Patient cohort
The current study selected ovarian cancer patients from June 2018 to November 2021 based on the pre-defined inclusion and exclusion criteria. Inclusion criteria were: (1) recurrent ovarian cancer; (2) surgery and platinum-based chemotherapy as the initial treatment or platinum-based chemotherapy alone; (3) PARP inhibitors as maintenance therapy. Exclusion criteria were: (1) disease progression within 6 months of remission with platinum-based chemotherapy; (2) no platinum-based chemotherapy; (3) maintenance therapy with a combination of PARP inhibitors and other targeted agents. Progression-free survival (PFS) was the time starting from the date of ovarian cancer diagnosis to the date of disease recurrence or progression in patients with recurrent ovarian cancer or discontinuation of follow-up.
Cell culture
The human BRCA2 mutated ovarian cancer PEO1 and wild-type BRCA ovarian cancer OVCAR3, SKOV3, and A2780 cell lines, as well as the mouse-derived ovarian cancer ID8 cell line were obtained from the Cancer Institute, Central South University. PEO1 and ID8 cells were cultured in DMEM High-Glucose medium (Gibco, Life Technologies, Eugene, OR, USA) containing 10% FBS (Menlo Park, CA, USA), 100 μg/mL penicillin, and 100 U/mL streptomycin. OVCAR3, SKOV3, and A2780 cells were cultured in RPMI 1640 (Gibco, Life Technologies) containing 10% FBS, 100 μg/mL penicillin, and 100 U/mL streptomycin. All cell lines were cultured at 37˚C with 5% CO2.
CCK8 test
Ovarian cancer cells were seeded into 96-well plates at 5×103/well and cultured for 24 hours. PEO1, OVCAR3, SKOV3, and A2780 cells were treated with vehicle control, niraparib, P4 plus niraparib (at concentration ratios of 1:10, 1:6, 1:4, 1:2, 1:1, and 1:0.5, respectively) or P4 for 48 hours. Then, 10 µl of the CCK8 reagent (KOO9-100; Zeta-Life) was added, and cells were incubated at 37˚C with 5% CO2 in the dark for 2 h. Optical density was measured at 450 nm on a spectrophotometer. Based on the manufacturer’s instructions, combination indexes (CI) at indicated fraction affected (FA) levels were calculated with the CompuSyn software by the Chou-Talalay method with nonconstant-ratio combinations. IC50 values were calculated from optical density values. CI values were between 0.9 and 1.1 (additive effect), between 0.8 and 0.9 (low synergistic effect), between 0.6 and 0.8 (moderate synergistic effect), between 0.4 and 0.6 (high synergistic effect), and between 0.2 and 0.4 (strong synergistic effect), respectively.
Colony formation assay
PEO1 and OVCAR3 cells were seeded into a 6 cm-dish at 1×103/dish and cultured for 24 h. Then, the cells were administered niraparib (10 µM), P4 (10 µM), niraparib + P4 or vehicle control for 48 h. After formation, cell colonies were fixed with 95% alcohol for 30 min, stained with 1% crystal violet for 1.5 h and washed with water. The colony formation rate was determined after drying.
Wound healing assay
PEO1, OVCAR3, SKOV3, and A2780 cells were seeded into 6-well plates at 5×105/well. When the cells were ~90% confluent, using a sterile 10 µl pipette tip, the middle of the well was scratched. Next, the cells were administered niraparib (10 µM), P4 (10 µM), niraparib + P4 or vehicle control for 48 h, and cultured in DMEM High-Glucose medium or RPMI1640 containing 5% FBS. Images were acquired at 0, 24 h, and 48 h after drug administration, and the healing area on the scratch was calculated with the Image J software.
Transwell migration assay
PEO1, OVCAR3, SKOV3, and A2780 cells were seeded into a Transwell chamber at 5×105/well, in 160 µL of DMEM High-Glucose medium or RPMI 1640 without serum. The chambers were placed on a 24-well plate containing 600 µL/well of DMEM High-Glucose medium or RPMI 1640 with 10% serum. After 6 h of culture, the cells were administered niraparib (10 µM), P4 (10 µM), niraparib + P4 or vehicle control for 48 h. Then, the membranes with cells were fixed with 4% paraformaldehyde for 40 min and stained with 0.1% crystal violet solution for 20 min. Cells on the inner membrane were wiped with a cotton swab, and the chamber was washed with PBS to remove the excess staining solution. After drying, the chambers were imaged under a fluorescent inverted microscope, and the numbers of migrated cells were determined.
Flow cytometry
Cells apoptosis and cell cycle distribution in PEO1, OVCAR3, SKOV3, and A2780 cells were detected by flow cytometry. Cells seeded at 1×106/well into 6-well plates were administered niraparib (10 µM), P4 (10 µM), niraparib + P4 or vehicle control for 24 h. Cells were collected by trypsinization and centrifugation. The Annexin V/PI double-staining kit (cat. no. KGAV113; Nanjing KeyGen Biotech Co., Ltd., Jiangsu, China) was used to determine the proportion of apoptotic cells. For cell cycle analysis, cells were washed with pre-cold PBS and fixed in cold 90% ethanol at 4℃ overnight. Then, the cells were washed with pre-chilled PBS, and the cell suspension was incubated with propidium iodide (PI) solution for 30 min and analyzed by flow cytometer.
Immunofluorescence
PEO1, OVCAR3, SKOV3, and A2780 cell slides were treated with niraparib (10 µM), P4 (10 µM), niraparib + P4 or vehicle control for 24 h. The cell slides were washed with PBS, fixed with 4% paraformaldehyde, and permeabilized with 0.1% TritonX-100 in PBS. The samples were incubated with primary antibodies targeting γH2AX (ab81299; 1:200; Abcam, Cambridge, MA, USA), SCD1 (ab19862; 1:200; Abcam), and PR (ab2765; 1:200; Abcam), respectively, overnight at 4℃. This was followed by incubation with fluorescent-labeled secondary antibodies (A11034; 1:1000; Invitrogen, Eugene, OR, USA) in the dark for 1 h. Then, counterstaining was performed with DAPI, before analysis under a confocal laser scanning fluorescence microscope.
Immunohistochemistry and immunofluorescence
Paraffin sections from ovarian cancer tissue and mouse tumor tissue samples were deparaffinized with xylene and hydrated with different concentrations of alcohol. Antigen retrieval was performed after heating in citrate buffer (Beyotime Biotechnology, Shanghai, China). The tissue samples were incubated at room temperature with an endogenous peroxidase blocker (CWBIO, Jiangsu, China). This was followed by overnight incubation at 4℃ with primary antibodies against Ki67 (ab6667; 1:200; Abcam), Caspase-1 (ab138483; 1:200; Abcam), PR (ab2765; 1:200; Abcam), or SCD1 (ab19862; 1:200; Abcam). For immunohistochemistry, the tissue sections were incubated with enhanced enzyme-labeled goat anti-mouse/rabbit IgG (AWS0003a/AWS0002a; 1:1000; Abiowell) at room temperature for 20 min, with subsequent staining with the DAB kit (CWBIO). For immunofluorescence, fluorescent-labeled secondary antibodies (A11034; diluted 1:1000 with BSA solution; Invitrogen) were added to the tissue samples, followed by incubation for 2 h at room temperature in the dark. After a PBS wash, 0.5 µg/mL DAPI 5% BSA solution was added (C1005; Beyotime Biotechnology) for staining and an appropriate amount of anti-fluorescence attenuation dose was added and mounted.
Animal experiments
Mouse ovarian in situ tumorigenesis model and intraperitoneal tumorigenesis experiment
Female C57BL/6 and BALB-nude mice at 6-7 weeks of age were purchased from SLA Laboratory. Female C57BL/6 mice were intraperitoneally injected with 1×105/mL ID8 cells, and female BALB-nude mice were injected 2×105/mL OVCAR3 cells into ovarian-tubal intrabursally or peritoneally. Then the mice were divided into 4 groups, including Vehicle, P4 (5 mg/kg P4 by intramuscular injection), Niraparib (50 mg/kg niraparib by gavage), and P4 plus Niraparib (5 mg/kg progesterone and 50 mg/kg niraparib) groups. Mice were continuously treated 3 times per week, and animal body weights and ascites were observed. Mice were sacrificed after 4 weeks of treatment, and tumor sizes, volumes, and tumor invasion and metastasis were recorded. Mouse tumors were fixed with 10% formalin, embedded in paraffin, and cut into 3 µm sections. Survival analysis of tumor-bearing mice after treatment with control, P4, niraparib, and P4 plus niraparib based on the methods mentioned above was also performed.
Quantitative real-time PCR (qRT-PCR)
Total RNA was extracted from OVCAR3, SKOV3, A2780, and PEO1 cells with TRIzol reagent (Ambion Inc., Austin, TX, USA). Total RNA was reverse transcribed into complementary DNA (cDNA) with PrimeScript RT reagent Kit with gDNA Eraser (TaKaRa Biotechnology, Dalian, China), following the manufacturer’s instructions. TB Green Premix Ex Taq II (TaKaRa Biotechnology) was used for qRT-PCR on a LightCycle 480 II instrument. All primers were synthesized by Sangon Biotechnology Company (Shanghai, China), and are listed in Supplementary Table 1. The relative mRNA expression was calculated by the 2-ΔΔct method.
Western blot
Proteins from OVCAR3, SKOV3, A2780 and PEO1 cells were extracted with RIPA lysis buffer, and protein concentrations were determined with the BCA kit. Each sample was loaded at 30 µg, subjected to SDS-PAGE electrophoresis and transferred onto PVDF membranes (Millipore, Burlington, MA, USA). PVDF membranes were blocked with 5% skimmed milk (Biosharp, Hefei, Anhui, China) for 2 h and incubated overnight at 4℃ with primary antibodies against PR, H2AX, and SCD1. Bound antibodies were then detected with HRP-conjugated secondary antibodies (AWS0003a; 1:1000; Abiowell). The ECL Western blotting substrate (Advansta, Munich, Germany) was used for visualization.
Statistical analysis
All data were obtained from three independent replicates, and the results were expressed as mean ± standard deviation (SD). Statistical analysis was performed with SPSS version 18.0 or GraphPad. Two-sample t-test or non-parametric test was used to compare group pairs. The Chi-square test was used for categorical data. Kaplan-Meier analysis was performed to assess the prognosis of ovarian cancer patients, and the log-rank test was performed to compare survival. P<0.05 was considered statistically significant.