You are on page 1of 11

Journal of Colloid and Interface Science 646 (2023) 370–380

Contents lists available at ScienceDirect

Journal of Colloid And Interface Science


journal homepage: www.elsevier.com/locate/jcis

Enhanced lysosomal escape of cell penetrating peptide-functionalized


metal–organic frameworks for co-delivery of survivin siRNA and oridonin
Mengru Cai 1, Yu Yao 1, Dongge Yin , Rongyue Zhu , Tingting Fu , Jiahui Kong , Kaixin Wang ,
Jing Liu , Aina Yao, Yidan Ruan , Wenjuan Shi , Qian Zhu , Jian Ni *, Xingbin Yin *
School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China

G R A P H I C A L A B S T R A C T

A R T I C L E I N F O A B S T R A C T

Keywords: In recent years, small interfering RNA (siRNA) has been widely used in the treatment of human diseases,
Metal organic frameworks especially tumors, and has shown great appeal. However, the clinical application of siRNA faces several chal­
Oridonin lenges. Insufficient efficacy, poor bioavailability, poor stability, and lack of responsiveness to a single therapy are
Survivin siRNA
the main problems affecting tumor therapy. Here, we designed a cell-penetrating peptide (CPP)-modified metal
CPP33
organic framework nanoplatform (named PEG-CPP33@ORI@survivin siRNA@ZIF-90, PEG-CPP33@NPs) for
targeted co-delivery of oridonin (ORI), a natural anti-tumor active ingredient) and survivin siRNA in vivo. This
can improve the stability and bioavailability of siRNA and the efficacy of siRNA monotherapy. The high drug-
loading capacity and pH-sensitive properties of zeolite imidazolides endowed the PEG-CPP33@NPs with lyso­
somal escape abilities. The Polyethylene glycol (PEG)-conjugated CPP (PEG-CPP33) coating significantly
improved the uptake in the PEG-CPP33@NPs in vitro and in vivo. The results showed that the co-delivery of ORI
and survivin siRNA greatly enhanced the anti-tumor effect of PEG-CPP33@NPs, demonstrating the synergistic
effect between ORI and survivin siRNA. In summary, the novel targeted nanobiological platform loaded with ORI
and survivin siRNA presented herein showed great advantages in cancer therapy, and provides an attractive
strategy for the synergistic application of chemotherapy and gene therapy.

* Corresponding authors.
E-mail addresses: 602054@bucm.edu.cn (J. Ni), yxbtcm@bucm.edu.cn (X. Yin).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.jcis.2023.04.126
Received 18 February 2023; Received in revised form 20 April 2023; Accepted 24 April 2023
Available online 28 April 2023
0021-9797/© 2023 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
M. Cai et al. Journal of Colloid And Interface Science 646 (2023) 370–380

1. Introduction [27]. Our group also developed a CPP44-modified MOF delivery system
for efficient drug delivery into HepG2 cells [23]. CPP33
Cancer has a significant impact on human health, and lung cancer is (RLWMRWYSPRTRAYG) can specifically penetrate non-small cell lung
the leading cause of cancer death [1]. Traditional tumor treatment cancer A549 cells, overcoming the barrier of the cell membrane to
methods mainly include surgical resection, chemotherapy, and radio­ improve drug absorption, and has great application potential in the
therapy, although still face great challenges, such as poor targeting and targeted delivery of anti-tumor drugs [28].
drug resistance [2]. In recent years, immunotherapy [3], gene therapy Survivin is a tumor-specific gene in the human genome that is highly
[4], hormone therapy [5], stem cell transplantation therapy [6], and expressed in lung cancer cells. As important inhibitors of apoptosis
tumor-targeted therapy [7] have become the focus of cancer research. At proteins, translated proteins have anti-apoptotic and cell cycle regula­
the core of RNA interference, small interfering RNA (siRNA) has become tory functions [29]. Oridonin (ORI) has received considerable attention
an indispensable tool to verify gene function, and has also been widely as an innovative anticancer drug. ORI induces apoptosis in A549 cells,
used in research on treating human diseases [8]. Many cancer-related and its mechanism may be related to the regulation of Bcl-2/Bax, PARP,
genes can be used as targets of siRNA, which provides unlimited possi­ caspase 3 and survivin signaling pathways [30]. Co-delivery of survivin
bilities for the application of siRNA in tumor therapy. siRNA can knock siRNA with ORI may enhance the anti-tumor effect and achieve better
down the expression of key genes and have the advantages of high efficacy. Here, we first synthesized two ZIFs (ZIF-8 and ZIF-90) with
knockout efficiency, high specificity, and no immunogenicity. Although different hydrophilicities to investigate their different protective effects
siRNA therapy has shown incomparable advantages in anti-tumor ac­ against siRNA, and finally screened ZIF-90 with a better protective effect
tivity, a single therapeutic agent is still insufficient to achieve satisfac­ for subsequent anti-tumor studies. We constructed a functional, pH-
tory anti-tumor efficacy. Currently, one of the most promising responsive, nanodrug delivery platform (named PEG-
combinations for dual delivery is the combination of nucleic acids and CPP33@ORI@survivin siRNA@ZIF-90, PEG-CPP33@NPs) to specif­
small-molecule drugs. The use of siRNA to inhibit the expression of the ically co-deliver ORI with survivin siRNA and enhance the chemo­
target protein at the gene level combined with chemotherapy drugs such sensitivity in vivo (Scheme 1). Specifically, ORI and survivin siRNA were
as doxorubicin (DOX) and cisplatin is an effective strategy to improve non-covalently adsorbed in ZIF-90 (named ORI@survivin siRNA@ZIF-
the sensitivity of chemotherapy [9]. The synergistic effects of genes and 90, NPs) and further coated with polyethylene glycol-CPP (PEG-
chemotherapy can enhance the activity and efficacy of a single drug and CPP33). The obtained PEG-CPP33@NPs had good tumor selectivity and
overcome the problem of insufficient efficacy associated with single pH sensitivity and could be effectively internalized into the cytoplasm.
drugs. Finally, the nanoformulation exhibited excellent gene-silencing effi­
However, the co-delivery of siRNA and chemotherapeutic agents ciency and anti-tumor effects. Our study provides insights into the
brings new problems owing to their easy degradation by nucleases in the combination of chemotherapy and gene therapy, and we expect to
plasma, rapid renal excretion, lack of targeting ability, transmembrane expand this strategy for a wide range of disease treatment applications.
barriers, lysosomal escape barriers, and differences in biodistribution
[10,11]. The use of nanocarriers to achieve the co-delivery of biological 2. Materials and methods
macromolecules and small-molecule drugs is the most attractive means
of overcoming difficulties in the clinical treatment of tumors [12–14]. 2.1. Materials
Metal-organic frameworks (MOFs) have high porosity, adjustable pore
size, and good biocompatibility, giving them great advantages in anti- Imidazole-2-formaldehyde (≥98%), 2-methylimidazole (≥98%), and
tumor synergistic therapy [15–18] and allowing them to encapsulate oridonin (≥98%) were purchased from Shanghai Yuanye Biotechnology
biological macromolecules and small-drug molecules with high drug- Co., Ltd. Zn(NO3)2⋅6H2O (99.99%) was obtained from Shanghai Aladdin
loading capacity. In particular, siRNA encapsulation in the cavity of Bio-Chem Technology Co., Ltd. Zn(CH3COO)2⋅2H2O (98%) was pro­
MOFs can significantly reduce structural changes in biomolecules, pre­ vided by Shanghai Macklin Biochemical Co., Ltd. 3-(4,5-dimethylth­
vent damage and degradation of their structure by the external envi­ iazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT, ≥98%) and
ronment, and increase their active sites [19,20]. Zeolitic imidazolate annexin V-FITC/PI were provided by Beijing Beyerdi Biotechnology Co.,
frameworks (ZIFs) have become a research hotspot in the field of Ltd. DEPC water was obtained from Beijing Rambolide Biotechnology
nanomaterials because of their unique advantages in constructing Co., Ltd. siRNA (antisense sequence: UAAUUCUUCAAACUGCUUCTT,
siRNA-based drug delivery systems, such as mild synthesis conditions, 98%) was purchased from Shanghai Jima Pharmaceutical Technology
good thermal stability, and pH responsiveness [21]. ZIF-8 and ZIF-90 are Co., Ltd. High rich glucose DMEM was purchased from Sigma-Aldrich
the most representative ZIFs [22]. ZIF-90 introduced aldehyde groups LLC. Fetal bovine serum (FBS) and a penicillin–streptomycin mixture
based on ZIF-8; therefore ZIF-90 was more hydrophilic than ZIF-8. The were obtained from Beijing Sijiqing Biotechnology Co., Ltd. N,N-
preparation of siRNA nanoparticles using the two as carriers allows for dimethylformamide (DMF, ≥99.5%) were purchased from Shanghai
the construction of a new RNAi delivery platform, which may be an Aladdin Bio-Chem Technology Co., LTD. Dimethyl sulfoxide (DMSO)
effective strategy to improve the inherent defects of siRNA. However, was obtained from Tianjin Guangfu Fine Chemical Research Institute.
whether ZIF hydrophilicity induces a protective effect against siRNA has
not yet been reported. 2.2. Synthesis of ZIF-8
Surface coatings such as chitosan, hyaluronic acid, and polyethylene
glycol play important roles in nanobiomedicine research [23,24]. Sur­ Zn(NO3)2⋅6H2O was weighed and dissolved in a beaker with deion­
face coatings can provide active sites for the conjugation of targeted ized water. The Zn(NO3)2⋅6H2O solution was poured into a conical flask
ligands, improve the stability and biosafety of nanoparticles, and pre­ containing an aqueous solution of 2-methylimidazole. After continuous
vent recognition and clearance by the immune system [25]. stirring for 15 min, the mixture was transferred into a centrifuge tube,
Nanotechnology-based targeted drug delivery systems present another and the products were collected by centrifugation for 20 min. The
important challenge. Cell-penetrating peptides (CPPs) are a class of products were washed with deionized water and dried in a vacuum-
small-molecule peptides with strong cell membrane penetration abilities drying oven. The prepared product, ZIF-8, was a white solid powder
[26]. CPPs are usually non-toxic, and their ability to target tumor cells that was odorless and was stored in a desiccator.
and transport various drugs (such as enzymes, RNA, and chemical
molecules) through the cell membrane and into the cell has been 2.3. Synthesis of ZIF-90
confirmed. Hade et al. used cell-penetrating peptide (CPPs) to deliver
microRNA (miRNAs) into cells, showing good tumor-homing ability Imidazole-2-formaldehyde was dissolved in DMF, Zn

371
M. Cai et al. Journal of Colloid And Interface Science 646 (2023) 370–380

(CH3COO)2⋅2H2O was added and stirred for 15 min, and the products SDS solution to terminate the reaction. Agarose gel electrophoresis was
were collected by centrifugation for 10 min. The products were washed then performed.
with methanol and dried in a vacuum-drying oven.

2.6. ORI loading


2.4. siRNA loading
ZIF-90 (10 mg) was added to the ORI solution (5 mg/mL, 15 mg) and
The ZIFs (1 mg) were added to the siRNA solution (2.5 μmol/L, 9
magnetically stirred for 24 h. The supernatant was collected by centri­
nmol) and stirred on a magnetic stirrer for 24 h. The supernatant was
fugation and the content of free ORI was determined using high-
collected by centrifugation for determination of free siRNA content by
performance liquid chromatography (HPLC). The precipitate was
gel electrophoresis, and the precipitate was freeze-dried and stored at
lyophilized and stored at − 20 ◦ C to obtain ORI@ZIF-90.
− 20 ◦ C. Subsequently, siRNA@ZIF-8 and siRNA@ZIF-90 were
synthesized.
2.7. Synthesis of ORI@siRNA@ZIF-90 (NPs)
2.5. Protective effect of ZIFs against siRNA
ZIF-90 (1 mg) was stirred with the ORI solution (5 mg/mL) for 24 h.
Free siRNA, siRNA@ZIF-8, and siRNA@ZIF-90 were incubated with After centrifugation, siRNA (9 nmol) was added and the mixture was
100 μL of 10% FBS at 37 ◦ C for 1, 2, 4, 6, 8, 10, and 12 h. At the cor­ stirred in DPEC water for 24 h. The precipitate was lyophilized and
responding time point, 10 μL of sample was added to 5 μL of 10 mg/mL stored at − 20 ◦ C to obtain the NPs.

Scheme 1. Schematic Illustration of the siRNA Protection of ZIF-8 and ZIF-90 and Anti-tumor Mechanism of PEG-CPP33@ORI@Survivin siRNA@ZIF-90.

372
M. Cai et al. Journal of Colloid And Interface Science 646 (2023) 370–380

2.8. Synthesis of PEG-CPP33@NPs injection. The hearts, livers, spleens, lungs, kidneys, and tumors were
collected for imaging using an IVIS imaging system. The data were
ZIF-90 (1 mg) was stirred with ORI solution (5 mg/mL) for 24 h. analyzed using Care Stream software.
After centrifugation, siRNA (9 nmol) was added and the mixture was
stirred in DPEC water for 24 h. After centrifugation and drying, PEG- 2.15. In vivo antitumor effect
CPP33 was added to the mixture and centrifuged to obtain the PEG-
CPP33@NPs. Thirty tumor-bearing mice were equally divided into five groups and
treated with normal saline, siRNA@ZIF-90, ORI@ZIF-90, NPs, and PEG-
2.9. In vitro cytotoxicity test CPP33@NPs (1.5 mg⋅kg− 1 siRNA and 3 mg⋅kg− 1 ORI) via intravenously
injection, respectively. The body weights and tumor volumes of the mice
Cells were seeded in a 96-well plate at a density of 5000 cells/well. were monitored every two days to explore the physiological influences
After culturing for 24 h, media containing different concentrations of and treatment efficiency of the NPs. Blood was harvested from the mice
NPs were added (siRNA: 10, 30, 50, 100, 150, 200, and 250 nM). After after treatment to detect the blood biochemical parameters.
culturing for 24 h, 20 μL 5% MTT solution was added to each well and
incubated for 4 h. Next, the supernatant was removed, and 150 μL of 2.16. Histopathological analysis
DMSO was added to each well. While providing protection from light,
the samples were placed on a shaker and shaken for 5 min. The absor­ The extracted organs (heart, liver, spleen, lungs, and kidneys) and
bance of each well was measured at an excitation wavelength of 490 nm tumors were preserved in formaldehyde (4%) and stained with hema­
using a microplate reader. toxylin and eosin (H&E).

2.10. Annexin V-FITC/PI measurement 2.17. Statistical methods

To investigate the cell apoptosis induced by NPs, cells were treated All experiments were performed with at least three independent
with NPs (siRNA: 125 nmol/L) for 24 h at 37 ◦ C. Cells were harvested, replications, and data were expressed as means ± standard deviations
washed with Phosphate buffer solution (PBS) and resuspended in 195 μL (SDs). Comparisons were performed using one-way analysis of variance;
annexin-V FITC binding solution. Next, 5 μL annexin V-FITC and 10 μL *P < 0.05 and **P < 0.001 were considered statistically significant.
PI were added and incubated with the cells for 20 min in the dark.
Finally, different cells were analyzed by flow cytometry (BD, LSRFor­ 3. Results and discussion
tessa SORP).
3.1. Synthesis of ZIF-8 and ZIF-90
2.11. Cell uptake study
ZIF-8 and ZIF-90 were synthesized by stirring at 25 ◦ C [31]. The
HepG2, A549, MDA-MB-231, and HT-29 cells were plated at a den­ Scanning electron microscope (SEM) images show that the synthesized
sity of 1 × 105 cells/well and cultured for 24 h. CPP33-PEG@NPs were ZIF-8 and ZIF-90 are circular in shape, with a particle size of approxi­
added to the plates at a final coumarin-6 concentration of 0.5 μg/ml. mately 100 nm (Fig. S1). The X-Ray Diffraction (XRD) pattern showed
After 4 h incubation, the cells were washed three times with PBS and that the synthesized ZIFs were completely crystalline, and the charac­
fixed with 4% paraformaldehyde for 10 min at 37 ◦ C. 4′ ,6-Diamidino-2- teristic diffraction peak was consistent with the standard pattern
Phenylindole (DAPI) was added for 5 min to stain the nuclei. Finally, the (Fig. S2). The Fourier Transform Ioncyclotron Resonance (FTIR) results
cells were imaged using a confocal laser scanning microscope (CLSM, illustrated the presence of characteristic peaks at the corresponding
Leica Microsystems, LAS X 3.5.5.19976). band positions of ZIF-8 and ZIF-90 (Fig. S3). ZIF-8 showed the stretching
vibration peak of the imidazole molecule C–– N at 1580 cm− 1, and ZIF-90
2.12. Evaluation of Intracellular Trafficking and Endosomal Escape showed the characteristic peak of the aldehyde group C– – O at 1680
cm− 1. As shown in Figs. S4–S5, the particle sizes of ZIF-8 and ZIF-90
To identify the intracellular location of CPP33-PEG@NPs, lysosome were 78.82 nm and 68.06 nm, respectively, and the zeta potentials
staining was performed on A549 cells. Briefly, A549 cells were incu­ were 13 mV and 9.99 mV, respectively, consistent with previous studies
bated with CPP33-PEG@NPs for 0.5, 4, 8, and 12 h and further treated [32–34]. The N in the organic ligands of ZIFs is easily protonated, giving
with LysoTracker Red (50 nM) for 30 min. The cells were imaged using a ZIFs a positive charge.
CLSM. The biosafety of ZIFs must be evaluated before conducting trans­
fection and drug-loading experiments. First, MTT assay showed that ZIF-
2.13. Animal and tumor-bearing mouse model 8 and ZIF-90 had no obvious cytotoxicity in the concentration range of
0–55 μg/mL, and the survival rate of A549 cells and normal human fi­
All animal experiments were performed in accordance with the broblasts (NHDFS) was more than 85% (Figs. S6–S7). Second, DAPI
guidelines approved by the Institutional Animal Care and Use Com­ staining showed that ZIF-8 and ZIF-90 (0–55 μg/mL) had no significant
mittee of the Beijing University of Chinese Medicine (BUCM-4- effect on nuclear morphology (Fig. S8). In addition, Annexin V-FITC/PI
2020123103-4138). BALB/c nude mice (8–10-weeks old, 20 g) were double staining assay also demonstrated that ZIF-8 and ZIF-90 (0–55 μg/
obtained from Sipeifu Biological Technology Co., Ltd. First, a tumor- mL) had no obvious apoptosis-inducing effect (Figs. S9–10). This dem­
bearing mouse model was established. A549 cells were subcutaneously onstrates that ZIF-8 and ZIF-90 exhibit good in vitro biological safety at
injected into the right forelimbs of the mice, and the tumor volume was certain doses.
monitored daily. The tumors were allowed to grow to an average volume
of 100 mm3 (V = W2 × L/2) prior to the study. 3.2. The construction of siRNA@ZIFs

2.14. In vivo fluorescence imaging siRNA was loaded by solvent adsorption [35]. Briefly, 1 mg of ZIF-8/
ZIF-90 was stirred with 9 nmol siRNA (2.5 mol/L) in DEPC water for 24
NPs were prepared and intravenously injected into tumor-bearing h. The drug loading capacity of ZIF-8 and ZIF-90 was 7.91 ± 0.13% and
mice. The mice were imaged using an IVIS imaging system (Caliper, 9.89 ± 0.19%, respectively (n = 3). The morphologies of ZIF-8 and ZIF-
Hopkington, MA, USA) before injection and at 2, 6, and 12 h after 90 were observed using SEM, and no significant changes were observed

373
M. Cai et al. Journal of Colloid And Interface Science 646 (2023) 370–380

after drug loading (Fig. S11). The dynamic light scattering results also (Fig. 1A–D). Interestingly, the protective effect of ZIF-90 against siRNA
confirmed that there was no significant change in size after drug loading was greater than that of ZIF-8, presumably because of the presence of
(Fig. S12). The FTIR spectra showed that the structure of ZIFs remained aldehyde ligands in ZIF-90, which make it more polar than ZIF-8 [37].
unchanged after drug loading (Fig. S13). The formation of siRNA@ZIFs ZIF-90 encapsulated hydrophilic siRNA and exhibited a better protective
was monitored by Ultraviolet–visible spectroscopy (UV–vis) absorption effect owing to steric hindrance. In addition, the effects of temperature
spectroscopy (Fig. S14). As shown in the figure, siRNA had a maximum and organic solvents on the stability of siRNA were investigated
absorption peak at 260 nm, whereas ZIF-8 showed no absorption peak (Figs. S15–16). Neither temperature nor organic solvent affected the
within 240–480 nm. When siRNA was mixed with ZIF-8, the mixed so­ stability of siRNA, which verified the double-stranded nature of the
lution exhibited an absorption peak at 260 nm, indicating that siRNA siRNA structure. Finally, we selected ZIF-90 as the siRNA vector for
was not coated with ZIF-8. Spectral scanning of siRNA@ZIF-8 showed subsequent experiments.
that the drug-loaded particles had no absorption peak at 260 nm, indi­
cating that siRNA was loaded into ZIF-8. ZIF-90 exhibits a maximum 3.3. Synthesis of NPs and PEG-CPP33@NPs
absorption peak at 290 nm. When siRNA was mixed with ZIF-90, the
maximum absorption peak of the mixed solution was approximately ORI regulates multiple signaling pathways in cells, including reactive
290 nm, and the shift in the maximum absorption peak was speculated oxygen species, Bcl-2/Bax, p53/p21, MAPK, miRNA, and fatty acid
to be related to the binding of siRNA and ZIF-90. The maximum ab­ synthase [38]. A large number of studies have emerged on the combi­
sorption peak of the drug-loaded particles was consistent with that of nation therapy of ORI with other drugs, such as cisplatin and cetuximab
ZIF-90, indicating that siRNA was successfully loaded into ZIF-90. [39,40]. In this study, the loading capacity of ZIF-90 on ORI was
siRNA is unstable under physiological conditions and is susceptible investigated. The content of ORI was detected using HPLC. The results
to degradation by nucleases in body fluids [36]. To verify the protection showed that the loading capacity of ORI for ZIF-90 was 27.28%.
of siRNA after ZIFs encapsulation, siRNA@ZIFs were dispersed in a Furthermore, 8.18% siRNA and 19.56% ORI were incorporated into the
medium containing serum or RNase A for 12 h. The results showed that pores of ZIF-90. siRNA and ORI enter the interior of ZIFs via electrostatic
after encapsulation with ZIF-8 or ZIF-90, the stability of siRNA in serum and π-π interactions [41–43]. To verify whether siRNA and ORI entered
and RNase A was improved compared to that of free siRNA, which may the pores of ZIF-90 or were absorbed on the surface of ZIF-90, Bru­
be due to the physical protection provided by the ZIFs scaffold nauer–Emmett–Teller (BET) analyses of ZIF-90 and ORI@siRNA@ZIF-

Fig. 1. (A) Protective effect of ZIFs against siRNA after different serum treatment times (a: free siRNA, b: siRNA@ZIF-8, c: siRNA@ZIF-90 incubated with 10% FBS at
37 ◦ C); B. relative activity of siRNA after different serum treatment times; (C) protective effect of ZIFs against siRNA after different RNase A treatment times (a: free
siRNA, b: siRNA@ZIF-8, c: siRNA@ZIF-90 incubated with RNase A at 37 ◦ C); (D) relative activity of siRNA after RNase A treatment times; (E) SEM of PEG-
CPP33@NPs; (F) FT-IR spectrum of PEG-CPP33@NPs; (G) zeta potential of PEG-CPP33@NPs; (H) siRNA release from PEG-CPP33@NPs at a pH 5.5 or pH 7.4;
and (I) ORI release from PEG-CPP33@NPs at pH 5.5 or pH 7.4. (n = 3, mean ± SD).

374
M. Cai et al. Journal of Colloid And Interface Science 646 (2023) 370–380

90 were also performed. The results are presented in Fig. S17. The BET characteristic peak of the NPs after PEGylation was found (Fig. S18),
specific surface area of ZIF-90 was 1419 m2/g, whereas that of indicating that PEGylation did not affect the crystal form of the NPs.
ORI@siRNA@ZIF-90 was 794 m2/g. According to the BET test results, Zeta potential results showed that PEG-CPP33@NPs transforms into a
the specific surface area and pore volume of ZIF-90 were significantly negative potential of –23 mv, which is opposite to the positive zeta
higher than those of ORI@siRNA@ZIF-90, indicating the successful potential of blank ZIF-90 (Fig. 1G). We speculated that PEG-CPP33
loading of siRNA and ORI. binding to the ZIF-90 surface caused a change in the zeta potential. A
To improve the distribution of ZIF-90 in vivo, we coated ZIF-90 with nanodrop microultraviolet spectrophotometer was used for ultraviolet
a CPP, CPP33, which specifically targets A549 cells [44]. The cysteine at full-wavelength scanning (Fig. S19). The siRNA had a maximum ab­
the end of CPP33 and the maleimide group at the end of mPEG2000-Mal sorption peak at 260 nm, ZIF-90 had a maximum absorption peak at 290
were used to construct the targeting ligand PEG-CPP33 via the addition nm, PEG-CPP33 had no UV absorption peak at 240–480 nm, and the UV
reaction of the sulfhydryl and maleimide groups. As shown in Fig. 1E, absorption peak of the NPs was consistent with that of the drug-carrying
the PEG-CPP33@NPs had a quasi-circular spherical shape, although the particles, indicating that the structure of the NPs did not change.
surface was no longer smooth, and the particle size was approximately The responsive release of drugs at tumor sites is a major research
100 nm (Fig. 1E). The FTIR spectrum showed that PEG-CPP33 had an- focus. Therefore, ORI and siRNA release rates in PEG-CPP33@NPs were
OH stretching vibration peak at 2880 cm− 1, –NH+ 3 deformation vibra­ evaluated in acidic (pH = 5.5) and neutral (pH = 7.4) PBS at 37 ◦ C. ORI
tion peaks at 1600–1575 cm− 1 and 1550–1504 cm− 1, and a COO- and survivin siRNA were released faster at a pH 5.0, than at a pH 7.4
symmetric stretching vibration peak at 1412 cm− 1 (Fig. 1F). The (Fig. 1H–I). At a pH 5.5, approximately 70% and 85% of ORI and sur­
bending and stretching vibration peaks of C–N appeared at 1220–1020 vivin siRNA, respectively, were released from the NPs within 24 h. In
cm− 1 and 1340–1280 cm− 1, and the vibration peak of CH in –CH2– contrast, approximately 38% and 40% of ORI and survivin siRNA,
appeared at 1466 cm− 1, indicating that PEG was successfully connected respectively, at a pH 7.4. This is due to the instability of ZIF-90 under
to the polypeptide chain CPP33. The infrared spectrum of PEG- acidic conditions [22,45], allowing the nanocarriers in acidic lysosomes
CPP33@NPs was similar to that of PEG-CPP33. PEG-CPP33@NPs to release more drugs, which facilitates tumor pH-responsive drug
retained the characteristic peaks of ZIF-90 at 1680 cm− 1 and 796 cm− 1, release [46].
although the peak intensity became weaker, indicating that PEG-CPP33
was modified on the surface of the drug-carrying particles and the
structure of the nanoparticles was intact. The XRD pattern of PEG- 3.4. Cellular uptake and lysosomal escape of NPs
CPP33@NPs was further examined, and no significant change in the
The specific selection of drugs for cells is the key to tumor-targeted

Fig. 2. CLSM images of the uptake of different drugs by different cells. (A) A549 cells; (B) HepG2 cells; (C) HT-29 cells; and (D) MDA-MB-231 cells. (a) Coumarin 6;
(b) Coumarin 6@ZIF-90; and (c) PEG-CPP33@Coumarin 6@ZIF-90.

375
M. Cai et al. Journal of Colloid And Interface Science 646 (2023) 370–380

therapy. Using Coumarin 6 as a fluorescent indicator, the specific small amount of siRNA was internalized into the cells (Fig. 3B). After 4 h
selectivity of PEG-CPP33 for A549 cells was investigated using a CLSM. of treatment, yellow fluorescence, formed by the superposition of green
The results showed that green fluorescence appeared in all four cell and red fluorescence, was observed in the cells, indicating that siRNA
types after incubation with coumarin 6 and Coumarin 6@ZIF-90, indi­ was swallowed by lysosomes. After co-incubation with cells for 8 h, the
cating that the targeting abilities of coumarin 6 and Coumarin 6@ZIF-90 yellow fluorescence decreased and the red fluorescence increased,
were not obvious (Fig. 2A–D). After incubating the cells with PEG- indicating that the siRNA successfully escaped from the lysosomes and
CPP33@Coumarin 6@ZIF-90, the intensity of green fluorescence in was released into the cytoplasm, which was more obvious after 12 h. The
the A549 cells was the highest, and no obvious green fluorescence was colocalization rates of Cy3-siRNA with lysosomes at 0.5, 4, 8, and 12 h
observed in the HepG2, HT-29, and MDA-MB-231 cells. This indicates were 0.45%, 94.34%, 58.13%, and 16.52%, respectively, which further
that PEG-CPP33@Coumarin 6@ZIF-90 is only taken up by A549 cells indicated that PEG-CPP33@Cy3-siRNAsur@ZIF-90 could achieve lyso­
and has high cell-targeting ability [47]. CPP33 promotes the internali­ somal escape of siRNA. After incubation with siRNA, the co-localization
zation of NPs in A549 cells through dynamin-dependent and clathrin/ signal (yellow) was significantly higher than that of PEG-CPP33@Cy3-
microcytosis/caveo-independent pathways [26,28]. siRNAsur@ZIF-90, and the level of co-localization was greater than
siRNASur was labeled with Cy3 for fluorescence, and the uptake of 90% at different times (Fig. S20), indicating that the escape of siRNA
siRNA, siRNA@ZIF-90, and PEG-CPP33@siRNA@ZIF-90 by A549 cells from the lysosomes was difficult. This enhanced lysosomal escape may
was measured using flow cytometry. As shown in Fig. 3A, only a small be related to the pH sponging effect of ZIF-90 [23].
amount of naked siRNA was taken up by A549 cells, most siRNA@ZIF-90
and PEG-CPP33@siRNA@ZIF-90 were internalized by the cells, and the 3.5. Survivin expression
drug-loaded nanoparticles showed the strongest fluorescence intensity
in the cells after modification with PEG-CPP33. This suggests that the The mRNA expression level was used as a direct indicator of the gene
encapsulation of ZIF-90 and modification of PEG-CPP33 could promote silencing effect of siRNA. Survivin mRNA expression was detected by
cellular uptake of the drug, and CPP33 in particular could further pro­ real-time Polymerase Chain Reaction (PCR) after NPs treatment. The
mote the internalization of the drug. results showed no significant difference between the cells treated with
Effective lysosomal escape is key to the therapeutic effects of siRNA. siRNA alone and the negative control group (Fig. 3C). The relative
After staining with LysoTracker Green, the lysosomes showed green expression of survivin mRNA in cells treated with siRNA@ZIF-90 and
fluorescence; Cy3-siRNAsur showed red fluorescence. After 0.5 h of in­ PEG-CPP33@siRNA@ZIF-90 was significantly lower than that in the
cubation with PEG-CPP33@Cy3-siRNAsur@ZIF-90, a small amount of negative control group. Among them, siRNA@ZIF-90 reduced the rela­
red fluorescence was observed inside the A549 cells, indicating that a tive expression of survivin mRNA by 57.41 ± 9.56%; PEG-

Fig. 3. (A) Intracellular fluorescence intensity of A549 cells incubated with siRNA, siRNA@ZIF-90, and PEG-CPP33@siRNA@ZIF-90 for 24 h; (B) CLSM images of the
localization of siRNA in A549 cells treated with PEG-CPP33@Cy3-siRNAsur@ZIF-90 for different times; (C) relative expression of survivin mRNA in A549 cells treated
with different nanoparticles for 24 h; (D) relative expression of survivin protein in A549 cells treated with different nanoparticles for 24 h; (E) images of Western blot:
(a) control; (b) siRNA; (c) siRNA@ZIF-90; and (d) PEG-CPP33@siRNA@ZIF-90; (F) cell viability of A549 cells treated with different drugs for 24 h; and (G) apoptosis
statistics of A549 cells treated with different drugs for 24 h. (n = 3, mean ± SD, *P < 0.05, **P < 0.001).

376
M. Cai et al. Journal of Colloid And Interface Science 646 (2023) 370–380

CPP33@siRNA@ZIF-90 reduced the relative expression of survivin 3.7. In vivo biodistribution of nanoformulations
mRNA by 69.34 ± 1.06%. This illustrates that CPP33-modified nano­
particles have a better survivin mRNA downregulation effect, possibly Having determined the in vitro biological processes of the nano­
owing to the enhanced efficacy of efficient delivery. formulations, we evaluated their tissue selectivity of the nano­
Survivin is a specific inhibitor of apoptosis in tumor cells. Down­ formulations. The biodistribution of nanoparticles in mice was observed
regulation of its mRNA level can reduce the expression of the survivin at 2, 6, and 12 h after tail vein injection of Cy3-siRNAsur@ZIF-90 and
protein and induce apoptosis of tumor cells, which plays an anti-tumor PEG-CPP33@Cy3-siRNAsur@ZIF-90 (siRNA, 2 nmol). Two hours after
role. Western blotting revealed that both siRNA@ZIF-90 and PEG- administration, the mice injected with PEG-CPP33@Cy3-siRNAsur@ZIF-
CPP33@siRNA@ZIF-90 downregulated survivin protein expression. As 90 showed strong fluorescence at the tumor site, whereas the fluores­
shown in Fig. 3D–E, no significant difference was observed in survivin cence of Cy3-siRNAsur@ZIF-90 was mainly distributed in the abdomen
protein expression in A549 cells treated with siRNA alone compared of the mice (Fig. 4A). Twelve hours after injection, PEG-CPP33@Cy3-
with the control group. After 24 h of treatment with siRNA@ZIF-90 and siRNAsur@ZIF-90 still accumulated the most at the tumor site, which
PEG-CPP33@siRNA@ZIF-90, the survivin protein expression levels in proved that PEG-CPP33 modification could retain the nanoparticles at
A549 cells decreased significantly compared with that in the control the tumor site for a long time. In addition, as shown in Fig. 4B, the
group, and the targeted drug-loaded nanoparticles reduced the protein unmodified PEG-CPP33 nanoparticles mainly accumulated in the liver,
expression more significantly. These results demonstrate that CPP33 which may be due to their uptake by the mononuclear phagocytic sys­
modification could further downregulate the expression of anti- tem [48].
apoptotic proteins in A549 cells, thereby showing a more significant After administration, the fluorescence intensity of each tissue and
pro-apoptotic effect. tumor was analyzed. The tumor sites of the mice in the Cy3-siRNA
sur
@ZIF-90 group showed a certain degree of fluorescence intensity,
3.6. In vitro cytotoxicity of nanoparticles although the liver and kidney showed stronger fluorescence (Fig. 4C).
Fluorescence at the tumor site in the PEG-CPP33@Cy3-siRNAsur@ZIF-
The cytotoxicity of ORI, survivin siRNA and the different nano­ 90 group was the strongest. There was also fluorescence accumulation in
particles in A549 cells was determined using the MTT assay. As shown in the liver and kidney, which was mainly related to the liver and kidney
Fig. 3F, survivin siRNA showed little cytotoxicity, whereas survivin being important metabolism and excretion organs for drugs [49,50].
siRNA@ZIF-90 showed dose-dependent cytotoxicity, which was attrib­ Compared with Cy3-siRNAsur@ZIF-90, the PEG-CPP33@Cy3-
uted to the enhanced uptake of survivin siRNA by ZIF-90 encapsulation. siRNAsur@ZIF-90 group showed stronger tumor accumulation,
Both ORI and ORI@ZIF-90 showed cytotoxicity, although survivin whereas other organs showed lower fluorescence values. This is due to
siRNA combined with ORI significantly improved the antitumor effects the ability of CPP33 to penetrate the cell membrane of human non-small
of individual chemotherapeutics. No significant difference was observed cell lung cancer A549 cells while minimizing transduction to normal
in the cytotoxicity of NPs before and after coupling with CPP33 peptide tissues [47].
chains, which may be due to similar intracellular drug content after 24 h
of incubation. Since both ORI and survivin siRNA inhibited survivin
protein expression, annexin V-FITC staining and FACS analysis were 3.8. Anti-tumor efficacy in vivo
used to detect the apoptosis rates induced by the different drugs. After
ORI and survivin siRNA treatment, the apoptosis rates were 35.2% and The in vivo anti-tumor effects of PEG-CPP33 were investigated in
10.9%, respectively (Fig. 3G). The apoptosis rates of survivin A549 tumor-bearing nude mice. A549 tumor-bearing nude mice were
siRNA@ZIF-90, ORI@ZIF-90, NPs, and PEG-CPP33@NPs were 30.13%, randomly divided into five groups and injected with saline, siRNA@ZIF-
36.45%, 51.37%, and 60.73%, respectively. The combination of ORI and 90, ORI@ZIF-90, NPs and PEG-CPP33@NPs (1.5 mg⋅kg− 1 siRNA and 3
survivin siRNA resulted in a higher apoptotic rate, which was consistent mg⋅kg− 1 ORI per injection) via the tail vein every 2 days for four
with the results of the MTT assay. consecutive days. The body weight and tumor volume of nude mice were
measured for 8 consecutive days. As shown in Fig. 5A, no significant
change was observed in the body weights of the mice in each group,

Fig. 4. In vivo biodistribution of nanoformulations. (A) In vivo fluorescence imaging at 2 h, 6 h and 12 h after injection of different nanoparticles; (B) fluorescence
imaging of major organs and tumors in mice 12 h after injection of different nanoformulations; (C) fluorescence intensity statistics of main organs and tumors of mice
12 h after injection of different nanoformulations. (a: Cy3-siRNAsur@ZIF-90; b: PEG-CPP33@Cy3-siRNAsur@ZIF-90; n = 3, mean ± SD, *P < 0.05, **P < 0.001).

377
M. Cai et al. Journal of Colloid And Interface Science 646 (2023) 370–380

Fig. 5. (A) Changes of body weight over time after injection of different nanoparticles (n = 6, mean ± SD); (B) changes in the tumor volumes of mice after injection
of different nanoparticles (n = 6, mean ± SD); (C) photo of the tumor after administration (a: saline, b: siRNA@ZIF-90, c: ORI@ZIF-90, d: NPs, and e: PEG-
CPP33@NPs); (D) tumor weight at the end of administration (n = 6, mean ± SD); and (E–I) expression of Bcl-2, Bax, PARP, caspase 3 and survivin in tumors (n
= 3, mean ± SD, *P < 0.05, **P < 0.001).

indicating that the nanoparticles had good biosafety and no significant promoting tumor apoptosis. Therefore, we determined that ORI exerts
toxicity. Changes in tumor volume in each group are shown in Fig. 5B–D. anti-tumor effects through multiple targets and enhances the tumor
Compared with the control group, siRNA@ZIF-90, ORI@ZIF-90, NPs, suppressive effect of siRNA through multiple pathways, such as caspase-
and PEG-CPP33@NPs showed tumor inhibition. The tumor inhibition 3, survivin, and Bcl-2/Bax.
rates of siRNA@ZIF-90, ORI@ZIF-90, NPs, and PEG-CPP33@NPs groups
were 7.28%, 27.54%, 49.87%, and 61.04%, respectively. In contrast, the
tumors in the NPs and PEG-CPP33@NPs groups were significantly 3.9. In vivo biosafety evaluation
smaller than those in the other groups, which may have been due to the
combined therapeutic effect of siRNA and ORI, making the anti-tumor Owing to the inevitable accumulation of nano-formulations in the
effect more significant. In addition, PEG-CPP33@NPs showed the liver and kidney of mice, the physiological and biochemical parameters
strongest anti-tumor effect, which may have been due to the specific of the mice were measured to evaluate possible liver and kidney dam­
targeting of PEG-CPP33@NPs to A549 cells to promote the local accu­ age. No significant differences were observed in ALT, AST, ALB, and
mulation of siRNA and ORI in tumor tissues. In conclusion, PEG- BUN concentrations among the groups, indicating no significant damage
CPP33@NPs exhibited good siRNA and DOX synergy and excellent to the liver and kidney function in each preparation (Fig. S21). In
targeting capability against A549 tumors. addition, hematological parameters were measured in the mice after
ORI can reportedly induce tumor cell apoptosis through multiple treatment (Fig. S22). No obvious abnormalities were observed on
pathways, such as caspase-3, survivin, and Bcl-2/Bax [51]. The syner­ routine blood examinations in any group. The tumor, heart, liver,
gistic application of ORI and other drugs can achieve an additive effect spleen, lungs, and kidneys of the mice in each group were collected and
on cancer treatment [52]. Fan et al. reported that ORI reversed cisplatin stained with H&E for observation, as shown in Fig. S23. No significant
resistance in non-small cell lung cancer [53]. To investigate apoptosis in histological differences were observed between the major organs in the
tumor tissues, survivin, Bcl-2/Bax, PARP, caspase 3 levels were treatment and control groups. No significant histopathological changes
measured by western blotting. In accordance with the above results, were observed in any of the major organs (heart, liver, spleen, lungs, or
survivin levels were significantly decreased in the NPs and PEG- kidneys), indicating that the nano-preparation was safe in vivo. In
CPP33@NPs groups, while the Bax/Bcl-2 ratio and PARP and caspase contrast, tumor morphology significantly changed after treatment with
3 levels were significantly higher than those in the other groups NPs and PEG-CPP33@NPs, further demonstrating the therapeutic effect
(Fig. 5E–I). In addition, PEG-CPP33 had a more significant effect on of the nano-preparations.

378
M. Cai et al. Journal of Colloid And Interface Science 646 (2023) 370–380

4. Conclusion Appendix A. Supplementary data

We constructed a nanoplatform with tumor-homing and enhanced Supplementary data to this article can be found online at https://doi.
lysosomal escape capabilities for synergy with chemotherapy and gene org/10.1016/j.jcis.2023.04.126.
therapy. The high porosity of MOFs enables the co-delivery of survivin
siRNA and ORI. The targeting ability of A549 cells was determined by References
coating a PEGylated CPP (CPP33) onto the surface. The results showed
that ZIF-8 and ZIF-90 exerted protective effects against siRNA. In [1] R.L. Siegel, K.D. Miller, H.E. Fuchs, et al., Cancer statistics, 2022, CA Cancer J. Clin.
72 (1) (2022) 7–33.
addition, ZIF-90, with an aldehyde structure, showed better siRNA [2] N.D. James, S.A. Hussain, E. Hall, et al., Radiotherapy with or without
protection in serum-containing media and RNase. This protective effect chemotherapy in muscle-Invasive bladder cancer, N. Engl. J. Med. 366 (16) (2012)
resulted in a significant increase in the stability of the siRNA, thereby 1477–1488.
[3] K. Odunsi, Immunotherapy in ovarian cancer, Ann. Oncol. 28 (2017) 1–7.
alleviating the vulnerability of the siRNA to circulation in vivo. In [4] Z. Mirza, S. Karim, Nanoparticles-based drug delivery and gene therapy for breast
contrast to previous studies, survivin siRNA enhanced lysosomal escape cancer: recent advancements and future challenges, Semin. Cancer Biol. 69 (2021)
and was specifically taken up by A549 cells [54,55]. In addition, we 226–237.
[5] C.J. Sweeney, Y.H. Chen, M. Carducci, et al., Chemohormonal therapy in
found that co-delivery of ORI with siRNA significantly increased the metastatic hormone-sensitive prostate cancer, N. Engl. J. Med. 373 (8) (2015)
apoptotic index of A549 cells and decreased the expression of survivin 737–746.
compared with the application of siRNA alone, indicating a good syn­ [6] P.L. McCarthy, K. Owzar, C.C. Hofmeister, et al., Lenalidomide after stem-cell
transplantation for multiple myeloma, N. Engl. J. Med. 366 (19) (2012)
ergistic effect between survivin siRNA and ORI [56]. The coating of PEG-
1770–1781.
CPP33 improved the tumor-homing ability of the nanoplatform, and the [7] N.E. Sounni, A. Noel, Targeting the tumor microenvironment for Cancer Therapy,
anticancer effect was significantly improved without obvious damage to Clin. Chem. 59 (1) (2013) 85–93.
normal tissues. Most importantly, this study provides a simple method [8] A. Wittrup, J. Lieberman, Knocking down disease: a progress report on siRNA
therapeutics, Nat. Rev. Genet. 16 (9) (2015) 543–552.
for the simultaneous delivery of siRNA and small-molecule drugs to [9] Y. Cao, H.Y. Huang, L.Q. Chen, et al., Enhanced lysosomal escape of pH-responsive
achieve effective tumor suppression. Notably, PEGylated CPP-coated Polyethylenimine-Betaine FUNCTIONALIZED Carbon Nanotube for the codelivery
MOFs offer a promising low-side-effect strategy for synergistic chemo­ of survivin small interfering RNA and doxorubicin, ACS Appl. Mater. Interfaces 11
(10) (2019) 9763–9776.
therapy/gene therapy and may accelerate the clinical translation of [10] P.H. Zhang, K.L. An, X.M. Duan, et al., Recent advances in siRNA delivery for
MOFs for cancer therapy. cancer therapy using smart nanocarriers, Drug Discovery Today 23 (4) (2018)
Availability of data and materials: 900–911.
[11] M.Y. Chen, C.Y. Dong, S. Shi, Nanoparticle-mediated siRNA delivery and
The datasets used and/or analyzed during the current study are multifunctional modification strategies for effective cancer therapy, Adv. Mater.
available from the corresponding author on reasonable request. Technol. 6 (10) (2021).
Declarations: [12] M. Zhang, X. Qin, Z. Zhao, et al., A self-amplifying nanodrug to manipulate the
Janus-faced nature of ferroptosis for tumor therapy, Nanoscale Horiz. 7 (2) (2022)
Ethics approval and consent to participate. 198–210.
This study is approved by the Institutional Animal Care and Use [13] Z.P. Zhao, Q. Li, X.H. Qin, An injectable hydrogel reshaping adenosinergic axis for
Committee of Beijing University of Chinese Medicine. cancer therapy, Adv. Funct. Mater. 32 (24) (2022) 2200801.
[14] X. Ren, N. Wang, Y. Zhou, et al., An injectable hydrogel using an
Consent for publication:
immunomodulating gelator for amplified tumor immunotherapy by blocking the
All authors agree to submit the manuscript for publication. arginase pathway, Acta Biomater. 124 (2022) 179–190.
[15] H. Tian, M. Zhang, G. Jin, et al., Cu-MOF chemodynamic nanoplatform via
modulating glutathione and H2O2 in tumor microenvironment for amplified cancer
CRediT authorship contribution statement therapy, J. Colloid. Interface Sci. 587 (2021) 358–366.
[16] Z. Xu, Z. Wu, S. Huang, et al., A metal-organic framework-based
Mengru Cai: Conceptualization, Investigation, Methodology, immunomodulatory nanoplatform for anti-atherosclerosis treatment, J. Control.
Release 354 (2023) 615–625.
Writing – original draft, Writing – review & editing. Yu Yao: Concep­ [17] R. Zheng, J. Guo, X. Cai, et al., Manganese complexes and manganese-based metal-
tualization, Investigation, Methodology, Data curation. Dongge Yin: organic frameworks as contrast agents in MRI and chemotherapeutics agents:
Visualization, Investigation. Rongyue Zhu: Validation. Tingting Fu: applications and prospects, Colloids Surf. B Biointerfaces 213 (2022), 112432.
[18] Q. Ding, Z. Xu, L. Zhou, et al., A multimodal metal-organic framework based on
Validation. Jiahui Kong: Investigation, Visualization. Kaixin Wang:
unsaturated metal site for enhancing antitumor cytotoxicity through chemo-
Investigation, Visualization. Jing Liu: Investigation, Validation. Aina photodynamic therapy, J. Colloid. Interface Sci. 621 (2022) 180–194.
Yao: . Yidan Ruan: Investigation, Validation. Wenjuan Shi: Formal [19] H. He, H. Han, H. Shi, et al., Construction of thermophilic lipase-embedded metal-
organic frameworks via biomimetic mineralization: Aa biocatalyst for ester
analysis, Investigation. Qian Zhu: Formal analysis, Investigation. Jian
hydrolysis and kinetic resolution, ACS Appl. Mater. Interfaces 8 (37) (2016)
Ni: Supervision, Project administration. Xingbin Yin: Project adminis­ 24517–24524.
tration, Funding acquisition. [20] F.K. Shieh, S.C. Wang, C.I. Yen, et al., Imparting functionality to biocatalysts via
embedding enzymes into nanoporous materials by a de novo approach: size-
selective sheltering of catalase in metal-organic framework microcrystals, J. Am.
Chem. Soc. 137 (13) (2015) 4276–4279.
Declaration of Competing Interest [21] H.N. Abdelhamid, Zeolitic imidazolate frameworks (ZIF-8) for biomedical
applications: a review, Curr. Med. Chem. 28 (34) (2021) 7023–7075.
The authors declare that they have no known competing financial [22] Z.Q. Jiang, Y.J. Wang, L. Sun, et al., Dual ATP and pH responsive ZIF-90
nanosystem with favorable biocompatibility and facile post-modification improves
interests or personal relationships that could have appeared to influence therapeutic outcomes of triple negative breast cancer in vivo, Biomaterials 197
the work reported in this paper. (2019) 41–50.
[23] M.R. Cai, W.L. Liang, K.X. Wang, et al., Aperture modulation of isoreticular metal
organic frameworks for targeted antitumor drug delivery, ACS Appl. Mater.
Data availability
Interfaces 14 (32) (2022) 36366–36378.
[24] M. Swierczewska, H.S. Han, K. Kim, et al., Polysaccharide-based nanoparticles for
Data will be made available on request. theranostic nanomedicine, Adv. Drug Deliv. Rev. 99 (2016) 70–84.
[25] M. Wang, M. Thanou, Targeting nanoparticles to cancer, Pharmacol. Res. 62 (2)
(2010) 90–99.
Acknowledgements [26] E. Kondo, K. Saito, Y. Tashiro, et al., Tumour lineage-homing cell-penetrating
peptides as anticancer molecular delivery systems, Nat. Commun. 3 (2012) 951.
[27] M.D. Hade, C.N. Suire, Z. Suo, An effective peptide-based platform for efficient
This research was funded by the Beijing Natural Science Foundation exosomal loading and cellular delivery of a microRNA, ACS Appl. Mater. Interfaces
(No. 7202121), Young Elite Scientists Sponsorship Program by CACM 15 (3) (2023) 3851–3866.
(2021-QNRC2-A03), and the National Natural Science Foundation of
China (No. 81703715).

379
M. Cai et al. Journal of Colloid And Interface Science 646 (2023) 370–380

[28] M. Qiu, J. Ouyang, Y.H. Wei, et al., Selective cell penetrating peptide- [42] M. Cai, L. Qin, L. You, et al., Functionalization of MOF-5 with mono-substituents:
functionalized envelope-type chimeric lipopepsomes boost systemic RNAi therapy effects on drug delivery behavior, RSC Adv. 10 (60) (2020) 36862–36872.
for lung tumors, Adv. Healthc. Mater. 8 (16) (2019) e1900500. [43] K. Buyens, M. Meyer, E. Wagner, et al., Monitoring the disassembly of siRNA
[29] D.C. Altieri, Survivin - the inconvenient IAP, Semin. Cell Dev. Biol. 39 (2015) polyplexes in serum is crucial for predicting their biological efficacy, J. Control.
91–96. Release 141 (1) (2010) 38–41.
[30] J.J. Li, Y.H. Wu, D. Wang, et al., Oridonin synergistically enhances the anti-tumor [44] S.L. Yu, H. Koo, S.I. Lee, et al., A Synthetic CPP33-conjugated HOXA9 active
efficacy of doxorubicin against aggressive breast cancer via pro-apoptotic and anti- domain peptide inhibits invasion ability of Non-small lung cancer cells,
angiogenic effects, Pharmacol. Res. 146 (2019) 104313. Biomolecules 10 (11) (2020) 1589.
[31] H. Kaur, G.C. Mohanta, V. Gupta, et al., Synthesis and characterization of ZIF-8 [45] X. Xiao, S. Liang, Y. Zhao, et al., Core-shell structured 5-FU@ZIF-90@ZnO as a
nanoparticles for controlled release of 6-mercaptopurine drug, J. Drug Deliv. Sci. biodegradable nanoplatform for synergistic cancer therapy, Nanoscale 12 (6)
Technol. 41 (2017) 106–112. (2020) 3846–3854.
[32] B. Liu, Y. Yang, H. Wu, et al., Zeolitic Imidazolate framework-8 triggers the [46] R.F. Zou, Q.Y. Gong, Z.Z. Shi, et al., A ZIF-90 nanoplatform loaded with an enzyme-
inhibition of arginine biosynthesis to combat Methicillin-resistant staphylococcus responsive organic small-molecule probe for imaging the hypoxia status of tumor
aureus, Small (2023) e2205682. cells, Nanoscale 12 (27) (2020) 14870–14881.
[33] H. Wang, Y. Chen, H. Wang, et al., DNAzyme-loaded metal-organic frameworks [47] C. Lin, X. Zhang, H. Chen, et al., Dual-ligand modified liposomes provide effective
(MOFs) for self-sufficient gene therapy, Angew. Chem. Int. Ed. Engl. 58 (22) (2019) local targeted delivery of lung-cancer drug by antibody and tumor lineage-homing
7380–7384. cell-penetrating peptide, Drug Deliv. 25 (1) (2018) 256–266.
[34] C.I. Yen, S.M. Liu, W.S. Lo, et al., Cytotoxicity of postmodified zeolitic imidazolate [48] H. Maeda, Macromolecular therapeutics in cancer treatment: the EPR effect and
framework-90 (ZIF-90) nanocrystals: correlation between functionality and beyond, J. Control. Release 164 (2) (2012) 138–144.
toxicity, Chemistry 22 (9) (2016) 2925–2929. [49] A. Kermanizadeh, B.K. Gaiser, H. Johnston, et al., Toxicological effect of
[35] M.A. Mohammad, S.A. Negar, E. Azra, Development of metal-organic frameworks engineered nanomaterials on the liver, Br. J. Pharmacol. 171 (17) (2014)
(ZIF-8) as low-cost carriers for sustained release of hydrophobic and hydrophilic 3980–3987.
drugs: in vitro evaluation of anti-breast cancer and anti-infection effect, J. Cluster [50] L.R. Guo, I. Panderi, D.D. Yan, et al., A Comparative study of hollow Copper sulfide
Sci. 1 (16) (2022). nanoparticles and hollow Gold Nanospheres on degradability and toxicity, Acs
[36] W. Ho, X.Q. Zhang, X.Y. Xu, Biomaterials in siRNA delivery: a comprehensive Nano 7 (2013) 8780–8793.
review, Adv. Healthcare Mater. 5 (12) (2016) 2715–2731. [51] Y. Gui, J. Cheng, Z. Chen, Oridonin improves the therapeutic effect of lentinan on
[37] X. Xu, K. Lin, Y. Wang, et al., A metal-organic framework based inner ear delivery lung cancer, Exp. Ther. Med. 22 (2) (2021) 886.
system for the treatment of noise-induced hearing loss, Nanoscale 12 (30) (2020) [52] J. Liermann, P. Naumann, F. Fortunato, et al., Phytotherapeutics oridonin and
16359–16365. ponicidin show additive effects combined with irradiation in pancreatic cancer in
[38] J. Pi, H. Jin, J.H. Jiang, et al., Single molecule force spectroscopy for in-situ Vitro, Radiol. Oncol. 51 (4) (2017) 407–414.
probing oridonin inhibited ROS-mediated EGF-EGFR interactions in living KYSE- [53] X. Fan, T. Wang, Z. Ji, et al., Synergistic combination therapy of lung cancer using
150 cells, Pharmacol. Res. 119 (2017) 479–489. lipid-layered cisplatin and oridonin co-encapsulated nanoparticles, Biomed.
[39] Z.W. He, X.L. Xiao, S. Li, et al., Oridonin induces apoptosis and reverses drug Pharmacother. 141 (2021), 111830.
resistance in cisplatin resistant human gastric cancer cells, Oncol. Lett. 14 (2) [54] K. Wang, X. Chen, F. Yan, et al., 5’-triphosphate-siRNA against survivin gene
(2017) 2499–2504. induces interferon production and inhibits proliferation of lung cancer cells in
[40] S.J. Cao, M.J. Xia, Y.W. Mao, et al., Combined oridonin with cetuximab treatment vitro, J. Immunother. 36 (5) (2013) 294–304.
shows synergistic anticancer effects on laryngeal squamous cell carcinoma: [55] J.L. Liu, Y. Wang, J. Jiang, et al., Inhibition of survivin expression and mechanisms
involvement of inhibition of EGFR and activation of reactive oxygen species- of reversing drug-resistance of human lung adenocarcinoma cells by siRNA, Chin.
mediated JNK pathway, Int. J. Oncol. 49 (5) (2016) 2075–2087. Med. J. (Engl.) 123 (20) (2010) 2901–2907.
[41] D. Ling, H. Li, W. Xi, et al., Heterodimers made of metal-organic frameworks and [56] B. Zheng, S. Yang, M. Wang, et al., Non-covalent nanocomplexes of folic acid and
upconversion nanoparticles for bioimaging and pH-responsive dual-drug delivery, reducible polyethylenimine for survivin siRNA delivery, Anticancer Res. 35 (10)
J. Mater. Chem. B 8 (6) (2020) 1316–1325. (2015) 5433–5441.

380

You might also like