Boosting Cancer Therapy with Organelle-Targeted Nanomaterials

May 28, 2019 - Continuous laser irradiation (2 mW/cm2, 3 min) resulted in the further ... This method opened a new avenue for designing nucleus-target...
0 downloads 0 Views 3MB Size
Subscriber access provided by BUFFALO STATE

Review

Boosting Cancer Therapy with Organelle-Targeted Nanomaterials peng gao, Wei Pan, Na Li, and Bo Tang ACS Appl. Mater. Interfaces, Just Accepted Manuscript • DOI: 10.1021/acsami.9b01370 • Publication Date (Web): 28 May 2019 Downloaded from pubs.acs.org on July 16, 2019

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Boosting Cancer Therapy with Organelle-Targeted Nanomaterials Peng Gao, Wei Pan, Na Li,* and Bo Tang* College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China. KEYWORDS: cancer therapy, organelle-targeted, nucleus, mitochondrion, endoplasmic reticulum, lysosome, Golgi apparatus, nanodrugs

ABSTRACT: The ultimate goal of cancer therapy is to eliminate malignant tumors while cause no damage to normal tissues. In the past decades, numerous nano-agents have been employed for cancer treatment owing to their unique properties over traditional molecular drugs. However, lack of selectivity and unwanted therapeutic outcomes severely limited the therapeutic index of traditional nanodrugs. Recently, a series of organelle-targeted nanomaterials that can accumulate into specific organelles (nucleus, mitochondrion, endoplasmic reticulum, lysosome, Golgi apparatus) intra cancer cells have received increasing interest. These rationally designed nanoagents can either directly destroy the subcellular structure or effectively deliver drugs into the proper targets, which can further activate certain cell death pathways, enabling them to boost the

ACS Paragon Plus Environment

1

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 2 of 85

therapeutic efficiency, lower drug dosage, reduce side effects, avoid multidrug resistance and prevent recurrence. In this review, the design principles, targeting strategies, therapeutic mechanisms, current challenges and potential future directions of organelle-targeted nanomaterials will be introduced.

1. Introduction Cancer is a group of diseases that can affect almost any part of human body, which has continuously brought heavy social burdens and severe health problems.1-6 Currently, the clinical cancer therapeutic efficiency is still unsatisfactory due to the lack of effective schemes. Therefore, developing novel medicine for highly efficient cancer treatment is a worldwide concern.7-9 Many strategies, including surgery, chemotherapy, radiotherapy, phototherapy, immune therapy etc., have been explored to eliminate malignant tumors and increase survival rates of patients.1-10 Unfortunately, only limited therapeutic effects can be achieved due to the intrinsic disadvantages of each approach.11-16 As an example, chemotherapy usually involves the frequent administration of high dosage therapeutic drugs with poor selectivity toward cancerous cells, which can bring deadly side effects to the patients, including liver/kidney toxicity, break down the immunity systems and so on.17,18 Meanwhile, via mutation, cancer cells can quickly become resistant to the drugs due to the development of drug resistance system, further restricting the therapeutic effects.19,20 Moreover, increasing evidences have suggested that chemotherapy may cause cancer metastasis, which can cause extra burden for patients.21-26 Accordingly, many efforts have been devoted to the modification of drugs to improve the bioavailabilities and reduce side effects. Nevertheless, chemical conjugation can reduce the pharmaceutical effects of the drugs, and only limited therapeutic drugs can be easily functionalized.27-30 Therefore, research focus has been shifted to the design and fabrication of

ACS Paragon Plus Environment

2

Page 3 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

novel drug delivery vehicles, including inorganic materials, organic materials as well as their hybrids.31-45 These studies revealed that both passive and active targeting modality of these vehicles can lead to positive outcomes, and some of them have been clinically employed due to their higher therapeutic index.46-56 However, nonspecific bio-distribution and relatively low therapeutic efficiency still have yet to be further resolved. In vivo experiments indicated that only a small percentage of the administered drugs can reach tumor sites.57-64 Thus, highly efficient tumor targeted therapy is still challenging and desired.65-73 Organelles are indispensable for cellular function.74-83 Lysosome takes part in digestion, autophagy and cellular defense; mitochondrion takes charge of the ATP synthesis, calcium ion cycle and apoptosis regulation; endoplasm reticulum and Golgi apparatus participate in protein fabrication and transportation, nucleus controls the gene expression as well as cell proliferation.84,90 Benefiting from the critical biological effects of diverse organelles, driving proper therapeutics into specific organelles may take new opportunities for cancer therapy.91-102 For example, the first line chemotherapeutic drug, cisplatin, can crosslink the tumor DNA and reduce the gene repression of a wide range of cancer cells.64,103 By delivering cisplatin into the nucleus of cancer cell, higher therapeutic index can be realized compared to diffusing it to the cytoplasm. Therefore, side effects and recurrence could be avoided.104 More importantly, organelle dysfunction can usually activate specific cell death signal, since organelles are closely related to diverse signal pathways, which can be employed for cancer treatment with enhanced efficiency and specificity.105-107 During the past several years, organelle-targeted therapeutics have arose increasing interest.108111

Earlier studies revealed that organelle-targeted nano-objects are beneficial for overcoming

several obstacles, including drug resistance, drug premature leakage, and high dosage

ACS Paragon Plus Environment

3

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 4 of 85

requirement.112-121 Moreover, by virtue of their superior designability, multi-functional nanosystems can be easily constructed by integrating multiple imaging and therapy modality into the designed nano-platforms, which will further potentiate cancer theranostics. This review mainly focuses on the recent achievements of functional nanomaterials for organelle-targeted therapy, which covers mitochondria-targeted therapy, nucleus-targeted therapy, lysosome-targeted therapy, endoplasmic reticulum, and Golgi apparatus-targeted therapy (Figure 1). The design principle, working mechanisms and advantages of diverse nano-formulations are illustrated. Meanwhile, their limitations and potential future development directions are also pointed out. The major objectives of the current review are to emphasize the broad potential of advanced organelle-targeted nanomaterials for cancer therapy applications, and to offer valuable information for medical chemists to further resolve involved challenges.

Figure 1. Schematic of the precise cancer therapy with boosted therapeutic index based on organelle-targeted nanomaterials. 2. Nucleus-targeted therapy As the most important organelle of eukaryotic cells, nucleus contains the vast majority of DNA and controls the gene expression of the cells.84,85,122 The main origin of cancer is believed to be the uncontrolled growth of cells caused by gene mutation, and many cancer treatment modality have

ACS Paragon Plus Environment

4

Page 5 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

been developed to inhibit the cellular proliferation by taking nucleus/nucleic acids as targets.123126

Therefore, nucleus-targeted cancer therapy has intrinsic superiority because the drugs are more

accessible to their ultimate targets.127-130 The nucleus is surrounded by two-layered nuclear envelope, which can separate the genomes from cytoplasm to ensure DNA replication and gene repression in a secure environment. To maintain the transport of diverse RNA as well as enzymes between nucleus and cytoplasm, the nuclear membrane is traversed by a series of protein machines (nuclear pore complexes, NPCs).105,131,132 Although some ultra-small nanoparticles might diffuse into the nucleus17,123,129, those nanoparticles still suffer from low nucleus-entry efficiency since they must penetrate a series of complicated biological membrane barriers in cancer cells. Therefore, vehicles with nucleartargeting ligands are critical for nucleus-targeted delivery. Up to now, many virus-inspired peptides that could interact with the NPCs has been used to modify nanoparticles to render them nucleus locating capacity. Chemical molecules with strong nucleus membrane receptor affinity and some other nucleus membrane opening strategies were also reported.118,127,133-136 (Table 1) Based on these nano-carriers/medicine, nucleus-targeted chemotherapy, photodynamic therapy, photothermal therapy, radiotherapy as well as combined therapy have been reported. Table 1. Typical nanomaterials for nucleus-targeted therapy.

Therapy modality

Nanomaterials

Chemotherapy

PEG-PCL QDs@mSiO2

PTT

Targeting mechanisms

Amidized TAT TAT

Acid responsive TAT activation, importin guided nuclear entry Enzyme responsive TAT exposure, importin guided nuclear entry Magnet guided tumor accumulation, receptor mediated endocytosis, importin guided nuclear entry Nanostructure enhanced uptake, charge accelerated nuclear entry ROS break nuclear membrane integrity Importin mediated nuclear entry Receptor-mediated targeting, importin mediated nuclear entry Importin mediated nuclear entry

Fe3O4@MSN

Magnet, TAT, FA

oligopeptide-drug aggregates RB functionalized POSS-PEG NPs Au nanorods Fe3O4



CuS@MSN PDT

Targeting ligands

MSNs

 TAT Tf, TAT TAT, RGD RGD, TAT

IC50

Cell type

Tumor model

Administration route (i.v./i.t.)

Treatment times

Treatment period

Refs

Bcap-37

i.v.

6

15

116



SKOV3 A549









133



HeLa









137

42 nM

A549/ 4T1 A549

4T1

i.v.

4

21

104











138

 

HeLa A549

HeLa A549

i.v. i.v.

1 1

15 14

128 139



HeLa

HeLa

i.v.

1

14

140



HeLa

HeLa

i.v.

1

15

141

(μg/mL)

Receptor-mediated targeting, importin mediated nuclear entry

2

ACS Paragon Plus Environment

5

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Peptide nanoparticles Gene therapy Combined therapy

UCNPs@TiO2-Ce6 AuNP aggregates

PKKKRK V TAT TAT

UCNP@MSN

TAT

Page 6 of 85

Acid mediated NLS exposure



HeLa

H22

i.v.

1

11

142

Importin mediated nuclear entry Photothermal triggered cargo release, importin mediated nuclear entry Importin mediated nuclear entry

 

MCF-7 A375

MCF-7 human melanoma

i.t. i.t.

1 10

14 20

122 143



MCF-7

MCF-7

i.t.

1

16

144

2.1 Nucleus-targeted chemotherapeutic agents As discussed above, a large number of clinical employed drugs are DNA replication associated toxins, including platinum based drugs (DNA crosslinker), doxorubicin (topoisomerase II inhibitor), 5-fluorouracil (thymidylate synthase inhibitor), camptothecin (topoisomerase I inhibitor) and so on.104,117,137,145,146 These drugs can inhibit DNA replication by interacting with double-standard DNA or deactivating related enzymes, which indicated that nucleus is the ultimate target for these chemotherapeutic drugs. Direct delivering drugs into the nucleus can enhance the therapeutic efficiency compared to traditional drug delivery strategies, because it significantly reduced the risks of these drugs being denaturalized or pumped out during their intracellular travel.127,147-149 Brinker’s group reported a mesoporous silica nanoparticles (MSNs) based platform for cancer nucleus-targeted drug delivery in 2011.80 MSNs was employed as the core of their “protocell”, which can load diverse nucleus localization signal (NLS) peptide functionalized model drugs (calcein, double-stranded DNA, red fluorescent protein, CdSe/ZnS quantum dots), the targeting peptide, fusogenic peptide and PEG co-modified lipid bilayer was encapsulated on the surface of the MSNs to reduce the immunogenicity, enhance the target efficiency and promote endosomal escape. The results revealed that this “protocell” significantly enhanced the intra-nuclear accumulation of NLS modified drugs, while the bare fluorescent protein and quantum dots loaded by MSNs stayed in cytoplasm. Compared to liposomes, higher drug loading efficiency and 106fold of therapy efficiency enhancement toward drug-resistant human hepatocellular carcinoma cell was observed, which proved the superiority of nucleus-targeted drug delivery strategy. Thereafter,

ACS Paragon Plus Environment

6

Page 7 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

MSNs, upconversion nanoparticles (UCNPs), polymeric nanoparticles, gold nanoparticles and quantum dots (QDs) have been investigated as nuclear-targeted chemotherapeutic drug vehicles.116,117,130,133,136,137,142,150-153

Figure 2. (A) Schematic illustration of the general preparation protocol of MSNs-TAT. (B) Schematic diagram of transport of DOX@MSNs-TAT across the nuclear membrane. (C) TEM images of MSNs with different sizes. Scale bars: 100 nm. CLSM (top) images and line-scan profiles (bottom) of fluorescence intensity for Hela cells incubated for 4 h with (D) free DOX, (E) DOX@MSNs (25 nm), and (F) DOX@MSNs-TAT (25 nm). The red fluorescence is from DOX, and the blue fluorescence is from DAPI. (G) Hela cell viabilities after 24 h of incubation with DOX-loaded MSNs and MSNs-TAT nanoparticles with different sizes at different DOX concentrations. Adapted and modified with permission from ref 117. Copyright 2012 American Chemical Society. Shi’s group investigated a series of TAT modified MSNs (MSNs-TAT) with different diameters (25, 50, 67 and 105 nm) for nucleus-targeted DOX delivery (Figure 2A-C).117 TAT peptides can bind with the import receptors importin α and β, and then translocate nanomaterials into the

ACS Paragon Plus Environment

7

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 8 of 85

nucleus. They found small MSNs-TAT (25 nm 50 nm) could efficiently enter the nucleus of drug resistant HeLa cells, while MSNs-TAT with larger sizes (67 and 105 nm) can only accumulate at the perinuclear region. They also demonstrated NLS peptides modified MSNs with suitable size can improve the anticancer effect of DOX (Figure 2D-G).

Figure 3. (A) Schematic illustration of the aTAT modified nanoparticles for enhanced blood circulation, tumor/lysosomal acidic microenvironment responsive activation and nucleus-targeted delivery. (B) Cellular uptake and intracellular localization of aTAT-PEG-PCL/nile red micelles at different time points: (a) 1, (b) 5, (c) 12, and (d) 24 h. (e-g) Lysosomal colocalization of aTATPEG-PCL/nile red in the cells after incubation for 5 h at 37 ºC. (h) An amplification of one cell in (c). (C) Blood circulation of the micelles. (D) In vivo tumor growth curves after treated with DOX or DOX-loaded micelles. (E) The concentration of DOX in tumor tissues after different treatments. Adapted and modified with permission from ref 116. Copyright 2012 American Chemical Society.

ACS Paragon Plus Environment

8

Page 9 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Although promising, it is noteworthy that the NLS peptides modified materials are usually positively charged, which can result in strong non-specific binding between nanoparticles and serum proteins and lead to the rapid blood clearance of nano-drugs. In the other hand, these nanoformulations with neutral or negative charge suffer from poor cell internalization. Thus, novel strategies that could overcome multiple biological barriers for hierarchical targeting are highly attractive. Shen et al. reported the amidation of TAT with succinyl amides as a charge shielding method for potentiating the in vivo targeting performance of TAT modified nano-carrier (Figure 3A).116 The cationic charge of TAT was reduced after amidation (aTAT). The amides in aTAT were quickly hydrolyzed in the acidic extracellular interstitium and lysosomes microenvironments owing to the acid sensitivity, afterwards the nuclear targeting capability of TAT was fully reactivated. aTAT was employed as the corona of DOX loaded poly(ethylene glycol)-block-poly(εcaprolactone) (PEG-PCL) micelles. In vitro and in vivo experiments suggested such drug delivery system significantly enhanced the anticancer efficiency of DOX on account of the prolonged blood circulation time and enhanced cancer cell nucleus drug accumulation (Figure 3B-E). Xing’s group reported the naturalization of cationic NLS peptide (CRRRQRRKKR) with a cathepsin B -responsive peptide (PGFK) linked anionic sequence (EEEEEE).133 Because of the shielding effect of anionic peptide, such peptide (CRRRQRRKKR-PGFK-EEEEEE) modified mesoporous silica coated quantum dots (CPP-QDs@mSiO2) located in cytoplasm of cells without cathepsin B expression, but were nucleus-targetable in cancer cells with cathepsin B due to the specific cleavage of the PGFK sequence. Benefiting from the outstanding fluorescent properties of QDs, imaging guided nucleus-targeted DOX delivery was performed in a series of cell lines. Unlike the rapid DOX expelling observed in drug resistant cells, DOX loaded CPP-QDs@mSiO2

ACS Paragon Plus Environment

9

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 10 of 85

specifically accumulated into the nucleus region of cancer cells, which demonstrated this drugloading system is a potential candidate for highly efficient and specific chemotherapy.

Figure 4. Schematic illustration of vasculature-to-cell membrane-to-nucleus sequential targeted drug delivery based on RGD and TAT peptides co-conjugated MSNs for effective cancer therapy. (A) CLSM images and line-scanning profiles of fluorescence intensity for HeLa cells incubated with (a1) free DOX, (a2) DOX@MSNs, (a3) DOX@MSNs-RGD, (a4) DOX@MSNs-TAT, and (a5) DOX@MSNs-RGD/TAT. (B) Effects of different groups on the growth of HeLa cancer xenografts. (C) Photographs of the mice taken before (day 0) and after 10 and 17 days of chemotherapy with different treatments. Adapted and modified with permission from ref 130. Copyright 2014 WILEY-VCH. To endow nanocarriers with tumor recognition capability and fulfill the requirement of system administration, Shi’s group reported tumor-targeting peptide RGD and TAT co-functionalized ultrasmall (30 nm) MSNs for vasculature-to-cell membrane-to-nucleus-targeted DOX delivery

ACS Paragon Plus Environment

10

Page 11 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

(Figure 4).130 The facile prepared nanosystem can first reach tumor vasculature, and then target the integrin αvβ3 on the cell membrane. After entering cancer cells, the nanosystem can further enter the nucleus under the direction of TAT (Figure 4A). This highly dispersed DOX loaded nanosystem exhibited 98.6% tumor growth inhibition rate during in vivo experiments (Figure 4BC). By introducing magnetic guidance performance, Qu et al. reported a Fe3O4 based triple targeted theranostic system.137 MSN encapsulated Fe3O4 (FMSN) was functionalized with TAT, and the positive charge of TAT was subsequently protected by citraconic anhydride (Cit) and folate (FA) modified charge reversible chitosan (FA-CS-Cit). After intravenously administration (i.v.), these nanoparticles could first accumulate in the tumor site under the guidance of local magnetic field (stage I). Subsequently, the FMSN-TAT/FA-CS-Cit nanoparticles could enter cancer cells under FA receptor mediated endocytosis (stage II). Due to the acid sensitivity of citraconic amides, the protective moiety detached from the FMSN-TAT in lysosomal environments, and the re-exposed TAT finally directed the nuclear targeted delivery (stage III). Anticancer drug CPT could be delivered into the nucleus of cancer cell more effectively, thereby higher therapeutic index and fewer side effects were demonstrated with this smart nanosystem. Moreover, FMSN-TAT/FA-CSCit nanoparticles were used as MRI guided therapy because Fe3O4 nanoparticles are superparamagnetic. Apart from the surface charge, the size distribution of nanoparticles is another key point closely associated with the pharmaceutical parameters of nano-drugs. Since nanoparticles with larger size (~100 nm) are beneficial for long-term circulation and tumor accumulation, while smaller particles are more penetrable in cancer tissue/cells, developing size reducible smart systems is beneficial for in vivo cancer therapy. Huang et al. reported a nanovehicle with stepwise size reduction and

ACS Paragon Plus Environment

11

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 12 of 85

on-demand NLS exposure for systemic delivering drugs into nucleus.142 The large sized nanoparticles (55 nm) were facile assembled from cationic and charge reversal anionic N-(2hydroxypropyl) methacrylamide (HPMA) polymer. The anti-cancer drug DOX and dualfunctional peptide R8NLS (cell-penetrating peptide (R8) and NLS tandem fused peptide) conjugate was connected with cationic HPMA via hydrazone bonds. Benefiting from their appropriate size and surface charge, these nanovehicles possess long-time circulation and enhanced tumor accumulation. After reached tumor tissues, the mild acidic tumor microenvironments caused the disassembly of these NPs into smaller conjugates (10 nm) through charge reversal, the exposed cationic peptides guaranteed enhanced cellular uptake rate. In the endo/lysosomes, these copolymers could release the drug-peptide conjugates owing to the acidic sensitivity of hydrazone bonds and thus facilitated the final intra-nuclear drug transferring. 4.5fold drug accumulation enhancement and higher antitumor efficiency were achieved with this smart drug delivery system. In addition to using NLS peptides152,154,155 (TAT, SV40 large T antigen, oligo-L-lysine and so on) as nuclear targeting ligands, many other strategies have been explored to enhance the intranuclear accumulation of chemotherapeutic drugs.105,107 Yang and co-workers reported a supramolecular drug self-assembly nanosystems for enhanced nuclear-targeted delivery.104 The complexes assembled from negatively charged HCPT-peptide (HP) amphiphile and positively charged cisplatin showed different morphology under different ratios: long nanofibers like complex I was assembled in 1 equivalent of cisplatin while nanoparticle like complex II was prepared in 1.5 equivalent of cisplatin. The cellular membrane barrier was subtly broken by the assemblies due to the formation of nanostructures, while the surface charge of the nanoformulations accelerated the intra-nuclear accumulation of two chemotherapy drugs.

ACS Paragon Plus Environment

12

Page 13 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Therefore, synergistic effects between nanostructures and surface charges rendered the complexes with enhanced drug release kinetics and significantly enhanced the intra-nuclear availability of the chemotherapeutic drugs, which is like the “Trojan Horse” to deliver fighting soldiers (anticancer drugs) into the castle by penetrating the walls (plasma membranes and nuclear envelope). Wu et al. recently demonstrated 1O2 as a novel nucleus envelope opening agent (Figure 5A).138 PEG and rose Bengal (RB) co-functionalized polyhedral oligomeric silsesquioxane (POSS) could self-assembled into nano-photosensitizers (PPR NPs). These nanoparticles first located in the acidic lysosomes after entered A549 cells. Under white light irradiation, the generation of 1O2 accelerated the lysosomal membrane disruption and photosensitizer release. Continuous laser irradiation (2 mW/cm2, 3 min) resulted in the oxidation of the nuclear membranes, accelerating the intra-nuclear photosensitizer accumulation. Molecular agents (10-hydroxycamptothecine, docetaxel and DiD) and inorganic nanomaterials (Prussian blue and gold nanorods) were all successfully delivered into the nuclei with this platform (Figure 5B-C). This method opened new avenue for designing nucleus-targeted drug delivery systems and would inspire more therapeutic systems.

Figure 5. (A) Schematic illustration of the pH-responsive and light-triggered nucleus-targeted delivery. (B) CLSM images of A549 cells stained by Hoechst 33342 after incubation with PPR

ACS Paragon Plus Environment

13

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 14 of 85

NPs (3 μM RB) for 2 h before and after white light irradiation. Scale bars = 10 μm. (C) The corresponding fluorescence intensity profiles of the areas marked by white lines in the confocal images in B. The green-shaded areas indicate the nuclear regions. Adapted and modified with permission from ref 138. Copyright 2018 American Chemical Society. 2.2 Nucleus-targeted photothermal agents Photothermal therapy (PTT), as an emerging cancer treatment modality, has been clinically employed for cancer treatment.43,44,140 The working mechanism of PTT is to induce heat damage in cancer cells through the hyperthermia generated from photothermal conversion agents.156-161 Benefiting from the employment of light, PTT undergoes high spatio-temporal controllability and good repeatability.158-161 However, the cancer cells may rapidly produce large amount of heat shock proteins to enhance their thermal resistant capability.162,163 As a result, long-term and high power density laser irradiation become essential to completely eliminate tumor tissue, which inevitably causes serious side effects to normal tissues due to the heat diffusion. To overcome this “Achilles’ Heel” of PTT, nucleus-targeted PTT has become an alternative modality. Nucleustargeted PTT can in situ denaturalize a large quantity of important enzymes and interdict the cancer proliferation, avoiding the requirement of long irradiation time or high laser power density.118 Vo-Dinh et al. reported the preparation of TAT functionalized gold nanostars for nucleustargeted PTT in vitro.118 Experiments revealed that the gold nanostars specifically accumulated in the nuclear region. Under ultra-low density (0.2 W/cm2, 3 min) laser irradiation, BT549 breast cancer cells were obviously eliminated, while the eliminating efficiency of non-targeted groups were insufficient under the same condition. This report demonstrated nucleus-targeted PTT as a novel therapeutic strategy for enhancing the efficiency of PTT.

ACS Paragon Plus Environment

14

Page 15 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Shi et al. reported the TAT functionalized gold nanorods (GNRs) as PTT agents for cancer therapy under ultralow energy intensity irradiation (Figure 6A-B).128 In vivo experiments revealed the nucleus-targeted GNRs significantly inhibited the growth of xenografted HeLa tumor under 0.2 W/cm2 light treatment for 5 min even though mild temperature increment was detected in the tumor region. The PTT efficiency was comparable to non-TAT modified GNRs (GNRs-NLS) irradiated with a high power density of 2 W/cm2 (Figure 6C-D). This work proved the great promise of nucleus-targeted therapy—it can eliminate malignant tumor under faint laser irradiation, with a density even below the maximal permissible exposure of skin. Besides gold nanomaterials, Fe3O4 has also been investigated for nucleus-targeted PTT. Yang et al. reported the transferrin and TAT co-functionalized Fe3O4 (IONP-20-TAT-Tf) for nucleustargeted PTT.139 Benefiting from the admirable photo-thermal conversion efficiency (about 37%) and 45-fold higher intra-nuclear accumulation, the reported monodispersed nanoparticles effectively ablated tumor xenograft under NIR irradiation (3 W/cm2, 5 min). Meanwhile, the Cy7 modified IONPs are promising candidates for MRI and near infra-red fluorescence imaging.

Figure 6. Schematic illustration of the (A) preparation and (B) nucleus-targeted photothermal therapy of GNRs-NLS. (C) Tumor growth curves of mice after different treatments during PTT

ACS Paragon Plus Environment

15

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 16 of 85

period. (D) Tumor weights of mice in different groups in 15 days of PTT. Schematic Illustration of the (E) synthesis of the CuS@MSN-TAT-RGD NPs and (F) in vivo tumor recurrence-targeted therapy. (G) Photographs of nude mice tumor recurrence with different treatments after tumor resection. Adapted and modified with permission from refs 128 and 140, respectively. Copyright 2017 American Chemical Society and 2018 American Chemical Society. Our group reported nucleus-targeted PTT as a promising strategy for preventing tumor recurrence (Figure 6E-G).140 CuS nanoparticles were employed as the photothermal agent due to its easy preparation, good biocompatibility, high NIR adsorption and molar extinction efficiency. After coating with amino-functionalized MSN layer, dual-targeted CuS@MSN-TAT-RGD can be obtained via further functionalization of TAT and RGD through EDC/NHS coupling reaction. Confocal experiments indicated the nanoparticles effectively accumulated in the nucleus of cancer cells under the guidance of TAT, which ensured the high anticancer efficiency. In vivo fluorescence and photoacoustic imaging experiments revealed the RGD functionalization was a beneficial step for boosting the tumor targeting efficiency of the photothermal agents. As a result, such dual-targeted CuS nano-agent was employed to eliminate the tumor completely with onetime treatment (1.5 W/cm2, 5 min), and also was successfully employed for preventing postoperative recurrence due to their superiority in destroying cancer cell nucleus function. 2.3 Nucleus-targeted photosensitizers Nucleus-targeted photodynamic therapy (PDT) is also promising for highly efficient cancer treatment.122,141,164 The intrinsic mechanism of PDT is that photosensitizers (PSs) can be excited by light with specific wavelength and the excited PSs could transfer energy to the biomolecules, H2O or oxygen molecules in the ground state, denaturalizing bio-species or generating highly reactive oxygen species (O2•−, 1O2, •OH, etc.) to further hamper cellular function.165-167

ACS Paragon Plus Environment

16

Page 17 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Unfortunately, the short diffusion distance and lifespan of ROS or excited PSs, hypoxia and reductive tumor microenvironments restricted the direct damage of critical biomolecules in cancer cells during PDT, resulting in unsatisfied therapy efficiency and requirement of repeated treatment.168 Directly delivering PSs into nucleus offers extraordinary superiority for PDT, which is beneficial for overcoming drug resistance, hypoxia and recurrence due to the direct dysfunction of nucleus with abundant ROS is a more powerful cancer therapy strategy. Ce6 is one of the most investigated PSs in PDT, it can generate 1O2 under red light irradiation in the existence of O2. However, the weak solubility, poor in vivo distribution of Ce6 and analogous molecular PSs sparked the interest of finding novel carriers for boosting the tumor selectivity and PDT efficiency. Just like traditional chemotherapeutic drugs, molecular PSs could also be expelled out by P-gp from the drug resistant cancer cells.122 Thus driving molecular PSs into nucleus has emerged as an alternative strategy for enhanced PDT effects.115 On the basis of previous work, Shi et al. reported the nucleus-targeted delivery of Ce6 with MSNs.141 MSNs were employed as the vehicles owing to their properties of easy preparation, high morphology, size and surface area controllability, ideal physiochemical stability and biocompatibility. The TAT and RGD cofunctionalized 30 nm MSNs showed admirable vasculature to nuclear targeting capacity, and significantly potentiated the PDT efficiency due to the in situ intra-nuclear ROS generation and DNA destroy. Extraordinary high tumor inhibition efficiency under ultralow power density (0.02 W/cm2) irradiation of 5 min was demonstrated, which also indicated the nucleus-targeted PDT systems had potential for deep-seated tumor treatment. In 2016, Han and co-workers demonstrated an amphiphilic chimeric peptide (PAPP-DMA) for tumor microenvironment activated nucleus-targeted PDT (Figure 7A-B).164 The peptide was constructed from alkylated protoporphyrin IX, PEG and 2,3-dimethylmaleic anhydride (DMA)

ACS Paragon Plus Environment

17

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 18 of 85

shielded NLS peptide PKKKRKV. The amphiphilic property enabled the peptides self-assembled into nanoparticles smaller than 200 nm. The zeta potential of assembled nanoparticles was −19.1 mV after DMA shielding, which is beneficial for enhancing the blood circulation period. In the acidic tumor microenvironment, DMA detached from the PKKKRKV, resulted in the surface charge of nanoassemblies reversion, which enhanced cancer cell internalization, and realized intranuclear photosensitizer accumulation after the re-exposure of cationic NLS peptide. Enhanced cancer PDT efficiencies were demonstrated both in vivo (200 mW/cm2, 10 min) and in vitro (10 mW/cm2, 2 min). (Figure 7C-E)

Figure 7. Schematic illustration of (A) tumor and intranuclear delivery of photosensitizer for enhanced PDT and (B) the synthesis of PAPP-DMA and the detachment of DMA in the acidic environment. (C) Relative tumor volume after different treatments. (D) Post-sacrifice tumor images at the 11th day. (E) Average tumor weight at the 11th day. Adapted and modified with permission from ref 164. Copyright 2016 WILEY-VCH. Since molecular PSs often suffer from efflux effects caused by the over expressed P-gp, nanoPSs should offer more opportunities for highly efficient PDT. Recently, our group reported a nucleus-targeted NIR responsive dual-PS with multiple ROS generation capacity for tumor

ACS Paragon Plus Environment

18

Page 19 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

treatment (Figure 8A-D).122 Molecular PS Ce6 was decorated onto the surface of pre-synthesized nanoscale core/shell upconversion@TiO2. Then nucleus-targeted peptide TAT was further decorated onto the surface of the dual-PS to obtain the nucleus-targetable UCNPs@TiO2-Ce6TAT. The successful FRET effects between UCNPs and TiO2 as well as Ce6 resulted in multiple ROS (O2•−, 1O2 and •OH) generation under 980 nm laser irradiation. Accurate nuclear location and efficient 980 nm laser triggered nuclear DNA damage was realized with UCNPs@TiO2-Ce6-TAT (Figure 8E-F). In vivo tests (1 W/cm2, 5 min) revealed the nucleus-targeted dual-PS could be used as a novel candidate for the treatment of drug resistant tumor (Figure 8G-H).

Figure 8. (A) Schematic illustration of the synthetic process of UCNPs@TiO2-Ce6-TAT, multiple ROS generation under NIR laser excitation and inducing DNA double strand breaks. High resolution TEM images of (B) oil acid coated UCNPs; (C) UCNPs; (D) UCNPs@TiO2; Scale bars are 25 nm. (E) Colocalization images of UCNPs@TiO2-Ce6-TAT and the nuclei. (F) The quantification of fluorescence intensity of the line scanning profiles of the corresponding confocal

ACS Paragon Plus Environment

19

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 20 of 85

images in E. (G) Photographs of the xenograft MCF-7/Dox tumor mice taken before treatment (0 days) and at 14 days with different treatments: control, UCNPs@TiO2-Ce6, UCNPs@TiO2-TAT, and UCNPs@TiO2-Ce6-TAT with irradiation for 5 min in all groups; H) Tumor growth curves of different treatment groups. Adapted and modified with permission from ref 122. Copyright 2016 Royal Society of Chemistry. 2.4 Nucleus-targeted gene silence systems Direct silencing of pathogenic genes or introducing therapeutic genes into cells, which were called gene therapy, have become a popular strategy for treating series of major disease including cancer in recent years.169 Gene therapy can be divided into the transcriptional level and the posttranscriptional level. Transcriptional level involves knocking down the DNA in the nucleus to stop the expression of diseased-related proteins, post-transcriptional level is to silence mRNA, siRNA or microRNA in the cytoplasm.155,169 Because of the easy degradation of therapeutic genes in cytoplasm, nucleus-targeted gene silencing systems should be better choices due its advantages of long-term silencing effects and avoidance of repetitive administration of post-transcriptional gene silencing agents. However, the major question regarding nucleus-targeted gene therapy is the absence of appropriate vehicles which could be capable of overcoming several intracellular barriers and deliver enough genes into nucleus. In 2014, Shi et al. reported the nucleus-targeted delivery of plasmid DNA with a micelle/precursor

co-templating

assembly

method

prepared

mesoporous

organosilica

nanoparticles (MONs) (Figure 9A-B).170 Benefiting from their large pore volume and small size, the MONs exhibited significantly reduced hemolytic effect and improved biocompatibility, which were used for highly efficient intra-nuclear gene delivery after surface modified with PEI and TAT (MONs-PTAT). Owing to the good plasmid DNA binding and protecting capacity, the transfection

ACS Paragon Plus Environment

20

Page 21 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

efficiency of the dual-modified carriers was as high as 36.5%, which was significantly higher than PEI (13.5%) or TAT (4.97%) mono-functionalized groups. The successful construction of organicinorganic hybrid nanoparticles with controllable structure and composition provided new opportunities to design new platforms for nucleus-targeted gene therapy. Recently, our group demonstrated nucleus-targeted delivery of siRNA with TAT modified 13 nm gold nanoparticles (AuNPs) to directly initiate the cancer associated DNA methylation.155 The nucleus-targeted gene delivery system was suggested could deliver siRNA into the nucleus of diverse cell lines, including MCF-7, HeLa and HepG2. The siRNA dramatically interacted with the promoter of cancer related Thymidine Kinase 1 (TK1), and long-term gene silencing was achieved due to the efficient suppression of the TK1 protein and TK1 mRNA expression. The silencing time was tested to be longer than 30 days, which enormously inhibited the in vivo tumor formation.

Figure 9. Schematic illustration of (A) the synthesis process and (B) the nucleus-targeted gene delivery of the MONs-PTAT@pDNA. Adapted and modified with permission from ref

170.

Copyright 2014 WILEY-VCH.

ACS Paragon Plus Environment

21

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 22 of 85

Figure 10. Schematic diagram of (A) the synthesis process of LACP and (B) laser-enhanced knock-outs of targeted genes by LACP in A375 cells. (C) Sizes of tumors treated by different CP formulations. (D) Weights of tumors treated by different CP formulations. Adapted and modified with permission from ref 143. Copyright 2018 WILEY-VCH. CRISPR systems are recently emerging gene editing techniques, which have been reported to be useful toolbox for a series of genetic diseases. However, the intracellular transfection efficiency as well as specificity are main challenges for their practical application. Compared to virus vehicles, nano-transfection agents hold virtues of easy preparation, high designability and controllability. Jiang’s group143 (Figure 10A-B) reported laser controlled nucleus-targeted delivery of Cas9-sgPlk-1 plasmids (CP). 20 nm cationic gold nanoparticles (AuNPs) were prepared via decorating AuNPs with TAT peptides, which were employed to condense CP through electrostatic interaction. After further encapsulation with lipids, multi-AuNPs-lipid complex (LACP) with good solubility was obtained. There are 4-15 AuNPs in each LACP, the plasmid encapsulation efficiency was 97%, under 514 nm laser irradiation for 20 min (24 mW/cm2), 79.4% CP was

ACS Paragon Plus Environment

22

Page 23 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

released due to the photothermal conversion effect of the Au core, the released CP could enter the nucleus under TAT guiding. By virtue of the enhanced intracellular uptake and controlled release manner, LACP exerted enhanced antitumor effect during the intratumoral injection (i.t.) experiments (Figure 10C-D). 2.5 Nucleus-targeted combined therapy systems Rational combination of two or more treating modality is helpful to potentiate the therapy index in many resistant cancers. Nucleus-targeted combined therapies have also been reported recently.144,147,148,171 Hwang et al. reported the simultaneous nucleus-targeted PDT and gene delivery with NIR responsive gold clusters.147 The TAT modified gold clusters (TAT peptide-Au NCs) possessed admirable photostability, biocompatibility and nucleus-targeting capacity. Compared to commercial LP2000 liposome gene carrier, 3.2-fold higher gene transfection efficiency was demonstrated owing to the enhanced cellular uptake and nucleus accumulation. TAT peptideAu NCs were also demonstrated as novel photosensitizers because they could cause intra-nuclear DNA damage by the 980 nm laser induced ROS (500 mW/cm2, 30 min). Therefore, TAT peptideAu NCs was a powerful platform for imaging guided nucleus targeted gene and PDT synergistic therapy. It is well known that many tumors are radiotherapy (RT) resistant due to their intrinsic hypoxia environments, and some cancer cells could stay at RT-insensitive phase (such as G1 and S phase). Therefore, delivering radiosensitizers into the nucleuses of cancer cells have intrinsic superiority. Further combination of nucleus-targeted RT with other treatment modality may cause extra efficiency. Shi’s group realized the nuclear targeted combined chemo-radio therapy with sub 50 nm yolk-shell structured upconversion-mesoporous silica (Figure 11A).144 Chemotherapy drug Mitomycin C, a type of DNA toxin, was loaded into the rattle-like nanoparticles and served as the

ACS Paragon Plus Environment

23

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 24 of 85

radiosenstitizer. Such nucleus-targeted chemo-radio therapy platform significantly enhanced the RT efficiency (8 Gy, 5 min) compared to monotherapy strategy or cytoplasmic localized combined therapy (Figure 11B-C). As the mechanisms of RT and PDT are both ROS inducing biomolecules damage, combining nucleus-targeted PDT and RT may also reduce the radio-dosage and irradiation time. Shi’s group further demonstrated the nucleus-targeted combined photodynamic-radio therapy with a UCNPs based core-shell nanocomposite (Figure 11D).171 By rational covalently coating/separation of radiosenstizer and photosensitizer on the out silica layers of UCNPs and further PEG/TAT functionalization, nucleus-targeted UCSPs-PEG/TAT nanoparticles were obtained to generate abundant ROS under either NIR or X-ray irradiation. As a result, synergistic enhancement effect was observed in the radioresistant HT-1080 cells. Significant tumor growth suppress effect was demonstrated (6 Gy, 5 min for RT; 1.5 W/cm2, 20 min for PDT), which further verified the significant potential of nucleus-targeted combination therapy (Figure 11E).

ACS Paragon Plus Environment

24

Page 25 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Figure 11. (A) Schematic illustration of nucleus-targeted chemo combined radiotherapy. (B) Tumor growth curves of MCF-7 tumor-bearing mice over a period of half a month after different treatments. (C) Relative MCF-7 tumor volumes of different groups after different treatments. Adapted and modified with permission from ref 144. Copyright 2015 Royal Society of Chemistry. (D) Schematic illustration of the nucleus-targeted PDT-RT combined therapy using UCSPsPEG/TAT. (E) Relative tumor volumes of HT-1080 tumor-bearing mice during half a month after different treatments. Adapted and modified with permission from ref 171. Copyright 2015 Elsevier Ltd. 3. Mitochondria-targeted therapy Mitochondrion is a two-layer membrane-coated organelle with the diameter of 0.5-1.0 μm.84 As the “power house” of eukaryotic cells, it takes charge of energy production, electron transport, calcium metabolism, ROS generation and even immunity regulation.172-174 Targeted delivery of therapeutic drugs to mitochondria and efficiently regulating mitochondrial function may offer great potentials for efficient tumor treatment, which should have diverse advantages over traditional cytoplasmic drug delivery modality on account of the extremely high biological significance of mitochondria. Interestingly, mitochondria possess negative potential because the protons intra mitochondria are pumped into the mitochondrial intermembrane space, and cancer cells usually undergo stronger mitochondrial transmembrane potential (-180 mV).173,175 Therefore, lipophilic molecular or nanodrugs with delocalized cationic structures may effectively accumulate into mitochondria.87 Some peptides or functional groups with high affinity to the mitochondrial protein have also been used for vehicle modification to deliver specific drugs into the mitochondria. (Table 2) In this section, we will introduce the recent advances of mitochondria-

ACS Paragon Plus Environment

25

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 26 of 85

targeted therapeutic agents for chemotherapy, PDT, PTT, RT, and combined therapy with enhanced therapeutic efficiency. Table 2. Typical nanomaterials for mitochondria-targeted therapy. Therapy modality

Nanomaterials

Cell type

Tumor model

Administration route (i.v./i.t.)

Treatment times

Treatment period

Refs

Chemotherapy



HeLa, MCF-7









175

Mitochondrial transmembrane potential mediated accumulation



Drug-resistant A549/cDDP

drug-resistant A549/cDDP

i.v.

5

26

176

TPP

GSH triggered TPP exposure

5.22

MCF-7

MCF-7

i.v.

1

18

177

UCNPs@TiO2

TPP

Mitochondrial transmembrane potential mediated accumulation



MCF-7

MCF-7

i.t.

1

14

178

PS-peptide NPs

MLS

ROS accelerated cellular internalization, cationic hydrophobic peptide mediated targeting

40

HeLa

H22

i.v.

2

10

179

AIE dots

FA, TPP

Receptor mediated targeting, mitochondrial transmembrane potential mediated accumulation

10

MCF-7









180

Fe3O4

TPP

Mitochondrial transmembrane potential mediated accumulation



HeLa

HeLa

i.v.

1

21

181

Photothermal molecule loaded F127 NPs

Biotin, TPP

Receptor mediated targeting, mitochondrial transmembrane potential mediated accumulation



4T1

4T1

i.v.

1

14

182

RT

TiO2–Au

TPP

Mitochondrial transmembrane potential mediated accumulation



MCF-7, 4T1

MCF-7, 4T1

i.t.

1

14

183

Combined therapy

MOFs

Ru(bpy)32+

Mitochondrial transmembrane potential mediated accumulation

20.13 ± 8.58 µM

MC38

MC38

i.v.

1

22

184

PDT

PTT

Targeting ligands

Targeting mechanisms

Nanodiamonds

FA, MLS peptide

Receptor and protein mediated targeting

Liposome

TPP

PLGA

IC50 (μg/mL)

3.1 Mitochondria-targeted chemotherapeutic agents As mentioned above, most chemotherapeutic drugs are DNA replication associated toxins. It is well known that mitochondria also contain DNA (mtDNA) and these double-stranded circular DNA molecules take charge of the synthesis of tens of proteins.185 Thus, mitochondria should be another critical target for chemotherapy, and delivering DNA toxins into mitochondria is another wise choice toward enhanced chemotherapy.119,186-194 Platinum based drugs are famous DNA crosslinker, which can interact with DNA effectively and stop the proliferation of mammalian cells. However, the advanced DNA repair systems in nucleus (nucleotide excision repair, NEP) can cause cancer metastasis and drug resistance during

ACS Paragon Plus Environment

26

Page 27 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

traditional chemotherapy.195 In comparison, the mtDNA can be destroyed by platinum based drugs more easily due to the lack of NEP system in mitochondria. Transferring DNA toxins into the mitochondria has opened a new avenue for potentiated cancer therapy. In 2014, Dhar and coworkers constructed a mitochondria-targeted platinum prodrug delivery system for detouring the intra-nuclear DNA repair (Figure 12A-B).195 The lipophilic cationic triphenylphosphonium (TPP) functionalization of cisplatin prodrug Platin-M was conducted by the strain-promoted alkyne-azide cycloaddition reaction, making the prodrugs able to diffuse into the mitochondria under the driving of transmembrane potential. To further achieve in vivo application, terminal TPP functionalized decomposable poly (lactic-co-glycolic acid)-block-polyethyleneglycol was prepared as the delivery vehicle. The optimized drug loading nanoparticles possessed long blood circulation time and high brain-penetrating properties, thus the nanoparticles could deliver and release abundant Platin-M into the mitochondria of neuroblastoma cells (Figure 12C-F). About 17-fold higher anticancer activity toward cisplatin resistant cells could be realized by this nanomedicine compared to the original cisplatin, suggesting that the targeted delivery of DNA toxins into the mitochondria is rather favorable for drug resistant cancer therapy application.

Figure 12. (A) Schematic diagram for mitochondria-targeted delivery of cisplatin prodrug using a targeted NP and the mechanism of action. (B) Synthesis of mitochondria-targeted Pt(IV) prodrug Platin-M. (C-F) Pt concentration variation in plasma and organs following the i.v. administration

ACS Paragon Plus Environment

27

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 28 of 85

of different NPs to male rats. Adapted and modified with permission from ref 195. Copyright 2014 National Academy of Sciences. Another important first-line chemotherapeutic drug, DOX, has also been reported to be significantly potentiated when delivered into mitochondria. Direct functionalization of chemotherapeutic drugs might compromise the bioactivity, thus novel nanovehicles are more optimal. Lo’s group reported the specific delivery of DOX into the mitochondria of cancer cells via functionalized nanodiamonds.175 The FA and mitochondria localizing sequence (MLS) peptides co-functionalized nanodiamonds possessed dual targeting capability. They first distinguished the cancer cells from normal cells based on the different expression levels of FA receptor. Subsequently these nanodiamonds could accumulate into the mitochondria under the guidance of MLS. With this system, the cellular drug uptake rate was dramatically increased, and the drug resistant MCF-7 cells were successfully killed owing to the enhanced mitochondrial DOX accumulation. Besides DNA toxins, many other chemotherapeutic drugs also take mitochondria as their targets, such as paclitaxel (PTX), a hydrophobic drug extracted from western taxus brevifolia. Lu et al. reported a mitochondria-targeted delivery system based on PTX liposomes.176 Mitochondriatargetable PTX liposomes were prepared via anchoring the mitochondria-targeted macromolecule TPGS1000-TPP to the liposomes and used for treating drug-resistant lung cancer. Owing to the suitable size distribution, prolonged blood circulation time, high cancer cell uptake rate and mitochondrial-specific PTX accumulation, the delivery system successfully induced the apoptosis of drug-resistant lung cancer through the activation of the mitochondrial apoptosis pathways ( including release of the cytochrome C, initiate of the cascade of caspase-9 and caspase-3 reactions,

ACS Paragon Plus Environment

28

Page 29 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

activation of the pro-apoptotic Bax and Bid proteins, and suppression of the anti-apoptotic Bcl-2 protein). To further improve the in vivo performances of liposomes, Gu and co-workers reported the dualfunctional liposomes for tumor pH/mitochondria-dual-targeted PTX delivery (Figure 13).196 DMA modified peptide

D[KLAKLAK]2

(KLA) was combined with 1, 2-distearoyl-sn-glycero-3-

phosphoethanolamine (DSPE) to obtain the dual-functional lipid (DKD). The DKD was further mixed with commercial lipids to construct the dual-functional liposomes. The surface charge of these liposomes was negative under healthy extracellular pH (7.4), which could reduce the protein adsorption and prolong the blood circulation time. Then the surface charge could be converted into positive in acidic tumor microenvironments due to the detachment of DMA (Figure 13A-B), facilitating the cancer cellular uptake. After entering lysosomes, the KLA peptide complete exposed and guided the mitochondrial PTX accumulation as well as subsequent drug release. These features endowed the smart liposomes (DKD/PTX-Lip) with high tumor growth inhibition (86.7%) in the drug-resistant lung cancer cells xenografted nude mice (Figure 13C-D), further proving the significance of mitochondria-targeted therapy.

Figure 13. Left: illustration of dual-functional liposome with pH response to the tumor microenvironment and mitochondria-targeted anticancer drug delivery. (A) Size distribution of

ACS Paragon Plus Environment

29

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 30 of 85

PTX-loaded DKD-Lip (DKD/PTX-Lip). (B) Zeta potential of DKD-Lips at different pH values within 12 h. (C) Tumor volume of mice after different treatments. (D) Tumor growth inhibition rates of the treatment groups. Adapted and modified with permission from ref 196. Copyright 2015 Elsevier Ltd. Redox responsive nanocarrier has been reported by Huang et al. for mitochondria-targeted drug delivery.177 FDA approved PLGA was used as the carrier of PTX, and TPP-modified amphiphilic polymer (C18-PEG2000-TPP) was selected as the mitochondria targeting group. To shield the positive charge of cationic TPP, disulfide bond connected amphiphilic polymer DLPE-S-SmPEG4000 was employed as the protective moiety, which could completely shield the positive charge of TPP and enable the drug delivery system to be injected in vivo. In the cytoplasm of cancer cells, TPP was re-exposed due to the cleavage of disulfide bond and subsequent detachment of PEG4000, which made the carriers capable of accumulating into the mitochondria and resulted in enhanced mitochondrial PTX accumulation. This system effectively activated the apoptosis pathway of MCF-7 cells and higher in vivo tumor suppression efficiency was realized compared to the non-responsive group. 3.2 Mitochondria-targeted photosensitizers Mitochondria-targeted PSs delivery is another novel strategy for boosting the PDT efficiency. Mitochondria are the main places for ROS production, which hold a series of regulation systems for maintaining cancer cellular homeostasis. Mitochondrial ROS overexpression may disrupt the dynamic balance of mitochondrial microenvironments and cause serious mitochondrial dysfunction due to the oxidation stress.197 Mitochondria-targeted ROS generation is therefore highly attractive for enhanced cancer treatment owing to the potential in reducing the photo/PSs dosage and side effects.178,198 Up to now, a variety of PSs with mitochondria-localization capacity

ACS Paragon Plus Environment

30

Page 31 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

have been designed and synthesized for enhanced PDT, which could be roughly divided as porphyrins, transitional-metals, aggregation-induced emission dyes and others.199-209

Figure 14. Schematic illustration of (A) the structure of the nano PS and ROS generation and (B) the NIR triggered nanophotosensitizer inducing domino effect on mitochondrial ROS burst for cancer therapy. Cell viability of MCF-7cells incubated with (C) UCNPs@TiO2-TPP and (D) UCNPs@TiO2 plus with 980 nm laser irradiation for different time. (E) Tumor growth curves and (F) mice body weight curves of different groups during 14 days. Adapted and modified with permission from ref 178. Copyright 2015 American Chemical Society. In 2015, our group reported the titanium dioxide-coated UCNPs (UCNPs@TiO2-TPP) for mitochondria-targeted PDT (Figure 14A-B).178 The nano-PSs possessed uniform size distribution and upconversion feature. Under 980 nm laser irradiation, it could effectively convert NIR light into UV light and activated TiO2 to generate abundant O2•−. Interestingly, the mitochondriatargeting capacity of UCNPs@TiO2-TPP make it rather favorable for highly efficient PDT due to

ACS Paragon Plus Environment

31

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 32 of 85

the induction of domino effect on ROS burst. When the mitochondrial ROS concentration reached a threshold value, the mitochondrial function could be disrupted, and triggered continuous ROS generation, thus further resulted in the activation of apoptosis pathway (including the reduction of mitochondrial membrane potential, release of cytochrome C, activation of caspase-3 and caspase-7 and so on) (Figure 14C-D). Upon 90 s NIR light (3 W/cm2) irradiation, the tumor growth of treating group was completely suppressed, confirmed mitochondria-targeting as a powerful tool for highly efficient cancer treatment (Figure 14E-F). Porphyrin and its derivatives are famous PSs suffering from poor solubility, easy photobleaching, and low efficiency. Mitochondria-targeted nanocarriers offered new chance to the PDT efficiency of porphyrin based PSs. Wu et al. reported a dual-targeted strategy to enhance the PDT efficiency of cancer.210 Cationic porphyrin derivative (MitoTPP) was effectively loaded onto the FA functionalized PEG modified nanographene oxide via π-π staking. In this PDT system, the fluorescence of MitoTPP was quenched initially and almost no 1O2 could be detected during laser irradiation, which reduced the potential side effects of the PSs. After entering the FA receptor overexpressed HeLa cells, MitoTPP could be released in the acidic lysosomal microenvironments and subsequently accumulated into the mitochondria under the guidance of TPP. Under light irradiation (10 mW/cm2, 30 min), in situ accumulation of 1O2 resulted in remarkable cancer cell apoptosis due to the induction of mitochondrial oxidation damage. Zhang’s group demonstrated a self-assembled system (PPK) for mitochondria-targeted PDT (Figure 15).179 Protoporphyrin IX (PpIX) was covalently linked with an amphiphilic peptide (KLAKLAK)2 (a mitochondria-targeted proapoptosis peptide) through a short PEG linker, the Ce6 functionalized peptides further self-assembled into spherical nanoparticles with narrow size distribution and good solubility (Figure 15A-B). These nanoparticles possessed photochemical

ACS Paragon Plus Environment

32

Page 33 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

internalization (PCI) feature under short light irradiation: they showed enhanced cell penetrating capacity due to the short-time laser irradiation resulted ROS−induced lipid peroxidation. After 6 min laser irradiation, the PSs entered cancer cells under ROS assistance and accumulated in the mitochondria effectively. More ROS generated here under 18 min light irradiation and resulted in cancer cell apoptosis. MTT assay revealed the cancer inhibition efficiency of the PSs combined with two steps (6+18 min) laser irradiation was higher than one-step 24 min irradiation. The system achieved higher in vivo anticancer efficiency owing to the synergetic mitochondrial-targeted PDT (220 mW/cm2, 20 min) and proapoptosis effect (Figure 15C-D).

Figure 15. Left: Chemical structure of PPK and schematic diagram of the mitochondria-targeted self-delivering process. (A) Hydrodynamic size and (B) TEM image of PPK in DI water. (C) Relative tumor volume after post-treatment. (D) The average tumor weight at the 12th day after different treatments. Adapted and modified with permission from ref 179. Copyright 2015 WILEY-VCH. In 2016, a programmed supramolecular system was reported by Ji and co-workers.211 Ce6 modified β-Cyclodextrin (β-CD) interacted with the adamantine (Ad) terminated tri-blocked peptide (Ad-CGKRK-GFLG- EE-T7) and further self-assembled into well-defined nanostructures.

ACS Paragon Plus Environment

33

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 34 of 85

T7 with highly affinity toward the cancer cell overexpressed transferrin receptor (TfR) endowed the nanosystem with high cancer cell uptake rate. Cathepsin B (CTSB) cleavable GFLG moiety and the mitochondria-targeted CGKRK peptide imposed the system responsive mitochondriatargeting capability. Therefore, higher PDT efficiency (IC50 = 0.9 mg L-1) was realized over nontargeted delivery systems, suggesting that the reported system can be used for highly efficient and specific cancer therapy. AIE-based PSs for mitochondria-targeted PDT have attracted increasing consideration recently, owing to their excellent photostability and the capability for overcoming aggregation-caused quenching phenomenon.207,212-215 Liu and Tang et al. reported an AIE dots for cancer cell-tomitochondria-targeted therapy.180 Dual-targeted AIE dots could be easily obtained by using lipidPEG as the encapsulation agent, FA and TPP as targeting group. The dual-targeted dots show enhanced PDT efficiency (100 mW/cm2, 10 min). The value of IC50 was 10 µg mL−1, which is lower than cellular or mitochondria-targeted nanoparticles owing to their higher mitochondrialaccumulation capability. By integrating TPP contained AIE based polymer with Tm3+ doped UCNPs, Gao et al. reported the first NIR activated mitochondria-targeted AIE based PDT system.212 The positive charge of this system was shielded by pH sensitive PEG layer (UCNP@PAIE-TPP-PEG) via benzoic imine bonds, which could avoid side effects caused by TPP. After entering cancer cells, the imine groups were split, and the UCNP@PAIE-TPP accumulated into mitochondria effectively. Under 980 nm laser irradiation (3W/cm2, 10 min), abundant ROS production resulted in mitochondrial ROS collapse as well as cancer cell apoptosis, suggesting that UCNP@PAIE-TPP-PEG is potential for enhanced in vivo cancer treatment (Figure 16 Left). Chao et al. reported the first example of AIEactive iridium(III) complexes (Ir1-Ir3) for mitochondria-targeted PDT (Figure 16 a-b).213 Ir1

ACS Paragon Plus Environment

34

Page 35 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

exhibits the most significant two-photon adsorption cross-sections (214 GM) and the most impressive lethality toward cancer cells, which make it an ideal candidate for highly efficient PDT (IC50 = 0.40 μM).

Figure 16. Left: schematic illustration for the pH responsive UCNP@PAIE-TPP-PEG NPs for mitochondria-targeted PDT. Adapted and modified with permission from ref 212. Copyright 2017 American Chemical Society. Right: (a) the structures of Ir1-Ir3. (b) Schematic illustration of mitochondria-targeted TPA-PDT with Ir1-Ir3. Adapted and modified with permission from ref 213. Copyright 2017 Royal Society of Chemistry. 3.3 Mitochondria-targeted photothermal agents In addition to chemotherapeutic drugs and ROS, mitochondria are also hypersensitive to heat. Mitochondria-targeted PTT can effectively reduce the activity of enzymes, disrupt the mitochondrial respiration, cause mitochondrial thermal damage, and further induce oxidation stress and activate mitochondrial apoptosis pathways. Therefore, design and preparation of mitochondria-targeted photothermal agents are promising for improving the tumor therapy efficiency, suppressing tumor growth and avoiding recurrence. In 2015, Kim et al. reported an iron oxide nanoparticle for mitochondria-targeted PTT.216 TPP coupled coumarin was surface functionalized onto iron oxide NPs via catechol group, yielding the

ACS Paragon Plus Environment

35

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 36 of 85

mitochondria-targeted “nanoheater” (Mito-CIO). Confocal co-localization experiments revealed that Mito-CIO localized in mitochondria with a high Pearson’s coefficient (0.871), while the coumarin modified iron oxide (CIO) mainly located in the endoplasmic reticulum. Under 740 nm NIR irradiation (2W/cm2, 10 min), Mito-CIO induced higher in vivo therapy efficiency than nontargetable CIO, demonstrating the superiority of mitochondrial-targeted PTT. It is well known that NO is an important cellular signal molecule. Employing gas signal molecules (NO, CO, H2 and so on) for sensitized cancer therapy has been reported to be an alternative modality. In 2017, Liu et al. developed a Ru nitrosyl and TPP co-functionalized graphene quantum dots (N-GQDs@Ru-NO@TPP) for mitochondrial-targeted PTT.217 The TPP modification imposed the nanoplatform with ideal mitochondrial-targeting capability and the photothermal effect of GQDs ensured the 808 nm laser irradiation triggered NO release. Benefiting from the synergistic effect between NO sensitizing and mitochondria-targeted PTT, this nanoplatform exerted admirable tumor growth inhibition capability and the tumor could even be eradicated after single treatment (1.0 W/cm2, 10 min). Compared to nanomaterials, molecular photothermal agents with high photothermal conversion efficiency are also quite attractive, owing to their well-defined structure and performances. Mitochondria-targeted molecular photothermal agents may accelerate the clinical translation of mitochondria-targeted PTT. However, the in vivo PTT of the cationic photothermal agents are restricted due to their poor tumor targeting capabilities, thus efforts should be devoted to realize the mitochondria-targeted delivery.218 The first example of cancer cell-to-mitochondria-targeted photothermal agent was reported by our group (Figure 17A-B).182 A cyanine derived biotin and TPP co-functionalized photothermal agent was designed and synthesized. To improve the water solubility, FDA approved F127 was employed to encapsulate the photothermal agents to obtain

ACS Paragon Plus Environment

36

Page 37 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

the tumor-mitochondria-targeted photothermal nano agents (Bio-PPh3-PT). The photo-to-thermal conversion efficiency of the prepared photothermal molecule was 37.8%. In vitro experiments demonstrated that the dark toxicity of Bio-PPh3-PT was negligible while the phototoxicity remained high, mostly owing to the biocompatibility of organic small molecules as well as the thermal sensitivity of mitochondria. In vivo fluorescence and photoacoustic imaging experiments revealed the good tumor targeting capability (Figure 17C-E). More importantly, higher cancer treatment efficiency was demonstrated during in vivo anticancer therapy (0.5 W/cm2, 5 min) and no significant side effects can be detected (Figure 17F-H).

Figure 17. (A) Schematic illustration of the dual-targeted PTT. (B) The molecular structure of Bio-PPh3-PT. (C) In vivo fluorescence imaging of 4T1 tumor-bearing mice after i.v. injection of Bio-PPh3-PT or PT at different time points and (D) the related normalized fluorescence intensities. (E) Photoacoustic imaging of 4T1 tumor-bearing mice after i.v. injection of Bio-PPh3-PT. (F)

ACS Paragon Plus Environment

37

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 38 of 85

Photographs of tumor-bearing mice with different treatments at day 1 and day 15. (G) Tumor growth curves and (H) the mice body weight curves of different groups. Adapted and modified with permission from ref 182. Copyright 2019 WILEY-VCH. 3.4 Mitochondria-targeted nano radio-sensitizers Clinically employed RT mainly use ionizing radiations to destroy the DNA and protein/enzymes of cancer cells to inhibit the growth of tumors. Nevertheless, the low tissue adsorption of tumor tissues toward radiations resulted the requirement of high energy and long-time radiation exposure, which usually resulted in serious side effects and limited therapeutic index.183 Moreover, chemotherapeutic drugs are frequently employed by doctors to ensure the elimination of cancer cells as more as possible, which usually cause extra burdens for cancer patients.219,220 In practice, cancer cells may stay in the RT insensitive phase, and the hypoxia tumor microenvironments may severely compromise the therapy efficiency. Therefore, it has sparked the interest of design and synthesis of radio-sensitizers, to enhance the radiation sensitivity of tumor tissues.219,220 The current methods for radio-sensitization including hypoxia relief, combination with other modality as well as radiation energy conversion enhancement with diverse sensitizers. Driving nano radiosensitizers into mitochondria is another wise choice toward enhanced cancer treatment. Recently, our group reported a mitochondria-targeted radio-sensitizer for triggering mitochondrial ROS burst with admirable RT efficiency (Figure 18).183 The ultra-small (3-5 nm) AuNPs modified 18 nm TiO2 showed enhanced O2•− productivity. After further modification with mitochondria-targeting ligand TPP, the obtained nano radio-sensitizer TiO2-Au-TPP NPs could effectively accumulate into the mitochondria. The mitochondria-specific ROS generation after Xray irradiation was observed and domino effect on ROS burst was demonstrated, which significantly potentiated the therapeutic effects of RT (Figure 18A-B). Upon one-time 6 Gy X-

ACS Paragon Plus Environment

38

Page 39 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

rays irradiation, MCF-7 tumor-baring nude mice as well as 4T1 tumor-baring Balb/c mice were effectively cured, suggesting that the mitochondria-targeted RT as an ultra-attractive cancer treatment modality (Figure 18C-E). High-Z element doping is another important strategy for improve the RT efficiency of nanomaterials. A mitochondria-targeted Gd-doped titania nanoparticles (TiO2(Gd)-TPP) was further developed by our group for enhanced cancer RT. Compared to the non-targeted group, the RT efficiency was effectively elevated by TiO2(Gd)-TPP owing to the generation of abundant ROS intra-mitochondria.221

Figure 18. Schematic illustration for (A) the preparation of the TiO2-Au-TPP NPs and (B) mitochondria-targeted RT with TiO2-Au-TPP NPs. Photographs of (C) the tumor bearing nude mice at 0 days and 14 days with different treatments. (D) The tumor growth curves and (E) body weight curves of different groups. Adapted and modified with permission from ref 183. Copyright 2018 Royal Society of Chemistry. 3.5 Mitochondria-targeted combined therapy systems

ACS Paragon Plus Environment

39

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 40 of 85

Combining two or more treating modality is another bright pathway for further leveraging the therapeutic index of mitochondria-targeted cancer treatment, which could avoid the inherent disadvantages and limitations of single treatment modality. Therefore, many efforts have been devoted to mitochondria-targeted combined therapy to further improve the therapeutic index of advanced tumors. Up to now, mitochondria-targeted chemo-PTT, chemo-PDT, PTT-PDT, and trimodal therapy, have been investigated.90,96,97,145,181,184,215,222-235 Zhang et al. reported a gold nanostar (AuNS) based nanovehicle for chemo-PTT.229 Mitochondria-targeting pro-apoptotic peptide TPP-KLA, biocompatible PEG5000 and cationic peptide R8 were attached onto AuNS via Au-S bonds. Then the nanocomposites and DOX were co-encapsulated by the cellular targeting ligand, hyaluronic acid, yielding the multi-targeted nanodrug AuNS-pep/DOX@HA. AuNS-pep/DOX@HA effectively localized in the tumor site via EPR effect and CD44 receptor-mediated recognition. After entered cancer cells, HA was degraded by HAase and DOX was released. The AuNS further accumulated into the mitochondria and PTT was triggered under NIR irradiation (1 W/cm2, 5 min). Owing to the high photothermal conversion efficiency, mitochondrial hyperthermia blocked the drug efflux pathway due to the inhibition of energy generation. 97% of the drug retained in cancer cells after 22 h incubation, which was much higher than the control groups. PDT combined with chemotherapy is an interesting strategy to design subcellular organelle destructive systems for highly efficient cancer therapy. In 2017, Tan et al. reported a mitochondriatargeted prodrug (PNPS) for NIR imaging guided synergistic chemo-PDT.145 NPS, a mitochondria-targeted NIR fluorescent probe, was coupled with the anticancer drug 5′-DFUR via a H2O2 responsive bisboronate group connection. After entering cancer cells, the bisboronate group was cleaved by H2O2 in the mitochondria and resulted the release of NPS and 5′-DFUR.

ACS Paragon Plus Environment

40

Page 41 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Subsequently, NIR triggered PDT and chemotherapy was activated. This prodrug offered an important signal for monitoring the real time drug release. Because the concentration of H2O2 in cancer cells (even up to 100 μM) was much higher than that in normal cells (~20 nM)87,236-238, the side effects was reduced. The IC50 values of PNPS towards HeLa and HepG2 cells under white light irradiation was 9.32 μM and 8.15 μM, respectively, indicating the promising application of the mitochondria-targeted prodrug for combined cancer theranostics.

Figure 19. Top: Scheme of the IR-DBI delivered by HSA via formation of dye-protein complex. (A) IR thermal images of 4T1-tumor-bearing mice injected with HSA@IR-DBI or PBS during 5 min NIR laser irradiation. (B) Tumor growth curves and (C) the survival rates of 4T1 tumor xenograft mice of different treatment groups. Adapted and modified with permission from ref 227. Copyright 2016 WILEY-VCH. In 2016, Shi and Qi et al. reported the first example of organic small molecule for mitochondriatargeted synchronous PTT-PDT.225 This heptamethine cyanine dyes derivative (PS 7) could target mitochondria via organic-anion transporting polypeptide mediation, and exhibited excellent mitochondrial retention capability owing to its lipophilic cationic feature. PS 7 exerted novel NIR

ACS Paragon Plus Environment

41

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 42 of 85

fluorescent characteristics and synergistic PDT/PTT effects. In vitro tests revealed that A549 cells were completely killed during synchronous PDT/PTT, while 29.3% cell viability remained after PTT treatment alone and 67.4% remained in PDT treatment alone. Interestingly, in vivo results demonstrated that the tumor growth in the PS treated A549 and 4T1 xenograft models were completely inhibited without tumor recurrence (1.5 W/cm2, 5 min). The survival rate was 100%, suggesting the significant potential of the reported PS 7 and the great promise of synchronous PDT/PTT. By using spherical magnetite (Fe3O4) as the core, PDA as the inner shell and mesoporous silica (mSiO2) as the outer shell, mitochondria-targeted PTT-PDT nanosystem was constructed after loading indocyanine green (ICG) and further surface modification of polyethylene glycol (PEG), transferrin (Tf) and TPP.181 The multifunctional nanocomposites (MMCNs) could be used for NIR fluorescence imaging and MRI, and the synergistic effects between mitochondria-targeted PDT and PTT significantly reduced the demand of high laser power during cancer photo-therapy (0.5 W/cm2, 5 min). Lin’s group reported the Hf and Ru dual-metal contained nanoscale MOFs for mitochondriatargeted RT-radiodynamic therapy (RDT).184 The integration of Ru(bpy)32+ contained frameworks endowed the Hf-based nMOFs novel cationic UiO topology and excellent mitochondria localization capability. Upon ultralow dosage (1 Gy) X-ray irradiation, in situ ROS generation (•OH from the Hf6 SBUs and 1O2 from Ru based PSs) in the mitochondria significantly depolarized the mitochondrial membrane potential of MC38 cells and further activated the apoptosis pathways. In vivo experiments revealed the admirable colorectal tumor suppressing capacity of the nMOFs, and no significant side effects could be observed.

ACS Paragon Plus Environment

42

Page 43 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

The integration of multiple cancer treatment modality into one simple system has also emerged as a potential strategy for further boosting mitochondria-targeted cancer treatment, which could offer fascinating superadditive therapeutic effects and reduce unwanted side effects. In 2017, Shi et al. reported an indocyanine green analog (IR-DBI) for mitochondria-targeted multimodal therapy (Figure 19).227 IR-DBI possesses a series of properties, including cancer and mitochondriaspecific accumulation, NIR emission and chemo/PDT/PTT multimodal therapeutic activities. The IC50 values of IR-DBI toward a series of human and mouse cancer cell lines remained low. IRDBI was biocompatible to normal human cells after incorporated into human serum albumin (HSA). The nanosized complex exhibited enhanced characteristics for imaging guided multimodal cancer therapy. Interestingly, in vivo tests revealed that the tumor growth was significantly depressed after 24 h tail vein injection plus 808 nm laser irradiation (1.5 W/cm2, 5 min), and the survival rate of treated tumor-bearing mouse was 100% after 90 days (Figure 19A-C). 4. Lysosome-targeted therapy Lysosomes are the main digest components in cells and they contain tens of hydrolases for degradation, repair and recirculation of diverse biomolecules, which make them critical in autophagy, secretion and membrane repair.83 Therefore, lysosomes are responsible for the cancer resistance during diverse therapies. Lysosome-targeted therapy has caused increasing interest in the past decade. The acidic lysosomal microenvironments (pH≈5.0) have been frequently employed for controlled drug release as well as activation of diverse therapeutic agents.201,203,239246

Lysosomal membrane permeabilization (LMP) and subsequent lysosomal enzymes release

have been noticed to be an important cell death pathway: the released enzymes are capable of digesting the whole cell and activating caspase- or other cell death pathways. Therefore, lysosome-

ACS Paragon Plus Environment

43

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 44 of 85

targeted/responsive toxins are another kind of therapeutic agents for enhanced cancer treatment.239242,247

There are many molecular/nano-formulations with lysosomal membrane disruption capability that are favorable for activation of LMP associated death pathways after entering cancer cells.105,248 Nanomaterials are rather attractive in lysosome targeting, because most ligand modified nanoformulations can finally reach lysosomes due to the lysosomal membrane fusion after entering cells via membrane receptor mediated internalization. And the nano-formulations can also be functionalized by lysosome targeting groups to improve the lysosomal accumulation and further enhancd the anticancer efficiency.240 Among the developed lysosomal membrane disruption strategies, alternating magnetic field (AMF) triggered LMP is highly admirable owing to the good controllability and non-invasive characteristics. The lysosomal membrane can be effectively broke by driving the nanomaterials via AMF after magnetic responsive nanomaterials reached the lysosomes, which could effectively trigger the leakage of lysosomal enzymes and subsequent cancer cell autolysis.248 Rinaldi’s group reported the activation of LMP death pathway by Epidermal Growth Factor (EGF) modified iron oxide magnetic nanoparticles (IO-MNPs). EGF receptor-mediated cell internalization resulted the MNPs located into lysosomes rapidly. After expose to AMF, the mechanic force caused the lysosomal contents release, and further triggered intracellular ROS production as well as protease cathepsin B activation, which significantly reduced the viability of cancer cells.249 This work suggested the potential application of remote control cancer cell fate via AMF. In addition to mechanistic force, ROS have also been employed for LMP activation. Other than the existence of multiple ROS scavenging enzymes in other places of cancer cells, the acidic and digestive lysosomal microenvironments restrict the activity of superoxide dismutase (SOD),

ACS Paragon Plus Environment

44

Page 45 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

peroxidase and so on, which make the local ROS concentration in lysosomes easier to be elevated.250-253 Therefore, lysosomal microenvironments are favorable for ROS induced apoptosis or LMP death pathway. Programmed ROS generation intra lysosomes of cancer cell should be another important point for enhanced cancer treatment.

Figure 20. (A) Schematic illustration of the mechanism of inducing and imaging LMP-dependent apoptosis in cancer cells by FITC-RR-Au-ZnO-RGD NPs. (B) TEM images of Au-ZnO hybrid NPs. (C) UV-Vis adsorption spectra of (a) Au NPs in hexane; (b) Au-ZnO NPs in hexane and (c) FITC-RR-Au-ZnO-RGD NPs in PBS. (D) The time-dependent confocal images of HepG2 cells after incubation with FITC-RR-Au-ZnO-RGD. The boxed areas are enlarged. Adapted and modified with permission from ref 106. Copyright 2014 Royal Society of Chemistry. Our group106 reported the Au-ZnO hybrid nanoparticles for lysosome-targeted LMP activation and real-time imaging (Figure 20A). FITC labeled protease cathepsin B substrate sequence ArgArg was grafted onto the Au NPs for selective tracing the activation of protease cathepsin B. RGD was modified onto ZnO to endowing the NPs with cancer targeting capacity (Figure 20B-C). The

ACS Paragon Plus Environment

45

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 46 of 85

resulting FITC-RR-Au-ZnO-RGD heterojunction structured NPs have high affinity to HepG2 cells and specifically localized to the lysosomes after entering cancer cells. ZnO NPs effectively catalyzed the ROS generation in the acidic lysosomal environments and successfully increased the permeability of lysosomal membrane, further resulting LMP-dependent apoptosis pathway activation in cancer cells (Figure 20D). This work demonstrated Au-ZnO as a promising platform for lysosome-targeted cancer cell treatment. In 2016, lysosomal activatable bis-styryl BODIPY based nanoparticles (BODIPY NPs) were reported by Huang and co-workers (Figure 21A).254 The nanoprecipitation method prepared BODIPY NPs possessed good water dispersibility, photostability and biocompatibility. The strong NIR adsorption of BODIPY NPs allowed them as the contrast agents for photoacoustic imaging, and the pH responsive feature made the NPs capable for lysosome-targeted PDT (200 mW/cm2, 10 min). Lan et al. reported the polythiophene nanoparticles (PT NPs) for lysosome-targeted twophoton excited (TPE) imaging and PDT.255 The energy dependent, clathrin- and caveolaindependent endocytic was found to be the main pathway for PT NPs endocytosis, diethylenediamine group endowed the PT NPs lysosome-targeting capacity. Under TPE, significant 1O2 generation in lysosomes was realized due to the ultra large TPA cross section (~3420 GM). PT NPs were also demonstrated for deep penetration (1300 μm) PDT and fluorescence imaging. Lysosome-targeted synergistic therapy was realized in the recent work of Mao and co-workers (Figure 21B).224 Reduced graphene oxide (rGO) was selected as the vehicle and PTT agent owing to the photothermal conversion efficiency as well as large surface area. The PEGylated Ru(II) based PS (Ru-PEG) was facile loaded onto rGO via π-π staking and hydrophobic interaction. The PDT effect of Ru-PEG was initially switched off due to the quenching effect of rGO, but would

ACS Paragon Plus Environment

46

Page 47 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

be switched on in the acidic lysosomal environments plus 808 nm laser irradiation, based on the photothermal enhanced PSs release in acidic microenvironments (Figure 21C). Thus, lysosometargeted combined PDT and PTT was realized under 808 nm (0.5 W/cm2, 5 min) and 450 nm (20 mW/cm2, 2 min) laser irradiation. With this system, significant synergistic effect was realized during in vitro and in vivo cancer treatment (Figure 21D-F).

Figure 21. (A) Schematic illustration of the construction of lysosome-targeted BODIPY NPs and their applications in lysosomal PAI and pH-activated PDT. Adapted and modified with permission from ref 254. Copyright 2016 American Chemical Society. (B) Schematic illustration of the construction of rGO-Ru-PEG. (C) Colocalization of rGO-Ru-PEG with lysosomes and mitochondria. (D) IR thermal images of A549 tumor-bearing mice under 808 nm laser irradiation. (E) Tumor growth curves of different groups of A549 tumor-bearing mice. (F) Photos of mice after various treatments taken at day 15. Adapted and modified with permission from ref 224. Copyright

ACS Paragon Plus Environment

47

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 48 of 85

2017 American Chemical Society. (G) TEM image of PTX@PAsp-g-(PEG-ICG) micelles, scale bar is 500 nm. (H) Statistical data of PAsp-g-(PEG-ICG) micelles’ diameter in (G). Cell viability of U-87 MG cells at (I) 48 h and (J) 72 h incubated with Taxol and PTX@PAsp-g-(PEG-ICG) micelles with/without laser irradiation. Adapted and modified with permission from ref 256. Copyright 2018 Royal Society of Chemistry. Although with promising application, lysosome-targeted therapeutics have not received as much interest as nucleus- or mitochondria-targeted modalities, mostly because many kinds of therapeutic agents including proteins and peptides are intolerant to the digestive acidic lysosomal microenvironments. Therefore, more investigations on lysosome-targeted therapy are desired. More importantly, it makes sense to devote more efforts to explore their working mechanisms and enhance the controllability as well as therapeutic efficiency of related therapeutic agents, because it should be beneficial for future personal medicines without resistance. 5. Endoplasmic reticulum/ Golgi apparatus-targeted therapy Endoplasmic reticulum (ER) and Golgi apparatus are closely related with protein/lipids synthesis and transfer, and they also play critical roles in tumor growth, drug resistance, cancer metastasis and immune escape.86 Specific therapeutic drugs accumulation in ER as well as Golgi apparatus may hamper the protein folding and result in stress signal, which will further cause cancer cell apoptosis. ER stress has been regarded as an important signal pathway to trigger cancer death, which makes ER another important target toward enhanced cancer treatment.257-260 In 2014, Chakravarty et al. reported an ER-targeted oxovanadium(IV) vitamin-B6 complex, [VO(HL)(acdppz)]Cl, for cancer PDT.261 By virtue of the over-expressed VB6 transporting membrane carrier (VTC) in cancer cells,

ACS Paragon Plus Environment

48

Page 49 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

[VO(HL)(acdppz)]Cl was specifically internalized by the MCF-7 cells and localized in the ER, while the internalization rate for normal counterparts of MCF-10A cells was slow. The ERtargeted [VO(HL)(acdppz)]Cl was biocompatible under dark but induced 1O2 production as well as ER stress under white light irradiation (2.4 mW/cm2, 1 h). Therefore, [VO(HL)(acdppz)]Cl was a potential candidate for ER stress activation and this work demonstrated ER as a potential target for highly efficient and specific cancer treatment. In 2016, Kwon and co-workers synthesized four Ir(III)-based complexes (TIr1-4). TIr1-4 with different energy levels exhibited TPA characteristics and ER location capabilities.262 TIr3 and TIr4 have high quantum yields for 1O2 and phosphorescence due to their well-matched energy levels. TIr3 could effectively induce cell apoptosis under hypoxia condition even under low density TPlaser irradiation (100 mW/cm2, 10 s). They further proved that the potential mechanism of ERtargeted PDT was the arbitrary protein oxidation and crosslinking via protein mass spectrometry analysis. This work offered a new idea for cancer therapy targeting the protein dysfunction associated cell death pathways. Very recently, Wang’s group developed an ER targeted comblike polymer, PAsp-g-(PEG-ICG), for enhanced chemotherapy (Figure 21G-H).256 Azido-modified PEG and ICG were grafted onto the alkynyl-functionalized poly(aspartic acid) (PAsp) through the 1,2,3-triazole connected click reaction. Owing to the rich carboxyl groups in PAsp, the PAsp-g-(PEG-ICG) effectively accumulated into the ER of SK-OV-3 cancer cells based on the coordination interaction with Ca2+ of ER lumen. PTX loaded nano micelles (PTX@PAsp-g-(PEG-ICG)) exhibited significant synergistic effect for enhanced in vivo cancer treatment owing to the ER targeted ROS production under laser irradiation (2 W/cm2, 5 min), which further proved the potential application of ERtargeting for enhanced cancer treatment (Figure 21I-J).

ACS Paragon Plus Environment

49

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 50 of 85

Yi’s group reported a pH responsive cyanine based photothermal agent (pH-PTT) for Golgi apparatus-targeted PTT.263 pH-PTT could be converted into conjugated structure with strong 808 nm adsorption under acidic microenvironments, and it was used for preparing nano anticancer agent after assembled with bovine serum albumin (BSA-pH-PTT). Interestingly, owing to the hypertrophic morphology of Golgi apparatus in cancer cells, the obtained BSA-pH-PTT exhibited cancer cell Golgi apparatus selectivity and could be activated by the local acidic microenvironment. Upon 808 nm laser irradiation (1.45 Wcm2, 10 min), BSA-pH-PTT significantly inhibited tumor growth due to the induction of Golgi apparatus hyperthermia. In contrast, lysosome-, ER- and Golgi apparatus-targeted nanodrugs have not receive as much interest as nucleus- and mitochondria-targeted therapeutics. Mostly because the targeting mechanisms are still unclear and development of related nanomaterials remains challengeable. (Table 3) Table 3. Typical nanomaterials for lysosome-, ER- and Golgi apparatus-targeted therapy. Organelles

Therapy modality

Nanomaterials

Targeting ligands

Targeting mechanisms

Cell type

Tumor model

Administration route (i.v./i.t.)

Treatment times

Treatment period

Refs

Lysosome

Catalytic therapy

Au-ZnO

RGD

Endocytic pathway mediated targeting

HepG2









106

PDT

Polythiophene NPs

Triethylen ediamine

Protonation induced targeting

HeLa









255

Synergistic therapy

rGO



Endocytic pathway mediated targeting

A549

A549

i.t.

1

15

224

Endoplasmic reticulum

Synergistic therapy

Comblike polymer NPs

Carboxyl group

Carboxyl-Ca(II) coordination induced targeting

U-87 MG

U-87 MG

i.v.

1

21

256

Golgi apparatus

PTT

BSA-Cyanine

BSA

Cancer cell Golgi apparatus hypertrophic resulted BSA accumulation

HepG-2









263

6. Summary and Perspectives

ACS Paragon Plus Environment

50

Page 51 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Organelles are critical components for cancer cell survival and proliferation. Owing to the specific destruction of tumor cell structures and activation of certain cell apoptosis pathways, therapeutic nanomaterials capable of targeting specific organelles have shown significant advantages over traditional drugs: they are rather favorable for reducing the drug dosage, boosting therapeutic index, avoiding multidrug resistance, recurrence, and detouring side effects. Organelletargeted chemotherapy, PDT, PTT, RT, gene therapy etc. have been widely investigated in recent years. These agents can be regarded as the new generation of therapeutics for cancer treatment and hold promise for clinical translation. Despite significant advances have been made, it still should be noteworthy that there are several barriers to be broke before preclinical studies: (a) current strategies for nanomaterials fabrication are still hard to fulfill the requirements of controlled and high-dosage preparation of well-defined nanostructures, while the pharmacokinetics behaviors and toxicity effects of nanomaterials are highly dependent on their morphology and size264,265; (b) targeting ligands on the surface of nanomaterials is another critical factor determining targeting efficiency, few efforts have been devoted into the optimization of the surface charge and ligand density of organelle-targeted nanomaterials; (c) there are multiple biological barriers to be overcome for i.v. administrated drugs reaching the organelles of interest, which depends on different and even opposite material properties, the available systems still cannot fulfill this demand; (d) nano-formulations that are capable of highly efficient cargo encapsulation and controlled release are desired for safe and efficient drug delivery, but the developed organelle-targeted nano-systems are still far from this objective; (e) the biodegradability as well as long-term toxicity of most employed nanomaterials have not been fully evaluated, biocompatible nanomaterials should be better candidates for clinical anticancer application;266-271 (f) multifunctional theranostic platforms are promising candidates for

ACS Paragon Plus Environment

51

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 52 of 85

precise cancer treatment, multiple imaging functions integrated organelle-targeted therapeutic systems have not been well investigated up to now; (g) nucleus and mitochondria-targeted therapy have attracted significant research interests, while much less attention have been devoted into the lysosome, ER, Golgi apparatus and other organelles, though they are also important targets for enhanced cancer treatment; (h) it is hard to compare the treatment potentials of different nanomaterials, due to the absence of standard methods for evaluating the therapeutic efficiency between different works. On the basis of the described challenges, we propose future works should focus on the following aspects: i) developing standard procedures for precise control of the size distribution and morphologies of nano-formulations, to ensure the repeatability of nano-drugs; ii) exploring more feasible cancer cell/organelle targeting ligands to enhance the subcellular location capability of nano-drugs; iii) designing smart platforms to overcome the multiple biological barriers in living systems, to further realize cancer cell subcellular structure-specific drug accumulation; iv) developing novel drug encapsulation as well as controlled release strategies for boosting the therapeutic index and avoiding the side effects; v) designing more therapeutic systems based on nontoxic/biodegradable nanomaterials, thoroughly evaluate their pharmacokinetics as well as clinical translation potential; vi) developing imaging guided theranostic systems for precise cancer treatment and even future personalized medicine; vii) paying more efforts to the therapeutic mechanisms of organelle-targeted nano-drugs, evaluating their immune effect, finding out the best recipes for specific cancer; viii) establishing universal evaluation protocols for comparing the therapeutic efficiency of different therapeutics, to further guide the design and synthesis of advanced organelle-targeted nano-drugs.

ACS Paragon Plus Environment

52

Page 53 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Overall, this review described the latest achievements of advanced organelle-targeted nanomaterials for boosting cancer therapy. Because it is still at the early stage, some molecular drugs were also included to highlight the significance and advantages of driving drugs into specific organelles. A list of recently published papers is provided for interested readers to further get to know the state of the art of this field.272-293 We believe this review will offer valuable information for the researchers to design better organelle-targeted nanomaterials, and more encouraging news can be brought to this field in the near future under the joint efforts from chemist to pharmaceutical scientists and clinical doctors. AUTHOR INFORMATION Corresponding Authors [email protected]; [email protected] ORCID Peng Gao: 0000-0002-3839-3167 Wei Pan: 0000-0002-2281-8749 Na Li: 0000-0002-0392-6672 Bo Tang: 0000-0002-8712-7025 Notes The authors declare no competing financial interests. ACKNOWLEDGMENT

ACS Paragon Plus Environment

53

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 54 of 85

This work was supported by National Natural Science Foundation of China (21535004, 91753111, 21874086, 21775094) and the Key Research and Development Program of Shandong Province (2018YFJH0502). REFERENCES 1.

Lucky, S. S.; Soo, K. C.; Zhang, Y., Nanoparticles in Photodynamic Therapy.

Chem. Rev. 2015, 115 (4), 1990-2042. 2.

Fan, W.; Yung, B.; Huang, P.; Chen, X., Nanotechnology for Multimodal

Synergistic Cancer Therapy. Chem. Rev. 2017, 117 (22), 13566-13638. 3.

Qian, J.; Tang, B. Z., AIE Luminogens for Bioimaging and Theranostics: From

Organelles to Animals. Chem 2017, 3 (1), 56-91. 4.

Yang, G.; Gong, H.; Liu, T.; Sun, X.; Cheng, L.; Liu, Z., Two-Dimensional Magnetic

WS2@Fe3O4 Nanocomposite with Mesoporous Silica Coating for Drug Delivery and Imaging-Guided Therapy of Cancer. Biomaterials 2015, 60, 62-71. 5.

Tian, J.; Ding, L.; Ju, H.; Yang, Y.; Li, X.; Shen, Z.; Zhu, Z.; Yu, J.-S.; Yang, C. J.,

A Multifunctional Nanomicelle for Real-Time Targeted Imaging and Precise Near-Infrared Cancer Therapy. Angew. Chem., Int. Ed. 2014, 53 (36), 9544-9549. 6.

Ma, Y.; Li, X.; Li, A.; Yang, P.; Zhang, C.; Tang, B., H2S-Activable MOF

Nanoparticle Photosensitizer for Effective Photodynamic Therapy against Cancer with Controllable Singlet-Oxygen Release. Angew. Chem., Int. Ed. 2017, 56 (44), 1375213756. 7.

Cheng, L.; Wang, C.; Feng, L.; Yang, K.; Liu, Z., Functional Nanomaterials for

Phototherapies of Cancer. Chem. Rev. 2014, 114 (21), 10869-10939. 8.

Xu, G.; Zeng, S.; Zhang, B.; Swihart, M. T.; Yong, K.-T.; Prasad, P. N., New

Generation Cadmium-Free Quantum Dots for Biophotonics and Nanomedicine. Chem.

Rev. 2016, 116 (19), 12234-12327. 9.

Zhao, Y.; Chen, F.; Li, Q.; Wang, L.; Fan, C., Isothermal Amplification of Nucleic

Acids. Chem. Rev. 2015, 115 (22), 12491-12545. 10.

Zhou, J.; Yu, G.; Huang, F., Supramolecular Chemotherapy Based on Host-Guest

Molecular Recognition: a Novel Strategy in the Battle Against Cancer with a Bright Future.

ACS Paragon Plus Environment

54

Page 55 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Chem. Soc. Rev. 2017, 46 (22), 7021-7053. 11.

Li, X.; Kim, J.; Yoon, J.; Chen, X., Cancer-Associated, Stimuli-Driven, Turn on

Theranostics for Multimodality Imaging and Therapy. Adv. Mater. 2017, 29 (23), 16068571606880. 12.

Sun, T.; Zhang, Y. S.; Pang, B.; Hyun, D. C.; Yang, M.; Xia, Y., Engineered

Nanoparticles for Drug Delivery in Cancer Therapy. Angew. Chem., Int. Ed. 2014, 53 (46), 12320-12364. 13.

Gao, W.; Cao, W.; Sun, Y.; Wei, X.; Xu, K.; Zhang, H.; Tang, B., AuNP Flares-

Capped Mesoporous Silica Nanoplatform for MTH1 Detection and Inhibition. Biomaterials 2015, 69, 212-221. 14.

Mei, J.; Leung, N. L. C.; Kwok, R. T. K.; Lam, J. W. Y.; Tang, B. Z., Aggregation-

Induced Emission: Together We Shine, United We Soar! Chem. Rev. 2015, 115 (21), 11718-11940. 15.

Zhou, W.; Gao, X.; Liu, D.; Chen, X., Gold Nanoparticles for in Vitro Diagnostics.

Chem. Rev. 2015, 115 (19), 10575-10636. 16.

Cheetham, A. G.; Chakroun, R. W.; Ma, W.; Cui, H., Self-Assembling Prodrugs.

Chem. Soc. Rev. 2017, 46 (21), 6638-6663. 17.

Li, Y.; Li, N.; Pan, W.; Yu, Z.; Yang, L.; Tang, B., Hollow Mesoporous Silica

Nanoparticles with Tunable Structures for Controlled Drug Delivery. ACS Appl. Mater.

Interfaces 2017, 9 (3), 2123-2129. 18.

Li, Y.; Chen, Y.; Pan, W.; Yu, Z.; Yang, L.; Wang, H.; Li, N.; Tang, B., Nanocarriers

with Multi-Locked DNA Valves Targeting Intracellular Tumor-Related mRNAs for Controlled Drug Release. Nanoscale 2017, 9 (44), 17318-17324. 19.

Li, Y.; Gao, X.; Yu, Z.; Liu, B.; Pan, W.; Li, N.; Tang, B., Reversing Multidrug

Resistance by Multiplexed Gene Silencing for Enhanced Breast Cancer Chemotherapy.

ACS Appl. Mater. Interfaces 2018, 10 (18), 15461-15466. 20.

Li, N.; Li, Y.; Gao, X.; Yu, Z.; Pan, W.; Tang, B., Multiplexed Gene Silencing in

Living Cells and in Vivo Using a DNAzymes-CoOOH Nanocomposite. Chem. Commun. 2017, 53 (36), 4962-4965. 21.

Daenen, L. G.; Roodhart, J. M.; van Amersfoort, M.; Dehnad, M.; Roessingh, W.;

Ulfman, L. H.; Derksen, P. W.; Voest, E. E., Chemotherapy Enhances Metastasis

ACS Paragon Plus Environment

55

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 56 of 85

Formation via VEGFR-1-Expressing Endothelial Cells. Cancer Res. 2011, 71 (22), 69766985. 22.

Gingis-Velitski, S.; Loven, D.; Benayoun, L.; Munster, M.; Bril, R.; Voloshin, T.;

Alishekevitz, D.; Bertolini, F.; Shaked, Y., Host Response to Short-Term, Single-Agent Chemotherapy Induces Matrix Metalloproteinase-9 Expression and Accelerates Metastasis in Mice. Cancer Res. 2011, 71 (22), 6986-6996. 23.

Volk-Draper, L.; Hall, K.; Griggs, C.; Rajput, S.; Kohio, P.; DeNardo, D.; Ran, S.,

Paclitaxel Therapy Promotes Breast Cancer Metastasis in a TLR4-Dependent Manner.

Cancer Res. 2014, 74 (19), 5421-5434. 24.

Luan, M.; Li, N.; Pan, W.; Yang, L.; Yu, Z.; Tang, B., Simultaneous Detection of

Multiple Targets Involved in the PI3K/AKT Pathway for Investigating Cellular Migration and Invasion with a Multicolor Fluorescent Nanoprobe. Chem. Commun. 2017, 53 (2), 356-359. 25.

Chang, Y. S.; Jalgaonkar, S. P.; Middleton, J. D.; Hai, T., Stress-Inducible Gene

Atf3 in the Noncancer Host Cells Contributes to Chemotherapy-Exacerbated Breast Cancer Metastasis. Proc. Natl. Acad. Sci. U. S. A. 2017, 114 (34), E7159-E7168. 26.

Luan, M.; Yu, L.; Li, Y.; Pan, W.; Gao, X.; Wan, X.; Li, N.; Tang, B., Visualizing

Breast Cancer Cell Proliferation and Invasion for Assessing Drug Efficacy with a Fluorescent Nanoprobe. Anal. Chem. 2017, 89 (19), 10601-10607. 27.

Kong, F.; Liang, Z.; Luan, D.; Liu, X.; Xu, K.; Tang, B., A Glutathione (GSH)-

Responsive Near-Infrared (NIR) Theranostic Prodrug for Cancer Therapy and Imaging.

Anal. Chem. 2016, 88 (12), 6450-6456. 28.

Louage, B.; Nuhn, L.; Risseeuw, M. D. P.; Vanparijs, N.; De Coen, R.; Karalic, I.;

Van Calenbergh, S.; De Geest, B. G., Well-Defined Polymer-Paclitaxel Prodrugs by a Grafting-from-Drug Approach. Angew. Chem., Int. Ed. 2016, 55 (39), 11791-11796. 29.

Bertin, P. A.; Smith, D.; Nguyen, S. T., High-Density Doxorubicin-Conjugated

Polymeric Nanoparticles via Ring-Opening Metathesis Polymerization. Chem. Commun. 2005, (30), 3793-3795. 30.

Du, J. Z.; Du, X. J.; Mao, C. Q.; Wang, J., Tailor-Made Dual pH-Sensitive Polymer-

Doxorubicin Nanoparticles for Efficient Anticancer Drug Delivery. J. Am. Chem. Soc. 2011, 133 (44), 17560-17563.

ACS Paragon Plus Environment

56

Page 57 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

31.

Culver, H. R.; Clegg, J. R.; Peppas, N. A., Analyte-Responsive Hydrogels:

Intelligent Materials for Biosensing and Drug Delivery. Acc. Chem. Res. 2017, 50 (2), 170178. 32.

Peng, F.; Su, Y.; Zhong, Y.; Fan, C.; Lee, S. T.; He, Y., Silicon Nanomaterials

Platform for Bioimaging, Biosensing, and Cancer Therapy. Acc. Chem. Res. 2014, 47 (2), 612-623. 33.

Cheng, Y.-J.; Luo, G.-F.; Zhu, J.-Y.; Xu, X.-D.; Zeng, X.; Cheng, D.-B.; Li, Y.-M.;

Wu, Y.; Zhang, X.-Z.; Zhuo, R.-X.; He, F., Enzyme-Induced and Tumor-Targeted Drug Delivery System Based on Multifunctional Mesoporous Silica Nanoparticles. ACS Appl.

Mater. Interfaces 2015, 7 (17), 9078-9087. 34.

Jiang, D.; Sun, Y.; Li, J.; Li, Q.; Lv, M.; Zhu, B.; Tian, T.; Cheng, D.; Xia, J.; Zhang,

L.; Wang, L.; Huang, Q.; Shi, J.; Fan, C., Multiple-Armed Tetrahedral DNA Nanostructures for Tumor-Targeting, Dual-Modality in Vivo Imaging. ACS Appl. Mater. Interfaces 2016, 8 (7), 4378-4384. 35.

Sukhorukova, I. V.; Zhitnyak, I. Y.; Kovalskii, A. M.; Matveev, A. T.; Lebedev, O. I.;

Li, X.; Gloushankova, N. A.; Golberg, D.; Shtansky, D. V., Boron Nitride Nanoparticles with a Petal-Like Surface as Anticancer Drug-Delivery Systems. ACS Appl. Mater.

Interfaces 2015, 7 (31), 17217-17225. 36.

Zhang, Y.; Ang, C. Y.; Li, M.; Tan, S. Y.; Qu, Q.; Luo, Z.; Zhao, Y., Polymer-Coated

Hollow Mesoporous Silica Nanoparticles for Triple-Responsive Drug Delivery. ACS Appl.

Mater. Interfaces 2015, 7 (32), 18179-18187. 37.

Merino, S.; Martin, C.; Kostarelos, K.; Prato, M.; Vazquez, E., Nanocomposite

Hydrogels: 3D Polymer-Nanoparticle Synergies for On-Demand Drug Delivery. ACS

Nano 2015, 9 (5), 4686-4697. 38.

Weng, Q.; Wang, B.; Wang, X.; Hanagata, N.; Li, X.; Liu, D.; Wang, X.; Jiang, X.;

Bando, Y.; Golberg, D., Highly Water-Soluble, Porous, and Biocompatible Boron Nitrides for Anticancer Drug Delivery. ACS Nano 2014, 8 (6), 6123-6130. 39.

Yin, W.; Yan, L.; Yu, J.; Tian, G.; Zhou, L.; Zheng, X.; Zhang, X.; Yong, Y.; Li, J.;

Gu, Z.; Zhao, Y., High-Throughput Synthesis of Single-Layer MoS2 Nanosheets as a Near-Infrared Photothermal-Triggered Drug Delivery for Effective Cancer Therapy. ACS

Nano 2014, 8 (7), 6922-6933.

ACS Paragon Plus Environment

57

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

40.

Page 58 of 85

Yang, K.; Feng, L.; Liu, Z., Stimuli Responsive Drug Delivery Systems Based on

Nano-Graphene for Cancer Therapy. Adv. Drug Delivery Rev. 2016, 105, 228-241. 41.

Song, G.; Chao, Y.; Chen, Y.; Liang, C.; Yi, X.; Yang, G.; Yang, K.; Cheng, L.;

Zhang, Q.; Liu, Z., All-in-One Theranostic Nanoplatform Based on Hollow TaOx for Chelator-Free Labeling Imaging, Drug Delivery, and Synergistically Enhanced Radiotherapy. Adv. Funct. Mater. 2016, 26 (45), 8243-8254. 42.

Zhou, W.; Wang, L.; Li, F.; Zhang, W.; Huang, W.; Huo, F.; Xu, H., Selenium-

Containing Polymer@Metal-Organic Frameworks Nanocomposites as an Efficient Multiresponsive Drug Delivery System. Adv. Funct. Mater. 2017, 27 (6), 16054651605472. 43.

Chen, W.; Ouyang, J.; Liu, H.; Chen, M.; Zeng, K.; Sheng, J.; Liu, Z.; Han, Y.;

Wang, L.; Li, J.; Deng, L.; Liu, Y.-N.; Guo, S., Black Phosphorus Nanosheet-Based Drug Delivery System for Synergistic Photodynamic/Photothermal/Chemotherapy of Cancer.

Adv. Mater. 2017, 29 (5), 1603864-1603870. 44.

Liu, T.; Wang, C.; Gu, X.; Gong, H.; Cheng, L.; Shi, X.; Feng, L.; Sun, B.; Liu, Z.,

Drug Delivery with PEGylated MoS2 Nano-Sheets for Combined Photothermal and Chemotherapy of Cancer. Adv. Mater. 2014, 26 (21), 3433-3440. 45.

Chen, Y.; Ye, D.; Wu, M.; Chen, H.; Zhang, L.; Shi, J.; Wang, L., Break-up of Two-

Dimensional MnO2 Nanosheets Promotes Ultrasensitive pH-Triggered Theranostics of Cancer. Adv. Mater. 2014, 26 (41), 7019-7026. 46.

Chimene, D.; Alge, D. L.; Gaharwar, A. K., Two-Dimensional Nanomaterials for

Biomedical Applications: Emerging Trends and Future Prospects. Adv. Mater. 2015, 27 (45), 7261-7284. 47.

Tao, W.; Zhu, X.; Yu, X.; Zeng, X.; Xiao, Q.; Zhang, X.; Ji, X.; Wang, X.; Shi, J.;

Zhang, H.; Mei, L., Black Phosphorus Nanosheets as a Robust Delivery Platform for Cancer Theranostics. Adv. Mater. 2017, 29 (1), 1603276-1603284. 48.

Wang, S.; Li, X.; Chen, Y.; Cai, X.; Yao, H.; Gao, W.; Zheng, Y.; An, X.; Shi, J.;

Chen, H., A Facile One-Pot Synthesis of a Two-Dimensional MoS2 /Bi2S3 Composite Theranostic Nanosystem for Multi-Modality Tumor Imaging and Therapy. Adv. Mater. 2015, 27 (17), 2775-2782. 49.

Jiang, Y.; Chen, J.; Deng, C.; Suuronen, E. J.; Zhong, Z., Click Hydrogels,

ACS Paragon Plus Environment

58

Page 59 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Microgels and Nanogels: Emerging Platforms for Drug Delivery and Tissue Engineering.

Biomaterials 2014, 35 (18), 4969-4985. 50.

Zhang, X.; Dong, Y.; Zeng, X.; Liang, X.; Li, X.; Tao, W.; Chen, H.; Jiang, Y.; Mei,

L.; Feng, S.-S., The Effect of Autophagy Inhibitors on Drug Delivery Using Biodegradable Polymer Nanoparticles in Cancer Treatment. Biomaterials 2014, 35 (6), 1932-1943. 51.

Bag, P. P.; Wang, D.; Chen, Z.; Cao, R., Outstanding Drug Loading Capacity by

Water Stable Microporous MOF: a Potential Drug Carrier. Chem. Commun. 2016, 52 (18), 3669-3672. 52.

Bai, L.; Phua, S. Z.; Lim, W. Q.; Jana, A.; Luo, Z.; Tham, H. P.; Zhao, L.; Gao, Q.;

Zhao, Y., Nanoscale Covalent Organic Frameworks as Smart Carriers for Drug Delivery.

Chem. Commun. 2016, 52 (22), 4128-4131. 53.

Liang, R.; Wei, M.; Evans, D. G.; Duan, X., Inorganic Nanomaterials for

Bioimaging, Targeted Drug Delivery and Therapeutics. Chem. Commun. 2014, 50 (91), 14071-14081. 54.

Orellana-Tavra, C.; Baxter, E. F.; Tian, T.; Bennett, T. D.; Slater, N. K. H.;

Cheetham, A. K.; Fairen-Jimenez, D., Amorphous Metal-Organic Frameworks for Drug Delivery. Chem. Commun. 2015, 51 (73), 13878-13881. 55.

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 (15), 4405-4418. 56.

Zhao, H.-X.; Zou, Q.; Sun, S.-K.; Yu, C.-S.; Zhang, X.-J.; Li, R.-J.; Fu, Y.-Y.,

Theranostic Metal-Organic Framework Core-Shell Composites for Magnetic Resonance Imaging and Drug Delivery. Chem. Sci. 2016, 7 (8), 5294-5301. 57.

Liu, J.; Zheng, X.; Yan, L.; Zhou, L.; Tian, G.; Yin, W.; Wang, L.; Liu, Y.; Hu, Z.;

Gu, Z.; Chen, C.; Zhao, Y., Bismuth Sulfide Nanorods as a Precision Nanomedicine for in Vivo Multimodal Imaging-Guided Photothermal Therapy of Tumor. ACS Nano 2015, 9 (1), 696-707. 58.

Wang, X.; Wang, M.; Lei, R.; Zhu, S. F.; Zhao, Y.; Chen, C., Chiral Surface of

Nanoparticles Determines the Orientation of Adsorbed Transferrin and Its Interaction with Receptors. ACS Nano 2017, 11 (5), 4606-4616. 59.

Yang, T.; Tang, Y.; Liu, L.; Lv, X.; Wang, Q.; Ke, H.; Deng, Y.; Yang, H.; Yang, X.;

ACS Paragon Plus Environment

59

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 60 of 85

Liu, G.; Zhao, Y.; Chen, H., Size-Dependent Ag2S Nanodots for Second Near-Infrared Fluorescence/Photoacoustics Imaging and Simultaneous Photothermal Therapy. ACS

Nano 2017, 11 (2), 1848-1857. 60.

Yuan, Y.; Chen, S.; Paunesku, T.; Gleber, S. C.; Liu, W. C.; Doty, C. B.; Mak, R.;

Deng, J.; Jin, Q.; Lai, B.; Brister, K.; Flachenecker, C.; Jacobsen, C.; Vogt, S.; Woloschak, G. E., 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 (12), 10502-10517. 61.

Zhao, R.; Han, X.; Li, Y.; Wang, H.; Ji, T.; Zhao, Y.; Nie, G., Photothermal Effect

Enhanced Cascade-Targeting Strategy for Improved Pancreatic Cancer Therapy by Gold Nanoshell@Mesoporous Silica Nanorod. ACS Nano 2017, 11 (8), 8103-8113. 62.

Zhou, Z.; Song, J.; Nie, L.; Chen, X., Reactive Oxygen Species Generating

Systems Meeting Challenges of Photodynamic Cancer Therapy. Chem. Soc. Rev. 2016,

45 (23), 6597-6626. 63.

Zhang, Z.; Wang, J.; Nie, X.; Wen, T.; Ji, Y.; Wu, X.; Zhao, Y.; Chen, C., Near

Infrared Laser-Induced Targeted Cancer Therapy Using Thermoresponsive Polymer Encapsulated Gold Nanorods. J. Am. Chem. Soc. 2014, 136 (20), 7317-7326. 64.

Liang, X. J.; Meng, H.; Wang, Y.; He, H.; Meng, J.; Lu, J.; Wang, P. C.; Zhao, Y.;

Gao, X.; Sun, B.; Chen, C.; Xing, G.; Shen, D.; Gottesman, M. M.; Wu, Y.; Yin, J. J.; Jia, L., Metallofullerene Nanoparticles Circumvent Tumor Resistance to Cisplatin by Reactivating Endocytosis. Proc. Natl. Acad. Sci. U. S. A. 2010, 107 (16), 7449-7454. 65.

Chen, Y.; Tan, C.; Zhang, H.; Wang, L., Two-Dimensional Graphene Analogues

for Biomedical Applications. Chem. Soc. Rev. 2015, 44 (9), 2681-2701. 66.

Yang, G.; Liu, J.; Wu, Y.; Feng, L.; Liu, Z., Near-Infrared-Light Responsive

Nanoscale Drug Delivery Systems for Cancer Treatment. Coord. Chem. Rev. 2016, 320, 100-117. 67.

Li, J.; Zheng, C.; Cansiz, S.; Wu, C.; Xu, J.; Cui, C.; Liu, Y.; Hou, W.; Wang, Y.;

Zhang, L.; Teng, I.-t.; Yang, H.-H.; Tan, W., Self-Assembly of DNA Nanohydrogels with Controllable Size and Stimuli-Responsive Property for Targeted Gene Regulation Therapy. J. Am. Chem. Soc. 2015, 137 (4), 1412-1415. 68.

Oh, S. S.; Lee, B. F.; Leibfarth, F. A.; Eisenstein, M.; Robb, M. J.; Lynd, N. A.;

ACS Paragon Plus Environment

60

Page 61 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Hawker, C. J.; Soh, H. T., Synthetic Aptamer-Polymer Hybrid Constructs for Programmed Drug Delivery into Specific Target Cells. J. Am. Chem. Soc. 2014, 136 (42), 15010-15015. 69.

Teplensky, M. H.; Fantham, M.; Li, P.; Wang, T. C.; Mehta, J. P.; Young, L. J.;

Moghadam, P. Z.; Hupp, J. T.; Farha, O. K.; Kaminski, C. F.; Fairen-Jimenez, D., Temperature

Treatment

of

Highly

Porous

Zirconium-Containing

Metal-Organic

Frameworks Extends Drug Delivery Release. J. Am. Chem. Soc. 2017, 139 (22), 75227532. 70.

Goenka, S.; Sant, V.; Sant, S., Graphene-Based Nanomaterials for Drug Delivery

and Tissue Engineering. J. Controlled Release 2014, 173, 75-88. 71.

Blanco, E.; Shen, H.; Ferrari, M., Principles of Nanoparticle Design for Overcoming

Biological Barriers to Drug Delivery. Nat. Biotechnol. 2015, 33 (9), 941-951. 72.

Felfoul, O.; Mohammadi, M.; Taherkhani, S.; de Lanauze, D.; Zhong Xu, Y.;

Loghin, D.; Essa, S.; Jancik, S.; Houle, D.; Lafleur, M.; Gaboury, L.; Tabrizian, M.; Kaou, N.; Atkin, M.; Vuong, T.; Batist, G.; Beauchemin, N.; Radzioch, D.; Martel, S., MagnetoAerotactic Bacteria Deliver Drug-Containing Nanoliposomes to Tumour Hypoxic Regions.

Nat. Nanotechnol. 2016, 11 (11), 941-947. 73.

Qiu, M.; Wang, D.; Liang, W.; Liu, L.; Zhang, Y.; Chen, X.; Sang, D. K.; Xing, C.;

Li, Z.; Dong, B.; Xing, F.; Fan, D.; Bao, S.; Zhang, H.; Cao, Y., Novel Concept of the Smart NIR-Light-Controlled Drug Release of Black Phosphorus Nanostructure for Cancer Therapy. Proc. Natl. Acad. Sci. U. S. A. 2018, 115 (3), 501-506. 74.

Mills, E. L.; Kelly, B.; Logan, A.; Costa, A. S. H.; Varma, M.; Bryant, C. E.;

Tourlomousis, P.; Dabritz, J. H. M.; Gottlieb, E.; Latorre, I.; Corr, S. C.; McManus, G.; Ryan, D.; Jacobs, H. T.; Szibor, M.; Xavier, R. J.; Braun, T.; Frezza, C.; Murphy, M. P.; O'Neill, L. A., Succinate Dehydrogenase Supports Metabolic Repurposing of Mitochondria to Drive Inflammatory Macrophages. Cell 2016, 167 (2), 457-470 e13. 75.

Shi, Y.; Massague, J., Mechanisms of TGF-beta Signaling from Cell Membrane to

the Nucleus. Cell 2003, 113 (6), 685-700. 76.

Vyas, S.; Zaganjor, E.; Haigis, M. C., Mitochondria and Cancer. Cell 2016, 166 (3),

555-566. 77.

Weinberg, S. E.; Sena, L. A.; Chandel, N. S., Mitochondria in the Regulation of

Innate and Adaptive Immunity. Immunity 2015, 42 (3), 406-417.

ACS Paragon Plus Environment

61

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

78.

Page 62 of 85

Zong, W. X.; Rabinowitz, J. D.; White, E., Mitochondria and Cancer. Mol. Cell

2016, 61 (5), 667-676. 79.

Weinberg, S. E.; Chandel, N. S., Targeting Mitochondria Metabolism for Cancer

Therapy. Nat. Chem. Biol. 2015, 11 (1), 9-15. 80.

Ashley, C. E.; Carnes, E. C.; Phillips, G. K.; Padilla, D.; Durfee, P. N.; Brown, P.

A.; Hanna, T. N.; Liu, J.; Phillips, B.; Carter, M. B.; Carroll, N. J.; Jiang, X.; Dunphy, D. R.; Willman, C. L.; Petsev, D. N.; Evans, D. G.; Parikh, A. N.; Chackerian, B.; Wharton, W.; Peabody, D. S.; Brinker, C. J., The Targeted Delivery of Multicomponent Cargos to Cancer Cells by Nanoporous Particle-Supported Lipid Bilayers. Nat. Mater. 2011, 10 (5), 389-397. 81.

Wang, M.; Kaufman, R. J., The Impact of the Endoplasmic Reticulum Protein-

Folding Environment on Cancer Development. Nat. Rev. Cancer 2014, 14 (9), 581-597. 82.

Phillips, M. J.; Voeltz, G. K., Structure and Function of ER Membrane Contact Sites

with Other Organelles. Nat. Rev. Mol. Cell Biol. 2016, 17 (2), 69-82. 83.

Yu, L.; McPhee, C. K.; Zheng, L.; Mardones, G. A.; Rong, Y.; Peng, J.; Mi, N.;

Zhao, Y.; Liu, Z.; Wan, F.; Hailey, D. W.; Oorschot, V.; Klumperman, J.; Baehrecke, E. H.; Lenardo, M. J., Termination of Autophagy and Reformation of Lysosomes Regulated by mTOR. Nature 2010, 465 (7300), 942-946. 84.

Zhu, H.; Fan, J.; Du, J.; Peng, X., Fluorescent Probes for Sensing and Imaging

within Specific Cellular Organelles. Acc. Chem. Res. 2016, 49 (10), 2115-2126. 85.

Xu, W.; Zeng, Z.; Jiang, J.-H.; Chang, Y.-T.; Yuan, L., Discerning the Chemistry in

Individual Organelles with Small-Molecule Fluorescent Probes. Angew. Chem., Int. Ed. 2016, 55 (44), 13658-13699. 86.

Moenner, M.; Pluquet, O.; Bouchecareilh, M.; Chevet, E., Integrated Endoplasmic

Reticulum Stress Responses in Cancer. Cancer Res. 2007, 67 (22), 10631-10634. 87.

Xu, Z.; Xu, L., Fluorescent Probes for the Selective Detection of Chemical Species

Inside Mitochondria. Chem. Commun. 2016, 52 (6), 1094-1119. 88.

Mills, E. L.; Kelly, B.; O'Neill, L. A. J., Mitochondria are the Powerhouses of

Immunity. Nat. Immunol. 2017, 18 (5), 488-498. 89.

Zhou, Z.; Liu, J.; Rees, T. W.; Wang, H.; Li, X.; Chao, H.; Stang, P. J.,

Heterometallic Ru–Pt Metallacycle for Two-Photon Photodynamic Therapy. Proc. Natl.

ACS Paragon Plus Environment

62

Page 63 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Acad. Sci. U. S. A. 2018, 115 (22), 5664-5669. 90.

Wang, Y.; Wei, G.; Zhang, X.; Huang, X.; Zhao, J.; Guo, X.; Zhou, S., Multistage

Targeting Strategy Using Magnetic Composite Nanoparticles for Synergism of Photothermal Therapy and Chemotherapy. Small 2017, 1702994-1703003. 91.

Feng, G.; Liu, J.; Zhang, C.-J.; Liu, B., Artemisinin and AIEgen Conjugate for

Mitochondria-Targeted and Image-Guided Chemo- and Photodynamic Cancer Cell Ablation. ACS Appl. Mater. Interfaces 2018, 10 (14), 11546-11553. 92.

Qiu, K.; Wang, J.; Song, C.; Wang, L.; Zhu, H.; Huang, H.; Huang, J.; Wang, H.;

Ji, L.; Chao, H., Crossfire for Two-Photon Photodynamic Therapy with Fluorinated Ruthenium (II) Photosensitizers. ACS Appl. Mater. Interfaces 2017, 9 (22), 18482-18492. 93.

Wu, C.; Wang, L.; Tian, Y.; Guan, X.; Liu, Q.; Li, S.; Qin, X.; Yang, H.; Liu, Y.,

“Triple-Punch” Anticancer Strategy Mediated by Near-Infrared Photosensitizer/CpG Oligonucleotides Dual-Dressed and Mitochondria-Targeted Nanographene. ACS Appl.

Mater. Interfaces 2018, 10 (8), 6942-6955. 94.

Tu, Z.; Qiao, H.; Yan, Y.; Guday, G.; Chen, W.; Adeli, M.; Haag, R., Directed

Graphene-Based

Nanoplatforms

for

Hyperthermia:

Overcoming

Multiple

Drug

Resistance. Angew. Chem., Int. Ed. 2018, 57 (35), 11198-11202. 95.

Wang, Z.; Chen, Y.; Zhang, H.; Li, Y.; Ma, Y.; Huang, J.; Liu, X.; Liu, F.; Wang, T.;

Zhang,

X.,

Mitochondria-Targeting

Polydopamine

Nanocomposites

as

Chemophotothermal Therapeutics for Cancer. Bioconjugate Chem 2018, 29 (7), 24152425. 96.

Liu, Y.; Li, H.; Xie, J.; Zhou, M.; Huang, H.; Lu, H.; Chai, Z.; Chen, J.; Hu, Y., Facile

Construction of Mitochondria-Targeting Nanoparticles for Enhanced Phototherapeutic Effects. Biomater. Sci. 2017, 5 (5), 1022-1031. 97.

Liu, J.; Liang, H.; Li, M.; Luo, Z.; Zhang, J.; Guo, X.; Cai, K., Tumor Acidity

Activating

Multifunctional

Nanoplatform

for

NIR-Mediated

Multiple

Enhanced

Photodynamic and Photothermal Tumor Therapy. Biomaterials 2018, 157, 107-124. 98.

Zhou, W.; Wang, X.; Hu, M.; Zhu, C.; Guo, Z., A Mitochondrion-Targeting Copper

Complex Exhibits Potent Cytotoxicity Against Cisplatin-Resistant Tumor Cells Through Multiple Mechanisms of Action. Chem. Sci. 2014, 5 (7), 2761-2770. 99.

He, H.; Wang, J.; Wang, H.; Zhou, N.; Yang, D.; Green, D. R.; Xu, B., Enzymatic

ACS Paragon Plus Environment

63

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 64 of 85

Cleavage of Branched Peptides for Targeting Mitochondria. J. Am. Chem. Soc. 2018, 140 (4), 1215-1218. 100.

Zheng, Y.; Ji, X.; Yu, B.; Ji, K.; Gallo, D.; Csizmadia, E.; Zhu, M.; Choudhury, M.

R.; De La Cruz, L. K. C.; Chittavong, V.; Pan, Z.; Yuan, Z.; Otterbein, L. E.; Wang, B., Enrichment-Triggered Prodrug Activation Demonstrated Through Mitochondria-Targeted Delivery of Doxorubicin and Carbon Monoxide. Nat. Chem. 2018, 10 (7), 787-794. 101.

Jeena, M. T.; Palanikumar, L.; Go, E. M.; Kim, I.; Kang, M. G.; Lee, S.; Park, S.;

Choi, H.; Kim, C.; Jin, S.-M.; Bae, S. C.; Rhee, H. W.; Lee, E.; Kwak, S. K.; Ryu, J.-H., Mitochondria Localization Induced Self-Assembly of Peptide Amphiphiles for Cellular Dysfunction. Nat. Commun. 2017, 8 (1), 26-35. 102.

Wang, H.; Gao, Z.; Liu, X.; Agarwal, P.; Zhao, S.; Conroy, D. W.; Ji, G.; Yu, J.;

Jaroniec, C. P.; Liu, Z.; Lu, X.; Li, X.; He, X., Targeted Production of Reactive Oxygen Species in Mitochondria to Overcome Cancer Drug Resistance. Nat. Commun. 2018, 9 (1), 562-577. 103.

Zhang, Y.; Wang, F.; Li, M.; Yu, Z.; Qi, R.; Ding, J.; Zhang, Z.; Chen, X., Self-

Stabilized Hyaluronate Nanogel for Intracellular Codelivery of Doxorubicin and Cisplatin to Osteosarcoma. Adv. Sci. 2018, 1700821-1700832. 104.

Cai, Y.; Shen, H.; Zhan, J.; Lin, M.; Dai, L.; Ren, C.; Shi, Y.; Liu, J.; Gao, J.; Yang,

Z., Supramolecular "Trojan Horse" for Nuclear Delivery of Dual Anticancer Drugs. J. Am.

Chem. Soc. 2017, 139 (8), 2876-2879. 105.

Chen, W. H.; Luo, G. F.; Zhang, X. Z., Recent Advances in Subcellular Targeted

Cancer Therapy Based on Functional Materials. Adv. Mater. 2018, 1802725-1802763. 106.

Gao, W.; Cao, W.; Zhang, H.; Li, P.; Xu, K.; Tang, B., Targeting Lysosomal

Membrane Permeabilization to Induce and Image Apoptosis in Cancer Cells by Multifunctional Au-ZnO Hybrid Nanoparticles. Chem. Commun. 2014, 50 (60), 81178120. 107.

Pan, L.; Liu, J.; Shi, J., Cancer Cell Nucleus-Targeting Nanocomposites for

Advanced Tumor Therapeutics. Chem. Soc. Rev. 2018, 47 (18), 6930-6946. 108.

Biswas, S.; Torchilin, V. P., Nanopreparations for Organelle-Specific Delivery in

Cancer. Adv. Drug Delivery Rev. 2014, 66, 26-41. 109.

Ma, X.; Gong, N.; Zhong, L.; Sun, J.; Liang, X.-J., Future of Nanotherapeutics:

ACS Paragon Plus Environment

64

Page 65 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Targeting the Cellular Sub-Organelles. Biomaterials 2016, 97, 10-21. 110.

Qiu, K.; Chen, Y.; Rees, T. W.; Ji, L.; Chao, H., Organelle-Targeting Metal

Complexes: from Molecular Design to Bio-Applications. Coord. Chem. Rev. 2019, 378, 66-86. 111.

Milane, L.; Trivedi, M.; Singh, A.; Talekar, M.; Amiji, M., Mitochondrial Biology,

Targets, and Drug Delivery. J. Controlled Release 2015, 207, 40-58. 112.

Mo, R.; Sun, Q.; Xue, J.; Li, N.; Li, W.; Zhang, C.; Ping, Q., Multistage pH-

Responsive Liposomes for Mitochondrial-Targeted Anticancer Drug Delivery. Adv. Mater. 2012, 24 (27), 3659-3665. 113.

Noh, I.; Lee, D.; Kim, H.; Jeong, C.-U.; Lee, Y.; Ahn, J.-O.; Hyun, H.; Park, J.-H.;

Kim, Y.-C., Enhanced Photodynamic Cancer Treatment by Mitochondria-Targeting and Brominated Near-Infrared Fluorophores. Adv. Sci. 2018, 5 (3), 1700481-1700491. 114.

Zhang, L.; Wang, D.; Yang, K.; Sheng, D.; Tan, B.; Wang, Z.; Ran, H.; Yi, H.;

Zhong, Y.; Lin, H.; Chen, Y., Mitochondria-Targeted Artificial “Nano-RBCs” for Amplified Synergistic Cancer Phototherapy by a Single NIR Irradiation. Adv. Sci. 2018, 18000491800063. 115.

Ling, D.; Bae, B.-C.; Park, W.; Na, K., Photodynamic Efficacy of Photosensitizers

Under an Attenuated Light Dose via Lipid Nano-Carrier-Mediated Nuclear Targeting.

Biomaterials 2012, 33 (21), 5478-5486. 116.

Jin, E.; Zhang, B.; Sun, X.; Zhou, Z.; Ma, X.; Sun, Q.; Tang, J.; Shen, Y.; Van Kirk,

E.; Murdoch, W. J.; Radosz, M., Acid-Active Cell-Penetrating Peptides for In Vivo TumorTargeted Drug Delivery. J. Am. Chem. Soc. 2013, 135 (2), 933-940. 117.

Pan, L.; He, Q.; Liu, J.; Chen, Y.; Ma, M.; Zhang, L.; Shi, J., Nuclear-Targeted Drug

Delivery of TAT Peptide-Conjugated Monodisperse Mesoporous Silica Nanoparticles. J.

Am. Chem. Soc. 2012, 134 (13), 5722-5725. 118.

Yuan, H.; Fales, A. M.; Vo-Dinh, T., TAT Peptide-Functionalized Gold Nanostars:

Enhanced Intracellular Delivery and Efficient NIR Photothermal Therapy Using Ultralow Irradiance. J. Am. Chem. Soc. 2012, 134 (28), 11358-11361. 119.

Biswas, S.; Dodwadkar, N. S.; Deshpande, P. P.; Torchilin, V. P., Liposomes

Loaded with Paclitaxel and Modified with Novel Triphenylphosphonium-PEG-PE Conjugate Possess Low Toxicity, Target Mitochondria and Demonstrate Enhanced

ACS Paragon Plus Environment

65

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 66 of 85

Antitumor Effects in Vitro and in Vivo. J. Controlled Release 2012, 159 (3), 393-402. 120.

Yang, G.; Xu, L.; Xu, J.; Zhang, R.; Song, G.; Chao, Y.; Feng, L.; Han, F.; Dong,

Z.; Li, B.; Liu, Z., Smart Nanoreactors for pH-Responsive Tumor Homing, MitochondriaTargeting, and Enhanced Photodynamic-Immunotherapy of Cancer. Nano Lett. 2018, 18 (4), 2475-2484. 121.

Zhou, F.; Wu, S.; Wu, B.; Chen, W. R.; Xing, D., Mitochondria-Targeting Single-

Walled Carbon Nanotubes for Cancer Photothermal Therapy. Small 2011, 7 (19), 27272735. 122.

Yu, Z.; Pan, W.; Li, N.; Tang, B., A Nuclear Targeted Dual-Photosensitizer for

Drug-Resistant Cancer Therapy with NIR Activated Multiple ROS. Chem. Sci. 2016, 7 (7), 4237-4244. 123.

Li, N.; Yu, Z.; Pan, W.; Han, Y.; Zhang, T.; Tang, B., A Near-Infrared Light-

Triggered Nanocarrier with Reversible DNA Valves for Intracellular Controlled Release.

Adv. Funct. Mater. 2013, 23 (18), 2255-2262. 124.

Jiang, Q.; Song, C.; Nangreave, J.; Liu, X.; Lin, L.; Qiu, D.; Wang, Z.-G.; Zou, G.;

Liang, X.; Yan, H.; Ding, B., DNA Origami as a Carrier for Circumvention of Drug Resistance. J. Am. Chem. Soc. 2012, 134 (32), 13396-13403. 125.

Chao, J.; Liu, H.; Su, S.; Wang, L.; Huang, W.; Fan, C., Structural DNA

Nanotechnology for Intelligent Drug Delivery. Small 2014, 10 (22), 4626-4635. 126.

Xing, H.; Hwang, K.; Lu, Y., Recent Developments of Liposomes as Nanocarriers

for Theranostic Applications. Theranostics 2016, 6 (9), 1336-1352. 127.

Li, Z.-Y.; Liu, Y.; Hu, J.-J.; Xu, Q.; Liu, L.-H.; Jia, H.-Z.; Chen, W.-H.; Lei, Q.; Rong,

L.; Zhang, X.-Z., Stepwise-Acid-Active Multifunctional Mesoporous Silica Nanoparticles for Tumor-Specific Nucleus-Targeted Drug Delivery. ACS Appl. Mater. Interfaces 2014, 6 (16), 14568-14575. 128.

Pan, L.; Liu, J.; Shi, J., Nuclear-Targeting Gold Nanorods for Extremely Low NIR

Activated Photothermal Therapy. ACS Appl. Mater. Interfaces 2017, 9 (19), 15952-15961. 129.

Huo, S.; Jin, S.; Ma, X.; Xue, X.; Yang, K.; Kumar, A.; Wang, P. C.; Zhang, J.; Hu,

Z.; Liang, X.-J., Ultrasmall Gold Nanoparticles as Carriers for Nucleus-Based Gene Therapy Due to Size-Dependent Nuclear Entry. ACS Nano 2014, 8 (6), 5852-5862. 130.

Pan, L.; Liu, J.; He, Q.; Shi, J., MSN-Mediated Sequential Vascular-to-Cell

ACS Paragon Plus Environment

66

Page 67 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Nuclear-Targeted Drug Delivery for Efficient Tumor Regression. Adv. Mater. 2014, 26 (39), 6742-6748. 131.

Sakiyama, Y.; Mazur, A.; Kapinos, L. E.; Lim, R. Y., Spatiotemporal Dynamics of

the Nuclear Pore Complex Transport Barrier Resolved by High-Speed Atomic Force Microscopy. Nat. Nanotechnol. 2016, 11 (8), 719-723. 132.

von Appen, A.; Kosinski, J.; Sparks, L.; Ori, A.; DiGuilio, A. L.; Vollmer, B.;

Mackmull, M. T.; Banterle, N.; Parca, L.; Kastritis, P.; Buczak, K.; Mosalaganti, S.; Hagen, W.; Andres-Pons, A.; Lemke, E. A.; Bork, P.; Antonin, W.; Glavy, J. S.; Bui, K. H.; Beck, M., In Situ Structural Analysis of the Human Nuclear Pore Complex. Nature 2015, 526 (7571), 140-143. 133.

Li, J.; Liu, F.; Shao, Q.; Min, Y.; Costa, M.; Yeow, E. K. L.; Xing, B., Enzyme-

Responsive Cell-Penetrating Peptide Conjugated Mesoporous Silica Quantum Dot Nanocarriers for Controlled Release of Nucleus-Targeted Drug Molecules and Real-Time Intracellular Fluorescence Imaging of Tumor Cells. Adv. Healthcare Mater. 2014, 3 (8), 1230-1239. 134.

Pan, L.; Liu, J.; He, Q.; Wang, L.; Shi, J., Overcoming Multidrug Resistance of

Cancer Cells by Direct Intranuclear Drug Delivery Using TAT-Conjugated Mesoporous Silica Nanoparticles. Biomaterials 2013, 34 (11), 2719-2730. 135.

Han, S.-S.; Li, Z.-Y.; Zhu, J.-Y.; Han, K.; Zeng, Z.-Y.; Hong, W.; Li, W.-X.; Jia, H.-

Z.; Liu, Y.; Zhuo, R.-X.; Zhang, X.-Z., Dual-pH Sensitive Charge-Reversal Polypeptide Micelles for Tumor-Triggered Targeting Uptake and Nuclear Drug Delivery. Small 2015,

11 (21), 2543-2554. 136.

Xiong, L.; Du, X.; Kleitz, F.; Qiao, S. Z., Cancer-Cell-Specific Nuclear-Targeted

Drug Delivery by Dual-Ligand-Modified Mesoporous Silica Nanoparticles. Small 2015, 11 (44), 5919-5926. 137.

Li, Z.; Dong, K.; Huang, S.; Ju, E.; Liu, Z.; Yin, M.; Ren, J.; Qu, X., A Smart

Nanoassembly for Multistage Targeted Drug Delivery and Magnetic Resonance Imaging.

Adv. Funct. Mater. 2014, 24 (23), 3612-3620. 138.

Zhu, Y. X.; Jia, H. R.; Pan, G. Y.; Ulrich, N. W.; Chen, Z.; Wu, F. G., Development

of a Light-Controlled Nanoplatform for Direct Nuclear Delivery of Molecular and Nanoscale Materials. J. Am. Chem. Soc. 2018, 140 (11), 4062-4070.

ACS Paragon Plus Environment

67

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

139.

Page 68 of 85

Peng, H.; Tang, J.; Zheng, R.; Guo, G.; Dong, A.; Wang, Y.; Yang, W., Nuclear-

Targeted Multifunctional Magnetic Nanoparticles for Photothermal Therapy. Adv.

Healthcare Mater. 2017, 6 (7), 1601289-1601300. 140.

Li, N.; Sun, Q.; Yu, Z.; Gao, X.; Pan, W.; Wan, X.; Tang, B., Nuclear-Targeted

Photothermal Therapy Prevents Cancer Recurrence with Near-Infrared Triggered Copper Sulfide Nanoparticles. ACS Nano 2018, 12 (6), 5197–5206. 141.

Pan,

L.;

Liu,

J.;

Shi,

J.,

Intranuclear

Photosensitizer

Delivery

and

Photosensitization for Enhanced Photodynamic Therapy with Ultralow Irradiance. Adv.

Funct. Mater. 2014, 24 (46), 7318-7327. 142.

Li, L.; Sun, W.; Zhong, J.; Yang, Q.; Zhu, X.; Zhou, Z.; Zhang, Z.; Huang, Y.,

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 (26), 4101-4113. 143.

Wang, P.; Zhang, L.; Zheng, W.; Cong, L.; Guo, Z.; Xie, Y.; Wang, L.; Tang, R.;

Feng, Q.; Hamada, Y.; Gonda, K.; Hu, Z.; Wu, X.; Jiang, X., Thermo-Triggered Release of CRISPR-Cas9 System by Lipid-Encapsulated Gold Nanoparticles for Tumor Therapy.

Angew. Chem., Int. Ed. 2018, 57 (6), 1491-1496. 144.

Fan, W.; Shen, B.; Bu, W.; Zheng, X.; He, Q.; Cui, Z.; Zhao, K.; Zhang, S.; Shi, J.,

Design of an Intelligent Sub-50 nm Nuclear-Targeting Nanotheranostic System for Imaging Guided Intranuclear Radiosensitization. Chem. Sci. 2015, 6 (3), 1747-1753. 145.

Liu, H.-W.; Hu, X.-X.; Li, K.; Liu, Y.; Rong, Q.; Zhu, L.; Yuan, L.; Qu, F.-L.; Zhang,

X.-B.; Tan, W., A Mitochondrial-Targeted Prodrug for NIR Imaging Guided and Synergetic NIR Photodynamic-Chemo Cancer Therapy. Chem. Sci. 2017, 8 (11), 7689-7695. 146.

Gao, P.; Liu, M.; Tian, J.; Deng, F.; Wang, K.; Xu, D.; Liu, L.; Zhang, X.; Wei, Y.,

Improving the drug delivery characteristics of graphene oxide based polymer nanocomposites through the "one-pot" synthetic approach of single-electron-transfer living radical polymerization. Appl. Surf. Sci. 2016, 378, 22-29. 147.

Vankayala, R.; Kuo, C.-L.; Nuthalapati, K.; Chiang, C.-S.; Hwang, K. C., Nucleus-

Targeting Gold Nanoclusters for Simultaneous In Vivo Fluorescence Imaging, Gene Delivery, and NIR-Light Activated Photodynamic Therapy. Adv. Funct. Mater. 2015, 25 (37), 5934-5945.

ACS Paragon Plus Environment

68

Page 69 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

148.

Roy, E.; Patra, S.; Madhuri, R.; Sharma, P. K., Carbon Dot/TAT Peptide Co-

Conjugated Bubble Nanoliposome for Multicolor Cell Imaging, Nuclear-Targeted Delivery, and Chemo/Photothermal Synergistic Therapy. Chem. Eng. J. 2017, 312, 144-157. 149.

Chen, H.; Wang, Z.; Zong, S.; Chen, P.; Zhu, D.; Wu, L.; Cui, Y., A Graphene

Quantum Dot-based FRET System for Nuclear-Targeted and Real-Time Monitoring of Drug Delivery. Nanoscale 2015, 7 (37), 15477-15486. 150.

Wang, J.; Shen, S.; Li, D.; Zhan, C.; Yuan, Y.; Yang, X., Photoswitchable Ultrafast

Transactivator of Transcription (TAT) Targeting Effect for Nanocarrier-Based On-Demand Drug Delivery. Adv. Funct. Mater. 2017, 28 (3), 1704806-1704817. 151.

Chen, D.; Li, B.; Cai, S.; Wang, P.; Peng, S.; Sheng, Y.; He, Y.; Gu, Y.; Chen, H.,

Dual Targeting Luminescent Gold Nanoclusters for Tumor Imaging and Deep Tissue Therapy. Biomaterials 2016, 100, 1-16. 152.

Fan, Y.; Li, C.; Li, F.; Chen, D., pH-Activated Size Reduction of Large Compound

Nanoparticles for in Vivo Nucleus-Targeted Drug Delivery. Biomaterials 2016, 85, 30-39. 153.

Liu, J.-N.; Bu, W.; Pan, L.-M.; Zhang, S.; Chen, F.; Zhou, L.; Zhao, K.-L.; Peng,

W.; Shi, J., Simultaneous Nuclear Imaging and Intranuclear Drug Delivery by NuclearTargeted Multifunctional Upconversion Nanoprobes. Biomaterials 2012, 33 (29), 72827290. 154.

Chen, Z.; Zhang, L.; He, Y.; Li, Y., Sandwich-Type Au-PEI/DNA/PEI-Dexa

Nanocomplex for Nucleus-Targeted Gene Delivery in Vitro and in Vivo. ACS Appl. Mater.

Interfaces 2014, 6 (16), 14196-14206. 155.

Li, N.; Yang, H.; Yu, Z.; Li, Y.; Pan, W.; Wang, H.; Tang, B., Nuclear-Targeted

siRNA Delivery for Long-Term Gene Silencing. Chem. Sci. 2017, 8 (4), 2816-2822. 156.

Shao, L.; Zhang, R.; Lu, J.; Zhao, C.; Deng, X.; Wu, Y., Mesoporous Silica Coated

Polydopamine Functionalized Reduced Graphene Oxide for Synergistic Targeted Chemo-Photothermal Therapy. ACS Appl. Mater. Interfaces 2017, 9 (2), 1226-1236. 157.

Xiang, H.; Xue, F.; Yi, T.; Tham, H. P.; Liu, J.-G.; Zhao, Y., Cu2–xS Nanocrystals

Cross-Linked

with

Chlorin

e6-Functionalized

Polyethylenimine

for

Synergistic

Photodynamic and Photothermal Therapy of Cancer. ACS Appl. Mater. Interfaces 2018,

10 (19), 16344-16351. 158.

Xu, Y.; Feng, T.; Yang, T.; Wei, H.; Yang, H.; Li, G.; Zhao, M.; Liu, S.; Huang, W.;

ACS Paragon Plus Environment

69

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 70 of 85

Zhao, Q., Utilizing Intramolecular Photoinduced Electron Transfer to Enhance Photothermal Tumor Treatment of Aza-BODIPY-Based Near-Infrared Nanoparticles.

ACS Appl. Mater. Interfaces 2018, 10 (19), 16299-16307. 159.

Chen, Z.; Zhao, P.; Luo, Z.; Zheng, M.; Tian, H.; Gong, P.; Gao, G.; Pan, H.; Liu,

L.; Ma, A., Cancer Cell Membrane–Biomimetic Nanoparticles for Homologous-Targeting Dual-Modal Imaging and Photothermal Therapy. ACS Nano 2016, 10 (11), 10049-10057. 160.

Lin, L.; Cong, Z.; Cao, J.; Ke, K.; Peng, Q.; Gao, J.; Yang, H.; Liu, G.; Chen, X.,

Multifunctional Fe3O4@Polydopamine Core–Shell Nanocomposites for Intracellular mRNA Detection and Imaging-Guided Photothermal Therapy. ACS Nano 2014, 8 (4), 3876-3883. 161.

Liu, Y.; Ai, K.; Liu, J.; Deng, M.; He, Y.; Lu, L., Dopamine ‐ Melanin Colloidal

Nanospheres: an Efficient Near ‐ Infrared Photothermal Therapeutic Agent for in Vivo Cancer Therapy. Adv. Mater. 2013, 25 (9), 1353-1359. 162.

Zhou, J.; Li, M.; Hou, Y.; Luo, Z.; Chen, Q.; Cao, H.; Huo, R.; Xue, C.; Sutrisno, L.;

Hao, L.; Cao, Y.; Ran, H.; Lu, L.; Li, K.; Cai, K., Engineering of a Nanosized Biocatalyst for Combined Tumor Starvation and Low-Temperature Photothermal Therapy. ACS Nano 2018, 12 (3), 2858-2872. 163.

Yang, Y.; Zhu, W.; Dong, Z.; Chao, Y.; Xu, L.; Chen, M.; Liu, Z., 1D Coordination

Polymer Nanofibers for Low-Temperature Photothermal Therapy. Adv. Mater. 2017, 29 (40), 1703588-1703599. 164.

Han, K.; Zhang, W.-Y.; Zhang, J.; Lei, Q.; Wang, S.-B.; Liu, J.-W.; Zhang, X.-Z.;

Han, H.-Y., Acidity-Triggered Tumor-Targeted Chimeric Peptide for Enhanced IntraNuclear Photodynamic Therapy. Adv. Funct. Mater. 2016, 26 (24), 4351-4361. 165.

Zhang, Y.; Wang, F.; Liu, C.; Wang, Z.; Kang, L.; Huang, Y.; Dong, K.; Ren, J.; Qu,

X., Nanozyme Decorated Metal-Organic Frameworks for Enhanced Photodynamic Therapy. ACS Nano 2018, 12 (1), 651-661. 166.

Fan, H.; Zhao, Z.; Yan, G.; Zhang, X.; Yang, C.; Meng, H.; Chen, Z.; Liu, H.; Tan,

W., A Smart DNAzyme-MnO(2) Nanosystem for Efficient Gene Silencing. Angew. Chem.,

Int. Ed. 2015, 54 (16), 4801-4805. 167.

Fan, W.; Huang, P.; Chen, X., Overcoming the Achilles' Heel of Photodynamic

Therapy. Chem. Soc. Rev. 2016, 45 (23), 6488-6519.

ACS Paragon Plus Environment

70

Page 71 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

168.

Yu, Z.; Zhou, P.; Pan, W.; Li, N.; Tang, B., A biomimetic nanoreactor for synergistic

chemiexcited photodynamic therapy and starvation therapy against tumor metastasis.

Nat. Commun. 2018, 9 (1), 5044-5052. 169.

Nair, J. K.; Willoughby, J. L.; Chan, A.; Charisse, K.; Alam, M. R.; Wang, Q.;

Hoekstra, M.; Kandasamy, P.; Kel'in, A. V.; Milstein, S.; Taneja, N.; O'Shea, J.; Shaikh, S.; Zhang, L.; van der Sluis, R. J.; Jung, M. E.; Akinc, A.; Hutabarat, R.; Kuchimanchi, S.; Fitzgerald, K.; Zimmermann, T.; van Berkel, T. J.; Maier, M. A.; Rajeev, K. G.; Manoharan, M., Multivalent N-Acetylgalactosamine-Conjugated siRNA localizes in Hepatocytes and Elicits Robust RNAi-Mediated Gene Silencing. J. Am. Chem. Soc. 2014, 136 (49), 1695816961. 170.

Wu, M.; Meng, Q.; Chen, Y.; Du, Y.; Zhang, L.; Li, Y.; Zhang, L.; Shi, J., Large-

Pore Ultrasmall Mesoporous Organosilica Nanoparticles: Micelle/Precursor Cotemplating Assembly and Nuclear-Targeted Gene Delivery. Adv. Mater. 2015, 27 (2), 215222. 171.

Fan, W.; Shen, B.; Bu, W.; Zheng, X.; He, Q.; Cui, Z.; Ni, D.; Zhao, K.; Zhang, S.;

Shi, J., Intranuclear Biophotonics by Smart Design of Nuclear-Targeting Photo-/RadioSensitizers Co-Loaded Upconversion Nanoparticles. Biomaterials 2015, 69, 89-98. 172.

Li, P.; Xie, T.; Duan, X.; Yu, F.; Wang, X.; Tang, B., A New Highly Selective and

Sensitive Assay for Fluorescence Imaging of •OH in Living Cells: Effectively Avoiding the Interference of Peroxynitrite. Chem. - Eur. J. 2010, 16 (6), 1834-1840. 173.

Roopa; Kumar, N.; Bhalla, V.; Kumar, M., Development and Sensing Applications

of Fluorescent Motifs within the Mitochondrial Environment. Chem. Commun. 2015, 51 (86), 15614-15628. 174.

Xiao, H.; Li, P.; Zhang, S.; Zhang, W.; Zhang, W.; Tang, B., Simultaneous

Fluorescence Visualization of Mitochondrial Hydrogen Peroxide and Zinc Ions in Live Cells and in Vivo. Chem. Commun. 2016, 52 (86), 12741-12744. 175.

Chan, M. S.; Liu, L. S.; Leung, H. M.; Lo, P. K., Cancer-Cell-Specific Mitochondria-

Targeted Drug Delivery by Dual-Ligand-Functionalized Nanodiamonds Circumvent Drug Resistance. ACS Appl. Mater. Interfaces 2017, 9 (13), 11780-11789. 176.

Zhou, J.; Zhao, W.-Y.; Ma, X.; Ju, R.-J.; Li, X.-Y.; Li, N.; Sun, M.-G.; Shi, J.-F.;

Zhang, C.-X.; Lu, W.-L., The Anticancer Efficacy of Paclitaxel Liposomes Modified with

ACS Paragon Plus Environment

71

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 72 of 85

Mitochondrial Targeting Conjugate in Resistant Lung Cancer. Biomaterials 2013, 34 (14), 3626-3638. 177.

Zhou, W.; Yu, H.; Zhang, L.-J.; Wu, B.; Wang, C.-X.; Wang, Q.; Deng, K.; Zhuo,

R.-X.; Huang, S.-W., Redox-Triggered Activation of Nanocarriers for MitochondriaTargeting Cancer Chemotherapy. Nanoscale 2017, 9 (43), 17044-17053. 178.

Yu, Z.; Sun, Q.; Pan, W.; Li, N.; Tang, B., A Near-Infrared Triggered

Nanophotosensitizer Inducing Domino Effect on Mitochondrial Reactive Oxygen Species Burst for Cancer Therapy. ACS Nano 2015, 9 (11), 11064-11074. 179.

Han, K.; Lei, Q.; Wang, S.-B.; Hu, J.-J.; Qiu, W.-X.; Zhu, J.-Y.; Yin, W.-N.; Luo, X.;

Zhang, X.-Z., Dual-Stage-Light-Guided Tumor Inhibition by Mitochondria-Targeted Photodynamic Therapy. Adv. Funct. Mater. 2015, 25 (20), 2961-2971. 180.

Feng, G.; Qin, W.; Hu, Q.; Tang, B. Z.; Liu, B., Cellular and Mitochondrial Dual-

Targeted Organic Dots with Aggregation-Induced Emission Characteristics for ImageGuided Photodynamic Therapy. Adv. Healthcare Mater. 2015, 4 (17), 2667-2676. 181.

Guo, R.; Peng, H.; Tian, Y.; Shen, S.; Yang, W., Mitochondria-Targeting Magnetic

Composite Nanoparticles for Enhanced Phototherapy of Cancer. Small 2016, 12 (33), 4541-4552. 182.

Wang, H.; Chang, J.; Shi, M.; Pan, W.; Li, N.; Tang, B., A Dual-Targeted Organic

Photothermal Agent for Enhanced Photothermal Therapy. Angew. Chem., Int. Ed. 2019,

58 (4), 1609-1073. 183.

Li, N.; Yu, L.; Wang, J.; Gao, X.; Chen, Y.; Pan, W.; Tang, B., A Mitochondria-

Targeted Nanoradiosensitizer Activating Reactive Oxygen Species Burst for Enhanced Radiation Therapy. Chem. Sci. 2018, 9 (12), 3159-3164. 184.

Ni, K.; Lan, G.; Veroneau, S. S.; Duan, X.; Song, Y.; Lin, W., Nanoscale Metal-

Organic Frameworks for Mitochondria-Targeted Radiotherapy-Radiodynamic Therapy.

Nat. Commun. 2018, 9 (1), 4321-4333. 185.

Wen, Y.; Liu, K.; Yang, H.; Liu, Y.; Chen, L.; Liu, Z.; Huang, C.; Yi, T., Mitochondria-

Directed Fluorescent Probe for the Detection of Hydrogen Peroxide near Mitochondrial DNA. Anal. Chem. 2015, 87 (20), 10579-10584. 186.

López, V.; Villegas, M. R.; Rodríguez, V.; Villaverde, G.; Lozano, D.; Baeza, A.;

Vallet-Regí, M., Janus Mesoporous Silica Nanoparticles for Dual Targeting of Tumor Cells

ACS Paragon Plus Environment

72

Page 73 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

and Mitochondria. ACS Appl. Mater. Interfaces 2017, 9 (32), 26697-26706. 187.

Zhang, Y.; Zhang, C.; Chen, J.; Liu, L.; Hu, M.; Li, J.; Bi, H., Trackable

Mitochondria-Targeting Nanomicellar Loaded with Doxorubicin for Overcoming Drug Resistance. ACS Appl. Mater. Interfaces 2017, 9 (30), 25152-25163. 188.

Yu, P.; Yu, H.; Guo, C.; Cui, Z.; Chen, X.; Yin, Q.; Zhang, P.; Yang, X.; Cui, H.; Li,

Y., Reversal of Doxorubicin Resistance in Breast Cancer by Mitochondria-Targeted pHResponsive Micelles. Acta Biomater. 2015, 14, 115-124. 189.

Khatun, Z.; Choi, Y. S.; Kim, Y. G.; Yoon, K.; Nurunnabi, M.; Li, L.; Lee, E.; Kang,

H. C.; Huh, K. M., Bioreducible Poly(ethylene glycol)-Triphenylphosphonium Conjugate as a Bioactivable Mitochondria-Targeting Nanocarrier. Biomacromolecules 2017, 18 (4), 1074-1085. 190.

Assanhou, A. G.; Li, W.; Zhang, L.; Xue, L.; Kong, L.; Sun, H.; Mo, R.; Zhang, C.,

Reversal of Multidrug Resistance by Co-Delivery of Paclitaxel and Lonidamine Ssing a TPGS and Hyaluronic Acid Dual-Functionalized Liposome for Cancer Treatment.

Biomaterials 2015, 73, 284-295. 191.

Chen, Z.; Zhang, L.; Song, Y.; He, J.; Wu, L.; Zhao, C.; Xiao, Y.; Li, W.; Cai, B.;

Cheng, H.; Li, W., Hierarchical Targeted Hepatocyte Mitochondrial Multifunctional Chitosan Nanoparticles for Anticancer Drug Delivery. Biomaterials 2015, 52, 240-250. 192.

Cao, J.-J.; Tan, C.-P.; Chen, M.-H.; Wu, N.; Yao, D.-Y.; Liu, X.-G.; Ji, L.-N.; Mao,

Z.-W., Targeting Cancer Cell Metabolism with Mitochondria-Immobilized Phosphorescent Cyclometalated Iridium(III) Complexes. Chem. Sci. 2017, 8 (1), 631-640. 193.

Marrache, S.; Dhar, S., The Energy Blocker Inside The Power House:

Mitochondria Targeted Delivery of 3-Bromopyruvate. Chem. Sci. 2015, 6 (3), 1832-1845. 194.

Tuo, J.; Xie, Y.; Song, J.; Chen, Y.; Guo, Q.; Liu, X.; Ni, X.; Xu, D.; Huang, H.; Yin,

S.; Zhu, W.; Wu, J.; Hu, H., Development of a Novel Berberine-Mediated MitochondriaTargeting Nano-Platform for Drug-Resistant Cancer Therapy. J. Mater. Chem. B 2016, 4 (42), 6856-6864. 195.

Marrache, S.; Pathak, R. K.; Dhar, S., Detouring of Cisplatin to Access

Mitochondrial Genome for Overcoming Resistance. Proc. Natl. Acad. Sci. U. S. A. 2014,

111 (29), 10444-10449. 196.

Jiang, L.; Li, L.; He, X.; Yi, Q.; He, B.; Cao, J.; Pan, W.; Gu, Z., Overcoming Drug-

ACS Paragon Plus Environment

73

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 74 of 85

Resistant Lung Cancer by Paclitaxel Loaded Dual-Functional Liposomes with Mitochondria Targeting and pH-Response. Biomaterials 2015, 52, 126-139. 197.

Yang, L.; Gao, P.; Huang, Y.; Lu, X.; Chang, Q.; Pan, W.; Li, N.; Tang, B., Boosting

the photodynamic therapy efficiency with a mitochondria-targeted nanophotosensitizer.

Chin. Chem. Lett. 2019, DOI: 10.1016/j.cclet.2019.03.032. 198.

Hou, Z.; Zhang, Y.; Deng, K.; Chen, Y.; Li, X.; Deng, X.; Cheng, Z.; Lian, H.; Li, C.;

Lin, J., UV-Emitting Upconversion-Based TiO2 Photosensitizing Nanoplatform: NearInfrared Light Mediated in Vivo Photodynamic Therapy via Mitochondria-Involved Apoptosis Pathway. ACS Nano 2015, 9 (3), 2584-2599. 199.

Hu, Q.; Gao, M.; Feng, G.; Liu, B., Mitochondria-Targeted Cancer Therapy Using

a Light-up Probe with Aggregation-Induced-Emission Characteristics. Angew. Chem., Int.

Ed. 2014, 53 (51), 14225-14229. 200.

Pereira, P. M. R.; Silva, S.; Bispo, M.; Zuzarte, M.; Gomes, C.; Girao, H.; Cavaleiro,

J. A. S.; Ribeiro, C. A. F.; Tome, J. P. C.; Fernandes, R., Mitochondria-Targeted Photodynamic Therapy with a Galactodendritic Chlorin to Enhance Cell Death in Resistant Bladder Cancer Cells. Bioconjugate Chem 2016, 27 (11), 2762-2769. 201.

Shen, Y.; Sun, Y.; Yan, R.; Chen, E.; Wang, H.; Ye, D.; Xu, J.-J.; Chen, H.-Y.,

Rational Engineering of Semiconductor QDs Enabling Remarkable 1O2 Production for Tumor-Targeted Photodynamic Therapy. Biomaterials 2017, 148, 31-40. 202.

Hammerer, F.; Poyer, F.; Fourmois, L.; Chen, S.; Garcia, G.; Teulade-Fichou, M.-

P.; Maillard, P.; Mahuteau-Betzer, F., Mitochondria-Targeted Cationic PorphyrinTriphenylamine Hybrids for Enhanced Two-Photon Photodynamic Therapy. Bioorg. Med.

Chem. 2017, 26 (1), 107-118. 203.

Du, E.; Hu, X.; Roy, S.; Wang, P.; Deasy, K.; Mochizuki, T.; Zhang, Y., Taurine-

Modified Ru(II)-Complex Targets Cancerous Brain Cells for Photodynamic Therapy.

Chem. Commun. 2017, 53 (44), 6033-6036. 204.

Zhao, E.; Deng, H.; Chen, S.; Hong, Y.; Leung, C. W. T.; Lam, J. W. Y.; Tang, B.

Z., A Dual Functional AEE Fluorogen as a Mitochondrial-Specific Bioprobe and an Effective Photosensitizer for Photodynamic Therapy. Chem. Commun. 2014, 50 (92), 14451-14454. 205.

Raza, M. K.; Gautam, S.; Garai, A.; Mitra, K.; Kondaiah, P.; Chakravarty, A. R.,

ACS Paragon Plus Environment

74

Page 75 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Monofunctional BODIPY-Appended Imidazoplatin for Cellular Imaging and MitochondriaTargeted Photocytotoxicity. Inorg. Chem. 2017, 56 (18), 11019-11029. 206.

Qiu, K.; Ouyang, M.; Liu, Y.; Huang, H.; Liu, C.; Chen, Y.; Ji, L.; Chao, H., Two-

Photon Photodynamic Ablation of Tumor Cells by Mitochondria-Targeted Iridium(III) Complexes in Aggregate States. J. Mater. Chem. B 2017, 5 (27), 5488-5498. 207.

Zheng, Y.; Lu, H.; Jiang, Z.; Guan, Y.; Zou, J.; Wang, X.; Cheng, R.; Gao, H., Low-

Power White Light Triggered AIE Polymer Nanoparticles with High ROS Quantum Yield for Mitochondria-Targeted and Image-Guided Photodynamic Therapy. J. Mater. Chem. B 2017, 5 (31), 6277-6281. 208.

Liu, Y.; Zhang, J.; Zuo, C.; Zhang, Z.; Ni, D.; Zhang, C.; Wang, J.; Zhang, H.; Yao,

Z.; Bu, W., Upconversion Nano-Photosensitizer Targeting into Mitochondria for Cancer Apoptosis Induction and Cyt c Fluorescence Monitoring. Nano Res. 2016, 9 (11), 32573266. 209.

Zhang, D.-Y.; Zheng, Y.; Zhang, H.; He, L.; Tan, C.-P.; Sun, J.-H.; Zhang, W.;

Peng, X.; Zhan, Q.; Ji, L.-N.; Mao, Z.-W., Ruthenium Complex-Modified Carbon Nanodots for Lysosome-Targeted One- and Two-Photon Imaging and Photodynamic Therapy.

Nanoscale 2017, 9 (47), 18966-18976. 210.

Xu, J.; Zeng, F.; Wu, H.; Yu, C.; Wu, S., Dual-Targeting Nanosystem for Enhancing

Photodynamic Therapy Efficiency. ACS Appl. Mater. Interfaces 2015, 7 (17), 9287-9296. 211.

Tong, H.; Du, J.; Li, H.; Jin, Q.; Wang, Y.; Ji, J., Programmed Photosensitizer

Conjugated Supramolecular Nanocarriers with Dual Targeting Ability for Enhanced Photodynamic Therapy. Chem. Commun. 2016, 52 (80), 11935-11938. 212.

Guan, Y.; Lu, H.; Li, W.; Zheng, Y.; Jiang, Z.; Zou, J.; Gao, H., Near-Infrared

Triggered Upconversion Polymeric Nanoparticles Based on Aggregation-Induced Emission and Mitochondria Targeting for Photodynamic Cancer Therapy. ACS Appl.

Mater. Interfaces 2017, 9 (32), 26731-26739. 213.

Liu, J.; Jin, C.; Yuan, B.; Liu, X.; Chen, Y.; Ji, L.; Chao, H., Selectively Lighting up

Two-Photon Photodynamic Activity in Mitochondria with AIE-Active Iridium(III) Complexes. Chem. Commun. 2017, 53 (12), 2052-2055. 214.

Gui, C.; Zhao, E.; Kwok, R. T. K.; Leung, A. C. S.; Lam, J. W. Y.; Jiang, M.; Deng,

H.; Cai, Y.; Zhang, W.; Su, H.; Tang, B. Z., AIE-Active Theranostic System: Selective

ACS Paragon Plus Environment

75

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 76 of 85

Staining and Killing of Cancer Cells. Chem. Sci. 2017, 8 (3), 1822-1830. 215.

Zhang, C.-J.; Hu, Q.; Feng, G.; Zhang, R.; Yuan, Y.; Lu, X.; Liu, B., Image-Guided

Combination Chemotherapy and Photodynamic Therapy Using a Mitochondria-Targeted Molecular Probe with Aggregation-Induced Emission Characteristics. Chem. Sci. 2015, 6 (8), 4580-4586. 216.

Jung, H. S.; Han, J.; Lee, J.-H.; Lee, J. H.; Choi, J.-M.; Kweon, H.-S.; Han, J. H.;

Kim, J.-H.; Byun, K. M.; Jung, J. H.; Kang, C.; Kim, J. S., Enhanced NIR RadiationTriggered Hyperthermia by Mitochondrial Targeting. J. Am. Chem. Soc. 2015, 137 (8), 3017-3023. 217.

Guo, M.; Xiang, H.-J.; Wang, Y.; Zhang, Q.-L.; An, L.; Yang, S.-P.; Ma, Y. C.;

Wang, Y.; Liu, J.-G., Ruthenium Nitrosyl Functionalized Graphene Quantum Dots as an Efficient Nanoplatform for NIR-Light-Controlled and Mitochondria-Targeted Delivery of Nitric Oxide Combined with Photothermal Therapy. Chem. Commun. 2017, 53 (22), 32533256. 218.

Jung, H. S.; Lee, J.-H.; Kim, K.; Koo, S.; Verwilst, P.; Sessler, J. L.; Kang, C.; Kim,

J. S., A Mitochondria-Targeted Cryptocyanine-Based Photothermogenic Photosensitizer.

J. Am. Chem. Soc. 2017, 139 (29), 9972-9978. 219.

Song, G.; Cheng, L.; Chao, Y.; Yang, K.; Liu, Z., Emerging Nanotechnology and

Advanced Materials for Cancer Radiation Therapy. Adv. Mater. 2017, 29 (32), 17009961701021. 220.

Yu, C. Y. Y.; Xu, H.; Ji, S.; Kwok, R. T. K.; Lam, J. W. Y.; Li, X.; Krishnan, S.; Ding,

D.; Tang, B. Z., Mitochondrion-Anchoring Photosensitizer with Aggregation-Induced Emission Characteristics Synergistically Boosts the Radiosensitivity of Cancer Cells to Ionizing Radiation. Adv. Mater. 2017, 29 (15), 1606167-1606175. 221.

Chen, Y.; Li, N.; Wang, J.; Zhang, X.; Pan, W.; Yu, L.; Tang, B., Enhancement of

Mitochondrial ROS Accumulation and Radiotherapeutic Efficacy Using a Gd-Doped Titania Nanosensitizer. Theranostics 2019, 9, 167-178. 222.

Han, K.; Zhu, J.-Y.; Jia, H.-Z.; Wang, S.-B.; Li, S.-Y.; Zhang, X.-Z.; Han, H.-Y.,

Mitochondria-Targeted Chimeric Peptide for Trinitarian Overcoming of Drug Resistance.

ACS Appl. Mater. Interfaces 2016, 8 (38), 25060-25068. 223.

Ju, E.; Li, Z.; Liu, Z.; Ren, J.; Qu, X., Near-Infrared Light-Triggered Drug-Delivery

ACS Paragon Plus Environment

76

Page 77 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Vehicle for Mitochondria-Targeted Chemo-Photothermal Therapy. ACS Appl. Mater.

Interfaces 2014, 6 (6), 4364-4370. 224.

Zhang, D.-Y.; Zheng, Y.; Tan, C.-P.; Sun, J.-H.; Zhang, W.; Ji, L.-N.; Mao, Z.-W.,

Graphene Oxide Decorated with Ru(II)-Polyethylene Glycol Complex for LysosomeTargeted Imaging and Photodynamic/Photothermal Therapy. ACS Appl. Mater. Interfaces 2017, 9 (8), 6761-6771. 225.

Luo, S.; Tan, X.; Fang, S.; Wang, Y.; Liu, T.; Wang, X.; Yuan, Y.; Sun, H.; Qi, Q.;

Shi, C., Mitochondria-Targeted Small-Molecule Fluorophores for Dual Modal Cancer Phototherapy. Adv. Funct. Mater. 2016, 26 (17), 2826-2835. 226.

Zhang, R. X.; Li, L. Y.; Li, J.; Xu, Z. S.; Abbasi, A. Z.; Lin, L.; Amini, M. A.; Weng,

W. Y.; Sun, Y.; Rauth, A. M.; Wu, X. Y., Coordinating Biointeraction and Bioreaction of a Nanocarrier Material and an Anticancer Drug to Overcome Membrane Rigidity and Target Mitochondria in Multidrug-Resistant Cancer Cells. Adv. Funct. Mater. 2017, 27 (39), 1700804-1700815. 227.

Tan, X.; Luo, S.; Long, L.; Wang, Y.; Wang, D.; Fang, S.; Ouyang, Q.; Su, Y.;

Cheng, T.; Shi, C., Structure-Guided Design and Synthesis of a Mitochondria-Targeting Near-Infrared Fluorophore with Multimodal Therapeutic Activities. Adv. Mater. 2017, 29 (43), 1704196-1704204. 228.

Yu, B.; Wei, H.; He, Q.; Ferreira, C. A.; Kutyreff, C. J.; Ni, D.; Rosenkrans, Z. T.;

Cheng, L.; Yu, F.; Engle, J. W.; Lan, X.; Cai, W., Efficient Uptake of (177)Lu-PorphyrinPEG

Nanocomplexes

by

Tumor

Mitochondria

for

Multimodal-Imaging-Guided

Combination Therapy. Angew. Chem., Int. Ed. 2018, 57 (1), 218-222. 229. Y.-X.;

Chen, S.; Lei, Q.; Qiu, W.-X.; Liu, L.-H.; Zheng, D.-W.; Fan, J.-X.; Rong, L.; Sun, Zhang,

X.-Z.,

Mitochondria-Targeting

"Nanoheater"

for

Enhanced

Photothermal/Chemo-Therapy. Biomaterials 2017, 117, 92-104. 230.

Xiong, H.; Du, S.; Ni, J.; Zhou, J.; Yao, J., Mitochondria and Nuclei Dual-Targeted

Heterogeneous Hydroxyapatite Nanoparticles for Enhancing Therapeutic Efficacy of Doxorubicin. Biomaterials 2016, 94, 70-83. 231.

Shin, W. S.; Park, S. K.; Verwilst, P.; Koo, S.; Lee, J. H.; Chi, S.-G.; Kim, J. S.,

Targeted combinational therapy inducing mitochondrial dysfunction. Chem. Commun. 2017, 53 (7), 1281-1284.

ACS Paragon Plus Environment

77

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

232.

Page 78 of 85

Song, Y.; Shi, Q.; Zhu, C.; Luo, Y.; Lu, Q.; Li, H.; Ye, R.; Du, D.; Lin, Y.,

Mitochondrial-Targeted Multifunctional Mesoporous Au@Pt Nanoparticles for Dual-Mode Photodynamic and Photothermal Therapy of Cancers. Nanoscale 2017, 9 (41), 1581315824. 233.

Yue, C.; Yang, Y.; Song, J.; Alfranca, G.; Zhang, C.; Zhang, Q.; Yin, T.; Pan, F.;

de la Fuente, J. M.; Cui, D., Mitochondria-Targeting Near-Infrared Light-Triggered Thermosensitive Liposomes for Localized Photothermal and Photodynamic Ablation of Tumors Combined with Chemotherapy. Nanoscale 2017, 9 (31), 11103-11118. 234.

Mou, J.; Lin, T.; Huang, F.; Shi, J.; Chen, H., A New Green Titania with Enhanced

NIR Absorption for Mitochondria-Targeted Cancer Therapy. Theranostics 2017, 7 (6), 1531-1542. 235.

Yue, C.; Yang, Y.; Zhang, C.; Alfranca, G.; Cheng, S.; Ma, L.; Liu, Y.; Zhi, X.; Ni,

J.; Jiang, W.; Song, J.; de la Fuente, J. M.; Cui, D., ROS-Responsive MitochondriaTargeting Blended Nanoparticles: Chemo- and Photodynamic Synergistic Therapy for Lung Cancer with On-Demand Drug Release upon Irradiation with a Single Light Source.

Theranostics 2016, 6 (13), 2352-2366. 236.

Peng, J.; Yang, Q.; Li, W.; Tan, L.; Xiao, Y.; Chen, L.; Hao, Y.; Qian, Z.,

Erythrocyte-Membrane-Coated Prussian Blue/Manganese Dioxide Nanoparticles as H2O2-Responsive Oxygen Generators To Enhance Cancer Chemotherapy/Photothermal Therapy. ACS Appl. Mater. Interfaces 2017, 9 (51), 44410-44422. 237.

Chen, Y.; Zhong, H.; Wang, J.; Wan, X.; Li, Y.; Pan, W.; Li, N.; Tang, B., Catalase-

like metal-organic framework nanoparticles to enhance radiotherapy in hypoxic cancer and prevent cancer recurrence. Chem. Sci. 2019, DOI: 10.1039/c9sc00747d. 238.

Liu, Y.; Ji, X.; Tong, W. W. L.; Askhatova, D.; Yang, T.; Cheng, H.; Wang, Y.; Shi,

J., Engineering Multifunctional RNAi Nanomedicine To Concurrently Target Cancer Hallmarks for Combinatorial Therapy. Angew. Chem., Int. Ed. 2018, 57 (6), 1510-1513. 239.

Wang, F.-X.; Chen, M.-H.; Lin, Y.-N.; Zhang, H.; Tan, C.-P.; Ji, L.-N.; Mao, Z.-W.,

Dual Functions of Cyclometalated Iridium(III) Complexes: Anti-Metastasis and LysosomeDamaged Photodynamic Therapy. ACS Appl. Mater. Interfaces 2017, 9 (49), 4247142481. 240.

Wang, T.; Wang, D.; Yu, H.; Wang, M.; Liu, J.; Feng, B.; Zhou, F.; Yin, Q.; Zhang,

ACS Paragon Plus Environment

78

Page 79 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Z.; Huang, Y.; Li, Y., Intracellularly Acid-Switchable Multifunctional Micelles for Combinational Photo/Chemotherapy of the Drug-Resistant Tumor. ACS Nano 2016, 10 (3), 3496-3508. 241.

Daum, S.; Reshetnikov, M. S. V.; Sisa, M.; Dumych, T.; Lootsik, M. D.; Bilyy, R.;

Bila, E.; Janko, C.; Alexiou, C.; Herrmann, M.; Sellner, L.; Mokhir, A., Lysosome-Targeting Amplifiers of Reactive Oxygen Species as Anticancer Prodrugs. Angew. Chem., Int. Ed. 2017, 56 (49), 15545–15549. 242. Highly

Huang, H.; Yu, B.; Zhang, P.; Huang, J.; Chen, Y.; Gasser, G.; Ji, L.; Chao, H., Charged

Ruthenium(II)

Polypyridyl

Complexes

as

Lysosome-Localized

Photosensitizers for Two-Photon Photodynamic Therapy. Angew. Chem., Int. Ed. 2015,

54 (47), 14049-14052. 243.

Ke, C.-J.; Chiang, W.-L.; Liao, Z.-X.; Chen, H.-L.; Lai, P.-S.; Sun, J.-S.; Sung, H.-

W., Real-Time Visualization of pH-Responsive PLGA Hollow Particles Containing a GasGenerating Agent Targeted for Acidic Organelles for Overcoming Multi-Drug Resistance.

Biomaterials 2013, 34 (1), 1-10. 244.

Xiang, H.-J.; Deng, Q.; An, L.; Guo, M.; Yang, S.-P.; Liu, J.-G., Tumor Cell Specific

and Lysosome-Targeted Delivery of Nitric Oxide for Enhanced Photodynamic Therapy Triggered by 808 nm Near-Infrared Light. Chem. Commun. 2016, 52 (1), 148-151. 245.

Li, M.; Tian, R.; Fan, J.; Du, J.; Long, S.; Peng, X., A Lysosome-Targeted BODIPY

as Potential NIR Photosensitizer for Photodynamic Therapy. Dyes Pigm. 2017, 147, 99105. 246.

Xiang, H.-J.; Guo, M.; An, L.; Yang, S.-P.; Zhang, Q.-L.; Liu, J.-G., A Multifunctional

Nanoplatform for Lysosome Targeted Delivery of Nitric Oxide and Photothermal Therapy Under 808 nm Near-Infrared Light. J. Mater. Chem. B 2016, 4 (27), 4667-4674. 247.

Nair, J. B.; Mohapatra, S.; Ghosh, S.; Maiti, K. K., 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 (12), 24032406. 248.

Dai, L.; Liu, J.; Luo, Z.; Li, M.; Cai, K., Tumor Therapy: Targeted Drug Delivery

Systems. J. Mater. Chem. B 2016, 4 (42), 6758-6772. 249.

Domenech, M.; Marrero-Berrios, I.; Torres-Lugo, M.; Rinaldi, C., Lysosomal

ACS Paragon Plus Environment

79

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 80 of 85

Membrane Permeabilization by Targeted Magnetic Nanoparticles in Alternating Magnetic Fields. ACS Nano 2013, 7 (6), 5091-5101. 250.

Jiao, X.; Li, Y.; Niu, J.; Xie, X.; Wang, X.; Tang, B., Small-Molecule Fluorescent

Probes for Imaging and Detection of Reactive Oxygen, Nitrogen, and Sulfur Species in Biological Systems. Anal. Chem. 2018, 90 (1), 533-555. 251.

Pan, W.; Wang, H.; Yang, L.; Yu, Z.; Li, N.; Tang, B., Ratiometric Fluorescence

Nanoprobes for Subcellular pH Imaging with a Single-Wavelength Excitation in Living Cells. Anal. Chem. 2016, 88 (13), 6743-6748. 252.

Zhu, B.; Li, P.; Shu, W.; Wang, X.; Liu, C.; Wang, Y.; Wang, Z.; Wang, Y.; Tang,

B., Highly Specific and Ultrasensitive Two-Photon Fluorescence Imaging of Native HOCl in Lysosomes and Tissues Based on Thiocarbamate Derivatives. Anal. Chem. 2016, 88 (24), 12532-12538. 253.

Xu, H.; Li, Q.; Wang, L.; He, Y.; Shi, J.; Tang, B.; Fan, C., Nanoscale optical probes

for cellular imaging. Chem. Soc. Rev. 2014, 43 (8), 2650-2661. 254.

Hu, W.; Ma, H.; Hou, B.; Zhao, H.; Ji, Y.; Jiang, R.; Hu, X.; Lu, X.; Zhang, L.; Tang,

Y.; Fan, Q.; Huang, W., Engineering Lysosome-Targeting BODIPY Nanoparticles for Photoacoustic Imaging and Photodynamic Therapy under Near-Infrared Light. ACS Appl.

Mater. Interfaces 2016, 8 (19), 12039-12047. 255.

Zhao, S.; Niu, G.; Wu, F.; Yan, L.; Zhang, H.; Zhao, J.; Zeng, L.; Lan, M.,

Lysosome-Targetable

Polythiophene

Nanoparticles

for

Two-Photon

Excitation

Photodynamic Therapy and Deep Tissue Imaging. J. Mater. Chem. B 2017, 5 (20), 36513657. 256.

Wan, J.; Sun, L.; Wu, P.; Wang, F.; Guo, J.; Cheng, J.; Wang, C., Synthesis of

Indocyanine Green Functionalized Comblike Poly(Aspartic Acid) Derivatives for Enhanced Cancer Cell Ablation by Targeting the Endoplasmic Reticulum. Polym. Chem. 2018, 9 (10), 1206-1215. 257.

Wang, Y.; Luo, S.; Zhang, C.; Liao, X.; Liu, T.; Jiang, Z.; Liu, D.; Tan, X.; Long, L.;

Wang, Y.; Chen, Z.; Liu, Y.; Yang, F.; Gan, Y.; Shi, C., An NIR-Fluorophore-Based Therapeutic Endoplasmic Reticulum Stress Inducer. Adv. Mater. 2018, 30, 18004751800483. 258.

Yu, L.; Wang, Q.; Yeung, K. W.; Fong, W. P.; Lo, P. C., A Biotinylated and

ACS Paragon Plus Environment

80

Page 81 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Endoplasmic Reticulum-Targeted Glutathione-Responsive Zinc(II) Phthalocyanine for Targeted Photodynamic Therapy. Chem. - Asian J. 2018, 13 (22), 3509-3517. 259.

Zhou, Y.; Cheung, Y. K.; Ma, C.; Zhao, S.; Gao, D.; Lo, P. C.; Fong, W. P.; Wong,

K. S.; Ng, D. K. P., Endoplasmic Reticulum-Localized Two-Photon-Absorbing Boron Dipyrromethenes as Advanced Photosensitizers for Photodynamic Therapy. J. Med.

Chem. 2018, 61 (9), 3952-3961. 260.

Dabrowski, J. M.; Arnaut, L. G., Photodynamic Therapy (PDT) of Cancer: From

Local to Systemic Treatment. Photochem. Photobiol. Sci. 2015, 14 (10), 1765-1780. 261.

Banerjee, S.; Dixit, A.; Shridharan, R. N.; Karande, A. A.; Chakravarty, A. R.,

Endoplasmic

Reticulum

Targeted

Chemotherapeutics:

the

Remarkable

Photo-

Cytotoxicity of an Oxovanadium(IV) Vitamin-B6 Complex in Visible Light. Chem.

Commun. 2014, 50 (42), 5590-5592. 262.

Nam, J. S.; Kang, M.-G.; Kang, J.; Park, S.-Y.; Lee, S. J. C.; Kim, H.-T.; Seo, J. K.;

Kwon, O.-H.; Lim, M. H.; Rhee, H.-W.; Kwon, T.-H., Endoplasmic Reticulum-Localized Iridium(III) Complexes as Efficient Photodynamic Therapy Agents via Protein Modifications. J. Am. Chem. Soc. 2016, 138 (34), 10968-10977. 263.

Xue, F.; Wen, Y.; Wei, P.; Gao, Y.; Zhou, Z.; Xiao, S.; Yi, T., A Smart Drug: a pH-

Responsive Photothermal Ablation Agent for Golgi Apparatus Activated Aancer Therapy.

Chem. Commun. 2017, 53 (48), 6424-6427. 264.

Wan, X.; Zhang, X.; Pan, W.; Liu, B.; Yu, L.; Wang, H.; Li, N.; Tang, B., Ratiometric

Fluorescent

Quantification

of

the

Size-Dependent

Cellular

Toxicity

of

Silica

Nanoparticles. Anal Chem 2019, DOI: 10.1021/acs.analchem.9b00633. 265.

Cardillo, D.; Tehei, M.; Hossain, M. S.; Islam, M. M.; Bogusz, K.; Shi, D.; Mitchell,

D.; Lerch, M.; Rosenfeld, A.; Corde, S.; Konstantinov, K., Synthesis-Dependent Surface Defects and Morphology of Hematite Nanoparticles and Their Effect on Cytotoxicity in Vitro. ACS Appl. Mater. Interfaces 2016, 8 (9), 5867-5876. 266.

Schutz, C. A.; Juillerat-Jeanneret, L.; Mueller, H.; Lynch, I.; Riediker, M.;

NanoImpactNet, C., Therapeutic Nanoparticles in Clinics and Under Clinical Evaluation.

Nanomedicine 2013, 8 (3), 449-467. 267.

Anselmo, A. C.; Mitragotri, S., A Review of Clinical Translation of Inorganic

Nanoparticles. AAPS J. 2015, 17 (5), 1041-1054.

ACS Paragon Plus Environment

81

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

268.

Page 82 of 85

Anselmo, A. C.; Mitragotri, S., Nanoparticles in the Clinic. Bioeng. Transl. Med.

2016, 1 (1), 10-29. 269.

Downing, N. S.; Aminawung, J. A.; Shah, N. D.; Krumholz, H. M.; Ross, J. S.,

Clinical Trial Evidence Supporting FDA Approval of Novel Therapeutic Agents, 20052012. JAMA 2014, 311 (4), 368-377. 270.

Hassan, S.; Prakash, G.; Ozturk, A.; Saghazadeh, S.; Sohail, M. F.; Seo, J.;

Dockmeci, M.; Zhang, Y. S.; Khademhosseini, A., Evolution and Clinical Translation of Drug Delivery Nanomaterials. Nano Today 2017, 15, 91-106. 271.

Bobo, D.; Robinson, K. J.; Islam, J.; Thurecht, K. J.; Corrie, S. R., Nanoparticle-

Based Medicines: A Review of FDA-Approved Materials and Clinical Trials to Date.

Pharm. Res. 2016, 33 (10), 2373-2387. 272.

Zhang, L.; Yi, H.; Song, J.; Huang, J.; Yang, K.; Tan, B.; Wang, D.; Yang, N.; Wang,

Z.; Li, X., Mitochondria-Targeted and Ultrasound-Activated Nanodroplets for Enhanced Deep-Penetration Sonodynamic Cancer Therapy. ACS Appl. Mater. Interfaces 2019, 11 (9), 9355-9366. 273.

Xu, J.; Yan, B.; Du, X.; Xiong, J.; Zhou, M.; Wang, H.; Du, Z., Acidity-Triggered

Zwitterionic Prodrug Nano-Carriers with AIE Properties and Amplification of Oxidative Stress for Mitochondria-Targeted Cancer Theranostics. Polym. Chem. 2019, 10 (8), 983990. 274.

Zhang, W.; Hu, X.; Shen, Q.; Xing, D., Mitochondria-Specific Drug Release and

Reactive Oxygen Species Burst Induced by Polyprodrug Nanoreactors Can Enhance Chemotherapy. Nat. Commun. 2019, 10 (1), 1704-1717. 275.

Liu, C.; Zhou, L.; Wei, F.; Li, L.; Zhao, S.; Gong, P.; Cai, L.; Wong, K. M., Versatile

Strategy To Generate a Rhodamine Triplet State as Mitochondria-Targeting Visible-Light Photosensitizers for Efficient Photodynamic Therapy. ACS Appl. Mater. Interfaces 2019,

11 (9), 8797-8806. 276.

Bao, Y. W.; Hua, X. W.; Li, Y. H.; Jia, H. R.; Wu, F. G., Endoplasmic Reticulum-

Targeted Phototherapy Using One-Step Synthesized Trace Metal-Doped CarbonDominated Nanoparticles: Laser-Triggered Nucleolar Delivery and Increased Tumor Accumulation. Acta Biomater. 2019, 88, 462-476. 277.

Li, H.; Yan, W.; Suo, X.; Peng, H.; Yang, X.; Li, Z.; Zhang, J.; Liu, D., Nucleus-

ACS Paragon Plus Environment

82

Page 83 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Targeted Nano Delivery System Eradicates Cancer Stem Cells by Combined Thermotherapy and Hypoxia-Activated Chemotherapy. Biomaterials 2019, 200, 1-14. 278.

Wang, F.; Zhang, Y.; Liu, Z.; Du, Z.; Zhang, L.; Ren, J.; Qu, X., A Biocompatible

Heterogeneous MOF-Cu Catalyst for In Vivo Drug Synthesis in Targeted Subcellular Organelles. Angew. Chem., Int. Ed. 2019, DOI: 10.1002/anie.201901760. 279.

Zhang, P.; Huang, H.; Banerjee, S.; Clarkson, G. J.; Ge, C.; Imberti, C.; Sadler, P.

J., Nucleus-Targeted Organoiridium-Albumin Conjugate for Photodynamic Cancer Therapy. Angew. Chem., Int. Ed. 2019, 58 (8), 2350-2354. 280.

Wang, W.; Liu, J.; Feng, W.; Du, S.; Ge, R.; Li, J.; Liu, Y.; Sun, H.; Zhang, D.;

Zhang, H.; Yang, B., Targeting Mitochondria with Au-Ag@Polydopamine Nanoparticles for Papillary Thyroid Cancer Therapy. Biomater. Sci. 2019, 7 (3), 1052-1063. 281.

Niu, N.; Zhou, H.; Liu, N.; Jiang, H.; Hussain, E.; Hu, Z.; Yu, C., A Smart Perylene

Derived Photosensitizer for Lysosome-Targeted and Self-Assessed Photodynamic Therapy. Chem. Commun. 2019, 55 (8), 1036-1039. 282.

Cheng, H.; Zheng, R. R.; Fan, G. L.; Fan, J. H.; Zhao, L. P.; Jiang, X. Y.; Yang, B.;

Yu, X. Y.; Li, S. Y.; Zhang, X. Z., Mitochondria and Plasma Membrane Dual-Targeted Chimeric Peptide for Single-Agent Synergistic Photodynamic Therapy. Biomaterials 2019, 188, 1-11. 283.

Zhang, T.; Li, Y.; Zheng, Z.; Ye, R.; Zhang, Y.; Kwok, R. T. K.; Lam, J. W. Y.; Tang,

B. Z., In Situ Monitoring Apoptosis Process by a Self-Reporting Photosensitizer. J. Am.

Chem. Soc. 2019, 141 (14), 5612-5616. 284.

Yao, D.; Yang, S.; Wang, Y.; Bian, K.; Yang, W.; Wang, D.; Zhang, B., An ALP-

Activatable and Mitochondria-Targeted Probe for Prostate Cancer-Specific Bimodal Imaging and Aggregation-Enhanced Photothermal Therapy. Nanoscale 2019, 11 (13), 6307-6314. 285.

Cao, Y.; Wu, T.; Zhang, K.; Meng, X.; Dai, W.; Wang, D.; Dong, H.; Zhang, X.,

Engineered Exosome-Mediated Near-Infrared-II Region V2C Quantum Dot Delivery for Nucleus-Target Low-Temperature Photothermal Therapy. ACS Nano 2019, 13 (2), 14991510. 286.

Wang, D.; Huang, H.; Zhou, M.; Lu, H.; Chen, J.; Chang, Y. T.; Gao, J.; Chai, Z.;

Hu, Y., A Thermoresponsive Nanocarrier for Mitochondria-Targeted Drug Delivery.

ACS Paragon Plus Environment

83

ACS Applied Materials & Interfaces 1 2 3 4 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 58 59 60

Page 84 of 85

Chem. Commun. 2019, 55 (28), 4051-4054. 287.

Cheng, Y.; Sun, C.; Liu, R.; Yang, J.; Dai, J.; Zhai, T.; Lou, X.; Xia, F., A

Multifunctional Peptide-Conjugated AIEgen for Efficient and Sequential Targeted Gene Delivery into the Nucleus. Angew. Chem., Int. Ed. 2019, 58 (15), 5049-5053. 288.

Tan, Y.; Yang, X.; Dai, S.; Lian, K.; Wen, L.; Zhu, Y.; Meng, T.; Liu, X.; Yuan, H.;

Hu, F., In Vivo Programming of Tumor Mitochondria-Specific Doxorubicin Delivery by A Cationic Glycolipid Polymer for Enhanced Antitumor Activity. Polym. Chem. 2019, 10 (4), 512-525. 289.

Gong, N.; Ma, X.; Ye, X.; Zhou, Q.; Chen, X.; Tan, X.; Yao, S.; Huo, S.; Zhang, T.;

Chen, S.; Teng, X.; Hu, X.; Yu, J.; Gan, Y.; Jiang, H.; Li, J.; Liang, X. J., Carbon-DotSupported Atomically Dispersed Gold as a Mitochondrial Oxidative Stress Amplifier for Cancer Treatment. Nat. Nanotechnol. 2019, 14 (4), 379-387. 290.

Cong, Y.; Ji, L.; Gao, Y. J.; Liu, F. H.; Cheng, D. B.; Hu, Z.; Qiao, Z. Y.; Wang, H.,

Microenvironment-Induced in Situ Self-Assembly of Polymer-Peptide Conjugates that Attack Solid Tumors Deeply. Angew. Chem., Int. Ed. 2019, 58 (14), 4632-4637. 291.

Chen, W.; Liu, J.; Wang, Y.; Jiang, C.; Yu, B.; Sun, Z.; Lu, L., A C5N2 Nanoparticle

Based Direct Nucleus Delivery Platform for Synergistic Cancer Therapy. Angew. Chem.,

Int. Ed. 2019, 58 (19), 6290-6294. 292. Y.;

Cheng, D. B.; Zhang, X. H.; Gao, Y. J.; Ji, L.; Hou, D.; Wang, Z.; Xu, W.; Qiao, Z. Wang,

H.,

Endogenous

Reactive

Oxygen

Species-Triggered

Morphology

Transformation for Enhanced Cooperative Interaction with Mitochondria. J. Am. Chem.

Soc. 2019, DOI: 10.1021/jacs.8b07727. 293.

Kirakci, K.; Zelenka, J.; Rumlova, M.; Cvacka, J.; Ruml, T.; Lang, K., Cationic

Octahedral Molybdenum Cluster Complexes Functionalized with Mitochondria-Targeting Ligands: Photodynamic Anticancer and Antibacterial Activities. Biomater. Sci. 2019, 7 (4), 1386-1392.

ACS Paragon Plus Environment

84

Page 85 of 85 1 2 3 4 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 58 59 60

ACS Applied Materials & Interfaces

Table of Contents

ACS Paragon Plus Environment

85