] [PubMed]
  98. Yin, Y.; Hu, B.; Yuan, X.; Cai, L.; Gao, H.; Yang, Q. Nanogel: A versatile nano-delivery system for biomedical applications. Pharmaceutics 202012, 290. [Google Scholar] [CrossRef] [PubMed]
  99. Zhou, Q.; Zhang, L.; Yang, T.; Wu, H. Stimuli-responsive polymeric micelles for drug delivery and cancer therapy. Int. J. Nanomed. 201813, 2921–2942. [Google Scholar] [CrossRef] [PubMed]
  100. Shi, H.; Xu, M.; Zhu, J.; Liu, Y.; He, Z.; Zhang, Y.; Xu, Q.; Niu, Y. Programmed co-delivery of platinum nanodrugs and gemcitabine by a clustered nanocarrier for precision chemotherapy for NSCLC tumors. J. Mater. Chem. B 20208, 332–342. [Google Scholar] [CrossRef] [PubMed]
  101. Wilson, D.S.; Dalmasso, G.; Wang, L.; Sitaraman, S.V.; Merlin, D.; Murthy, N. Orally delivered thioketal nanoparticles loaded with TNF-α-siRNA target inflammation and inhibit gene expression in the intestines. Nat. Mater. 20109, 923–928. [Google Scholar] [CrossRef]
  102. Cheng, R.; Feng, F.; Meng, F.; Deng, C.; Feijen, J.; Zhong, Z. Glutathione-responsive nano-vehicles as a promising platform for targeted intracellular drug and gene delivery. J. Control. Release 2011152, 2–12. [Google Scholar] [CrossRef] [PubMed]
  103. Volpatti, L.R.; Matranga, M.A.; Cortinas, A.B.; Delcassian, D.; Daniel, K.B.; Langer, R.; Anderson, D.G. Glucose-responsive nanoparticles for rapid and extended self-regulated insulin delivery. ACS Nano 201914, 488–497. [Google Scholar] [CrossRef]
  104. Tang, W.; Chen, C. Hydrogel-based colloidal photonic crystal devices for glucose sensing. Polymers 202012, 625. [Google Scholar] [CrossRef]
  105. Yin, R.; He, J.; Bai, M.; Huang, C.; Wang, K.; Zhang, H.; Yang, S.-M.; Zhang, W.; Yang, S.-M. Engineering synthetic artificial pancreas using chitosan hydrogels integrated with glucose-responsive microspheres for insulin delivery. Mater. Sci. Eng. C 201996, 374–382. [Google Scholar] [CrossRef]
  106. Liu, C.; Kou, Y.; Zhang, X.; Dong, W.; Cheng, H.; Mao, S. Enhanced oral insulin delivery via surface hydrophilic modification of chitosan copolymer based self-assembly polyelectrolyte nanocomplex. Int. J. Pharm. 2019554, 36–47. [Google Scholar] [CrossRef]
  107. Liu, D.; Jiang, G.; Yu, W.; Li, L.; Tong, Z.; Kong, X.; Yao, J. Oral delivery of insulin using CaCO3-based composite nanocarriers with hyaluronic acid coatings. Mater. Lett. 2017188, 263–266. [Google Scholar] [CrossRef]
  108. Sonaje, K.; Chen, Y.J.; Chen, H.L.; Wey, S.-P.; Juang, J.-H.; Nguyen, H.-N.; Hsu, C.-W.; Lin, K.J.; Sung, H.W. Enteric-coated capsules filled with freeze-dried chitosan/poly(g-glutamic acid) nanoparticles for oral insulin delivery. Biomaterials 201031, 3384–3394. [Google Scholar] [CrossRef]
  109. Li, L.; Jiang, G.; Yu, W.; Liu, D.; Chen, H.; Liu, Y.; Tong, Z.; Kong, X.; Yao, J. Preparation of chitosan-based multifunctional nanocarriers overcoming multiple barriers for oral delivery of insulin. Mater. Sci. Eng. C 201770, 278–286. [Google Scholar] [CrossRef]
  110. Ding, Y.; Hao, Y.; Yuan, Z.; Tao, B.; Chen, M.; Lin, C.; Liu, P.; Caib, K. A dual-functional implant with an enzyme-responsive effect for bacterial infection therapy and tissue regeneration. Biomater. Sci. 20208, 1840–1854. [Google Scholar] [CrossRef]
  111. Datta, L.P.; Chatterjee, A.; Acharya, K.; De, P.; Das, M. Enzyme responsive nucleotide functionalized silver nanoparticles with effective antimicrobial and anticancer activity. New J. Chem. 201741, 1538–1548. [Google Scholar] [CrossRef]
  112. Gu, G.; Xia, H.; Hu, Q.; Liu, Z.; Jiang, M.; Kang, T.; Miao, D.; Tu, Y.; Pang, Z.; Song, Q.; et al. PEG-co-PCL nanoparticles modified with MMP-2/9 activatable low molecular weight protamine for enhanced targeted glioblastoma therapy. Biomaterials 201334, 196–208. [Google Scholar] [CrossRef]
  113. Gunawan, S.T.; Kempe, K.; Hagemeyer, C.E.; Caruso, F.; Bonnard, T.; Cui, J.; Alt, K.; Law, L.S.; Wang, X.; Westein, E.; et al. Multifunctional thrombin-activatable polymer capsules for specific targeting to activated platelets. Adv. Mater. 201527, 5153–5157. [Google Scholar] [CrossRef] [PubMed]
  114. Mann, E.; Sunni, M.; Bellin, M.D. Secretion of Insulin in Response to Diet and Hormones. Pancreapedia 2020, 1–16. [Google Scholar] [CrossRef]
  115. Mansoor, S.; Kondiah, P.P.D.; Choonara, Y.E.; Pillay, V. Polymer-based nanoparticle strategies for insulin delivery. Polymers 201911, 1380. [Google Scholar] [CrossRef]
  116. Jamwal, S.; Ram, B.; Ranote, S.; Dharela, R.; Chauhan, G.S. New glucose oxidase-immobilized stimuli-responsive dextran nanoparticles for insulin delivery. Int. J. Biol. Macromol. 2019123, 968–978. [Google Scholar] [CrossRef]
  117. Takemoto, Y.; Ajiro, H.; Asoh, T.-A.; Akashi, M. Fabrication of surface-modified hydrogels with polyion complex for controlled release. Chem. Mater. 201022, 2923–2929. [Google Scholar] [CrossRef]
  118. George, A.; Shah, P.A.; Shrivastav, P.S. Natural biodegradable polymers based nano-formulations for drug delivery: A review. Int. J. Pharm. 2019561, 244–264. [Google Scholar] [CrossRef] [PubMed]
  119. Ling, K.; Wu, H.; Neish, A.S.; Champion, J.A. Alginate/chitosan microparticles for gastric passage and intestinal release of therapeutic protein nanoparticles. J. Control. Release 2019295, 174–186. [Google Scholar] [CrossRef] [PubMed]
  120. Pham, S.H.; Choi, Y.; Choi, J. Stimuli-responsive nanomaterials for application in antitumor therapy and drug delivery. Pharmaceutics 202012, 630. [Google Scholar] [CrossRef] [PubMed]
  121. Jo, Y.; Choi, N.; Kim, K.; Koo, H.-J.; Choi, J.; Kim, H.N. Chemoresistance of cancer cells: Requirements of tumor microenvironment-mimicking in vitro models in anti-cancer drug development. Theranostics 20188, 5259–5275. [Google Scholar] [CrossRef] [PubMed]
  122. Li, N.; Cai, H.; Jiang, L.; Hu, J.; Bains, A.; Hu, J.; Gong, Q.; Luo, K.; Gu, Z. Enzyme-sensitive and amphiphilic pegylated dendrimer-paclitaxel prodrug-based nanoparticles for enhanced stability and anticancer efficacy. ACS Appl. Mater. Interfaces 20179, 6865–6877. [Google Scholar] [CrossRef] [PubMed]
  123. Hu, Q.; Katti, P.S.; Gu, Z. Enzyme-responsive nanomaterials for controlled drug delivery. Nanoscale 20146, 12273–12286. [Google Scholar] [CrossRef]
  124. Basel, M.T.; Shrestha, T.B.; Troyer, D.L.; Bossmann, S.H. Protease-sensitive, polymer-caged liposomes: A method for making highly targeted liposomes using triggered release. ACS Nano 20115, 2162–2175. [Google Scholar] [CrossRef]
  125. Thamphiwatana, S.; Gao, W.; Pornpattananangkul, D.; Zhang, Q.; Fu, V.; Li, J.; Li, J.; Obonyo, M.; Zhang, L. Phospholipase A2-responsive antibiotic delivery via nanoparticle-stabilized liposomes for the treatment of bacterial infection. J. Mater. Chem. B 20142, 8201–8207. [Google Scholar] [CrossRef]
  126. Cai, H.; Wang, X.; Zhang, H.; Sun, L.; Pan, D.; Gong, Q.; Gu, Z.; Luo, K. Enzyme-sensitive biodegradable and multifunctional polymeric conjugate as theranostic nanomedicine. Appl. Mater. Today 201811, 207–218. [Google Scholar] [CrossRef]
  127. Cheng, R.; Meng, F.; Deng, C.; Klok, H.-A.; Zhong, Z. Dual and multi-stimuli responsive polymeric nanoparticles for programmed site-specific drug delivery. Biomaterials 201334, 3647–3657. [Google Scholar] [CrossRef]
  128. Yu, B.; Song, N.; Hu, H.; Chen, G.; Shen, Y.; Cong, H. A degradable triple temperature-, pH-, and redox-responsive drug system for cancer chemotherapy. J. Biomed. Mater. Res. Part A 2018106, 3203–3210. [Google Scholar] [CrossRef]
  129. Chen, W.; Du, J. Ultrasound and PH dually responsive polymer vesicles for anticancer drug delivery. Sci. Rep. 20133, srep02162. [Google Scholar] [CrossRef] [PubMed]
  130. Lai, J.J.; Hoffman, J.M.; Ebara, M.; Hoffman, A.S.; Estournès, C.; Wattiaux, A.; Stayton, P.S. Dual magnetic-/Temperature-responsive nanoparticles for microfluidic separations and assays. Langmuir 200723, 7385–7391. [Google Scholar] [CrossRef] [PubMed]
  131. Jiang, W.; Mo, F.; Lin, Y.; Wang, X.; Xu, L.; Fu, F. Tumor targeting dual stimuli responsive controllable release nanoplatform based on DNA-conjugated reduced graphene oxide for chemo-photothermal synergetic cancer therapy. J. Mater. Chem. B 20186, 4360–4367. [Google Scholar] [CrossRef]
  132. Bhattacharya, D.; Behera, B.; Sahu, S.K.; Ananthakrishnan, R.; Maiti, T.K.; Pramanik, P. Design of dual stimuli responsive polymer modified magnetic nanoparticles for targeted anti-cancer drug delivery and enhanced MR imaging. New J. Chem. 201540, 545–557. [Google Scholar] [CrossRef]
  133. Chen, Y.; Wei, W.; Zhu, Y.; Luo, J.; Liu, R.; Liu, X. Synthesis of temperature/PH dual-stimuli-response multicompartmental microcapsules via pickering emulsion for preprogrammable payload release. ACS Appl. Mater. Interfaces 202012, 4821–4832. [Google Scholar] [CrossRef]
  134. Poudel, K.; Gautam, M.; Jin, S.G.; Choi, H.-G.; Yong, C.S.; Kim, J.O. Copper sulfide: An emerging adaptable nanoplatform in cancer theranostics. Int. J. Pharm. 2019562, 135–150. [Google Scholar] [CrossRef]
  135. Zhang, P.; Gao, Z.; Cui, J.; Hao, J. Dual-stimuli-responsive polypeptide nanoparticles for photothermal and photodynamic therapy. ACS Appl. Bio Mater. 20203, 561–569. [Google Scholar] [CrossRef]
  136. Lu, H.; Zhao, Q.; Wang, X.; Mao, Y.; Chen, C.; Gao, Y.; Sun, C.; Wang, S. Multi-stimuli responsive mesoporous silica-coated carbon nanoparticles for chemo-photothermal therapy of tumor. Colloids Surf. B 2020190, 110941. [Google Scholar] [CrossRef]
  137. Shi, Y.; Shan, S.; Li, C.; Song, X.; Zhang, C.; Chen, J.; You, J.; Xiong, J. Application of the tumor site recognizable and dual-responsive nanoparticles for combinational treatment of the drug-resistant colorectal cancer. Pharm. Res. 202037, 1–14. [Google Scholar] [CrossRef]
  138. Hu, Q.; Wang, Y.; Xu, L.; Chen, D.; Cheng, L. Transferrin conjugated PH- and redox-responsive poly(Amidoamine) Dendrimer conjugate as an efficient drug delivery carrier for cancer therapy. Int. J. Nanomed. 202015, 2751–2764. [Google Scholar] [CrossRef]
  139. Liang, Y.; Zhang, J.; Tian, B.; Wu, Z.; Svirskis, D.; Han, J. A NAG-guided nano-delivery system for redox-and pH-triggered intracellularly sequential drug release in cancer cells. Int. J. Nanomed. 202015, 841. [Google Scholar] [CrossRef]
  140. Huang, Y.; Tang, Z.; Peng, S.; Zhang, J.; Wang, W.; Wang, Q.; Lin, W.; Lin, X.; Zu, X.; Luo, H.; et al. pH/redox/UV irradiation multi-stimuli responsive nanogels from star copolymer micelles and Fe3+ complexation for “on-demand” anticancer drug delivery. React. Funct. Polym. 2020149, 104532. [Google Scholar] [CrossRef]
  141. Bardajee, G.R.; Khamooshi, N.; Nasri, S.; Vancaeyzeele, C. Multi-stimuli responsive nanogel/hydrogel nanocomposites based on κ-carrageenan for prolonged release of levodopa as model drug. Int. J. Biol. Macromol. 2020153, 180–189. [Google Scholar] [CrossRef]
  142. Xu, X.; Wang, X.; Luo, W.; Qian, Q.; Li, Q.; Han, B.; Liu, C. Triple cell-responsive nanogels for delivery of drug into cancer cells. Colloids Surf. B 2018163, 362–368. [Google Scholar] [CrossRef]
  143. Zhang, Y.M.; Liu, Y.H.; Liu, Y. Cyclodextrin-based multistimuli-responsive supramolecular assemblies and their biological functions. Adv. Mater. 202032, 1806158. [Google Scholar] [CrossRef]
  144. Lu, J.; Luo, B.; Chen, Z.; Yuan, Y.; Kuang, Y.; Wan, L.; Yao, L.; Chen, X.; Jiang, B.; Liu, J.; et al. Host-guest fabrication of dualresponsive hyaluronic acid/mesoporous silica nanoparticle based drug delivery system for targeted cancer therapy. Int. J. Biol. Macromol. 2020146, 363–373. [Google Scholar] [CrossRef]
  145. Xiong, D.; Wen, L.; Peng, S.; Xu, J.; Zhang, L. Reversible cross-linked mixed micelles for PH triggered swelling and redox triggered degradation for enhanced and controlled drug release. Pharmaceutics 202012, 258. [Google Scholar] [CrossRef]
  146. Chen, H.; Fan, X.; Zhao, Y.; Zhi, D.; Cui, S.; Zhang, E.; Lan, H.; Du, J.; Zhang, Z.; Zhang, S.; et al. Stimuli-responsive polysaccharide enveloped liposome for targeting and penetrating delivery of survivin-shRNA into breast tumor. ACS Appl. Mater. Interfaces 202012, 22074–22087. [Google Scholar] [CrossRef]
  147. Ruttala, H.B.; Ramasamy, T.; Poudel, B.K.; Ruttala, R.R.T.; Jin, S.G.; Choi, H.-G.; Ku, S.K.; Yong, C.S.; Kim, J.O. Multi-responsive albumin-lonidamine conjugated hybridized gold nanoparticle as a combined photothermal-chemotherapy for synergistic tumor ablation. Acta Biomater. 2020101, 531–543. [Google Scholar] [CrossRef]
  148. Cuggino, J.C.; Gatti, G.; Picchio, M.L.; Maccioni, M.; Gugliotta, L.M.; Igarzabal, C.I.A. Dually responsive nanogels as smart carriers for improving the therapeutic index of doxorubicin for breast cancer. Eur. Polym. J. 2019116, 445–452. [Google Scholar] [CrossRef]
  149. Yang, J.; Wei-Yahng, Y. Metal-organic framework-based cancer theranostic nanoplatforms. View 20201, e20. [Google Scholar] [CrossRef]
  150. Li, Y.; Wu, Y.; Li, T.; Yang, X.; Li, Z.; Yang, X.; Chen, J.; Wan, J.; Xiao, C.; Guan, J.; et al. A simple glutathione-responsive turn-on theranostic nanoparticle for dual-modal imaging and chemo-photothermal combination therapy. Nano Lett. 201919, 5806–5817. [Google Scholar] [CrossRef]
  151. Halas, N.J. Connecting the dots: Reinventing optics for nanoscale dimensions. Proc. Natl. Acad. Sci. USA 2009106, 3643–3644. [Google Scholar] [CrossRef]
  152. Lal, S.; Clare, S.E.; Halas, N.J. Nanoshell-enabled photothermal cancer therapy: Impending clinical impact. Accounts Chem. Res. 200841, 1842–1851. [Google Scholar] [CrossRef]
  153. Ali, M.R.K.; Wu, Y.; Ghosh, D.; Do, B.H.; Chen, K.; Dawson, M.R.; Fang, N.; Sulchek, T.A.; El-Sayed, M.A. Nuclear membrane-targeted gold nanoparticles inhibit cancer cell migration and invasion. ACS Nano 201711, 3716–3726. [Google Scholar] [CrossRef]
  154. Dreaden, E.C.; Mackey, M.A.; Huang, X.; Kangy, B.; El-Sayed, M.A. Beating cancer in multiple ways using nanogold. Chem. Soc. Rev. 201140, 3391–3404. [Google Scholar] [CrossRef]
  155. Cao, J.; Shi, X.; Gurav, D.D.; Huang, L.; Su, H.; Li, K.; Niu, J.; Zhang, M.; Wang, Q.; Jiang, M.; et al. Metabolic fingerprinting on synthetic alloys for medulloblastoma diagnosis and radiotherapy evaluation. Adv. Mater. 202032, 2000906. [Google Scholar] [CrossRef]
  156. Xu, W.; Wang, L.; Dai, H.; Sun, X.; Huang, L.; Su, H.; Wei, X.; Chen, C.-C.; Lou, J.; Dai, H.; et al. Diagnosis and prognosis of myocardial infarction on a plasmonic chip. Nat. Commun. 202011, 1–9. [Google Scholar] [CrossRef]
  157. Huang, L.; Gurav, D.D.; Price, C.-A.H.; Velliou, E.; Liu, J.; Qian, K.; Wu, S.; Xu, W.; Vedarethinam, V.; Yang, J.; et al. A multifunctional platinum nanoreactor for point-of-care metabolic analysis. Matter 20191, 1669–1680. [Google Scholar] [CrossRef]
  158. Shu, W.; Wang, Y.; Liu, C.; Li, R.; Pei, C.; Lou, W.; Lin, S.; Di, W.; Wan, J. Construction of a plasmonic chip for metabolic analysis in cervical cancer screening and evaluation. Small Methods 20194, 190046. [Google Scholar] [CrossRef]
  159. Baffou, G.; Cichos, F.; Quidant, R. Applications and challenges of thermoplasmonics. Nat. Mater. 202019, 946–958. [Google Scholar] [CrossRef]
  160. Annesi, F.; Pane, A.; Bartolino, R.; De Sio, L.; Losso, M.A.; Guglielmelli, A.; Lucente, F.; Petronella, F.; Placido, T.; Comparelli, R.; et al. Thermo-plasmonic killing of Escherichia coli tg1 bacteria. Materials 201912, 1530. [Google Scholar] [CrossRef]
  161. Guglielmelli, A.; Rosa, P.; Perotto, G.; De Sio, L.; Contardi, M.; Prato, M.; Mangino, G.; Miglietta, S.; Petrozza, V.; Pani, R.; et al. Biomimetic keratin gold nanoparticle-mediated in vitro photothermal therapy on glioblastoma multiforme. Nanomedicine 20214. [Google Scholar] [CrossRef]
  162. De Angelis, B.; DePalo, N.; Petronella, F.; Quintarelli, C.; Curri, M.L.; Pani, R.; Calogero, A.; Locatelli, F.; De Sio, L. Stimuli-responsive nanoparticle-assisted immunotherapy: A new weapon against solid tumours. J. Mater. Chem. B 20208, 1823–1840. [Google Scholar] [CrossRef]
  163. Renard, D.; Tian, S.; Lou, M.; Neumann, O.; Yang, J.; Bayles, A.; Solti, D.; Nordlander, P.; Halas, N.J. Uv-resonant al nanocrystals: Synthesis, silica coating, and broadband photothermal response. Nano Lett. 202021. [Google Scholar] [CrossRef]

This entry is adapted from the peer-reviewed paper 10.3390/nano11020396

This entry is offline, you can click here to edit this entry!