Materials
1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N [amin (polyethylene glycol)-2000] (DSPE-PEG2000-NH2) and Egg phosphatidylcholine (EPC) were obtained from AVT Pharmaceutical Technology Co., Ltd. (Shanghai, China). Lyso-Tracker Green was from Beyotime Biotechnology Co., Ltd. (Shanghai, China). Single-chain variable fragment (scFv) was provided by Merry Bio Co., Ltd. (Nanjing, China). Cholesterol, Hoechst33258, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), Agarose and 4,6-diamino-2-phenylindole (DAPI) were acquired from Solarbio (Beijing, China). Nine arginine (R9) peptide was synthesized from Shanghai Taopu Biotechnology Co., Ltd. (Shanghai, China). DSPE-PEG-cRGD was synthesized by Xi’an Ruixi Biological Technology Co., Ltd. (Xi’an, China). Irinotecan base (IRI), Doxorubicin Hydrochloride (DOX·HCl) were purchased from Meilun Biotechnology Co., Ltd. (Dalian, China). FAP1 Ab-AF5344 and alpha-SMA Ab-AF1032 were obtained from Affinity Biosciences LTD (Jiangsu, China).
Cell culture and animals
Embryonic mouse fibroblasts (NIH 3T3) and mouse CT-26 colon carcinoma cells were obtained from cell bank of Chinese Academy of Sciences (Shanghai, China). Both cells were cultured in the culture media containing 10% fetal bovine serum with addition of 1% penicillin–streptomycin. DMEM and RPMI-1640 media were selected for NIH 3T3 and CT-26 cells, respectively. The cells were cultured at 37 ºC containing 5% CO2. For the incubation of CT-26/NIH 3T3 co-cultured cells, both cells were mixed at the ratio of 1:2.
Female BALB/c mice (14–16 g) were provided by Pengyue Experimental Animal Co., Ltd. (Jinan, China). All animal experiments were conducted in accordance with the Regulations on Animal Control issued by the Ministry of Health of the People's Republic of China and the Ethical Review of Animal Experiments issued by Weifang Medical University (2017–025).
Synthesis of DSPE-PEG-R9
R9 peptide were dissolved in DMSO and activated by EDC for 10–15 min, followed by addition of NHS with stirring for 2 h. Afterwards, DSPE-PEG2000-NH2 and a small amount of triethylamine were added to react for 24 h under the protection of nitrogen. The reaction solution was dialyzed for 48 h and then lyophilized to obtain DSPE-PEG-R9. The structure of DSPE-PEG-R9 was characterized by 1H-NMR. The yield and reaction percent were calculated as follows:
$$Yield \left( \% \right) = \frac{mass \,of \,DSPE - PEG - R9}{{mass\, of\, total}} \times 100\%$$
$${Reaction}\, {percent}\left( \% \right) = \frac{{{Area}({R}9)/{Number}({R}9)}}{{{Area}({PEG})/{Number}({PEG})}} \times 100\%$$
In which Area(PEG) and Area(R9) represent the peak area of “alkyl in PEG” and “guanidyl in R9”, repectively. While Number(PEG) and Number(R9) represent the hydrogen proton numbers of “alkyl in PEG” and “guanidyl in R9”, respectively.
Preparation of liposomes
Before the preparation of liposomes, DSPE-PEG-R9 was obtained by one-step amide reaction using DSPE-PEG2000-NH2 and R9 peptide. IRI-RGD/R9-Lip were prepared via the following procedures. IRI, EPC, DSPE-PEG-cRGD, DSPE-PEG-R9 and cholesterol were dissolved in 2 mL ethanol to form a lipophilic solution, in which the mole ratio of DSPE-PEG-cRGD: DSPE-PEG-R9: EPC was controlled at 5:5:90. The solution were slowly injected into 5 mL PBS and stirred at 60 ºC for 1 h. IRI-loaded liposomes (IRI-RGD/R9-Lip) were prepared after ultrasonication and filtered through 0.45 μm and 0.22 μm membrane for 3 times, respectively [42]. Afterwards, scFv was added and electrostatic adsorbed on the surface of IRI-RGD/R9-Lip to obtain co-loaded liposomes (IRI-RGD/R9-sLip).
Besides, IRI-loaded liposomes without cRGD and R9 modification (IRI-Lip) were prepared as control group. IRI-RGD-Liposomes (IRI-RGD-Lip) were also prepared with the mole ratio of DSPE-PEG-cRGD: EPC at 5:95. Additionally, doxorubicin base (DOX) was obtained after DOX·HCl was dehydrochlorinated, and DOX loaded liposomes (DOX-Lip) were prepared for fluorescent trace. All the liposomes were prepared using the same method.
Characterization of liposomes
The morphologies of IRI-RGD/R9-sLip was visualized by transmission electron microscopy (TEM). Particle sizes, polydispersity index (PDI) and zeta potentials of liposomes were determined by Malvern Zetasizer Nano ZS90. To evaluate the stability of IRI-RGD/R9-sLip, the size changes was analyzed for 14 days after the samples diluted with RPMI-1640 or PBS, respectively. Besides, after the liposomes demulsified by 10% Triton X-100, the concentrations of IRI or DOX were measured by ultraviolet (UV) spectrophotometer at the UV absorption wavelength of 360 nm or 480 nm, respectively. Encapsulation efficiency (EE%) and drug loading (DL%) were figured out using the equations as below:
$$EE\% = \frac{{W_{encapsulated drug} }}{{W_{total drug} }} \times 100\%$$
$$DL\% = \frac{{W_{encapsulated drug} }}{{W_{liposomes} }} \times 100\%$$
In vitro release of IRI and scFv
The release profiles of IRI were studied using the dialysis bag method, in which 0.5% Tween-80 were added into PBS (pH 7.4) solution to meet the sink condition [43]. Briefly, 1 mL free IRI or IRI-Lip were transferred into dialysis bags (MWCO = 3500) respectively and incubated with 30 mL releasing medium at 37 ºC under 100 rpm shaking. At each time interval, 2 mL solution was extracted and supplemented with fresh release medium of the equal volume. The drug contents were analyzed by UV spectrophotometer at the absorption wavelength of 360 nm.
Regarding the release profiles of anti-FAP scFv, in consideration only after binding to the FAP on CAFs, scFv would be shed from the liposomes. Herein, 300μL R9-sLip were firstly incubated with activated NIH3T3 cells, and PBS group was set as control group to eliminate the influence of cell secretions. At different time points, 150μL cell supernatant solution was extracted and diluted with 150μL PBS. The content of scFv was determined by BCA microprotein quantitative kit.
In vitro cytotoxicity assay
In vitro cytotoxicity of free IRI, free scFv, IRI-Lip, IRI-RGD-Lip, IRI-RGD/R9-Lip and IRI-RGD/R9-sLip were evaluated using MTT assay. 150 μL of CT-26 cells (5 \(\times\) 103 cells/well) or CT-26/NIH 3T3 co-cultured cells (the ratio of CT-26 to NIH 3T3 cells at 1:2) were seeded in 96-well plates and cultured overnight [44]. Different concentrations of IRI or scFv were added respectively and incubated for 48 h. Afterwards, 20 μL of MTT (5 mg/mL) was added to each well and incubated for another 4 h. Finally, MTT medium was replaced by 150 μL of DMSO. The absorbance was measured at 490 nm using a microplate reader (ELX800, Bio-Tek, USA). Besides, blank liposomes were incubated with CT-26 cells or CT-26/NIH 3T3 co-cultured cells for 48 h to evaluate their cell viability.
The normal NIH 3T3 cells can be activated by tumor cells or tumor cell supernatant to become activated NIH 3T3 cells (CAFs). In brief, NIH 3T3 cells alone were cultured for 8 h and then pre-treated with supernatant of CT-26 cells for 16 h. After that, the cells were treated with IRI, RGD/R9-Lip and RGD/R9-sLip for 48 h and evaluated by MTT assay in activated NIH3T3 cells (incubated with CT-26 supernatants).
Cell migration assay
Firstly, CT-26 cells (4 \(\times\) 105 cells/well) or CT-26/NIH 3T3 co-cultured cells were inoculated in six-well plates. After the cells grown to 90–100%, three lines were scraped from each plate with the sterile tip of a spear. Each well was washed twice with PBS and treated with different preparations. For the dosage of free IRI, IRI-Lip, IRI-RGD-Lip and IRI-RGD/R9-sLip, the concentration of IRI in all the formulations were fixed at 15 μg/mL. Inverted fluorescence microscopy was used to obtain the images at 0 h and 24 h. The migration rate was evaluated by area detection methods, which was measured by ImageJ software and calculated as
$${Migration} {rate} = \frac{{{Width}_{{0{h}}} - {Width}_{{24{h}}} }}{{{Width}_{{0{h}}} }} \times 100\%$$
Western blot analysis
After washing cells with PBS for three times and adding cell lysis buffer, the cells were scraped off with a cell scrape and centrifuged at 14,000 g for 5 min. The concentration of the supernatant was quantified by Bicinchoninic acid (BCA) kit. The separated proteins that run on SDS-PAGE gels (12%) transferred to polyvinylidene difluoride (PVDF) membrane, and the sealant containing 5% milk was used to block the membrane for 2 h. After the membrane washed with TBST for three times, the primary antibody was added and incubated overnight. The membrane was further washed to remove the unbound primary antibody and then incubated with secondary antibody at room temperature for 1 h. Ultra-sensitive ECL chemiluminescence solution was added to observe the protein bands.
In vitro cellular uptake
DOX-loaded liposomes were prepared to analyze the intracellular accumulation of drugs in substitution for IRI. CT-26 cells (5 × 104 cells/well) were seeded in 24-well plates containing round glass sheet. The DOX-Lip or DOX-RGD-Lip at the DOX concentration of 10 μg/mL were added for 1 h. To study the effect of RGD on cells uptake efficiency, the cells were firstly incubated with free RGD solution at the concentration of 1 mg/mL for 4 h, followed by incubation with DOX-RGD-Lip for 1 h. Each plate was washed three times with PBS. 4% tissue fixing fluid was applied to immobilize cells and then washed away. The nuclei were stained with DAPI and washed three times with PBS. Finally, the round glass was placed on the slide, and observed using a confocal laser scanning microscope (CLSM). Besides, the cells were collected and the fluorescence intensity was quantified by flow cytometry.
Endosomal escape
To verify the endosomal escape ability of DSPE-PEG-R9, CT-26 cells (2 × 105cells/well) were seeded on confocal dish. After incubation for 24 h, the cells were incubated with DOX-Lip or DOX-R9-Lip for 0.5 h, 1 h, 2 h and 4 h. After washed with PBS, the cells were incubated with Lysotracker Green (70 nM) and Hoechst 33,258 for 30 min. The cells were observed under CLSM. The colocalization ratio of lysosome and liposome (Mander’s coefficients) was calculated by ImageJ [45, 46].
3D Tumor spheroids
Co-cultured cells (ratio of CT-26 to NIH 3T3 cells was 1:2, among which CT-26 2 × 103 cells/well) were seeded onto 96-well plates with 1% agarose. After seven days of culture, a 3D tumor model was generated. DOX-Lip, DOX-RGD-Lip and DOX-RGD/R9-Lip were incubated with tumor spheroids for 6 h. DOX fluorescence signals at different depths were scanned by CLSM.
Tumor penetration in vivo
The tumor-bearing mice model was established by subcutaneous injection of 0.1 mL CT-26/NIH 3T3 co-cultured cells (ratio of CT-26 to NIH 3T3 cells was 1:2, among which CT-26 1 × 107 cells/mL) at the right hind legs in mice. When the tumor grew to approximately 200 mm3, DOX-Lip, DOX-RGD-Lip and DOX-RGD/R9-Lip were injected intravenously at a DOX concentration of 3 mg/kg. After 12 h, the mice were sacrificed and the tumors were fixed with 4% paraformaldehyde. The different depth sections of tumor tissues were cryo-sectioned by freezing microtome at the thickness of 10 μm, then the nuclei were stained with DAPI and the sections were observed by CLSM.
In addition, a 3D fluorescence imaging system was applied to further verify the penetration of R9. In briefly, CT-26/NIH 3T3 co-cultured cells tumor-bearing mice models were administrated with IR-780-Lip, IR-780-RGD-Lip and IR-780-RGD/R9-Lip at the IR-780 concentration of 0.1 mg/mL. At 8, 12 and 24 h post administration, the fluorescence distribution at the tumor site was observed by 3D fluorescence imaging system.
In vivo animal imaging
BALB/c mice were subcutaneously injected with CT-26 cells (1 × 107 cells/mL) or CT-26/NIH 3T3 co-cultured cells. IR-780 was selected as the imaging tracker in vivo due to its strong absorption at around 780 nm, which was beneficial to monitor the distribution in the body in real time. After the tumors grew about 100 mm3, free IR-780, IR-780-Lip, IR-780-RGD-Lip and IR-780-RGD/R9-sLip were injected intravenously at the IR-780 concentration of 0.1 mg/mL. At different time intervals post administration, the fluorescence imaging was observed using the in vivo imaging system (IVIS). 24 h later, the mice were sacrificed to dissect the organs and tumors for further ex vivo imaging.
Moreover, orthotopic tumor model was first attempted to further evaluate the in vivo biodistribution. BABL/c mice were anesthetized with chloral hydrate (4%) and the abdomen was cut to expose cecum. CT-26 cells (1 × 106 cells suspended in 25μL of PBS/Matrigel, 1:1 v/v) were injected into the colon wall to establish orthotopic tumor model. After 10 days, free IR-780, IR-780-Lip, IR-780-RGD-Lip and IR-780-RGD/R9-sLip were injected intravenously at the IR-780 concentration of 0.1 mg/mL. At different time intervals post administration, the fluorescence imaging was observed using the in vivo imaging system (IVIS). 24 h later, the mice were sacrificed to dissect the organs and tumors for further ex vivo imaging.
In vivo antitumor effects
The antitumor effects were evaluated in both CT-26 cells (1 × 107 cells/mL) and CT-26/NIH 3T3 co-cultured cells tumor-bearing mice models, respectively. After the tumor grew to approximately 150 mm3, the mice were randomly divided into six groups (n = 5) and intravenously administrated with saline, free IRI, IRI-Lip, IRI-RGD-Lip, IRI-RGD/R9-Lip or IRI-RGD/R9-sLip every 2 days for 14 days (IRI was 20 mg/kg, scFv 0.334 mg/kg). The body weights and the tumor size were recorded every 2 days. Tumor volumes (V) were calculated as V = (tumor length × tumor width2)/2.
After treatment, the major organs were dissected from the sacrificed mice and the tumor weights were measured. Organs and tumors were fixed with 4% paraformaldehyde. All tissues were sectioned and embedded in paraffin, for hematoxylin and eosin (H&E) staining. Apoptosis of tumor tissues was detected by Colorimetric TUNEL Apoptosis Assay Kit. Ki-67 immunohistochemistry was performed according to the instructions to identify cell proliferation. Dewaxing and antigenic repair were performed on the sections. The tissue was blocked with bovine serum albumin (BSA). Subsequently, the expression of FAP or α-SMA was detected according to the instructions of rabbit polymer assay system. The above tissues were observed under microscope. The percentage of positive FAP or α-SMA area to the total area was quantified using ImageJ software.
Orthotopic tumor model was also attempted for further evaluation of in vivo antitumor effects. The mice were randomly divided into three groups (n = 3): (1) saline, (2) free IRI, (3) IRI-RGD/R9-sLip. After 7 times of administration, the mice were sacrificed and their colon tissues were collected for inhibitory evaluation. Afterwards, H&E staining was also carried out to explore the proliferative status of tumor.
Evaluation of lung metastasis
Colorectal cancer is closely associated with distant metastasis, especially to the liver and lungs. To build a lung metastasis model of colorectal cancer, intravenous injection of CT-26 cells is widely applied in many studies [47,48,49,50,51]. Lung metastasis model was built by intravenously injecting of 0.1 mL CT-26 cells (1 × 107 cells/mL) into the mice. All mice were randomly divided into 7 groups (n = 3) and the lungs of healthy mice served as negative control [47, 52]. One day later, mice with lung metastases were treated with Saline, free IRI, IRI-Lip, IRI-RGD-Lip, IRI-RGD/R9-Lip or IRI-RGD/R9-sLip (IRI 20 mg/kg, scFv 0.334 mg/kg). Changes in body weight of mice were detected during the treatment. After 7 times of administration, the mice were sacrificed and their lungs were collected for H&E staining and the numbers of lung nodules were recorded.
Statistical analysis
All the data were presented as the mean ± standard deviation (SD). The differences between two groups were evaluated by Student’s t test using GraphPad Prism 8.0. P < 0.05 was considered to be statistical significance.