Skip to main content

Near-infrared photoresponsive drug delivery nanosystems for cancer photo-chemotherapy

Abstract

Drug delivery systems (DDSs) based on nanomaterials have shown a promise for cancer chemotherapy; however, it remains a great challenge to localize on-demand release of anticancer drugs in tumor tissues to improve therapeutic effects and minimize the side effects. In this regard, photoresponsive DDSs that employ light as an external stimulus can offer a precise spatiotemporal control of drug release at desired sites of interest. Most photoresponsive DDSs are only responsive to ultraviolet-visible light that shows phototoxicity and/or shallow tissue penetration depth, and thereby their applications are greatly restricted. To address these issues, near-infrared (NIR) photoresponsive DDSs have been developed. In this review, the development of NIR photoresponsive DDSs in last several years for cancer photo-chemotherapy are summarized. They can achieve on-demand release of drugs into tumors of living animals through photothermal, photodynamic, and photoconversion mechanisms, affording obviously amplified therapeutic effects in synergy with phototherapy. Finally, the existing challenges and further perspectives on the development of NIR photoresponsive DDSs and their clinical translation are discussed.

Introduction

Chemotherapy is one of the most common treatment strategies in the clinic for cancer, which however often has the issues of low therapeutic efficacy, intrinsic drug resistance and severe side effects [1,2,3]. Although nanomaterial-based drug delivery systems (DDSs) can mitigate the problems [4,5,6,7], ineluctable accumulation of therapeutic drugs in healthy tissues is still prominent. In contrast, stimuli-responsive DDSs with controllable on-demand drug release profiles have been demonstrated to allow highly specific cancer treatment with reduced toxic concerns to normal tissues [8,9,10]. Within the exciting stimuli-responsive DDSs, typical controlled release of drugs mainly relies on some endogenous reactions in the biological systems, such as, cleavages of reactive oxygen species (ROS)-responsive moieties by oxidative stress [11,12,13], charge reversal of polyelectrolytes in acidic microenvironment [14,15,16], proteolysis of peptide substrates by enzymes [17,18,19], cleavages of disulfide linkers under reductive conditions [20,21,22], and reduction of hypoxia-responsive moieties in hypoxic conditions [23, 24]. However, these DDSs still display an insignificant tumor selectivity due to undesirable release in circulation and off-target release caused by endogenous processes in healthy tissues [25,26,27].

External stimuli can provide an improved spatiotemporal control of drug release, and thus have received tremendous attention in the field of DDSs [28,29,30]. In particular, a promising toolbox is the construction of photoresponsive DDSs based on external light that has the advantages of noninvasive nature, ease of production, simplicity of operation, good controllability over both wavelength and intensity, and high spatiotemporal resolution [31,32,33]. Although there are various light-sensitive moieties that are responsive to ultraviolet (UV)–visible light, their applications in DDSs are restricted by the high phototoxicity of UV light and shallow tissue penetration depths for both UV and visible lights (< 1 mm) [34,35,36]. In contrast, near-infrared (NIR) light (650–950 nm) displays negligible phototoxicity and can penetrate more deeply into biological tissues (1–3.5 mm) [37]. In the presence of suitable optical materials as the transducers, NIR light can be converted into heat for photothermal therapy (PTT), ROS for photodynamic therapy (PDT), and UV/visible photons for photoregulation [38,39,40]. Therefore, it is feasible to integrate thermal-, ROS- and short-wavelength light-sensitive components into DDSs to achieve photoresponsive drug release for precise cancer therapy.

Herein, we summarize the recent development of NIR photoresponsive DDSs with on-demand drug release profiles for cancer photo-chemotherapy. They are first classified into (i) photothermal responsive DDSs, (ii) photodynamic responsive DDSs, and (iii) photoconversion responsive DDSs based on three different photoresponsive mechanisms. The constructions, NIR light triggered drug release profiles of these DDSs, and their applications for cancer therapy are then introduced. At last, a brief conclusion and discussion of the existing challenges and further perspectives are given.

Classification of photoresponsive DDSs

NIR photoresponsive DDSs can be classified into three categories: (i) photothermal responsive DDSs, (ii) photodynamic responsive DDSs, and (iii) photoconversion responsive DDSs according to different mechanisms. For cancer therapy, DDSs are typically administered via intravenous injection, and accumulate into tumor tissues though the enhanced permeability and retention (EPR) effect [41]. The representative NIR photoresponsive DDSs used for synergetic treatments of tumors in living animals are listed in Table 1.

Table 1 Summary of representative NIR photoresponsive DDSs for cancer therapy

Photothermal responsive DDSs achieve on-demand release of drugs via nanomaterial-mediated PTT under NIR laser irradiation to generate heat to destroy thermal-responsive materials (Fig. 1) [42]. Such a class of DDSs are generally constructed via integrating thermal-responsive components into nanomaterials containing drugs and photothermal agents [43, 44]. Due to the excellent photothermal conversion efficacy, NIR-absorbing dyes, polydopamine, Prussian blue, carbon, copper sulfide (CuS), bismuth sulfide (Bi2S3), and gold (Au) nanoparticles have been widely utilized to fabricate photothermal responsive DDSs.

Fig. 1
figure 1

Schematic illustration of NIR photoresponsive DDSs

Photodynamic responsive DDSs rely on photosensitizer-mediated PDT to allow NIR triggered drug release (Fig. 1) [45]. PDT utilizes photosensitizers and light irradiation to generate ROS to induce cancer cell death [46]. Meanwhile, the hypoxic condition of tumors will be aggravated due to the continuous consumption of oxygen molecules in PDT process [47]. In view of this, integrations of ROS- and hypoxia-sensitive moieties into DDSs enable the development of ROS- and hypoxia-responsive DDSs, respectively [31]. The common photosensitizers for PDT include tetraphenylchlorin (TPC), indocyanine green (ICG), Rose bengal (RB), chlorin e6 (Ce6), porphyrin, pheophorbide A (PhA), boron dipyrromethene (BODIPY), conjugated polymer (CP), and semiconducting polymer (SP). Among them, ICG has been approved by the US Food and Drug Administration (FDA) for the clinical diagnosis and phototherapy [31].

Photoconversion responsive DDSs are fabricated through integrating UV/visible light-sensitive components (such as 2-nitrobenzyl, spiropyran, coumarin, 7-nitroindoli, donor–acceptor Stenhouse adduct, and azobenzene) into upconverting nanosystems with loadings or conjugations of drugs [32]. Under NIR laser irradiation, upconversion materials convert NIR light into UV or visible light that can destroy these sensitive moieties [48], allowing for on-demand drug release (Fig. 1). The major upconversion materials used in such a type of DDSs are upconverting nanoparticles (UCNPs).

Photothermal responsive DDSs

Due to the existence of thermal-responsive materials that exhibit conspicuous changes of their physical properties with temperature [5], a wide variety of photothermal responsive DDSs have been developed for cancer therapy. Based on the structures and components of nanocarriers, photothermal responsive DDSs mainly consist of photothermal responsive liposomes and photothermal responsive phase change material (PCM)-based hollow nanostructures [49,50,51].

Photothermal responsive liposomes

Thermal responsive liposomes transformed from natural and/or synthetic fatty acids have been widely used to synthesize DDSs, especially for photothermal responsive DDSs [52,53,54]. Xia’s group synthesized a photothermal responsive liposome consisting of an eutectic mixture of natural fatty acids, a chemotherapeutic drug, doxorubicin (DOX) and a NIR dye (IR780) for NIR-triggered drug release [55]. Biocompatible PCM of eutectic mixture with a well-defined melt point at 39 °C was constructed using lauric acid and stearic acid. IR780 served as a photothermal agent to increase the temperature under 808 nm NIR laser irradiation, inducing the melt of liposomes for drug release. The cell death of human lung A549 cancer cells after treatment with DOX/IR780 DDSs upon NIR laser irradiation reached to 90%, which was 1.34-fold higher than that without laser irradiation, verifying the photothermal-controlled chemotherapeutic effect.

Soon afterwards, photothermal responsive liposomes with photocontrolled drug release profiles have been extensively developed and used for synergetic PTT/chemotherapy [56]. For example, a thermosensitive liposome was designed to contain CuS, DOX, and a NIR dye (MBA) encapsulated with stearic acid and lauric acid acting as the PCM [57]. The formed thermosensitive liposome (CuS-DOX-MBA@PCM) showed a relatively low eutectic point close to the body normal temperature. MBA and DOX could be released from this DDS upon 808 nm laser irradiation, while negligible MBA and DOX releases were observed without laser irradiation. As such, CuS-DOX-MBA@PCM afforded a high efficacy in killing A549 cancer cells and inhibiting growth of ehrlich ascites carcinoma (EAC) tumors in living mice after NIR-triggered drug release. In another study, Wang and Zhao et al. developed a thermosensitive liposome bilayers coated mesoporous carbon nanoparticle (MCN) to achieve NIR light triggered on-demand release of DOX for synergistic cancer PTT-chemotherapy [58]. 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(poly(ethylene glycol))] (DSPE-PEG), 1-stearoyl-2-hydroxy-sn-glycero-3-phosphatidylcholine (MSPC), and 1,2-dipalmitoyl-sn-glycero-3-phosphatidylcholine (DPPC) were used to construct surface liposome bilayers with a phase transition temperature (Tm) of 40.7 °C. The release of DOX from DOX/MCN upon 808 nm laser irradiation was sharply increased compared to that without laser irradiation. After treatment with DOX/MCN and NIR laser irradiation, the growth of murine breast 4T1 tumors in living mice was much slower than that of mice after other treatments, indicating superior therapeutic effect of synergistic PTT/chemotherapy.

Photothermal responsive liposomes can also achieve improved penetration of drugs into deep tumors for further enhanced therapeutic efficacy. As demonstrated in a recent study of Zhou’s group [59], a degradable photothermal responsive liposome was constructed via a nanoprecipitation of cyclic arginine-glycine-aspartic acid (cRGD)-conjugated DSPE-PEG, DPPC, cholesterol, a photosensitizer (ICG), and poly(amidoamine) (PAMAM) dendrimers grafting cisplatin prodrug (PAM/Pt) (Fig. 2a). ICG was loaded in the out layer and PAM/Pt was encapsulated inside the formed liposome (PAM/Pt@IcLipo). PAM/Pt@IcLipo showed a long blood circulation time and an excellent targeting capability to tumors via cRGD-mediated targeting pathway. Under NIR laser irradiation at 808 nm, the photothermal effect of ICG raised the local temperature, which resulted in the destruction of PAM/Pt@IcLipo and release of tiny PAM/Pt (~ 8.6 nm). These tiny PAM/Pt could penetrate into deep tumor tissues to exert chemotherapy (Fig. 2b). Because of the synergistic action of PTT/chemotherapy and improved drug penetration depth, the highest therapeutic efficacy (91.1%) in suppressing the growth of 4T1 tumors was achieved through PAM/Pt@IcLipo injection plus 808 nm laser irradiation.

Fig. 2
figure 2

(Reproduced from Ref. [59] with permission from Elsevier, copyright 2020)

a Schematic illustration of formation of degradable liposomal PAM/Pt@IcLipo DDSs. b Schematic illustration of PAM/Pt@IcLipo DDSs for tumor vessel targeting, NIR light-triggered drug release and deep penetration in tumor tissues: (1) extending the circulation time of PAM/Pt in the blood; (2) targeting the tumor blood vessels; (3) being triggered to release PAM/Pt nanoparticles with a size less than 10 nm through NIR laser irradiation

To further increase the therapeutic effect, several photothermal responsive liposomes have been developed for multimodal cancer therapy. As an example, a photothermal responsive liposome with loadings of ICG and DOX and conjugation of gadolinium chelates was synthesized by Fan and coworkers for imaging-guided PTT/PDT/chemotherapy [60]. The PCM core consisted of auric acid and stearic acid at a weight ratio of 4:1 showed a sharp melting point at 39 °C. Under NIR laser irradiation at 808 nm, ICG mediated photothermal effect and increased temperature to melt the PCM core, which enabled on-demand release of DOX. In contrast, the drug release from this liposome was negligible without laser irradiation. In addition, ICG exerted PDT to produce ROS under NIR laser irradiation. As a result, this liposome upon NIR laser irradiation mediated a synergistic PTT/PDT/chemotherapy, greatly inhibiting the growth of human cervical HeLa tumors in living nude mice. Similarly, Cui’s group developed a mitochondria-targeting photothermal responsive liposome via self-assembly of triphenylphosphine (TPP)-coupled DSPE-PEG, DPPC, 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), cholesterol, a photosensitizer (IR-780) and lonidamine (LON) [61]. TPP-binding improved the internalization of the liposome into mitochondria of cancer cells. Laser irradiation at 808 nm triggered IR-780 to elevate the local temperature, which could be utilized for PTT and induced the release of LON from the thermosensitive liposome for chemotherapy. Meanwhile, IR-780 generated ROS in mitochondria for PDT to increase therapeutic efficiency. Such liposome-mediated synergetic PTT/PDT/chemotherapy led to complete eradication of murine Lewis lung LL/2 tumors in nude mice without recurrence within 50 days. In another study, an “all-in-one” photothermal responsive liposome was developed based on a small-molecule dye (DPP-BT) for second NIR fluorescence and photoacoustic dual-modal imaging guided synergetic PTT/PDT/chemotherapy [62]. DPP-BT and DOX were co-encapsulated into organic PCM core of lauric acid and stearic acid (eutectic point at 39 °C) with an out liposome layer of folic acid-modified DSPE-PEG and lecithin. DPP-BT not only served as a dual-modal contrast agent for imaging, but also acted as a therapeutic agent for PTT and PDT. Under NIR laser irradiation at 730 nm, DOX was effectively released from the formed P(DPP-BT/DOX) nanoparticles through NIR-induced PTT of DPP-BT. Upon accumulation of P(DPP-BT/DOX) into tumors of HeLa tumor-bearing mice, the temperature of tumor regions increased under laser irradiation and reached around 54 °C within 4 min, which was enough for PTT effect and to induce the melt of PCM matrix for DOX release. Both in vitro and in vivo results showed that such a liposome resulted in a remarkable antitumor efficacy via synergetic PTT/PDT/chemotherapy triggered by a single NIR laser irradiation.

Different from above photothermal responsive liposomes, Dong’s group recently developed a photothermal-pH-hypoxia multi-responsive liposome for synergistic cancer PTT/PDT/chemotherapy with negligible skin phototoxicity [63]. Such a multi-stimuli responsive liposome consisted of an eutectic PCM mixture of linoleic acid and stearyl alcohol with a melting point at around 43.9 °C, cell-penetrating peptide (CPP, YGRKKRRQRRR) modified DSPE-PEG, DSPE-PEG, a diethylamino group-flanked aza-BODIPY derivative (ENAB) as the pH-responsive photosensitizer, and a hypoxia-specific prodrug, tirapazamine (TPZ). Under acidic pH in tumor microenvironment, ENAB within the formed TENAB nanoparticle turned “off” its charge-transfer state, generating ROS for PDT and heat for PTT upon NIR laser irradiation at 808 nm. Meanwhile, the generated heat melted the PCM coating for release of TPZ (Fig. 3), while negligible release of TPZ was observed without laser irradiation. Subsequently, in the hypoxic condition aggravated by PDT, TPZ was activated into cytotoxic form for chemotherapy. As a result, TENAB nanoparticle afforded a synergistic action of PTT/PDT/chemotherapy upon NIR laser irradiation, completely eradicating HeLa tumors in living nude mice.

Fig. 3
figure 3

(Reproduced from Ref. [63] with permission from Elsevier, copyright 2019)

Schematic illustration of photothermal-pH-hypoxia multi-responsive TENAB nanoparticle for synergistic therapy

Via loading anticancer drugs and genes, photothermal responsive liposomes can be developed for synergistic PTT/chemo-/gene therapy. For example, Nie’s group synthesized a thermo-sensitive co-polymer, poly(2-(2-methoxyethoxy) ethyl methacrylate-co-oligo (ethylene glycol) methacrylate)-co-2-(dimethylamino) ethyl methacrylate-b-poly(d,ʟ-lactide-co-glycolic acid) and used it to construct a liposome with co-encapsulations of DOX and paclitaxel (PTX) and absorption of small interfering RNAs against surviving [64]. The liposome was surface coated with polydopamine, which conferred it with photothermal effect and also protected the burst release of drugs. Under NIR laser irradiation at 808 nm, polydopamine generated heat via PTT effect, resulting in collapse of nanoparticles and subsequent release of drugs within tumors. Such a synergistic action of PTT/chemo-/gene therapy resulted in complete regression of human breast MDA-MB-231 tumors, with a decrease in the chemotherapeutic drug dosage to about 1/20 of conventional dose.

Apart from phase transitions of PCM, the controlled drug release from photothermal responsive liposomes can also be achieved via photothermal-mediated generation of gas. For example, Liang’s group fabricated a thermoresponsive cerasome-forming liposome via self-assembly of a NIR agent, 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindotricarbocyanine iodide (DiR), cerasome-forming lipid, DSPE-PEG, a hyperthermia-responsive ammonium bicarbonate (ABC) and DOX for NIR light-triggered drug release and synergistic cancer PTT/chemotherapy [65]. Due to the good stability of cerasome, both DOX and DiR retained in DDSs at body temperature without obvious releases during blood circulation. Under NIR laser irradiation at 760 nm, DiR-mediated photothermal effect to increase temperature, resulting in the degradation of ABC to generate CO2 bubbles. The generated CO2 bubbles increased the permeability of cerasome bilayers to allow DOX release. As such, a high therapeutic effect in eradicating tumors of living mice was achieved via an intravenous injection of DOX/DiR/ABC cerasome liposome followed by laser irradiation of tumors. In another study, Du and coworkers constructed a dual aptamer-modified and gold nanoshell coated thermosensitive liposome with encapsulation of ABC in the internal aqueous phase and an anti-cancer drug, docetaxel (DTX) in the phospholipid layer [66]. Similarly, gold nanoshell-mediated photothermal effect under 808 nm laser irradiation induced the generation of CO2 bubbles, which increased the permeability of bilayers for drug release. In addition to PTT and chemotherapy, the AS1411 aptamer that could dispute the functions of nucleolin via binding to nucleolin in nuclear membrane was used as a biotherapeutic agent. As a result, this DDS showed a high therapeutic efficacy in suppressing the growth of murine sarcoma S180 tumors via synergistic PTT/chemotherapy/biological therapy.

Photothermal responsive PCM-based hollow nanostructures

The combinations of hollow nanostructures and PCM can be utilized to construct photothermal responsive hollow DDSs [67]. The photothermal effect of optical materials in nanocarriers elevates local temperatures, which induces the phase transition of PCM from a solid to a liquid, leading to precise release of drugs from hollow cavities.

Mesoporous silica nanoparticles (MSNs) have been used to develop photothermal responsive DDSs for cancer therapy [68,69,70]. For example, Kim and Yong et al. constructed a mesoporous silica coated silver–gold hollow nanoplatform to precisely regulate the release of 5-fluorouracil (anticancer drug) for prostate cancer therapy and photothermal therapy [70]. The mesopores were capped with a thermosensitive PCM (lauric acid), which allowed for remote, precise, and spatiotemporal control of drug release via silver–gold nanoshell-mediated photothermal heating under NIR laser irradiation at 808 nm. Such a nanoplatform thus showed a synergistic effect in killing cancer cells. Since MSNs do not have photothermal effect, other photothermal agents are required to construct these MSN-based photothermal responsive DDSs.

Qian’s group developed a photothermal responsive DDS based on rod-based urchin-like Bi2S3 hollow nanoparticles (termed as U-BSHM) to allow precise release of chemotherapeutic agents for synergistic PTT/chemotherapy [71]. A sacrificial template engaged polyol route was used to synthesize U-BSHM as the photothermal agent with a photothermal conversion efficiency of 26.8%. U-BSHM was loaded with DOX and encapsulated with PCM of 1-tetradecanol (the melting point at around 38 °C) as the “gatekeeper” to form the photothermal responsive DDS (termed as PD@BS) (Fig. 4a). The release of DOX was rapidly increased upon NIR laser irradiation at 808 nm compared to that without laser irradiation (Fig. 4b). The cell viability of MDA-MB-231 cancer cells gradually decreased with the prolonged NIR laser irradiation time and elevated concentrations of DDSs, which was due to the synergetic action of PTT and released DOX induced by NIR laser irradiation. The temperature of MDA-MB-231 tumor regions of living mice after intratumoral injection of PD@BS gradually increased to a plateau of around 49 °C under NIR laser irradiation, which was enough to induce the release of DOX and ablate tumors. As a result, the therapeutic effect of PD@BS was obviously improved with NIR laser irradiation (Fig. 4c).

Fig. 4
figure 4

(Reproduced from Ref. [71] with permission from Elsevier, copyright 2020)

a Schematic illustration of thermosensitive urchin-like Bi2S3 hollow nanoparticles as photothermal responsive DDSs for photothermal-chemotherapy. b Cumulative release of DOX from PD@BS induced by cyclic “turn-on” and “turn-off” of NIR light at different times. c The growth curves of MDA-MB-231 tumors in different groups during a period of 12 days

Several other photothermal agents have also been used to induce the phase changes of PCM to achieve on-demand releases of chemotherapeutic drugs and synergistic effects of PTT/chemotherapy. Cai and co-workers constructed a photothermal responsive DDS based on hollow magnetic Prussian blue nanoparticles for NIR light-triggered PTT/chemotherapy [72]. Such a DDS consisted of a Prussian blue coated hollow iron oxide magnetic nanoparticle as the carrier and photothermal agent, DOX as the chemotherapeutic drug, and a biocompatible PCM of 1-pentadecanol as the “drug-janitor” for controlled-release in response to increased temperature (> 42 °C). In such a system, the release of DOX from DDS was significantly increased upon 808 nm laser irradiation due to Prussian blue-mediated photothermal effect. After treatments, this system exerted synergistic PTT/chemotherapy and afforded an enhanced therapeutic efficacy in inhibiting the growth of human hepatoma HepG2 tumors compared to sole chemotherapy and PTT. Similarly, Guo’s group developed a mesoporous carbon nanoparticle (MCN)-based DDS filled with DOX and 1-tetradecano as the PCM in hollow cavities for NIR light-triggered release of DOX [73]. The cumulative DOX release was significantly increased under 808 nm laser irradiation via MCN-mediated photothermal effect. A much higher intracellular DOX level was observed in MCF-7/ADR cells after treatment with MCN-based DDS plus laser irradiation relative to those without laser irradiation. The apoptosis evaluation showed that the highest percentage of total apoptosis (49.7%) was caused by MCN-based DDS treatment plus NIR laser irradiation, which was 2.48-fold higher relative to that of free DOX treatment alone. This suggested an enhanced therapeutic effect induced by the NIR light-triggered PTT and drug release.

In addition to PTT/chemotherapy, photothermal responsive PCM-based hollow nanostructures have also been developed for synergistic PTT/PDT/chemotherapy. For example, hollow mesoporous ZrO2-coated Nd3+-doped UCNPs were used as the photothermal nanocarriers to load DOX, Ce6, and 1-tetradecanol (PCM), forming a DDS (termed as UCNPs@ZrO2-Ce6/DOX/PCM) [74]. Due to the UCNP-mediated PTT, the release efficiency upon 808 nm NIR laser irradiation reached 44.12%. The inhibitory effect of HeLa cancer cells after treatment of UCNPs@ZrO2-Ce6/DOX/PCM and laser irradiation reached nearly 100%, which was 3.2-, 2.3-, and 1.6-fold higher than that in sole PTT, free DOX and PTT + PDT group, respectively. An excellent in vivo synergistic antitumor efficacy was also verified in murine cervical U14 tumor-bearing mice after treatment with this nanosystem plus NIR laser irradiation. Similarly, Xu’s group constructed another photothermal responsive DDS (termed as HPDC) with a high photothermal conversion efficiency of 44.13% for synergistic PTT/PDT/chemotherapy [75]. Such a HPDC consisted of four key components: hollow mesoporous CuS nanoparticle as both the photothermal agent and nanocarrier, surface coated 1-tetradecanol as a PCM, DOX as a chemotherapy drug and Ce6 as a photodynamic photosensitizer. NIR laser irradiation at 808 nm increased local temperature via CuS-mediated PTT, which obviously increased the release of both DOX and Ce6. Their cumulative release reached 72.78 and 74.4% after six cycles of NIR laser irradiation, respectively, while only approximately 5% DOX or Ce6 in total was released without laser irradiation. Via HPDC-mediated synergistic PTT/PDT/chemotherapy, the growth of 4T1 tumors in living mice was almost completely inhibited, while other treatments failed to effectively suppress tumor growth.

This section summarizes the recent constructions of photothermal responsive liposomes and PCM-based hollow nanostructures for NIR photoactivated drug release through photothermal effect mediated phase transition of thermal-sensitive materials. The liposomes have intrinsic advantages of good biocompatibility and biodegradability, low toxicity and immunogenicity, tunable physicochemical and biophysical properties and unique capability of loading both lipophilic and hydrophilic drugs [76]. However, liposomes often exhibit low stability because the phospholipid can be easily oxidized and hydrolyzed. Moreover, the use of organic solvent or high temperature during the liposome fabrication process may affect the bioactivity of drug molecules [77]. The hollow nanostructure-based DDSs exhibit various advantages including excellent chemical stability, high drug loading capability, and abundant surface chemical groups for further functionalization [78,79,80]. However, they generally have the drawbacks of poor biodegradability and long-term toxic concerns in living bodies [81]. These disadvantages of photothermal responsive DDSs should be considered to facilitate their translation for clinical medicine.

Photodynamic responsive DDSs

In the typical process of PDT, photosensitizers not only convert light energy into ROS, but also deplete oxygen to increase the tumor hypoxia to a certain extent [47]. Hence, it is feasible to construct photodynamic responsive DDSs via integrating ROS- or hypoxia-cleavable moieties into DDSs.

ROS-responsive DDSs

ROS-responsive DDSs have been applied to selectively release various drugs into target tissues, which can be achieved via photo-controlled cleavage of ROS-responsive linkers [12, 82, 83]. For instance, Schnermann et al. reported the utilization of NIR light to cleave antibody–drug conjugates containing a cyanine photocage [84]. Such conjugates consisted of a heptamethine cyanine fluorophore serving as the photocaging component, combretastatin A4 (CA4) acting as the potent inhibitor of microtubule polymerization and a human epidermal growth factor receptor (EGFR)-binding monoclonal antibody. This linker strategy utilized carbamate functional groups as the antibody attachment points, which ensured the release of CA4 drugs from antibodies triggered by ROS generation form fluorophore under NIR laser irradiation at 690 nm. Moreover, the fluorescence signal of this system provided a useful marker to verify the accumulation of conjugates, while the loss of fluorescence signal after excitation by NIR light indicated drug release.

Another representative example of ROS-responsive DDS was demonstrated by Liu’ s group [85]. In this system, DOX was covalently conjugated to an organic conjugated polyelectrolyte (CPE) through a ROS-cleavable dithioketal linker. PEG chains and cRGD were also conjugated to the backbone of CPE to improve its solubility and target specificity to cancer cells, respectively. CPE was utilized as the photosensitizer to generate ROS upon white light irradiation, not only exerting PDT, but also triggering the cleavage of dithioketal linkers for on-demand DOX release, which permitted a synergistic cancer therapy with an enhanced therapeutic effect. The ROS-cleavable dithioketal linker was also utilized by Xie’s group to construct a photoactivatable biomimetic dimeric prodrug DDS with red blood cell (RBC) membrane camouflage for synergistic cancer PDT/chemotherapy (Fig. 5a, b) [86]. It has been demonstrated that cell membrane camouflaged nanomaterials exhibit a prolonged blood circulation, and tumor targeting ability, thus enabling an enhanced tumor accumulation and improved therapeutic outcomes [87,88,89,90]. After the effective cellular internalization of biomimetic prodrug DDS by cancer cells, the photosensitizer, tetraphenylchlorin (TPC) embedded in the inner core could generate ROS upon NIR light irradiation at 638 nm, leading to synergetic therapy through photodynamic disruption of the cellular endosomes, and meanwhile, cleavage of the dithioketal linkers to amplify the release of PTX (Fig. 5c). The therapeutic effect was significantly increased via such a biomimetic prodrug DDS-mediated synergistic PDT/chemotherapy.

Fig. 5
figure 5

(Reproduced from Ref. [86] with permission from American Chemical Society, copyright 2018)

a Schematic illustration of synthesis and mechanism of RBC membrane-coated dimeric prodrug DDSs with NIR light triggered on-demand drug release for synergistic PDT/chemotherapy. b Schematic illustration of mechanism of 1O2 induced activation of PTX2-dithioketal prodrug. c The degradation of PTX2-dithioketal in DDSs over time upon irradiated by 638 nm laser lamp (100 mW/cm2) tested by high-performance liquid chromatography (HPLC)

In addition, a singlet oxygen (1O2) sensitive bis-(alkylthio)alkene (BATA) linker has been used to construct ROS-responsive DDSs to achieve on-demand chemotherapy upon irradiation of light [91]. An interesting example was demonstrated by Liu and coworkers [92]. In their system, photosensitizer (Ce6) doped mesoporous silica nanorods (CMSNRs) were coated with bovine serum albumin (BSA) via the BATA linkers and then modified with PEG, forming a drug delivery nanocarrier that was able to load small drug molecules such as DOX, or larger cargos such as cis-Pt(IV) prodrug conjugated third generation dendrimer (G3-Pt). Upon NIR laser irradiation at 660 nm with a low power density (down to 5–50 mW/cm2), the doped Ce6 generated 1O2 to effectively cleave the BATA linkers, which induced detachments of BSA-PEG from the surface of nanocarriers and thus triggered a release of loaded DOX or G3-Pt. As a result, both the in vitro and in vivo therapeutic effects were enhanced through the on-demand release of therapeutic cargos under NIR light irradiation.

Diselenide bond (Se–Se) that can be cleaved by ROS has shown to be another good candidate as light-sensitive linker for the construction of ROS-responsive DDSs [93]. Xu and colleagues developed a light‐responsive micelle with co-encapsulation of photosensitizer (porphyrin) and DOX based on the diselenide bond [94]. The 1O2 generated from porphyrin under light (600–780 nm) irradiation facilitated the cleavage of diselenide bonds, resulting in disruption of micelles and on-demand release of DOX. Similarly, Yang’s group developed a light‐responsive poly(methacrylic acid) (PMAA)-based nanogel system based on diselenide-cross-linkers [95]. ICG was loaded into nanogels to generate 1O2 upon NIR laser irradiation at 785 nm, which induced disassembly of diselenide‐crosslinked nanogels and thus achieved on-demand release of loaded DOX.

In another study, a ROS-responsive DDS was constructed via self-assembly of PEG-stearamine (C18) conjugate with the ROS-cleavable thioketal linkers and co-loaded DOX and a photosensitizer pheophorbide A (PhA) for enhanced cancer PDT/chemotherapy [96]. Upon NIR laser irradiation at 670 nm, the formed DDS (termed as PTS-DP) generated 1O2 due to the photodynamic effect of PhA, and the generated 1O2 cleaved thioketal linkers in polymer conjugates, leading to rapid dissociation of DDSs for DOX release. The gradual elevation of local ROS levels mediated by PDT synergized with NIR light-triggered release of DOX, showing an enhanced efficacy to treat murine colorectal CT26 tumors.

In addition, the ROS sensitivity of aminoacrylate groups was used to develop a ROS-responsive supramolecular DDS with an optimized loading ratio of the photosensitizers and prodrugs for cancer therapy [97]. A diblock copolymer was synthesized to consist of PEG, poly-ʟ-glutamic acid (PGA) and β-cyclodextrin (β-CD). Adamantane-conjugated aza-BODIPY (Ada-BODIPY) and PTX (Ada-PTX) were used as the photosensitizer and prodrug guest molecules, respectively. Via a strong interaction between β-CD and the adamantane units, the supramolecular DDS was formed. Under 660 nm laser irradiation, the photosensitizers generated ROS for PDT, and meanwhile to cleave the ROS-sensitive aminoacrylate groups in Ada-PTX to allow PTX release. Hence, the supramolecular DDS enabled a synergistic action of PDT/chemotherapy for inhibition of HeLa tumor growth.

Hypoxia-responsive DDSs

In view of aggravated hypoxia in tumor microenvironment after PDT process, hypoxia-sensitive moieties have been used to construct hypoxia-responsive DDSs for synergistic PDT/chemotherapy. For example, Gu’s group constructed a novel DDS to realize NIR light triggered ROS generation and subsequent hypoxia-activated drug release [98]. This DDS was constructed via using a ROS-generating and hypoxia-responsive 2-nitroimidazole-grafted conjugated polymer (CP-NI) to encapsulate DOX through a double-emulsion-based solvent evaporation/extraction method (Fig. 6a). Upon 635 nm laser irradiation, the formed DDS (termed as DOX/CP-NI) generated 1O2 to permit PDT. Meanwhile, the dissolved oxygen was rapidly consumed due to the generation of 1O2, leading to a local hypoxic microenvironment. As a result, hydrophobic 2-nitroimidazole groups in the CP-NI were converted to hydrophilic 2-aminoimidazoles under the hypoxic condition, leading to the dissociation of DDS, and subsequent release of DOX. As expected, about 60% of DOX was released from the DOX/CP-NI at pH 7.4 after laser irradiation, but the released amount was very low without laser irradiation. Such a light-activated and hypoxia-responsive nanocarrier exerted synergistic PDT/chemotherapy, resulting in complete inhibition of the growth of HeLa tumors (Fig. 6b). In another similar study, 2-nitroimidazole derivative conjugated PEG amphoteric polymer-based DDSs with loadings of Ce6, gene probe, and a hypoxia-activated prodrug TPZ were fabricated through self-assembly for synergistic PDT/chemotherapy [99]. Upon NIR laser irradiation at 670 nm, Ce6-mediated PDT induced a hypoxic condition, which resulted in disassembly of DDSs for drug release and also activated the antitumor effect of TPZ for improved therapeutic effect. In vivo studies verified the greatly improved anti-cancer activity of such a DDS-mediated synergistic PDT/chemotherapy in inhibiting human breast MCF-7 tumor growth compared to sole PDT.

Fig. 6
figure 6

(Reproduced from Ref. [98] with permission from Wiley-VCH, copyright 2016)

a Schematic of the formation and mechanism of light-activated hypoxia-responsive DOX/CP-NI. b Schematic of DOX/CP-NI mediated generation of ROS and induction of a local hypoxic environment for hypoxia-responsive release of DOX into cell nucleus for synergistic anticancer efficacy

In another study, Pu’s group constructed a semiconducting polymer nanoparticles (SPNs)-based DDS for hypoxia-activated photodynamic cancer therapy [100]. The SPN-based DDS was formed via a self-assembly of the amphiphilic semiconducting brush polymers that comprised a photodynamic SP grafted with PEG and conjugated with a chemotherapeutic drug (bromoisophosphoramide mustard intermediate, IPM-Br) through a hypoxia-cleavable linker. Upon NIR laser irradiation at 808 nm, the SPN core generated 1O2 for PDT, and meanwhile, depleted oxygen to further promote tumor hypoxia. The hypoxia-cleavable linker was then cleaved by hypoxia and thus on-demand release of IPM-Br was achieved. After systemic administration, this DDS effectively accumulated at the tumor of living mice due to their small size and stealthy PEG grafting. Under NIR laser irradiation, the SPN-based DDS exerted synergistic PDT/chemotherapy and effectively inhibited the growth of 4T1 tumors.

The recent development of photodynamic responsive DDSs for PDT-synergistic chemotherapy has been introduced in this section. Although controllable drug releases from nanocarriers have been demonstrated, photodynamic responsive DDSs still have some unsatisfied drawbacks that need to be overcome. These DDSs can also respond to endogenous ROS or hypoxia in the biological systems, which may result in off-target drug release and thus cause side effects. In addition, the majority of photosensitizers in these systems are small molecular organic dyes that display poor optical stability upon light exposure [101]. Therefore, development of photosensitizers with good photostability will be a key step to realize wide applications of photodynamic responsive DDSs.

Photoconversion responsive DDSs

Photosensitive moieties, such as 2-nitrobenzyl, coumarin, 7-nitroindoli, and azobenzene have been widely used to construct DDSs, whereas the majority of these moieties typically respond to short-wavelength deep-blue or UV light [102,103,104,105], which greatly limits in vivo applications of these DDSs. To address this issue, upconverting materials, such as UCNPs can be integrated into DDSs to convert NIR light into short-wavelength light sources [106].

Photoconversion-induced drug release from DDSs can be achieved via photolysis strategy. Li and colleagues encapsulated an 7-amino-coumarin derivative caged chlorambucil prodrug (termed as ACCh) into yolk–shell structured nanocarriers with UCNPs as moveable core and mesoporous silica as shell [107]. Under NIR laser irradiation at 980 nm, the UCNP core upconverted NIR light into UV emission (365 nm), effectively triggering the photolysis of ACCh prodrug to release chlorambucil, which facilitated the diffusion of activated drugs from nanocarriers through pores in the silica shell. This photoconversion responsive DDS afforded a high therapeutic efficacy in inhibiting the growth of S180 tumors and increasing survival of tumor-bearing living mice. In another study, Han’s group reported an organic-chromophore-based triplet–triplet annihilation upconversion (TTA-UC) strategy for NIR light-triggered cancer chemotherapy [108]. The developed DDSs consisted of MSNs loaded with TTA-UC molecules as the core, and an amphiphilic polymer encapsulated coumarin-chlorambucil prodrugs as the shell. Upon laser irradiation at 650 nm, TTA-UC molecules upconverted far-red light into blue emission to activate the prodrugs via photolysis. More than 48% activation of the prodrugs was achieved within 30 min, and a maximum photorelease of around 82% of prodrugs after 60 min of laser irradiation. As a result, the TTA-UC-based DDS mediated drug release allowed effective inhibition of 4T1 tumor growth in living mice.

UCNP-mediated NIR-to-UV conversion can also be utilized for photoreduction of platinum(IV) (Pt(IV)) prodrugs to form toxic Pt(II) [109]. A representative example was presented by Lin and coworkers, in which, photoconversion responsive DDSs were constructed via conjugating UCNPs with Pt(IV) prodrugs and a monolayer of PEG [110]. Under 980 nm NIR laser irradiation, UCNPs excited UV light to activate Pt(IV) prodrug into Pt(II) drug. More interestingly, the Pt(IV) prodrug-conjugated UCNPs under 980 nm laser irradiation showed a high efficiency in inhibiting the growth of murine hepatoma H22 tumors than that under direct UV light irradiation, although UV light could also effectively activate the Pt(IV) prodrugs. Similarly, Xing’s group developed a photoconversion responsive DDSs via conjugating UCNPs with Pt(IV) prodrugs and a short peptide probe [111]. Upon NIR light irradiation at 980 nm, the converted emission from UCNPs locally activated the Pt(IV) prodrug and thus efficiently induced a potent antitumor efficacy.

In a recent study, a Pt(IV) prodrug based charge-convertible DDS with the loading of NaYF4:Yb,Tm UCNPs and surface coating of a layer anionic PEG-poly(allylamine hydrochloride)-dimethyl-maleic acid polymer was developed for cancer therapy [112]. After response to the mild acidic stimulus (pH ~ 6.5) of tumor extracellular microenvironment, the anionic polymer underwent a charge-shifting to form a cationic polymer, leading to electrostatic repulsion and releases of positive Pt(IV)-UCNPs that could effectively bind to the negative cell membrane for cell internalization. Under 980 nm laser irradiation, UCNPs emitted UV light to efficiently activate the Pt(IV) prodrugs into highly cytotoxic Pt(II). Such a NIR photoresponsive smart DDS thus displayed a markedly enhanced tumor ablation efficacy in murine cervical U14 tumor-bearing Kunming mice.

In addition to photolysis and photoreduction, photoswitchable strategy has been used to develop photoconversion responsive DDSs. Shi’s group developed a photoconversion responsive DDS based on photoswitchable strategy to achieve NIR light-triggered release of anticancer drugs [113]. Mesoporous silica coated UCNPs were inside installed with “photomechanical” azobenzene (azo) groups acting as the “stirrer” and surface conjugated with a transactivator of transcription peptide to enable an enhanced cellular internalization into cancer cells, following by loading of DOX through formation of strong hydrogen bonds and charge interactions with the surface silanol groups. Upon NIR laser irradiation at 980 nm, UCNPs emitted UV and visible light, which triggered the switch of azo molecules between trans and cis isomer. Such a reversible photoisomerization created a continuous rotation-inversion movement, leading to the release of DOX from such a DDS. As a result, the DOX release percentage reached a maximum of 80% under intermittent NIR laser irradiation, while less than 5% of DOX was released without laser irradiation. This photoconversion responsive DDS was verified to show a high efficacy in killing cancer cells.

Another azo-based DDS with controllable intracellular drug release upon NIR photoirradiation was constructed by Ju’s group for cancer therapy [114]. Such a DDS was constructed by assembling azo-functionalized DNA strands on poly(acrylic acid) (PAA)-modified UCNPs, followed by loading of DOX into the DNA helix (Fig. 7a). Under NIR laser irradiation at 980 nm, UCNPs emitted both UV and visible lights to fuel continuous photoisomerization of azo, which induced controllable DOX release due to the hybridization and dehybridization of cyclic DNA (Fig. 7b). The maximum DOX release amount reached 86.7% after 30 min of NIR laser irradiation. Through assembling a nuclear localizable HIV-1 trans-activator of transcription (TAT) peptide and hyaluronic acid (HA) onto the surface of DDS, targeting release of DOX inside cancer cell nucleus was achieved upon NIR laser irradiation (Fig. 7c). Monitoring of tumor growths after different treatments showed that this DDS (UCNPs/DOX-TAT-HA)-mediated therapy had a significantly improved chemotherapeutic outcome for HepG2 tumors in living mice.

Fig. 7
figure 7

(Reproduced from Ref. [114] with permission from Wiley-VCH, copyright 2019)

a Schematic illustration of assembly of UCNPs-LAAzoBCAzo/DOX. The enlarged section delineates the continuous photoisomerization of azo and cyclic hybridization and dehybridization of LAAzo and LB (DNA strands LAAzo, LCAzo with 3 azo moieties per DNA strand). b Synthesis of UCNPs/DOX-TAT-HA. c Illustration of HA-mediated endocytosis, TAT-mediated nuclear targeting and NIR-triggered drug release in living cells

A NIR photoswitchable cage mimicking DDS was developed through anchoring a photochromic spiropyran onto mesoporous silica coated UCNPs with loading of curcumin for cancer therapy [115]. The hydrophobic spiropyran formed a compact layer on silica shells to conceal curcumin inside the channels of nanocarrier without unexpected drug release. Upon NIR irradiation at 980 nm, UCNPs effectively converted NIR light to UV emission light that induced the conformational transformation of spiropyran molecules from hydrophobic to hydrophilic state. Such a NIR photoirradiation caused hydrophobicity-hydrophilicity switch achieved on-demand release of curcumin with good bioactivity for cancer chemotherapy. Moreover, the UV/visible light produced by UCNPs activated curcumin to initiate the generation of ROS, further improving the therapeutic efficiency. Such a photoconversion responsive DDS was demonstrated to exhibit a significantly enhanced antitumor efficiency in 4T1 tumor-bearing mice.

In another study, a multifunctional UCNP-based micelle with NIR photocontrolled drug release was developed for combinational cancer PDT and chemotherapy [116]. The micelle was formed via modifying UCNP with a photosensitive amphiphilic copolymers poly(4,5-dimethoxy-2-nitrobenzyl methacrylate)-PEG (PNBMA-PEG) and a photosensitizer (RB), followed by loading with a hydrophobic anticancer drug (a histone deacetylase inhibitor). Under NIR laser irradiation at 980 nm, UCNPs emitted UV, 540 and 650 nm luminescence bands. The UV light activated photocleavable PNBMA segments to induce the hydrophobic-to-hydrophilic transition of micelle cores, triggering a rapid drug release for NIR-controlled chemotherapy. The emitted 540 nm light could activate RB molecules to produce 1O2 for NIR-induced PDT. Further surface modification with a neuroendocrine tumor-targeting ligand allowed high tumor accumulation of micelles in a human medullary thyroid TT tumor model, thus achieving the highest antitumor efficacy.

In this section, we have introduced the utilization of UCNPs to construct NIR photoconversion responsive DDSs for cancer therapy. NIR photoactivated on-demand release of drugs can be realized via the use of UCNPs with unique intrinsic optical properties and photocleavable, photoswitchable, or photoreductive moieties. UCNPs often show excellent stability against photochemical degradation [117], and thus these photoconversion responsive DDSs have a great potential for cancer therapy. However, the relatively low quantum yields of UCNPs and in vivo safety concerns of inorganic rare elements in UCNPs need to be overcome for their further clinical applications [41].

Conclusion and perspectives

Nanomaterial-based DDSs potentially improve the therapeutic effects and reduce side effects of chemotherapeutic drugs. However, less than 5% dosage of DDSs after systemic administration can reach tumor tissues [5], which often limits their therapeutic outcomes. The development of advanced DDSs with on-demand drug release profiles are highly desired. In this regard, NIR photoresponsive DDSs have received tremendous attention because of their unique advantages. Herein, we have summarized the recent development of NIR photoresponsive DDSs for cancer photo-chemotherapy. Based on three different photoresponsive mechanisms, these DDSs are constructed to integrate optical materials, anti-cancer drugs and responsive moieties. Upon NIR laser irradiation, optical materials convert NIR light into heat, ROS or short-wavelength light, which not only enables PTT and/or PDT, but also results in the destruction of responsive moieties for on-demand release of drugs in tumor tissues. As a result, these DDSs often afford improved therapeutic effects to reject tumors.

Although the promising achievements of NIR photoresponsive DDSs, there are several crucial issues that greatly restrict their clinical translation. First, the tissue penetration depth of NIR light has been improved compared to UV/visible light, which however is still less than 1 cm. Thus, the uses of photoresponsive DDSs are only suitable for superficial tumors such as melanoma. Since the second NIR light (NIR-II, 1000–1700 nm) shows a further increased tissue penetration depth of around 3–5 cm, development of NIR-II photoresponsive DDSs can overcome this dilemma [118]. Alternatively, the combination of light delivery technologies can be adopted to achieve a deep delivery of light sources in biological tissues [119]. Second, photon conversion efficiencies of optical components in NIR photoresponsive DDSs still need to be enhanced to improve the therapeutic efficacies upon laser irradiation at a low power density. High laser power density may induce photodamage to skill following the guidelines of American National Standard Institute (ANSI) [120]. Some strategies such as molecule acceptor doping, light-harvesting unit integration, and incorporation of different optical materials have shown great potential in amplifying the photon conversion efficiencies [37]. Third, the in vivo long-term biocompatibility and biodegradability of NIR photoresponsive DDSs is questionable and the products of photoirradiation may cause some safety concerns. To address this issue, it is necessary to systemically evaluate their biosafety in living subjects. Alternatively, efforts can be made to enhance their biodegradability and/or reduce their dimensions for a rapid clearance via renal and/or hepatic excretions [121,122,123,124]. Fourth, the variety of photoresponsive components is very limited, and their manufactures require long processing time and high cost of production, which greatly hinders the large-scale manufacturing of photoresponsive DDSs for clinical and translational applications. Exploration of facile inexpensive manufacturing methodology and/or development of new photoresponsive components is desirable to achieve their clinical translation. At last, it is often difficult to identify the tumor regions and the optimal therapeutic windows for NIR laser irradiation. Additional imaging agents can be integrated into photoresponsive DDSs to realize imaging-guided cancer photo-chemotherapy.

In addition to anticancer drugs, NIR photoresponsive DDSs can be used for on-demand release of other agents to achieve different therapeutic purposes. For example, Chang’s group reported the use of Prussian blue nanocubes to mediate photothermic activation of a tumor suppressor gene (p53) for PTT-synergistic gene therapy of tumors [125]. Via integrating photoresponsive components with immunotherapeutic molecules into a single nanoplatform, it is probable to achieve photoactivation of cancer immunotherapy using NIR photoresponsive DDSs [45]. Furthermore, the feasibility of NIR photoresponsive DDSs for the treatments of diseases other than cancer such as neurodegenerative, cardiovascular, infectious, and autoimmune diseases can be explored. Overall, with the progression of extensive research that will enable a better understanding of the current state of art, NIR photoresponsive DDSs should be available for clinical applications in the near future.

Availability of data and materials

Not applicable.

Abbreviations

DDSs:

Drug delivery systems

NIR:

Near-infrared

ROS:

Reactive oxygen species

UV:

Ultraviolet

PTT:

Photothermal therapy

PDT:

Photodynamic therapy

PCM:

Phase change material

CuS:

Copper sulfide

Bi2S3 :

Bismuth sulfide

TPC:

Tetraphenylchlorin

ICG:

Indocyanine green

RB:

Rose bengal

Ce6:

Chlorin e6

PhA:

Pheophorbide A

BODIPY:

Boron dipyrromethene

CP:

Conjugated polymer

SP:

Semiconducting polymer

UCNPs:

Upconverting nanoparticles

DOX:

Doxorubicin

MCN:

Mesoporous carbon nanoparticle

DSPE-PEG:

1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(poly(ethylene glycol))]

MSPC:

1-Stearoyl-2-hydroxy-sn-glycero-3-phosphatidylcholine

DPPC:

1,2-Dipalmitoyl-sn-glycero-3-phosphatidylcholine

DiR:

1,1′-Dioctadecyl-3,3,3′,3′-tetramethylindotricarbocyanine iodide

ABC:

Ammonium bicarbonate

DTX:

Docetaxel

CPP:

Cell-penetrating peptide

cRGD:

Cyclic arginine-glycine-aspartic acid

PAMAM:

Poly(amidoamine)

TPP:

Triphenylphosphine

DSPC:

1,2-Distearoyl-sn-glycero-3-phosphocholine

LON:

Lonidamine

PTX:

Paclitaxel

TPZ:

Tirapazamine

MSNs:

Mesoporous silica nanoparticles

CA4:

Combretastatin A4

EGFR:

Epidermal growth factor receptor

RBC:

Red blood cell

TPC:

Tetraphenylchlorin

BATA:

Bis-(alkylthio)alkene

BSA:

Bovine serum albumin

PMAA:

Poly(methacrylic acid)

PGA:

Poly-ʟ-glutamic acid

β-CD:

β-Cyclodextrin

HPLC:

High-performance liquid chromatography

SPNs:

Semiconducting polymer nanoparticles

Pt(IV):

Platinum (IV)

PAA:

Poly(acrylic acid)

TAT:

Trans-activator of transcription

HA:

Hyaluronic acid

ANSI:

American National Standard Institute

References

  1. Markman JL, Rekechenetskiy A, Holler E, Ljubimova JY. Nanomedicine therapeutic approaches to overcome cancer drug resistance. Adv Drug Deliv Rev. 2013;65:1866–79.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Browning RJ, Reardon PJT, Parhizkar M, Pedley RB, Edirisinghe M, Knowles JC, Stride E. Drug delivery strategies for platinum-based chemotherapy. ACS Nano. 2017;11:8560–78.

    CAS  PubMed  Google Scholar 

  3. Kim J, Yung BC, Kim WJ, Chen X. Combination of nitric oxide and drug delivery systems: tools for overcoming drug resistance in chemotherapy. J Control Release. 2017;263:223–30.

    CAS  PubMed  Google Scholar 

  4. Hu Q, Sun W, Wang C, Gu Z. Recent advances of cocktail chemotherapy by combination drug delivery systems. Adv Drug Deliv Rev. 2016;98:19–34.

    CAS  PubMed  Google Scholar 

  5. Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nat Mater. 2013;12:991–1003.

    CAS  PubMed  Google Scholar 

  6. Masood F. Polymeric nanoparticles for targeted drug delivery system for cancer therapy. Mater Sci Eng C. 2016;60:569–78.

    CAS  Google Scholar 

  7. Yang K, Feng L, Liu Z. Stimuli responsive drug delivery systems based on nano-graphene for cancer therapy. Adv Drug Deliv Rev. 2016;105:228–41.

    CAS  PubMed  Google Scholar 

  8. Zhu Y, Shi J, Shen W, Dong X, Feng J, Ruan M, Li Y. Stimuli-responsive controlled drug release from a hollow mesoporous silica sphere/polyelectrolyte multilayer core-shell structure. Angew Chem Int Ed. 2005;12:5083–7.

    Google Scholar 

  9. Ganta S, Devalapally H, Shahiwala A, Amiji M. A review of stimuli-responsive nanocarriers for drug and gene delivery. J Control Release. 2008;126:187–204.

    CAS  PubMed  Google Scholar 

  10. Yin Q, Shen J, Zhang Z, Yu H, Li Y. Reversal of multidrug resistance by stimuli-responsive drug delivery systems for therapy of tumor. Adv Drug Deliv Rev. 2013;65:1699–715.

    CAS  PubMed  Google Scholar 

  11. Yin W, Ke W, Chen W, Xi L, Zhou Q, Mukerabigwi JF, Ge Z. Integrated block copolymer prodrug nanoparticles for combination of tumor oxidative stress amplification and ROS-responsive drug release. Biomaterials. 2019;195:63–74.

    CAS  PubMed  Google Scholar 

  12. Saravanakumar G, Kim J, Kim WJ. Reactive-oxygen-species-responsive drug delivery systems: promises and challenges. Adv Sci. 2017;4:1600124.

    Google Scholar 

  13. Lee SH, Gupta MK, Bang JB, Bae H, Sung HJ. Current progress in reactive oxygen species (ROS)-responsive materials for biomedical applications. Adv Healthc Mater. 2013;2:908–15.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhou T, Zhou X, Xing D. Controlled release of doxorubicin from graphene oxide based charge-reversal nanocarrier. Biomaterials. 2014;35:4185–94.

    CAS  PubMed  Google Scholar 

  15. Zhang Y, Teh C, Li M, Ang CY, Tan SY, Qu Q, Korzh V, Zhao Y. Acid-responsive polymeric doxorubicin prodrug nanoparticles encapsulating a near-infrared dye for combined photothermal-chemotherapy. Chem Mater. 2016;28:7039–50.

    CAS  Google Scholar 

  16. Xu C, Yan Y, Tan J, Yang D, Jia X, Wang L, Xu Y, Cao S, Sun S. Biodegradable nanoparticles of polyacrylic acid-stabilized amorphous CaCO3 for tunable pH-responsive drug delivery and enhanced tumor inhibition. Adv Funct Mater. 2019;29:1808146.

    Google Scholar 

  17. Liu J, Zhang B, Luo Z, Ding X, Li J, Dai L, Zhou J, Zhao X, Ye J, Cai K. Enzyme responsive mesoporous silica nanoparticles for targeted tumor therapy in vitro and in vivo. Nanoscale. 2015;7:3614–26.

    CAS  PubMed  Google Scholar 

  18. Lee SJ, Jeong Y-I, Park H-K, Kang DH, Oh J-S, Lee S-G, Lee HC. Enzyme-responsive doxorubicin release from dendrimer nanoparticles for anticancer drug delivery. Int J Nanomed. 2015;10:5489.

    CAS  Google Scholar 

  19. Hu Q, Katti PS, Gu Z. Enzyme-responsive nanomaterials for controlled drug delivery. Nanoscale. 2014;6:12273–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Chen J, Ding J, Wang Y, Cheng J, Ji S, Zhuang X, Chen X. Sequentially responsive shell-stacked nanoparticles for deep penetration into solid tumors. Adv Mater. 2017;29:1701170.

    Google Scholar 

  21. Chi Y, Yin X, Sun K, Feng S, Liu J, Chen D, Guo C, Wu Z. Redox-sensitive and hyaluronic acid functionalized liposomes for cytoplasmic drug delivery to osteosarcoma in animal models. J Control Release. 2017;261:113–25.

    CAS  PubMed  Google Scholar 

  22. Sun B, Luo C, Yu H, Zhang X, Chen Q, Yang W, Wang M, Kan Q, Zhang H, Wang Y. Disulfide bond-driven oxidation-and reduction-responsive prodrug nanoassemblies for cancer therapy. Nano Lett. 2018;18:3643–50.

    CAS  PubMed  Google Scholar 

  23. Li Y, Lu A, Long M, Cui L, Chen Z, Zhu L. Nitroimidazole derivative incorporated liposomes for hypoxia-triggered drug delivery and enhanced therapeutic efficacy in patient-derived tumor xenografts. Acta Biomater. 2019;83:334–48.

    CAS  PubMed  Google Scholar 

  24. Liu H, Xie Y, Zhang Y, Cai Y, Li B, Mao H, Liu Y, Lu J, Zhang L, Yu R. Development of a hypoxia-triggered and hypoxic radiosensitized liposome as a doxorubicin carrier to promote synergetic chemo-/radio-therapy for glioma. Biomaterials. 2017;121:130–43.

    CAS  PubMed  Google Scholar 

  25. Sanderson RJ, Hering MA, James SF, Sun MM, Doronina SO, Siadak AW, Senter PD, Wahl AF. In vivo drug-linker stability of an anti-CD30 dipeptide-linked auristatin immunoconjugate. Clin Cancer Res. 2005;11:843–52.

    CAS  PubMed  Google Scholar 

  26. Dorywalska M, Strop P, Melton-Witt JA, Hasa-Moreno A, Farias SE, Galindo Casas M, Delaria K, Lui V, Poulsen K, Loo C. Effect of attachment site on stability of cleavable antibody drug conjugates. Bioconjug Chem. 2015;26:650–9.

    CAS  PubMed  Google Scholar 

  27. Gorovits B, Krinos-Fiorotti C. Proposed mechanism of off-target toxicity for antibody-drug conjugates driven by mannose receptor uptake. Cancer Immunol Immunother. 2013;62:217–23.

    CAS  PubMed  Google Scholar 

  28. Karimi M, Sahandi Zangabad P, Baghaee-Ravari S, Ghazadeh M, Mirshekari H, Hamblin MR. Smart nanostructures for cargo delivery: uncaging and activating by light. J Am Chem Soc. 2017;139:4584–610.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Hyun DC, Lu P, Choi SI, Jeong U, Xia Y. Microscale polymer bottles corked with a phase-change material for temperature-controlled release. Angew Chem Int Ed. 2013;52:10468–71.

    CAS  Google Scholar 

  30. Moon GD, Choi S-W, Cai X, Li W, Cho EC, Jeong U, Wang LV, Xia Y. A new theranostic system based on gold nanocages and phase-change materials with unique features for photoacoustic imaging and controlled release. J Am Chem Soc. 2011;133:4762–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Li J, Pu K. Semiconducting polymer nanomaterials as near-infrared photoactivatable protherapeutics for cancer. Acc Chem Res. 2020;53:752–62.

    CAS  PubMed  Google Scholar 

  32. Xiong Q, Lim Y, Li D, Pu K, Liang L, Duan H. Photoactive nanocarriers for controlled delivery. Adv Funct Mater. 2020;30:1903896.

    CAS  Google Scholar 

  33. Shaker MA, Younes HM. Photo-irradiation paradigm: mapping a remarkable facile technique used for advanced drug, gene and cell delivery. J Control Release. 2015;217:10–26.

    CAS  PubMed  Google Scholar 

  34. Fomina N, McFearin C, Sermsakdi M, Edigin O, Almutairi A. UV and near-IR triggered release from polymeric nanoparticles. J Am Chem Soc. 2010;132:9540–2.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Gnanasammandhan MK, Idris NM, Bansal A, Huang K, Zhang Y. Near-IR photoactivation using mesoporous silica-coated NaYF4:Yb, Er/Tm upconversion nanoparticles. Nat Protoc. 2016;11:688.

    CAS  PubMed  Google Scholar 

  36. Li J, Pu K. Development of organic semiconducting materials for deep-tissue optical imaging, phototherapy and photoactivation. Chem Soc Rev. 2019;48:38–71.

    CAS  PubMed  Google Scholar 

  37. Li J, Duan H, Pu K. Nanotransducers for near-infrared photoregulation in biomedicine. Adv Mater. 2019;31:1901607.

    Google Scholar 

  38. Hessel CM, Pattani VP, Rasch M, Panthani MG, Koo B, Tunnell JW, Korgel BA. Copper selenide nanocrystals for photothermal therapy. Nano Lett. 2011;11:2560–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Vankayala R, Kuo CL, Nuthalapati K, Chiang CS, Hwang KC. Nucleus-targeting gold nanoclusters for simultaneous in vivo fluorescence imaging, gene delivery, and NIR-light activated photodynamic therapy. Adv Funct Mater. 2015;25:5934–45.

    CAS  Google Scholar 

  40. Jayakumar MKG, Idris NM, Zhang Y. Remote activation of biomolecules in deep tissues using near-infrared-to-UV upconversion nanotransducers. Proc Natl Acad Sci USA. 2012;109:8483–8.

    CAS  PubMed  Google Scholar 

  41. Yang G, Liu J, Wu Y, Feng L, Liu Z. Near-infrared-light responsive nanoscale drug delivery systems for cancer treatment. Coord Chem Rev. 2016;320–321:100–17.

    Google Scholar 

  42. Luo L, Zhu C, Yin H, Jiang M, Zhang J, Qin B, Luo Z, Yuan X, Yang J, Li W. Laser immunotherapy in combination with perdurable PD-1 blocking for the treatment of metastatic tumors. ACS Nano. 2018;12:7647–62.

    CAS  Google Scholar 

  43. Li H, Li J, Ke W, Ge Z. A near-infrared photothermal effect-responsive drug delivery system based on indocyanine green and doxorubicin-loaded polymeric micelles mediated by reversible diels-alder reaction. Macromol Rapid Commun. 2015;36:1841–9.

    CAS  PubMed  Google Scholar 

  44. Ryu T-K, Baek S-W, Kang R-H, Jeong K-Y, Jun D-R, Choi S-W. Photodynamic and photothermal tumor therapy using phase-change material nanoparticles containing chlorin e6 and nanodiamonds. J Control Release. 2018;270:237–45.

    CAS  PubMed  Google Scholar 

  45. Li J, Cui D, Huang J, He S, Yang Z, Zhang Y, Luo Y, Pu K. Organic semiconducting pro-nanostimulants for near-infrared photoactivatable cancer immunotherapy. Angew Chem Int Ed. 2019;58:12680–7.

    CAS  Google Scholar 

  46. Cheng L, Wang C, Feng L, Yang K, Liu Z. Functional nanomaterials for phototherapies of cancer. Chem Rev. 2014;114:10869–939.

    CAS  PubMed  Google Scholar 

  47. Henderson BW, Fingar VH. Relationship of tumor hypoxia and response to photodynamic treatment in an experimental mouse tumor. Cancer Res. 1987;47:3110–4.

    CAS  PubMed  Google Scholar 

  48. He S, Krippes K, Ritz S, Chen Z, Best A, Butt H-J, Mailänder V, Wu S. Ultralow-intensity near-infrared light induces drug delivery by upconverting nanoparticles. Chem Commun. 2015;51:431–4.

    CAS  Google Scholar 

  49. Zhen X, Xie C, Jiang Y, Ai X, Xing B, Pu K. Semiconducting photothermal nanoagonist for remote-controlled specific cancer therapy. Nano Lett. 2018;18:1498–505.

    CAS  PubMed  Google Scholar 

  50. Shen S, Zhu C, Huo D, Yang M, Xue J, Xia Y. A hybrid nanomaterial for the controlled generation of free radicals and oxidative destruction of hypoxic cancer cells. Angew Chem Int Ed. 2017;56:8801–4.

    CAS  Google Scholar 

  51. Mathiyazhakan M, Wiraja C, Xu C. A concise review of gold nanoparticles-based photo-responsive liposomes for controlled drug delivery. Nano-Micro Lett. 2018;10:10.

    Google Scholar 

  52. Lokerse WJ, Bolkestein M, ten Hagen TL, de Jong M, Eggermont AM, Grüll H, Koning GA. Investigation of particle accumulation, chemosensitivity and thermosensitivity for effective solid tumor therapy using thermosensitive liposomes and hyperthermia. Theranostics. 2016;6:1717.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Mazzotta E, Tavano L, Muzzalupo R. Thermo-sensitive vesicles in controlled drug delivery for chemotherapy. Pharmaceutics. 2018;10:150.

    CAS  PubMed Central  Google Scholar 

  54. Dong Z, Feng L, Chao Y, Hao Y, Chen M, Gong F, Han X, Zhang R, Cheng L, Liu Z. Amplification of tumor oxidative stresses with liposomal fenton catalyst and glutathione inhibitor for enhanced cancer chemotherapy and radiotherapy. Nano Lett. 2018;19:805–15.

    Google Scholar 

  55. Zhu C, Huo D, Chen Q, Xue J, Shen S, Xia Y. A eutectic mixture of natural fatty acids can serve as the gating material for near-infrared-triggered drug release. Adv Mater. 2017;29:1703702.

    Google Scholar 

  56. Zangabad PS, Mirkiani S, Shahsavari S, Masoudi B, Masroor M, Hamed H, Jafari Z, Taghipour YD, Hashemi H, Karimi M. Stimulus-responsive liposomes as smart nanoplatforms for drug delivery applications. Nanotechnol Rev. 2018;7:95–122.

    CAS  PubMed  Google Scholar 

  57. Yuan Z, Qu S, He Y, Xu Y, Liang L, Zhou X, Gui L, Gu Y, Chen H. Thermosensitive drug-loading system based on copper sulfide nanoparticles for combined photothermal therapy and chemotherapy in vivo. Biomater Sci. 2018;6:3219–30.

    CAS  PubMed  Google Scholar 

  58. Li X, Wang X, Sha L, Wang D, Shi W, Zhao Q, Wang S. Thermosensitive lipid bilayer-coated mesoporous carbon nanoparticles for synergistic thermochemotherapy of tumor. ACS Appl Mater Interfaces. 2018;10:19386–97.

    CAS  PubMed  Google Scholar 

  59. Xiong X, Xu Z, Huang H, Wang Y, Zhao J, Guo X, Zhou S. A NIR light triggered disintegratable nanoplatform for enhanced penetration and chemotherapy in deep tumor tissues. Biomaterials. 2020;245:119840.

    CAS  PubMed  Google Scholar 

  60. Dai Y, Su J, Wu K, Ma W, Wang B, Li M, Sun P, Shen Q, Wang Q, Fan Q. Multifunctional thermosensitive liposomes based on natural phase-change material: near-infrared light-triggered drug release and multimodal imaging-guided cancer combination therapy. ACS Appl Mater Interfaces. 2019;11:10540–53.

    CAS  PubMed  Google Scholar 

  61. Yue C, Yang Y, Song J, Alfranca G, Zhang C, Zhang Q, Yin T, Pan F, Jesús M, Cui D. Mitochondria-targeting near-infrared light-triggered thermosensitive liposomes for localized photothermal and photodynamic ablation of tumors combined with chemotherapy. Nanoscale. 2017;9:11103–18.

    CAS  PubMed  Google Scholar 

  62. Wang Q, Dai Y, Xu J, Cai J, Niu X, Zhang L, Chen R, Shen Q, Huang W, Fan Q. All-in-one phototheranostics: single laser triggers NIR-II fluorescence/photoacoustic imaging guided photothermal/photodynamic/chemo combination therapy. Adv Funct Mater. 2019;29:1901480.

    Google Scholar 

  63. Chen D, Tang Y, Zhu J, Zhang J, Song X, Wang W, Shao J, Huang W, Chen P, Dong X. Photothermal-pH-hypoxia responsive multifunctional nanoplatform for cancer photo-chemo therapy with negligible skin phototoxicity. Biomaterials. 2019;221:119422.

    CAS  PubMed  Google Scholar 

  64. Ding Y, Su S, Zhang R, Shao L, Zhang Y, Wang B, Li Y, Chen L, Yu Q, Wu Y. Precision combination therapy for triple negative breast cancer via biomimetic polydopamine polymer core-shell nanostructures. Biomaterials. 2017;113:243–52.

    CAS  PubMed  Google Scholar 

  65. Sun S, Sun S, Sun Y, Wang P, Zhang J, Du W, Wang S, Liang X. Bubble-manipulated local drug release from a smart thermosensitive cerasome for dual-mode imaging guided tumor chemo-photothermal therapy. Theranostics. 2019;9:8138.

    PubMed  PubMed Central  Google Scholar 

  66. Zhao F, Zhou J, Su X, Wang Y, Yan X, Jia S, Du B. A smart responsive dual aptamers-targeted bubble-generating nanosystem for cancer triplex therapy and ultrasound imaging. Small. 2017;13:1603990.

    Google Scholar 

  67. Wu S, Liu X, Ren J, Qu X. Glutathione depletion in a benign manner by MoS2-based nanoflowers for enhanced hypoxia-irrelevant free-radical-based cancer therapy. Small. 2019;15:1904870.

    CAS  Google Scholar 

  68. Hernandez Montoto A, Montes R, Samadi A, Gorbe M, Terrés JM, Cao-Milan R, Aznar E, Ibanez J, Masot R, Marcos MD. Gold nanostars coated with mesoporous silica are effective and nontoxic photothermal agents capable of gate keeping and laser-induced drug release. ACS Appl Mater Interfaces. 2018;10:27644–56.

    CAS  PubMed  Google Scholar 

  69. Yang J, Shen D, Zhou L, Li W, Li X, Yao C, Wang R, El-Toni AM, Zhang F, Zhao D. Spatially confined fabrication of core-shell gold nanocages@mesoporous silica for near-infrared controlled photothermal drug release. Chem Mater. 2013;25:3030–7.

    CAS  Google Scholar 

  70. Poudel BK, Soe ZC, Ruttala HB, Gupta B, Ramasamy T, Thapa RK, Gautam M, Ou W, Nguyen HT, Jeong J-H. In situ fabrication of mesoporous silica-coated silver–gold hollow nanoshell for remotely controllable chemo-photothermal therapy via phase-change molecule as gatekeepers. Int J Pharm. 2018;548:92–103.

    CAS  PubMed  Google Scholar 

  71. Zhang C, Li D, Pei P, Wang W, Chen B, Chu Z, Zha Z, Yang X, Wang J, Qian H. Rod-based urchin-like hollow microspheres of Bi2S3: facile synthesis, photo-controlled drug release for photoacoustic imaging and chemo-photothermal therapy of tumor ablation. Biomaterials. 2020;237:119835.

    CAS  PubMed  Google Scholar 

  72. Li J, Zhang F, Hu Z, Song W, Li G, Liang G, Zhou J, Li K, Cao Y, Luo Z. Drug “pent-up” in hollow magnetic Prussian blue nanoparticles for NIR-induced chemo-photothermal tumor therapy with trimodal imaging. Adv Healthc Mater. 2017;6:1700005.

    Google Scholar 

  73. Hussain A, Guo S. NIR-triggered release of DOX from sophorolipid-coated mesoporous carbon nanoparticles with the phase-change material 1-tetradecanol to treat MCF-7/ADR cells. J Mater Chem B. 2019;7:974–85.

    CAS  PubMed  Google Scholar 

  74. Feng L, Gai S, He F, Dai Y, Zhong C, Yang P, Lin J. Multifunctional mesoporous ZrO2 encapsulated upconversion nanoparticles for mild NIR light activated synergistic cancer therapy. Biomaterials. 2017;147:39–52.

    CAS  PubMed  Google Scholar 

  75. Li Q, Sun L, Hou M, Chen Q, Yang R, Zhang L, Xu Z, Kang Y, Xue P. Phase-change material packaged within hollow copper sulfide nanoparticles carrying doxorubicin and chlorin e6 for fluorescence-guided trimodal therapy of cancer. ACS Appl Mater Interfaces. 2018;11:417–29.

    PubMed  Google Scholar 

  76. Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and challenges of liposome assisted drug delivery. Front Pharmacol. 2015;6:286.

    PubMed  PubMed Central  Google Scholar 

  77. Van Tran V, Moon J-Y, Lee Y-C. Liposomes for delivery of antioxidants in cosmeceuticals: challenges and development strategies. J Control Release. 2019;300:114–40.

    PubMed  Google Scholar 

  78. Du X, Zhao C, Zhou M, Ma T, Huang H, Jaroniec M, Zhang X, Qiao SZ. Hollow carbon nanospheres with tunable hierarchical pores for drug, gene, and photothermal synergistic treatment. Small. 2017;13:1602592.

    Google Scholar 

  79. Li Y, Li N, Pan W, Yu Z, Yang L, Tang B. Hollow mesoporous silica nanoparticles with tunable structures for controlled drug delivery. ACS Appl Mater Interfaces. 2017;9:2123–9.

    CAS  PubMed  Google Scholar 

  80. Song G, Chao Y, Chen Y, Liang C, Yi X, Yang G, Yang K, Cheng L, Zhang Q, Liu Z. All-in-one theranostic nanoplatform based on hollow TaOx for chelator-free labeling imaging, drug delivery, and synergistically enhanced radiotherapy. Adv Funct Mater. 2016;26:8243–54.

    CAS  Google Scholar 

  81. Li Y, Shi J. Hollow-structured mesoporous materials: chemical synthesis, functionalization and applications. Adv Mater. 2014;26:3176–205.

    CAS  PubMed  Google Scholar 

  82. Saravanakumar G, Lee J, Kim J, Kim WJ. Visible light-induced singlet oxygen-mediated intracellular disassembly of polymeric micelles co-loaded with a photosensitizer and an anticancer drug for enhanced photodynamic therapy. Chem Commun. 2015;51:9995–8.

    CAS  Google Scholar 

  83. Li J, Cui D, Jiang Y, Huang J, Cheng P, Pu K. Near-infrared photoactivatable semiconducting polymer nanoblockaders for metastasis-inhibited combination cancer therapy. Adv Mater. 2019;31:1905091.

    CAS  Google Scholar 

  84. Nani RR, Gorka AP, Nagaya T, Kobayashi H, Schnermann MJ. Near-IR light-mediated cleavage of antibody-drug conjugates using cyanine photocages. Angew Chem Int Ed. 2015;54:13635–8.

    CAS  Google Scholar 

  85. Yuan Y, Liu J, Liu B. Conjugated-polyelectrolyte-based polyprodrug: targeted and image-guided photodynamic and chemotherapy with on-demand drug release upon irradiation with a single light source. Angew Chem Int Ed. 2014;53:7163–8.

    CAS  Google Scholar 

  86. Pei Q, Hu X, Zheng X, Liu S, Li Y, Jing X, Xie Z. Light-activatable red blood cell membrane-camouflaged dimeric prodrug nanoparticles for synergistic photodynamic/chemotherapy. ACS Nano. 2018;12:1630–41.

    CAS  PubMed  Google Scholar 

  87. Li J, Zhen X, Lyu Y, Jiang Y, Huang J, Pu K. Cell membrane coated semiconducting polymer nanoparticles for enhanced multimodal cancer phototheranostics. ACS Nano. 2018;12:8520–30.

    CAS  PubMed  Google Scholar 

  88. Zhen X, Cheng P, Pu K. Recent advances in cell membrane-camouflaged nanoparticles for cancer phototherapy. Small. 2019;15:1804105.

    Google Scholar 

  89. Chen Z, Zhao P, Luo Z, Zheng M, Tian H, Gong P, Gao G, Pan H, Liu L, Ma A, Cai L. Cancer cell membrane-biomimetic nanoparticles for homologous-targeting dual-modal imaging and photothermal therapy. ACS Nano. 2016;10:10049–57.

    CAS  PubMed  Google Scholar 

  90. Sun H, Su J, Meng Q, Yin Q, Chen L, Gu W, Zhang P, Zhang Z, Yu H, Wang S, Li Y. Cancer-cell-biomimetic nanoparticles for targeted therapy of homotypic tumors. Adv Mater. 2016;28:9581–8.

    CAS  PubMed  Google Scholar 

  91. Lee J, Park J, Singha K, Kim WJ. Mesoporous silica nanoparticle facilitated drug release through cascade photosensitizer activation and cleavage of singlet oxygen sensitive linker. Chem Commun. 2013;49:1545–7.

    CAS  Google Scholar 

  92. Yang G, Sun X, Liu J, Feng L, Liu Z. Light-responsive, singlet-oxygen-triggered on-demand drug release from photosensitizer-doped mesoporous silica nanorods for cancer combination therapy. Adv Funct Mater. 2016;26:4722–32.

    CAS  Google Scholar 

  93. Ma N, Li Y, Xu H, Wang Z, Zhang X. Dual redox responsive assemblies formed from diselenide block copolymers. J Am Chem Soc. 2009;132:442–3.

    Google Scholar 

  94. Han P, Li S, Cao W, Li Y, Sun Z, Wang Z, Xu H. Red light responsive diselenide-containing block copolymer micelles. J Mater Chem B. 2013;1:740–3.

    CAS  PubMed  Google Scholar 

  95. Tian Y, Zheng J, Tang X, Ren Q, Wang Y, Yang W. Near-infrared light-responsive nanogels with diselenide-cross-linkers for on-demand degradation and triggered drug release. Part Part Syst Charact. 2015;32:547–51.

    CAS  Google Scholar 

  96. Uthaman S, Pillarisetti S, Mathew AP, Kim Y, Bae WK, Huh KM, Park I-K. Long circulating photoactivable nanomicelles with tumor localized activation and ROS triggered self-accelerating drug release for enhanced locoregional chemo-photodynamic therapy. Biomaterials. 2020;232:119702.

    CAS  PubMed  Google Scholar 

  97. Chen H, Zeng X, Tham HP, Phua SZF, Cheng W, Zeng W, Shi H, Mei L, Zhao Y. NIR-light-activated combination therapy with a precise ratio of photosensitizer and prodrug using a host-guest strategy. Angew Chem Int Ed. 2019;58:7641–6.

    CAS  Google Scholar 

  98. Qian C, Yu J, Chen Y, Hu Q, Xiao X, Sun W, Wang C, Feng P, Shen QD, Gu Z. Light-activated hypoxia-responsive nanocarriers for enhanced anticancer therapy. Adv Mater. 2016;28:3313–20.

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Zhang K, Zhang Y, Meng X, Lu H, Chang H, Dong H, Zhang X. Light-triggered theranostic liposomes for tumor diagnosis and combined photodynamic and hypoxia-activated prodrug therapy. Biomaterials. 2018;185:301–9.

    CAS  PubMed  Google Scholar 

  100. Cui D, Huang J, Zhen X, Li J, Jiang Y, Pu K. A semiconducting polymer nano-prodrug for hypoxia-activated photodynamic cancer therapy. Angew Chem Int Ed. 2019;58:5920–4.

    CAS  Google Scholar 

  101. Wang Z, Sun M, Liu T, Tan X, Zhang H, Zhang X, He Z, Sun J. A surfactant-like chemotherapeutic agent as a nanocarrier for delivering photosensitizers against cancer: a facile drug-delivering-drug strategy. Int J Pharm. 2019;562:313–20.

    CAS  PubMed  Google Scholar 

  102. Senthilkumar T, Zhou L, Gu Q, Liu L, Lv F, Wang S. Conjugated polymer nanoparticles with appended photo-responsive units for controlled drug delivery, release, and imaging. Angew Chem Int Ed. 2018;57:13114–9.

    CAS  Google Scholar 

  103. Kienzler MA, Reiner A, Trautman E, Yoo S, Trauner D, Isacoff EY. A red-shifted, fast-relaxing azobenzene photoswitch for visible light control of an ionotropic glutamate receptor. J Am Chem Soc. 2013;135:17683–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Gu Z, Yan L, Tian G, Li S, Chai Z, Zhao Y. Recent advances in design and fabrication of upconversion nanoparticles and their safe theranostic applications. Adv Mater. 2013;25:3758–79.

    CAS  PubMed  Google Scholar 

  105. Xiang J, Ge F, Yu B, Yan Q, Shi F, Zhao Y. Nanocomplexes of photolabile polyelectrolyte and upconversion nanoparticles for near-infrared light-triggered payload release. ACS Appl Mater Interfaces. 2018;10:20790–800.

    CAS  PubMed  Google Scholar 

  106. Boyer J-C, Carling C-J, Gates BD, Branda NR. Two-way photoswitching using one type of near-infrared light, upconverting nanoparticles, and changing only the light intensity. J Am Chem Soc. 2010;132:15766–72.

    CAS  PubMed  Google Scholar 

  107. Zhao L, Peng J, Huang Q, Li C, Chen M, Sun Y, Lin Q, Zhu L, Li F. Near-infrared photoregulated drug release in living tumor tissue via yolk–shell upconversion nanocages. Adv Funct Mater. 2014;24:363–71.

    CAS  Google Scholar 

  108. Huang L, Zhao Y, Zhang H, Huang K, Yang J, Han G. Expanding anti-stokes shifting in triplet-triplet annihilation upconversion for in vivo anticancer prodrug activation. Angew Chem Int Ed. 2017;56:14400–4.

    CAS  Google Scholar 

  109. He S, Li C, Zhang Q, Ding J, Liang X-J, Chen X, Xiao H, Chen X, Zhou D, Huang Y. Tailoring platinum(IV) amphiphiles for self-targeting all-in-one assemblies as precise multimodal theranostic nanomedicine. ACS Nano. 2018;12:7272–81.

    CAS  PubMed  Google Scholar 

  110. Dai Y, Xiao H, Liu J, Yuan Q, Ma P, Yang D, Li C, Cheng Z, Hou Z, Yang P, Lin J. In vivo multimodality imaging and cancer therapy by near-infrared light-triggered trans-platinum pro-drug-conjugated upconverison nanoparticles. J Am Chem Soc. 2013;135:18920–9.

    CAS  PubMed  Google Scholar 

  111. Min Y, Li J, Liu F, Yeow EK, Xing B. Near-infrared light-mediated photoactivation of a platinum antitumor prodrug and simultaneous cellular apoptosis imaging by upconversion-luminescent nanoparticles. Angew Chem Int Ed. 2014;53:1012–6.

    CAS  Google Scholar 

  112. Xu J, Kuang Y, Lv R, Yang P, Li C, Bi H, Liu B, Yang D, Dai Y, Gai S. Charge convertibility and near infrared photon co-enhanced cisplatin chemotherapy based on upconversion nanoplatform. Biomaterials. 2017;130:42–55.

    CAS  PubMed  Google Scholar 

  113. Liu J, Bu W, Pan L, Shi J. NIR-triggered anticancer drug delivery by upconverting nanoparticles with integrated azobenzene-modified mesoporous silica. Angew Chem Int Ed. 2013;52:4375–9.

    CAS  Google Scholar 

  114. Zhang Y, Zhang Y, Song G, He Y, Zhang X, Liu Y, Ju H. A DNA-azobenzene nanopump fueled by upconversion luminescence for controllable intracellular drug release. Angew Chem Int Ed. 2019;58:18207–11.

    CAS  Google Scholar 

  115. Liu C, Zhang Y, Liu M, Chen Z, Lin Y, Li W, Cao F, Liu Z, Ren J, Qu X. A NIR-controlled cage mimicking system for hydrophobic drug mediated cancer therapy. Biomaterials. 2017;139:151–62.

    CAS  PubMed  Google Scholar 

  116. Chen G, Jaskula-Sztul R, Esquibel CR, Lou I, Zheng Q, Dammalapati A, Harrison A, Eliceiri KW, Tang W, Chen H. Neuroendocrine tumor-targeted upconversion nanoparticle-based micelles for simultaneous NIR-controlled combination chemotherapy and photodynamic therapy, and fluorescence imaging. Adv Funct Mater. 2017;27:1604671.

    PubMed  PubMed Central  Google Scholar 

  117. Bagheri A, Arandiyan H, Boyer C, Lim M. Lanthanide-doped upconversion nanoparticles: emerging intelligent light-activated drug delivery systems. Adv Sci. 2016;3:1500437.

    Google Scholar 

  118. Jiang Y, Li J, Zhen X, Xie C, Pu K. Dual-peak absorbing semiconducting copolymer nanoparticles for first and second near-infrared window photothermal therapy: a comparative study. Adv Mater. 2018;30:1705980.

    Google Scholar 

  119. Maruoka Y, Nagaya T, Sato K, Ogata F, Okuyama S, Choyke PL, Kobayashi H. Near infrared photoimmunotherapy with combined exposure of external and interstitial light sources. Mol Pharm. 2018;15:3634–41.

    CAS  PubMed  Google Scholar 

  120. Younis MR, Wang C, An R, Wang S, Younis MA, Li Z-Q, Wang Y, Ihsan A, Ye D, Xia X-H. Low power single laser activated synergistic cancer phototherapy using photosensitizer functionalized dual plasmonic photothermal nanoagents. ACS Nano. 2019;13:2544–57.

    CAS  PubMed  Google Scholar 

  121. Yu M, Zhou J, Du B, Ning X, Authement C, Gandee L, Kapur P, Hsieh JT, Zheng J. Noninvasive staging of kidney dysfunction enabled by renal-clearable luminescent gold nanoparticles. Angew Chem Int Ed. 2016;55:2787–91.

    CAS  Google Scholar 

  122. Repenko T, Rix A, Ludwanowski S, Go D, Kiessling F, Lederle W, Kuehne AJ. Bio-degradable highly fluorescent conjugated polymer nanoparticles for bio-medical imaging applications. Nat Commun. 2017;8:470.

    PubMed  PubMed Central  Google Scholar 

  123. Lei T, Guan M, Liu J, Lin H-C, Pfattner R, Shaw L, McGuire AF, Huang T-C, Shao L, Cheng K-T. Biocompatible and totally disintegrable semiconducting polymer for ultrathin and ultralightweight transient electronics. Proc Natl Acad Sci USA. 2017;114:5107–12.

    CAS  PubMed  Google Scholar 

  124. Huang P, Chen Y, Lin H, Yu L, Zhang L, Wang L, Zhu Y, Shi J. Molecularly organic/inorganic hybrid hollow mesoporous organosilica nanocapsules with tumor-specific biodegradability and enhanced chemotherapeutic functionality. Biomaterials. 2017;125:23–37.

    CAS  PubMed  Google Scholar 

  125. Liu Y, Shu G, Li X, Chen H, Zhang B, Pan H, Li T, Gong X, Wang H, Wu X. Human HSP70 promoter-based Prussian blue nanotheranostics for thermo-controlled gene therapy and synergistic photothermal ablation. Adv Funct Mater. 2018;28:1802026.

    Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by the Key Project of Natural Science Foundation of Anhui Provincial Department of Education, China (KJ2019A0402), and the Provincial Quality Engineering Key Project of Chinese Institutions of Higher Learning (2019mooc590).

Author information

Authors and Affiliations

Authors

Contributions

XW, ZX, XZ and HS write the original draft manuscript; JL and ZX review and edit the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jingchao Li or Zongyu Xie.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that there is no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, X., Xuan, Z., Zhu, X. et al. Near-infrared photoresponsive drug delivery nanosystems for cancer photo-chemotherapy. J Nanobiotechnol 18, 108 (2020). https://doi.org/10.1186/s12951-020-00668-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-020-00668-5

Keywords