Multifunctional nanoplatforms for subcellular delivery of drugs in cancer therapy

Multifunctional nanoplatforms for subcellular delivery of drugs in cancer therapy

Progress in Materials Science 107 (2020) 100599 Contents lists available at ScienceDirect Progress in Materials Science journal homepage: www.elsevi...

3MB Sizes 0 Downloads 67 Views

Progress in Materials Science 107 (2020) 100599

Contents lists available at ScienceDirect

Progress in Materials Science journal homepage: www.elsevier.com/locate/pmatsci

Multifunctional nanoplatforms for subcellular delivery of drugs in cancer therapy Xing Guoa, Xiao Weia, Zi Chenb, Xiaobin Zhanga, Guang Yanga, Shaobing Zhoua,

T



a Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China b Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA

A R T IC LE I N F O

ABS TRA CT

Keywords: Nanoparticle Targeted delivery Cancer therapy Organelle-targeting Stimuli-sensitive

To achieve highly effective treatment to diseases, successful delivery of drugs with a carrier into the specific organelles (nucleus, mitochondria, lysosomes, etc.) is of great importance. Targeted delivery based on nanoparticle (NP)-based drug delivery systems (NDDSs) is mainly focused on cell-membrane targeting. In this review we summarize researches on organelle-specific drug delivery with multifunctional NPs. Many effective strategies are introduced for functionalizing these NPs by altering their chemical composition or by grafting functional groups onto their surface for improving organelle-targeting ability. Only when the concentration of released drugs becomes high enough will they interact with molecular targets on specific organelles to induce

Abbreviations: Ab, antibody; ABC, ammonium bicarbonate; Ad, adenoviruses; ANG, angiopep; ApDCs, aptamer-drug conjugates; AR, aspect ratio; ATP, adenosine triphosphate; AuNPs, gold nanoparticles; B55, residues 83-137 of mature bindin; BBB, blood-brain barrier; CME, clathrin-mediated endocytosis; CPPs, cell penetrating peptides; CPT, camptothecin; CvME, caveolae-mediated endocytosis; DLCs, delocalized lipophilic cations; DMMA, dimethylmaleic anhydride; DOPE, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine; DOTAP, 1,2-dioleyl-3-trimethylammonium-propane; DOX, doxorubicin; DQA, 1,1′-decamethylene bis (4-aminoquinaldiniumchloride); EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; EGFP, enhanced green fluorescent protein; ELS, endoplasmic reticulum localization sequence; EPR, enhanced permeation and retention; ER, endoplasmic reticulum; ERHNPs, electro-responsive hydrogel nanoparticles; Fab, antigen-binding fragment; FPs, fusogenic peptides; FR, folate receptor; GA, Golgi apparatus; GALA, glutamic acid-alanine-leucine-alanine; Gb3, globotriaosylceramide; GI, gastrointestinal; GILT, gamma interferon-inducible lysosomal thiol; GLS, Golgi localization sequence; GSH, glutathione; HeLa, human cervix adenocarcinoma; HepG2, human hepatoma; HIV, human immunodeficiency virus; HNPs, hexagonal nanoprisms; HPMA, N-(2-hydroxypropyl) methacrylamide; HT, HEAT; IA, imidazoacridinone; ICP, inductively-coupled plasma-atomic emission spectroscopy; IDS, intracellularly detachable subgroups; IMM, inner mitochondrial membrane; IMS, intermembrane space; Ir, iridium; IUSs, intervening interunit spacers; LAMP, lysosomal-associated membrane protein; LCST, lower critical solution temperature; LCVs, large compound vesicles; LIMP, lysosomal integral membrane protein; LMP, lysosomal membrane permeabilization; LRP, lipoprotein receptor-related protein; LSDs, lysosomal storage diseases; M6P, mannose-6-phosphate; MAPK, mitogen-activated protein kinase; MCF-7, human breast cancer; MD, molecular dynamics; MDR, multidrug resistance; MHC, major histocompatibility complex; MSNP, mesoporous silica nanoparticle; mTOR, mammalian target of rapamycin; MTS, mitochondrial targeting signal; N9, murine microglial cell line; NCs, nanoconjugates; NDDSs, nanoparticle-based drug delivery systems; NE, nuclear envelope; NER, nucleotide excision repair; NLSs, nuclear localization signals; NP, nanoparticle; NPCs, nuclear pore complexes; Nups, nucleoporins; OLL, oligolysine; OMM, outer mitochondrial membrane; PI3K, phosphoinositide 3-kinase; PAMAM, poly(amidoamine); PCL, poly(ε-caprolactone); PEG, poly(ethylene glycol); PEI, poly (ethyleneimine); P-gp, p-glycoprotein; Phis, poly-histidine; PLGA, poly(lactide-co-glycolide); PLA, poly(lactic acid); PLL, poly(L-lysine); PS, polystyrene; PSD, poly(methacryloyl sulfadimethoxine); QDs, quantum dots; R8, octaarginine; RAW 264.7, murine macrophage cell line; RGD, arginylglycyl-aspartic acid; ROS, reactive oxygen species; SNAP, synaptosome-associated protein; SNPs, spherical nanoparticles; STxB, Shiga toxin B; SV40, simian virus 40; TAT, HIV transactivator protein; Tf, transferrin; TfR, transferrin receptor; TPP, triphenylphosphonium; UPR, unfolded protein response; VAMP, vesicle-associated membrane protein; VNB, vapor nanobubble ⁎ Corresponding author. E-mail address: [email protected] (S. Zhou). https://doi.org/10.1016/j.pmatsci.2019.100599 Received 15 April 2017; Received in revised form 25 August 2019; Accepted 26 August 2019 Available online 26 August 2019 0079-6425/ © 2019 Elsevier Ltd. All rights reserved.

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

tumor cell apoptosis. Organelle-specific delivery will become one of the primary goals for targeted drug delivery research.

1. Introduction The success of traditional cancer treatments such as chemotherapy and radiation therapy are often constrained by drug-associated side effects, because of the lack of targeted delivery techniques for transporting anticancer compounds to the neoplastic tissue and the fact that the drug concentration at the tumor site is typically low. Targeted delivery of anticancer agents is important to overcome these limitations. Recent developments in nanoparticle (NP)-based drug delivery systems (NDDSs) have demonstrated that they can carry drugs to pathological tissues via cell-membrane targeting and release these drugs in the cytoplasm [1,2]. After successful delivery into the targeted cell, a drug should get access to a particular organelle (endo/lysosome, mitochondria, Golgi apparatus, nucleus, etc.) to be effective [3]. Organelle-specific delivery has become one of the primary goals for targeted drug delivery research [4]. Many chemotherapeutics are limited in clinic application due to the poor water solubility, unmanageable toxicities, and low therapeutic efficiency. One elegant strategy to solve these problems is the use of NDDSs [5–9]. NPs templates with tunable composition, size, shape, and surface chemistry can be used as universal platforms in a variety of disease treatments. Most NDDSs researches have focused on cell-membrane targeting; for an excellent review of recent developments in the field we point the reader to reviews here [10]. Relatively few review articles have covered research on organelle-specific drug delivery. In this review, we provided a comprehensive overview on recent research towards multifunctional NPs for organelle-specific drug delivery. When nanomedicines are injected into blood, they can first accumulate in tumor tissues via the well-known enhanced permeation and retention (EPR) effect [2,11]. They will be internalized into tumor cells via different pathways which depend on their size, shape, stiffness [12], and surface chemistry. To increase drug delivery efficiency, both passive and active targeted delivery strategies are used. After these NPs enter the cytoplasm, it is important for the carried cargos to reach specific organelles. An effective method is to functionalize these NPs by altering their chemical composition or by grafting functional groups onto their surface. Only when the concentration of released drugs becomes high enough will they interact with molecular targets on specific organelles to induce tumor cell apoptosis.

2. Internalization strategies 2.1. Internalization mechanisms According to the physicochemical properties of particles and the types of cells, there are two major internalization mechanisms: phagocytosis and pinocytosis. Phagocytosis is the process of cell eating, usually occurs in internalization of large particles; and pinocytosis is also called as cell drinking, which occurs when small particles enter the cell. These internalization pathways are quite important since they determine the NPs fate through various possible subcellular compartments. For instance, NPs internalized through clathrin-mediated endocytosis will locate in lysosomes, while those internalized through caveolin-mediated endocytosis will not [13]. In clathrin-mediated endocytosis internalization, NPs must escape from the lysosomes as soon as possible in order to prevent

Fig. 1. Internalization mechanisms of NPs by cells and respective size limitations. Large particles usually generate a phagocytic response; particles > 1 μm (purple spheres) are internalized through micropinocytosis; while uptake of smaller particles can be assisted by a couple of pathways. According to size variations, 120 nm-NPs (green spheres) are internalized through clathrin-mediated endocytosis (CME), 90 nm-NPs (orange spheres) are internalized through CME- and caveolae-mediated endocytosis (CvME)-independent endocytosis, and 60 nm-NPs (blue spheres) are internalized through CvME. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.) 2

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

loading degradation. After that, the cargo can be delivered to the desired subcellular compartments including the cytosol, the mitochondria, or the nucleus. The cellular uptake pathway of particles is heavily dependent on particle size (Fig. 1). 2.1.1. Phagocytosis Phagocytosis mostly occurs in professional phagocytes, including polymorphonuclear leucocytes and mononuclear cells such as macrophage and monocytes [14]. In contrast, non-professional phagocytes like epithelial cells and endothelial cells show a much weaker phagocytic activity [15]. Particles smaller than 10 μm can be taken up by macrophage, which is the main obstacle that hinders the drug delivery to the targeted cell. Macrophages can take in particles up to 10 μm in size and are one of the major barriers that limit the effective delivery of vehicles to the desired location. It remains a grand challenge to develop NPs that are capable of evading macrophage clearance and sustaining good bio-distribution. Generally, particles > 1 μm generate a phagocytic response [16,17]. 2.1.2. Pinocytosis Pinocytosis, the non-phagocytic endocytosis, is restricted to specialized cells and occurs in all cell types [18]. For cancer-associated delivery, tumor cells belong to non-phagocytic cells, can take in particles up to 500 nm [19], therefore, pinocytosis is more relevant to the uptake of NPs by tumor cells. There are four main pinocytotic mechanisms, which will be described as following. 2.1.2.1. Clathrin-mediated endocytosis (CME). This is the main pathway for cellular entry. CME happens with the assistance of clathrin, a cytoplasmic protein, which assembles into the coated pits during the process. CME allows for the cellular internalization by decorating NPs with particular ligands, which are capable of recognizing the specific receptors overexpressed on the cell membranes [15]. The coated pits develop into vesicles after endocytosis, followed by entry into the endo/lysosomes to release the NPs [14]. Therefore, NPs have to escape form the endo/lysosomes for further subcellular delivery after being internalized by CME. 2.1.2.2. Caveolae-mediated endocytosis (CvME). Caveolae is small flask-shaped membrane invagination [18], commonly found in many cells, especially the adipocytes [20]. Various proteins are involved in endocytosis and trancytosis through CvME, like cavin, dynamin, synaptosome-associated protein and vesicle-associated membrane protein [21,22]. Unlike the CME, CvME bypasses the endo/lysosomal compartments. Thus, delivery of proteins and DNA into cells through CvME can effectively prevent the therapeutics from degrading by endo/lysosomes [23,24]. Compared to the CME, CvME possesses slower process and forms smaller vesicles [25]. 2.1.2.3. CME- and CvME-independent endocytosis. This pathway refers to the internalization of the particles by the cells without clathrin and caveolin. Vesicles formed by clathrin- and caveolin-independent endocytosis are around 90 nm in diameter. The internalized particles are matured to become early and late endosomes. There are not many NPs reported that can be internalized by the CME- and CvME-independent endocytosis, and understanding of interactions between NPs with cells in such pathway is still in a nascent stage. 2.1.2.4. Macropinocytosis. Macropinocytosis occurs in many cells, especially macrophages [26]. For macropinocytotic pathway, NPs are transferred into the cells by forming an invagination around them at the plasma membrane, termed as a macropinosome [27]. Once inside the cells, these macropinosomes are fused with endo/lysosomal compartments or recycle the content to cell membrane [26]. Macropinocytosis is an actin-driven process like phagocytosis, but the macropinosomes are much larger, commonly several microns. This endocytic pathway is generally nonspecific to take in large particles [28]. 2.2. Targeting A key goal of drug delivery system is to unload their payloads at the desired tissue. Targeting comprises passive and active targeting. The biophysicochemical characteristics of NPs, such as particle size, shape, charge and targeting ligands can influence the interaction of NPs with cells. The following is a brief discussion about the passive and active targeting of NPs-based drug delivery systems. 2.2.1. Passive targeting The blood vessels in tumor tissues are frequently leaky, so NPs can passively target tumors via the EPR effect [29]. The EPR effect has become an effective means for delivery of anticancer drugs to tumors, whether using polymer conjugates, liposomes or micelles. Passive targeting of NPs is mainly affected by the biophysicochemical properties of them, which have an influence on cellular uptake, toxicity, and exocytosis. Thus, studying the relationship between the NPs biophysicochemical characteristics and their interaction with cells provides an elegant method for designing effective nanocarriers. 2.2.1.1. Size. The particle size greatly influences the process of cellular internalization. It may affect the uptake efficiency and kinetics, the internalization mechanism, and the subcellular distribution. NPs larger than 500 nm can be phagocytized by macrophages, while smaller NPs can be endocytized by phagocytic or non-phagocytic cells regardless of the method of administration [19,30,31]. Internalization by non-phagocytic cells like tumor cells is desirable to achieve therapeutic effects, and macrophages usually clears large particles from the body quickly, which is undesirable. Particle size also influences the process of crossing the cell membrane. Kulkarni et al. prepared a couple of commercial 3

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

polystyrene NPs with various size (20, 50, 100, 200, and 500 nm), and investigated their ability to transport across the gastrointestinal barrier and the blood-brain barrier [32]. The results showed that the 100 nm and 200 nm polystyrene NPs were able to bypass the two barriers. The size can also affect the subcellular distribution of NPs. For instance, Lovrić et al. proved that smaller quantum dots with a diameter of 2.2 nm could enter the nucleus of murine microglial (N9) cells, while the larger quantum dots approximately 5.2 nm were limited in the nuclear entry with distribution throughout the cytoplasm [33]. In another study, Huang et al. investigated the cellular uptake of gold nanoparticles (AuNPs) with different diameters by human breast cancer (MCF-7) cells. It was found that smaller AuNPs internalized faster. Furthermore, the two AuNPs measuring in 2 nm and 6 nm were located in both the cytoplasm and the nucleus, while the AuNPs measuring in 15 nm were hardly found in nucleus [34]. Tang et al. incubated three CdSe/ZnS quantum dots (3, 4.8 and 8 nm) with human cervix adenocarcinoma (HeLa) cells, the inductively-coupled plasma-atomic emission spectroscopy (ICP) analysis showed that only 3 nm particles were able to accumulate in the nucleus [35]. 2.2.1.2. Shape. Numerous shapes of NPs are able to be constructed due to the rapid development in the technologies. Spheres, rods, cylinders, spherocylinders have been researched to date [36–38]. Non-spherical NPs with various aspect ratios instead of spheres aroused wide interest since the aspect ratio (AR) is a key factor in biological processes, especially the cellular uptake. Gratton et al. reported that the uptake of cylindrical NPs was relevant to their aspect ratios to a large extent [36]. The high-aspect-ratio cylinders (AR = 3) were internalized about four times faster than the low-aspect-ratio cylinders (AR = 1) of similar volumes. In order to study the influence of AR on the cellular internalization, Meng et al. designed a series of mesoporous silica NPs (MSNPs) with different lengths. After incubation with HeLa cells, the extent of internalization was dependent on the variation of AR [39]. MSNPs with AR ranging from 2.1 to 2.5 could deliver more therapeutics to HeLa cells. Vácha et al. reported molecular dynamics (MD) simulations of the cellular uptake of ligand-coated NPs with varying shapes [38]. They showed that the passive endocytosis of spherocylinders is more efficient than that of spheres. Interestingly, study of endocytosis of spherocylinders and cylinders demonstrated that AR has no significant effects on both particles. However, endocytosis is suppressed for cylinders since there are defects at the sharp edges, which prevented the separation of the two membranes. It has shown that shape has a profound impact on the endocytosis of NPs by the macrophages as well as the tumor cells [40]. To design a shape which can avoid uptake by macrophages has garnered considerable attention. Champion et al. demonstrated that a macrophage is more able to internalize a spherical-shape particle, while worm-shaped particles exhibited negligible uptake by macrophages of the same volume [41]. Lin et al. compared the behaviors of spherical NPs (SNPs) and hexagonal nanoprisms (HNPs) in vitro [42]. This study on phagocytosis of HNPs and SNPs showed that HNPs can efficiently decrease phagocytosis more than by SNPs from 91.6 ± 0.8% to 68.1 ± 2.4%. Hu et al. synthesized a polymer-drug conjugate with a reduction-labile linker [43]. A couple of morphologies could be formed by the prodrug. The in vitro cell study indicated that the fastest internalization rate was displayed by the spiky staggered lamellae, and the large compound vesicles (LCVs) showed slower cellular entry while particles with spherical shape internalized slowest compared to the other two. They also investigated the internalization mechanism of the particles with various morphologies. The result indicated that the endocytic pathway of the staggered lamellae and LCVs was mainly by CME- and CvME-independent endocytosis, which was distinct from that of smooth disks and spheres. Interestingly, all other particles apart from the spheres exhibited nuclear drug delivery and more significant cytotoxicity. Furthermore, the in vivo study illustrated that the staggered lamellae and the smooth disks circulated longer in blood compared with LCVs and spheres. 2.2.1.3. Surface charge. The shape and size of NPs contribute markedly to their interaction with cells, however, the surface charge is the dominant factor that influences internalization. It is well-known that positively charged NPs tend to have a higher rate of cell uptake in comparison with the neutral and negatively charged NPs [44]. He et al. studied the relationship between the surface charge of chitosan grafted NPs and the cellular entry rate. Slight surface charge differences remarkably influenced the internalization [31]. It has been shown that the uptake by macrophages increased as the surface charge’s magnitude increased (either positive or negative), however, the NPs with more positive charge and less negative charge were more efficiently internalized by the non-phagocytic cells. In our previous study, we designed a charge-reversal polymer micelle system, which was negatively charged under neutral conditions, but would reverse to carry positive charge triggered by the tumor extracellular pH [45]. We proved that these chargereversal micelles were capable of cancer cell entry quickly at pH 6.8, which is a result of electrostatic interaction, while the cellular uptake of these micelles at pH 7.4 is negligible. This illustrates that the micelles with a positively charged surface, rather than those with a negatively charged surface, can be internalized more by the cells. Most recently, Zhou et al. reported a polymer-drug conjugate that could be triggered by the γ-glutamyl transpeptidase on the cell membrane to generate cationic primary amines. The resulted positively charged conjugate subsequently underwent caveolae-mediated endocytosis and transcytosis, which lead to the fast extravasation and efficient transcytosis of cancer cells [46]. Not just size and surface properties affect the interaction between NPs and cells, it is strongly dependent on cell types as well. Liu et al. investigated the surface charge effect on cell interaction with NPs. AuNPs with both positive charge and negative charge were treated with nonphagocytic human hepatoma (HepG2) cells and murine macrophage (RAW 264.7) cells [47]. There’s no significant difference in uptake by RAW 264.7 cells, whereas HepG2 cells took in much more positively charged AuNPs than AuNPs with negative charge. 2.2.2. Active targeting Although the EPR effect shows an obvious histological difference between normal and tumor tissues, the exploitation of this alone 4

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

is often not adequate. Active targeting has been efficiently exploited to increase the cancer cell entry of NPs and improve the therapeutic effects of their cargos. NPs with actively targeted ligands recognize and bind to specific receptors, which are expressed a lot on cancer cells and minimal on normal cells. The ligands usually include folate, transferrin, aptamers, antibodies, and peptides. 2.2.2.1. Folate. Folate has been one of the most extensively utilized small molecule ligands for targeted drug delivery. Folate is small and less prone to degradation than biomolecular ligands. It has a high affinity to folate receptor (FR), which is up-regulated on the surface of many human tumor cells [48]. The binding of folate to FR has a very high affinity (KD ∼ 10-9), thus, the folate conjugates can deliver cargos targeted to the cancer cells instead of being internalized by the normal cells [49]. Our group developed several folate-attached micelle systems, one example being a polymeric prodrug system for tumor targeting and pH-triggered drug release [50]. The anticancer drug, doxorubicin (DOX), is chemically conjugated to the polymer backbone via pH-responsive hydrazone linkages, and folate bared on the surface of the micelles facilitates the endocytosis by the FR over-expressing tumor cells. Another folate-attached micelle system is a star-shaped vehicle connected with folate as the targeting ligand and the disulfide bond between the hydrophilic and hydrophobic segments can be cleaved by glutathione (GSH) in cytoplasm [51]. More recently, we designed a dual-targeting and dual-responsive polymer micelle system, which possesses both passive targeting ability by a charge-reversal induced electrostatic interaction and an active targeting effect by FR-mediated endocytosis [45]. This dual-targeting vehicle can be internalized by the tumor cells more quickly than the single-targeting one. The results indicate that all of these folate conjugates possessed excellent actively targeting efficacy to FR-overexpressed cancer in vitro and the best therapeutic effect in vivo. 2.2.2.2. Transferrin. Transferrin (Tf) is another ligand that selectively binds to specific membrane-bound receptors on cells for targeting via receptor-mediated endocytosis. Transferrin is an 80-kDa glycoprotein, which selectively binds to its receptor TfR [52]. Since TfR is overexpressed in the tumor cells, Tf-decorated NPs show potential application in the target-cell-specific drug delivery [53]. Roy et al. developed a Tf-targeted chemotherapeutic delivery system [54]. In vitro cell study demonstrated that Tf-targeted group obviously induced more HeLa cells death in comparison with those without Tf decoration. In another study, Sarisozen et al. simultaneously loaded paclitaxel and curcumin into the Tf-modified micelles [55]. The in vitro 3D cell culture experiments proved that the targeted micelles penetrated the spheroid more efficiently than the non-targeted micelles, and combining paclitaxel with curcumin yielded better results than either individually. 2.2.2.3. Aptamers. Aptamers are single-stranded oligonucleotides that fold by intramolecular interactions into unique conformations [56]. Aptamers include DNA, RNA or modified oligonucleotides resistant to nuclease degradation [57]. The use of aptamers to decorate NPs remains an attractive targeting strategy given their small size (∼15 kD), lack of immunogenicity, as well as their easy penetration and targeting to tumor cells [58]. Various aptamer-modified NPs have been prepared to successfully deliver cargos to tumor sites [59–61]. However, the development of aptamer-drug conjugates (ApDCs) faces problems such as complicated preparation and low controllability. In order to resolve these problems, Wang et al. developed a therapeutic module that facilitated solid synthesis of ApDCs [62]. By using this module, they synthesized a photo-cleavable phosphoramdite, which could both target tumor cells specifically and release drugs triggered by light. 2.2.2.4. Antibodies. An antibody (Ab) is a large Y-shaped glycoprotein that can target specific antigens present on the cell membrane. The antibody-antigen interactions can induce antitumor effects caused by multiple mechanisms, such as interfering with ligandreceptor binding and suppression of protein expression [63]. The usage of Abs as targeting ligands has been extensively investigated over the last decade and has resulted in multiple successful treatments [64–66]. However, Abs-targeted NPs still encounter many limitations due to the large molecule weight (∼150 kDa) as well as a hydrodynamic radius of 15–20 nm, instability in organic solvents, and rapid NPs clearance caused by the potential immunogenicity of Abs [67]. Current technology has overcome some of these limitations, for example, by using antigen-binding fragment (Fab) of Abs as targeting ligands, but the monomers and dimers of the Fab recognition patterns still result in large molecules with sizes around 50 and 100 kDa, respectively [2,68]. One method to prolong blood circulation is to remove the crystallizable fragment (i.e., using only Fab fragments), however, these Abs-NPs are still cleared faster than undecorated NPs [69]. 2.2.2.5. Peptides. Peptides are smaller than antibodies, and more stable, less immunogenic, and much easier for NPs conjugation [58]. Screening of potential peptides is usually completed by using a combinatorial phage library, which results in ligands with length ranging from 10 to 15 amino acids. They have high affinity with specific tumor targets [70]. The most studied peptide sequence is that of arginyl-glycyl-aspartic acid (RGD) which strongly binds to integrin ανβ3 overexpressed on endothelial and specific cancer cells [71]. The recognition of the RGD sequence to the ανβ3 integrin has the potential to be exploited for cancer therapy. Kim et al. attached RGD to polymer-coated oncolytic adenoviruses (Ad) vectors to ensure Ad can be selectively delivered to tumor cells [72]. Enhanced transduction efficiency and incremental cytotoxicity were displayed in RGD-conjugated group, which also showed significant in vivo antitumor effect against lung orthotropic tumor. Another peptide sequence capable of targeting behavior is angiopep-2 (ANG), which specifically binds to the low-density lipoprotein receptor-related protein. This receptor is overexpressed on the glioma cells and blood-brain barrier (BBB). ANG-NPs can penetrate the BBB to suppress the neuronal discharges and provide an effective therapy for epilepsy. In order to enhance the brain delivery of phenytoin sodium, Ying et al. decorated one electro-sensitive hydrogel NPs with ANG and studied their antiepileptic activity by using an amygdala kindling model [73]. They found that the ANGmodified NPs could cross the BBB and release the phenytoin sodium within the brain during epileptiform abnormalities. 5

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

3. Intracellular delivery After NPs pass through the first barrier in blood and arrive in the tumor, they must be internalized by cancer cells. The intracellular specific target of NPs depends on the drug action, for instance, gene and antisense oligonucleotides should be transported to nucleus, proapoptotic drugs and mitochondrial DNA to mitochondria, enzymes for lysosomal storage disease and siRNA for mRNA interference to lysosomes, drugs for neurodegenerative disorders and endoplasmic reticulum (ER) storage stress to the Golgi/endoplasmic reticulum (Fig. 2). However, the intracellular delivery of NPs represents a challenge since the cellular internalization of them is energy-dependent, which is different from the endocytosis of small molecules that cross the membrane by random diffusion. NPs will be trapped in the endo/lysosomes even if they have been successfully internalized, and subsequently, the degradation of the NPs together with drugs takes place in acidic conditions. As a consequence, just a tiny part of the loaded drugs can be delivered once in the cytoplasm. To solve this problem, a variety of strategies have been developed as described below. 3.1. Cell penetrating peptides (CPPs) CPPs are a class of short amphiphilic peptides with up to 30 amino acids, commonly used to facilitate cellular uptake of NPs. CPPs are also known as membrane translocating sequences, protein transduction domains and Trojan peptides [74]. Because of the abundance in lysine or arginine, these peptides are highly cationic. CPPs are divided into three classes based on origin: derived CPPs, chimeric CPPs and synthetic CPPs [75,76]. The TAT peptide (human immunodeficiency virus (HIV) transactivator protein), derived from the transactivator activator of human immunodeficiency virus, is one of the frequently used derived CPPs that gain access to the cells [75,77,78]. Chimeric peptides, for instance transportan (TP), are consisting of regions from the neuropeptide galanin and the wasp venom mastoparan. Polyarginine is the best studied synthetic peptide, the cationic nature of which efficiently promote cell entry [79]. Regardless of in-depth investigation of the cell penetrating activity, the internalization mechanism of CPPs is still controversial. Three main possibilities for CPPs crossing the membranes have been proposed: direct penetration into cells, translocation through forming a transitory structure and translocation through endocytosis-mediated pathway (energy-dependent pathway)

Fig. 2. NPs with various biophysicochemical characteristics for subcellular targeting. The orange box represents organelle-related diseases, the purple box represents drugs that act on organelles, and the blue box represents the motifs for subcellular targeting. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.) 6

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

[80–82]. The pathway of CPPs-mediated delivery mainly depends on the concentration of CPPs, the type of cell, and the lipid composition of membrane [83,84]. The endocytosis-mediated pathway is the main internalization mechanism for CPPs according to many studies. CPPs-mediated delivery possesses significant advantages, such as quick internalization, ease of synthesis, and lack of toxicity. CPPs have been conjugated to a variety of NPs to facilitate their cellular uptake, including gold NPs, quantum dots (QDs), magnetic QDs, polymeric micelles, and liposomes. 3.1.1. TAt TAT, derived from the 86-mer trans-activating transactivator activator of human immunodeficiency virus, is one of the most thoroughly studied and extensively used CPPs for intracellular delivery [85,86]. TAT is comprised of 13 amino acids containing two lysine and six arginine residues (GRKKRRQRRRPPQ), which contribute to its basicity and hydrophilicity. In 1999, Josephson et al. fabricated dextran coated superparamagnetic iron oxide NPs derivatized with TAT [87], which is the first report of internalizing NPs using TAT. In another study, TAT-derivatized magnetic NPs displayed a 100-fold higher lymphocytes accumulation over the TAT-free group [88]. TAT-mediated delivery involves multiple mechanisms. Torchilin et al. prepared an intracellular delivery system of TATfunctionalized liposomes, which exhibited efficient cellular uptake even at low temperature and in the presence of metabolic inhibitors, indicating a non-energy dependent process of internalization [89]. Nevertheless, another study showed that TAT-decorated liposomes entered the ovarian carcinoma cells via clathrin-coated pits or caveolin-dependent endocytosis instead of direct cytosolic delivery by plasma membrane translocation [90]. Even if these researchers proved that the TAT-mediated cellular entry is governed by various pathways, whether the energy-dependent and energy-independent modes of transport would occur concurrently still remains unknown. The cell internalization efficiency of TAT-conjugated NPs can be determined by many properties of NPs, including particle size, shape and surface property as described in Section 2.2.1. To promote the cell entry of TAT-conjugated NPs, the TAT concentration and surface arrangement in solution should be considered as well. Todorova et al. simultaneously carried out computational simulations and experiments to study the influence of the two factors on cell permeation (Fig. 3) [91]. By altering the TAT concentrations from 0 to 9%, the higher concentration made the NPs unstable and reduced cell entry, the lower concentration lead to the decrease of the bare NPs exposure; and 5.4% TAT displayed the optimal cell internalization. Moreover, they found that the arrangement of TAT also affected the cell penetration with a relatively even distribution resulting in effective membrane interaction while the high TAT density would lead to the conformational collapse because of the repulsive interactions, in turn hinder the membrane penetration. Recently, Ming et al. studied the relationship between TAT coating density of PEG-PCL micelles and the cell internalization behavior. They found that the increase of TAT density resulted in the increase of membrane-anchoring and internalization rate, and also accelerated the energy-independent pathway [92]. 3.1.2. Penetratin Penetratin, another frequently used CPP, is derived from the homeodomain (residues 43-58) of Drosophila antennapedia [93]. This 16-amino-acid peptide with a N-terminal can be synthesized and facilitate cell entry [93]. In contrast, the same peptide without N-terminal or C-terminal have a limited access to the cells, indicating a necessity of this sequence for efficient internalization. Penetratin internalizes cells either by endocytosis [94–96] or endocytosis along with direct membrane translocation [97,98]. Penetratin possesses the ability to bind heparin sulfates [99] or chondroitin sulfates [100] with high-affinity, offering assistance to endocytosis as a unique internalization pathway for penetratin. In the condition of ionic detergents, penetratin is able to form multimers [93], followed by electrostatically interaction with anionic phospholipids and penetration into the cell membrane [101]. The penetratin/lipid interaction is a function of the vesicle lipid compositions. A high lipid/peptide ratio (325/1) resulted in the αhelical conformation of penetratin, while a low lipid/peptide ratio (10/1) gave the antiparallel β-sheets [102]. Simultaneously, the lipid bilayer organization and the lipid acyl chain direction can be changed by penetratin upon deep insertion into membrane bicelles [103]. Penetratin has been chemically conjugated to a variety of drugs or NPs to facilitate their cellular entry. Penetratin was reported to be functionalized to polymer NPs and the resulting penetratin-NPs displayed increased cell entry [104]. Furthermore, the in vivo biodistribution test indicated obvious brain accumulation enhancement with minimal uptake by non-target sites.

Fig. 3. Illustration of the influence of TAT concentration and distribution in solution on cellular internalization efficiency [91]. 7

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

3.1.3. Octaarginine (R8) Peptides consisting of merely arginine residues, namely oligoarginines, have been widely used for membrane penetration [105]. Although both arginine and lysine are representative basic amino acids, oligoarginines usually promote cellular uptake more efficiently than oligolysines with the same number of residues, mainly due to the fact that more hydrogen bonds form in arginine [106–108]. Among the oligoarginines, R8 with 8 arginine residues exhibits maximized internalization, while others with more or fewer arginine residues were less effective [105]. R8 has been successfully utilized for intracellular transport of several NPs, including multifunctional envelope-type NPs [109] and lipid NPs [110] for gene delivery, as well as PEGylated liposomes for DOX delivery [111]. The use of transporter conjugates can not only gain the cellular uptake but has also been considered as a promising strategy to overcome the drug resistance. The conjugation of R8 to a representative small-molecule therapeutic agent (Taxol) through a bioactivatable disulfide linker results in significantly improved therapeutic effect against malignant cells that are resistant to Taxol alone [112]. The mechanism of this transporter-drug conjugate for overcoming multidrug resistance (MDR) mainly result from a different mode of entry and physical properties of it in comparison with free drug. El-Sayed et al. studied the effect of R8 decoration on endo/lysosomal escape of liposomes and the escape modes of them [113]. They found that the liposomes with R8 modification were capable of endo/lysosomal escape through liposomes-endo/lysosomal membrane fusion at either neutral condition (pH 7.4) or acidic condition (pH 5.5). As a control, the fusion of octalysine (K8) modified liposomes only took place at pH 7.4.

Fig. 4. Schematic illustration of tumor tissue penetration and cellular internalization of NDDSs [125]. 8

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

3.2. Endosomal escape Endosomes usually are the first intracellular compartments encountered by NPs. The endosomes are acidic (pH 5–6) compared to extracellular environment and late evolve into more acidic lysosomes (pH 4–5). Besides, the lysosomal enzymes perhaps lead to the degradation of the carriers and the cargos [114]. Therefore, NPs have to escape from the endo/lysosomes as quickly as possible to get access to the intended subcellular target. There are a variety of approaches that have been successfully applied in endosomal escape, mainly through adopting fusogenic peptides and cationic materials [115]. 3.2.1. Fusogenic peptides (FPs) FPs are acid-responsive amphiphilic peptides, usually utilized to assist NPs in escaping from the endo/lysosomes due to their membrane destabilizing activity. The majority amino acid sequences of FPs are derived as viral fusion protein segments that come from such sources as influenza and HIV. These peptides exist as random coils under the physiological pH condition attributing to the electrostatic repulsion between anionic carboxyls. Once triggered by a lower pH in the environment of late endosomes or lysosomes, the carboxyls become protonated, followed by the formation of an α-helix secondary structure of the peptides. Finally, the structured peptides would merge with the endo/lysosomal membrane and thus escape from the endo/lysosomes successfully [116–118]. A synthetic 30-amino-acid peptide GALA with a glutamic acid- alanine-leucine-alanine repeat produced a membrane permeabilization effect at endo/lysosomal pH, endowing NPs with an efficient cytosolic delivery of loaded cargos [119,120]. In a recent work by Niikura et al., a sea urchin fertilization protein derived peptide, B55 (residues 83-137 of mature bindin), could simultaneously facilitate endo/lysosomal escape of various macromolecules and in turn promote cytosolic delivery [121]. 3.2.2. Cationic materials Another approach for endosomal escape is achieved by using cationic materials as carriers. Extensively studied cationic materials are usually cationic polymers and cationic lipids, most of which are rich in amine groups, making them positively charged [122]. Polyethyleneimine (PEI) has been one of the most representative and frequently used materials that exhibit endosomal escape. The capability of PEI to escape is driven by two mechanisms. PEI interacts with the endosomal membrane to make it porous, thus allowing the cargos to leak out into the cytosol. The other mechanism is the proton sponge effect of PEI [123]. The buffering action of PEI obtained by capturing hydrogen protons in the endo/lysosomes causes the osmotic swelling of this organelle, followed by endosome bursting and cargo release. Many cationic materials are able to interact with negatively charged genes or proteins to generate nanocomplexes, which inhibits their hydrolysis by lysosomal enzymes [124]. These nanocomplexes are usually positively charged, enhancing penetration into cells as a result of electronic interaction [115]. 3.3. Stimuli-sensitive NDDSs NDDSs that respond to environmental stimuli followed by intracellular delivery of therapeutics have been extensively explored. The stimuli can be external like the application of a magnetic field, ultrasound, heat, and light, or internal like changes in pH values, reduction conditions, and enzyme activity (Fig. 4) [125]. By allowing NDDSs to respond to stimulus, they are capable of delivering the cargos intracellularly, making further subcellular targeting possible. 3.3.1. Temperature sensitive Temperature is a widely studied stimulus. Compared with normal tissues, the temperature of tumor tissue is abnormal [126,127]. Tumor cells can be effectively killed by hyperthermia in the range of 40–43 °C due to the activation of apoptotic mechanism [128]. Additionally, the tumor can be heated artificially to increase the pore size of the perivascular site and quicken the blood flow, resulting in enhanced NPs accumulation in tumor. A commonly used temperature-sensitive material is poly(N-isopropylacrylamide), which possesses a lower critical solution temperature (LCST) in solution. Barhoumi et al. developed a photothermally targeted thermosensitive polymer-masked NPs based on poly(N-isopropylacrylamide)-co-polyacrylamide copolymer [129]. By conjugating this copolymer to the silica core-gold shell NPs, the peptide ligand decorated on the NPs surface can be masked at the physiological temperature. Upon NIR light trigger, the gold NPs convert it into heat, which causes the collapse of the polymer chain, followed by the exposure of the peptide ligand and binding the corresponding receptor integrin β1 on the cell surface. A relatively novel thermoresponsive carrier for intracellular delivery is fabricated by encapsulating ammonium bicarbonate (ABC) into liposomes [130–132]. Once triggered by hyperthermia (42 °C), ABC would decompose to generate CO2 bubbles, which destroyed the liposomal membrane and released the cargo. This bubble-generating liposomal system has the potential to observe the thermo-triggered drug release process as well, because ultrasound imaging could be enhanced as the generated CO2 bubbles are hyperechogenic. 3.3.2. pH sensitive The physiological pH in normal tissues is ∼7.4 while in tumors it ranges from 6 to 7 [133]. Lysosomes usually possess a pH of 4 to 5. Study on pH sensitive drug delivery systems dates back to the 1970s. The initial research was focused on liposomes that contain pH sensitive phospholipid components such as phosphatidylethanolamine, which induced destabilization of the vehicle upon acid trigger, followed by the release of the cargo [134]. Similar materials which are intrinsically sensitive to lower pH include PEI, poly(Llysine) (PLL), poly(amidoamine) (PAMAM) chitosan, etc., as discussed in Section 3.2.2. In addition to these lysosomal pH responsive materials, NPs prepared by polymers that respond to weak acid within tumor tissues have been successfully developed as well, such as poly-histidine (PHis) [135,136] and poly(methacryloyl sulfadimethoxine) (PSD) [137]. Another strategy for endowing a carrier 9

10

ER-Golgi network

Endo/lysosomes

Mitochondria

DOX

Nucleus

STxB-SN-38 prodrug

TPP-STxB conjugate

Porphyrin

Liposomes

MNPs

KLA peptide-modified liposomes DQAsomes TPCL NPs Mito-CIO Fe3O4 NPs

FA-PLL/amide-CPT conjugate FPAMAM/amide-CPT conjugate TAT-MSNs

PELEss-DA micelles

TAT-MSNs

Formulation

SN-38

IA

NH4HCO3

DOX

PTX

Ce6

CPT

Agent

Organelle

Table 1 Methods that are targeted to organelles for cancer therapy.

STxB binds to the glycolipid Gb3 receptor, then transported via endosomes and the GA to the ER by retrograde route; the disulfide bond can be broken triggered by GSH, and SN-38 would be released from the conjugate By conjugating STxB to the photocytotoxic porphyrin compound TPP(p-O-β-D-GluOH)3, the resulted TPP-STxB exhibited a high photocytotoxic activity against Gb3-expressing HeLa cells

MNPs targeted to EGFR-overexpressing cancer cells induce LMP when an AMF was applied, subsequently cathepsin B released from lysosomes can activate caspase-dependent death pathways and further induce cancer cell death Upon heating, NH4HCO3 in liposomes creates cavitation with CO2, which disrupted lysosomal membranes to release proteolytic enzymes into the cytosol By illuminating the MDR cancer cells that are preloading IA in the lysosomes, the cells would be eradicated based on photodestruction of lysosomes by IA and tumor cell lysis via formation of ROS

Mito-CIO quickly elevated the intracellular temperature of HeLa cells upon NIR irradiation due to their mitochondrial localization, enhancing the cytotoxicity in HeLa cells

DLCs and DQAsomes show high affinity with mitochondrial membranes; drugs delivered into mitochondria induces apoptosis of MDR cancer cells through mitochondrial signaling pathways by release of cytochrome c and activation of caspase-9 and −3

The intranuclear-accumulated Ce6 can generate ROS upon irradiation right inside nuclei to destroy DNA instantaneously

HeLa cells

TAT peptide binds importin α and β and subsequently targets the NPCs of cancer cells to facilitate intranuclear entry; DOX delivered into the nucleus causes DNA damage and/or inhibit topoisomerase to induce cell death PELEss-DA micelles with size-changeable ability can deshield PEI shell triggered by GSH to become PELA micelles that are smaller than nuclear pores, which directly delivers DOX into nucleus to overcome MDR Targeted charge-reversal conjugates carry CPT to the nucleus to cause cell death

[168] [169]

HeLa cells

[167] HT-29 cells

MDR A549/K1.5 and 2008/ MRP1 cells

[130,131]

[166]

MDA-MB-231 cells

HT1080 cells

[163] [164] [165]

[162]

PTX-resistant A549 cells MCF-7/ADR cells HeLa and HepG2 cells HeLa cells

[161]

[160]

[149]

[159]

[157,158]

Refs.

HeLa cells

SKOV-3 and MCF-7 cells

MCF-7/ADR cells

Cancer

Mechanisms

X. Guo, et al.

Progress in Materials Science 107 (2020) 100599

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

with a pH response is introducing via an acid-labile linkage. The linkages that respond to the tumor microenvironment include benzoic-imine [138] and 2,3-dimethylmaleic anhydride (DMMA) [139], and those that respond to the lysosomal pH include hydrazine [140,141], β-thiopropionate [142], imine [143,144], ortho ester [145,146], and ketal [147]. pH-sensitive NPs that react to the tumor acidic environment can be applied in tumor targeting by decorating DMMA on the amino groups of PEI [148] or PLL [149] to form charge-reversal carriers, while those responding to a lower pH have been widely used in endosomal escape as mentioned in Section 3.2.2 or subcellular targeted delivery, which will be further discussed in Section 4. 3.3.3. Reduction sensitive Redox potential differs in normal organs and tumors, as well as in intracellular and extracellular environments; this unique feature can also be introduced in intracellular drug delivery system. The reductive GSH concentration in tumor cells is 100–1000 times higher than that in blood due to the rapid plasmic enzymatic degradation; GSH in tumor tissue is also 100 times higher than in normal tissues [150]. In addition to the high concentration of GSH in cytosol, the endo/lysosomes also possess a reductive environment, which has the potential to assist NPs in endo/lysosomal escape [151]. In particular, the reductive environment in endo/ lysosomes is induced by a specific gene, γ-interferon-inducible lysosomal thiol reductase (GILT), which resembles cysteine rather than GSH [152]. Indeed, the lysosomal redox potential is modulated by cysteine such as thiols, which can be converted to disulfide bonds under oxidative condition. Similarly, disulfide bonds convert to thiols in the presence of reductants; this inter-conversion plays a key role in biological process maintaining the cellular native protein structure [153]. According to this mechanism, the reduction sensitive NPs that possess disulfide linkages can keep stable in physiological condition, while rapid thiol-disulfide exchange occurs triggered by reducing agents, promoting an intracellular drug delivery. Disulfide linkages can be introduced in the polymer structure, or between polymer and drugs to form conjugates [51,154,155]. Besides, other linkages including thioether bond, selenoether bond, diselenide bond, carbon bond and carbon-carbon bond can also be used for designing redox-responsive drug delivery systems [156]. 4. Organelle-specific targeting Certain organelles are associated with various diseases (Fig. 2), but therapeutic molecules often fail to reach their subcellular target after being internalized by the cell. Recent research has focused on organelle targeting as a new vector for drug-delivery. (Table 1). The necessity and strategies to target each organelle are addressed in this section. 4.1. Nuclear targeting The nucleus is usually the final target of a therapeutic after crossing a series of biological barriers. This important organelle regulates gene expression and exchange proteins with cytosol to control cellular process [170]. Fig. 5 shows the structure of a typical nucleus. The nuclear envelope with bilayer membrane is perforated by nuclear pore complexes (NPCs), and separates the nucleus from the cytosol. The nucleoporins (Nups), constituent proteins of NPCs, determine the NPCs' structural and functional aspects. The human nuclear genome possesses 3.2 × 109 base pairs, of which 2% codes for around 30,000 [171–173]. A variety of gene-associated diseases may occur if the 30,000 genes mutate. Therefore, the nucleus is the most significant subcellular organelle target in nanomedicine. There exist various therapeutic targets in the nucleus, including proteins, nuclear receptors, and DNA. Most anticancer drugs are DNA toxins, working on nuclear DNA to interfere with replication or inhibit topoisomerase to cause cell death. Thus, they must enter the nucleus to work. Even though NPs are internalized by the desired cells, it is still difficult to get access into the nucleus. It is reported that DNA less than 0.1% from cytoplasm were capable of nuclear entry [174]. Furthermore, multidrug resistance (MDR) has become an increasingly serious problem in cancer treatment. The P-glycoprotein (P-gp) overexpressed on MDR cell membrane can actively efflux various anticancer drugs outside of the cell. Therefore, it is an urgent task to design a vehicle that directly and

Fig. 5. Schematic representation of nuclear entry of the drug-loaded NPs through the nuclear pore complexes. 11

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

effectively transports the drugs into the nucleus to elicit their pharmacological responses and bypass P-gp. Drugs or NPs can move across the NPCs via both passive and active delivery. Passive delivery of cytoplasm to nucleus occurs by using ions, small molecules, and NPs of up to 9 nm [175]. Macromolecules larger than 39 nm (40 KDa) are not capable of nuclear entry either upon the occurrence of mitosis, which makes the nuclear membrane break down, or through active delivery. It has been reported that the nucleolin in the cytoplasm of breast cancer cells could actively and continuously migrated into the nucleus, endowing the nucleolin with natural nuclear targeting function [176]. Besides, nuclear entry of larger particles can rely on nuclear transport receptor-mediated delivery, resulting from the specific binding of the oligopeptide sequences to the receptors. These receptors are called nuclear localization signals (NLSs) [177], and they can ferry the vehicles via the NPCs. A series of functional NPs, including MSNPs [156,157,161], micelles [178–181], gold NPs [182–184], QDs [35,185,186], drug conjugates [187], peptide assemblies [188,189], etc., have been reported to be capable of nucleus targeting through conjugation of NLSs to the surface of the NPs. Among all the NLSs, the TAT peptide has been proved to be an efficient moiety for delivering NPs to cell nuclei through binding import receptors importin α and β [190,191]. The nuclear transport kinetics are highly dependent on the surface density of NLS. Maity et al. proved that QDs decorated with 83, 246, and 265 NLS peptides could effectively accumulate in the nucleus of HeLa cells with an average amount of 30.4%, 43.3%, and 49.0% of the intracellular QDs, respectively [186]. In contrast, less than 30% of the QDs entered the nucleus decorating with 63, 231, and 308 peptides. An alternative nuclear transport receptor-mediated delivery relies on the interaction between epidermal growth factor receptor (EGFR) and the karyopherin family of nuclear transport proteins [192–195]. EGFR that are particularly pronounced in cancer cells has a key role in nuclear delivery of NPs [196–199]. For instance, the nuclear EGFR is related to an increased resistance to chemotherapy [200] and inversely associated with survival in a couple of tumor [201,202]. Thus, nuclear delivery of genotoxic NPs through EGFR/karyopherin-mediated translocation has the potential to be applied for malignant tumor therapy. Methods of nuclear delivery of EGFR has been to modify with targeting moieties and exposure to stresses, for instance, through irradiation [203,204]. Yuan et al. developed EGFR-targeted NPs composed of a Fe3O4 core and a TiO2 shell (Fe3O4@TiO2) to facilitate cellular uptake and translocation into nucleus (Fig. 6) [205]. The Fe3O4@TiO2 NPs were modified with a couple of peptides to form peptide nanoconjugates (NCs). To select the NCs with the most affinity to EGFR and karyopherin-β, their EGFR-binding affinity as well as the NCs-EGFR/karyopherin-β interaction were investigated. The result indicated that an 11-amino-acid peptide bound with EGFR best, they called it B-loop peptide. By using X-ray fluorescence microscopy to image and quantify intracellular NCs directly, they confirmed the increased nuclear delivery of the EGFR-targeted B-loop NCs. They also showed that light activation of targeted NCs in cell nuclei significantly destroyed the double helix of DNA, probably due to the successful EGFR nuclear accumulation. An alternative strategy for nuclear delivery of NPs larger than nuclear pores is to destroy the integrity of nuclear membranes. For instance, Zhu et al. fabricated a nanoplatform for direct nuclear delivery via disrupting the nuclear membrane upon light irradiation [206]. The NPs consisting of PEG, polyamine-containing polyhedral oligomeric silsesquioxane and photosensitizer rose bengal could effectively disrupt the lysosomal structures through light-triggered singlet oxygen oxidation, followed by accumulating in the perinuclear region and augmenting the permeability of the nuclear membrane via inducing lipid peroxidation, allowing the influx of NPs. In another work, Houthaeve et al. provided a method based on vapor nanobubble (VNB)-mediated photoporation, which allows for temporarily perturbing the integrity of nuclear envelope (NE) [207]. Upon wide-field laser illumination, the gold NPs around the nucleus after endocytic uptake or electroporation-mediated delivery produced short-lived VNB, resulting in minute mechanical damage to the NE, subsequently creating small pores, which promoted nuclear entry of macromolecules that are too large to pass the NE. In this case, nuclear photoporation provides prospective potential for boosting nuclear delivery. In addition to endowing the NPs with an actively targeted ligand and disrupting the nuclear membrane, another promising strategy is to shrink the NPs on demand [208–212]. Limited by the diameter of nucleopores, NPs should be small enough for nuclear entry. However, NPs with such a small size of diameter usually have a short blood half-life as well as low tumor accumulation. To

Fig. 6. Interactions of EGFR-targeted NCs and non-targeted NCs with EGFR extracellularly (a) and intracellularly (b) [205]. 12

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

solve this dilemma, Fan et al. developed size-changeable NPs composed of oligolysine/iridium (III) (OLL-Ir) compound, which are sized 128 nm in physiological condition but dissociate into ∼18 nm complexes in a lysosomal environment [213]. After lysosomal escape, the OLL-Ir NPs can enter the nucleus freely due to their small size and the active targeting moiety of oligolysine segments. In our previous work, a size changeable polymeric micelle system with a dual-shell composed of mPEG-PLA-ss-PEI-DMMA (PELEss-DA) was developed for nuclear targeting (Fig. 7) [158]. These micelles could enter the cancer cells via electrostatic interaction resulting from the surface negative charge reversing to a positive charge in acidic tumor tissues. After cellular uptake and lysosomal location, the micelles become larger triggered by the excess H+ in the lysosomes and can easily escape through the proton sponge effect. PEI shells are deshielded by the cleavage of disulfide bond between poly(lactic acid) (PLA) and PEI under a high concentration of glutathione in the cytoplasm to generate new micelles that are smaller than nucleopores. Consequently, they can enter the nucleus and release cargo to cause cancer cell death. Li et al. developed a similar delivery system with size changeable and charge reversal capabilities [214]. A nuclear-targeting peptide, R8NLS, was conjugated to DOX to generate DoxR8NLS, which was further grafted to the N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer backbone via a hydrazone linkage to form P-DoxR8NLS. This cationic prodrug was then cross-linked with the anionic DMMA-modified HPMA to form a complex, PNV [214]. This nanosystem underwent a stepwise size reduction from ∼55 nm to ∼10 nm in tumor extracellular conditions with DOX release triggered by an acidic endo/ lysosomal environment. 4.2. Mitochondrial targeting Mitochondria are power-generated organelles with bilayer membrane, transforming oxygen and nutrients into adenosine triphosphate (ATP) and distribute energy throughout the cell [215]. This process is necessary for sustaining life and supporting organ function. Apart from ATP production, mitochondria participate in many other biological activities like management of reactive oxygen species (ROS) and biosynthesis of amino acid [216,217]. The abnormalities in the mitochondrial functions have been implicated in a series of neurodegenerative diseases. In addition, mitochondrial dysfunction is related to characteristics of tumor cells, such as impaired apoptosis [218,219]. Mutations in apoptotic-related genes usually lead to the MDR of tumor cells [220]. Hence, mitochondria are important action site for cancer therapy [221]. However, the highly complex structure of this organelle makes mitochondrial delivery a difficult task (Fig. 8). The inner mitochondrial membrane (IMM) is dense with a 3-fold higher protein/lipid ratio and membrane potential than those of cell membrane [222]. These properties assist mitochondria in sequestering various materials, making them difficult to penetrate [223]. A series of chemotherapeutics including paclitaxel [224], etoposide [225], betulinic acid [226], and ceramide [220] act on mitochondria to trigger apoptosis; delivering these drugs directly to mitochondria is a promising concept for cancer therapy. There have been several methods to achieve mitochondrial targeting. The first approach is implemented by using delocalized lipophilic cations (DLCs), which can reduce the activation energy related to deionization that must take place before its uptake through the hydrophobic IMM [227,228]. To increase the ionic radius, the cations are spread over a large surface area, resulting in an effective reduction in the interaction with surrounding water molecules [229]. These cations move across the IMM driven by their large negative potential, leading to a higher concentration of mitochondria-targeted conjugates in the matrix. Triphenylphosphonium (TPP) is the most common example of DLCs for mitochondrial targeting, and has a single positive charge stabilized over three phenyl groups [230]. Besides the delocalized positive charge, TPP possesses a large hydrophobic surface area, facilitating the interaction with IMM and subsequently permitting uptake into the mitochondrial matrix [231]. TPP can be conjugated to small molecules including natural/synthetic antioxidants [232–238], spin-traps [239] and superoxide probes [240,241] as well as hydrophilic polymers/amphiphilic copolymers for mitochondrial targeting [242–248]. A series of TPP-decorated NPs have been extensively studied for mitochondria-specific bio-imaging, diagnosis and therapy, such as Fe3O4 NPs [165], nanoprobe [249,250], carbon dots

Fig. 7. Illustration of size changeable polymeric micelles for cancer therapy by direct nuclear drug delivery [159]. 13

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

Fig. 8. Schematic description of mitochondrion structure and antitumor mechanism of mitochondrial targeted NPs. The therapeutics cause mitochondrial membrane damage and subsequently induces ROS (reactive oxygen species) generation, inhibits the production of ATP (adenosine triphosphate), and finally induces cancer cell death.

Fig. 9. (A) Illustration of occurrence of hyperthermia induced by NIR-triggered Mito-CIO. (B) CLSM images showing HeLa cells after incubation with different agents [165]. 14

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

[251,252], gold NPs [253,254], MSNPs [255], and polymer NPs [164,256,257]. Cho et al. developed a synergistic anticancer drug delivery nanocarrier based on TPP-conjugated poly(ε-caprolactone) (TPCL) [164]. Different morphologies of TPCL NPs including nanovesicles, nanofibers, and nanosheets have been formed depending on the preparation method. After simultaneous encapsulation of DOX and its hydrochloride, TPCL NPs exhibit enhanced mitochondrial uptake over free cargo, showing an improved tumor-killing activity. Jung et al. reported a TPP-modified and NIR-triggered system, Mito-CIO, loading with coumarin for fluorescence observation (Fig. 9) [165]. In vitro cell test showed that Mito-CIO entered the mitochondria while the non-targeted CIO were accumulated in the ER. Besides, a higher cytotoxicity was displayed by Mito-CIO over CIO under NIR irradiation. For the in vivo study, the NIR-triggered Mito-CIO significantly inhibited tumor growth compared to other groups. Both the in vitro and in vivo test indicated that hyperthermia in mitochondria were more efficient to induce tumor death than in ER. Mitochondrial targeted liposomes made of positively charged amphiphilic molecules can also be used as potential mitochondrial transporters. The most intensively investigated examples are DQAsomes, which are formed by dicationic mitochondriotropic compounds dequalinium [258]. DQAsomes can transport drugs and DNA to mitochondria due to their high mitochondrial affinity [259,260]. A novel liposome carrier for mitochondrial targeting is R8 moieties bearing MITO-Porter that fuse with mitochondrial membrane to get access [261–263]. The endocytic mechanism of this carrier was dependent on R8 density with high density resulting in micropinocytosis, while low density gave a CME-mediated uptake [264]. More recently, Bae et al. reported that DQA80s nanosomes consisting of dequalinium/1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)/ 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), showed increased cellular uptake and more cytotoxicity than DQAsomes [265]. The potential anticancer effect of DQA80s was probably due to the production of ROS via mitogen-activated protein kinase (MAPK) signaling pathways, loss of mitochondrial membrane potential, and the caspase-3 activation [265]. The third strategy for mitochondrial targeting is to use the mitochondrial targeting signal (MTS) peptide [223]. Although the Nterminal of MTS peptide are nonspecific, they still possess parallel characteristics to some extent [266]. MTS exhibited significant mitochondrial targeting ability by successfully delivering various therapeutics into this organelle [267–270]. However, due to the limitation of the polypeptide sequence length and requirement for cell penetrating vehicle like positively charged polymers or liposomes, using this strategy for mitochondrial targeting is uncommon [271]. In addition to the above-mentioned methods, it has

Fig. 10. Schematic illustration of the lysosomal targeted conjugate [288]. 15

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

been reported that the temperature of mitochondria was approximately 10 ℃ higher than other organelles [272], such a temperature difference can be exploited for thermo-responsive drug delivery to mitochondria [273]. By sequestering the therapeutics within the mitochondria, it is potentially to bypass the MDR, such as the membrane-associated Pgp pump [274–276]. This is because P-gp only pumps the drugs in the cytosol out of the cells [277]. The commonly used chemotherapeutics including DOX and platinum usually face the problem of P-gp even if delivered intracellular. The DOX-MITO-Porter fabricated by loading DOX into a MITO-Porter, could effectively deliver DOX to mitochondria of human DOX-resistant renal (OS-RC2) cells, consequently bypassed the P-gp and killed the cells [261]. In addition to P-gp, another protein that resist to platinum is nucleotide excision repair (NER) [278]. Mitochondria delivery of platinum to bypass NER is an effective strategy to kill platinumresistant cells [279–281]. Leukemia resistant to chlorambucil is mainly due to the overexpression of antiapoptotic proteins in Bcl2 family, which in turn inactivates apoptosis [282]. By activation of apoptosis, a mitochondrial targeted chlorambucil, mtCbl, successfully overcame the chlorambucil resistance of leukemic cells [283]. 4.3. Endo/lysosomal targeting Lysosomes are membrane-bound subcellular organelles found in nearly all animal-like eukaryotic cells. The hydrolytic enzymes in lysosomes can digest a couple of cellular materials, including proteins, nucleic acids and carbohydrates. Various pH-dependent NDDSs were designed to inhibit degradation in these compartments. The lysosomal-associated diseases like lysosomal storage diseases (LSDs) limit cell transport [284]. Enzyme replacement therapy is still the best treatment for LSDs [285,286]. Mannose6phosphate receptors (MPRs) can bind with the Golgi apparatus and then transported to the endo/lysosomal system [287], increase macrophage delivery of the enzyme in Gaucher’s disease (one LSD). Considering the overexpression in the endo/lysosomal compartments of a variety of cell types, MPRs are prospective targets for endo/lysosomal delivery. Hoogendoorn et al. described a strategy to develop an MPR ligand [288], and attached it to a cathepsin probe to generate a conjugate (Fig. 10). The MPR ligandmodified conjugate displayed an enhanced targeting activity; while the non-targeted control showed minimal lysosomal accumulation. In addition to MPRs, another receptor overexpressed on lysosomes is integral membrane protein LIMP-2, which has an affinity with β-glucocerebrosidase (β-GCase). This process depends on lysosomal pH that LIMP-2 binds to β-GCase in early endosomes while releasing it in more acidic late lysosomes [289]. Zhao et al. investigated the mechanism of pH-triggered combination and separation between LIMP-2 and β-GCase by using structural simulation [290]. The results indicated that a higher pH at both pH 5.5 and 6.5 lead to the transformation of Histidine 150 to threonine, which in turn locked LIMP-2, followed by binding to β-GCase. In contrast, a lower pH in late lysosomal compartment made Histidine 150 protonated, resulting in the change in conformation and separation between βGCase and LIMP-2. The major functions of lysosomes are degradation of macromolecules and release of enzymes intracellularly. The lysosomal protease cathepsin B that assists in maintaining the homeostasis of cells has been proved to take part in cell death pathways [291]. Upon particular trigger, the lysosomal membrane becomes permeable, followed by release of cathepsin B [292–294], which in turn activate the lysosomal pathway of apoptosis and further induce cell death [295]. Therefore, lysosomes are thought to be related to cancer cell progression. Magnetic nanoparticles (MNPs) can be used to selectively permeabilize cancer lysosomes. By applying an alternating magnetic field, Domenech et al. demonstrated that the EGFR-targeting MNPs effectively made the lysosomal membrane permeable after treatment with EGFR-expressed tumor cells [166]. Lysosomes are highly associated with drug resistance in cancer resulting from the sequestration of anticancer drugs in their acidic environment. For instance, Gotink et al. reported that the expression of lysosomal-associated membrane protein 1 (LAMP-1) and LAMP-2 in the resistant cells increased after being treated with sunitinib, an antiangiogenic tyrosine kinase inhibitor [296]. This finding proved that lysosomal sequestration could be a novel mechanism of sunitinib resistance. A series of therapeutic approaches for lysosomal targeting have attempted to overcome MDR. One approach has been to treat lysosomes with photosensitizers, which

Fig. 11. Illustration of Gb3-mediated retrograde delivery of drug-conjugated Shiga toxins for Golgi-ER network targeting. 16

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

can be triggered under irradiation and oxidize the lysosomal membrane, and finally release the therapeutics to avoid sequestration in lysosomes [297]. Adar et al. developed a novel light activating strategy to overcome MDR. By illuminating the imidazoacridinoneloaded cancer cells, lysosomes are destroyed and cells become sensitive to drugs. Therefore, lysosomal photodestruction is a promising approach to overcome MDR [167]. 4.4. Golgi apparatus (GA)/endoplasmic reticulum (ER) targeting GA and ER are closely associated organelles and they have similar structures. The membranes of ER form sacs, also called cisternae, where proteins are folded and transferred to GA [298]. GA and ER play a key role in the retrograde trafficking pathway, which bypasses the acidic pH and degrading enzymes in the endo/lysosomes [299]. There’s more than one mechanism involved in retrograde transport from GA to ER, such as coatomer protein complex-I (COPI)-dependent and COPI-independent pathways [300,301]. Overexpression of Golgi-associated proteins, pharmacological agents, and certain pathological changes can lead to profound alterations in the GA, which causes various neurodegenerative disorders [302–304]. In addition, ER dysfunction causes an unfolded protein response, which inhibits protein synthesis and protein folding. Both GA and ER are relevant to tumor and potentially targets for anticancer therapeutics [305]. Targeting of therapeutic agents to the ER-Golgi network has been applied in cancer diagnosis and therapy [299,306]. The therapeutics delivered to these intracellular compartments should enter the cells by CME-independent endocytosis for purpose of avoiding sequestration within endo/lysosomes. A series of strategies have been demonstrated for ER-Golgi targeting. One effective approach is to aim at the mammalian (or mechanistic) target of rapamycin (mTOR). mTOR is a phosphatidylinositol 3-kinase (PI3K) -related kinase that responds to the surrounding and cellular nutrition and energy conditions. mTOR signal can be activated via diverse stimuli, including nutrients, ATP and stress signals [307]. mTOR is highly correlated with cancer. For instance, activation of mTOR in certain tumor cells leads to PI3K inhibition. In contrast, inactivation of mTOR results in an increase in PI3K activity and the subsequent decrease of the antiproliferative effect of mTOR inhibition, killing the tumor [308]. It is worth mentioning that ER-Golgi network is main place where mTOR exists [309]. Hence, mTOR is an appealing therapeutic target in the ER-Golgi network. Another targeting strategy with significant potential is to conjugate a therapeutic moiety to protein toxins (Shiga or cholera toxins) that are capable of targeting the ER [306]. This approach takes advantage of retrograde transport [309], which is illustrated in Fig. 11. The Shiga toxin receptor, globotriaosylceramide (Gb3 or CD77), is highly expressed in most common tumors [310,311]. By conjugating to Shiga toxin subunit, topoisomerase inhibitors and imino sugar Nbutyldeoxynojiromycin have been successfully delivered to the ER [168,312]. 5. Conclusion and outlook The goal of nanoparticle-based drug delivery is to selectively deliver drugs to a target, which decreases side effects and increases therapeutic efficiency. It can accelerate the development of personalized and precision medicine. To fabricate nanocarriers with potential applications in clinical practices, an in-depth understanding of internalization mechanisms is important. NPs enter cells via different routes, which in turn determine where the NPs end up within the cell. The therapeutic outcome can be influenced by controlling the uptake route of NPs. In this review, several targeting strategies have been discussed, such as optimizing the physicochemical properties of NPs, decorating actively targeted ligands, or using cell penetrating peptides. Subcellular targeting remains a blooming research area. To achieve the goal of transporting drugs directly into organelles, new NPs requires a higher level of complexity. Despite this challenge, some efforts have been made in recent years to target organelles. Endosomes are the first type of intracellular compartment encountered by NPs, which they must escape in order to avoid destruction. Fusogenic peptides and cationic materials have been proven to successfully promote endosomal escape. To deliver the nanomedicines to the site of drug action (genes to nucleus, proapoptotic drugs to the mitochondria, enzymes for LSDs to the lysosomes, etc.), various approaches have been successfully developed. A detailed understanding of the intracellular trafficking of subcellular targeted nanomedicines, as well as their time-dependent fate and drug release profile inside the organelles still remains lacking. Although the task is challenging, sequential multistage targeting is the next step in NP design, and it is hopeful that this new paradigm will be successfully applied in clinic in the upcoming years. Acknowledgements This work was partially supported by the China National Funds for Distinguished Young Scientists (51725303), National Natural Science Foundation of China (Nos. 51373138, 21574105, 51603172), and Sichuan Province Youth Science and Technology Innovation Team (Grant No. 2016TD0026). Z.C. acknowledge the support from Society in Science, the Branco Weiss Fellowship, administered by ETH Zürich. References [1] [2] [3] [4]

Wilhelm S, Tavares AJ, Dai Q, Ohta S, Audet J, Dvorak HF, et al. Analysis of nanoparticle delivery to tumours. Nat Rev Mater 2016;1:16014. Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer 2017;17:20. Torchilin VP. Recent approaches to intracellular delivery of drugs and DNA and organelle targeting. Annu Rev Biomed Eng 2006;8:343–75. Torchilin VP, Khaw B-A, Weissig V. Intracellular targets for DNA delivery: nuclei and mitochondria. Somat Cell Mol Genet 2002;27:49–64.

17

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

[5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] [56] [57]

Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol 2015;33:941–51. Chauhan VP, Jain RK. Strategies for advancing cancer nanomedicine. Nat Mater 2013;12:958–62. Guo X, Wang L, Wei X, Zhou S. Polymer-based drug delivery systems for cancer treatment. J Polym Sci Pol Chem 2016;54:3525–50. Hubbell JA, Chilkoti A. Nanomaterials for drug delivery. Science 2012;337:303–5 Bourzac K. Nanotechnology: carrying drugs. Nature 2012;491:S58–60. Couvreur P. Nanoparticles in drug delivery: past, present and future. Adv Drug Deliv Rev 2013;65:21–3. Nicolas J, Mura S, Brambilla D, Mackiewicz N, Couvreur P. Design, functionalization strategies and biomedical applications of targeted biodegradable/biocompatible polymer-based nanocarriers for drug delivery. Chem Soc Rev 2013;42:1147–235. Maeda H, Nakamura H, Fang J. The EPR effect for macromolecular drug delivery to solid tumors: improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Drug Deliver Rev 2013;65:71–9. Huang C, Butler PJ, Tong S, Muddana HS, Bao G, Zhang S. Substrate stiffness regulates cellular uptake of nanoparticles. Nano Lett 2013;13:1611–5. Bareford LM, Swaan PW. Endocytic mechanisms for targeted drug delivery. Adv Drug Deliver Rev 2007;59:748–58. Han CZ, Juncadella IJ, Kinchen JM, Buckley MW, Klibanov AL, Dryden K, et al. Macrophages redirect phagocytosis by non-professional phagocytes and influence inflammation. Nature 2016;539:570–4. Freeman SA, Grinstein S. Phagocytosis: how macrophages tune their non-professional counterparts. Curr Biol 2016;26:R1279–82. Harashima H, Sakata K, Funato K, Kiwada H. Enhanced hepatic uptake of liposomes through complement activation depending on the size of liposomes. Pharm Res 1994;11:402–6. Koval M, Preiter K, Adles C, Stahl PD, Steinberg TH. Size of IgG-opsonized particles determines macrophage response during internalization. Exp Cell Res 1998;242:265–73. Conner SD, Schmid SL. Regulated portals of entry into the cell. Nature 2003;422:37–44. Rejman J, Oberle V, Zuhorn I, Hoekstra D. Size-dependent internalization of particles via the pathways of clathrin-and caveolae-mediated endocytosis. Biochem J 2004;377:159–69. Doherty GJ, McMahon HT. Mechanisms of endocytosis. Annu Rev Biochem 2009;78:857–902. Schnitzer JE, Liu J, Oh P. Endothelial caveolae have the molecular transport machinery for vesicle budding, docking, and fusion including VAMP, NSF, SNAP, annexins, and GTPases. J Biol Chem 1995;270:14399–404. Nabi IR. Cavin fever: regulating caveolae. Nat Cell Biol 2009;11:789–91. Carver LA, Schnitzer JE. Caveolae: mining little caves for new cancer targets. Nat Rev Cancer 2003;3:571–81. Rejman J, Conese M, Hoekstra D. Gene transfer by means of lipo-and polyplexes: role of clathrin and caveolae-mediated endocytosis. J Liposome Res 2006;16:237–47. Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov 2010;9:615–27. Mukherjee S, Ghosh RN, Maxfield FR. Endocytosis. Physiol Rev 1997;77:759–803. Swanson JA. Shaping cups into phagosomes and macropinosomes. Nat Rev Mol Cell Biol 2008;9:639–49. Sahay G, Alakhova DY, Kabanov AV. Endocytosis of nanomedicines. J Control Release 2010;145:182–95. Farokhzad OC, Langer R. Impact of nanotechnology on drug delivery. ACS Nano 2009;3:16–20. May RC, Machesky LM. Phagocytosis and the actin cytoskeleton. J Cell Sci 2001;114:1061–77. He C, Hu Y, Yin L, Tang C, Yin C. Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles. Biomaterials 2010;31:3657–66. Kulkarni SA, Feng S-S. Effects of particle size and surface modification on cellular uptake and biodistribution of polymeric nanoparticles for drug delivery. Pharm Res 2013;30:2512–22. Lovrić J, Bazzi HS, Cuie Y, Fortin GR, Winnik FM, Maysinger D. Differences in subcellular distribution and toxicity of green and red emitting CdTe quantum dots. J Mol Med 2005;83:377–85. Huang K, Ma H, Liu J, Huo S, Kumar A, Wei T, et al. Size-dependent localization and penetration of ultrasmall gold nanoparticles in cancer cells, multicellular spheroids, and tumors in vivo. ACS Nano 2012;6:4483–93. Tang PS, Sathiamoorthy S, Lustig LC, Ponzielli R, Inamoto I, Penn LZ, et al. The role of ligand density and size in mediating quantum dot nuclear transport. Small 2014;10:4182–92. Gratton SE, Ropp PA, Pohlhaus PD, Luft JC, Madden VJ, Napier ME, et al. The effect of particle design on cellular internalization pathways. Proc Natl Acad Sci USA 2008;105:11613–8. Decuzzi P, Godin B, Tanaka T, Lee S-Y, Chiappini C, Liu X, et al. Size and shape effects in the biodistribution of intravascularly injected particles. J Control Release 2010;141:320–7. Vácha R, Martinez-Veracoechea FJ, Frenkel D. Receptor-mediated endocytosis of nanoparticles of various shapes. Nano Lett 2011;11:5391–5. Meng H, Yang S, Li Z, Xia T, Chen J, Ji Z, et al. Aspect ratio determines the quantity of mesoporous silica nanoparticle uptake by a small GTPase-dependent macropinocytosis mechanism. ACS Nano 2011;5:4434–47. Champion JA, Mitragotri S. Role of target geometry in phagocytosis. Proc Natl Acad Sci 2006;103:4930–4. Champion JA, Mitragotri S. Shape induced inhibition of phagocytosis of polymer particles. Pharm Res 2009;26:244–9. Lin S-Y, Hsu W-H, Lo J-M, Tsai H-C, Hsiue G-H. Novel geometry type of nanocarriers mitigated the phagocytosis for drug delivery. J Control Release 2011;154:84–92. Hu X, Hu J, Tian J, Ge Z, Zhang G, Luo K, et al. Polyprodrug amphiphiles: hierarchical assemblies for shape-regulated cellular internalization, trafficking, and drug delivery. J Am Chem Soc 2013;135:17617–29. Alexis F, Pridgen E, Molnar LK, Farokhzad OC. Factors affecting the clearance and biodistribution of polymeric nanoparticles. Mol Pharm 2008;5:505–15. Guo X, Shi C, Yang G, Wang J, Cai Z, Zhou S. Dual-responsive polymer micelles for target-cell-specific anticancer drug delivery. Chem Mater 2014;26:4405–18. Zhou Q, Shao S, Wang J, Xu C, Xiang J, Piao Y, et al. Enzyme-activatable polymer-drug conjugate augments tumour penetration and treatment efficacy. Nat Nanotechnol 2019;14:799–809. Liu X, Huang N, Li H, Jin Q, Ji J. Surface and size effects on cell interaction of gold nanoparticles with both phagocytic and nonphagocytic cells. Langmuir 2013;29:9138–48. Sudimack J, Lee RJ. Targeted drug delivery via the folate receptor. Adv Drug Deliver Rev 2000;41:147–62. Xia W, Low PS. Folate-targeted therapies for cancer. J Med Chem 2010;53:6811–24. Guo X, Shi C, Wang J, Di S, Zhou S. pH-triggered intracellular release from actively targeting polymer micelles. Biomaterials 2013;34:4544–54. Shi C, Guo X, Qu Q, Tang Z, Wang Y, Zhou S. Actively targeted delivery of anticancer drug to tumor cells by redox-responsive star-shaped micelles. Biomaterials 2014;35:8711–22. Choi CHJ, Alabi CA, Paul W, Davis ME. Mechanism of active targeting in solid tumors with transferrin-containing gold nanoparticles. Proc Natl Acad Sci USA 2010;107:1235–40. Sahoo SK, Ma W, Labhasetwar V, Sahoo SK, Ma W, Labhasetwar V. Efficacy of transferrin-conjugated paclitaxel-loaded nanoparticles in a murine model of prostate cancer. Int J Cancer 2004;112:335–40. Roy D, Berguig GY, Ghosn B, Lane D, Braswell S, Stayton PS, et al. Synthesis and characterization of transferrin-targeted chemotherapeutic delivery systems prepared via RAFT copolymerization of high molecular weight PEG macromonomers. Polym Chem 2014;5:1791–9. Sarisozen C, Abouzeid AH, Torchilin VP. The effect of co-delivery of paclitaxel and curcumin by transferrin-targeted PEG-PE-based mixed micelles on resistant ovarian cancer in 3-D spheroids and in vivo tumors. Eur J Pharm Biopharm 2014;88:539–50. Wilson DS, Szostak JW. In vitro selection of functional nucleic acids. Annu Rev Biochem 1999;8:611–47. Zeyu X, Farokhzad OC. Aptamer-functionalized nanoparticles for medical applications: challenges and opportunities. ACS Nano 2012;6:3670–6.

18

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

[58] Gu FX, Karnik R, Wang AZ, Alexis F, Levy-Nissenbaum E, Hong S, et al. Targeted nanoparticles for cancer therapy. Nano Today 2007;2:14–21. [59] Guizhi Z, Jing Z, Erqun S, Michael D, Kejing Z, Chen L, et al. Self-assembled, aptamer-tethered DNA nanotrains for targeted transport of molecular drugs in cancer theranostics. Proc Natl Acad Sci USA 2013;110:7998–8003. [60] Wang CH, Kang ST, Lee YH, Luo YL, Huang YF, Yeh CK. Aptamer-conjugated and drug-loaded acoustic droplets for ultrasound theranosis. Biomaterials 2012;33:1939–47. [61] Bagalkot V, Gao X. siRNA-Aptamer chimeras on nanoparticles: preserving targeting functionality for effective gene silencing. ACS Nano 2011;5:8131–9. [62] Ruowen W, Guizhi Z, Lei M, Yan X, Haibin M, Mao Y, et al. Automated modular synthesis of aptamer-drug conjugates for targeted drug delivery. J Am Chem Soc 2014;136:2731–4. [63] Von MM, Adams GP, Weiner LM. Monoclonal antibody therapy for cancer. Annu Rev Med 2003;54:343–69. [64] Adams GP, Weiner LM, Adams GP, Weiner LM. Monoclonal antibody therapy of cancer. Nat Biotechnol 2005;23:1147–57. [65] Brannon-Peppas L, Blanchette JO. Corrigendum to “Nanoparticle and targeted systems for cancer therapy”. Adv Drug Deliver Rev 2009;61:364. [66] Weiner LM, Rishi S, Shangzi W. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat Rev Immunol 2010;10:317–27. [67] Weinberg WC, Frazier-Jessen MR, Wu WJ, Weir A, Hartsough M, Keegan P, et al. Development and regulation of monoclonal antibody products: challenges and opportunities. Cancer Metast Rev 2005;24:569–84. [68] Allen TM. Ligand-targeted therapeutics in anticancer therapy. Nat Rev Cancer 2002;2:750–63. [69] Simard P, Leroux JC. In vivo evaluation of pH-sensitive polymer-based immunoliposomes targeting the CD33 antigen. Mol Pharm 2010;7:1098–107. [70] Renee B, Prendergast JKA, Goldstein NI. Identification of cancer targets and therapeutics using phage display. Curr Opin Drug Disc 2006;9:363–9. [71] Desgrosellier JS, Cheresh DA, Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer 2010;10:9–22. [72] Kim J, Nam HY, Choi JW, Yun C-O, Kim SW. Efficient lung orthotopic tumor-growth suppression of oncolytic adenovirus complexed with RGD-targeted bioreducible polymer. Gene Ther 2014;21:476–83. [73] Xiaoying Y, Yi W, Jiao L, Jiaxing Y, Cenglin X, Lina L, et al. Angiopep-conjugated electro-responsive hydrogel nanoparticles: therapeutic potential for epilepsy. Angew Chem Int Edit 2014;53:12436–40. [74] Andaloussi SE, Hassane FS, Boisguerin P, Sillard R, Langel Ü, Lebleu B. Cell-penetrating peptides. Trends Pharmacol Sci 2000;21:99–103. [75] Deshayes S, Morris MC, Divita G, Heitz F. Cell-penetrating peptides: tools for intracellular delivery of therapeutics. CMLS-Cell Mol Life Sci 2005;62:1839–49. [76] Snyder EL, Dowdy SF. Recent advances in the use of protein transduction domains for the delivery of peptides, proteins and nucleic acids in vivo. Drug Del 2005;2:43–51. [77] Michelle S, Mudit T, Mauro G, Bernard L, Eric V. Different mechanisms for cellular internalization of the HIV-1 Tat-derived cell penetrating peptide and recombinant proteins fused to Tat. Eur J Biochem 2002;269:494–501. [78] Jones SW, Christison R, Bundell K, Voyce CJ, Brockbank SM, Newham P, Lindsay MA. Characterisation of cell-penetrating peptide-mediated peptide delivery. Brit J Pharmacol 2005;145:1093–102. [79] Lindgren M, Langel Ü. Classes and prediction of cell-penetrating peptides. Meth Mol Biol 2011;683:3–19 Matjaz Z, Ulo L. Cell-penetrating peptides: mechanism and kinetics of cargo delivery. Adv Drug Deliver Rev 2005;57:529–45. [80] Wender PA, Galliher WC, Goun EA, Jones LR, Pillow TH. The design of guanidinium-rich transporters and their internalization mechanisms. Adv Drug Deliver Rev 2008;60:452–72. [81] Polanco C, Samaniego JL, Castañón-González JA, Buhse T, Sordo ML. Characterization of a possible uptake mechanism of selective antibacterial peptides. Acta Biochim Pol 2013;60:629–33. [82] Wu X, Gehring W. Cellular uptake of the Antennapedia homeodomain polypeptide by macropinocytosis. Biochem Bioph Res Co 2014;443:1136–40. [83] Koppelhus U, Awasthi SK, Zachar V, Holst HU, Ebbesen P, Nielsen PE. Cell-dependent differential cellular uptake of PNA, peptides, and PNA-peptide conjugates. Antisense Nucleic A 2002;12:51–63. [84] Young SC, Allan ED. Cell penetrating peptides and the mechanisms for intracellular entry. Curr Pharm Biotechno 2014;15:192–9. [85] Fawell S, Seery J, Daikh Y, Moore C, Chen LL, Pepinsky B, et al. Tat-mediated delivery of heterologous proteins into cells. Proc Natl Acad Sci USA 1994;91:664–8. [86] Guo X, Wang L, Duval K, Fan J, Zhou S, Chen Z. Dimeric drug polymeric micelles with acid-active tumor targeting and FRET-traceable drug release. Adv Mater 2018;30:1705436. [87] Josephson L, Tung CH, Moore A, Weissleder R. High-efficiency intracellular magnetic labeling with novel superparamagnetic-Tat peptide conjugates. Bioconjugate Chem 1999;10:186–91. [88] Wunderbaldinger P, Josephson L, Weissleder R. Tat peptide directs enhanced clearance and hepatic permeability of magnetic nanoparticles. Bioconjugate Chem 2002;13:264–8. [89] Torchilin VP, Rammohan R, Weissig V, Levchenko TS. TAT peptide on the surface of liposomes affords their efficient intracellular delivery even at low temperature and in the presence of metabolic inhibitors. Proc Natl Acad Sci USA 2001;98:8786–91. [90] Fretz MM, Koning GA, Mastrobattista E, Jiskoot W, Storm G. OVCAR-3 cells internalize TAT-peptide modified liposomes by endocytosis. BBA-Biomembranes 2004;1665:48–56. [91] Todorova N, Chiappini C, Mager M, Simona B, Patel II, Stevens MM, et al. Surface presentation of functional peptides in solution determines cell internalization efficiency of TAT conjugated nanoparticles. Nano Lett 2014;14:5229–37. [92] Ming Y, Xiao Y, Tian Y, Zhou S. Cell-membrane penetration of Tat-conjugated polymeric micelles: effect of Tat coating density. Macromol Biosci 2019;19:1800364. [93] Derossi D, Joliot AH, Chassaing G, Prochiantz A. The third helix of the Antennapedia homeodomain translocates through biological membranes. J Biol Chem 1994;269:10444–50. [94] Thorén PEG, Daniel P, Petter I, Mattias GR, Agneta O, Bengt N. Uptake of analogs of penetratin, Tat(48–60) and oligoarginine in live cells. Biochem Bioph Res Co 2003;307:100–7. [95] Eiriksdotur E, Mager I, Lehto T. Cellular internalization kinetics of (luciferin-)cell-penetrating peptide conjugates. Bioconjugate Chem 2010;21:1662–72. [96] Amand HL, Fant K, Nordén B, Ek E. Stimulated endocytosis in penetratin uptake: effect of arginine and lysine. Biochem Bioph Res Co 2008;371:621–5. [97] Chen-Yu J, Diane D, Fabienne B, Alves ID, Gérard C, Sandrine S. Translocation and endocytosis for cell-penetrating peptide internalization. J Biol Chem 2009;284:33957–65. [98] Watkins CL, Dirk S, Shiroh F, Jones AT. Low concentration thresholds of plasma membranes for rapid energy-independent translocation of a cell-penetrating peptide. Biochem J 2009;420:179–89. [99] Letoha T, Keller-Pintér A, Kusz E, Kolozsi C, Bozsó Z, Tóth G, et al. Cell-penetrating peptide exploited syndecans. BBA-Biomembranes 2010;1798:2258–65. [100] Hao Y, Shan L, Huawei C, Lin W, Shengfu L, Youping L, et al. Chondroitin sulfate as a molecular portal that preferentially mediates the apoptotic killing of tumor cells by penetratin-directed mitochondria-disrupting peptides. J Biol Chem 2010;285:25666–76. [101] Binder H, Lindblom G. Charge-dependent translocation of the Trojan peptide penetratin across lipid membranes. Biophys J 2003;85:982–95. [102] Magzoub M, Kilk K, Eriksson LEG, Langel Ü, Gräslund A. Interaction and structure induction of cell-penetrating peptides in the presence of phospholipid vesicles. BBA-Biomembranes 2001;1512:77–89. [103] Wenyi Z, Smith SO. Mechanism of penetration of Antp(43–58) into membrane bilayers. Biochemistry 2005;44:10110–8. [104] Xia H, Gao X, Gu G, Liu Z, Hu Q, Tu Y, et al. Penetratin-functionalized PEG-PLA nanoparticles for brain drug delivery. Int J Pharm 2012;436:840–50. [105] Futaki S, Suzuki T, Ohashi W, Yagami T, Tanaka S, Ueda K, et al. Arginine-rich Peptides. J Biol Chem 2001;276:5836–40. [106] Florent P, Masamichi N, Toshihide T, Shiroh F, Lazar AN, Coleman AW, et al. Anionic fullerenes, calixarenes, coronenes, and pyrenes as activators of oligo/ polyarginines in model membranes and live cells. J Am Chem Soc 2005;127:1114–5. [107] Takeuchi T, Kosuge M, Tadokoro A, Sugiura Y, Nishi M, Kawata M, et al. Direct and rapid cytosolic delivery using cell-penetrating peptides mediated by

19

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

pyrenebutyrate. ACS Chem Biol 2006;1:299–303. [108] Guterstam P, Fhirose M. Elucidating cell-penetrating peptide mechanisms of action for membrane interaction, cellular uptake, and translocation utilizing the hydrophobic counter-anion pyrenebutyrate. BBA-Biomembranes 2009;1788:2509–17. [109] Khalil IA, Kogure K, Futaki S, Hama S, Akita H, Ueno M, et al. Octaarginine-modified multifunctional envelope-type nanoparticles for gene delivery. Gene Ther 2007;14:682–9. [110] Hayashi Y, Yamauchi J, Khalil IA, Kajimoto K, Akita H, Harashima H. Cell penetrating peptide-mediated systemic siRNA delivery to the liver. Int J Pharm 2011;419:308–13. [111] Biswas S, Dodwadkar NS, Deshpande PP, Parab S, Torchilin VP. Surface functionalization of doxorubicin-loaded liposomes with octa-arginine for enhanced anticancer activity. Eur J Pharm Biopharm 2013;84:517–25. [112] Dubikovskaya EA, Thorne SH, Pillow TH, Contag CH, Wender PA. Overcoming multidrug resistance of small-molecule therapeutics through conjugation with releasable octaarginine transporters. Proc Natl Acad Sci USA 2008;105:12128–33. [113] El-Sayed A, Khalil I, Kogure K, Futaki S, Harashima H. Octaarginine-and octalysine-modified nanoparticles have different modes of endosomal escape. J Biol Chem 2008;283:23450–61. [114] Dunn WA. Autophagy and related mechanisms of lysosome-mediated protein degradation. Trends Cell Biol 1994;4:139–43. [115] Dominska M, Dykxhoorn DM. Breaking down the barriers: siRNA delivery and endosome escape. J Cell Sci 2010;123:1183–9. [116] Shete HK, Prabhu RH, Patravale VB. Endosomal escape: a bottleneck in intracellular delivery. J Nanosci Nanotechnol 2014;14:460–74. [117] Hatakeyama H, Ito E, Akita H, Oishi M, Nagasaki Y, Futaki S, et al. A pH-sensitive fusogenic peptide facilitates endosomal escape and greatly enhances the gene silencing of siRNA-containing nanoparticles in vitro and in vivo. J Control Release 2009;139:127–32. [118] Endoh T, Ohtsuki T. Cellular siRNA delivery using cell-penetrating peptides modified for endosomal escape. Adv Drug Deliver Rev 2009;61:704–9. [119] Nakase I, Futaki S. Combined treatment with a pH-sensitive fusogenic peptide and cationic lipids achieves enhanced cytosolic delivery of exosomes. Sci Rep 2015;5:10112–24. [120] Nishimura Y, Takeda K, Ezawa R, Ishii J, Ogino C, Kondo A. A display of pH-sensitive fusogenic GALA peptide facilitates endosomal escape from a Bionanocapsule via an endocytic uptake pathway. Nanobiotech 2014;12:2248–52. [121] Niikura K, Horisawa K, Doi N. A fusogenic peptide from a sea urchin fertilization protein promotes intracellular delivery of biomacromolecules by facilitating endosomal escape. J Control Release 2015;212:85–93. [122] Zhang S, Zhao B, Jiang H, Wang B, Ma B. Cationic lipids and polymers mediated vectors for delivery of siRNA. J Control Release 2007;123:1. [123] Neu M, Fischer D, Kissel T. Recent advances in rational gene transfer vector design based on poly(ethylene imine) and its derivatives. J Gene Med 2005;7:992–1009. [124] Lächelt U, Wagner E. Nucleic acid therapeutics using polyplexes: a journey of 50 years (and beyond). Chem Rev 2015;115:11043–78. [125] Torchilin VP. Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery. Nat Rev Drug Discov 2014;13:813–27. [126] Jayasundar R, Singh VP. In vivo temperature measurements in brain tumors using proton MR spectroscopy. Neurol India 2002;50:436–9. [127] Yahara T, Koga T, Yoshida S, Nakagawa S, Deguchi H, Shirouzu K. Relationship between microvessel density and thermographic hot areas in breast cancer. Surg Today 2003;33:243–8. [128] Issels R. Hyperthermia adds to chemotherapy. Eur J Cancer 2008;44:2546–54. [129] Barhoumi A, Wang W, Zurakowski D, Langer RS, Kohane DS. Photothermally targeted thermosensitive polymer-masked nanoparticles. Nano Lett 2014;14:3697–701. [130] Chen KJ, Liang HF, Chen HL, Wang Y, Cheng PY, Liu HL, et al. A thermoresponsive bubble-generating liposomal system for triggering localized extracellular drug delivery. ACS Nano 2013;7:438–46. [131] Chung MF, Chen KJ, Liang HF, Liao ZX, Chia WT, Xia Y, et al. A liposomal system capable of generating CO_2 bubbles to induce transient cavitation, lysosomal rupturing, and cell necrosis. Angew Chem 2012;51:10236–40. [132] Cheng Y, Zou T, Dai M, He XY, Peng N, Wu K, et al. Doxorubicin loaded tumor-triggered targeting ammonium bicarbonate liposomes for tumor-specific drug delivery. Colloid Surface B 2019;178:263–8. [133] Fleige E, Quadir MA, Haag R. Stimuli-responsive polymeric nanocarriers for the controlled transport of active compounds: concepts and applications. Adv Drug Deliver Rev 2012;64:866–84. [134] Wang CY, Huang L. Highly efficient DNA delivery mediated by pH-sensitive immunoliposomes. Biochemistry-US 1989;28:9508–14. [135] Qiu L, Qiao M, Chen Q, Tian C, Long M, Wang M, et al. Enhanced effect of pH-sensitive mixed copolymer micelles for overcoming multidrug resistance of doxorubicin. Biomaterials 2014;35:9877–87. [136] Zhang X, Chen D, Ba S, Zhu J, Zhang J, Hong W, et al. Poly(l-histidine) based triblock copolymers: pH induced reassembly of copolymer micelles and mechanism underlying endolysosomal escape for intracellular delivery. Biomacromolecules 2014;15:4032–45. [137] Sara B, Marian V-C, Chiara B, Michela B, Stefano S. pH-sensitive stearoyl-PEG-poly(methacryloyl sulfadimethoxine) decorated liposomes for the delivery of gemcitabine to cancer cells. Eur J Pharm Biopharm 2014;88:670–82. [138] Minghui Z, Junjie L, Shizhong L, Zhishen G. Dual pH-triggered multistage drug delivery systems based on host-guest interaction-associated polymeric nanogels. Chem Commun 2014;50:7824–7. [139] Wang Y, Min SS, Levinson NS, Sung H-W, Xia Y. Stimuli-responsive materials for controlled release of theranostic agents. Adv Funct Mater 2014;24:4206–20. [140] Cao Y, Li Y, Hu XY, Zou X, Xiong S, Lin C, et al. Supramolecular nanoparticles constructed by DOX-based prodrug with water-soluble Pillar[6]arene for selfcatalyzed rapid drug release. Chem Mater 2015;27:1110–9. [141] Wei X, Wang Y, Xiong X, Guo X, Zhang L, Zhang X, et al. Codelivery of a π-π stacked dual anticancer drug combination with nanocarriers for overcoming multidrug resistance and tumor metastasis. Adv Func Mater 2016;26:8266–80. [142] Wu W-X, Qu L, Liu B-Y, Zhang W-W, Wang N, Yu X-Q. Lipase-catalyzed synthesis of acid-degradable poly(β-thioether ester) and poly(β-thioether ester-colactone) copolymers. Polymer 2015;59:187–93. [143] Nair JB, Saswat M, Surajit G, Maiti KK. Novel lysosome targeted molecular transporter built on a guanidinium-poly-(propylene imine) hybrid dendron for efficient delivery of doxorubicin into cancer cells. Chem Commun 2015;51:2403–6. [144] Deng H, Zhang Y, Wang X, Cao Y, Liu J, Deng L, et al. Balancing the stability and drug release of polymer micelles by the coordination of dual-sensitive cleavable bonds in cross-linked core. Acta Biomater 2015;11:126–36. [145] Ji R, Cheng J, Yang T, Song CC, Li L, Du FS, et al. Shell-sheddable, pH-sensitive supramolecular nanoparticles based on ortho ester-modified cyclodextrin and adamantyl PEG. Biomacromolecules 2014;15:3531–9. [146] Ji R, Cheng J, Song CC, Du FS, Liang DH, Li ZC. Acid-sensitive polypseudorotaxanes based on ortho ester-modified cyclodextrin and pluronic F-127. ACS Macro Lett 2015;4:65–9. [147] Chen D, Sun J. In vitro and in vivo evaluation of PEG-conjugated ketal-based chitosan micelles as pH-sensitive carriers. Polym Chem 2014;6:998–1004. [148] Peisheng X, Edward AVK, Yihong Z, William JM, Maciej R, Shen YQ. Targeted charge-reversal nanoparticles for nuclear drug delivery. Angew Chem Int Ed 2007;46:4999–5002. [149] Zhou Z, Shen Y, Tang J, Fan M, Kirk EAV, Murdoch WJ, et al. Charge-reversal drug conjugate for targeted cancer cell nuclear drug delivery. Adv Funct Mater 2009;19:3580–9. [150] Torchilin V. Multifunctional and stimuli-sensitive pharmaceutical nanocarriers. Eur J Pharm Biopharm 2009;71:431–44. [151] Ru C, Fang F, Fenghua M, Chao D, Jan F, Zhiyuan Z. Glutathione-responsive nano-vehicles as a promising platform for targeted intracellular drug and gene delivery. J Control Release 2011;152:2–12. [152] Li J, Huo M, Wang J, Zhou J, Mohammad JM, Zhang Y, et al. Redox-sensitive micelles self-assembled from amphiphilic hyaluronic acid-deoxycholic acid conjugates for targeted intracellular delivery of paclitaxel. Biomaterials 2012;33:2310–20. [153] Roy D, Cambre JN, Sumerlin BS. Future perspectives and recent advances in stimuli-responsive materials. Prog Polym Sci 2010;35:278–301.

20

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

[154] Wang Y, Wei G, Zhang X, Xu F, Xiong X, Zhou S. A step-by-step multiple stimuli-responsive nanoplatform for enhancing combined chemo-photodynamic therapy. Adv Mater 2017;29:1605357. [155] Wang J, Yang G, Guo X, Tang ZM, Zhong ZD, Zhou SB. Redox-responsive polyanhydride micelles for cancer therapy. Biomaterials 2014;35:3080–90. [156] Sun B, Luo C, Zhang X, Guo M, Sun M, Yu H, et al. Probing the impact of sulfur/selenium/carbon linkages on prodrug nanoassemblies for cancer therapy. Nat Commun 2019;10:3211. [157] Pan L, He Q, Liu J, Chen Y, Ma M, Zhang L, et al. Nuclear-targeted drug delivery of TAT peptide-conjugated monodisperse mesoporous silica nanoparticles. J Am Chem Soc 2012;134:5722–5. [158] Qianjun H, Jianlin S. MSN anti-cancer nanomedicines: chemotherapy enhancement, overcoming of drug resistance, and metastasis inhibition. Adv Mater 2014;26:391–411. [159] Guo X, Wei X, Jing Y, Zhou S. Size changeable nanocarriers with nuclear targeting for effectively overcoming multidrug resistance in cancer therapy. Adv Mater 2015;27:6450–6. [160] Shen Y. Charge-reversal polyamidoamine dendrimer for cascade nuclear drug delivery. Nanomedicine 2010;5:1205–17. [161] Pan L, Liu J, Shi J. Intranuclear photosensitizer delivery and photosensitization for enhanced photodynamic therapy with ultralow irradiance. Adv Funct Mater 2014;24:7318–27. [162] Lei J, Li L, He X, Yi Q, He B, Cao J, et al. Overcoming drug-resistant lung cancer by paclitaxel loaded dual-functional liposomes with mitochondria targeting and pH-response. Biomaterials 2015;52:126–39. [163] Wang H, Yin H, Yan F, Sun M, Du L, Peng W, et al. Folate-mediated mitochondrial targeting with doxorubicin-polyrotaxane nanoparticles overcomes multidrug resistance. Oncotarget 2015;6:2827–42. [164] Dong YC, Cho H, Kwon K, Yu M, Lee E, Kang MH, et al. Triphenylphosphonium-conjugated poly(ε-caprolactone)-based self-assembled nanostructures as nanosized drugs and drug delivery carriers for mitochondria-targeting synergistic anticancer drug delivery. Adv Funct Mater 2015;25:5479–91. [165] Jung HS, Han J, Lee J, Lee JH, Choi J, Kweon H, et al. Enhanced NIR radiation-triggered hyperthermia by mitochondrial targeting. J Am Chem Soc 2015;137:3017–23. [166] Maribella D, Ileana M-B, Madeline T-L, Carlos R. Lysosomal membrane permeabilization by targeted magnetic nanoparticles in alternating magnetic fields. ACS Nano 2013;7:5091–101. [167] Adar Y, Stark M, Bram EE, Nowak-Sliwinska P, Van DBH, Szewczyk G, Sarna T, Skladanowski A. Imidazoacridinone-dependent lysosomal photodestruction: a pharmacological Trojan horse approach to eradicate multidrug-resistant cancers. Cell Death Dis 2012;3:e293. [168] Alaoui AE, Schmidt F, Amessou M, Sarr M, Decaudin D, Florent JC, et al. Shiga toxin-mediated retrograde delivery of a topoisomerase I inhibitor prodrug. Angew Chem Int Ed 2007;46:6469–72. [169] Amessou M, Carrez D, Patin D, Sarr M, Grierson DS, Croisy A, et al. Retrograde delivery of photosensitizer (TPPp-O-β-GluOH)3 selectively potentiates its photodynamic activity. Bioconjugate Chem 2008;19:532–8. [170] Cremer T, Cremer C. Chromosome territories, nuclear architecture and gene regulation in mammalian cells. Nat Rev Genet 2001;2:292–301. [171] Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, et al. Initial sequencing and analysis of the human genome. Nature 2001;409:860–921. [172] Smith MY, Evans CA, Holt RA. The sequence of the human genome. Science 2001;291:1304–51. [173] Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 2009;10:R25. [174] Zanta MA, Belguise-Valladier P, Behr JP. Gene delivery: a single nuclear localization signal peptide is sufficient to carry DNA to the cell nucleus. P Natl Acad Sci 1999;96:91–6. [175] Kau TR, Way JC, Silver PA. Nuclear transport and cancer: from mechanism to intervention. Nat Rev Cancer 2004;4:106–17. [176] Li X, Wu X, Yang H, Li L, Ye Z, Rao Y. A nuclear targeted Dox-aptamer loaded liposome delivery platform for the circumvention of drug resistance in breast cancer. Biomed Pharmacother 2019;117:109072. [177] Cokol M, Nair R, Rost B. Finding nuclear localization signals. EMBO Rep 2000;1:411–5. [178] Erlei J, Zhang B, Sun X, Ma X, Sun Q, Tang J, et al. Acid-active cell-penetrating peptides for in vivo tumor-targeted drug delivery. J Am Chem Soc 2013;135:933–40. [179] Yu J, Xie X, Xu X, Zhang L, Zhou X, Yu H, et al. Development of dual ligand-targeted polymeric micelles as drug carriers for cancer therapy in vitro and in vivo. J Mater Chem B 2014;2:2114–26. [180] Wang GH, Cai YY, Du JK, Li L, Li Q, Yang HK, et al. TAT-conjugated chitosan cationic micelle for nuclear-targeted drug and gene co-delivery. Colloid Surface B 2018;162:326–34. [181] Jing Y, Xiong X, Ming Y, Zhao J, Guo X, Yang G, et al. 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;7:1700974. [182] Yang C, Uertz J, Yohan D, Chithrani BD. Peptide modified gold nanoparticles for improved cellular uptake, nuclear transport, and intracellular retention. Nanoscale 2014;6:12026–33. [183] Hsiangkuo Y, Fales AM, Tuan VD. TAT peptide-functionalized gold nanostars: enhanced intracellular delivery and efficient NIR photothermal therapy using ultralow irradiance. J Am Chem Soc 2012;134:11358–61. [184] Desplancq D, Groysbeck N, Chiper M, Weiss E, Frisch B, Strub JM, et al. Cytosolic diffusion and peptide-assisted nuclear shuttling of peptide-substituted circa 102 gold atom nanoclusters in living cells. ACS Appl Nano Mater 2018;1:4236–46. [185] Narayanan K, Yen SK, Dou Q, Padmanabhan P, Sudhaharan T, Ahmed S, et al. Mimicking cellular transport mechanism in stem cells through endosomal escape of new peptide-coated quantum dots. Sci Rep 2013;3:2184. [186] Maity AR, Stepensky D. Nuclear and perinuclear targeting efficiency of quantum dots depends on density of peptidic targeting residues on their surface. J Control Release 2017;257:32–9. [187] Wlodarczyk MT, Dragulska SA, Camacho-Vanegas O, Dottino PR, Jarzęcki AA, Martignetti JA, et al. Platinum (II) complex-nuclear localization sequence peptide hybrid for overcoming platinum resistance in cancer therapy. ACS Biomater Sci Eng 2018;9:463–7. [188] Tomizaki KY, Kishioka K, Kataoka S, Miyatani M, Ikeda T, Komada M, et al. Non-covalent loading of anti-cancer doxorubicin by modularizable peptide selfassemblies for a nanoscale drug carrier. Molecules 2017;22:1916. [189] Hao X, Li Q, Guo J, Ren X, Feng Y, Shi C, et al. Multifunctional gene carriers with enhanced specific penetration and nucleus accumulation to promote neovascularization of HUVECs in vivo. ACS Appl Mater Inter 2017;9:35613–27. [190] Patel SS, Belmont BJ, Sante JM, Rexach MF. Natively unfolded nucleoporins gate protein diffusion across the nuclear pore complex. Cell 2007;129:83–96. [191] Alber F, Dokudovskaya S, Veenhoff LM, Zhang W, Kipper J, Devos D, et al. The molecular architecture of the nuclear pore complex. Nature 2007;450:695–701. [192] Hsu S, Hung M. Characterization of a novel tripartite nuclear localization sequence in the EGFR family. J Biol Chem 2007;282:10432–40. [193] Lo HW, Hsu SC, Ali-Seyed M, Gunduz M, Xia W, Wei Y, et al. Nuclear interaction of EGFR and STAT3 in the activation of the iNOS/NO pathway. Cancer Cell 2005;7:575–89. [194] Hui-Wen L, Mohamed AS, Yadi W, Geoffrey B, Sheng-Chieh H, Mien-Chie H. Nuclear-cytoplasmic transport of EGFR involves receptor endocytosis, importin beta1 and CRM1. J Cell Biochem 2006;98:1570–83. [195] Liao HJ, Carpenter G. Role of the Sec61 translocon in EGF receptor trafficking to the nucleus and gene expression. Mol Biol Cell 2007;18:1064–72. [196] Jaganathan S, Yue P, Paladino DC, Bogdanovic J, Huo Q, Turkson J. A functional nuclear epidermal growth factor receptor, src and stat3 heteromeric complex in pancreatic cancer cells. PLoS ONE 2011;6:e19605. [197] Klaus D, Claus M, Birgit F, Martin S, Rainer K, Peter H, et al. Nuclear epidermal growth factor receptor modulates cellular radio-sensitivity by regulation of chromatin access. Radiother Oncol 2011;99:317–22. [198] Traynor AM, Weigel TL, Oettel KR, Yang DT, Zhang C, Kim KM, et al. Nuclear EGFR protein expression predicts poor survival in early stage non-small cell lung cancer. Lung Cancer 2013;81:138–41.

21

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

[199] Hsu FT, Liu HS, Ali AA, Tsai PH, Kao YC, Lu CF, et al. Assessing the selective therapeutic efficacy of superparamagnetic erlotinib nanoparticles in lung cancer by using quantitative magnetic resonance imaging and a nuclear factor kappa-B reporter gene system. Nanomed Nanotechnol 2018;14:1019–31. [200] Li C, Iida M, Dunn EF, Ghia AJ, Wheeler DL. Nuclear EGFR contributes to acquired resistance to cetuximab. Oncogene 2009;28:3801–13. [201] Lo HW, Xia W, Wei Y, Ali-Seyed M, Huang SF, Hung MC. Novel prognostic value of nuclear epidermal growth factor receptor in breast cancer. Cancer Res 2005;65:338–48. [202] Xia W, Wei Y, Yi D, Liu J, Chang B, Yu YL, et al. Nuclear expression of epidermal growth factor receptor is a novel prognostic value in patients with ovarian cancer. Mol Carcinogen 2009;48:610–7. [203] Klaus D, Claus M, Birgit F, Martin S, Uma R, Luka M, et al. Radiation-induced epidermal growth factor receptor nuclear import is linked to activation of DNAdependent protein kinase. J Biol Chem 2005;280:31182–9. [204] Klaus D, Claus M, Birgit F, Martin S, Rainer K, Peter H, et al. Nuclear EGFR shuttling induced by ionizing radiation is regulated by phosphorylation at residue Thr654. Febs Lett 2010;584:3878–84. [205] Yuan Y, Chen S, Paunesku T, Gleber SC, Liu WC, Doty CB, et al. Epidermal growth factor receptor targeted nuclear delivery and high-resolution whole cell X-ray imaging of Fe3O4@TiO2 nanoparticles in cancer cells. ACS Nano 2013;7:10502–17. [206] Zhu YX, Jia HR, Pan GY, Ulrich NW, Chen Z, Wu FG. Development of a light-controlled nanoplatform for direct nuclear delivery of molecular and nanoscale materials. J Am Chem Soc 2018;140:4062–70. [207] Houthaeve G, Xiong R, Robijns J, Luyckx B, Beulque Y, Brans T, et al. Targeted perturbation of nuclear envelope integrity with vapor nanobubble-mediated photoporation. ACS Nano 2018;12:7791–802. [208] Wang H, Li Y, Bai H, Shen J, Chen X, Ping Y, et al. A cooperative dimensional strategy for enhanced nucleus-targeted delivery of anticancer drugs. Adv Funct Mater 2017;27:1700339. [209] Lai YH, Chiang CS, Kao TH, Chen SY. Dual-drug nanomedicine with hydrophilic F127-modified magnetic nanocarriers assembled in amphiphilic gelatin for enhanced penetration and drug delivery in deep tumor tissue. Int J Nanomed 2018;13:3011. [210] Lu Z, Long Y, Cun X, Wang X, Li J, Mei L, et al. A size-shrinkable nanoparticle-based combined anti-tumor and anti-inflammatory strategy for enhanced cancer therapy. Nanoscale 2018;10:9957–70. [211] Niu Y, Zhu J, Li Y, Shi H, Gong Y, Li R, et al. Size shrinkable drug delivery nanosystems and priming the tumor microenvironment for deep intratumoral penetration of nanoparticles. J Control Release 2018;277:35–47. [212] Liu Y, Li L, Li L, Zhou Z, Wang F, Xiong X, et al. Programmed drug delivery system based on optimized “size decrease and hydrophilicity/hydrophobicity transformation” for enhanced hepatocellular carcinoma therapy of doxorubicin. Nanomedicine 2018;14:1111–22. [213] Fan Y, Li C, Cao H, Li F, Chen D. The intranuclear release of a potential anticancer drug from small nanoparticles that are derived from intracellular dissociation of large nanoparticles. Biomaterials 2012;33:4220–8. [214] Li L, Sun W, Zhong J, Yang Q, Zhu X, Zhou Z, et al. Multistage nanovehicle delivery system based on stepwise size reduction and charge reversal for programmed nuclear targeting of systemically administered anticancer drugs. Adv Funct Mater 2015;25:4101–13. [215] Hollenbeck PJ, Saxton WM. The axonal transport of mitochondria. J Cell Sci 2005;118:5411–9. [216] Zamzami N, Kroemer G. The mitochondrion in apoptosis: how Pandora's box opens. Nat Rev Mol Cell Bio 2001;2:67–71. [217] Mcbride HM, Neuspiel M, Wasiak S. Mitochondria: more than just a powerhouse. Curr Biol 2006;16:R551–60. [218] Modica-Napolitano JS, Singh KK. Mitochondrial dysfunction in cancer. Mitochondrion 2004;4:755–62. [219] Galluzzi L, Morselli E, Kepp O, Vitale I, Rigoni A, Vacchelli E, et al. Mitochondrial gateways to cancer. Mol Aspects Med 2010;31:1–20. [220] Costantini P, Jacotot E, Decaudin D, Kroemer G. Mitochondrion as a novel target of anticancer chemotherapy. J Natl Cancer I 2000(92):1042–53. [221] Murphy MP, Smith RA. Drug delivery to mitochondria: the key to mitochondrial medicine. Adv Drug Deliver Rev 2000;41:235–50. [222] Smith RAJ, Hartley RC, Cochemé HM, Murphy MP. Mitochondrial pharmacology. Trends Pharmacol Sci 2012;33:341–52. [223] Yamada Y, Harashima H. Mitochondrial drug delivery systems for macromolecule and their therapeutic application to mitochondrial diseases. Adv Drug Deliver Rev 2008;60:1439–62. [224] Kidd JF, Pilkington MF, Schell MJ, Fogarty KE, Skepper JN, Taylor CW, et al. Paclitaxel affects cytosolic calcium signals by opening the mitochondrial permeability transition pore. J Biol Chem 2002;277:6504–10. [225] Robertson JD, Gogvadze V, Zhivotovsky B, Orrenius S. Distinct pathways for stimulation of cytochrome release by etoposide. J Biol Chem 2000;275:32438–43. [226] André N, Braguer D, Brasseur G, Gonçalves A, Lemesle-Meunier D, Guise S, et al. Paclitaxel induces release of cytochrome c from mitochondria isolated from human neuroblastoma cells. Cancer Res 2000;60:5349–53. [227] Kelley SO, Stewart KM, Mourtada R. Development of novel peptides for mitochondrial drug delivery: amino acids featuring delocalized lipophilic cations. Pharm Res 2011;28:2808–19. [228] Ross MF, Kelso GF, Blaikie FH, James AM, Cochemé HM, Filipovska A, et al. Lipophilic triphenylphosphonium cations as tools in mitochondrial bioenergetics and free radical biology. Biochemistry (Moscow) 2005;70:222–30. [229] Yousif LF, Stewart KM, Kelley SO. ChemInform abstract: targeting mitochondria with organelle-specific compounds: strategies and applications. ChemBioChem 2009;10:1939–50. [230] Murphy MP. Targeting lipophilic cations to mitochondria. BBA-Bioenergetics 2008;1777:1028–31. [231] Murphy MP, Smith RAJ, Murphy MP, Smith RA. Targeting antioxidants to mitochondria by conjugation to lipophilic cations. Annu Rev Pharmacol Toxicol 2007;47:629–56. [232] Gang C, Zielonka J, Mcallister D, Hardy M, Ouari O, Joseph J, et al. Antiproliferative effects of mitochondria-targeted cationic antioxidants and analogs: role of mitochondrial bioenergetics and energy-sensing mechanism. Cancer Lett 2015;365:96–106. [233] Antonenko YN, Avetisyan AV, Cherepanov DA, Knorre DA, Korshunova GA, Markova OV, et al. Derivatives of rhodamine 19 as mild mitochondria-targeted cationic uncouplers. J Biol Chem 2011;286:17831–40. [234] Prime TA, Blaikie FH, Cameron E, Nadtochiy SM, James AM, Dahm CC, et al. A mitochondria-targeted S-nitrosothiol modulates respiration, nitrosates thiols, and protects against ischemia-reperfusion injury. Proc Natl Acad Sci USA 2009;106:10764–9. [235] Dikalova AE, Bikineyeva AT, Klaudia B, Nazarewicz RR, Louise MC, William L, et al. Therapeutic targeting of mitochondrial superoxide in hypertension. Circ Res 2010;107:106–16. [236] Lei W, Xie J, Hou Y, Jiang G, Zhang H, Wang P, et al. Mitochondria-targeting properties and photodynamic activities of porphyrin derivatives bearing cationic pendant. J Photoch Photobio B 2010;98:167–71. [237] Eftekhari A. The application of novel mitochondria-targeted antioxidants: current strategies and future perspectives. J Adv Chem Pharm Mater 2018;1:1–2. [238] Amorim R, Benfeito S, Teixeira J, Cagide F, Oliveira PJ, Borges F. Targeting mitochondria: the road to mitochondriotropic antioxidants and beyond. In: Oliveira PJ, editor.Mitochondrial biology and experimental therapeutics; 2018. p 333–58. New York: Springer. [239] Hardy M, Poulhés F, Rizzato E, Rockenbauer A, Banaszak K, Karoui H, et al. Mitochondria-targeted spin traps: synthesis, superoxide spin trapping, and mitochondrial uptake. Chem Res Toxicol 2014;27:1155–65. [240] Shchepinova MM, Cairns AG, Prime TA, Logan A, James AM, Hall AR, et al. MitoNeoD: a mitochondria-targeted superoxide probe. Cell Chem Biol 2017;24:1285–98. [241] Han X, Wang R, Song X, Yu F, Lv C, Chen L. A mitochondrial-targeting near-infrared fluorescent probe for bioimaging and evaluating endogenous superoxide anion changes during ischemia/reperfusion injury. Biomaterials 2018;156:134–46. [242] Theodossiou TA, Zili S, Katsarou ME, Dimitris T. Mitochondrial delivery of doxorubicin by triphenylphosphonium-functionalized hyperbranched nanocarriers results in rapid and severe cytotoxicity. Pharm Res 2013;30:2832–42. [243] Jing Q, Zhi L, Ye T, Man W, Zhongwei N. Multifunctional self-assembled polymeric nanoprobes for FRET-based ratiometric detection of mitochondrial H_2 O_2 in living cells. Chem Commun 2015;51:3641–4. [244] Sean M, Shanta D. Engineering of blended nanoparticle platform for delivery of mitochondria-acting therapeutics. Proc Natl Acad Sci USA 2012;109:16288–93.

22

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

[245] Khatun Z, Choi YS, Kim YG, Yoon K, Nurunnabi M, Li L, et al. Bioreducible poly (ethylene glycol)-triphenylphosphonium conjugate as a bioactivable mitochondria-targeting nanocarrier. Biomacromolecules 2017;18:1074–85. [246] Wang Y, Wei G, Zhang X, Huang X, Zhao J, Guo X, et al. Multistage targeting strategy using magnetic composite nanoparticles for synergism of photothermal therapy and chemotherapy. Small 2018;14:1702994. [247] Xu Y, Wang S, Chan HF, Liu Y, Li H, He C, et al. Triphenylphosphonium-modified poly (ethylene glycol)-poly (ε-caprolactone) micelles for mitochondriatargeted gambogic acid delivery. Int J Pharm 2017;522:21–33. [248] Zhang S, Li J, Hu S, Wu F, Zhang X. Triphenylphosphonium and D-α-tocopheryl polyethylene glycol 1000 succinate-modified, tanshinone IIa-loaded lipidpolymeric nanocarriers for the targeted therapy of myocardial infarction. Int J Nanomed 2018;13:4045. [249] Hu Q, Gao M, Feng G, Liu B. Mitochondria-targeted cancer therapy using a light-up probe with aggregation-induced-emission characteristics. Angew Chem Inter Ed 2014;53:14225–9. [250] Yin C, Zhu H, Xie C, Zhang L, Chen P, Fan Q, et al. Organic nanoprobe cocktails for multilocal and multicolor fluorescence imaging of reactive oxygen species. Adv Funct Mater 2017;27:1700493. [251] Wang B, Wang Y, Wu H, Song X, Guo X, Zhang D, et al. A mitochondria-targeted fluorescent probe based on TPP-conjugated carbon dots for both one- and twophoton fluorescence cell imaging. RSC Adv 2014;4:49960–3. [252] Wu X, Sun S, Wang Y, Zhu J, Jiang K, Leng Y, et al. A fluorescent carbon-dots-based mitochondria-targetable nanoprobe for peroxynitrite sensing in living cells. Biosens Bioelectron 2017;90:501–7. [253] Zhuang Q, Jia H, Du L, Li Y, Zhao C, Huang S, et al. Targeted surface-functionalized gold nanoclusters for mitochondrial imaging. Biosens Bioelectron 2014;55:76–82. [254] Chen S, Lei Q, Qiu WX, Liu LH, Zheng DW, Fan JX, et al. Mitochondria-targeting “Nanoheater” for enhanced photothermal/chemo-therapy. Biomaterials 2017;117:92–104. [255] López V, Villegas MR, Rodríguez V, Villaverde G, Lozano D, Baeza A, et al. Janus mesoporous silica nanoparticles for dual targeting of tumor cells and mitochondria. ACS Appl Mater Int 2017;9:26697–706. [256] Choi YS, Kwon K, Yoon K, Huh KM, Kang HC. Photosensitizer-mediated mitochondria-targeting nanosized drug carriers: subcellular targeting, therapeutic, and imaging potentials. Int J Pharm 2017;520:195–206. [257] Xing L, Lyu JY, Yang Y, Cui PF, Gu LQ, Qiao JB, et al. pH-Responsive de-PEGylated nanoparticles based on triphenylphosphine-quercetin self-assemblies for mitochondria-targeted cancer therapy. Chem Commun 2017;53:8790–3. [258] Weissig V, Lizano C, Torchilin VP. Micellar delivery system for dequalinium—a lipophilic cationic drug with anticarcinoma activity. J Liposome Res 2008;8:391–400. [259] Weissig V, Lasch J, Erdos G, Meyer H, Rowe T, Hughes J. DQAsomes: a novel potential drug and gene delivery system made from dequalinium™. Pharm Res 1998;15:334–7. [260] D’Souza GGM, Rammohan R, Cheng SM, Torchilin VP, Weissig V. DQAsome-mediated delivery of plasmid DNA toward mitochondria in living cells. J Control Release 2003;92:189–97. [261] Yamada Y, Munechika R, Kawamura E, Sakurai Y, Sato Y, Harashima H. Mitochondrial delivery of doxorubicin using MITO-Porter kills drug-resistant renal cancer cells via mitochondrial toxicity. J Pharm Sci 2017;106:2428–37. [262] Yamada Y, Akita H, Kamiya H, Kogure K, Yamamoto T, Shinohara Y, et al. A liposome-based carrier system for delivery of macromolecules into mitochondria via membrane fusion. BBA-Biomembranes 2008;1778:423–32. [263] Yamada Y, Harashima H. Delivery of bioactive molecules to the mitochondrial genome using a membrane-fusing, liposome-based carrier, DF-MITO-Porter. Biomaterials 2012;33:1589–95. [264] Khalil IA, Kogure K, Futaki S, Harashima H. High density of octaarginine stimulates macropinocytosis leading to efficient intracellular trafficking for gene expression. J Biol Chem 2006;281:3544–51. [265] Bae Y, Jung MK, Lee S, Song SJ, Mun JY, Green ES, et al. Dequalinium-based functional nanosomes show increased mitochondria targeting and anticancer effect. Eur J Pharm Biopha 2018;124:104–15. [266] Abe Y, Shodai T, Muto T, Mihara K, Torii H, Nishikawa SI, et al. Structural basis of presequence recognition by the mitochondrial protein import receptor Tom20. Cell 2000;100:551–60. [267] Erlich-Hadad T, Hadad R, Feldman A, Greif H, Lorberboum-Galski H. TAT-MTS-MCM fusion proteins reduce MMA levels and improve mitochondrial activity and liver function in MCM-deficient cells. J Cell Mol Med 2018;22:1601–13. [268] Stojanovski D, Johnston AJ, Streimann I, Hoogenraad NJ, Ryan MT. Import of nuclear-encoded proteins into mitochondria. Exp Physiol 2003;88:57–64. [269] Mukhopadhyay A, Ni L, Yang C-S, Weiner H. Bacterial signal peptide recognizes HeLa cell mitochondrial import receptors and functions as a mitochondrial leader sequence. Cell Mol Life Sci 2005;62:1890–9. [270] Mc DLL, Wellinger RE. A novel approach for organelle-specific DNA damage targeting reveals different susceptibility of mitochondrial DNA to the anticancer drugs camptothecin and topotecan. Nucleic Acids Res 2009;37:e26. [271] Adlam VJ, Harrison JC, Porteous CM, James AM, Smith RAJ, Murphy MP, et al. Targeting an antioxidant to mitochondria decreases cardiac ischemiareperfusion injury. FASEB J 2005;19:1088–95. [272] Chretien D, Benit P, Ha HH, Keipert S, El-Khoury R, Chang YT, et al. Mitochondria are physiologically maintained at close to 50 °C. PLoS Biol 2018;16:e2003992. [273] Wang D, Huang H, Zhou M, Lu H, Chen J, Chang YT, et al. A thermoresponsive nanocarrier for mitochondria-targeted drug delivery. Chem Commun 2019;55:4051–4. [274] Bosc C, Selak MA, Sarry JE. Resistance is futile: targeting mitochondrial energetics and metabolism to overcome drug resistance in cancer treatment. Cell Metab 2017;26:705–7. [275] Wang H, Gao Z, Liu X, Agarwal P, Zhao S, Conroy DW, et al. Targeted production of reactive oxygen species in mitochondria to overcome cancer drug resistance. Nat Commun 2018;9:562. [276] Szakács G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov 2006;5:219. [277] Mourtada R, Fonseca SB, Wisnovsky SP, Pereira MP, Wang X, Hurren R, et al. Re-directing an alkylating agent to mitochondria alters drug target and cell death mechanism. PLoS ONE 2013;8:e6023. [278] Martin LP, Hamilton TC, Schilder RJ. Platinum resistance: the role of DNA repair pathways. Clin Cancer Res 2008;14:1291–5. [279] Li J, He X, Zou Y, Chen D, Yang L, Rao J, et al. Mitochondria-targeted platinum (ii) complexes: dual inhibitory activities on tumor cell proliferation and migration/invasion via intracellular trafficking of β-catenin. Metallomics 2017;9:726–33. [280] Ledoux SP, Wilson GL, Beecham EJ, Stevnsner T, Wassermann K, Bohr VA. Repair of mitochondrial DNA after various types of DNA damage in Chinese hamster ovary cells. Carcinogenesis 1992;13:1967–73. [281] Wisnovsky SP, Wilson JJ, Radford RJ, Pereira MP, Chan MR, Laposa RR, et al. Targeting mitochondrial DNA with a platinum-based anticancer agent. Chem Biol 2013;20:1323–8. [282] Panasci L, Paiement JP, Christodoulopoulos G, Belenkov A, Malapetsa A, Aloyz R. Chlorambucil drug resistance in chronic lymphocytic leukemia: the emerging role of DNA repair. Clin Cancer Res 2001;7:454–61. [283] Fonseca SB, Pereira MP, Rida M, Marcela G, Horton KL, Hurren R, et al. Rerouting chlorambucil to mitochondria combats drug deactivation and resistance in cancer cells. Chem Biol 2011;18:445–53. [284] Castino R, Démoz M, Isidoro C. Destination ‘Lysosome’: a target organelle for tumour cell killing? J Mol Recognit 2003;16:337–48. [285] Kudo M, Bao M, Souza AD, Ying F, Pan H, Roe BA, et al. The alpha-and beta-subunits of the human UDP-N-acetylglucosamine:lysosomal enzyme N-acetylglucosamine-1-phosphotransferase [corrected] are encoded by a single cDNA. J Biol Chem 2005;280:36141–9. [286] And GAG, Hopkin RJ. Enzyme therapy for lysosomal storage disease: principles, practice, and prospects. Annu Rev Genom Hum. G 2003;4:403–36.

23

Progress in Materials Science 107 (2020) 100599

X. Guo, et al.

[287] Ni X, Canuel M, Morales CR. The sorting and trafficking of lysosomal proteins. Histol Histopathol 2006;21:899–913. [288] Hoogendoorn S, van Puijvelde GH, Kuiper J, van der Marel GA, Overkleeft HS. A multivalent ligand for the mannose-6-phosphate receptor for endolysosomal targeting of an activity-based probe. Angew Chem Int Ed 2014;53:10975–8. [289] Reczek D, Schwake M, Schröder J, Hughes H, Blanz J, Jin X, et al. LIMP-2 is a receptor for lysosomal mannose-6-phosphate-independent targeting of betaglucocerebrosidase. Cell 2007;131:770–83. [290] Zhao Y, Ren J, Padill-Parra S, Fry EE, Stuart DI. Lysosome sorting of β-glucocerebrosidase by LIMP-2 is targeted by the mannose 6-phosphate receptor. Nat Commun 2014;5:4321–32. [291] Roberta C, Deborah P, Marina D, Marco G, Chiara A, Elisabetta R, et al. Lysosomal proteases as potential targets for the induction of apoptotic cell death in human neuroblastomas. Int J Cancer 2002;97:775–9. [292] Denamur S, Tyteca D, Bambeke FOV, Tulkens PM, Courtoy PJ. Role of oxidative stress in lysosomal membrane permeabilization and apoptosis induced by gentamicin, an aminoglycoside antibiotic. Free Radical Bio Med 2011;51:1656–65. [293] Hamdiye E, Maria B, Juan C, Ulf B, Shoshan MC, Stig L. Induction of lysosomal membrane permeabilization by compounds that activate p53-independent apoptosis. Proc Natl Acad Sci USA 2005;102:192–7. [294] Ostenfeld M, Høyer-Hansen M, Bastholm L, Fehrenbacher N, Olsen OD, Groth-Pedersen L, et al. Anti-cancer agent siramesine is a lysosomotropic detergent that induces cytoprotective autophagosome accumulation. Autophagy 2008;4:487–99. [295] Boya P, Kroemer G. Lysosomal membrane permeabilization in cell death. Oncogene 2008;27:6434–51. [296] Gotink KJ, Broxterman HJ, Mariette L, Haas RR, De Henk D, Honeywell RJ, et al. Lysosomal sequestration of sunitinib: a novel mechanism of drug resistance. Clin Cancer Res 2011;17:7337–46. [297] Selbo PK, Høgset A, Prasmickaite L, Berg K. Photochemical internalisation: a novel drug delivery system. Tumor Biol 2002;23:103–12. [298] Rao M, Alving CR. Delivery of lipids and liposomal proteins to the cytoplasm and Golgi of antigen-presenting cells. Adv Drug Deliver Rev 2000;41:171–88. [299] Tarragó-Trani MT, Storrie B. Alternate routes for drug delivery to the cell interior: pathways to the Golgi apparatus and endoplasmic reticulum. Adv Drug Deliver Rev 2007;59:782–97. [300] Sandvig K, Bergan J, Dyve AB, Skotland T, Torgersen ML. Endocytosis and retrograde transport of Shiga toxin. Toxicon 2010;56:1181–5. [301] Bonifacino JS, Rojas R. Retrograde transport from endosomes to the trans-Golgi network. Nat Rev Mol Cell Biol 2006;7:568–79. [302] Vella F. Molecular biology of the cell (third edition): By B Alberts, D Bray, J Lewis, M Raff, K Roberts and J D Watson. pp 1361. Garland Publishing, New York and London. Mol Biol Cell New York Garland 1994;22:600–51. [303] Aridor M, Hannan LA. Traffic Jam: a compendium of human diseases that affect intracellular transport processes. Traffic 2000;1:836–51. [304] Yoshida H. ER stress and diseases. Febs J 2007;274:630–58 Minamino T, Komuro IM. Endoplasmic reticulum stress as a therapeutic target in cardiovascular disease. Circ Res 2010;107:1071–82. [305] Donald W, Joanna S, Dagmara MG, Chris H, Zbigniew D. ER-Golgi network–a future target for anti-cancer therapy. Leukemia Res 2009;33:1440–7. [306] Rajendran L, Knölker HJ, Kai S. Subcellular targeting strategies for drug design and delivery. Nat Rev Drug Discov 2010;9:29–42. [307] Helena P, José Manuel L, Paula S. The mTOR signalling pathway in human cancer. Int J Mol Sci 2012;13:1886–918. [308] Hanahan D, Weinberg R. Hallmarks of cancer: the next generation. Cell 2011;144:646–74. [309] Nan RM, Xiangyu L, Bertram PG, Zheng XFS. FKBP12-rapamycin-associated protein or mammalian target of rapamycin (FRAP/mTOR) localization in the endoplasmic reticulum and the Golgi apparatus. J Biol Chem 2004;279:772–8. [310] Gariépy J. The use of Shiga-like toxin 1 in cancer therapy. Crit Rev Oncol Hemat 2001;39:99–106. [311] Furukawa K, Yokoyama K, Sato T, Wiels J, Hirayama Y, Ohta M, et al. Expression of the Gb3/CD77 synthase gene in megakaryoblastic leukemia cells. J Biol Chem 2002;277:11247–54. [312] Dan JS, Puri V, Marks DL, Butters TD, Dwek RA, Pagano RE, et al. Glucosylceramide modulates membrane traffic along the endocytic pathway. J Lipid Res 2002;43:1837–45.

Shaobing Zhou received his B.S. degree in polymer chemistry from Sichuan University (1996) and his Ph.D. in organic chemistry from Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences (2003). He was promoted in 2005 to Full Professor in the School of Materials Science and Engineering, Southwest Jiaotong University. His research interests include the synthesis and characterization of biodegradable polymers, drug-delivery systems, shape-memory polymer composites, and electrospun biodegradable polymer fibers as a tissue engineering scaffold. He is the author or coauthor of more than 120 refereed articles and 15 Chinese patents/patent applications. He has been cited over 5000 times and has an h-index of 45.

24