Int J App Pharm, Vol 15, Issue 6, 2023, 38-44Review Article

EMERGING TRENDS IN STIMULI-SENSITIVE DRUG DELIVERY SYSTEM: A COMPREHENSIVE REVIEW OF CLINICAL APPLICATIONS AND RECENT ADVANCEMENTS

THAMARAI SELVAN DHANDAPANI1, VIJAYARAGHAVAN KRISHNAN1, BALAGEE MUTHUKUMAR2, VIVEKANANDAN ELANGO1, SAKTHI SHANMUGA JEYANDAR LAKSHMANAN1, SAM HARRISON SAM JENKINSON3, DHANDAPANI NAGASAMY VENKATESH1*

1Department of Pharmaceutics, JSS College of Pharmacy, (JSS Academy of Higher Education and Research, Mysuru) Ooty - 643 001. Nilgiris. Tamil Nadu, India. 2Department of Pharmaceutical Analysis, JSS College of Pharmacy, (JSS Academy of Higher Education and Research, Mysuru) Ooty - 643 001. Nilgiris. Tamil Nadu. India. 3Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, (JSS Academy of Higher Education and Research, Mysuru) Ooty - 643 001. Nilgiris. Tamil Nadu. India.
*Corresponding author: Dhandapani Nagasamy Venkatesh; *Email: nagasamyvenkatesh@jssuni.edu.in

Received: 27 Jul 2023, Revised and Accepted: 26 Aug 2023


ABSTRAC

The combination of fields such as applied physics, biology, computational modeling and analysis, pharmaceuticals, chemistry, optics, and material science study has been made easier for the rise of stimuli-sensitive drug delivery systems. This study aimed to overcome the shortcomings of conventional therapeutic approaches by concentrating on the most recent developments in stimuli-sensitive drug delivery systems, which are intended to accomplish the targeted release of drugs in specified areas. This review aims to provide an overview of stimuli-sensitive drug delivery systems and recent advancements between 2015 and 2023 by focusing on their ability to respond to exogenous and endogenous stimuli. In recent years, significant progress has been made in developing innovative stimuli-responsive drug delivery platforms that can trigger various external stimuli, such as light, temperature, magnetic fields, and ultrasound. These exogenous stimuli-responsive systems enable on-demand drug release at specific target sites, allowing for personalized and patient-centric treatment strategies. Notable breakthroughs include photoresponsive nanocarriers, thermosensitive hydrogels, and magnetic nanoparticles, all designed to respond to specific cues for controlled drug delivery.

Keywords: Stimuli-sensitive drug delivery systems, Exogenous stimuli, Endogenous stimuli, Recent advancements


INTRODUCTION

A stimuli-sensitive drug delivery system is a sophisticated platform designed to respond intelligently to specific triggers from external sources (exogenous stimuli) and internal cues within the body (endogenous stimuli). These triggers include light, temperature, magnetic fields, ultrasound, pH levels, enzymatic activity, and more. By exploiting the potential of these stimuli, stimuli-sensitive drug delivery systems can achieve accurate drug release at specific locations, maximizing therapeutic results and minimizing potential adverse reactions [1].

This article will explore the fascinating world of stimuli-sensitive drug delivery systems. We will focus on two crucial aspects: the diverse range of exogenous and endogenous stimuli that can trigger drug release and the recent ground-breaking advancements that have propelled this field to new heights. By obtaining a comprehensive understanding of the fundamental principles and the latest innovations, we can better appreciate the immense potential of intelligent drug administration systems in revolutionizing patient treatments and ushering in a new era of personalized medicine. So, let us embark on this journey through cutting-edge advancements in drug delivery, where science meets resourcefulness to redefine the panorama of healthcare.

Stimuli-sensitive drug delivery systems

A recent advanced method for drug targeting is the Stimuli-sensitive drug delivery system. In a stimuli-sensitive drug delivery system, the medication is directly delivered to the target site or released whenever necessary, thereby reducing the adverse effects of medication in other tissues [2]. Medication is selectively accumulated in the target site for a prolonged period in a highly controlled manner for enhancing the therapeutic activity in the stimuli-sensitive drug delivery system [3]. An ideal stimuli-sensitive drug delivery system should meet several critical criteria. Firstly, the material used to create the system’s matrix should be biocompatible or at least not adversely affect the body. Secondly, the medication should be able to be encapsulated within the system without losing its effectiveness. Thirdly, the release of the drug should be able to be triggered in a non-invasive way without requiring any external devices. Finally, the system should ensure that no medication is released until it is activated or turned on [4].

Classification is based on the mechanism by which drug release

Endogenous stimulus

The term "endogenous stimulus," also called intrinsic stimulus, pertains to a scenario wherein the triggering signal originates within the body. This signal is generated explicitly by factors such as the internal pH level, redox activity, and enzyme activity. These drug delivery systems initiate the administration of medications by controlling the conditions within the tissues, increasing the activity of particular enzymes, facilitating the interaction between antibodies and antigens, and identifying specific configurations of host-guest molecules [5].

pH-responsive drug delivery system

pH-susceptible polymers are a kind of polymers that demonstrate a reply to fluctuations in the encompassing pH levels. These polymers can be sorted into two categories: (A) polymers with ionizable functional groups and (B) polymers containing acid-degradable bonds. The ionization of the connected acidic or fundamental groups is prompted by the change in environmental pH, which causes cross-linking and modifications to the polymer's swelling properties [6]. To demonstrate, when subjected to a low pH environment, polyacid polymers undergo a decrease in size, whereas, in a high pH environment, they encounter an extension due to the protonation of their acidic components [7]. Combining pH-sensitive drug-release systems with other stimuli, redox, or temperature triggers enhances the accuracy and efficacy of these systems in responding to multiple parameters. For example, PEG-grafted PMAAc demonstrates notable sensitivity to changes in pH levels. For instance, To achieve specific delivery of 5-Fluorouracil (5-FU) to the colon, researchers developed a method utilizing citrus pectin nanoparticles (E-CPNs) coated with Eudragit S100 [8]. Polymers that respond to changes in pH commonly encompass chitosan, albumin, gelatin, interpenetrating networks of poly(acrylic acid) (PAAc) and chitosan, poly(methacrylic acid-g-ethylene glycol) [P(MAA-g-EG)], poly(ethylene imine) (PEI), poly(N, N-dialkylamino ethyl methacrylates) (PDAAEMA), and poly(lysine). Table 1 shows the pH valves of different fluids in the body.

Table 1: pH valves of different fluids in the body

Site pH value Reference
Small intestinal fluid 7.5–8.0 [9]
Gastric juice 1.0–3.5 [9]
Bile 7.8 [9]
Plasma 7.38–7.42 [9]
Saliva 6.0–7.0 [9]
Golgi apparatus 6.0–6.7 [9]
Pancreatic juice 8.0–8.3 [9]
Lysosome 4.5–5.0 [9]
Coliform fluid 5.5–7.0 [9]
Tumor microenvironment 6.5–6.8 [9]
Endosome 4-7 [9]

Redox-responsive drug delivery systems

Compared to normal cells, the environment inside tumor tissues is distinguished by a substantial decrease in its circumstances, frequently called a reducing environment. It is essential to incorporate redox-sensitive chemical elements into the formulation of the drug distribution systems for utilizing the tumor cells' reducing environment for accurate drug distribution. Redox-sensitive chemical elements such as Sulfur Bonds, Amide-Thioether Linkage, Tetrasulfur Bonds, Platinum Conjugation, and the chemical substances responsible for the reduction microenvironment are glutathione [10]. The intracellular levels of Glutathione Sulfhydryl (GSH) in normal cells typically range from 1 to 10 mmol, while the extracellular GSH levels are significantly lower, ranging from 2 to 20 µM. However, GSH levels are more than four times higher in tumor cells than in healthy cells. GSH demonstrates remarkable antioxidant properties [11]. NADPH, along with its oxidized form NADP+, is an additional biomolecule that enhances the reducing capacity of tumor cells [12].

Enzyme-responsive drug delivery system

Enzyme-reactive systems offer a captivating approach for forming responsive drug carriers due to the potential irregularities in enzyme levels within the microenvironment of diseased sites [13]. Smart carriers or binders equipped with drug payloads will facilitate the targeted liberation of drugs at specific locations through enzymatic splitting. This splitting can occur between encapsulation or covalent linkage between the carriers/binders and the drug. The action of varied enzymes initiates the liberation of the drug. Different materials are employed in systems that respond to enzymes, with proteases, kinases, phosphatases, and endonucleases being among the most commonly utilized enzyme categories [14]. In reality, diseases frequently show the imbalance or disturbance of one or multiple enzymatic functions. Precisely, tumor tissues often exhibit increased levels of proteases, which aid in the invasion and spread of tumor cells. This makes proteases an appealing focus for delivering drugs selectively. By comprehending the structural characteristics necessary for targeting a specific enzyme, it becomes possible to create modified medicine carriers that release the medication specifically within tumor microenvironments [15]. When creating enzyme-reactive materials, several crucial factors are typically considered to ensure their effectiveness [16]. These factors include considering (a) the chemical and physical properties of the material, (b) the enzyme concentration in the substrate or material, and (c) how the substrate is attached or fixed to the material [17]. For instance, an eight-amino acid string known as the Gly-Pro-Leu-Gly-Ile-Ala-Gly-Gln (GPLGIAGQ) sequence was developed by Torchilin and coworkers as a sensitive connector for attaching long-chain PEG to liposomes and containing the cell-penetrating peptide TATp. MMP2's octapeptide GPLGIAGQ breakdown exposed TATp, which improved tumor cells' ability to absorb liposome particles [18]. Fig. 1 illustrates the Enzyme-dependent drug release profile of the Enzyme-responsive drug delivery system.

Fig. 1: Represents the mechanism of the enzyme-responsive drug delivery system

Exogenous stimulus

An external stimulus refers to an outside signal applied to Nanocarriers to provoke the liberation of the drug, such as a heat change, sound waves, magnetism field, or electrical field [19].

Temperature stimulus drug delivery system

Temperature can function as a stimulus in two ways: External when the temperature is applied from outside the body, or internal when certain illness conditions lead to a natural increase in body temperature [20]. In water, thermosensitive polymer solutions exhibit a phenomenon called the Lower Critical Solution Temperature (LCST). The polymer solution remains in a singular phase when the temperature is beneath this threshold. However, when the temperature surpasses the LCST, the polymer chains collapse and gather, leading to phase separation, where water is expelled from the solution [21]. This specific quality has been utilized in the design of temperature-responsive nanocarriers, mainly based on this property. Some frequently used PNIPAAm, poly(N-vinylalkylamide), poly(N-vinyl caprolactam), poly(N, N-diethyl acrylamide), electronics, Pluronic’s, phosphorene derivatives, and polysaccharide derivatives are examples of thermosensitive polymers. When establishing the phase transition temperature (Tp) of thermosensitive nanocarriers, it is crucial to consider certain factors. (a) The Tp (transition temperature) should be near the body's typical temperature (around 37 °C), avoiding temperatures lower than this. By doing this, medication release before the application of local hyperthermia is prevented or reduced. (b) The Tp should fall within a relatively small temperature range (about 4-5 °C) and be below the body's acceptable temperature. (c) When approaching the Tp, the architecture of thermosensitive nanocarriers should experience considerable changes that enable faster release and maximize efficiency. For instance, Phase-Change Materials (PCMs) are Temperature-responsive drug delivery systems to create a new method for releasing drugs in response to changes in temperature. They specifically employed two PCMs: 1-tetradecane, which liquefies at 38-39 °C, and dodecanoic acid, which liquefies at 43-46 °C [22]. To construct this system, they imgded small particles containing FITC-dextran (a fluorescent dye) into a matrix composed of the PCM. These particles were shaped like spheres or rods. When the temperature was beneath the liquefaction point of the PCM, nothing occurred because the PCM was hydrophobic (repelled water). Nonetheless, when the temperature increased beyond the liquefaction point, the PCM commenced melting, causing the particles to be released and eventually enabling FITC-dextran to be discharged from them [23]. Table 2 states that thermosensitive polymer and their LCST.

Table 2: Represents different thermosensitive polymer

Type of thermosensitive micelle Composition of thermosensitive micelles Encapsulated drug LCST (◦C) References
Thermo-responsive shell Poly(butyl methacrylate) Adriamycin 32.5 [24]
P(NIPAAm-co-HMAAm) with poly(D,L-lactide) Adriamycin 37–42.5 [25]
Poly(ε-caprolactone) with P(NIPAAm-co-HMAAm) Cinnarizine 29.5–35.2 [26]
Thermo-responsive core pHPMAmDL-b-PEG Methotrexate 41 [27]
P(IPAAm-co-HMAAm)-biotin-PEG Paclitaxel 10–65 [28]

Light-responsive drug delivery system

Systems for delivering photoresponsive drugs can adapt their structure and composition in response to exposure to light. Light is an easily accessible, effective, and non-intrusive external stimulus that can be effortlessly acquired and controlled by adjusting various factors such as brightness, wavelength, exposure duration, and beam size [29]. This allows for accurate concentration and management of the light stimulus. Ultraviolet (UV), observable light, and Near-Infrared (NIR) are commonly employed light sources. Three major categories can be used to categorize light-manipulated medication delivery systems: those relying on photoisomerization, photochemistry, and photothermal effects to release medications. In photoisomerization-based systems, the hydrogels undergo structural shifts from straight to bent when exposed to light [30]. In photochemical-based systems, light-triggered reactions modify the hydrogel's network structure and configuration, facilitating drug release. On the contrary, the photothermal reaction utilizes materials capable of converting light into heat energy. This heat energy subsequently disrupts the drug carrier's sensitivity to temperature changes [31]. Numerous photo-sensitive polymers containing azobenzene or spirogyra, such as PAA, PHPM Am, and PNIPAM, have been documented in the literature [32].

Magnetic-responsive drug delivery system

Magnetic drug directing involves using outer magnetic fields to control the motion of magnetic medicine transporters within the body, guiding them toward the desired position. For magnetic medicine directing to be efficient, a dependable magnet system is necessary for steering the drug transporters toward the intended target [33]. Pharmaceutical applications have extensively studied the use of Superparamagnetic Iron Oxide Nanoparticles (SPIONs). Superparamagnetism occurs when the size of ferrimagnetic or ferromagnetic particles diminishes below a certain threshold, resulting in the display of magnetic properties. Superparamagnetic iron oxide nanoparticles, including magnetite (Fe3O4) and maghemite (γ-Fe2O3), possess significant potential in nanomedicine. The regulated motion of SPIONs in the bloodstream relies on a dynamic equilibrium between the magnetic and hydrodynamic forces acting upon them [34]. PVA and alginate-dispersed magnetic microspheres are some polymers that show magnetic-responsive drug delivery. Fig. 2 illustrates the magnetic-responsive drug delivery system's magnetic stimuli-dependent drug release profile.

Fig. 2: Represents magnetic responsive drug delivery system

Ultrasound-responsive drug delivery system

Ultrasound signals are a kind of mechanical signals that have elevated frequencies (≥20 kHz) [35]. These signals can be directed and transmitted through specific substances or mediums. Two primary impacts of ultrasound are believed to contribute to this procedure: thermal and non-thermal impacts. The thermal impact of ultrasound involves transforming sound energy into heat, increasing the temperature within the targeted tissue [36]. This temperature elevation can disturb the cell membrane and enhance the permeability of blood vessels. In cancer treatment, this effect has been harnessed to trigger the release of drugs from temperature-sensitive liposomes, microbubbles, or polymeric micelles. The non-thermal effect of ultrasound is mainly linked to a phenomenon known as cavitation. Cavitation can occur when small or tiny bubbles within the tissue respond to ultrasound signals [37]. Two categories of cavitation exist: non-inertial and inertial. Non-inertial cavitation comprises a continuous cycle of bubbles expanding and contracting, which can be intensified by agents that respond to ultrasound. On the other hand, inertial cavitation involves the forceful collapse of bubbles, producing high-speed microstreams and free radicals [38]. PLA poly(lactic acid), PAH poly(allylamine hydrochloride), a polymer which responsive to ultrasound, are PFC perfluorocarbon, PVA polyvinyl alcohol, PLGA poly(lactic-co-glycolic acid), PFO-PLLA perfuoroctanol-poly(lactic acid) [39].

Electro-responsive drug delivery system

The utilization of electric field shocks has the potential to magnify the permeability of cellular membranes. The electrical potential disparity generated by employing an electric field is responsible for the aimed discharge of medications by producing a differentiation between the internal and external potentials of the cell [40]. Utilizing an electric field to the cell membrane causes alterations in polarity, ionic potency, and pH, leading to variations in the overall osmotic pressure within polymers (recognized as electro-osmosis) [41]. This, consequently, triggers flexing, expansion, contraction, or disintegration of the polymer, resulting in the discharge of the active component. For several reasons, electric fields are preferred over alternate external stimuli [42]. Primarily, they provide the benefit of being effortlessly manageable and applicable. Secondly, they do not necessitate intricate and elaborate instruments. Lastly, they can be smoothly integrated into the progression of chip-based apparatus. Polypyrrole ferrocene and carbon nanotubes are commonly encountered as typical examples of electro-responsive materials used in pharmaceutical delivery applications [43].

Table 3: Represents the recent advancements in stimuli-sensitive drug delivery systems

Stimulus Product name Drug loaded Mechanism Targeting area Reference
Temperature-responsive drug delivery system DOX@PAM AND DOX@PIPAM Doxorubicin (DOX) This depolarization negatively affected the electrostatic interactions between the positively charged nanoparticles and mitochondrial membranes, thereby hindering mitochondrial targeting.

Lung Cancer

Mitochondria-targeted delivery

[44]
Poly(N-isopropyl acrylamide-co-acrylic acid) nanofiber Crystal violet (CV) or gentamicin The nanofibers showed responsive behavior to changes in temperature and pH, with the ability to swell and release their loaded contents between 31 and 34 °C. Wound healing [45]

(PNIPAM-FPA-DMA) copolymer-based hydrogel

with PEO90 dihydrazone as cross-linker

Doxorubicin (DOX) Enhanced matrix mobility beyond the LCST of PNIPAM, inducing coil-to-globule transitions, facilitates drug release. Cancer cells [46]
Chitosan grafted PNIPAM-based nano gel assembly. Curcumin The enhanced mobility of the matrix due to the coil-to-globule transition of PNIPAM, occurring above its LCST (lower critical solution temperature), facilitates the release of drugs. Controlled release [47]
Light-responsive drug delivery system NP [CPP] Doxorubicin (DOX) The caging group of DEACM is eliminated through photo-cleavage using UV light at 400 nm, 50 mW cm^-2, for a duration of 1 min. Choroidal neovascularization CNV [48]
AKBA@ZnO nanoparticles Acetyl-11-keto-β-boswellic acid (AKBA). Zinc oxide (ZnO) nanoparticles loaded with AKBA (acetyl-11-keto-β-boswellic acid) and designed for UV-controlled drug release. Polymorphous Light Eruption (PLE) [49]
Gelatin PAD and Alginate 5-fluorouracil (5-FU) 3D printed scaffolds for accurate structure control and drug release Breast cancer [50]
Ultrasonic responsive drug delivery system Bioelectrets Curcumin Ultrasonic stimulation at 90% power (Pmax = 1200 W) produced a current of 0.472 nA. This current produces the heat that makes carnauba wax melt and releases drugs. Sustained release for chronic inflammatory diseases [51]
MSN: MSN-Ce6 Doxorubicin (DOX) Drug-loaded mesoporous silica nanoparticles (MSN-DOX-Ce6) entered the bloodstream, passively targeting tumors through the EPR effect, releasing DOX and Ce6 to enhance drug concentration around tumor cells, with subsequent sonication-triggered activation leading to combined antitumor effects of DOX and Ce6. Breast Cancer [52]

LPs (MFL)+MBs:

DMPC|DOTAP|DSPE-

MPEG2k+

SonoVue

DTX MFLs are efficiently fused with the cell membrane for drug delivery within cells. MB+FUS induced sonoporation in vascular cells, enhancing the EPR effect. Breast Cancer [53]

LPs (Enzyme sensitive,

ES)+MBs:

POPC|CHOL|PCL+

SonoVue

Doxorubicin (DOX) PEG cleavage of coated LPs by MMP enzymes resulted in higher intracellular uptake than NES-LPs. However, the VIR: Doxil-like>ES>NES. Tumor growth was reduced by 58%, 39%, and 21%, respectively.

Prostatic

cancer

[54]
Electric responsive drug delivery system Smart skin bandage acrylamide and polyethylene glycol dimethyl acrylate

Curcumin

Graphene oxide (GO), gelatin, or trypsin

The water affinity and release profiles of curcumin, Slow and fast release profiles were achieved at 0 V and 24 V, respectively, while intermediate kinetics were observed at 12 V and 48 V Wound healing [55]
Chiston-gold nanocomposite fluorouracil (CGNC-FU) 5-fluorouracil (5-FU). The CGNC encapsulates the drug molecules within its 3D network at higher pH levels. It undergoes a reversible gel-to-sol transition upon exposure to lower pH conditions, releasing the drug. Cancer cells [56]
Electro-conductive hydrogels (ECHs)-Poly(ethyleneimine) (PEI) and 1-vinylimidazole (VI) polymer Indomethacin Volumes of poly(ethyleneimine) above 2.6 ml and 0.7 ml achieved optimal electro-responsive drug release (0.8 mg) for indomethacin, with swelling levels ranging from 25% to 45%. Controlled personalized drug delivery [57]
Macroporous polypyrrole (PPy) thin films Dexamethasone Macroporous polypyrrole (PPy) thin films sowed better electrical responsiveness and released more of the drug. Posterior uveitis [58]
Nanocomposite film Polypyrrole/graphene oxide nanocomposite film Acrylamide and N, N0-ethylene bisacrylamide Electrochemical reduction The application of an electric voltage results in the quicker release of anionic drugs, whereas cationic drugs are released more slowly Dexamethasone [59]
Magnetic responsive drug delivery system Fe3O4@carbon(C)/ZnO-doxorubicin (DOX)-folic acid (FA) nanoparticles Doxorubicin (DOX)

The ZnO “gatekeeper” component could degrade in the acidic tumor microenvironment, resulting in controlled drug release that responds to changes in pH.

The carbon shell of the nanocomposites can convert light into heat, enabling photothermal therapy.

Cancer cell [60]
Paclitaxel loaded in Pluronic F-68 Paclitaxel Magnetic hyperthermia causes the lipid layer to melt, facilitating drug release. Targeted drug delivery [61]
Manganese ferrite (MnFe2O4)-chitosan and alginate sodium Curcumin Magnetic hyperthermia Tumor cells [62]
Redox responsive drug-responsive system Xyl-SS-Cur/5-FUSA Curcumin and 5-FU When there is a high glutathione concentration, the nanoparticles release the drugs more efficiently. Cancer cell [63]
DOX@MSNs-CAIX particles Doxorubicin hydrochloride (DOX) Doxorubicin hydrochloride (DOX) released the drug in response to glutathione (GSH), a molecule found in high levels in cancer cells. Breast cancer cells [64]
MP3/ACPP/AE105@NPs Metal complex Nanotherapeutics enhanced reactive oxygen species (ROS) production by suppressing TrxR (thioredoxin reductase) activity and modulating metastasis-associated proteins. by inhibiting FAK (focal adhesion kinase) Breast cancer [65]
Chitosan/stearic acid nanoparticles (CSSA NPs) Curcumin and doxorubicin Under the influence of redox stimuli from cancer cells, the substance degrades specifically at the tumor site. Colorectal cancer [66]

CONCLUSION

This article briefly overviews the unique features shown by responsiveness-triggered polymeric carriers. Moreover, it emphasizes their encouraging abilities in the domain of aimed medication transportation. Before designing Drug Delivery Systems (DDS), it is pivotal to consider diverse vital factors like biocompatibility, biodegradability, non-toxicity, and safe elimination. These factors enforce notable restrictions that necessitate conscientious evaluation. Despite many products undergoing clinical trials, intelligent pharmaceutical delivery systems reveal noteworthy potential. They are actively employed in various fields, such as illness detection, molecular visualization, and precise administration of cancer-fighting medicines to tumors. The recent advancements in stimuli-sensitive drug delivery systems have demonstrated immense potential in revolutionizing healthcare and personalized medicine. However, challenges related to scalability, cost-effectiveness, and regulatory approval remain to be addressed. Continued research and collaboration between academia, industry, and regulatory bodies will be crucial to unlock the full potential of these transformative technologies for patient benefit. Furthermore, in the upcoming years, the fusion of knowledge in focused medication transportation and progressions in intelligent medication transportation systems should give importance to the namely, molecular visualization and focused distribution of anticancer medications to tumors.

ACKNOWLEDGEMENT

The authors would like to thank the Department of Science and Technology-Fund for Improvement of Science and Technology Infrastructure (DST-FIST) and Promotion of University Research and Scientific Excellence (DST-PURSE) and Department of Biotechnology-Boost to University Interdisciplinary Life Science Departments for Education and Research program (DBT-BUILDER) for the facilities provided in our department.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All the authors have contributed equally.

CONFLICTS OF INTERESTS

The authors declare no conflict of interest

REFERENCES

  1. Liu D, Yang F, Xiong F, Gu N. The smart drug delivery system and its clinical potential. Theranostics. 2016;6(9):1306-23. doi: 10.7150/thno.14858, PMID 27375781.

  2. Gupta M, Sharma V. Targeted drug delivery system a review. Res J Chem Sci. 2011;1:135-8.

  3. Muller RH, Keck CM. Challenges and solutions for the delivery of biotech drugs-a review of drug nanocrystal technology and lipid nanoparticles. J Biotechnol. 2004;113(1-3):151-70. doi: 10.1016/j.jbiotec.2004.06.007, PMID 15380654.

  4. Elema Buzu. Stimuli-sensitive drug delivery systems: a review, scieniQ Publisher; 2020. https://www.researchgateresearchgate.net/publication/363095261.

  5. Raza A, Rasheed T, Nabeel F, Hayat U, Bilal M, Iqbal HMN. Endogenous and exogenous stimuli-responsive drug delivery systems for programmed site-specific release. Molecules. 2019;24(6):1117. doi: 10.3390/molecules24061117, PMID 30901827.

  6. Chen K, Gou W, Wang X, Zeng C, Ge F, Dong Z. UV-cured fluoride-free polyurethane functionalized textile with pH-induced switchable superhydrophobicity and underwater superoleophobicity for controllable oil/water separation. ACS Sustainable Chem Eng. 2018 Oct 29;6(12):16616-28. doi: 10.1021/acssuschemeng.8b03851.

  7. Tan RYH, Lee CS, Pichika MR, Cheng SF, Lam KY. PH-responsive polyurethane for the advancement of biomedical and drug delivery. Polymers. 2022;14(9):1672. doi: 10.3390/polym14091672, PMID 35566843.

  8. Garcia Fernandez L, Mora Boza A, Reyes Ortega F. pH-responsive polymers: properties, synthesis, and applications. Smart Polym Appl. 2019:45-86. doi: 10.1016/B978-0-08-102416-4.00003-X.

  9. Mu Yongxu, Gong L, Peng T, Yao J, Lin Z. Advances in pH-responsive drug delivery systems. OpenNano. 2021;5:(100031):2021.100031. doi: 10.1016/j.onano.2021.100031.

  10. Mutalabisin MF, Chatterjee B, Jaffri JMd. PH responsive polymers in drug delivery. Res J Pharm Technol. 2018;11(11):5115-22. doi: 10.5958/0974-360X.2018.00934.4.

  11. Abed HF, Abuwatfa WH, Husseini GA. Redox-responsive drug delivery systems: a chemical perspective. Nanomaterials (Basel). 2022;12(18):3183. doi: 10.3390/nano12183183, PMID 36144971.

  12. Rao NV, Ko H, Lee J, Park JH. Recent progress and advances in stimuli-responsive polymers for cancer therapy. Front Bioeng Biotechnol. 2018;6(6):110. doi: 10.3389/fbioe.2018.00110, PMID 30159310.

  13. Hu Q, Katti PS, Gu Z. Enzyme-responsive nanomaterials for controlled drug delivery. Nanoscale. 2014;6(21):12273-86. doi: 10.1039/c4nr04249b, PMID 25251024.

  14. Rana A, Adhikary M, Singh PK, Das BC, Bhatnagar S. ’Smart’ drug delivery: a window to future of translational medicine. Front Chem. 2022;10:1095598. doi: 10.3389/fchem.2022.1095598, PMID 36688039.

  15. Uthaman S, Huh KM, Park IK. Tumor microenvironment-responsive nanoparticles for cancer theragnostic applications. Biomater Res. 2018;22:22. doi: 10.1186/s40824-018-0132-z, PMID 30155269.

  16. Fang JH, Lee YT, Chiang WH, Hu SH. Magneto-responsive virus-mimetic nanocapsules with dual heat-triggered sequential-infected multiple drug-delivery approach for combinatorial tumor therapy. Small. 2015;11(20):2417-28. doi: 10.1002/smll.201402969, PMID 25604032.

  17. Antoniou AI, Giofre S, Seneci P, Passarella D, Pellegrino S. Stimulus-responsive liposomes for biomedical applications. Drug Discov Today. 2021;26(8):1794-824. doi: 10.1016/j.drudis.2021.05.010, PMID 34058372.

  18. Rana A, Adhikary M, Singh PK, Das BC, Bhatnagar S. "Smart” drug delivery: a window to future of translational medicine. Front Chem. 2022;10:1095598. doi: 10.3389/fchem.2022.1095598, PMID 36688039.

  19. Liu G, Lovell JF, Zhang L, Zhang Y. Stimulus-responsive nanomedicines for disease diagnosis and treatment. Int J Mol Sci. 2020 Sep 2;21(17):6380. doi: 10.3390/ijms21176380, PMID 32887466.

  20. Bordbar Khiabani A, Gasik M. Smart hydrogels for advanced drug delivery systems. Int J Mol Sci. 2022;23(7):3665. doi: 10.3390/ijms23073665, PMID 35409025.

  21. Choi SW, Zhang Y, Xia Y. A temperature-sensitive drug release system based on phase-change materials. Angew Chem Int Ed Engl. 2010;49(43):7904-8. doi: 10.1002/anie.201004057, PMID 20839209.

  22. Liechty WB, Kryscio DR, Slaughter BV, Peppas NA. Polymers for drug delivery systems. Annu Rev Chem Biomol Eng. 2010;1:149-73. doi: 10.1146/annurev-chembioeng-073009-100847, PMID 22432577.

  23. Shao P, Wang B, Wang Y, Li J, Zhang Y. The application of thermosensitive nanocarriers in controlled drug delivery. Journal of Nanomaterials. 2011;2011:1-12. doi: 10.1155/2011/389640.

  24. Wei H, Zhang X, Cheng C, Cheng SX, Zhuo RX. Self-assembled, thermosensitive micelles of a star block copolymer based on PMMA and PNIPA am for controlled drug delivery. Biomaterials. 2007;28(1):99-107. doi: 10.1016/j.biomaterials.2006.08.030, PMID 16959312.

  25. Chung JE, Yokoyama M, Yamato M, Aoyagi T, Sakurai Y, Okano T. Thermo-responsive drug delivery from polymeric micelles constructed using block copolymers of poly(N-isopropylacrylamide) and poly(butyl methacrylate). J Control Release. 1999;62(1-2):115-27. doi: 10.1016/S0168-3659(99)00029-2.

  26. Ieiri I, Takane H, Hirota T, Otsubo K, Higuchi S. Genetic polymorphisms of drug transporters: pharmacokinetic and pharmacodynamic consequences in pharmacotherapy. Expert Opin Drug Metab Toxicol. 2006;2(5):651-74. doi: 10.1517/17425255.2.5.651, PMID 17014387.

  27. Choi C, Chae SY, Nah J. Thermosensitive poly(N-isopropyl acrylamide)-b-poly(ε-caprolactone) nanoparticles for efficient drug delivery system. Polymer Polymer. 2006;47(13):4571-80. doi: 10.1016/j.polymer.2006.05.011.

  28. Cheng C, Wei H, Shi BX, Cheng H, Li C, Gu ZW. Biotinylated thermoresponsive micelle self-assembled from double-hydrophilic block copolymer for drug delivery and tumor target. Biomaterials. 2008;29(4):497-505. doi: 10.1016/j.biomaterials.2007.10.004, PMID 17959241.

  29. Soga O, Van Nostrum CF, Fens M, Rijcken CJ, Schiffelers RM, Storm G. Thermosensitive and biodegradable polymeric micelles for paclitaxel delivery. J Control Release. 2005;103(2):341-53. doi: 10.1016/j.jconrel.2004.12.009, PMID 15763618.

  30. Bustamante Torres M, Romero Fierro D, Arcentales Vera B, Palomino K, Magana H, Bucio E. Hydrogels classification according to the physical or chemical interactions and as stimuli-sensitive materials. Gels. 2021;7(4):182. doi: 10.3390/gels7040182, PMID 34842654.

  31. Tomatsu I, Peng K, Kros A. Photoresponsive hydrogels for biomedical applications. Advanced Drug Delivery Reviews. 2011;63(14-15):1257-66. doi: 10.1016/j.addr.2011.06.009.

  32. Cabane E, Zhang X, Langowska K, Palivan CG, Meier W. Stimuli-responsive polymers and their applications in nanomedicine. Biointerphases. 2012 Dec;7(1-4):9. doi: 10.1007/s13758-011-0009-3, PMID 22589052.

  33. Nehls EM, Rosales AM, Anseth KS. Enhanced user-control of small molecule drug release from a poly(ethylene glycol) hydrogel via azobenzene/cyclodextrin complex tethers. J Mater Chem B. 2016;4(6):1035-9. doi: 10.1039/C5TB02004B, PMID 27127630.

  34. Liu JF, Jang B, Issadore D, Tsourkas A. Use of magnetic fields and nanoparticles to trigger drug release and improve tumor targeting. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2019;11(6):e1571. doi: 10.1002/wnan.1571, PMID 31241251.

  35. O’Brien WD Jr. Ultrasound-biophysics mechanisms. Prog Biophys Mol Biol. 2007;93(1-3):212-55. doi: 10.1016/j.pbiomolbio.2006.07.010, PMID 16934858.

  36. Al-Jamal KT, Bai J, Wang JT-W, Protti A, Southern P, Bogart L. Magnetic drug targeting: preclinical in vivo studies, mathematical modeling, and extrapolation to humans. Nano Lett. 2016;16(9):5652-60. doi: 10.1021/acs.nanolett.6b02261, PMID 27541372.

  37. Rapoport NY, Kennedy AM, Shea JE, Scaife CL, Nam KH. Controlled and targeted tumor chemotherapy by ultrasound-activated nanoemulsions/microbubbles. J Control Release. 2009;138(3):268-76. doi: 10.1016/j.jconrel.2009.05.026, PMID 19477208.

  38. Abdo GG, Zagho MM, Khalil A. Recent advances in stimuli-responsive drug release and targeting concepts using mesoporous silica nanoparticles. Emergent Mater. 2020;3(3):407-25. doi: 10.1007/s42247-020-00109-x.

  39. Bhatnagar S, Kwan JJ, Shah AR, Coussios CC, Carlisle RC. Exploitation of sub-micron cavitation nuclei to enhance ultrasound-mediated transdermal transport and penetration of vaccines. J Control Release. 2016;238:22-30. doi: 10.1016/j.jconrel.2016.07.016, PMID 27417040.

  40. Hart FX, Palisano JR. The application of electric fields in biology and medicine. Electric Field; 2018. doi: 10.5772/intechopen.71683.

  41. Wei P, Cornel EJ, Du J. Ultrasound-responsive polymer-based drug delivery systems. Drug Deliv Transl Res. 2021;11(4):1323-39. doi: 10.1007/s13346-021-00963-0, PMID 33761101.

  42. SG, SG, SS, KG, OB, Smart VT. Polymers and their applications: a review. Int J Curr Pharm Rev Res. 2017;8(03). doi: 10.25258/ijcprr.v8i03.9220.

  43. Kolosnjaj Tabi J, Gibot L, Fourquaux I, Golzio M, Rols MP. Electric field-responsive nanoparticles and electric fields: physical, chemical, biological mechanisms and therapeutic prospects. Adv Drug Deliv Rev. 2019;138:56-67. doi: 10.1016/j.addr.2018.10.017, PMID 30414494.

  44. Lin X, Wu X, Chen X, Wang B, Xu W. Intellective and stimuli-responsive drug delivery systems in eyes. Int J Pharm. 2021;602:120591. doi: 10.1016/j.ijpharm.2021.120591, PMID 33845152.

  45. Seyfoddin A, Chan A, Chen WT, Rupenthal ID, Waterhouse GI, Svirskis D. Electro-responsive macroporous polypyrrole scaffolds for triggered dexamethasone delivery. Eur J Pharm Biopharm. 2015;94:419-26. doi: 10.1016/j.ejpb.2015.06.018, PMID 26141345.

  46. Xu X, Liu Y, Fu W, Yao M, Ding Z, Xuan J. Poly(N-isopropylacrylamide)-based thermoresponsive composite hydrogels for biomedical applications. Polymers. 2020;12(3):580. doi: 10.3390/polym12030580, PMID 32150904.

  47. Luckanagul JA, Pitakchatwong C, Ratnatilaka Na Bhuket P, Muangnoi C, Rojsitthisak P, Chirachanchai S. Chitosan-based polymer hybrids for thermo-responsive nanogel delivery of curcumin. Carbohydr Polym. 2018;1(181):1119-27. doi: 10.1016/j.carbpol.2017.11.027.

  48. Parvathy R. Chandran and N, Sandhyarani. An electric field responsive drug delivery system Based on chitosan–gold nanocomposites for site-specific and controlled delivery of 5-fluorouracil. RSC Advances; 2014. doi: 10.1039/c4ra07551j.

  49. Zhang P, Lu T, Xia X, Wu L, Shao L, Zhou J. How biomimetic amino modified mesoporous silica xerogel regulates loading and in vitro sustained delivery of levorotary ofloxacin. Mater Sci Eng C Mater Biol Appl. 2020;107:110266. doi: 10.1016/j.msec.2019.110266, PMID 31761238.

  50. Svanstrom A, Rosendahl J, Salerno S, Leiva MC, Gregersson P, Berglin M. Optimized alginate-based 3D printed scaffolds as a model of patient-derived breast cancer microenvironments in drug discovery. Biomed Mater. 2021;16(4). doi: 10.1088/1748-605X/ac0451, PMID 34030145.

  51. Li Junfei, Xie Y, Zou X, Li Zhengze, Liu W, Liu G. Ultrasonic/electrical dual stimulation response nanocomposite bio electret for controlled precision drug release. Materials Today Bio. 2023. doi: 10.1016/j.mtbio.2023.100665.

  52. Entzian K, Aigner A. Drug delivery by ultrasound-responsive nanocarriers for cancer treatment. Pharmaceutics. 2021 Jul 26;13(8):1135. doi: 10.3390/pharmaceutics13081135, PMID 34452096.

  53. Liang J, Liu J, Jin X, Yao S, Chen B, Huang Q. Versatile nanoplatforms loaded with doxorubicin and graphene quantum dots/methylene blue for drug delivery and chemo photothermal/photodynamic synergetic cancer therapy. ACS Appl Bio Mater. 2020;3(10):7122-32. doi: 10.1021/acsabm.0c00942, PMID 35019372.

  54. Entzian K, Aigner A. Drug delivery by ultrasound-responsive nanocarriers for cancer treatment. Pharmaceutics. 2021;13(8):1135. doi: 10.3390/pharmaceutics13081135, PMID 34452096.

  55. Indermun S, Choonara YE, Kumar P, du Toit LC, Modi G, Luttge R. An interfacially plasticized electro-responsive hydrogel for transdermal electro-activated and modulated (TEAM) drug delivery. Int J Pharm. 2014;462(1-2):52-65. doi: 10.1016/j.ijpharm.2013.11.014, PMID 24257244.

  56. di Luca M, Vittorio O, Cirillo G, Curcio M, Czuban M, Voli F. Electro-responsive graphene oxide hydrogels for skin bandages: the outcome of gelatin and trypsin immobilization. Int J Pharm. 2018;546(1-2):50-60. doi: 10.1016/j.ijpharm.2018.05.027, PMID 29758346.

  57. Choonara S, Yahya Kumar P. An interfacially plasticized electro-responsive hydrogel for transdermal electro-activated and modulated (TEAM). Drug Deliv. 2020;462(1-2):52-65.

  58. Zhang B, Molino PJ, Harris AR, Yue Z, Moulton SE, Wallace GG. Conductive and protein-resistant polypyrrole films for dexamethasone delivery. J Mater Chem B. 2016;4(15):2570-7. doi: 10.1039/C5TB00574D, PMID 32263280.

  59. Curcio M, Spizzirri UG, Cirillo G, Vittorio O, Picci N, Nicoletta FP. On-demand delivery of ionic drugs from electro-responsive CNT hybrid films. RSC Adv. 2015;5(56):44902-11. doi: 10.1039/C5RA05484B.

  60. Sauraj, Vinay Kumar, Kumar B, Priyadarshi R, Deeba F, Kulshreshtha A. Redox responsive xylan-SS-curcumin prodrug nanoparticles for dual drug delivery in cancer therapy. Mater Sci Eng C Mater Biol Appl. 2020;107:110356. doi: 10.1016/j.msec.2019.110356, PMID 31761247.

  61. Oliveira RR, Carriao MS, Pacheco MT, Branquinho LC, de Souza ALR, Bakuzis AF. Triggered release of paclitaxel from magnetic solid lipid nanoparticles by magnetic hyperthermia. Mater Sci Eng C Mater Biol Appl. 2018;92:547-53. doi: 10.1016/j.msec.2018.07.011, PMID 30184781.

  62. Jardim KV, Palomec Garfias AFA, Chaker B, Bao JA, Marquez Beltran SNC. Novel magneto-responsive nanoplatforms based on MnFe2O4 nanoparticles layer-by-layer functionalized with chitosan and sodium alginate for magnetically controlled release of curcumin. Mater Sci Eng C. 2018;92:184-95. doi: 10.1016/j.msec.2018.06.039.

  63. Senapati S, Mahanta AK, Kumar S, Maiti P. Controlled drug delivery vehicles for cancer treatment and their performance. Signal Transduct Target Ther. 2018;3(7):7. doi: 10.1038/s41392-017-0004-3, PMID 29560283.

  64. Entzian K, Aigner A. Drug delivery by ultrasound-responsive nanocarriers for cancer treatment. Pharmaceutics. 2021;13(8):1135. doi: 10.3390/pharmaceutics13081135, PMID 34452096.

  65. Radu ER, Semenescu A, Voicu SI. Recent advances in stimuli-responsive doxorubicin delivery systems for liver cancer therapy. Polymers (Basel). 2022;14(23):5249. doi: 10.3390/polym14235249, PMID 36501642.

  66. Sood A, Gupta A, Bharadwaj R, Ranganath P, Silverman N, Agrawal G. Biodegradable disulfide crosslinked chitosan/stearic acid nanoparticles for dual drug delivery for colorectal cancer. Carbohydr Polym. 2022;294:119833. doi: 10.1016/j.carbpol.2022.119833, PMID 35868778.