Porphyrin Composite Photosensitizer: A Facile Way to

Mar 22, 2019 - Engineered cells detect tiny tumors. Detecting cancer early is difficult. Imaging approaches, like positron-emission tomography (PET), ...
0 downloads 0 Views 690KB Size
Subscriber access provided by - Access paid by the | UCSB Libraries

Biological and Medical Applications of Materials and Interfaces

G-Quadruplex/Porphyrin Composite Photosensitizer: A Facile Way to Promote Absorption Redshift and Photodynamic Therapy Efficacy Meng Cheng, Yunxi Cui, Jing Wang, Jing Zhang, Li-Na Zhu, and De-Ming Kong ACS Appl. Mater. Interfaces, Just Accepted Manuscript • DOI: 10.1021/acsami.9b02695 • Publication Date (Web): 22 Mar 2019 Downloaded from http://pubs.acs.org on March 24, 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 10 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

G-Quadruplex/Porphyrin Composite Photosensitizer: A Facile Way to Promote Absorption Redshift and Photodynamic Therapy Efficacy Meng Chenga, Yun-Xi Cuib, Jing Wangb, Jing Zhangb, Li-Na Zhua,*, De-Ming Kongb Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, China Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry, Nankai University, Tianjin, 300071, China KEYWORDS Composite photosensitizer, photodynamic therapy, cationic porphyrin, G-quadruplex, MnO2 nanosheet. a

b

ABSTRACT: Photosensitizer is one of the most important elements of photodynamic therapy (PDT). Herein, we reported a novel strategy to prepare a new series of composite photosensitizers. The composite photosensitizer was prepared by simply mixing DNA G-quadruplexes with a hydrophilic porphyrin (TMPipEOPP)4+•4I−. Compared to the conventional porphyrin photosensitizers, the excitation wavelength of the composite one has been ~50 nm red-shifted (from 650 to 700 nm), which is beneficial to the penetration of the light. Moreover, the composite photosensitizer showed about 7.4-fold increase of light absorption efficiency, thus greatly enhancing the singlet oxygen (1O2) generation capacity and PDT efficacy. What's more, the introduction of nucleic acids in the composite photosensitizer could also provide some extra charming properties such as the targeted recognition ability conferred by aptamer and high capability to assemble with various of drug carriers. We demonstrated that the composite photosensitizer could be easily assembled with MnO2 nanosheet. The obtained nanodevice integrated the merits of composite photosensitizer and MnO2 nanosheet, thus showing strong near-infrared absorption, high 1O2 generation efficiency, avoided nonideal 1O2 consumption by glutathione and in situ O2 generation to relieve tumor hypoxia. This nanodevice showed greatly improved PDT efficacy both in vitro and in vivo, presenting huge potential for applications in clinical therapy for tumors.

INTRODUCTION Malignant tumors seriously threaten human health and social development.1 In recent years, majority of treatment of tumors depends on surgical resection, chemotherapy and radiotherapy, these methods suffer from unsatisfactory clinical efficacy, severe side effects, poor prognosis and low patient survival rate.2-4 To address those challenges, targeting therapy has been developed extensively over the years by using highly specific molecular targeted drugs as "high-precision smart bombs" to initiate apoptosis of the cancer cells selectively.1 As one of the most promising targeted therapeutic strategies, photodynamic therapy (PDT) has been widely applied in tumor treatment.4-9 Photosensitizer, light and molecular oxygen (O2) are the three key components of PDT. An ideal photosensitizer should be efficiently convert O2 to reactive oxygen species (ROS, mainly singlet oxygen 1O2) upon irradiation, resulting apoptosis of cancer cells through the reaction between ROS and biological macromolecules.10-12 On the other hand, the photosensitizer should perform no cytotoxicity in dark. Compared with the traditional methods of cancer treatment, PDT shows the advantages of tiny trauma, good selectivity, low toxic and side effects, negligible drug resistance, repeatable application, and capability to be combined with other therapies. The development of PDT is always synchronous with the improvement of the photosensitizers.13-15 Porphyrin and its derivatives have been identified as important candidates in PDT photosensitizer synthesis, because of the high photosensitivity, good 1O2 quantum yield, and chemical

versatility of those compounds.16-22 However, the majority of currently reported porphyrin-based photosensitizers suffer from the drawbacks of poor water solubility (results in molecular aggregation under physiological conditions and limited pharmacokinetic properties), short wavelength of absorption light (result in low tissue penetration ability and reduced potential in the treatment of deep tumor) and low 1O2 generation efficiency (result in poor PDT efficiency).23 Improvement of the penetration depth and PDT efficacy is always the major concern in PDT studies.24 In addition, the non-specificity of the photosensitizers may cause accumulation of them in both cancer cells and normal cells, which not only reduces the efficiency of PDT but also increases the risk of side effects. Therefore, it is still urgent to develop a kind of hydrophilic photosensitiers with good tumor-specificity and high 1O2 generation efficiency under the stimulation of near-infrared (NIR) light. Besides photosensitizers, porphyrin and its derivatives have been widely applied in many fields taking advantage of their excellent optical and photovoltaic properties. For example, they are considered as promising probes and specific ligands of G-quadruplex, a kind of unique nucleic acid structures discovered to distribute broadly in human genome, showing great potential in specific G-quadruplex-probing and tumortargeted anticancer drug design.25-35 Recently, we have designed and synthesized series of water soluble cationic porphyrins with high G-quadruplex specificity.32-35 Intriguingly, we found that the G-quadruplex/porphyrin complexes formed by simple mixing of them showed a new

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

Scheme 1. Preparation of G-quadruplex/porphyrin composite photosensitizer and its application in PDT treatment of solid tumor after assembly with MnO2 nanosheet.

RESULT AND DISUSSION Characterization of composite photosensitizer. A water soluble cationic porphyrin (TMPipEOPP)4+·4I− (abbreviated as TMPipEOPP in the following) was selected as the model porphyrin to construct the photosensitizer (Figure 1a).34 Figure 1b showed the UV-Vis absorption spectra of free TMPipEOPP and the mixtures of TMPipEOPP and DNA with different structures such as single-stranded, double-stranded and Gquadruplex ones (see Table S1 for the detailed sequences). As a result, free TMPipEOPP shows a strong Soret absorption peak at 421 nm and four weak Q absorption bands centered at 521, 558, 594 and 650 nm. The strongest absorption was located at the Soret band, which was not the ideal wavelength for PDT due to the high photocytotoxicity and poor tissue penetration of the light at this band. In fact, light with a wavelength of 650 nm was usually selected because it was more close to the NIR window (650-900 nm), performing better PDT efficiency.39 However, the TMPipEOPP showed very weak absorption at 650 nm, the molar absorption

coefficient was estimated to be about 5700 L·mol-1·cm-1 according to Figure 1b. This means that the 1O2 generation efficiency is inevitably compromised.

(a)

(b)

N+ I -

NH N O

N+

O

I-

N HN

Absorbance

IN+

O

0.9 0.6

0.0

AS1411/TMPipEOPP

1.5

0.9 0.6

0.0

500

600

700

1 5 800

Wavelength (nm) +

AS1411+K +TMPipEOPP +

AS1411+DNase I+K +TMPipEOPP

1.2

+

1.2

0.3

400

(d)

TMPipEOPP

Blank

1.8

2

4 3

0.3

+ I- N

(c)

Free TMPipEOPP (1) C-MYC AT (2) AS1411 LD (3) Hum24 GC (4) KRAS CtDNA (5) Oxy28 ssDNA

1.2

O

Absorbance

absorption band in NIR region (~700 nm). The absorption intensity of the new band was much stronger than that of Qband absorption of free porphyrin. This observation suggested that G-quadruplex/porphyrin complex might be used as a new kind of photosensitizers which may response to NIR light. Inspired by such an observation, we herein reported a simple strategy to confer porphyrin-based photosensitizers with NIRresponse, great 1O2 generation efficiency and tumor-targeted specificity. In the proposed strategy, the new kind of composite photosensitizers was prepared by simply mixing of cationic porphyrin derivatives and G-quadruplexes. Compared to the traditional porphyrin photosensitizer that in response to 650 nm light, the novel G-quadruplex/porphyrin composite photosensitizer could give response to ~700 nm excitation light. And the efficiency of the 1O2 generation based on such an excitation is much higher. In addition, this composite photosensitizer could be easily assembled with some reported nano-drug carriers (e.g., graphene oxide (GO) or MnO2 nanosheet)36-38 to overcome its limitations and gain extra charming properties. Here, we have demonstrated that the assembly of the proposed photosensitizer with MnO2 nanosheet could further improve the tumor PDT efficacy (Scheme 1).

A417nm

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 10

650 nm 690 nm 0

1

2

AS1411+K +DNase I+TMPipEOPP

0.9 0.6 0.3 0.0

3

4

5

6

7

Time ( min)

8

9 10

+

AS1411+K +TMPipEOPP+DNase I

400

500

600

700

800

Wavelength (nm)

Figure 1. (a) Chemical structure of TMPipEOPP; (b) UV-Vis absorption spectra of TMPipEOPP and its mixture with differently structural DNAs. Free TMPipEOPP (black line); single-stranded DNA (green line); double-stranded DNA (blue line); G-quadruplex DNA (red line). (c) Time-dependent changes in DPBF absorbance at 417 nm in the absence and presence of TMPipEOPP or AS1411/TMPipEOPP complex upon irradiation with 650 nm or 690 nm laser. (d) Absorption spectra of the mixture of AS1411 and TMPipEOPP after treatment with DNase I by different ways. The order of reagent addition was shown in the figure. In (b) and (d), [TMPipEOPP] = [DNA] = 5 μM. In (c), [TMPipEOPP] = [DNA] = 50 nM. As shown in Figure 1b, single-stranded and double-stranded DNAs showed few effects on the TMPipEOPP absorption spectrum. When mixed with DNA G-quadruplexes, however, a new absorption band appeared centered at around 700 nm. More interestingly, the intensity of this new absorption was much higher than that of the Q-band absorption of free TMPipEOPP. For example, the complex formed by TMPipEOPP and G-quadruplex AS1411 gave a molar absorption coefficient of ~47000 L·mol-1·cm-1 at 700 nm, which was 7.4 times higher than that of free TMPipEOPP at 650 nm. Therefore, it is reasonable to assume that Gquadruplex/TMPipEOPP complex might be developed as a new kind of composite photosensitizers that can shift the excitation light from the margin to the middle of NIR window and show improved 1O2 generation efficiency due to enhanced light absorption ability. To verify this hypothesis, we investigated the effects of five G-quadruplex DNAs, including parallel (C-MYC, AS1411 and KRAS), anti-parallel (Oxy28) and hybrid type (Hum24), on TMPipEOPP absorption spectrum. All of the employed Gquadruplexes could bind to TMPipEOPP to form complexes with enhanced NIR absorption, and parallel G-quadruplexes showed stronger absorption than anti-parallel and hybrid ones. We selected AS1411/TMPipEOPP complex for subsequent composite photosensitizer studies based on two reasons: first, this complex shows the strongest NIR absorption, and second, the AS1411 is an aptamer of nucleolin, kind of cancer biomarker over expressed in numerous cancer cells. Job plot

ACS Paragon Plus Environment

analysis suggested that TMPipEOPP bound to AS1411 with a stoichiometry of 1:1 (Figure S1). Scatchard analysis confirmed this binding stoichiometry and showed that the binding constant (Ka) was (1.97±0.52) × 107 M−1 (Figure S2), thus demonstrating the formation of a stable complex. Then, light-induced 1O2 generation capacity of the proposed AS1411/TMPipEOPP composite photosensitizer was measured and compared with free TMPipEOPP by using 1,3diphenylisobenzofuran (DPBF) as the 1O2 indicator. As shown in Figure 1c, irradiation time-dependent decrease in DPBF absorption at 417 nm was observed upon irradiation of free TMPipEOPP with 650 nm laser, confirming the light-induced 1O generation by TMPipEOPP. On the contrary, irradiation 2 with 690 nm laser led to no changes in DPBF absorption signal, which was consistent with above observation that free TMPipEOPP could not absorb light with 690 nm wavelength. Notably, the AS1411/TMPipEOPP complex showed much higher 1O2 generation efficiency than free TMPipEOPP when either 650 or 690 nm laser was used. What's more, the AS1411/TMPipEOPP complex could lead to about 50% decrease of DPBF absorbance as short as 2 min upon 690 nm laser irradiation, and free TMPipEOPP with same concentration could only lead to about 43% DPBF absorbance decrease even after 10 min of irradiation with 650 nm laser. Besides the strong absorption at 700 nm, Gquadruplex/TMPipEOPP complex also showed a much higher absorption than free TMPipEOPP at 650 nm. Taken AS1411/TMPipEOPP complex as an example, the molar absorption coefficient is about 14700 L·mol-1·cm-1 at 650 nm, 3.6 times that of free TMPipEOPP. Correspondingly, this composite photosensitizer also showed obviously higher 1O2 generation capacity than free TMPipEOPP under the irradiation of 650 nm laser. In addition, similar 1O2 generation patterns were also observed for C-MYC/TMPipEOPP, demonstrated the proposed strategy is versatile to construct Gquadruplex/porphyrin photosensitizers for general applications. Biostability is an important character for photosensitizer. It is well known that DNA is susceptible to degrade by nucleases that are ubiquitous in physiological environments. As shown in Figure 1d, when AS1411 was treated with deoxyribonuclease I (DNase I), which is responsible for more than 90% of deoxynuclease activity in human plasma,40-41 for 2 h before mixing with TMPipEOPP, almost no absorption could be observed at 700 nm for the mixture of AS1411 and TMPipEOPP, implying that the DNA oligonucleotide has been degraded completely. When AS1411 folded into G-quadruplex in the presence of K+ before and then incubated with DNase I, a weak 700 nm light absorption could also be observed after addition of TMPipEOPP, indicating that G-quadruplex could partially resist nuclease degradation. As for the AS1411/TMPipEOPP complex, however, the absorption spectrum was almost unaffected by DNase I. This result suggested that the binding of TMPipEOPP could protect AS1411 from enzymolysis, thus endowing the complex with high biostability. The complex was so stable that the absorption spectrum was almost unchanged even after incubation with DNase I for 10 h (Figure S3). Herein, 100 U/mL DNase was used. This concentration was much higher than that in human serum (0.36±0.20 U/mL),42 therefore, the composite photosensitizer might keep stable in physiological environment and hold great potential for in vivo applications.

In vitro PDT evaluation. At first, confocal laser scanning microscopy (CLSM) analysis was used to compare the cell internalization efficiencies of TMPipEOPP, AS1411/TMPipEOPP and C-MYC/TMPipEOPP complex. As shown in Figure 2, TMPipEOPP-treated HeLa cells (human epithelial carcinoma cells) showed brighter fluorescence upon excitation at 405 nm (the excitation wavelength of free TMPipEOPP) than that at 458 nm (an appropriate excitation wavelength of G-quadruplex/TMPipEOPP complexes). In contrast, AS1411/TMPipEOPP complex showed a reverse result for the cell imaging. The 458 nm excitation initiated much brighter fluorescence than 405 nm excitation, indicating that AS1411/TMPipEOPP complex kept intact in cancer cells. Moreover, these results suggested that both TMPipEOPP and AS1411/TMPipEOPP complex could efficiently get into cancer cells. However, by comparing the fluorescence intensities of TMPipEOPP and AS1411/TMPipEOPP complex, it can be found that the cancer cell internalization efficiency of AS1411/TMPipEOPP complex seems was higher than free TMPipEOPP itself. In fact, although TMPipEOPP can be internalized into cancer cells, it is not suitable for in vivo application because the positively charge of the compound will cause cytotoxicity due to the inevitable interactions with the negatively charged biocomponents,38 thus increased the tendency of phagocytosis by the macrophage.43-44 After complexing with DNA G-quadruplex, the composite structure was a negatively charged complex (the Zeta potential of AS1411/TMPipEOPP is -5.2±0.3 mV), thus increasing the possibility of in vivo applications. Interestingly, the two investigated G-quadruplex/TMPipEOPP complexes showed distinctly different fluorescence intensities in cancer cells. As shown in Figure 2, AS1411/TMPipEOPP performed much brighter fluorescence than C-MYC/TMPipEOPP. After excluding the possibility of different fluorescence efficiencies (Figure S4), it can be attributed to the contribution of AS1411 aptamer. The recognition of this aptamer to the nucleolin, a protein overexpressed on cancer cell surface specially, could promote the accumulation of AS1411/TMPipEOPP towards cancer cells. Then, via the synergy of endocytosis and AS1411-induced microphinocytosis pathways,45-47 AS1411/TMPipEOPP complex could be efficiently internalized in cancer cells.

(a)

Bright field

405 nm

(b)

458 nm

Bright field

TMPipEOPP

TMPipEOPP

AS1411/ TMPipEOPP

AS1411/ TMPipEOPP

C-MYC/ TMPipEOPP

C-MYC/ TMPipEOPP

(c) 100 80

60 TMPipEOPP 40

AS1411/ TMPipEOPP

650 nm

Bright field

690 nm

(d)100

650 nm Laser 690 nm Laser C-MYC/ TMPipEOPP

20 0

Photosensitizer

Cell viability (%)

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

Cell viability (%)

Page 3 of 10

80 60 40 20 0

0.05 0.25 0.5 1 2.5 5 Photosensitizer concentraion (M)

Figure 2. In vitro PDT evaluation of Gquadruplex/TMPipEOPP composite photosensitizers. (a) CLSM images of HeLa cells incubated with TMPipEOPP, AS1411/TMPipEOPP or C-MYC/TMPipEOPP complex. (b) 1O 2 generation in HeLa cells treated with TMPipEOPP,

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

(a) 180 150

AS1411T33/TMPipEOPP MnO2@AS1411T33/TMPipEOPP MnO2@AS1411T33/TMPipEOPP+GSH

120 90 60 30 0 600

650

700

750

(b) Intensity (%)

2 generation in living cells was tested by using 2',7'dichlorodihydrofluorescein diacetate (DCFH-DA) as the fluorescent 1O2 probe. HeLa cells were preincubated with different photosensitizers for 2 h and then exposed under 650 nm or 690 nm laser for 5 min. In such situation, the 1O2 produced by the irradiation of the porphyrin would oxidize the DCFH-DA to DCF, thus generating fluorescent signal. As expected, cells treated with only TMPipEOPP showed brighter fluorescence of DCF upon 650 nm irradiation than that upon 690 nm irradiation. On the contrast, much brighter DCF fluorescence was given by AS1411/TMPipEOPP-treated cells after irradiation with 690 nm laser compared to the 650 nm one. HeLa cells treated with AS1411/TMPipEOPP complex and 690 nm laser showed the brightest fluorescence signal, suggesting the best in vitro PDT efficacy, which might be attributed to the synergetic contributions of high cancer cell internalization of the composite photosensitizer and improved 1O generation efficiency upon 690 nm irradiation. 2 In vitro PDT efficacies of the composite photosensitizers were also evaluated through comparing with those of free TMPipEOPP. After incubation with TMPipEOPP, AS1411/TMPipEOPP or C-MYC/TMPipEOPP (equivalent concentration of TMPipEOPP: 0.5 μΜ) for 3 h, HeLa cells were irradiated by 650 nm or 690 nm laser for 3.5 min, followed by incubation with fresh culture medium for 24 h. Then, the cell viability was measured by standard MTT (3(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. Except that free TMPipEOPP showed higher PDT efficacy upon 650 nm irradiation, both AS1411/TMPipEOPP and C-MYC/TMPipEOPP gave better PDT efficacies upon 690 nm irradiation. As expected, treatment with AS1411/TMPipEOPP and 690 nm laser gave the best PDT outcome, whose cell viability (21.1%) was much lower than that given by TMPipEOPP and 650 nm laser-treated group (47.5%). Without irradiation treatment, no discernible decrease in viability was observed for HeLa cells treated with TMPipEOPP, AS1411/TMPipEOPP or C-MYC/TMPipEOPP, indicating that both free TMPipEOPP and composite photosensitizers are biocompatible with no inherent toxicity. Assembly of composite photosensitizer with drug carriers. Among the components of the composite photosensitizers, Gquadruplexes are formed by G-rich DNA sequences. Various attractive properties and easy-to-modification merit of DNA make the assembly of the composite photosensitizers with reported drug carriers via simply operating ways possible. Such an assembly might overcome the shortcomings of the composite photosensitizers and endow the composite photosensitizers with some charming properties, thus improving PDT efficacy and promoting their clinical applications. As examples, the assembly of the composite photosensitizer AS1411/TMPipEOPP with GO and MnO2 nanosheet was investigated. Absolutely different results were given by these

25

MnO2 MnO2@AS1411T33/TMPipEOPP

20 15 10 5 0 10

800

100

Size (d. nm)

Wavelength (nm)

(c)

0 -3

(d)2.0

-6 -9 -12 -15

AS1411T33/TMPipEOPP MnO2 MnO2@AS1411T33/TMPipEOPP

Absorbance

1O

two drug carriers. The assembly with GO significantly hampered the PDT efficacy of the composite photosensitizer. However, the assembly with MnO2 nanosheet showed greatly improved PDT efficacy. The details of GO@AS1411T33/TMPipEOPP assembly can be found in supporting information file (Figure S5). The potential use of MnO2@AS1411T33/TMPipEOPP assembly as a novel composite photosensitizer was investigated in detail in the following sections. To achieve the facile assembly of the AS1411/TMPipEOPP complex with MnO2 nanosheet, a poly(T) sequence with 33 deoxythymidine nucleotides was added to the 3′-end of AS1411. Via strong physisorption between single-stranded poly(T) nucleobases and MnO2 nanosheet,48 the DNA oligonucleotide of AS1411T33 could be facilely assembled on the MnO2 nanosheet surface. Through this simple way, the MnO2@AS1411T33/TMPipEOPP assembly could be prepared. The successful assembly between AS1411T33/TMPipEOPP and MnO2 nanosheet was verified by several characterization techniques including UV-Vis absorption, fluorescence, dynamic light scattering (DLS) and Zeta potential analysis (Figure 3). After mixing with MnO2 nanosheet, the fluorescence of the AS1411T33/TMPipEOPP complex was significantly quenched, indicating that the composite photosensitizer was successfully loaded on MnO2 nanosheets. Such a fluorescence change was insensitive to pH (Figure S6), an important physiological parameter,49,50 demonstrating that the prepared MnO2@AS1411T33/TMPipEOPP assembly was stable in a wide pH range (e.g. 5.5-8.0). Such an assembly increased the average hydrodynamic diameter of MnO2 nanosheet from 218 nm to 230 nm. Correspondingly, the Zeta potential was changed from -5.2±0.3 mV for AS1411T33/TMPipEOPP, -11.4±0.3 mV for MnO2 to 12.4±0.5 mV for MnO2@AS1411T33/TMPipEOPP. The negatively charged property could make the assembly keep stable in physiological environments and thus successfully reach the target sites. By comparing Meaningfully, relatively strong absorption band centered at 700 nm could also be observed for the MnO2@AS1411T33/TMPipEOPP assembly, suggesting that the AS1411T33/TMPipEOPP complex was not destroyed by MnO2, and the assembly still has the potential to be used as NIR light-responsive PDT photosensitizer.

Fluorescence (a.u.)

AS1411/TMPipEOPP or C-MYC/TMPipEOPP followed by irradiation with 650 nm or 690 nm laser. (c) In vitro phototoxicity of TMPipEOPP, AS1411/TMPipEOPP and CMYC/TMPipEOPP upon 650 nm or 690 nm irradiation. (d) Dark cytotoxicity of (Black) TMPipEOPP, (Red) AS1411/TMPipEOPP and (Green) C-MYC/TMPipEOPP. In (a~c), equivalent TMPipEOPP concentration of 0.5 μM was used.

Zeta potential (mV)

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 10

1.6

1000

MnO2 AS1411T33/TMPipEOPP MnO2@AS1411T33/TMPipEOPP

1.2 0.8 0.4 0.0

400

500

600

700

800

Wavelength ( nm)

Figure 3. (a) Fluorescence, (b) DLS, (c) Zeta potential and (d) UV-Vis absorption spectral characterization of the assembly between AS1411T33/TMPipEOPP and MnO2 nanosheet.

ACS Paragon Plus Environment

more suitable for tumor microenvironments than AS1411/TMPipEOPP, thus holding huge promise in PDT treatment of solid tumors. CLSM images of HeLa cells incubated with MnO2@AS1411T33/TMPipEOPP showed bright red fluorescence (Figure 4b), demonstrating that MnO2@AS1411T33/TMPipEOPP could be efficiently internalized into cancer cells and AS1411T33/TMPipEOPP would be released from MnO2 nanosheet in cells, resulting in the recovery of the MnO2-quenched fluorescence (Figure 3a). Different from MnO2@TMPipEOPP that showed brighter free TMPipEOPP fluorescence, MnO2@AS1411T33/TMPipEOPP

(a)1.5 AS1411/TMPipEOPP

MnO2@AS1411T33 /TMPipEOPP

Control GSH H2O2

Control GSH H2O2

1.2 0.9

(b)

Bright field 405 nm

458 nm

TMPipEOPP AS1411/ TMPipEOPP

0.6 MnO2@ AS1411T33/ TMPipEOPP

0.3 0.0

0

(c)

3

6

9

0

3

Time ( min)

Bright field 650 nm

6

9

Bright field 690 nm

(d)100

MnO2@ TMPipEOPP

60 40 20 0

(e)

Bright field 650 nm Bright field 690 nm

MnO2@ AS1411T33/ TMPipEOPP

Cell viability (%)

(f)100 AS1411/ TMPipEOPP

650 nm Laser 690 nm Laser

3/

MnO2@ AS1411T33/ TMPipEOPP

80

3 1T 41 S1 @A O PP E O2 Mn MPip T

Cell viability (%)

AS1411/ TMPipEOPP

@ O2 P Mn EOP Pip TM

In vitro PDT evaluation of MnO2@AS1411T33/TMPipEOPP assembly. Using DPBF as 1O 1O 2 probe, the 2 generation abilities of the composite photosensitizer before and after assembling with MnO2 nanosheet were compared. As shown in Figure 4a, MnO2@AS1411T33/TMPipEOPP assembly showed an obviously decreased 1O2 generation efficiency than AS1411/TMPipEOPP. This result indicated that MnO2 nanosheet could inhibit 1O2 generation, thus conferring the assembly with low phototoxicity during circulation.51 After being internalized into cancer cells, MnO2 nanosheet might be rapidly degraded via the synergy of several ways including reduction by overexpressed glutathione (GSH) and reaction with endogenous H2O2 in acidic organelles (e.g., lysosomes and endosomes), thus resulting a burst release of the composite photosensitizers in cancer cells (Figure S7). GSH is a well-known 1O2 scavenger.52 The intracellular GSH level (0.5~10 mM) is about 200-fold higher than that in plasma (2~20 μM), and cancer cells have about 4 times GSH level enhancement compared to healthy cells.53 The overexpression of GSH in cancer cells becomes an inevitable factor that inhibits the PDT efficiency. As shown in Figure 4a, in the presence of 0.45 mM GSH, the signal change of DPBF probe was greatly inhibited in 690 nm laser-irradiated AS1411/TMPipEOPP system, confirming the consumption of 1O by GSH. In contrast, addition of GSH enlarged the DPBF 2 signal change in 690 nm laser-irradiated MnO2@AS1411T33/TMPipEOPP system due to the reaction between GSH and MnO2 nanosheet. On one hand, such a reaction caused the degradation of MnO2 nanosheet and thus the release of AS1411T33/TMPipEOPP composite photosensitizer (Figure S7). As a result, inhibition of 1O2 generation by MnO2 nanosheet would be overcome. On the other hand, such a reaction converted active GSH to inactive glutathione disulfide (GSSG),24,47,54-56 subsequently relieve the consumption of 1O2 by GSH. Notably, Since the GSH level in plasma is much lower than that in cells and MnO2@AS1411T33/TMPipEOPP has high resistance to enzymolysis (Figure S3), the assembly can keep stable in transmission paths, and be successfully transported to targets sites. Increase in H2O2 expression level is another important feature of cancer cells.24, 57 The overexpressed H2O2 can also result in the degradation of MnO2 nanosheet, accompanied by decomposition of H2O2 to O2, one of the three key elements of PDT. The increase of O2 consumption caused by rapid proliferation of cancer cells and inadequate O2 supplement due to abnormal and dysfunctional tumor blood vessels make solid tumors present a hypoxic environment. Since PDT treatment is highly dependent on O2, such a hypoxic feature has greatly restricted the development of tumor PDT. Reaction between MnO2 and endogenous H2O2 might provide a way for in situ generation of O2 in cancer cells, thus overcoming the shortcoming of hypoxia and improving the PDT outcome in some degree. Addition of H2O2 improved the 1O2 generation by MnO2@AS1411T33/TMPipEOPP to a comparable level to that by AS1411/TMPipEOPP, but gave ignorable effect on AS1411/TMPipEOPP. Since the experiments were conducted in an open-air but not hypoxic condition, the O2 supplement by H2O2 decomposition showed no obvious effect, but H2O2triggered MnO2 nanosheet degradation could result in the release of AS1411T33/TMPipEOPP composite photosensitizer. Collectively, the MnO2@AS1411T33/TMPipEOPP might be

/ 11 P 14 P AS pEO Pi TM

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

A417nm

Page 5 of 10

Photosensitizer 650 nm Laser 690 nm Laser

80 60 40 20 0

AS1411/ TMPipEOPP

MnO2@AS1411T33/ TMPipEOPP

1O

Figure 4. (a) 2 generation by AS1411/TMPipEOPP or MnO2@AS1411T33/TMPipEOPP in the absence or presence of 0.45 mM GSH or 3 mM H2O2. DPBF was used as 1O2 probe and its absorption signal change at 417 nm was monitored upon the irradiation of 690 nm laser. (b) CLSM images of HeLa cells incubated with MnO2@TMPipEOPP, AS1411/TMPipEOPP or MnO2@AS1411T33/TMPipEOPP. (c) 1O generation in HeLa cells treated with 2 MnO2@TMPipEOPP, AS1411/TMPipEOPP or MnO2@AS1411T33/TMPipEOPP followed by irradiation with 650 nm or 690 nm laser. (d) In vitro phototoxicity of MnO2@TMPipEOPP, AS1411/TMPipEOPP and MnO2@AS1411T33/TMPipEOPP upon 650 nm or 690 nm irradiation. (e) 1O2 generation in HeLa cells treated with AS1411/TMPipEOPP or MnO2@AS1411T33/TMPipEOPP under hypoxic condition followed by irradiation with 650 nm or 690 nm laser. (f) In vitro phototoxicity of AS1411/TMPipEOPP and MnO2@AS1411T33/TMPipEOPP under hypoxic condition. Equivalent TMPipEOPP concentration of 0.5 μM was used in these experiments. gave higher fluorescence intensity under the excitation at 458 nm than under 405 nm excitation, suggesting that AS1411T33/TMPipEOPP complex but not free TMPipEOPP was released. The released AS1411T33/TMPipEOPP could then be used as composite photosensitizer to produce 1O2 with

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

In vivo PDT treatment of the tumor model. For in vivo PDT applications, HeLa cells were xenografted into the flank region on the dorsum of female BALB/c-nu mice (7–8 weeks) to prepare tumor-bearing mouse models. When the tumor volume reached about 60 mm3, 18 mice were randomly divided into six groups and treated in different ways. 1) saline; 2) AS1411/TMPipEOPP; 3) MnO2@AS1411T33/TMPipEOPP; 4) saline+NIR; 5) AS1411/TMPipEOPP+NIR; 6) MnO2@AS1411T33/TMPipEOPP+NIR (Figure 5). At approximately 4 h after intravenous injection, the tumor sites were irradiated with a 690 nm laser (100 mW/cm2). Irradiation was performed for three times at an interval of 5 min, and each time was lasted for 5 min. The tumor sizes and body weights of the mice were measured every day. The tumor growth profiles showed that the tumor growth was greatly inhibited in AS1411/TMPipEOPP+NIR and MnO2@AS1411T33/TMPipEOPP+NIR groups, demonstrating the feasibility of G-quadruplex/TMPipEOPP as NIRresponsive photosensitizer for tumor PDT. MnO2@AS1411T33/TMPipEOPP gave a better PDT outcome than AS1411/TMPipEOPP, confirming the positive effects of MnO2 on PDT of solid tumors. In MnO2@AS1411T33/TMPipEOPP+NIR-treated group, not only the proliferation of tumor was completely inhibited, but sufficient tumor ablation was also achieved. After 9 days therapy, the tumor size was sharply decreased to about 30% of its previous value. For comparison, fast increase in tumor volume was observed for other four groups. In addition, there was no noticeable difference in mouse body weight and the weights of main organs (heart, liver, spleen, lung, kidney and stomach) among the six groups, indicating that both the composite photosensitizer and its assembly with MnO2 nanosheets are biocompatible.

0.12 0.09 0.06 0.03 0.00

1

2

3

4

5

Group

6

2 1 0

(e) Organ weight (g)

22 20 18 16

Group 1 Group 2 Group 3

14 12

Group 4 Group 5 Group 6

1.2 0.9 0.6

1

2

ACS Paragon Plus Environment

3

4

5

6

Time (days)

7

8

9

0

1

2

3

4

5

6

7

8

9

Time ( days) Group 1 Group 2 Group 3 Group 4 Group 5 Group 6

0.3 0.0

0

Group 1 Group 2 Group 3 Group 4 Group 5 Group 6

3

1.5

(d)24

10

Relative tumor volume (V/V0)

(c) 4

(b) 0.15 Tumor weight (g)

high efficiency upon 690 nm laser excitation. In addition, due to the reduced 1O2 scavenging effect caused by MnO2 degradation-induced GSH consumption, MnO2@AS1411T33/TMPipEOPP-treated HeLa cells showed brighter DCF fluorescence than AS1411/TMPipEOPP-treated ones. Correspondingly, MnO2@AS1411T33/TMPipEOPP showed obviously improved in vitro PDT efficiency compared to AS1411/TMPipEOPP. Using 690 nm laser as the excitation light source, the viability of HeLa cells was greatly decreased from 21.1% to 5.8% when the photosensitizer was changed from AS1411/TMPipEOPP complex to MnO2@AS1411T33/TMPipEOPP assembly. Because manganese is a necessary biological element, MnO2 nanosheet is relatively safe nanomaterial with good biocompatibility. Correspondingly, no dark cytotoxicity was observed for MnO2@AS1411T33/TMPipEOPP when its concentration was lower than 100 μg/mL (equivalent concentration of TMPipEOPP: 2.5 μΜ) (Figure S8), which was higher than that used in PDT treatment of cancer cells (0.5 μΜ TMPipEOPP). As mentioned above, the reaction of MnO2 with H2O2 can relieve tumor hypoxia by in situ producing O2. To demonstrate this, above cellular experiments were conducted in an inclosed plastic glove, from which air was completely extruded to mimic the hypoxic environment. In such a hypoxic environment, 1O2 generation was greatly inhibited in AS1411/TMPipEOPP-treated cells. On the contrary, bright DCF fluorescence could still be observed in MnO2@AS1411T33/TMPipEOPP-treated ones though the fluorescence intensity was also lower than that given in aerobic condition. By carefully examining Figure 4c and 4e, it could be found that the intensity deference of DCF fluorescence between AS1411/TMPipEOPP and MnO2@AS1411T33/TMPipEOPP-treated cells was amplified in hypoxic environment compared to that in aerobic environment, suggesting the enhanced DCF fluorescence given by MnO2@AS1411T33/TMPipEOPP in hypoxic environment was not attributed to GSH consumption alone. Correspondingly, in hypoxic environment, both AS1411/TMPipEOPP and MnO2@AS1411T33/TMPipEOPPinduced PDT killing of HeLa cells were greatly inhibited, but the cell viability of MnO2@AS1411T33/TMPipEOPP-treated group was obviously lower than AS1411/TMPipEOPP-treated one, suggesting that MnO2@AS1411T33/TMPipEOPP might also work in hypoxic tumor environment. These results demonstrated that MnO2-mediated in situ generation of O2 indeed is able to overcome the adverse effect of tumor hypoxia on PDT in some degree. It was demonstrated that MnO2 nanosheet could also successfully assemble with other G-quadruplex/TMPipEOPP composite photosensitizers (e.g., C-MYCT33/TMPipEOPP), and give positive effects on PDT efficacy (Figure S9). We also tried to assemble AS1411T33/TMPipEOPP and CMYCT33/TMPipEOPP with GO, a widely used drug carrier. However, GO gave a negative effect on the PDT applications of the composite photosensitizers (Figure S5). Different from the burst decomposition of MnO2, GO is stable in cancer cells. As a result, AS1411T33/TMPipEOPP (or CMYCT33/TMPipEOPP) cannot be released from the GO@AS1411T33/TMPipEOPP (or GO@CMYCT33/TMPipEOPP) assembly, and the generation of 1O2 was greatly inhibited.

Body weight (g)

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 6 of 10

Heart Liver Spleen Lung Kidney Stomach

Page 7 of 10 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 5. In vivo PDT of tumor-bearing mice. (a) Representative photos of mice before and after different treatments. (b) Photos of the tumors dissected from the mice and their weights after 9 days treatment. (c) Tumor growth profiles of different groups; (d) Mouse body weight changes after different treatments; (e) Organ weight in various groups after 9 days treatment. Groups 1 → 6: saline; AS1411/TMPipEOPP; MnO2@AS1411T33/TMPipEOPP; saline+NIR; AS1411/TMPipEOPP+NIR; MnO2@AS1411T33/TMPipEOPP+NIR. Histological analysis of dissected solid tumors and main organs showed that obvious cell apoptosis and fragmentation of the tumor tissues were only observed for AS1411/TMPipEOPP+NIR and MnO2@AS1411T33/TMPipEOPP+NIR groups (Figure 6), indicating that the tumors were severely damaged in these two groups after PDT treatment. On the contrary, almost no necrosis or apoptosis in the tumor tissues was shown by other four groups. In addition, in all groups, no perceptible pathological changes were observed for the main organs of mice, suggesting that the composite photosensitizer and its assembly with MnO2 has negligible side effects on normal tissues and organs, and is highly safe for in vivo antitumor application.

vivo experiments, thus showing huge potential for clinical PDT treatment of solid tumors.

ASSOCIATED CONTENT Supporting Information The Supporting Information include: Experimental details, Figure S1-S9, and Table S1.

AUTHOR INFORMATION Corresponding Author * Corresponding to: L.-N. Zhu, Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, China, E-mail address: [email protected] (L.-N. Zhu) * Corresponding to: D.-M. Kong, College of Chemistry, Nankai University, Tianjin, 300071, China, E-mail address: [email protected] (D.-M. Kong)

ACKNOWLEDGMENT This work was supported by the National Natural Science Foundation of China [no. 21371130], the Natural Science Foundation of Tianjin City [no. 15JCYBJC48300, 16JCYBJC19900], and the Innovation Fund of Tianjin University.

REFERENCES

Figure 6. Hematoxylin and eosin (H&E) images of six main organs and tumor tissues collected from mice after different treatments. Groups 1-6 are identical to those in Figure 5.

CONCLUSION In summary, we proposed a facile way to improve the penetration depth and PDT efficacy of photosensitizers. We designed a new kind of composite photosensitizers by simply mixing G-quadruplexes and water soluble cationic porphyrin TMPipEOPP. Compared to the conventional porphyrin based photosensitizer, the new composite photosensitizers show the following advantages: 1) Red-shifted excitation light wavelength (from 650 to 700 nm); 2) Enhanced NIR absorption (7.4-fold increase of molar absorption coefficient) and thus improved 1O2 generation efficiency; 3) Extra charming properties provided by nucleic acids such as ease-tomodification and facile assembly with other drug carriers. As an example, we demonstrated that the composite photosensitizers could be successfully assembled with MnO2 nanosheet. The assembly integrated the advantages of the composite photosensitizer and MnO2 nanosheet (reduction of 1O consumption by GSH and in situ supplement of O ), and 2 2 showed greatly improved PDT efficacy for both in vitro and in

(1) Teo, R. D.; Hwang, J. Y.; Termini, J.; Gross, Z.; Gray, H. B., Fighting Cancer with Corroles. Chem. Rev. 2017, 117, 2711-2729. (2) Fan, W.; Yung, B.; Huang, P.; Chen, X., Nanotechnology for Multimodal Synergistic Cancer Therapy. Chem. Rev. 2017, 117, 13566-13638. (3) Ulbrich, K.; Holá, K.; Šubr, V.; Bakandritsos, A.; Tuček, J.; Zbořil, R., Targeted Drug Delivery with Polymers and Magnetic Nanoparticles: Covalent and Noncovalent Approaches, Release Control, and Clinical Studies. Chem. Rev. 2016, 116, 5338-5431. (4) Cheng, L.; Wang, C.; Feng, L.; Yang, K.; Liu, Z., Functional Nanomaterials for Phototherapies of Cancer. Chem. Rev. 2014, 114, 10869-10939. (5) Liu, Y.; Hou, W.; Xia, L.; Cui, C.; Wan, S.; Jiang, Y.; Yang, Y.; Wu, Q.; Qiu, L.; Tan, W., ZrMOF Nanoparticles as Quenchers to Conjugate DNA Aptamers for Target-Induced Bioimaging and Photo Dynamic Therapy. Chem. Sci. 2018, 9, 7505-7509. (6) Lan, G.; Ni, K.; Xu, Z.; Veroneau, S. S.; Song, Y.; Lin, W., Nanoscale Metal–Organic Framework Overcomes Hypoxia for Photodynamic Therapy Primed Cancer Immunotherapy. J. Am. Chem. Soc. 2018, 14, 5670-5673. (7) Lu, K.; He, C.; Guo, N.; Chan, C.; Ni, K.; Weichselbaum, R. R.; Lin, W., Chlorin-Based Nanoscale Metal–Organic Framework Systemically Rejects Colorectal Cancers via Synergistic Photodynamic Therapy and Checkpoint Blockade Immunotherapy. J. Am. Chem. Soc. 2016, 138, 12502-12510. (8) Wang, Y.; Huang, X.; Tang, Y.; Zou, J.; Wang, P.; Zhang, Y.; Si, W.; Huang, W.; Dong, X., A Light-Induced Nitric Oxide ControlLable Release Nano-Platform Based on Dike to Pyrrolopyrrole Derivatives for pH-Responsive Photodynamic/Photothermal Synergistic Cancer Therapy. Chem. Sci. 2018, 9, 8103-8109. (9) Duan, X.; Chan, C.; Guo, N.; Han, W.; Weichselbaum, R. R.; Lin, W., Photodynamic Therapy Mediated by Nontoxic Core–Shell Nanoparticles Synergizes with Immune Checkpoint Blockade to Elicit Antitumor Immunity and Antimetastatic Effect on Breast Cancer. J. Am. Chem. Soc. 2016, 138, 16686-16695. (10) Cho, Y.; Choi, Y., Graphene Oxide–Photosensitizer Conjugate as a Redox-Responsive Theranostic Agent. Chem. Commun. 2012, 48, 9912-9914. (11) Monro, S.; Colón, K. L.; Yin, H.; Roque, J.; Konda, P.; Gujar, S.; Thummel, R. P.; Lilge, L.; Cameron, C. G.; McFarland, S. A.,

ACS Paragon Plus Environment

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

Transition Metal Complexes and Photodynamic Therapy from a Tumor-Centered Approach: Challenges, Opportunities, and Highlights from the Development of TLD1433. Chem. Rev. 2019, 119, 797-828. (12) Lucky, S. S.; Soo, K. C.; Zhang, Y., Nanoparticles in Photodynamic Therapy. Chem. Rev. 2015, 115, 1990-2042. (13) Zhang, J.; Jiang, C.; Figueiró Longo, J. P.; Azevedo, R. B.; Zhang, H.; Muehlmann, L. A., An Updated Overview on The Development of New Photosensitizers for Anticancer Photodynamic Therapy. Acta Pharm. Sin. B 2018, 8, 137-146. (14) Abrahamse, H.; Hamblin, Michael R., New Photosensitizers for Photodynamic Therapy. Biochem. J. 2016, 473, 347-364. (15) Li, X.; Lee, S.; Yoon, J., Supramolecular Photosensitizers Rejuvenate Photodynamic Therapy. Chem. Soc. Rev. 2018, 47, 11741188. (16) Kou, J.; Dou, D.; Yang, L., Porphyrin Photosensitizers in Photodynamic Therapy and Its Applications. Oncotarget 2017, 8, 81591-81603. (17) Washington, I.; Brooks, C.; Turro, N. J.; Nakanishi, K., Porphyrins as Photosensitizers To Enhance Night Vision. J. Am. Chem. Soc. 2004, 126, 9892-9893. (18) Maruani, A.; Savoie, H.; Bryden, F.; Caddick, S.; Boyle, R.; Chudasama, V., Site-Selective Multi-Porphyrin Attachment Enables the Formation of a Next-Generation Antibody-Based photodynamic Therapeutic. Chem. Commun. 2015, 51, 15304-15307. (19) Yap, S. Y.; Price, T. W.; Savoie, H.; Boyle, R. W.; Stasiuk, G. J., Selective Radiolabelling with 68Ga under Mild Conditions: a Route Towards a Porphyrin PET/PDT Theranostic Agent. Chem. Commun. 2018, 54, 7952-7954. (20) Hisamatsu, Y.; Umezawa, N.; Yagi, H.; Kato, K.; Higuchi, T., Design and Synthesis of a 4-Aminoquinoline-Based Molecular Tweezer that Recognizes Protoporphyrin IX and Iron(III) Protoporphyrin IX and its Application as a Supramolecular Photosensitizer. Chem. Sci. 2018, 9, 7455-7467. (21) Ethirajan, M.; Chen, Y.; Joshi, P.; Pandey, R. K., The Role of Porphyrin Chemistry in Tumor Imaging and Photodynamic Therapy. Chem. Soc. Rev. 2011, 40, 340-362. (22) Wang, D.; Niu, L.; Qiao, Z.-Y.; Cheng, D.-B.; Wang, J.; Zhong, Y.; Bai, F.; Wang, H.; Fan, H., Synthesis of Self-Assembled Porphyrin Nanoparticle Photosensitizers. ACS Nano 2018, 12, 37963803. (23) Singh, S.; Aggarwal, A.; Bhupathiraju, N. V. S. D. K.; Arianna, G.; Tiwari, K.; Drain, C. M., Glycosylated Porphyrins, Phthalocyanines, and Other Porphyrinoids for Diagnostics and Therapeutics. Chem. Rev. 2015, 115, 10261-10306. (24) Xu, J.; Han, W.; Yang, P.; Jia, T.; Dong, S.; Bi, H.; Gulzar, A.; Yang, D.; Gai, S.; He, F.; Lin, J.; Li, C., Tumor Microenvironment-Responsive Mesoporous MnO2-Coated Upconversion Nanoplatform for Self-Enhanced Tumor Theranostics. Adv. Funct. Mater. 2018, 28, 1803804. (25) Zamiri, B., Reddy, K., Macgregor, R. B., Pearson, C. E. TMPyP4 Porphyrin Distorts RNA G-quadruplex Structures of the Disease-associated r(GGGGCC)n Repeat of the C9orf72 Gene and Blocks Interaction of RNA-binding Proteins. J. Biol. Chem. 2014, 289, 4653-4659. (26) Xiong, Y.-X.; Huang, Z.-S.; Tan, J.-H., Targeting Gquadruplex Nucleic Acids with Heterocyclic Alkaloids and Their Derivatives. Eur. J. Med. Chem. 2015, 97, 538-551. (27) Ma, D.-L.; Che, C.-M.; Yan, S.-C., Platinum(II) Complexes with Dipyridophenazine Ligands as Human Telomerase Inhibitors and Luminescent Probes for G-Quadruplex DNA. J. Am. Chem. Soc. 2009, 131, 1835-1846. (28) Wang, P.; Leung, C.-H.; Ma, D.-L.; Yan, S.-C.; Che, C.-M., Structure-Based Design of Platinum(II) Complexes as c-myc Oncogene Down-Regulators and Luminescent Probes for GQuadruplex DNA. Chem. Eur. J. 2010, 16, 6900-6911. (29) Ma, D.-L.; Zhang, Z.; Wang, M.; Lu, L.; Zhong, H.-J.; Leung, C.-H., Recent Developments in G-Quadruplex Probes. Chem. Biol. 2015, 22, 812-828. (30) Zhang, L.-N.; Zhang, R.; Cui, Y.-X.; Liu, K.-K.; Kong, D.-M.; Li, X.-Z.; Zhu, L.-N., Highly Specific G-Quadruplex Recognition

Page 8 of 10

covering Physiological pH Range by a New Water-Soluble Cationic Porphyrin with Low Self-Aggregation Tendency. Dyes Pigm. 2017, 145, 404-417. (31) Huo, Y.-F.; Zhu, L.-N.; Liu, K.-K.; Zhang, L.-N.; Zhang, R.; Kong, D.-M., Water-Soluble Cationic Metalloporphyrins: Specific GQuadruplex-Stabilizing Ability and Reversible Chirality of Aggregates Induced by AT-Rich DNA. Inorg. Chem. 2017, 56, 63306342. (32) Huang, X.-X.; Zhu, L.-N.; Wu, B.; Huo, Y.-F.; Duan, N.-N.; Kong, D.-M., Two Cationic Porphyrin Isomers Showing Different Multimeric G-Quadruplex Recognition Specificity Against Monomeric G-Quadruplexes. Nucleic Acids Res. 2014, 42, 87198731. (33) Wu, B.; Kong, D.-M.; Zhu, L.-N., Specific Recognition and Stabilization of Monomeric and Multimeric G-Quadruplexes by Cationic Porphyrin TMPipEOPP under Molecular Crowding Conditions. Nucleic Acids Res. 2013, 41, 4324-4335. (34) Zhu, L.-N.; Zhao, S.-J.; Wu, B.; Li, X.-Z.; Kong, D.-M., A New Cationic Porphyrin Derivative (TMPipEOPP) with Large Side Arm Substituents: A Highly Selective G-Quadruplex Optical Probe. PLoS ONE 2012, 7, e35586. (35) Zhang, R.; Cheng, M.; Zhang, L.-M.; Zhu, L.-N.; Kong, D.M., Asymmetric Cationic Porphyrin as a New G-Quadruplex Probe with Wash-Free Cancer-Targeted Imaging Ability Under Acidic Microenvironments. ACS Appl. Mater. Inter. 2018, 10, 13350-13360. (36) Georgakilas, V.; Tiwari, J. N.; Kemp, K. C.; Perman, J. A.; Bourlinos, A. B.; Kim, K. S.; Zboril, R., Noncovalent Functionalization of Graphene and Graphene Oxide for Energy Materials, Biosensing, Catalytic, and Biomedical Applications. Chem. Rev. 2016, 116, 5464-5519. (37) Wang, H.; Zhang, Q.; Chu, X.; Chen, T.; Ge, J.; Yu, R., Graphene Oxide–Peptide Conjugate as an Intracellular Protease Sensor for Caspase-3 Activation Imaging in Live Cells. Angew. Chem. Int. Ed. Engl. 2011, 123, 7203-7207. (38) Fan, W.; Bu, W.; Shen, B.; He, Q.; Cui, Z.; Liu, Y.; Zheng, X.; Zhao, K.; Shi, J., Intelligent MnO2 Nanosheets Anchored with Upconversion Nanoprobes for Concurrent pH-/H2O2-Responsive UCL Imaging and Oxygen-Elevated Synergetic Therapy. Adv. Mater. 2015, 27, 4155-4161. (39) Jiang, Y.; Li, J.; Zhen, X.; Xie, C.; Pu, K., Dual-Peak Absorbing Semiconducting Copolymer Nanoparticles for First and Second Near-Infrared Window Photothermal Therapy: A Comparative Study. Adv. Mater. 2018, 30, 1705980. (40) Zhu, G.; Hu, R.; Zhao, Z.; Chen, Z.; Zhang, X.; Tan, W., Noncanonical Self-Assembly of Multifunctional DNA Nanoflowers for Biomedical Applications. J. Am. Chem. Soc. 2013, 135, 1643816445. (41) Han, G.-M.; Jia, Z.-Z.; Zhu, Y.-J.; Jiao, J.-J.; Kong, D.-M.; Feng, X.-Z., Biostable L-DNA-Templated Aptamer-Silver Nanoclusters for Cell-Type-Specific Imaging at Physiological Temperature. Anal. Chem. 2016, 88, 10800-10804. (42) Zheng, X.; Peng, R.; Jiang, X.; Wang, Y.; Xu, S.; Ke, G.; Fu, T.; Liu, Q.; Huan, S.; Zhang, X., Fluorescence Resonance Energy Transfer-Based DNA Nanoprism with a Split Aptamer for Adenosine Triphosphate Sensing in Living Cells. Anal. Chem. 2017, 89, 1094110947. (43) Sato, Y.; Matsui, H.; Sato, R.; Harashima, H., Neutralization of Negative Charges of siRNA Results in Improved Safety and Efficient Gene Silencing Activity of Lipid Nanoparticles Loaded with High Levels of SiRNA. J. Control. Release 2018, 284, 179-187. (44) Bao, W.; Liu, X.; Lv, Y.; Lu, G.-H.; Li, F.; Zhang, F.; Liu, B.; Li, D.; Wei, W.; Li, Y., Nanolongan with Multiple On-Demand Conversions for Ferroptosis–Apoptosis Combined Anticancer Therapy. ACS Nano 2019, 13, 260-273. (45) Li, J.; Zhong, X.; Cheng, F.; Zhang, J.-R.; Jiang, L.-P.; Zhu, J.-J., One-Pot Synthesis of Aptamer-Functionalized Silver Nanoclusters for Cell-Type-Specific Imaging. Anal. Chem. 2012, 84, 4140-4146. (46) Reyes-Reyes, E. M.; Teng, Y.; Bates, P. J., A New Paradigm for Aptamer Therapeutic AS1411 Action: Uptake by

ACS Paragon Plus Environment

Page 9 of 10 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

Macropinocytosis and its Stimulation by a Nucleolin-Dependent Mechanism. Cancer Res. 2010, 70, 8617-8629. (47) Reyes-Reyes, E. M.; Šalipur, F. R.; Shams, M.; Forsthoefel, M. K.; Bates, P. J., Mechanistic Studies of Anticancer Aptamer AS1411 Reveal a Novel Role for Nucleolin in Regulating Rac1 Activation. Mol. Oncol. 2015, 9, 1392-1405. (48) Chen, F.; Bai, M.; Cao, K.; Zhao, Y.; Wei, J.; Zhao, Y., Fabricating MnO2 Nanozymes as Intracellular Catalytic DNA Circuit Generators for Versatile Imaging of Base-Excision Repair in Living Cells. Adv. Funct. Mater. 2017, 27, 1702748. (49) Li, X.; Gao, X.; Shi, W.; Ma, H., Design Strategies for WaterSoluble Small Molecular Chromogenic and Fluorogenic Probes. Chem. Rev. 2014, 114 , 590-659. (50) Zhou, J.; Ma, H., Design Principles of Spectroscopic Probes for Biological Applications. Chem. Sci. 2016, 7, 6309-6315. (51) Fan, H.; Zhao, Z.; Yan, G.; Zhang, X; Yang, C; Meng, H; Chen, Z; Liu, H and Tan, W, A Smart DNAzyme–MnO2 Nanosystem for Efficient Gene Silencing. Angew. Chem. Int. Ed. Engl. 2015, 54, 4801-4805. (52) To, T.-L.; Fadul, M. J.; Shu, X., Singlet Oxygen Triplet Energy Transfer-Based Imaging Technology for Mapping Protein– Protein Proximity in Intact Cells. Nat. Commun. 2014, 5, 4072.

(53) Li, S.; Zou, Q.; Li, Y.; Yuan, C.; Xing, R.; Yan, X., Smart Peptide-Based Supramolecular Photodynamic Metallo-Nanodrugs Designed by Multicomponent Coordination Self-Assembly. J. Am. Chem. Soc. 2018, 140, 10794-10802. (54) Fan, H.; Yan, G.; Zhao, Z.; Hu, X.; hang, W.; Liu, H.; Fu, X.; Fu, T.; Zhang, X.-B.; Tan, W., A Smart Photosensitizer–Manganese Dioxide Nanosystem for Enhanced Photodynamic Therapy by Reducing Glutathione Levels in Cancer Cells. Angew. Chem. Int. Ed. Engl 2016, 55, 5477-5482 (55) Bi, H.; Dai, Y.; Yang, P.; Xu, J.; Yang, D.; Gai, S.; He, F.; An, G.; Zhong, C.; Lin, J., Glutathione and H2O2 Consumption Promoted Photodynamic and Chemotherapy Based on Biodegradable MnO2–Pt@Au25 Nanosheets. Chem. Eng. J. 2019, 356, 543-553. (56) Wang, H.-B.; Li, Y.; Bai, H.-Y.; Liu, Y.-M., DNA-Templated Au Nanoclusters and MnO2 Sheets: A Label-Free and Universal Fluorescence Biosensing Platform. Sens. Actuators B: Chem. 2018, 259, 204-210. (57) Yang, Y.; Wang, C.; Tian, C.; Guo, H.; Shen, Y.; Zhu, M., Fe3O4@MnO2@PPy Nanocomposites Overcome Hypoxia: MagneticTargeting-Assisted Controlled Chemotherapy and Enhanced Photodynamic/Photothermal Therapy. J. Mater. Chem. B 2018, 6, 6848-6857.

ACS Paragon Plus Environment

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 10

Table of Contents

ACS Paragon Plus Environment

10