Next Article in Journal
Neuraminidase Inhibitor of Garcinia atroviridis L. Fruits and Leaves Using Partial Purification and Molecular Characterization
Next Article in Special Issue
Preparation and Characterization of Controlled-Release Floating Bilayer Tablets of Esomeprazole and Clarithromycin
Previous Article in Journal
Artificial Solar Light-Driven APTES/TiO2 Photocatalysts for Methylene Blue Removal from Water
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Stimuli-Responsive Drug Delivery Systems for the Diagnosis and Therapy of Lung Cancer

1
Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
2
Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
3
Cancer Center, Zhejiang University, Hangzhou 310003, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 29 December 2021 / Revised: 24 January 2022 / Accepted: 26 January 2022 / Published: 30 January 2022
(This article belongs to the Special Issue Design and Development of Novel Responsive Carrier for Drug Delivery)

Abstract

:
Lung cancer is the most commonly diagnosed cancer and the leading cause of cancer death worldwide. Numerous drugs have been developed to treat lung cancer patients in recent years, whereas most of these drugs have undesirable adverse effects due to nonspecific distribution in the body. To address this problem, stimuli-responsive drug delivery systems are imparted with unique characteristics and specifically deliver loaded drugs at lung cancer tissues on the basis of internal tumor microenvironment or external stimuli. This review summarized recent studies focusing on the smart carriers that could respond to light, ultrasound, pH, or enzyme, and provided a promising strategy for lung cancer therapy.

Graphical Abstract

1. Introduction

Lung cancer is the most diagnosed cancer in the world, with over two million new cases in 2020 (Figure 1) [1]. The current therapeutics for lung cancer mainly include surgery, drug therapy, and radiotherapy [2]. Indeed, the development of either small-molecule anticancer drugs or biologics, such as monoclonal antibodies for immunological targets has dramatically increased the clinical efficacy for the treatment of lung cancer in recent years [3]. However, problems remain unsolved, such as relatively low clinical response rates of monoclonal antibodies, unwanted adverse effects of targeting drugs, and drug resistance after a duration of exposure to certain agents [4,5]. To further enhance the drug effectiveness, one important strategy is to deliver drug cargoes specifically into lung cancer tissue using tailored carrier systems, which would confer ‘desirable’ properties to therapeutic agents and meanwhile compensate significant drawbacks in clinical applications [6,7].
Drug delivery systems are generally composed of carriers and therapeutic agents, and the conjugation of these two components would improve drug stability, desirable drug accumulation, and drug release, thus providing a promising strategy for lung cancer therapy [8]. Recently, intelligent drug carrier systems could precisely deliver drugs on the basis of unique tumoral microenvironments or external stimulus, such as pH, enzymes, reactive oxygen species (ROS), photodynamic, and so on [9,10,11,12,13]. Those stimuli-responsive drug carriers such as micelles [14], liposomes [15], hydrogels [16], and nanoparticles [17] have been fabricated and undergo cleavage of chemical bonds or conformational change to release drugs at a specific pattern.
With the deep understanding of lung cancer studies, researchers have realized the unique tumor microenvironment that occurs during the progression of lung cancer, such as acidic tumoral environment [18], elevated intracellular glutathione (GSH) conditions [19], and high levels of ROS [20], thereby provides an opportunity for the application of drug delivery systems for lung cancer therapy. In addition to stimuli-based nanocarriers that are sensitive to internal (temperature, pH, redox, enzymes reactions) environment, external (ultrasound, electric field, light, magnetic field) conditions could also be utilized to trigger fast drug release at a designated localization [21,22,23,24]. Additionally, nanocarriers can be organic, inorganic, or hybrid and follow either an active mechanism or a passive mechanism for tumor targeting.
Herein, we reviewed the recent studies on stimuli-responsive drug delivery systems for the diagnosis and therapy of lung cancer. These drug delivery systems could respond to pH, enzyme, ROS, magnetic field, photodynamic, or ultrasound, aiming to enhance drug efficacy and meanwhile minimize side effects (Table 1).

2. Light-Responsive Nanocarriers

Because of its relative safety and noninvasive character, light has been widely applied for remotely controlled drug delivery [25]. Short-wavelength light, including ultraviolet (UV) and visible light, can be utilized to destruct photolabile groups directly for on-demand drug release. However, their limited penetration ability hinders their biomedical application. In contrast to short-wavelength light, near-infrared (NIR) light (780–2500 nm) is able to penetrate deeper through the tissues, and this feature of NIR is preferable for remote control of the desired drug release. To take advantage of both short-wavelength and NIR light, up-conversion nanoparticles (UCNPs), which are capable of transferring NIR light to short-wavelength light, are utilized to fulfill both deep penetration and short-wavelength light-responsive drug release. For example, Ming-Fong Tsai et al. used a UV-responsive o-nitrobenzyl ester (ONB) containing amphiphilic block copolymer to construct a polymersome [26]. Then core-shell UCNPs and doxorubicin (DOX) were co-encapsulated to the polymersome, enabling NIR light-inducing photolysis and on-demand drug release for enhanced chemotherapy of lung cancer.
Apart from the immediate breakdown of photolabile groups and structural transformation of the therapeutic system triggered by external light sources, some nanoparticles can be activated by light to generate reactive oxygen species (ROS) or increase local temperatures, which can be applied in killing tumor cells [27,28]. These therapeutic strategies are known as photodynamic therapy (PDT) and photothermal therapy (PTT), respectively. For instance, the core-shell gold nanocage@manganese dioxide (AuNC@MnO2) nanoparticles were developed by Ruijing Liang et al. to simultaneously ablate primary triple-negative breast cancer and prevent lung metastases via oxygen-boosted immunogenic PDT [29]. Gold nanomaterials can be utilized not only as PDT agents but also PTT agents. Jianli Chen et al. fabricated titania-coated gold nanobipyramids and loaded anticancer drug combretastatin A-4 phosphate to induce synergistic chemotherapy and PTT to lung cancer with 1064 nm laser irradiation [30].
Accumulating evidence indicates that the combining of various therapeutic modalities (e.g., PTT, PDT, immunotherapy, chemotherapy) is a promising strategy for enhanced anti-tumor efficiency with minimized side effects. Additionally, the ROS or thermal effects generated during PDT or PTT can also trigger the drug release in the meantime. For instance, afatinib was loaded into the poly(l-lysine)-conjugated chlorin e6 (Ce6) derivative nanoparticle and covered by hyaluronic acid (HA) shells by Man Zhang et al. [31]. Upon NIR irradiation, the photosensitizer Ce6 generated ROS which induced the oxidation of the thioether linker and subsequently triggered the release of afatinib for improved therapeutic efficacy in non-small-cell lung cancer (NSCLC) treatment. Similarly, Chongchong Wang et al. fabricated a palladium nanosheet (PdNS) to carry carbon monoxide via reaction with transition metals, which could be destroyed by the heat generated from the PdNS upon 808 nm laser irradiation [32].
Among multiple therapeutic modalities, immunotherapy is now regarded as the first-line therapy for many cancer indications and revolutionized the field of oncology during the past decade [33]. Specifically, it has been demonstrated that phototherapy can trigger immunogenic cell death (ICD) of tumor cells [34]. However, with the immunosuppressive tumor microenvironment, as well as multiple mechanisms involved, adaptive immune resistance may restrain the anti-tumor activity of the ICD cascade. Thus, immunotherapeutic molecules are sometimes introduced into the delivery system for enhanced immunotherapy-assisted synergistic treatment. Additionally, light-controlled release can further raise the specificity [35,36]. For instance, Jingchao Li et al. proposed a second near-infrared (NIR-II) photothermal immunotherapy using a semiconducting polymer nanoadjuvant (SPNIIR), which was composed of a semiconducting polymer nanoparticle core, a toll-like receptor agonist R848, and a thermally responsive lipid shell (DPPC) [37]. Under the irradiation of a NIR-II laser, the thermal effect of the semiconducting polymer nanoparticle core caused the removal of the DPPC shell and induced the on-demand release of R848. Consequently, the synergistic photothermal immunotherapy could suppress primary tumors and eliminate lung metastasis in vivo (Figure 2).
Light-triggered drug release can also be combined with other stimuli, including internal stimuli (e.g., pH [38,39,40], enzyme [41], glutathione [42,43], and other external stimuli (e.g., radio frequency [44]), for enhanced targeted therapy to lung cancer. For example, as the tumor microenvironment is enriched in esterase, introducing the esterase-labile ester bond to the therapeutic system can achieve tumor microenvironment-responsive drug release in situ. Combined with the photoexcited effect, the encapsulated drug is able to be released to its maximum extent [45]. Similarly, introducing a disulfide bond to a photosensitive therapeutic system can fabricate a dual-responsive (GSH/light) therapeutic agent as well [42]. Radio frequency stimulation is also a widely adopted noninvasive therapeutic tool by generating heat like laser irradiation. Based on this concept, Animesh Pan et al. loaded DOX and iron oxide nanoparticles (IONs) coated with a gold nanoshell into the layersome to perform combined hyperthermia and triggered drug release via radio frequency or NIR stimulation. Compared to the single PTT or radio frequency treatment, the DOX and nanoparticles-loaded layersome with dual stimulation displayed a higher therapeutic effect on non-small cell lung cancer A549 cells [44].
Light-responsive therapeutics exhibit multiple advantages as mentioned above and hold great potential for clinical treatment of lung cancer [41]. Furthermore, molecules that absorb light and generate heat or ROS sometimes can also emit fluorescence or transfer the heat to photoacoustic (PA) signal and thus are employed for imaging diagnosis during the therapeutic process. For instance, indocyanine green (ICG) is a typical photothermal molecule and can also be utilized for NIR fluorescence and PA imaging [43]. Other fluorescent molecules, such as IR780 [46,47], Cy7 [48], and NIR770 [41], are also applied as theranostic agents to the treatment of lung cancer. Ziying Li et al. [48] established a chitosan-based nanocomplex CE7Q/CQ/S to deliver molecular-targeted drug erlotinib (Er), Survivin shRNA-expressing plasmid (SV), and Cy7 for simultaneous NIR fluorescence imaging and monitored chemo/gene/photothermal tri-therapies therapy for NSCLC bearing epidermal growth factor receptor (EGFR) mutations. With the guidance of NIR imaging, the therapy was more accurate, and the therapeutic outcome was able to be observed in real-time.

3. Ultrasound-Responsive Nanocarriers

Ultrasound (usually defined as > 20 kHz) is widely used for diagnosis and therapy in the clinic [49]. Due to its merits in clinical application, including cost-effectiveness, simplicity, and particularly noninvasiveness, ultrasound has been adopted as an external stimulus for smart therapeutics to trigger amplified therapeutic effects. Micro-/nanobubbles, liposomes, liquid perfluorocarbon droplets, micelles, or mesoporous silica nanoparticles (MSN) have been developed as ultrasound-responsive drug carriers after rational design and synthesis.
The fundamental mechanisms underlying ultrasound-mediated therapy mainly include thermal effect, mechanical effect, and chemical effect [50]. The thermal effects are attributed to acoustic energy produced by propagating ultrasound. Surrounding biological tissues can absorb part of the energy and thus lead to a temperature increase in the respective areas. Relative high temperature is able to kill cancer cells directly, while hyperthermia (above 80 °C) caused by high intensity focused ultrasound (HIFU) may be associated with undesired complications, such as second- and third-degree skin burns [51]. On the other hand, the temperature increase induced by ultrasound could probably trigger the thermal instability of the drug delivery system and enable targeted controlled drug release.
The mechanical effects are mainly generated from ultrasound pressure, acoustic streaming, and ultrasound-induced oscillation or cavitation [49], among which cavitation is often leveraged for drug delivery owing to its specific influence on biological processes. For example, upon exposure to intense ultrasound energy, perfluoropentane containing nanobubbles would go through rapid bubble destruction triggered by rapid contraction and expansion of the bubbles. The bubble shell can be weakened by this mechanical stress until the bubble ruptures, leading to the drug release in situ [52]. Sonoporation is the process of pore formation in a cell membrane upon exposure to ultrasound and belongs to one of the cavitation effects, which could facilitate the intracellular transport of drugs. Moreover, cavitation can also widen the interspace between endothelial cells and thus enhance the penetration into adjacent tissues [50]. However, unwanted cavitation effects may take place in the presence of residual air bubbles, and thus the implementation of ultrasound in the treatment of lung cancer would lead to undesired drug release in the process of drug transportation.
The chemical effects of ultrasound mediated treatment can also be called sonodynamic therapy (SDT). Oxygen, sonosensitizer, and appropriate ultrasound are three necessary components to complete the process of SDT, and the generation of ROS upon focused ultrasound exposure can cause site-specific profound damage to tumor tissues [53]. The combinatorial treatment of SDT with other therapeutic strategies, such as chemotherapy and chemodynamic therapy (CDT), has a synergistic effect in the treatment of lung cancer [54]. For example, Shiyan Fu et al. synthesized PEGylated Co2Fe2O4 nanoflowers (CFP) [55]. This CFP occupying multivalent elements (Co2+/3+and Fe2+/3+) exhibited strong Fenton-like and catalase-like activity. Moreover, CFP could also be employed for high-performance SDT as a brand-new sonosensitizer attributed to the ultrasound-triggered electron (e)/hole (h+) pair separation from the energy band. After efficient accumulation in the tumorous region as revealed by magnetic resonance imaging, CFP could generate OH for CDT relying on Fenton-like reactions and generate molecular oxygen due to the catalase-like activity which may promote the production of 1O2 for SDT. Combined SDT/CDT could further efficiently trigger ICD and thus synergistically suppress primary and distant tumors, as well as lung metastasis.
Thermal, mechanical, and chemical effects are not entirely independent and sometimes two or three of them may result in a synergistically therapeutic modality. For instance, Xiaotu Ma et al. fabricated a cerasomal perfluorocarbon nanodroplet (D-vPCs-O2) with an atomic layer of polyorganosiloxane and pH-sensitive tumor-targeting peptide [55]. Oxygen and doxorubicin were co-loaded into the nanodroplets. HIFU was utilized to trigger the release of cargoes and simultaneously enhance ultrasound imaging, therefore achieving imaging-guided drug delivery. Mild-temperature HIFU (M-HIFU) could also be applied to slightly elevate tumor temperature and accelerate tumor blood flow. Consequently, ultrasound-triggered oxygen release and temperature elevation jointly relieved tumor hypoxia and alleviated multiple drug resistance, and these two effects jointly enhanced the drug therapeutic efficacy to lung metastasis [56] (Figure 3).
Two ultrasound parameters, acoustic frequency, and intensity, are often manipulated to induce desired biological effects. For example, Yichen Liu et al. constructed a functionalized smart nano sonosensitizer (EXO-DVDMS) by loading sinoporphyrin sodium (DVDMS), which was an excellent porphyrin sensitizer with both therapeutic and diagnostic features, onto homotypic tumor cell-derived exosomes [53]. A guided-ultrasound (US1, 2 W, 3 min) was first introduced to promote the accumulation of EXO-DVDMS in the tumor region, and subsequently, the therapeutic-ultrasound (US2, 3 W, 3 min) was applied for SDT, thus enhancing the targeted delivery of DVDMS to primary as well as metastatic lung tumors. In addition, other external stimuli, such as magnetic fields, can be incorporated with ultrasound-responsive delivery systems and achieve precisely controlled release. Senay Hamarat Sanlier et al. fabricated liposome-based nanobubbles [57]. Pemetrexed and pazopanib were conjugated with peptide and then attached to the surface of magnetic nanoparticles. After the functionalized magnetic nanoparticles were encapsulated into the liposomes, pemetrexed and pazopanib carrying nanobubble systems with magnetic responsiveness and ultrasound sensitivity were constructed for NSCLC targeted delivery [58]. Furthermore, the inclusion of magnetic nanoparticles can not only enable the magnetic field-guided targeted delivery but also considerably improve both the stability and phase conversion efficiency of nanodroplets.

4. PH-Responsive Nanocarriers

The lactic acid and certain end products produced by lung cancer cells, which are related to an abnormally fast metabolism and proliferation, lead to a more acidic environment (pH 5.7–6.9) in tumor tissues than normal physiological pH (pH 7.4) [59,60]. pH-sensitive nanoparticles could maximize drug release in the pulmonary tumor microenvironment and minimize drug release en route to the tumor, enhancing the accumulation of nanosystems in the tumor tissue and meanwhile improving the reliability and safety of targeted therapy for lung cancer.
Lee and coworkers found that the CHEMS-based liposomes could be efficiently triggered by the acidic pH and these drug-loaded carriers exhibited an outstanding anti-tumor effect in NSCLC. Folate receptor beta (FRβ), which was usually overexpressed in M2 tumor-associated macrophages (TAMs) and NSCLC cells, was associated with the poor prognosis of NSCLC patients [61]. Since cholesteryl hemisuccinate(CHEMS) was unstable in acidic conditions, the conjugation of CHEMS onto PEG-Folate was used to construct pH-sensitive liposomes to achieve targeted drug release. As a result, these liposomes showed a faster drug release profile at pH 6.5 than that at neutral pH, and a burst release was observed at pH 4.0. Acylhydrazone bond could also be used as a pH-sensitive linker between polyethylene glycol (PEG) and tumor-targeted hyaluronic acid (HA), resulting in HA-ERL/BEV-LPH nanoparticles to treat NSCLC. In vitro release profiles of HA-ERL/BEV-LPH nanoparticles demonstrated that pH-sensitive adipic acid dihydrazide (ADH) could effectively control drug release in the acidic pH and release drugs faster than that at physiological pH [62]. The poly-γ-benzyl-l-glutamate and an amphiphilic copolymer d-α-tocopherol polyethylene glycol succinate mixed micellar system could control the release of DOX by changing the secondary structures of poly-γ-benzyl-l-glutamate. The DOX-loaded mixed micelles exhibited great anti-tumor efficacy in human lung cancer A549 cells-bearing nude mice [63].
Additionally, the acid pH of the tumor environment also could trigger charge reversal to promote cellular internalization and nuclear entry in the treatment of lung cancer (Figure 4). Nanocarriers with positive surface charge usually bear short blood circulation half-life due to an unspecific adsorption, whereas the addition of TAT peptide would overcome this drawback by improving drug uptake of tumor cells. Anhydride (DA) groups can be utilized to mask the positive charges of TAT. Once the carrier accumulated in the tumor acidic environment, a charge reversal from negative to positive occurred and the targeting ability of TAT was recovered. Zhou et al. generated a DA-TAT carrier for pH-triggered cell uptake and nuclear targeting, which possessed beneficial effects in treating lung metastasis [64]. Similarity, Zhao et al. developed a pH-responsive poly(histidine) (PHis) based polymer consisting of a cationic lipid core and a triblock copolymer methoxy poly(ethylene glycol)-poly(histidine)-poly(sulfadimethoxine) (mPEG-PHis-PSD or PHD). Acidic pH transformed PSD from a negative to neutral charge, which resulted in a fast dissociation from lipid core, thus achieving tumor-selective accumulation, effective internalization, and efficient anti-tumor activity for NSCLC therapy [65].
Prodrug-based nanosystem was a great choice with high drug-loading capacity and could load additional drugs for synergistic treatment [66,67]. Ma and coworkers constructed a pH-sensitive doxorubicin (DOX) prodrug for lung cancer therapy. The hydrophilic segment U11-PEG was introduced to DOX by pH-responsive PHis. Curcumin (CUR) was loaded into DOX-based nanoparticles as a secondary anti-tumor drug. When the prodrug-based codelivery system U11-DOX/CUR nanoparticles were exposed to the tumor acidic microenvironment, DOX and CUR were released simultaneously because of the protonation of pHis. This study suggested that the U11-DOX/CUR nanoparticles are pH-responsive systems and had a potent anti-tumor effect on lung tumor cells [68]. Cis-aconitic anhydride-modified doxorubicin (CAD) was designed for pH-sensitive drug release in another study. CAD showed specific distribution in the tumor tissues after 12 h post-injection, exhibiting excellent lung tumor-targeting ability of these nanoparticles. The acid-responsive cis-aconityl linkage between the cis-aconitic anhydride (CA) and antitumor drug DOX could be hydrolyzed, and the release of DOX would accelerate the linkage breakdown once the nanoparticles reached tumor tissues [69].

5. Enzyme-Responsive Nanocarriers

Enzymes are essential biomolecules that maintain normal functions of living organisms, e.g., growth, development, metabolism, aging, disease, and immunity. Aberrant enzyme expression was commonly observed in multiple disease-associated microenvironments and cells, especially lung cancer [70,71]. The overexpressed enzymes mainly include matrix metalloproteinases (MMPs), hyaluronidase (HAase), esterase, NAD(P)H, and quinone oxidoreductase1 (NQO1). Enzyme-responsive nanoparticles have attracted considerable attention owing to their selectivity, effectiveness, and rapidity of enzymatic reactions in lung cancer treatment [72,73,74]. For example, MMPs are a family of proteolytic zinc-dependent secreted endopeptidases that can specifically degrade a variety of compositions in extracellular matrices (ECMs). Abnormally high expression of MMPs within lung tumor tissues could be utilized for the development of enzyme-responsive drug delivery systems [72,75]. Based on the concept that MMP-9 and MMP-2 can specifically degrade collagen and basement membrane, gelatin or the MMP-responsive peptides are generally conjugated onto the surface of nanoparticles for targeted lung cancer therapy [76]. Guo et al. conjugated MPEG to PCL using an MMP-2 sensitive peptide linker (GPLGIAGQ), resulting in lung targeted Cur-P-NPs, which exhibited a superior drug release profile than that of the non-responsive control [77]. Likewise, an engineered shell composed of phosphorylcholine (PC) and enzyme-responsive peptides was constructed by Kang and coworkers. The functional vehicle facilitated the precise delivery of loaded protein drugs once reaching the lung tumor sites; then the therapeutic agents would bind to pulmonary tumor cell surface receptors to suppress cell growth [78]. Gianneschi designed an MMP-9-sensitive nanocarrier to deliver the immunotherapeutic small molecule (1V209) for selective immune activation. The aforementioned platform enhanced drug efflux and inhibited in vivo lung tumor metastasis [78]. In addition, gelatin can also be used as a substrate for MMPs, and be incorporated as the carrier skeleton. Verma designed a smart inhalable nanocarrier by complexation of gelatin with cisplatin for lung tumor therapy. After the degradation of gelatin, cisplatin was exposed to physiological salts and exchanged with chloride ions, leading to the fast release of cisplatin at the tumor site [79].
Apart from MMPs, HAase is usually combined with other sensitive patterns to generate dual- or multi-responsive nanoparticles to improve lung cancer therapeutic effectiveness. Hyaluronic acid (HA) was used to construct the hydrophilic shell, while hydrophobic compounds could be introduced into the HA backbone by environmentally responsive bonds. For example, Tang’s group utilized pH-sensitive hydrazone bonds to construct enzyme and pH dual-responsive hyaluronic acid nanoparticles [80]. In a similar study, He and coworkers designed lung cancer cells’ active-targeting, enzyme, and ROS-sensitive nanoparticles named HPGBCA to deliver afatinib for NSCLC therapy. Poly(glycidylbutylamine) (PGBA) was a cationic amphiphilic compound with a ROS-sensitive thioether linker. The anionic HA shell could actively target CD44 receptor-overexpressed tumor cells and mask the positive charge of PGBA for long circulation in the bloodstream. When HPGBCA reached HAase-enriched lung tumor sites, the HA shell was degraded to expose positively charged cores and accelerate the lysosomal escape. Subsequently, the Ce6 of HPGBCA could produce ROS under NIR irradiation to trigger the oxidation of ROS-sensitive linkers for drug release [31].
The esterase-sensitive nanocarrier is also a great choice in enzyme-responsive drug delivery systems for the lung. A nanoparticle named HAPBA, which was designed by Cho and coworkers for lung cancer therapy, would release drugs at esterase-enriched tumor tissue environment. Ester bonds were used to join 4-Phenylbutyric acid (PBA) and HA backbone for the quick release of curcumin and PBA. PBA was not only the hydrophobic segment in the structure of these nanoparticles but also an efficient inhibitor of histone deacetylase (HDAC). The cleavage of ester bonds realized the rapid release of curcumin and PBA, exhibiting efficient tumor growth suppression in lung adenocarcinoma [81]. Similarly, Ren et al. designed a gold nanorod–curcumin conjugate held together by an esterase-labile ester bond. This conjugate showed a rapid and sustained release of curcumin. In the absence of esterase, encapsulated drugs were completely restricted inside the nanoparticles. When the concentration of esterase increased, an abrupt curcumin release was observed, suggesting that ester hydrolysis was an essential trigger of drug release. As a result, the introduction of the ester bond enhanced the inhibitory effects of the nanorod–curcumin conjugate on human lung cancer A549 cells [45].
NQO1 enzyme is a cytosolic reductase that is abnormally overexpressed in multiple cancers, including lung cancer [82,83]. Trimethyl-locked quinone propionic acid (QPA) reacts with NQO1 to form a lactone-based group via intramolecular cyclization. To take advantage of the fundamental features of NQO1, an NQO1-responsive nanoparticle termed QPA-P was designed by Kim et al. for lung cancer therapy. Poly(ethylene glycol) (PEG) was used as the hydrophilic segment and QPA-locked polycaprolactone (PCL), which was conditionally triggered by NQO1, was the hydrophobic tail that imparted amphiphilic property to QPA-P. After a cascade two-step cyclization process with the NQO1 enzyme, the particle size of QPA-P increased and loaded DOX rapidly released into surrounding medium and tumor cells, indicating that NQO1-sensitive micelles were promising for drug delivery in lung cancer [84].
There are still some issues that should be considered. The drug release efficiency totally depends on the specific enzyme concentration of lung tumor cells. Hence, the type and the stage of the tumor must be taken into full consideration. Furthermore, the stability of enzyme-responsive nanoparticles in the physiological environment is also important.

6. Discussion and Perspective

In the last ten years, lung cancer therapy has attracted intense attention not only because of the fact that lung cancer is still the most common type of cancer in the world but also considering the fast development of targeted therapy drugs for lung cancer. However, those medicines that have already benefited millions of patients are faced with undesirable adverse effects and thus, cause discontinuation of the treatment, which would substantially undermine the effectiveness of therapeutic drugs. Stimuli-responsive carriers provide a promising solution to overcome the drawbacks of traditional medicine, mainly through enhancing the accumulation of drugs and precisely releasing drugs at lung tumor sites. However, major challenges still remain and hinder the wide application of these drug carriers. Most of these carriers are complicated in terms of structure and formulation, and it is difficult to control the stability, integrity, and physiochemical property in the scale-up production. Additionally, the tumor microenvironment is one of the critical obstacles that devastate the transport of drug carriers and also greatly affect the drug release under certain circumstances, whereas few drug carriers have fully considered the complexity of the lung cancer microenvironment at the time of original design and development. In spite of these current hurdles, scientists are making tremendous efforts to fabricate drug carriers and optimize their features.
It is worth mentioning that several stimuli-responsive systems have already been approved by the FDA and are currently under clinical trials. For example, the first heat-activated nanocarrier (lyso-thermosensitive liposomal doxorubicin, LTLD, ThermoDox®) that was utilized in clinical trials has shown a promising advantage in improving the overall survival of primary liver cancer patients [85]. Some other systems, such as ferumoxytol and oral iron, which could be used for the treatment of iron deficiency anemia in the setting of chronic kidney disease and enhanced MRI, have shown their clinical applications in therapeutic functions and cancer diagnosis [86]. An intelligent, target-controlled liposome formulation (LiPlaCis®) has been evaluated for its therapeutic effects in prostate cancer treatment [87]. These inspiring results elicited that the exploration of unique targeted biomarkers was necessary to utilize interior stimulus, and meanwhile, the precise manipulation of external stimulus would lead to a sophisticated release of cargo drugs from nanocarriers. In addition, the strategy to combine those drug delivery systems which are based on tumor microenvironment or external stimuli with other anticancer drugs or immunoregulatory agents might display synergistic effects in lung cancer therapy.
Overall, the path to find the most suitable stimuli-responsive drug delivery systems for lung cancer therapy still requires numerous contributions in the near future.

Author Contributions

Conceptualization, B.Z. and J.H.; methodology, J.W.; software, Y.L.; validation and investigation, X.L.; resources, N.L. All authors have read and agreed to the published version of the manuscript.

Funding

Special thanks to Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Westlake University, for their financial support in this project. This work was supported by the National Natural Science Foundation (82073297), Hangzhou Major Science and Technology Project (20172016A01), the Natural Science Foundation of Zhejiang Province (LY20H310005, LQ22H180009), the Clinical Pharmacy of Hangzhou Medical Key Discipline (2021-21-16), and the Postdoctoral Research Foundation of China (2021M702876).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wang, M.; Herbst, R.S.; Boshoff, C. Toward personalized treatment approaches for non-small-cell lung cancer. Nat. Med. 2021, 27, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
  2. Sands, J.; Tammemagi, M.C.; Couraud, S.; Baldwin, D.R.; Borondy-Kitts, A.; Yankelevitz, D.; Lewis, J.; Grannis, F.; Kauczor, H.U.; von Stackelberg, O.; et al. Lung Screening Benefits and Challenges: A Review of The Data and Outline for Implementation. J. Thorac. Oncol. 2021, 16, 37–53. [Google Scholar] [CrossRef] [PubMed]
  3. Thai, A.A.; Solomon, B.J.; Sequist, L.V.; Gainor, J.F.; Heist, R.S. Lung cancer. Lancet 2021, 398, 535–554. [Google Scholar] [CrossRef]
  4. Horvath, L.; Thienpont, B.; Zhao, L.; Wolf, D.; Pircher, A. Overcoming immunotherapy resistance in non-small cell lung cancer (NSCLC)—Novel approaches and future outlook. Mol. Cancer 2020, 19, 141. [Google Scholar] [CrossRef] [PubMed]
  5. Tumbrink, H.L.; Heimsoeth, A.; Sos, M.L. The next tier of EGFR resistance mutations in lung cancer. Oncogene 2021, 40, 1–11. [Google Scholar] [CrossRef]
  6. Li, J.; Burgess, D.J. Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm. Sin. B 2020, 10, 2110–2124. [Google Scholar] [CrossRef]
  7. Mi, P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 2020, 10, 4557–4588. [Google Scholar] [CrossRef]
  8. Davoodi, P.; Lee, L.Y.; Xu, Q.; Sunil, V.; Sun, Y.; Soh, S.; Wang, C.H. Drug delivery systems for programmed and on-demand release. Adv. Drug Deliv. Rev. 2018, 132, 104–138. [Google Scholar] [CrossRef]
  9. Di, J.; Xie, F.; Xu, Y. When liposomes met antibodies: Drug delivery and beyond. Adv. Drug Deliv. Rev. 2020, 154, 151–162. [Google Scholar] [CrossRef]
  10. Hoffman, A.S.; Lai, J.J. Three significant highlights of controlled drug delivery over the past 55 years: PEGylation, ADCs, and EPR. Adv. Drug Deliv. Rev. 2020, 158, 2–3. [Google Scholar] [CrossRef]
  11. Yu, H.; Jin, F.; Liu, D.; Shu, G.; Wang, X.; Qi, J.; Sun, M.; Yang, P.; Jiang, S.; Ying, X.; et al. ROS-responsive nano-drug delivery system combining mitochondria-targeting ceria nanoparticles with atorvastatin for acute kidney injury. Theranostics 2020, 10, 2342–2357. [Google Scholar] [CrossRef] [PubMed]
  12. Zhou, N.; Zhi, Z.; Liu, D.; Wang, D.; Shao, Y.; Yan, K.; Meng, L.; Yu, D. Acid-responsive and biologically degradable polyphosphazene nanodrugs for efficient drug delivery. ACS Biomater. Sci. Eng. 2020, 6, 4285–4293. [Google Scholar] [CrossRef] [PubMed]
  13. Duan, W.; Ji, S.; Guan, Y.; Mu, X.; Fang, S.; Lu, Y.; Zhou, X.; Sun, J.; Li, Z. Esterase-responsive polypeptide vesicles as fast-response and sustained-release nanocompartments for fibroblast-exempt drug delivery. Biomacromolecules 2020, 21, 5093–5103. [Google Scholar] [CrossRef] [PubMed]
  14. Lu, L.; Zhao, X.; Fu, T.; Li, K.; He, Y.; Luo, Z.; Dai, L.; Zeng, R.; Cai, K. An iRGD-conjugated prodrug micelle with blood-brain-barrier penetrability for anti-glioma therapy. Biomaterials 2020, 230, 119666. [Google Scholar] [CrossRef]
  15. Almeida, B.; Nag, O.K.; Rogers, K.E.; Delehanty, J.B. Recent progress in bioconjugation strategies for liposome-mediated drug delivery. Molecules 2020, 25, 5672. [Google Scholar] [CrossRef]
  16. Mo, F.; Jiang, K.; Zhao, D.; Wang, Y.; Song, J.; Tan, W. DNA hydrogel-based gene editing and drug delivery systems. Adv. Drug Deliv. Rev. 2021, 168, 79–98. [Google Scholar] [CrossRef] [PubMed]
  17. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef] [PubMed]
  18. Shi, M.; Zhang, J.; Huang, Z.; Chen, Y.; Pan, S.; Hu, H.; Qiao, M.; Chen, D.; Zhao, X. Stimuli-responsive release and efficient siRNA delivery in non-small cell lung cancer by a poly(l-histidine)-based multifunctional nanoplatform. J. Mater. Chem. B 2020, 8, 1616–1628. [Google Scholar] [CrossRef]
  19. Feng, W.; Zong, M.; Wan, L.; Yu, X.; Yu, W. pH/redox sequentially responsive nanoparticles with size shrinkage properties achieve deep tumor penetration and reversal of multidrug resistance. Biomater. Sci. 2020, 8, 4767–4778. [Google Scholar] [CrossRef]
  20. Gai, C.; Liu, C.; Wu, X.; Yu, M.; Zheng, J.; Zhang, W.; Lv, S.; Li, W. MT1DP loaded by folate-modified liposomes sensitizes erastin-induced ferroptosis via regulating miR-365a-3p/NRF2 axis in non-small cell lung cancer cells. Cell Death Dis. 2020, 11, 751. [Google Scholar] [CrossRef]
  21. Akbaba, H.; Erel-Akbaba, G.; Kotmakci, M.; Baspinar, Y. Enhanced cellular uptake and gene silencing activity of survivin-siRNA via ultrasound-mediated nanobubbles in lung cancer cells. Pharm. Res. 2020, 37, 165. [Google Scholar] [CrossRef] [PubMed]
  22. Bomzon, Z.; Urman, N.; Wenger, C.; Giladi, M.; Weinberg, U.; Wasserman, Y.; Kirson, E.D.; Miranda, P.C.; Palti, Y. Modelling tumor treating fields for the treatment of lung-based tumors. Annu. Int. Conf. IEEE Eng. Med. Biol. Soc. 2015, 2015, 6888–6891. [Google Scholar] [PubMed]
  23. Diaz, D.; Vidal, X.; Sunna, A.; Care, A. Bioengineering a light-responsive encapsulin nanoreactor: A potential tool for in vitro photodynamic therapy. ACS Appl. Mater. Interfaces 2021, 13, 7977–7986. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, D.; Hong, Y.; Li, Y.; Hu, C.; Yip, T.C.; Yu, W.K.; Zhu, Y.; Fong, C.C.; Wang, W.; Au, S.K.; et al. Targeted destruction of cancer stem cells using multifunctional magnetic nanoparticles that enable combined hyperthermia and chemotherapy. Theranostics 2020, 10, 1181–1196. [Google Scholar] [CrossRef] [PubMed]
  25. Li, F.; Qin, Y.; Lee, J.; Liao, H.; Wang, N.; Davis, T.P.; Qiao, R.; Ling, D. Stimuli-responsive nano-assemblies for remotely controlled drug delivery. J. Control. Release 2020, 322, 566–592. [Google Scholar] [CrossRef] [PubMed]
  26. Tsai, M.-F.; Lo, Y.-L.; Soorni, Y.; Su, C.-H.; Sivasoorian, S.S.; Yang, J.-Y.; Wang, L.-F. Near-infrared light-triggered drug release from ultraviolet- and redox-responsive polymersome encapsulated with core–shell upconversion nanoparticles for cancer therapy. ACS Appl. Bio. Mater. 2021, 4, 3264–3275. [Google Scholar] [CrossRef] [PubMed]
  27. Ko, S.; Park, J.Y.; Oh, Y.K. A microbial siderophore-inspired self-gelling hydrogel for noninvasive anticancer phototherapy. Cancer Res. 2019, 79, 6178–6189. [Google Scholar] [CrossRef] [Green Version]
  28. Lee, C.; Lim, K.; Kim, S.S.; Thien, L.X.; Lee, E.S.; Oh, K.T.; Choi, H.G.; Youn, Y.S. Near infrared light-responsive heat-emitting hemoglobin hydrogels for photothermal cancer therapy. Colloids Surf. B Biointerfaces 2019, 176, 156–166. [Google Scholar] [CrossRef]
  29. Liang, R.; Liu, L.; He, H.; Chen, Z.; Han, Z.; Luo, Z.; Wu, Z.; Zheng, M.; Ma, Y.; Cai, L. Oxygen-boosted immunogenic photodynamic therapy with gold nanocages@manganese dioxide to inhibit tumor growth and metastases. Biomaterials 2018, 177, 149–160. [Google Scholar] [CrossRef]
  30. Chen, J.L.; Zhang, H.; Huang, X.Q.; Wan, H.Y.; Li, J.; Fan, X.X.; Luo, K.Q.; Wang, J.; Zhu, X.M.; Wang, J. Antiangiogenesis-combined photothermal therapy in the second near-infrared window at laser powers below the skin tolerance threshold. Nanomicro Lett. 2019, 11, 93. [Google Scholar] [CrossRef]
  31. Zhang, M.; Zhang, X.; Cai, S.; Mei, H.; He, Y.; Huang, D.; Shi, W.; Li, S.; Cao, J.; He, B. Photo-induced specific intracellular release EGFR inhibitor from enzyme/ROS-dual sensitive nano-platforms for molecular targeted-photodynamic combinational therapy of non-small cell lung cancer. J. Mater. Chem. B 2020, 8, 7931–7940. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, C.; Li, Y.; Shi, X.; Zhou, J.; Zhou, L.; Wei, S. Use of an NIR-light-responsive CO nanodonor to improve the EPR effect in photothermal cancer treatment. Chem. Commun. 2018, 54, 13403–13406. [Google Scholar] [CrossRef] [PubMed]
  33. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
  34. Jin, F.; Qi, J.; Zhu, M.; Liu, D.; You, Y.; Shu, G.; Du, Y.; Wang, J.; Yu, H.; Sun, M.; et al. NIR-Triggered Sequentially Responsive Nanocarriers Amplified Cascade Synergistic Effect of Chemo-Photodynamic Therapy with Inspired Antitumor Immunity. ACS Appl. Mater. Interfaces 2020, 12, 32372–32387. [Google Scholar] [CrossRef] [PubMed]
  35. Jiang, Y.; Huang, J.; Xu, C.; Pu, K. Activatable polymer nanoagonist for second near-infrared photothermal immunotherapy of cancer. Nat. Commun. 2021, 12, 742. [Google Scholar] [CrossRef] [PubMed]
  36. Feng, B.; Hou, B.; Xu, Z.; Saeed, M.; Yu, H.; Li, Y. Self-amplified drug delivery with light-inducible nanocargoes to enhance cancer immunotherapy. Adv. Mater. 2019, 31, e1902960. [Google Scholar] [CrossRef] [PubMed]
  37. Li, J.; Yu, X.; Jiang, Y.; He, S.; Zhang, Y.; Luo, Y.; Pu, K. Second near-infrared photothermal semiconducting polymer nanoadjuvant for enhanced cancer immunotherapy. Adv. Mater. 2021, 33, e2003458. [Google Scholar] [CrossRef] [PubMed]
  38. Du, P.; Yan, J.; Long, S.; Xiong, H.; Wen, N.; Cai, S.; Wang, Y.; Peng, D.; Liu, Z.; Liu, Y. Tumor microenvironment and NIR laser dual-responsive release of berberine 9-O-pyrazole alkyl derivative loaded in graphene oxide nanosheets for chemo-photothermal synergetic cancer therapy. J. Mater. Chem. B 2020, 8, 4046–4055. [Google Scholar] [CrossRef]
  39. Xing, Y.; Zhou, Y.; Zhang, Y.; Zhang, C.; Deng, X.; Dong, C.; Shuang, S. Facile fabrication route of janus gold-mesoporous silica nanocarriers with dual-drug delivery for tumor therapy. ACS Biomater. Sci. Eng. 2020, 6, 1573–1581. [Google Scholar] [CrossRef]
  40. Li, J.; Zhang, Z.; Deng, H.; Zheng, Z. Cinobufagin-loaded and folic acid-modified polydopamine nanomedicine combined with photothermal therapy for the treatment of lung cancer. Front. Chem. 2021, 9, 637754. [Google Scholar] [CrossRef]
  41. Gou, S.; Yang, J.; Ma, Y.; Zhang, X.; Zu, M.; Kang, T.; Liu, S.; Ke, B.; Xiao, B. Multi-responsive nanococktails with programmable targeting capacity for imaging-guided mitochondrial phototherapy combined with chemotherapy. J. Control. Release 2020, 327, 371–383. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, J.; Li, F.; Zheng, J.; Li, B.; Zhang, D.; Jia, L. Redox/NIR dual-responsive MoS2 for synergetic chemo-photothermal therapy of cancer. J. Nanobiotechnol. 2019, 17, 78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Zhu, Q.; Fan, Z.; Zuo, W.; Chen, Y.; Hou, Z.; Zhu, X. Self-distinguishing and stimulus-responsive carrier-free theranostic nanoagents for imaging-guided chemo-photothermal therapy in small-cell lung cancer. ACS Appl. Mater. Interfaces 2020, 12, 51314–51328. [Google Scholar] [CrossRef] [PubMed]
  44. Pan, A.; Jakaria, M.G.; Meenach, S.A.; Bothun, G.D. Radiofrequency and near-infrared responsive core–shell nanostructures using layersome templates for cancer treatment. ACS Appl. Bio. Mater. 2020, 3, 273–281. [Google Scholar] [CrossRef] [PubMed]
  45. Zhu, F.; Tan, G.; Jiang, Y.; Yu, Z.; Ren, F. Rational design of multi-stimuli-responsive gold nanorod-curcumin conjugates for chemo-photothermal synergistic cancer therapy. Biomater. Sci. 2018, 6, 2905–2917. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, X.; Li, X.; Sun, S.; Wang, P.; Ma, X.; Hou, R.; Liang, X. Anti-tumor metastasis via platelet inhibitor combined with photothermal therapy under activatable fluorescence/magnetic resonance bimodal imaging guidance. ACS Appl. Mater. Interfaces 2021, 13, 19679–19694. [Google Scholar] [CrossRef] [PubMed]
  47. Yang, Z.; Cheng, R.; Zhao, C.; Sun, N.; Luo, H.; Chen, Y.; Liu, Z.; Li, X.; Liu, J.; Tian, Z. Thermo- and pH-dual responsive polymeric micelles with upper critical solution temperature behavior for photoacoustic imaging-guided synergistic chemo-photothermal therapy against subcutaneous and metastatic breast tumors. Theranostics 2018, 8, 4097–4115. [Google Scholar] [CrossRef]
  48. Li, Z.; Zhu, L.; Liu, W.; Zheng, Y.; Li, X.; Ye, J.; Li, B.; Chen, H.; Gao, Y. Near-infrared/pH dual-responsive nanocomplexes for targeted imaging and chemo/gene/photothermal tri-therapies of non-small cell lung cancer. Acta Biomater. 2020, 107, 242–259. [Google Scholar] [CrossRef] [PubMed]
  49. Cai, X.; Jiang, Y.; Lin, M.; Zhang, J.; Guo, H.; Yang, F.; Leung, W.; Xu, C. Ultrasound-responsive materials for drug/gene delivery. Front. Pharmacol. 2019, 10, 1650. [Google Scholar] [CrossRef] [PubMed]
  50. Entzian, K.; Aigner, A. Drug delivery by ultrasound-responsive nanocarriers for cancer treatment. Pharmaceutics 2021, 13, 1135. [Google Scholar] [CrossRef]
  51. Awad, N.S.; Paul, V.; AlSawaftah, N.M.; Ter Haar, G.; Allen, T.M.; Pitt, W.G.; Husseini, G.A. Ultrasound-responsive nanocarriers in cancer treatment: A review. ACS Pharmacol. Transl. Sci. 2021, 4, 589–612. [Google Scholar] [CrossRef] [PubMed]
  52. Baspinar, Y.; Erel-Akbaba, G.; Kotmakci, M.; Akbaba, H. Development and characterization of nanobubbles containing paclitaxel and survivin inhibitor YM155 against lung cancer. Int. J. Pharm. 2019, 566, 149–156. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, Y.; Bai, L.; Guo, K.; Jia, Y.; Zhang, K.; Liu, Q.; Wang, P.; Wang, X. Focused ultrasound-augmented targeting delivery of nanosonosensitizers from homogenous exosomes for enhanced sonodynamic cancer therapy. Theranostics 2019, 9, 5261–5281. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, Y.; Khan, A.R.; Yang, X.; Shi, Y.; Zhao, X.; Zhai, G. A sonosensitiser-based polymeric nanoplatform for chemo-sonodynamic combination therapy of lung cancer. J. Nanobiotechnol. 2021, 19, 57. [Google Scholar] [CrossRef] [PubMed]
  55. Fu, S.; Yang, R.; Ren, J.; Liu, J.; Zhang, L.; Xu, Z.; Kang, Y.; Xue, P. Catalytically active CoFe2O4 nanoflowers for augmented sonodynamic and chemodynamic combination therapy with elicitation of robust immune response. ACS Nano 2021, 15, 11953–11969. [Google Scholar] [CrossRef] [PubMed]
  56. Ma, X.; Yao, M.; Shi, J.; Li, X.; Gao, Y.; Luo, Q.; Hou, R.; Liang, X.; Wang, F. High intensity focused ultrasound-responsive and ultrastable cerasomal perfluorocarbon nanodroplets for alleviating tumor multidrug resistance and epithelial-mesenchymal transition. ACS Nano 2020, 14, 15904–15918. [Google Scholar] [CrossRef]
  57. Lee, J.Y.; Crake, C.; Teo, B.; Carugo, D.; de Saint Victor, M.; Seth, A.; Stride, E. Ultrasound-enhanced siRNA delivery using magnetic nanoparticle-loaded chitosan-deoxycholic acid nanodroplets. Adv. Healthc. Mater. 2017, 6, 1601246. [Google Scholar] [CrossRef] [Green Version]
  58. Hamarat Sanlier, S.; Ak, G.; Yilmaz, H.; Unal, A.; Bozkaya, U.F.; Taniyan, G.; Yildirim, Y.; Yildiz Turkyilmaz, G. Development of ultrasound-triggered and magnetic-targeted nanobubble system for dual-drug delivery. J. Pharm. Sci. 2019, 108, 1272–1283. [Google Scholar] [CrossRef]
  59. Kanamala, M.; Wilson, W.R.; Yang, M.; Palmer, B.D.; Wu, Z. Mechanisms and biomaterials in pH-responsive tumour targeted drug delivery: A review. Biomaterials 2016, 85, 152–167. [Google Scholar] [CrossRef]
  60. Shi, J.; Ren, Y.; Ma, J.; Luo, X.; Li, J.; Wu, Y.; Gu, H.; Fu, C.; Cao, Z.; Zhang, J. Novel CD44-targeting and pH/redox-dual-stimuli-responsive core-shell nanoparticles loading triptolide combats breast cancer growth and lung metastasis. J. Nanobiotechnol. 2021, 19, 188. [Google Scholar] [CrossRef]
  61. Park, Y.I.; Kwon, S.H.; Lee, G.; Motoyama, K.; Kim, M.W.; Lin, M.; Niidome, T.; Choi, J.H.; Lee, R. pH-sensitive multi-drug liposomes targeting folate receptor beta for efficient treatment of non-small cell lung cancer. J. Control Release 2021, 330, 1–14. [Google Scholar] [CrossRef] [PubMed]
  62. Pang, J.; Xing, H.; Sun, Y.; Feng, S.; Wang, S. Non-small cell lung cancer combination therapy: Hyaluronic acid modified, epidermal growth factor receptor targeted, pH sensitive lipid-polymer hybrid nanoparticles for the delivery of erlotinib plus bevacizumab. Biomed. Pharmacother. 2020, 125, 109861. [Google Scholar] [CrossRef] [PubMed]
  63. Shih, F.Y.; Jiang, W.P.; Lin, X.; Kuo, S.C.; Huang, G.J.; Hou, Y.C.; Chang, C.S.; Liu, Y.; Chiang, Y.T. A Novel pH-tunable secondary conformation containing mixed micellar system in anticancer treatment. Cancers 2020, 12, 503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Jing, Y.; Xiong, X.; Ming, Y.; Zhao, J.; Guo, X.; Yang, G.; Zhou, S. A Multifunctional micellar nanoplatform with pH-triggered cell penetration and nuclear targeting for effective cancer therapy and inhibition to lung metastasis. Adv. Healthc. Mater. 2018, 7, e1700974. [Google Scholar] [CrossRef]
  65. Shi, M.; Zhao, X.; Zhang, J.; Pan, S.; Yang, C.; Wei, Y.; Hu, H.; Qiao, M.; Chen, D.; Zhao, X. pH-responsive hybrid nanoparticle with enhanced dissociation characteristic for siRNA delivery. Int. J. Nanomed. 2018, 13, 6885–6902. [Google Scholar] [CrossRef] [Green Version]
  66. Zhang, R.; Ru, Y.; Gao, Y.; Li, J.; Mao, S. Layer-by-layer nanoparticles co-loading gemcitabine and platinum (IV) prodrugs for synergistic combination therapy of lung cancer. Drug Des. Devel. Ther. 2017, 11, 2631–2642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Yu, J.; Li, W.; Yu, D. Atrial natriuretic peptide modified oleate adenosine prodrug lipid nanocarriers for the treatment of myocardial infarction: In vitro and in vivo evaluation. Drug Des. Devel. Ther. 2018, 12, 1697–1706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Hong, Y.; Che, S.; Hui, B.; Yang, Y.; Wang, X.; Zhang, X.; Qiang, Y.; Ma, H. Lung cancer therapy using doxorubicin and curcumin combination: Targeted prodrug based, pH sensitive nanomedicine. Biomed. Pharmacother. 2019, 112, 108614. [Google Scholar] [CrossRef]
  69. Xia, F.; Hou, W.; Zhang, C.; Zhi, X.; Cheng, J.; de la Fuente, J.M.; Song, J.; Cui, D. pH-responsive gold nanoclusters-based nanoprobes for lung cancer targeted near-infrared fluorescence imaging and chemo-photodynamic therapy. Acta Biomater. 2018, 68, 308–319. [Google Scholar] [CrossRef]
  70. Chen, H.; Jin, Y.; Wang, J.; Wang, Y.; Jiang, W.; Dai, H.; Pang, S.; Lei, L.; Ji, J.; Wang, B. Design of smart targeted and responsive drug delivery systems with enhanced antibacterial properties. Nanoscale 2018, 10, 20946–20962. [Google Scholar] [CrossRef]
  71. Sharma, A.; Kim, E.J.; Shi, H.; Lee, J.Y.; Chung, B.G.; Kim, J.S. Development of a theranostic prodrug for colon cancer therapy by combining ligand-targeted delivery and enzyme-stimulated activation. Biomaterials 2018, 155, 145–151. [Google Scholar] [CrossRef] [PubMed]
  72. Shahriari, M.; Zahiri, M.; Abnous, K.; Taghdisi, S.M.; Ramezani, M.; Alibolandi, M. Enzyme responsive drug delivery systems in cancer treatment. J. Control Release 2019, 308, 172–189. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, C.; Chen, S.; Wang, Y.; Liu, X.; Hu, F.; Sun, J.; Yuan, H. Lipase-triggered water-responsive “Pandora’s Box” for cancer therapy: Toward induced neighboring effect and enhanced drug penetration. Adv. Mater. 2018, 30, e1706407. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, X.; Gu, M.; Toh, T.B.; Abdullah, N.L.B.; Chow, E.K. Stimuli-responsive nanodiamond-based biosensor for enhanced metastatic tumor site detection. SLAS Technol. 2018, 23, 44–56. [Google Scholar] [CrossRef] [Green Version]
  75. Egeblad, M.; Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat. Rev. Cancer 2002, 2, 61–74. [Google Scholar] [CrossRef]
  76. Fan, Y.; Yuan, S.; Huo, M.; Chaudhuri, A.S.; Zhao, M.; Wu, Z.; Qi, X. Spatial controlled multistage nanocarriers through hybridization of dendrimers and gelatin nanoparticles for deep penetration and therapy into tumor tissue. Nanomedicine 2017, 13, 1399–1410. [Google Scholar] [CrossRef]
  77. Han, M.; Huang-Fu, M.Y.; Guo, W.W.; Guo, N.N.; Chen, J.; Liu, H.N.; Xie, Z.Q.; Lin, M.T.; Wei, Q.C.; Gao, J.Q. MMP-2-sensitive HA end-conjugated poly(amidoamine) dendrimers via click reaction to enhance drug penetration into solid tumor. ACS Appl. Mater. Interfaces 2017, 9, 42459–42470. [Google Scholar] [CrossRef]
  78. Sidi, L.; Luyang, C.; Kai, H.; Ning, C.; Qi, Z.; Kaikai, Y.; Hongzhao, Q.; Chaoyong, L.; Yanli, T.; Xin, H.; et al. Extracellular Delivery: Tumor microenvironment-tailored weakly cell-interacted extracellular delivery platform enables precise antibody release and function. Adv. Funct. Mater. 2019, 29, 1970301. [Google Scholar]
  79. Vaghasiya, K.; Ray, E.; Singh, R.; Jadhav, K.; Sharma, A.; Khan, R.; Katare, O.P.; Verma, R.K. Efficient, enzyme responsive and tumor receptor targeting gelatin nanoparticles decorated with concanavalin-A for site-specific and controlled drug delivery for cancer therapy. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 123, 112027. [Google Scholar] [CrossRef]
  80. Ren, Q.; Liang, Z.; Jiang, X.; Gong, P.; Zhou, L.; Sun, Z.; Xiang, J.; Xu, Z.; Peng, X.; Li, S.; et al. Enzyme and pH dual-responsive hyaluronic acid nanoparticles mediated combination of photodynamic therapy and chemotherapy. Int. J. Biol. Macromol. 2019, 130, 845–852. [Google Scholar] [CrossRef]
  81. Lee, S.Y.; Hong, E.H.; Jeong, J.Y.; Cho, J.; Seo, J.H.; Ko, H.J.; Cho, H.J. Esterase-sensitive cleavable histone deacetylase inhibitor-coupled hyaluronic acid nanoparticles for boosting anticancer activities against lung adenocarcinoma. Biomater. Sci. 2019, 7, 4624–4635. [Google Scholar] [CrossRef] [PubMed]
  82. Gong, Q.; Yang, F.; Hu, J.; Li, T.; Wang, P.; Li, X.; Zhang, X. Rational designed highly sensitive NQO1-activated near-infrared fluorescent probe combined with NQO1 substrates in vivo: An innovative strategy for NQO1-overexpressing cancer theranostics. Eur. J. Med. Chem. 2021, 224, 113707. [Google Scholar] [CrossRef] [PubMed]
  83. Pradubyat, N.; Sakunrangsit, N.; Mutirangura, A.; Ketchart, W. NADPH: Quinone oxidoreductase 1 (NQO1) mediated anti-cancer effects of plumbagin in endocrine resistant MCF7 breast cancer cells. Phytomedicine 2020, 66, 153133. [Google Scholar] [CrossRef] [PubMed]
  84. Park, J.; Jo, S.; Lee, Y.M.; Saravanakumar, G.; Lee, J.; Park, D.; Kim, W.J. Enzyme-triggered disassembly of polymeric micelles by controlled depolymerization via cascade cyclization for anticancer drug delivery. ACS Appl. Mater. Interfaces 2021, 13, 8060–8070. [Google Scholar] [CrossRef]
  85. Borys, N.; Dewhirst, M.W. Drug development of lyso-thermosensitive liposomal doxorubicin: Combining hyperthermia and thermosensitive drug delivery. Adv. Drug Deliv. Rev. 2021, 178, 113985. [Google Scholar] [CrossRef]
  86. Nardecchia, S.; Sánchez-Moreno, P.; Vicente, J.; Marchal, J.A.; Boulaiz, H. Clinical trials of thermosensitive nanomaterials: An overview. Nanomaterials 2019, 9, 191. [Google Scholar] [CrossRef] [Green Version]
  87. Zahednezhad, F.; Zakeri-Milani, P.; Shahbazi Mojarrad, J.; Valizadeh, H. The latest advances of cisplatin liposomal formulations: Essentials for preparation and analysis. Expert Opin. Drug Deliv. 2020, 17, 523–541. [Google Scholar] [CrossRef]
Figure 1. The number of new cases and deaths from cancer worldwide in 2020.
Figure 1. The number of new cases and deaths from cancer worldwide in 2020.
Molecules 27 00948 g001
Figure 2. NIR-II light-responsive semiconducting polymer nanoadjuvant (SPNIIR) is designed and applied for synergetic photothermal immunotherapy, not only to the primary and distant tumors but also the metastasis in the lung [37]. PTT, photothermal therapy; TLR, toll-like receptor; DC, dendritic cell; R848, a TLR agonist; TAA, tumor-associated antigens; ATP, adenosine triphosphate; CRT, calreticulin; HMGB1, high mobility group box 1 protein.
Figure 2. NIR-II light-responsive semiconducting polymer nanoadjuvant (SPNIIR) is designed and applied for synergetic photothermal immunotherapy, not only to the primary and distant tumors but also the metastasis in the lung [37]. PTT, photothermal therapy; TLR, toll-like receptor; DC, dendritic cell; R848, a TLR agonist; TAA, tumor-associated antigens; ATP, adenosine triphosphate; CRT, calreticulin; HMGB1, high mobility group box 1 protein.
Molecules 27 00948 g002
Figure 3. Ultrasound-responsive nanodroplets are designed, fabricated, and capable of inhibiting tumor metastasis in the lung [56]. HIFU, high intensity focused ultrasound; MDR, multi-drug resistance; EMT, epithelial-mesenchymal transition; P-gp, P-glycoprotein; TGF-β1, Transforming Growth Factor-β1; HSPs, heat-shock proteins.
Figure 3. Ultrasound-responsive nanodroplets are designed, fabricated, and capable of inhibiting tumor metastasis in the lung [56]. HIFU, high intensity focused ultrasound; MDR, multi-drug resistance; EMT, epithelial-mesenchymal transition; P-gp, P-glycoprotein; TGF-β1, Transforming Growth Factor-β1; HSPs, heat-shock proteins.
Molecules 27 00948 g003
Figure 4. The concept of pH-triggered cell penetration and nuclear targeting for effective cancer therapy for lung metastatic lung cancer. (1) The schematic diagram of charge reversal; (2) The schematic illustration of targeted transport and enhanced uptake of nanoparticles. (3) Nuclear targeting of positively charged nanoparticles [64].
Figure 4. The concept of pH-triggered cell penetration and nuclear targeting for effective cancer therapy for lung metastatic lung cancer. (1) The schematic diagram of charge reversal; (2) The schematic illustration of targeted transport and enhanced uptake of nanoparticles. (3) Nuclear targeting of positively charged nanoparticles [64].
Molecules 27 00948 g004
Table 1. Brief information of stimuli-responsive nanocarriers that is discussed in this review.
Table 1. Brief information of stimuli-responsive nanocarriers that is discussed in this review.
StimuliSpecific ConditionsNanocarriersDiagnostic /ImagingTherapeuticsReference
LightNear-infrared (NIR) lightGold nanocage@manganese dioxide (AuNC@MnO2) nanoparticlesLee et al., 2019
Titania-coated gold nanobipyramids Chen et al., 2019
Poly(l-lysine)-conjugated chlorin e6 (Ce6) derivative nanoparticle Zhang et al., 2020
Palladium nanosheet (PdNS) Wang et al., 2018
Semiconducting polymer nanoadjuvant (SPNIIIR) Li et al., 2021
CE7Q/CQ/SLi et al., 2020
Short-wavelength and NIR lightO-nitrobenzyl ester modified polymersome with up-conversion nanoparticles Tsai et al., 2021
UltrasoundMechanical effectPerfluoropentane containing nanobubbles Baspinar et al., 2019
Chemical effectPEGylated Co2Fe2O4 nanoflowers (CFP)Fu et al., 2021
Synergistically therapeutic modalityCerasomal perfluorocarbon nanodroplet (D-vPCs-O2)Ma et al., 2020
EXO-DVDMSLiu et al., 2019
Liposome-based nanobubblesLee et al., 2017;
pHpH 5.7–6.9CHEMS-based liposomes; HA-ERL/BEV-LPH nanoparticles; DOX-loaded mixed micelles;
DA-TAT carrier; mPEG-PHis-PSD; U11-DOX/CUR nanoparticles; Cis-aconitic anhydride-modified doxorubicin
Park et al., 2021; Pang et al., 2020; Shih et al., 2020; Jing et al., 2018; Shi et al., 2018; Hong et al., 2019; Xia et al., 2018
EnzymeMMP-2Cur-P-NPs Han et al., 2017
MMP-9MMP-9-sensitive nanocarrier Sidi et al., 2019
MMPsA smart inhalable nanocarrier Vaghasiya et al., 2021
HAaseHPGBCA Ren et al., 2019
EsteraseGold nanorod–curcumin conjugate, HAPBA Zhu et al., 2018; Lee et al., 2019
NQO1QPA-P Park et al., 2021
√ Nanocarriers that designed for diagnostic/therapeutic applications.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lin, X.; Wu, J.; Liu, Y.; Lin, N.; Hu, J.; Zhang, B. Stimuli-Responsive Drug Delivery Systems for the Diagnosis and Therapy of Lung Cancer. Molecules 2022, 27, 948. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27030948

AMA Style

Lin X, Wu J, Liu Y, Lin N, Hu J, Zhang B. Stimuli-Responsive Drug Delivery Systems for the Diagnosis and Therapy of Lung Cancer. Molecules. 2022; 27(3):948. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27030948

Chicago/Turabian Style

Lin, Xu, Jiahe Wu, Yupeng Liu, Nengming Lin, Jian Hu, and Bo Zhang. 2022. "Stimuli-Responsive Drug Delivery Systems for the Diagnosis and Therapy of Lung Cancer" Molecules 27, no. 3: 948. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27030948

Article Metrics

Back to TopTop