Mice
Six-to-eight-week-old immunodeficient NOD-SCID female mice were purchased from the Model Animal Resource Information Platform of Nanjing University. Mice were maintained under specific pathogen-free facilities at Sichuan University. All procedures met the requirements of the National Institutes of Health and Institutional Animal Care and Use Committee. The animal experiments were approved by the West China Hospital of Sichuan University Biomedical Ethics Committee (Ethical approval document: 2018-061).
Tissue microarray and blood samples
Human tissue microarrays for immunohistochemistry (IHC) were purchased from Xi’an Alenabio and Shanghai Outdo Biotech of China. Blood samples were taken from healthy donors and were used for isolation of human peripheral blood mononuclear cells (PBMCs).
Cell lines and culture conditions
Human NSCLC cells A549 (which have the KRASQ61H mutation), H460 (which have the KRASG12S mutation), BC cells T24 (which have the HRASG12V mutation) and HEK293T cell lines were purchased from ATCC. A549, H460 and HEK293T cell lines were maintained in Dulbecco’s modified Eagle medium (Gibco) with 10% fetal bovine serum (Gibco) and 2 mmol/L L-glutamine. T24 cell line was maintained in McCoy’s 5A Medium (Gibco) with 10% fetal bovine serum and 2 mmol/L L-glutamine.
PBMCs from healthy donors were isolated using density gradient centrifugation and activated by culturing with anti-CD3 mAb (OKT3, 100 ng/mL, BioLegend), anti-CD28 mAb (CD28.2, 100 ng/mL, Sino Biological) and recombinant human interleukin-2 (IL-2) (100 units/mL, Life Science) in X-Vivo medium (Lonza) supplemented with 10% fetal bovine serum (heat inactivation at 56℃ for 30 minutes) and 2 mM L-glutamine and for 3 days.
Construction and production of B7-H3 × CD3 BiAb
The anti-B7-H3 single-chain variable fragment (scFv) sequence was derived from a highly specific monoclonal antibody against B7-H3 (clone mAb-J42) generated by our group using a standard hybridoma technique. cDNAs encoding the anti-B7-H3 scFv and anti-CD3 scFv (according to published amino acid sequences) were synthesized by commercial gene synthesis service (Genewiz). The two scFvs were linked by a G4S linker to construct a recombinant single-chain BiAb. The cDNAs were subcloned into an expression vector with a His tag at the C-terminal for protein purification.
HEK293T cells were transfected with the vector described above and cultured in the FreeStyle serum-free medium (Thermo Fisher Scientific) at 37℃, 5% CO2. After 7 days, culture supernatant was harvested and pre-cleaned by 0.45-µm filters. The BiAb was purified on Ni-NTA affinity columns and subsequently subjected to size exclusion chromatography. To assess the molecular mass of the BiAb, obtained samples were subjected to SDS-PAGE and stained with Coomassie brilliant blue.
Immunofluorescence staining
Cells were incubated in 24-well plates under standard cell culture conditions (5 × 103 cells per well). After 12 hours, cells were blocked with 5% BSA for 15 minutes, stained with B7-H3 antibody (Abcam, ab227679) for 1 hours, Cy3-conjugated secondary antibody (Beyotime, A0516) for 40 minutes and DAPI (Beyotime) in the dark. Images were captured on a fluorescence microscopy.
Tumor tissues from the T cell group mice were collected and immediately froze at -80 °C. Sections were fixed in pre-chilled acetone-methanol (1:1) for 20 minutes at -20 °C and then allowed to air-dry for 10 minutes before being blocked with 5% BSA for 30 minutes. Subsequently, Sections were stained with B7-H3 antibody (Abcam, ab227679) for 1 hour, FITC-conjugated secondary antibody (Beyotime, A0562) for 40 minutes and DAPI (Beyotime) in the dark. Images were captured on a fluorescence microscopy.
Flow cytometry
B7-H3 expression level on tumor cells was analyzed by flow cytometry. Cells were collected by centrifugation and incubated with the human B7-H3 antibody (BioLegend, 331605) for 20 minutes in the dark. After washing three times with PBS, the cells were resuspended in 500 µL and analyzed using a NovoCyte™ Flow Cytometer (ACEA Bioscience) according to the manufacturer’s protocols. For T cell phenotype analyses, human CD4 (BioLegend, 357419), CD8 (BioLegend, 344729), CD25 (BioLegend, 302629) and CD69 (BioLegend, 310909) antibodies were used and experiments were performed on a Fortessa flow cytometer (BD).
For apoptosis detection, Annexin V staining was measured by FITC-annexin-V Apoptosis Detection Kit I (4A Biotech). A549 and H460 cells (5 × 105) were treated for 48 hours with 10 µM trametinib (MCE). Cells were collected and resuspended in 1 × Binding Buffer, 100 µL solution (1 × 105 cells) was used to stain cells with 5 µL FITC annexin V for 15 minutes in the dark followed by the addition of 0.4 mL of 1 × Binding Buffer and 10 µL 7-AAD. Flow cytometry analysis was performed on a NovoCyte™ Flow Cytometer (ACEA Bioscience) according to the manufacturer’s protocols.
For T cell proliferation assay, T cells were initially stained with carboxy fluorescein succinimidyl ester (CFSE) (Beyotime) and cultured in the presence or absence of 1 µM trametinib. After 48 hours, cell proliferation was carried out by flow cytometry.
Western blotting
Total proteins were extracted from cells after treatment with the indicated concentrations of trametinib for 48 hours and quantified by BCA protein assay kit (Beyotime). Then, equal amount of proteins (10 µg) was subjected to SDS-PAGE and transferred to polyvinylidene difluoride membranes. After that, the membranes were blocked with 5% milk for 1 hour. Subsequently, the membranes were stained with B7-H3 (CST, 14058S), P-MEK1/2 (CST, 3958S), P-ERK1/2 (CST 4370S) and β-actin (ZSGB-BIO, TA09) antibodies for 1 hour, HRP-conjugated secondary antibody (Beyotime, A0208) for 1 hour. Images were captured by a ChemiScope 6000 Touch (Clinx).
Cell viability assay
Cells were seeded in 96-well plates and incubated overnight prior to treatment. After 48 hours with the indicated concentrations of trametinib, 10 µL of the Cell Counting Kit-8 solution (Beyotime) was added to each well and incubated for 2 hours in the dark. Absorbance at 450 nm was measured in a microplate reader.
Cytotoxicity Assays
A 2D and 3D coculture models of tumor cells with human T cells were used to assess the cytotoxicity. In the 2D coculture model, A549, H460 or T24 cells were co-cultured with T cells at an E:T ratio of 1:4, 1:1 and 4:1, together with 1 µM trametinib alone or in combination with 5 µg/mL BiAb. Images were captured at 12 and 24 hours. To assess the effect of cytotoxicity, Chromium-51 assay was carried out as described [14]. Tumor cells were labeled with sodium chromate (molecular formula, Na251CrO4) and incubated with T cells at an E:T ratio of 1:4, 1:1 and 4:1 for 4 h. Then the radioactivity of the supernatants was measured by a gamma counter. The percentage of specific lysis was calculated by the formula: (test release-spontaneous release)/(maximal release-spontaneous release) × 100.
For the 3D spheroid model, the method was performed according to our previous description [37]. In brief, 1 × 105 cells were added to the Matrigel-coated wells and cultured in serum-free DMEM (Gibco) with 2% B-27 supplement (Gibco), 20 ng/ml human EGF (Sino Biological) and 20 ng/ml human bFGF (Sino Biological). After 5 days, human T cells were stained with CFSE (Beyotime) and added to the wells at the E:T ratio of 1:1, together with 5 µg/mL BiAb alone or in combination with 1 µM trametinib for 12 hours. Cells were stained with DAPI (Beyotime) in the dark and images were captured on a fluorescence microscopy.
Analysis of Cytokine Secretion
Tumor cells were coculture with T cells alone or together with 5 µg/ml BiAb in 24-well plates at different E:T ratio with. After 24 h, the supernatant was collected to evaluate the IFN-γ secretion by ELISA kits (BioLegend) according to the manufacturer’s protocols.
In vivo experiments
In the H460 and T24 xenograft experiments, 2 × 106 H460 or T24 cells were subcutaneously injected into NOD-SCID mice and were randomly divided into four groups consisted of n = 5 per group. From the tenth day on, trametinib (0.6 mg/kg) or vehicle control was administered for 10 consecutive days via oral gavage. On day 13, all mice were intravenously treated with 8 × 106 T cells and from the day on, mice were intravenously treated with 100U IL-2 or in combination with 2 mg/kg BiAb or PBS for seven consecutive days. The mice in the combination treatment group received both trametinib and the BiAb at the above doses and schedule. The vehicle control of trametinib was a mixture of 30% PEG400, 0.5% Tween80 and 5% propylene glycol. Bodyweight and tumor sizes were measured every three days. The tumor volume was calculated using the following equation: (length × width × width)/2.
IHC assay
Tumor, Heart, liver, spleen, lung and kidney sections from mice were preprocessed by paraformaldehyde and embedded in paraffin. After slicing into sections, slides were performed with H&E staining. Tumor paraffin sections were immunostained with CD3 antibody (Servicebio, GB13014). All procedures followed the manufacturer’s protocol. In brief, tissue sections were incubated at 65 °C for 1 h to retrieve antigenicity, blocked with PBS containing 10% normal goat serum for 30 min at room temperature, and then incubated with primary antibody at 4 ℃ overnight. The sections were then incubated with secondary antibodies, and the staining was detected with 3,3’diaminobenzidine (ZSGB-Bio).
Statistical analysis
All in vitro results were representative of three separate experiments. Data were presented as the mean ± standard deviation in figure legends. Statistical analyses were performed using GraphPad Prism 7.0. The difference between various experimental and control groups was examined by Student′s t-test and considered significant at *, P < 0.05; **, P < 0.01; ***, P < 0.001. For bioinformatic analysis of B7-H3, the relationship between B7-H3 expression and prognosis was performed using the dataset of the Kaplan-Meier Plotter (KM Plotter) [38]. The meta-analysis and mRNA expression of B7-H3 in tumor and normal tissues was analyzed by using the Oncomine [39]. The association of B7-H3 expression and the tumor stage were examined by data mining in Oncomine or The Cancer Genome Atlas (TCGA).