Abstract Background The cellular tumor protein p53 (TP53) is a tumor suppressor gene that is frequently mutated in human cancers. Among various cancer types, the very aggressive high‐grade serous ovarian carcinoma (HGSOC) exhibits the highest prevalence of TP53 mutations, present in >96% of cases. Despite intensive efforts to reactivate p53, no clinical drug has been approved to rescue p53 function. In this study, our primary objective was to administer in vitro‐transcribed (IVT) wild‐type (WT) p53‐mRNA to HGSOC cell lines, primary cells, and orthotopic mouse models, with the aim of exploring its impact on inhibiting tumor growth and dissemination, both in vitro and in vivo. Methods To restore the activity of p53, WT p53 was exogenously expressed in HGSOC cell lines using a mammalian vector system. Moreover, IVT WT p53 mRNA was delivered into different HGSOC model systems (primary cells and patient‐derived organoids) using liposomes and studied for proliferation, cell cycle progression, apoptosis, colony formation, and chromosomal instability. Transcriptomic alterations induced by p53 mRNA were analyzed using RNA sequencing in OVCAR‐8 and primary HGSOC cells, followed by ingenuity pathway analysis. In vivo effects on tumor growth and metastasis were studied using orthotopic xenografts and metastatic intraperitoneal mouse models. Results Reactivation of the TP53 tumor suppressor gene was explored in different HGSOC model systems using newly designed IVT mRNA‐based methods. The introduction of WT p53 mRNA triggered dose‐dependent apoptosis, cell cycle arrest, and potent long‐lasting inhibition of HGSOC cell proliferation. Transcriptome analysis of OVCAR‐8 cells upon mRNA‐based p53 reactivation revealed significant alterations in gene expression related to p53 signaling, such as apoptosis, cell cycle regulation, and DNA damage. Restoring p53 function concurrently reduces chromosomal instability within the HGSOC cells, underscoring its crucial contribution in safeguarding genomic integrity by moderating the baseline occurrence of double‐strand breaks arising from replication stress. Furthermore, in various mouse models, treatment with p53 mRNA reduced tumor growth and inhibited tumor cell dissemination in the peritoneal cavity in a dose‐dependent manner. Conclusions The IVT mRNA‐based reactivation of p53 holds promise as a potential therapeutic strategy for HGSOC, providing valuable insights into the molecular mechanisms underlying p53 function and its relevance in ovarian cancer treatment. Keywords: apoptosis, cell cycle, chromosomal instability, HGSOC metastatic intraperitoneal mouse model, HGSOC orthotopic Xenograft model, high‐grade serous ovarian cancer, in vitro‐transcribed p53‐mRNA, liposomal IVT mRNA delivery system, patient‐derived organoid, patient‐derived primary cancer cell __________________________________________________________________ List of abbreviations APC Adenomatous polyposis coli protein BAK B‐cell lymphoma 2 (BCL‐2) antagonist/killer BAX BCL‐2 associated X‐protein BCL‐2 B‐cell lymphoma 2 Bid BH3 interacting domain death agonist Bub1 Mitotic checkpoint serine/threonine kinase BUB1B BUB1 mitotic checkpoint serine/threonine kinase B CDC20 Cell division protein 20 homolog CDK1 Cyclin‐dependent Kinase 1 EGFP Enhanced Green Fluorescence Protein FAS Tumor necrosis factor receptor superfamily member 6 GSK3β Glycogen synthase kinase 3β H2AX H2A histone family member X HGSOC high‐grade serous ovarian cancer IVT in vitro transcription MDM2 Double minute 2 homolog protein MYC Master regulator of cell cycle entry and proliferative metabolism Noxa NADPH oxidase activator 1 p14ARF ARF tumor suppressor p16 inhibitor of cyclin‐dependent kinase 4 (CDK4) and CDK6 P21/(CDKN1A) Cyclin‐dependent kinase inhibitor 1 p27 Inhibitor of cyclin‐dependent kinase 4 (CDK4) and CDK6 PARP Poly (ADP‐ribose) polymerase PLK1 Polo‐like Kinase 1 PLK3 Polo‐like Kinase 3 Puma The p53 upregulated modulator of apoptosis TP53 Tumor Protein p53 XIAP X‐linked inhibitor of apoptosis 1. BACKGROUND High‐grade serous ovarian cancer (HGSOC) is the most lethal gynecological malignancy. It interacts intensively with the tumor stroma, grows rapidly, metastasizes extensively, and shows an aggressive course [[42]1, [43]2]. Ovarian cancer (OC) cells remain within the peritoneal cavity and disseminate only to the mesothelium‐lined surface [[44]3]. The current standard of care includes cytoreductive surgery, chemotherapy (platinum compounds, paclitaxel [[45]4], and cyclophosphamide [[46]5]), bevacizumab (avastin) [[47]6], and poly‐ADP‐ribose polymerase (PARP) inhibitors [[48]7]. Nevertheless, approximately 70% of patients experience relapse within 3 years following surgery and platinum‐based chemotherapy and succumb to disease progression [[49]8]. The tumor protein p53 (TP53) gene is the most frequently mutated human tumor suppressor gene, mostly through point mutations, whereby amino acid substitutions lead to the disruption of tumor protein p53 binding to DNA [[50]9, [51]10, [52]11]. Thus, p53 protects the human genome by counteracting cellular stress and DNA damage [[53]12]. Multiple reports have shown that tumor cells harboring functionally inactive p53 are resistant to chemotherapeutics because their mechanism of action involves DNA damage, leading to the activation of wild‐type (WT) p53. The Cancer Genome Atlas Research Network revealed that HGSOC is characterized by inactive or truncated TP53 in up to 96% of all cases [[54]13]. Moreover, mutations in TP53 seem to be early events in tumorigenesis, likely in OC precursor lesions, supporting the importance of mutated TP53 as a driver of this malignancy [[55]14]. Approaches aimed at re‐establishing the WT function of p53 have the unique ability to improve outcomes in patients with many types of cancer. Several small molecules have been developed to counteract the loss of p53 function, mostly by stabilizing the native conformation of p53 [[56]15, [57]16], like PRIMA‐1, which reactivates mutant p53, induces apoptosis in cancer cells [[58]15, [59]17], and is currently in clinical trials (ClinicalTrials.gov). In our laboratory, silencing of Cyclin B1 upregulated the ARF tumor suppressor, p14ARF, which inhibits mouse double minute 2 homolog protein (MDM2) and reduces p53‐S315 phosphorylation by Cyclin‐dependent kinase 1 (CDK1)/Cyclin B1, thereby modulating two mechanisms that stabilize and activate p53 [[60]18]. Other small molecules have been developed to reinstate specific p53 functions [[61]15, [62]19, [63]20], or nonsense mutations by promoting transcriptional readthrough [[64]21]. Nevertheless, different studies have provided evidence of off‐target effects associated with small molecules used for p53 reactivation [[65]22, [66]23, [67]24]. Thus, whether off‐target activities or restoration of p53 function are the origin of therapeutic responses is currently being investigated. With benefits such as scalable manufacturing, few adverse effects, and high efficiency [[68]25, [69]26], novel strategies based on mRNA therapeutics have emerged as effective and safe approaches for fighting predominantly infectious diseases. Although mRNA therapies for protein replacement are gaining attention in several clinical settings, their use in cancer remains limited [[70]27, [71]28]. To explore whether mutant TP53‐bearing HGSOC cells respond to p53 rescue, we tested an in vitro transcribed (IVT), codon‐optimized p53‐mRNA in human and murine models and achieved robust and persistent protein expression using diverse IVT‐mRNAs. We aimed to investigate the impact of prolonged p53‐mRNA treatment on the induction of cell cycle arrest, apoptosis, and chromosomal stability in HGSOC, a tumor type characterized by TP53 loss and chromosomal instability [[72]29]. Finally, we assessed the ability of p53‐mRNA to inhibit tumor cell proliferation and dissemination in xenograft models, highlighting its potential for HGSOC therapy. 2. MATERIALS AND METHODS 2.1. Generation of mRNA Plasmid templates containing open reading frames of Enhanced Green Fluorescent Protein (EGFP), luciferase, or Flag‐p53 were amplified using PCR, restoring functionality to the T7 promoter sequence and adding a 120A tail via the reverse primer. The primer pair used for cloning TP53 into Flag vector are: 5’‐ATGGAGGAGCCGCAGTCAGATCC‐3’, forward primer; 5’‐TCAGTCTGAGTCAGGCCCTTCTG‐3’, reverse primer. These constructs were capped (Cap 1) using CleanCap® AG (Tebu‐Bio, Offenbach, Hessen, Germany) in a reaction where uridine‐5’‐triphosphate was fully substituted with Pseudo‐U. After the IVT, DNase (Tebu‐Bio, Offenbach, Hessen, Germany) was used to remove any residual DNA template from the reaction. Thorough purification of mRNAs included the use of DNase to degrade the DNA template and a spin column (silica membrane; Tebu‐Bio, Offenbach, Hessen, Germany) to remove the digested template, excess nucleoside triphosphates (NTPs), salts, and unused capping analogs. A Qiagen RNeasy (silica membrane) spin column was used to remove the digested template, excess NTPs, salts, and unused capping analogs. Antarctic phosphatase (Tebu‐Bio, Offenbach, Hessen, Germany) was then used to cleave residual immunogenic 5′ triphosphate from any uncapped material. A second RNeasy spin column was used to remove phosphatase from the reaction. Purified mRNA was purchased from TriLink (Tebu‐Bio). 2.2. Patients and tissue samples This study was conducted according to the “REporting recommendations for tumor MARKer prognostic studies” [[73]30]. Following the acquisition of informed consent, samples of ovarian carcinoma were obtained from individuals diagnosed with epithelial ovarian cancer at the Department of Gynecology at Goethe University Hospital in Frankfurt am Main, Germany from January 2015 to December 2022. The validation of these samples involved assessment by 3 pathologists, with exclusion criteria applied to patients with alternative tumor types. To establish primary patient‐derived OC cell cultures, we analyzed samples from 8 patients who underwent surgical resection (Supplementary Table [74]S1). Sufficient archival materials for cell culture analyses were available for samples with validated diagnoses. The local research ethics committee approved the studies on human tissue (permit number: SGO‐1‐2017), and the samples were processed anonymously. The assessment of progression‐free survival (PFS) extended from the surgical procedure to the point of disease relapse or progression. Cases with a PFS duration of fewer than 6 months were designated as chemoresistant, whereas durations surpassing this threshold were characterized as chemosensitive. 2.3. Cell lines, primary cells, and transfection Human ovarian cancer cell lines SKOV3, OVCAR‐3, OVCAR‐4, OVCAR‐5, and OVCAR‐8 were purchased from the DCTD Tumor Repository (Bethesda, MD, USA), maintained in RPMI 1640 medium (Thermo Fisher Scientific, Dreieich, Hessen, Germany), containing 10% fetal bovine serum (FBS; Thermo Fisher Scientific) and 1% penicillin‐streptomycin (#17060063, Thermo Fisher Scientific). All cell lines were cultured in a humidified incubator of 5% CO[2] at 37°C. Primary cells were isolated from OC tissues derived from in‐house surgery using a tumor dissociation (Max Miltenyi 130‐095‐929, Bergisch Gladbach, NRW Germany) and tumor cell isolation (Max Miltenyi 130‐108‐339) kits, according to the manufacturer's instructions. Briefly, the samples were stored in a solution comprising Advanced DMEM/F12 (Thermo Fisher Scientific), 2 mmol/L HEPES (Thermo Fisher Scientific), 1 × GlutaMAX‐I (Thermo Fisher Scientific), and 200 U/mL penicillin/streptomycin (Thermo Fisher Scientific). Upon arrival at the laboratory, the tissue specimens were put in a sterile petri dish on ice. Following this, the tumor was cut into a sterile 10 mm square. A section of the dissected tissue was stored at −80°C in 2 mL tubes using Recovery Cell Culture Freezing Medium (Thermo Fisher Scientific) for subsequent utilization. In the process of preparing organoids, tissue samples were collected in 50 mL tubes with HBSS 1 × (Thermo Fisher Scientific) and incubated on ice for of 5 minutes. We performed DNA transfections using jetPEI Transfection Reagent (Polyplus, Illkirch, Bas‐rhin, France) or LTX (Invitrogen, Waltham, MA, USA) (1 μg IVT mRNA: 2 μL Lipofectamine Messenger MAX Transfection Reagent [Invitrogen]), according to the previously described [[75]31]. mRNA transfections were performed using the Lipofectamine Messenger MAX Transfection Reagent (Invitrogen), according to the manufacturer's instructions. Ovarian cancer cells were obtained by cell scraper and then lysed in radioimmunoprecipitation assay, RIPA buffer (Waltham, MA, USA). After centrifuging for 5 min at 15,000 ×g at 4°C, cytoplasmic protein was collected in the supernatant. 2.4. Colony formation assay Two thousand cells were seeded into 6‐well plates. The transfected cells were washed and incubated in fresh medium for 2 weeks. The colonies were fixed using 70% ethanol and stained with 0.5 μg Coomassie Brilliant Blue (Biorad, Feldkirchen, Bavaria, Germany). The number of grown colonies was counted, and images were captured using an AxioObserver Z1 microscope (Zeiss, Göttingen, Lower Saxony, Germany) and ChemiDoc MP system (BioRad, Hercules, CA, USA). 2.5. Organoids Following the in‐house surgery, tissues were collected in a storage medium (Dulbecco's Modified Eagle's Medium [DMEM; Thermo Fisher Scientific, 1% penicillin‐streptomycin [Thermo Fisher Scientific]) on ice and processed immediately. Fresh tissues were cut into small pieces (2‐4 mm) and rinsed 3 times with wash medium (500 mL DMEM, 1% penicillin‐streptomycin, and 2 g bovine serum albumin [BSA; Thermo Fisher Scientific]). Subsequently, the sample was left on ice for 2 min to settle, and the supernatant was removed. Finally, the tissue was kept in 5‐20 mL digestion medium (20 mL Advanced DMEM, 20 g BSA, and 10 μmol/L Y‐27632), depending on the sample size, for up to 30 min at 37°C. Every 5‐10 min, the sample was shaken and the stage of digestion was examined under a microscope. The digestion process was terminated with an equal volume of wash medium, as soon as the tissue was digested into single cells or small cell clusters. After that, the cell suspension was filtered through a 100 μm‐filter and spun down at 200 × g for 5 min at 4°C, and the supernatant was then removed. Red blood cells as components of the pellet were lysed with ammonium‐chloride‐potassium buffer (Red lysis buffer: 0.15 M NH[4]Cl, 10 mmol/L KHCO[3], and 0.1 mmol/L ethylenediaminetetraacetic acid) for 2‐3 min at room temperature and transferred to an Eppendorf tube. After a second wash step (200 × g, 5 min, 4°C), the pellet was resuspended in a wash medium (#356231, Corning, Kaiserslautern, NRW, Germany) and mixed with a triple volume Base Membrane Extract (BME; cat. # BME 001‐05, Cultrex UltiMatrix Reduced Growth Factor Basement Membrane Extract, R+D Systems, Wiesbaden, Hessen, Germany), or Corning Matrigel. Finally, 3D drops of the mixture (10‐20 μL) were seeded on a 12‐well suspension plate. The plate was flipped upside down and incubated in a cell culture incubator for 30 min at 37°C, 5% CO[2]. Finally, 1 mL growth medium (38.65 mL Advanced DMEM/F12 [Thermo Fischer, Dreieich, Hessen, Germany], 10 μmol/L Y‐27632 [Biozol, Eching, Bavaria, Germany], 0.25 μmol/L A83‐01 [Tocris, Bristol, Somerset, UK], 1 mL 1 × B27 supplement, 5 mmol/L Nicotinamide [Sigma, Frankfurt, Hessen, Germany], 1.25 mmol/L N‐acetycysteine [Sigma], 50 ng/mL human Epidermal Growth Factor [Peprotech, Cranburry, NJ, USA), 50 ng/mL Neuregulin [Peprotech], 5 ng/mL FGF‐Basic [Peprotech, Cranburry, NJ, USA], 10 ng/mL hFGF‐10 [Peprotech], 200 ng/mL primocin [InvivoGen, San Diego, CA, USA], 50 ng/mL R‐Spondin [Peprotech], and 100 ng/mL Noggin [R&D system, Mineapolis, MN, USA]), were added to each well. The medium was replaced twice weekly. The organoids were passaged according to their size and density. 2.6. Western blot (WB) and antibodies Cell protein extracts were prepared by lysis in the RIPA buffer (Sigma) supplemented with protease inhibitors (complete protease inhibitor cocktail, Roche, Mannheim, Rheinland Pfalz Germany). Protein extracts (25 μg) were separated using sodium dodecyl sulfate‐polyacrylamide gel electrophoresis and transferred onto polyvinylidene difluoride membranes using a TransBlot Turbo Transfer System (Bio‐Rad, Feldkirchen, Bavaria, Germany). The membranes were blocked with tris‐buffered saline with 0.05% Tween^® 20 (Sigma, Frankfurt, Hesen, Germany) containing 2% BSA Thermo Fisher Scientific. The following antibodies were used at the indicated concentrations: mouse monoclonal Plk1 (F‐8:sc‐17783) (1:1000; Santa Cruz, Biotechnology Heidelberg, Germany), GFP (B‐2) (1:1000; Santa Cruz), Cyclin A (H‐432) (1:1000; Santa Cruz), CDK1 (sc‐54) (1:1000; Santa Cruz), KIF4A (sc‐365144) (1:1000; Santa Cruz), c‐Myc (sc‐764) (1:1000; Santa Cruz), p53 (sc‐126) (1:1000; Santa Cruz), p73 (sc‐17823) (1:1000; Santa Cruz), MDM2 (sc‐965) (1:1000; Santa Cruz), H‐Ras (sc‐30) (1:1000; Santa Cruz), Luciferase‐HRP (sc‐57604) (1:100; Santa Cruz), K‐Ras (sc‐30) (1:1000; Santa Cruz), CDK4 (sc‐260; Santa Cruz), BRCA‐2 (sc‐293185) (1:1000; Santa Cruz), β‐Actin (AC‐15:A1978) (1:200; Sigma‐Aldrich, Taufkirchen, Germany), Flag (M2:A8592) (1:1000; Sigma‐Aldrich), PLK1 (35‐206, #05‐844) (1:1000; Millipore, Schwalbach, Germany), Cyclin B1 (#4138) (1:1000; Millipore), Aurora B (#3094) (1:1000; Millipore), GAPDH (ab 9485) (1:1000; Abcam, Cambridge, Cambridgeshire, England), Bak (ab 32371) (1:1000; Abcam), XIAP (ab 28151) (1:1000; Abcam), Aurora A (#14475) (1:1000; Cell Signaling, Frankfurt, Hessen, Germany), Cyclin E (#4129) (1:1000; Cell Signaling), p53‐pS20 (#9287) (1:1000; Cell Signaling), p21 (#2947) (1:1000; Cell Signaling), p27 (#3686) (1:1000; Cell Signaling), p16 (#18769) (1:1000; Cell Signaling), PARP (#9542) (1:1000; Cell Signaling), Casp.3 (#9665) (1:1000; Cell Signaling), Puma (#98672) (1:1000; Cell Signaling), Noxa (#14766) (1:1000; Cell Signaling), Fas (#8023) (1:1000; Cell Signaling), TOP2A (#12286) (1:1000; Cell Signaling), BIRC5 (#2802) (1:1000; Cell Signaling), Nup98 (#2598) (1:1000; Cell Signaling), BID (#2002) (1:1000; Cell Signaling), PLK3 (#4896) (1:1000; Cell Signaling), Casp.9 (#65832) (1:1000; Cell Signaling), Bub1 (#4116) (1:1000; Cell Signaling), cdc25c (#4688) (1:1000; Cell Signaling), AKT3 (#14982) (1:1000; Cell Signaling), Cyclin D (#55506) (1:1000; Cell Signaling), GSKB (#12456) (1:1000; Cell Signaling), pGSK3B(Ser9) (#5558) (1:1000; Cell Signaling), β‐Catenin (#8480) (1:1000; Cell Signaling), p‐β‐Catenin(S33/37/T41) (#9561) (1:1000; Cell Signaling), Bub1B (A300‐386A) (1:1000; Biomol GmbH, Hamburg, Germany), cdc20 (Ls‐C357796) (1:1000; Biozol), Bcl‐2 (#610538) (1:1000; BD Biosciences, Heidelberg, Germany), Bax (AF 820) (1:1000; R&D, Abingdon, Oxfordshire, UK), and HRP‐conjugated secondary antibodies (1:5000; GE Healthcare and Jackson Laboratory, Bar Harbor, ME, USA. The ECL WB Substrate (Millipore) was used for detection. 2.7. Cell cycle, cell viability, and proliferation assays For cell cycle analysis, cells were harvested, washed with phosphate‐buffered saline (PBS), fixed in chilled 70% ethanol at 4°C for 30 min, treated with 1 mg/mL RNase A (Sigma‐Aldrich), and stained with 100 μg/mL propidium iodine for 30 min. Cell cycle quantification was performed using a FACSCalibur instrument and CellQuest Pro software (both BD Biosciences). The Caspase‐Glo 3/7 assay kit (Promega, Waldorf, Hessen, Germany) was used according to the manufacturer's instructions. The measured luminescence (RLU) was presented as the mean value ± standard deviation (n  =  3). Apoptotic loss of membrane asymmetry was analyzed by staining with PE Annexin V and 7‐AAD (BD Biosciences) and quantified using a FACSCalibur instrument. For the apoptosis assays, non‐synchronized cells were treated with the test compounds for the indicated times. Cell viability assays were conducted with the Cell Titer‐Blue^® Cell Viability assay (Promega) using fluorescence as a read‐out, according to the manufacturer's instructions (excitation/emission wavelengths: 562 nm/615 nm). The significance of differences between populations of data was assessed using the Student's two‐tailed test (*P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001). Proliferation assays were conducted using the Cell Titer‐Blue® Cell Viability assay (Promega), as described [[76]32]. In brief, cells were seeded in 96‐well plates and transfected with P53‐mRNA or control mRNA. After, the fluorescence was measured at the indicated time points using a Victor X4 Multilabel Counter (Victor X4, PerkinElmer, Rodgau, Hessen, Germany). 2.8. Luciferase assay Luciferase assays were performed using the Luciferase Assay System (Promega). Briefly, the organoids were harvested and dissociated into single cells using TrypLE and mechanical dispersion. Luciferase activity in the cells was measured according to the manufacturer's instructions. Briefly, the cells were lysed in lysis buffer. Afterwards, 20 μL cell lysate were mixed with 100 μL luciferase assay reagent (Promega, Waldorf, Hessen, Germany) and the luminescence was immediately recorded using a plate‐reading luminometer (Victor X4, PerkinElmer, Rodgau, Germany). 2.9. Lactate dehydrogenase (LDH) cytotoxic assay The cell culture medium was collected and diluted (1:100) in an LDH storage buffer (Promega, Waldorf, Hessen, Germany). For a single reaction, 50 μL of diluted cell culture medium was mixed with 50 μL of LDH Detection Enzyme Mix (Promega) and 0.25 μL of reductase substrate (Promega). After a 60‐min incubation period at room temperature, luminescence was recorded using a plate‐reading luminometer (Victor X4, PerkinElmer, Rodgau, Germany). 2.10. Chromosome spreads The cells were treated overnight with 3.3 μmol/L Nocodazol (Sigma Aldrich). The next day, the cells were harvested by mitotic shake off and hypotonically swollen in 40% medium/60% tap water for 20 min at 37°C. The cells were fixed with freshly prepared Carnoy's solution (75% methanol and 25% acetic acid), and the fixative was changed several times. For spreading, the cells in Carnoy's solution were dropped onto pre‐chilled glass slides. Slides were dried at room temperature for 24 h and stained with 4’,6‐diamidino‐2‐phenylindole (Thermo Fisher Scientific). Chromosome number per condition was counted using an AxioObserver.Z1 microscope with an HCX PL APO CS 63.0 × 1.4 oil UV objective (Zeiss). Graphic representation of the results was performed using GraphPad Prism software (GraphPad.com). 2.11. RNA sequencing (RNA‐seq) The RNA samples were used for library preparation using the NEBNext Ultra RNA Library Prep Kit (Illumina, SanDiego, CA, USA). Indices were included for multiple samples. Briefly, mRNA was purified from the total RNA using poly T oligo‐attached magnetic beads (Thermo Fisher Scientific). After fragmentation, first‐strand cDNA was synthesized using random hexamer primers (Thermo Fisher Scientific), followed by second‐strand cDNA synthesis. The library was prepared by end‐repair, A‐tailing, adapter ligation, and size selection. After amplification and purification, insert size of the library was validated using Agilent 2100 (Agilent, Santa Calra, CA, USA) and quantified using quantitative PCR (qPCR). Libraries were then sequenced on an Illumina NovaSeq 6000 S4flow cell with PE150, according to the results of library quality control and expected data volume. Gene expression data were analyzed as previously reported [[77]33]. Briefly, unpaired t‐tests were performed between cells transfected with p53‐mRNA and the EGFP control, considering group variances. A 2‐tailed Student's t‐test was performed for equal variances or Welch's test was used for unequal variances. All calculated P‐values were adjusted using the Benjamini and Hochberg false discovery rate (FDR) method to account for the possibility of false‐positive results due to multiple tests. Genes with an FDR q < 0.05 and an absolute fold change (FC) > 1.5 were further analyzed using Ingenuity Pathway Analysis (IPA) software (Qiagen, Hilden, Bavaria, Germany) to identify overrepresented canonical pathways. Heatmaps of RNA‐seq data were normalized to z‐score by subtracting the mean from each value and dividing by the standard deviation. 2.12. TCF/LEF reporter assay The TCF/LEF reporter assay was purchased from BPS Bioscience (#60500) (San Diego, CA, USA) and performed according to the manufacturer's instructions. Briefly, cells were seeded in a 96‐well plate one day prior to transfection to achieve a cellular confluence of 90% on the day of the transfection. The following day, a transfection mixture, consisting of Lipofectamine Messenger MAX Transfection Reagent (1 μL of the TCF/LEF reporter plasmid) and 1 μg of IVT mRNA, was prepared for each well. After a 25‐min incubation period at room temperature, the mixture was introduced into the cells. Subsequently, a 24‐h incubation at 37°C was carried out, and a dual luciferase assay was conducted using a plate‐reading luminometer (Victor X4, PerkinElmer, Rodgau, Hessen, Germany) to record the assay results. 2.13. Animal experiments All animal experiments were approved by the regional council of Darmstadt and performed under the supervision of the Division of Laboratory Animal Medicine (Goethe University, Medical School, and Georg‐Speyer‐Haus). For the OC orthotopic mouse model, 6‐8‐week‐old female nude mice (body weight: 25‐30 g) were purchased from Charles River Laboratories (Sulzfeld, Bavaria, Germany) and housed under a 12 h‐light/dark schedule. Twenty minutes before the surgery, the mice received analgesia (butorphanol 3 mg/kg and carprofen 2 mg/kg, subcutaneously), were kept under isoflurane anesthesia, transferred onto a heating pad to prevent hypothermia, inspected with the dorsal side up under a microscope, and kept anesthetized during the whole procedure. To confirm that the animals were under deep anesthesia, the toes of the hind paws were pinched using forceps. The procedure was performed in the absence of a pedal reflex. The eyes were covered with ointment to avoid drying. The optimal point of incision to reach the ovary was located on the dorsal side, to the right of the middle, and under the ribs. The area was disinfected with 70% ethanol, anthethe skin was then gently lifted using forceps and cut into pieces. The skin was separated from the abdominal wall using blunt‐ended scissors. Subsequently, the abdominal wall was cut, and the abdominal cavity was opened. The ovarian fat pad was visible adjacent to the ovary and oviduct under the microscope. The fat pad was gently pulled out of the abdominal cavity with forceps, placed on a sterile gauze, soaked in saline, and held with forceps to maintain a stable position. Approximately 1 × 10^6 Luc‐OVCAR‐8 cells were resuspended in 5 μL PBS containing 2% Matrigel and injected into the space between the bursa and ovary with a Hamilton 10 μL‐syringe (30 G needle). Once the needle was inserted into the bursa, a second person pushed the plunger, while the surgeon maintained the correct position of the needle under a microscope. While injecting the cell suspension, expansion of the bursa was observed, indicating that the tumor cells remained in the space between the bursa and ovary. The ovarian fat pad was then placed back into the abdominal cavity. Finally, both sites of the abdominal wall were gently placed together, and the skin was closed with wound clips. On days one and two after the surgery, the animals received analgesia (butorphanol 3 mg/kg and carprofen 2 mg/kg, subcutaneously). One week after the surgery, the wound clamps were removed. The surgery was associated with minimal pain, stress, and harm to the animals. All the mice survived the surgery and showed no behavioral abnormalities after the procedure. For the intraperitoneal OC, 6‐8‐week‐old female nude mice (body weight: 22‐28 g) were used. The injection area was disinfect infected with 70% ethanol. The skin was gently lifted with forceps, and 2 × 10^6 OVCAR‐8 cells were injected. 2.14. In vivo imaging system (IVIS) IVIS or bioluminescence imaging (BLI) was applied to monitor tumor growth and metastases. The mice were anesthetized with isoflurane. Approximately 100 μL of luciferin (Promega, Waldorf, Hessen, Germany) was administrated subcutaneously 15 min before measuring the bioluminescence. Emission of light signals was determined using an IVIS Lumina II Multispectral Imaging System (PerkinElmer, Rodgau, Hessen, Germany) [[78]34]. The first measurement was performed on the day of cell injection, followed by a second imaging seven days after the surgery. Thereafter, imaging was performed once a week. 2.15. Tissue processing for anatomical and histological examination For gross anatomical evaluation of the control and treated mice, the animals were anesthetized using isoflurane gas, injected with 100 μL luciferin, and kept in an isoflurane chamber for 15 min to allow the luciferin to dissociate in the organs. The animals were sacrificed by cervical dislocation, placed in dorsal recumbency, and fixed with needle pins. The skin and body walls were opened at the linea alba, and pictures of each animal's body cavities were taken. Subsequently, gross macroscopic analyses of the liver, kidneys, uterus and ovaries, spleen, omentum, small and large intestines, lungs, and peritoneal surfaces were performed. Noticeable changes in organ morphology have also been previously reported. The organs and structures were excised and placed in six‐well plates (all the organs except the intestine) or Petri dishes (small and large intestines) filled with PBS to keep the organs moist. IVIS measurements were performed, and all the organs were kept in PBS‐buffered formalin for histological and immunohistochemical analyses or on dry ice for further molecular analyses. Whole ovaries, including the ovarian bursa, and samples from the liver, spleen, lungs, and kidneys were carefully dissected. Intestinal tissue preparation and Swiss rolling of the small intestine and colon were performed as described [[79]35]. The specimens were fixed in 10% neutral‐buffered formalin for 48 h at room temperature. The formalin was removed by repeated washing with PBS. After dehydration in graded ethanol and exchange of ethanol with Xylol, the tissues were infiltrated with paraffin and embedded in blocks. Further, 5 μm sections taken from different parts of the blocks were stained with hematoxylin/eosin and microscopically evaluated with respect to overall organ histology and the presence of tumors. 2.16. Statistical methods All experiments were performed at least in triplicates. Standardization and statistical analysis were performed as previously described [[80]36]. Statistical analyses were performed using Microsoft Excel and GraphPad Prism. For paired t‐tests, all experimental groups were compared with their respective groups. The Student's t‐test and Wilcoxon test were used to determine statistical significance between two groups, unless otherwise indicated. P value < 0.05 was considered statistically different. Significant differences (*P ≤ 0.05, **P ≤ 0.01, and ***P ≤ 0.001) are indicated by asterisks. 3. RESULTS 3.1. Mammalian vector‐based expression of p53 induced apoptosis in HGSOC cells As no sufficiently specific drug is available to rescue functional WT p53 in cancer cells bearing a loss‐of‐function p53 mutant, we considered reactivating the p53 gene using different experimental strategies in HGSOC cell lines and primary OC cells. First, 5 known OC cell lines were studied for p53 expression (Supplementary Figure [81]S1A). The expression of p53 was not detected in SKOV3 or OVCAR‐5 cells, in line with previous observations that classified them as p53 null [[82]37] (Supplementary Figure [83]S1A). In contrast, OVCAR‐3, ‐4, and ‐8 cells showed abundant p53 protein expression and low to undetectable levels of the classical p53‐target gene, p21. This was due to mutations within the DNA‐binding domain at R248Q and L130V or a single‐nucleotide mutation at the splice junction (c.376‐1G > A) [[84]38, [85]39, [86]40], which inhibited the expression of the major negative p53 regulator, MDM2 [[87]41], within a negative feedback loop. For further analysis, we selected different HGSOC cell lines, including OVCAR‐8 [[88]42], which harbor different genomic alterations that were confirmed by sequencing (Supplementary Figure [89]S1B). Exogenous expression of Flag‐tagged WT p53 driven by a mammalian vector system for 48 h in mutant TP53‐bearing OVCAR‐8 cells significantly reduced cell proliferation compared to that in the controls (Supplementary Figure [90]S1C). We examined cells transfected with increasing amounts of the WT p53‐expressing vector (Supplementary Figure [91]S1D) by FACS 24 h after transfection and observed a moderate increase in the percentage of apoptotic cells in the sub‐G1 phase (Supplementary Figure [92]S1E), accompanied by increased levels of caspase 3/7 activity and annexin staining (Supplementary Figure [93]S1F). Western blot experiments for the detection of cleaved caspase‐3 and PARP corroborated increasing apoptosis (Supplementary Figure [94]S1D). To further assess the cell cycle progression, in the WB experiments, we observed an upregulation of CDK inhibitors (p16 and p27) and p21 upon a vector‐prone increase of p53 expression (Supplementary Figure [95]S1D). The expression of PLK1 and CDK1, two key players in cell cycle regulation, was reduced at higher amounts of transfected p53‐mRNA likely due to increased apoptosis (Supplementary Figure [96]S1D‐F). Next, we demonstrated the cytotoxicity of WT p53 in a dose‐dependent manner through in vitro assays assessing the colony‐forming ability compared to the controls (Supplementary Figure [97]S1G). Similar experiments in additional HGSOC cell lines (OVCAR‐3 and ‐5) corroborated growth inhibition, cell cycle arrest, induction of apoptosis, and reduction in colony‐forming ability upon WT p53 expression (Supplementary Figure [98]S2). Collectively, these data indicate that rescuing p53 function by vector‐based re‐expression of WT p53 induced cell cycle dysregulation and death in HGSOC cells and occurs in a dose‐dependent manner. 3.2. Design, generation, and characterization of IVT mRNAs in HGSOC cell lines Recently, the therapeutic use of IVT mRNA as a vaccine has fueled great hope in the fight against infectious diseases. However, whether this class of biological agents is suitable for improving precision medicine targeting cancer remains elusive. To address this unanswered question, we used an IVT system based on a bacterial vector including a T7 RNA Polymerase promoter for the transcription of mRNAs (human p53, firefly luciferase [Luc], and EGFP), considering aspects of translational efficacy and decay rates of mRNA [[99]26] (Supplementary Figure [100]S3A). In vitro‐transcribed mRNAs like IVT‐Flag‐tagged p53‐mRNA, dubbed here as p53‐mRNA, were analyzed using agarose gel electrophoresis for quality control (Supplementary Figure [101]S3B). Based on the clinical advantages of liposomes as carriers of chemotherapeutics over conventional chemotherapy due to reduced therapeutically side effects and increased anti‐cancer activity [[102]43, [103]44], we exploited a liposomal system using Lipofectamine Messenger MAX Transfection Reagent^® by testing its supportive quality for the delivery of IVT‐mRNA (EGFP‐mRNA, Luc‐mRNA) to the HGSOC cells. To quantitatively study the uptake of the control EGFP‐mRNA, EGFP expression was measured using FACS as a surrogate marker for transfection efficiency. At 24 h following transfection with EGFP‐mRNA (0.1‐2 μg), a dose‐dependent increase in EGFP expression was detected (Supplementary Figure [104]S3C). Although 0.25 μg EGFP‐mRNA was sufficient for expression in 90% of cells, at higher doses (0.5‐2 μg), 97%‐100% of cells showed EGFP fluorescence, supporting an excellent transfection efficiency in the HGSOC cells (Supplementary Figure [105]S3C). This observation was confirmed by WB, which showed high levels of EGFP at doses of ≥ 0.25 μg EGFP‐mRNA (Supplementary Figure [106]S3C). In time kinetics, a high percentage of EGFP‐expressing cells (92.2%‐99.7%) persisted for 10 days following transfection and declined to baseline levels (≤5.5%) on day 15 (Supplementary Figure [107]S3D). The EGFP expression was corroborated for a period of 7 days by WB (Supplementary Figure [108]S3D). As a second control IVT‐mRNA, Luc‐mRNA transfection was monitored by WB, and activity measurements showed high enzymatic activity in cells transfected with 0.25‐2 μg Luc mRNA (Supplementary Figure [109]S3E). To further validate the usefulness of our approach for the expression of a protein of choice driven by IVT mRNA in the HGSOC, we tested additional cell lines (OVCAR‐3, ‐4, ‐5, and‐8) and primary cells using immunofluorescence (Supplementary Figure [110]S3F), FACS (Supplementary Figure [111]S3G), and WB (Supplementary Figure [112]S3H). Strong expression with 72% and 86% could be detected in OVCAR‐5 and primary HGSOC cells, respectively, and very strong expression (≥ 95%) in the OVCAR‐3, ‐4 and ‐8 cells (Supplementary Figure [113]S3G‐H) transfected with 1 μg EGFP‐mRNA. Collectively, different experiments demonstrated the high efficiency of liposomal‐based IVT mRNA transfection and expression, irrespective of the OC cell line, primary cells, or type of mRNA. 3.3. Characterization of mRNA expression in primary human cells and a mouse model We investigated the time kinetics of EGFP and Luc mRNA expression in primary HGSOC cells. The EGFP expression using 1 μg mRNA could be detected in FACS studies on day one with 85% positive cells decreasing to 74% on day 11 and by WB for at least 7 days (Supplementary Figure [114]S4A). Upon transfection of Luc‐mRNA, enzymatic activity was detectable for 7 days, and WB signals were detectable for 4 days (Supplementary Figure [115]S4B). Organoids from primary HGSOC showed efficient Luc activity or EGFP expression for up to 5 days (Supplementary Figure [116]S4C‐D). To monitor the duration of Luc activity in the mice by in vivo BLI, we transfected the OVCAR‐8 cells with Luc mRNA (Supplementary Figure [117]S4E) for subsequent application to the mice: (I) orthotopic implantation of 1 × 10^6 OVCAR‐8 cells transfected with 2 μg Luc mRNA; (II) intraperitoneal injection of 2 × 10^6 OVCAR‐8 cells transfected with 4 μg Luc mRNA, or (III) intraperitoneal injection of 10 μg liposomal Luc‐mRNA (Supplementary Figure [118]S4F). Luc activity was detectable for 2‐3 days in all three experimental settings. Overall, the liposomal IVT mRNA was well suited to support efficient protein expression in primary human cells in cell culture and In vivo over several days. 3.4. mRNA‐based reactivation of p53 in HGSOC cells induces cell cycle arrest and cell death We aimed to rescue p53 function in different preclinical models by transfecting HGSOC cell lines and primary cells with p53‐mRNA. Low amounts of p53‐mRNA (10‐250 ng) strongly induced p53 protein expression in OVCAR‐8 cells (Figure [119]1A). An increase in cells in G1 was observed at 100‐500 ng p53‐mRNA 24 h post‐transfection compared to the controls (Figure [120]1B), supporting the reactivation of p53 activity. Strong induction of the cell cycle inhibitors, p21, p16, and p27, was readily visible at low doses of p53‐mRNA (≥ 0.1 μg) in parallel with the downregulation of critical regulators, such as PLK1, Aurora A, CDK1, and Cyclin A/B, corroborating our results using a vector‐based re‐expression of p53 (Figure [121]1C). These observations correlated with a significant drop in cellular viability by ∼2‐fold after 48 h at a dose of 0.5 μg (Figure [122]1D), accompanied by the rise of different indicators of cell death, such as Puma, Noxa, and Fas, cleaved PARP, cleaved Caspase‐3 compared with untreated controls (Figure [123]1C), and increased caspase 3/7 activities (Figure [124]1E), and Annexin staining (Figure [125]1F). Loss of colony formation ability was induced by increasing the concentration of p53‐mRNA (Figure [126]1G). We additionally performed a cytotoxicity assay, which confirmed previous observations of the impact of IVT P53 mRNA on ovarian cancer cells using apoptosis and colony forming assays (Figure [127]1H). Moreover, we tested p53‐null HGSOC cells, such as OVCAR‐5, for their sensitivity to re‐expression of WT p53. Within 24 h following the p53‐mRNA transfection, prominent signs of cell death, including strong PARP cleavage (Supplementary Figure [128]S5A), high levels of cells in the sub‐G1 phase (Supplementary Figure [129]S5B), and high levels of caspase 3/7 (Supplementary Figure [130]S5C) were observed. The performed cytotoxicity assays also confirmed previous observations of the impact of IVT P53 mRNA on OVCAR‐5 cancer cells using the LDH‐Glo cytotoxicity Assay (Supplementary Figure [131]S5D). Overall, our observations indicated that rescuing p53 function by transfection of WT p53‐mRNA induced cell death in different HGSOC cell lines. FIGURE 1. FIGURE 1 [132]Open in a new tab Liposomal transfection of in vitro‐transcribed WT p53‐mRNA induces cell cycle arrest and apoptosis in the HGSOC cell line OVCAR‐8. (A) OVCAR‐8 cells were transfected with increasing amounts of liposomal IVT Flag‐tagged WT p53‐mRNA. Cell lysates were subjected to WB using Flag‐ and β‐Actin‐antibodies. (B) The dose‐dependent cell cycle distribution of Nocodazole‐synchronized cells is represented as a bar graph (upper panel; n = 3). Cell lysates were subjected to WB using Flag‐, Cyclin E1‐, Cyclin B1‐, PLK1‐, and β‐Actin‐antibodies (lower panel). (C) Cell lysates were subjected to WB for key mediators of cell cycle regulation using Flag‐, p53 (pS20)‐, p53‐, p21‐, p27‐, p16‐, PLK1‐, Cyclin A1‐, Cyclin B1‐, Cyclin E1‐, Aurora A‐, CDK1‐, and β‐Actin‐antibodies (upper panel). The same cell lysates were subjected to WB for regulators of apoptosis using PARP‐, Caspase‐3‐, Puma‐, Noxa‐, and Fas‐antibodies (lower panel). (D) Cells were mock‐ or 0.5 μg p53‐mRNA‐transfected, and proliferation was measured using a CellTiter‐Blue Cell Viability assay, 24 h post‐treatment. ^** P < 0.01, ^*** P < 0.001, Student's t‐test, unpaired and two‐tailed. (E) The dose‐dependent Caspase‐3/7 activities were determined using a Caspase‐Glo 3/7 assay. ^*** P < 0.001, Student's t‐test, unpaired and two‐tailed. (F) Graphical representation of Annexin V positive (%) cells. FACS analysis of transfected cells, gated for Annexin V positive (early apoptotic), and Annexin V/7AAD positive (late apoptosis) populations is demonstrated. n  =  3 for each dose; ^** P < 0.01, ^*** P < 0.001. (G) Representative images show the number of 3D colonies (dose‐dependent) on day 10 (left panel). The dose‐dependent distribution of colonies is represented as bar graph (right panel; n = 3). ^* P < 0.05, ^*** P < 0.001, Student's t‐test, unpaired and two‐tailed. (H) Graphical representation of the cell cytotoxity caused by increasing concentrations of transfected p53 mRNA. n  =  3 for each concentration; ^* P < 0.5, ^**P < 0.01.Aurora‐A, Aurora Kinase A; CDK1, cyclin‐dependent kinase 1; FACS, Fluorescence‐Activated Cell SortingA; Fas, Tumor necrosis factor receptor superfamily member 6; HGSOC, High‐Grade Serous Ovarian Cancer; IVT, in vitro‐transcribed; Noxa, NADPH oxidase activator 1; p53, tumor protein p53; PARP, Poly (ADP‐ribose) polymerase; PLK1, Polo‐like kinase 1; WB, western blot. 3.5. Reinstating p53 reduces numerical chromosomal instability (nCIN) in the HGSOC cell line OVCAR‐8 Chromosomal instability is a fundamental feature of OC and most human OC cells exhibiting aneuploidy [[133]45, [134]46, [135]47]. The OVCAR‐8 cell line is classified as a hyperdiploid cell line with a high numerical complexity and modal number of chromosomes ranging between 50‐59 [[136]45]. Thus, we aimed to determine whether restoring functional WT p53 could reduce the number of chromosomes in cells to a near‐diploid status. Therefore, the cells were treated every two days for 28 days with low doses of p53‐mRNA, and the distribution of chromosomes was assessed at the end of the incubation period (Figure [137]2A‐B). The mean chromosome number was reduced from 53 to 47 upon restoration of WT p53 (Figure [138]2C‐D). FIGURE 2. FIGURE 2 [139]Open in a new tab Restoring p53 function reduces nCIN in OVCAR‐8 cells. (A) Treatment schedule. OVCAR‐8 cells were kept in culture for 28 d and treated with 200 ng p53‐mRNA every 48 h. Metaphase spreading of untreated controls and treated OVCAR‐8 cells were prepared at the end of the incubation. Chromosomes were stained with Hoechst. (B) Western blot of p53‐mRNA expressing OVCAR‐8 using Flag‐ and β‐Actin‐antibodies. (C) Histogram plots of the distribution of chromosome numbers in controls and cells treated with 200 ng p53‐mRNA. (D) Quantification of the number of chromosomes in cells with/without p53‐mRNA‐treatment (upper panel). The results were analyzed statistically, mean ± SD, ^**** P < 0.001. Representative images of metaphase chromosome spreads (lower panel). Green, H2AX pSer139; blue, DAPI. Scale bar = 20 μm. (E) IF images showing OVCAR‐8 and OVCAR‐8 + mRNA p53 cells with H2AX pSer193 foci staining 15 days post‐treatment with mRNA p53 (upper left panel). Scale bar = 20 μm. Quantification of H2AX pSer139 foci 14 days post‐treatment with mRNA p53 (lower left panel). The results are presented as mean ± SEM, n = 85 cells, ^*** P ≤ 0.001. OVCAR‐8 cells were kept in culture for 14 days and treated with 200 ng p53‐mRNA every 48 h (lower right panel). Cell lysates were subjected to WB using Flag‐ and β‐Actin‐antibodies. d, day; H2AX pSer 139, H2A histone family member X phosphorylated at Ser 139; nCIN, numerical chromosomal instability; p53, tumor protein p53; SD, standard deviation; IF, Immunofluorescence; SEM, standard error of the mean. The HGSOC subtype is characterized by increased replicative stress [[140]48], resulting from various factors, including frequent loss of TP53 [[141]49]. This loss of TP53 function contributes to replicative stress by compromising the DNA damage response mechanisms [[142]49], which may partly explain the increased genetic instability observed in the OVCAR‐8 cells. To quantify the extent of replicative stress in the OVCAR‐8 cells before and after the restoration of p53 function, we assessed Ser139‐phosphorylated H2AX foci (γ‐H2AX) as a reliable marker of replicative stress and the presence of DNA double‐strand breaks [[143]50] after 7 and 14 days. We found that the baseline level of γ‐H2AX foci decreased significantly over the treatment period from an average of 8 to 3 foci after 14 days in the OVCAR‐8 and p53 mRNA‐treated OVCAR‐8 cells (Figure [144]2E). This result provided further evidence for the critical role played by p53 in maintaining genomic integrity in the HGSOC cells, most likely by counteracting replicative stress. 3.6. mRNA‐based rescue of p53 function in primary HGSOC cells induces cell cycle arrest and cell death To further validate the rescue of p53 expression in a preclinical scenario, we compared the effects of p53‐mRNA transfection in primary normal ovarian and HGSOC cells. The expression of p53 was detected in all the primary samples, except the keratinocytes (Figure [145]3A). The first comparison of HGSOC and corresponding normal ovarian tissue from the same patient revealed higher expression of p53 in tumor cells, despite equal amounts of transfected p53‐mRNA (Figure [146]3A). Analysis of additional patient‐derived tissues confirmed that the expression of p53 and caspase 3/7 activity in all the tumor samples (Figure [147]3B) were significantly higher than the corresponding levels in the normal ovarian cells. In contrast, nearly the same levels of EGFP expression were observed in the normal and tumor cells upon transfection with equal amounts of IVT‐EGFP mRNA (Supplementary Figure [148]S6A). After testing the transfection efficacy of fluorescent mRNA in the normal and HGSOC patient‐derived cells, no difference was detected (Supplementary Figure [149]S6B), suggesting that different transfection efficiencies did not seem to be the reason for the different p53 levels compared to the primary cells (HGSOC vs. normal). FIGURE 3. FIGURE 3 [150]Open in a new tab Liposomal transfection of p53‐mRNA results in higher p53 expression in primary HGSOC cells compared to their normal counterparts. (A) Primary human cells of different origins (tumor, normal) and OVCAR‐8 cells were transfected with p53‐mRNA. Cell lysates were subjected to WB using Flag‐ and β‐Actin‐antibodies. (B) Primary human HGSOC cells (tumor, normal) derived from patients P3‐8 were transfected with 0.5 μg p53‐mRNA. Cell lysates were subjected to WB using Flag‐ and β‐Actin‐antibodies (upper panel). (lower left) Expression efficacy was depicted as bar graphs (lower left panel). Caspase 3/7 activities were determined using a Caspase‐Glo 3/7 assay (lower right panel). ^** P < 0.01, ^*** P < 0.001. Student's t‐test, unpaired and two‐tailed. (C) Lysates of mock‐ or p53‐mRNA‐transfected normal ovarian or tumor HGSOC (patient 2) cells were subjected to WB using Flag‐, p53‐, p21‐, CDK1‐, p27‐, p16‐, and β‐Actin‐antibodies (left panel). The normalized levels of p53 expression are represented as a bar graph (right panel). (D) The same cell lysates were subjected to WB using PARP‐, Puma‐, and Noxa‐antibodies (left panel). Caspase‐3/7 activities of mock‐ or p53‐mRNA‐transfected normal and tumor HGSOC cells were determined using a Caspase‐Glo 3/7 assay (right panel; n = 3). (E) Representative images show the 3D colonies of mock‐transfected cells on day 10 (left panel). Representative images show the 3D colonies of p53 mRNA‐transfected cells on day 10 (right panel; n = 3). ^* P < 0.05, ^** P < 0.01, ^*** P < 0.001. Wilcoxon test, unpaired and two‐tailed. (F) Graphical representation of the cell cytotoxicity caused by increasing concentrations of transfected p53 mRNA. n  =  3 for each concentration; ^*P < 0.05. (G) Representative images show organoids on day 4 after transfection with 2 μg p53 mRNA (left panel). Lysates of HGSOC organoids (patient 3) mock‐ or p53‐mRNA‐transfected cells were subjected to WB using Flag‐ and β‐actin‐antibodies (middle left panel). Organoid size was determined using ImageJ (middle right panel; n = 30). ^*** P < 0.001; Student's t‐test, unpaired and two‐tailed. Caspase‐3/7 activities of HGSOC organoids mock‐ or p53‐mRNA‐transfected were determined using the Caspase‐Glo 3/7 assay (right panel; n = 3). ^* P < 0.05; Student's t‐test, unpaired and two‐tailed. CDK1, cyclin‐dependent kinase 1; HUVEC, human umbilical vein endothelial cells; N, normal tissue; Noxa, NADPH oxidase activator 1: P (1‐8), patient‐derived material (n = 8 patients); p53, tumor protein p53; PARP, Poly (ADP‐ribose) polymerase; Puma, p53 upregulated modulator of apoptosis; T, tumor tissue. The WB examination revealed the upregulation of the inhibitors, p21 [[151]51], p16, and p27 (Figure [152]3C) and apoptosis (Figure [153]3D), which was stronger in the p53‐mRNA‐transfected primary HGSOC cells compared to their normal counterparts, correlating with a more pronounced reduction in the number of HGSOC colonies (Figure [154]3E), which was corroborated by the LDH‐Glo cytotoxicity assay (Figure [155]3F). Analysis of different HGSOC patients‐derived organoids also demonstrated a significant decrease in volume and an increase in caspase 3/7 activity upon p53‐mRNA‐treatment under 3D cell culture conditions (Figure [156]3G). In summary, although the expression of p53 reduced the viability of both types of primary cells (normal vs. HGSOC), the effect was significantly stronger in the primary HGSOC cells compared to that in the normal cells, which could be due to elevated levels of p53 in the tumor cells. 3.7. Transcriptome alterations in OVCAR‐8 cells upon rescue of WT p53 functions Our observations of reduced proliferative activity and increased cell death in the cell lines and primary cells of HGSOC origin support the model of rescued p53 function upon p53 re‐expression by mRNA transfection. To deepen our understanding of the underlying biological processes and address this aspect in a relatively unbiased manner, we transfected the OVCAR‐8 cells with 1 μg p53‐mRNA or mock control (EGFP mRNA) in three independent experiments. In total, 25,540 human genes were profiled by RNA‐seq 24 h after the transfection (Supplementary Figure [157]S7A‐B). Both groups were compared using unpaired t‐tests, considering group variance. Transcriptomic analysis revealed that 3,061 transcripts were upregulated, and 1,662 were downregulated (Figure [158]4A‐B). These significant genes were further analyzed with the IPA software to identify overrepresented canonical pathways associated with the p53‐mRNA transfection (Figure [159]4C, Supplementary Figure [160]S7C). FIGURE 4. FIGURE 4 [161]Open in a new tab Identification of differentially expressed genes in p53‐mRNA‐transfected OVCAR‐8 cells compared to mock‐transfected counterparts. (A) Heatmap of normalized gene expression represented by z scores of OVCAR‐8 cells treated with mock (EGFP mRNA) or p53‐mRNA from three independent RNA‐seq experiments (Student's t tests or Welch tests, depending on the variance, corrected by FDR q adjusted P‐value < 0.05 and absolute FC > 1.5). (B) Volcano plot showing the DEGs in the RNA‐seq between OVCAR‐8 cells treated with EGFP vs. p53‐mRNA (red dots = upregulated genes, blue dots = down‐regulated genes). (C) Selected overrepresented Ingenuity pathways from pathway enrichment analysis of DEGs with FDR‐adjusted P‐values < 0.05 and FC > 1.5. Pathways with a positive z score, representing a global upregulation of the pathway genes, are depicted in orange; pathways with a negative z score, indicating a global gene downregulation in the pathway, are depicted in blue; and pathways with no specific pattern are depicted in gray. (D) Heatmap of all genes corresponding to the “p53 signaling pathway” from IPA, indicating the P‐value, FDR‐adjusted P‐value and FC for the comparison of EGFP with p53‐mRNA. Genes with validation by WB are highlighted in red. (E) Cell lysates were subjected to WB for p53 signaling pathways using p21, MDM2, Puma, p73, Fas, and CDK1 antibodies. CDK1, cyclin‐dependent kinase 1; DEG, differentially‐expressed gene; EGFP, enhanced green fluorescent protein; Fas, tumor necrosis factor receptor superfamily member 6; FC, fold change; FDR, false‐discovery rate; IPA, Ingenuity Pathway Analysis; MDM2, double minute 2 homolog protein; Puma, p53 upregulated modulator of apoptosis; Noxa, NADPH oxidase activator 1; p53, tumor protein p53; RNA‐seq, RNA sequencing. Consistent with the pro‐apoptotic functions of p53 as a tumor suppressor involved in the induction of cell cycle arrest and apoptosis, our IPA revealed an overrepresentation of several related pathways upon p53‐mRNA transfection, such as “p53 signaling”, “apoptosis signaling”, and several “cell cycle” pathways (Figure [162]4C). To confirm these findings, we investigated all genes involved in the IPA “p53 signaling” pathway and we realized that most of them (51/81, 63%) were differentially expressed with FDR q < 0.05 and absolute FC >1.5 (Figure [163]4D). Notably, most known downstream targets of p53 signaling (96%) were coordinately upregulated by mRNA transfection of p53‐mRNA (Figure [164]4D). We further validated these results by performing WB of selected proteins from the p53 signaling pathway, including p21 (CDKN1A), MDM2, Puma (BBC3), p73, FAS (CD95), and CDK1 (Figure [165]4E). Their expression in the OVCAR‐8 cells was substantially upregulated or downregulated by p53‐mRNA in agreement with transcriptomic analysis. Most genes in the apoptosis signaling pathway (44/68, 65%) were significantly deregulated (Supplementary Figure [166]S8A). We extensively validated 12 of these proteins using WB (Supplementary Figure [167]S8B) to demonstrate that p53‐mRNA transfection induces apoptosis in the OVCAR‐8 cells. Several “cell cycle” pathways, including “cell cycle control of chromosomal replication”, “cell cycle: G2/M DNA damage checkpoint regulation”, and “cyclins and cell cycle regulation”, were significantly overrepresented, as well as “Mitotic roles of Polo‐like kinase” [[168]52, [169]53], and they showed a global downregulation (negative z score) (Figure [170]4C). Most of the genes from these pathways were differentially expressed (Supplementary Figure [171]S9A‐B) and validated using WB (Supplementary Figure [172]S9C), confirming the downregulation of the cell cycle and mitotic roles of PLK1. Remarkably, the cellular senescence pathway was also upregulated and may contribute to cell death along with the induction of apoptosis (Figure [173]4C). Next, we carried out a transcriptome comparison between the primary HGSOC cells from patient one and the OVCAR‐8 cells, of which both were p53‐mRNA‐transfected (Figure [174]5A). The high variability among the primary samples resulted in low significance after FDR correction for multiple testing. Thus, we considered the significance of the transcripts of primary HGSOC cells with P < 0.01 and FC > 1.5. A total of 156 transcripts were upregulated, and 115 were downregulated (Figure [175]5B‐C). Several genes from the primary HGSOC cells were significant and similar to the pathways identified in the OVCAR‐8 cells (Figure [176]5D), supporting the relevance of both datasets. FIGURE 5. FIGURE 5 [177]Open in a new tab Analysis of differentially expressed genes in primary HGSOC cells. (A) Primary HGSOC cells from patient 1 were transfected with p53‐mRNA or EGFP‐mRNA. Cell lysates were subjected to WB using EGFP, Flag, and β‐Actin antibodies. (B‐C) Heatmap (B) and volcano plot (C) showing the DEGs in the RNA‐seq between primary HGSOC cells treated with EGFP vs. p53‐mRNA (P value < 0.01 and absolute FC > 1.5; red dots = upregulated genes, blue dots = down‐regulated genes). (D) Transcriptome analysis of DEGs in common with OVCAR‐8 and primary HGSOC cells for selected significant overrepresented Ingenuity pathways. EGFP, enhanced green fluorescent protein; FC, fold change; DEG, differentially‐expressed gene; HGSOC, high‐grade serous ovarian cancer; RNA‐seq, RNA sequencing; TP53, tumor protein p53. Consistent with the paramount function of p53 in OC development, our IPA showed multiple transcriptional alterations in the “ovarian cancer signaling” pathway. Many genes in this pathway (55/76, 72%), including Wnt/β‐catenin signaling, were significantly deregulated (Figure [178]6A‐B). The Wnt/β‐catenin pathway promotes chemoresistance, metastasis, and cancer stem cell self‐renewal in OC by the aberrant activation of Wnt/β‐catenin signaling, leading to‐catenin hyperactivity [[179]54]. Treatment with p53‐mRNA resulted in upregulation of AXIN1, which functions as a scaffolding protein for components of the β‐catenin destruction complex, including APC, CK1PP2A, and GSK3β. Additionally, deregulation was observed in members of the frizzled receptor, Wnt, and TCF/LEF families (Figure [180]6A‐B). Overall, these transcriptional alterations led to a moderate reduction of β‐catenin (Figure [181]6B) and to the descending activity of Wnt signaling using a TCF/LEF reporter assay in the p53‐mRNA‐treated cells (Figure [182]6C). FIGURE 6. FIGURE 6 [183]Open in a new tab OC signaling pathway upregulation upon treatment of OVCAR‐8 cells with IVT p53‐mRNA.(A) Heatmap of all genes corresponding to the OC Signaling pathway from IPA, indicating the P value, FDR‐adjusted P value, and FC for the comparison of EGFP with p53‐mRNA in OVCAR‐8 cells. (B) WB analysis using Flag‐, EGFP‐, GSK3B‐, p‐GSK3B‐, β‐Catenin‐, p‐β‐Catenin‐, AKT3‐, Cyclin D‐, CDK4‐, BRCA2‐, K‐Ras‐, PLK1‐, and β‐Actin‐antibodies. (C) TCF/LEF luciferase reporter responses of cells transfected with increasing doses of p53‐mRNA. WB analysis using Flag‐ and β‐Actin‐antibodies. (ser9), phosphorylated Glycogen synthase kinase 3β AKT3, V‐Akt murine thymoma viral oncogene homolog 3; at serine 9; BRCA2, breast cancer gene 2; CDK4, cyclin‐dependent kinase 4; CTNNB1, β‐Catenin; EGFP, enhanced green fluorescence protein; FC, fold change; GSK3β, glycogen synthase kinase 3β; p‐ GSK3β IPA, Ingenuity Pathway Analysis; WB, western blot; IVT, in vitro transcribed; K‐ras, kristen murine sarcoma virus 2 homolog; OC, ovarian cancer; p53, tumor protein p53; PLK1, Polo‐like kinase 1. 3.8. Orthotopical application of p53‐mRNA‐treated OVCAR‐8 cells resulted in a dose‐dependent reduction of tumor growth in an HGSOC Xenograft model To explore the viability of the p53‐mRNA‐treated OC cells in a physiological microenvironment, an orthotopic disease model was used. First, we tested whether stable Luc‐expressing OVCAR‐8 cells (OVCAR‐8/Luc) showed an altered response to p53‐reactivation. Treatment with 100 ng p53‐mRNA induced only moderate cell death, as evidenced by the low expression of apoptotic markers/activation, low caspase 3/7 activity, and moderate inhibition of cell growth, whereas transfection with 500 ng p53‐mRNA induced a robust apoptotic response and strong drop in colony‐forming efficacy (Supplementary Figure [184]S10A‐C). Thus, the stable expression of Luc in OVCAR‐8 cells did not alter the response to mRNA‐dependent reactivation of p53 compared to that in WT cells. One day prior to the xenograft experiment, the OVCAR‐8/Luc cells were transfected with mock‐ or p53‐mRNA (100 ng or 500 ng) (Figure [185]7A). To establish an orthotopic model, a Matrigel/OVCAR‐8/Luc cell suspension was injected into the right ovary of each mouse. The determination of tumor volumes at 1× per week for an observation period of 10 weeks by BLI demonstrated growth retardation in tumors originating from cells treated with 100 ng p53‐mRNA and a complete block of cellular proliferation upon treatment with 500 ng p53‐mRNA (Figure [186]7B). To verify the luciferase activity in vivo, the mice were sacrificed at the end of the study and subjected to anatomical/histological examination of all organs by two independent veterinary histologists. The inspection of removed ovaries based on the determination of volume (Figure [187]7C) and BLI measurements (Figure [188]7D) confirmed the dose‐dependent size reduction of the ovary tumors by p53 mRNA treatment supporting the In vivo BLI measurements (Figure [189]7B). Histological examination revealed huge tumor masses and profoundly altered morphology due to the vast carcinoma tissue in the right ovaries injected with mock‐treated OVCAR‐8/Luc cells. In the 100 ng p53‐mRNA‐treated group, the right ovaries exhibited low to medium amounts of tumors (Figure [190]7E, d‐e) or normal histology. The right ovaries of the 500 ng p53‐mRNA‐treated group and all other organs showed normal histology (Figure [191]7E, g‐h, Supplementary Figure [192]S10D). In all treatment groups (mock, 100 ng, and 500 ng p53‐mRNA), the left ovaries that did not receive OVCAR‐8/Luc cells showed normal histology with follicular and luteal structures, as well as stromal tissue (Figure [193]7E, c‐f‐i). In the mice injected with mock‐treated cells, metastases were detected in the intestine; metastatic activity in the 100‐ng group was low and absent in the 500‐ng group (Figure [194]7D‐F). Overall, these animal data revealed dose‐dependent growth retardation and blocked dissemination of p53‐mRNA‐treated OVCAR‐8/Luc cells under orthotopic conditions. FIGURE 7. FIGURE 7 [195]Open in a new tab Application of liposomal p53‐mRNA to OVCAR‐8 cells prevents intrabursal growth and dissemination in an orthotopic mouse model. (A) Schematic design of the orthotopic murine model. (B) Bioluminescence imaging. One day before, Luc‐expressing OVCAR‐8 cells were transfected (mock, 100 ng p53‐mRNA, or 500 ng p53‐mRNA) (left panel). Approximately 1 × 10^6 cells per mouse were injected into the bursa (right ovary) of nude mice on day 0 and imaged using the IVIS Lumina system following D‐luciferin application. Eight weeks following orthotopic application of OVCAR‐8 cells (mock‐ or p53‐mRNA‐treated) representative images of the mice (one per week) at baseline are shown. Photon flux is indicated in the pseudocolored heat map. BLI was performed at the indicated time points and data were presented in a graphic (right panel). Photon flux was normalized to baseline values obtained from the mice on day 0. ^* P < 0.05, ^** P < 0.01. Wilcoxon test, unpaired and two‐tailed. (C) Representative photographs of removed ovaries 10 weeks after intrabursal application of mock‐ or p53‐mRNA‐transfected (100 ng, 500 ng) OVCAR‐8 cells in mice (left panel, n = 7 for each group). Analysis of extracted ovaries. The dose‐dependent tumor volumes are represented as bar graph (right panel). ^* P < 0.05, ^** P < 0.01. Wilcoxon test, unpaired and two‐tailed. (D) BLI of removed, tumor‐bearing organs 10 weeks after intrabursal application of mock‐ (n = 7), or p53‐mRNA‐transfected (100 ng, 500 ng) OVCAR‐8 cells (n = 7 mice for each group) represented as bar graph. ^** P < 0.01. Wilcoxon test, unpaired and two‐tailed. (E) Hematoxylin and eosin‐stained sections of ovaries injected with p53‐mRNA‐transfected (100 ng, 500 ng) (n = 7) or mock‐ (n = 7) OVCAR‐8 cells at the end of the experiment (10 weeks). Scale bars, 300 μm. (F) Representative hematoxylin and eosin‐stained sections of metastases at the small intestine and colon following intrabursal injection of mock‐, or p53‐mRNA‐transfected (100 ng, 500 ng) OVCAR‐8 cells (after 10 weeks). Scale bars, 300 μm.BLI, Bio‐layer Interferometry; IVIS, In vivo imaging system; p53, tumor protein p53. 3.9. Intraperitoneal application of OVCAR‐8 cells followed by intraperitoneal treatment with p53‐mRNA inhibited dissemination of tumor cells To further validate the clinical relevance of p53‐mRNA in metastatic HGSOC, we tested whether the scattered tumor cells within the peritoneum were accessible to liposomal p53‐mRNA. The intraperitoneal administration of drugs for OC was shown to be more efficacious than systemic administration and was recommended by the National Cancer Institute [[196]55]. Along these lines, we injected 2 × 10^6 OVCAR‐8/Luc cells intraperitoneally and administered p53‐mRNA 4‐6 h later via the intraperitoneal route. The mice were treated for three weeks with 2 intraperitoneal injections per week with p53‐mRNA (0.16 mg/kg) or control. Although the OVCAR‐8/Luc cells treated with the control grew exponentially, the intraperitoneal treatment with the liposomal p53‐mRNA completely blocked the tumor cell growth (Figure [197]8A). During the entire observation period, the body weights of the mice in both groups developed normally (Figure [198]8B). FIGURE 8. FIGURE 8 [199]Open in a new tab Intraperitoneal injection of OVCAR‐8 cells followed by intraperitoneal treatment of mice with liposomal p53‐mRNA prevents tumor formation and organ dissemination in a xenograft mouse model. (A) Bioluminescence imaging of luciferase‐expressing metastatic OVCAR‐8 cell‐bearing mice upon treatment with liposomal p53‐mRNA (upper panel). Approximately 2 × 10^6 OVCAR‐8 cells stably expressing luciferase were i.p. injected followed by treatment with liposomal p53‐mRNA for three weeks (2 × 5 μg liposomal p53‐mRNA per week). The color scale represents p/sec/cm^2/steradian. Tumor burden of control and liposomal p53‐mRNA‐treated nude mice determined weekly by bioluminescence over a period of 9 weeks (lower panel; n = 8). ^* P < 0.05, ^** P < 0.01, ^*** P < 0.001. Wilcoxon test, unpaired and two‐tailed. (B) Determination of the body weight over the entire observation period. (C) Inspection of peritoneal structures and organs for tumor dissemination following laparotomy. (D) Bioluminescence imaging of removed murine organs (control vs. p53‐mRNA treatment). (E) Hematoxylin and eosin‐stained representative sections of ovary enclosed by bursal peritoneum and fat pad (a, e), small intestine (b, f), colon (c, g) and fat/peritoneum (d, h) of mice ip injected with OVCAR‐8 cells alone (control a‐d) or followed by i.p. application of p53‐mRNA (e‐h). In the control group animals, prominent tumor cell aggregates are visible associated with serosal surfaces and fat (arrows in a‐d) in contrast to the p53‐mRNA‐treated animals. Scale bars 300 μm.i.p., intraperitoneal. In the control mice receiving intraperitoneal OVCAR‐8/Luc cells and intraperitoneal mock treatment, gross anatomical analysis revealed tumor masses on the peritoneal surfaces (Figure [200]8C‐D). Most of these metastases were weakly attached to the organs. Large tumors were also detected in fatty tissues, such as the omentum, ovarian fat pad, and mesentery (Figure [201]8C‐D). In particular, larger tumor masses attached to the small and large intestines lead to adhesion between intestinal structures, implicating strictures of the gut. The IVIS measurements of the omentum and intestine revealed the most prominent signals, correlating with the histological results (Figure [202]8D‐E). In the mice treated with p53‐mRNA, gross anatomical and histological analyses did not reveal tumor masses in the peritoneal cavity, and all the organs appeared normal upon visual inspection. The IVIS measurements and histological inspection did not provide evidence of tumor cell dissemination (Figure [203]8D‐E). 4. DISCUSSION In this study, we sought to restore p53 functionality in HGSOC using IVT p53 mRNA. Re‐expression of WT p53 resulted in a significant reduction in cell proliferation, induction of apoptosis, and perturbations in cell cycle dynamics in the HGSOC cells. The liposomal delivery system demonstrated high transfection efficiency and sustained mRNA expression. Furthermore, the p53 IVT‐mRNA transfection in primary HGSOC cells yielded similar outcomes, including cell cycle arrest, apoptosis induction, and diminished cell viability, compared to normal ovarian cells. Transcriptome analysis revealed notable alterations in pathways related to p53 signaling, apoptosis, and cell cycle regulation. In an in vivo setting, p53 IVT mRNA treatment effectively inhibited tumor growth and prevented tumor cell dissemination within the peritoneal cavity. These findings underscore the potential utility of mRNA‐based strategies for reactivating p53 and combating HGSOC by inducing programmed cell death, orchestrating cell cycle checkpoints, and reducing tumor progression and metastasis. Mutations in p53 occur in up to 96% of patients with HGSOC [[204]13]. Researchers have attempted to reactivate p53 and suppress OC growth using WT p53 gene therapy [[205]56]. However, therapies using WT Ad‐p53 (Gendicine, Advexin, SCH‐58500), liposomal WT p53, or p53‐specific conditionally replicative adenoviral vectors have not yet been approved as p53‐based drugs for HGSOC therapy. Recent studies have demonstrated that novel IVT‐mRNA‐based therapeutics are powerful tools for developing novel vaccines [[206]57]. Here, we explored the potential role of IVT mRNA for the reactivation of tumor suppressor functions in cancer. We first examined the functional aspects of mRNA expression in HGSOC cells using different IVT mRNA species (EGFP, luciferase, and Flag‐p53). The translation of eukaryotic mRNA is a precisely controlled procedure in which the 5’ Cap, 3’, and 5’ untranslated regions (UTRs) contribute significantly to the stability of the mRNA [[207]26]. To reach this aim, we used here mRNA co‐transcriptionally capped with m7G(5’)ppp(5’) (2’OMeA)pG. The 5’ UTR contains an eIF4G aptamer that was previously shown to improve the translation efficacy of IVT mRNA. Additionally, in all the three mRNA used here, the four canonical bases (ACGU) were replaced by ACGPseudo‐U, which was found to improve the functionality of IVT mRNA [[208]58]. Our IVT‐EGFP‐mRNA with optimized structural features achieved 72%‐99% expression in the HGSOC cell lines and up to 83% expression in the primary cells using liposomal transfection. High EGFP expression (≥ 90%) lasted over 10 days in the cell lines and 4‐11 days in 2D‐cultured primary cells and in tumor organoids for 5 days, suggesting that liposomal IVT‐mRNA transfection enables efficient, long‐lasting expression in HGSOC cells. Interestingly, comparison of the primary HGSOC samples with their normal counterparts, both transfected with equal doses, showed much higher p53 expression in the tumor cells, resulting in more robust anti‐proliferative effects in tumor cells. The p53 was previously found to be present at low levels in resting cells, but multiple cancer‐related stress stimuli, such as hyper‐induction of the HSP90 system, provoke post‐translational modifications of p53 to modulate its protein stability [[209]59]. Moreover, mutant p53 has been reported to be involved in the metabolic adaptation of cancer cells by sustaining the Warburg effect. By increasing glucose intake in tumor cells, mutant p53 hinders autophagy‐dependent degradation caused by glucose deprivation [[210]60]. Thus, the reprogramming of cancer cells increases the stability of mutant p53 through different mechanisms. Exogenous WT p53 expressed upon mRNA transfection may utilize these mechanisms for stabilization. Our findings support the idea that restoring WT p53 function induces cell cycle arrest, apoptosis, and tumor suppression [[211]61]. Sequencing of RNA revealed coordinated upregulation of multiple p53 target genes promoting cell death, including p21, MDM2, PUMA, GADD45B, FAS, BAX, and FBXW7, and downregulation of centromere‐related components such as BUB1, BUB1B, CDC20, APC, and PLK1, along with crucial regulators of cell cycle progression (CDK1/Cyclin B1, CDK4/Cyclin D, CDK2/Cyclin E, PLK1, and PLK4), providing evidence for deregulated cell cycle machinery and cell division. Importantly, these key targets agree with those of ongoing clinical trials investigating CDK1 and PLK1 inhibitors (ribociclib, rocovitine, palbociclib, and volasertib) alone or in combination with standard chemotherapy for OC treatment [[212]62]. Thus, p53 reactivation exhibits pleiotropic effects; among the most important effects is the simultaneous inhibition of multiple OC‐relevant kinases. Studies have linked functional loss of p53 to aneuploidy in vitro and In vivo [[213]29, [214]63–[215]65]. However, recent studies have suggested that the concurrent loss of p53 and p73 directly affects CIN in cancer cells [[216]66, [217]67, [218]68]. Notably, both transcription factors exhibit shared target genes, among which CDKN1A (p21) is critical for the inhibition of CDK1 and CDK2. However, p21 contributes to cell cycle arrest in the G1 and G2 phases, and its absence introduces various mitotic aberrations in tumor cells [[219]51, [220]69, [221]70]. Schmidt et al. [[222]71] revealed that the inactivation of p53/p73 or the absence of p21 drives CIN in a CDK1‐dependent manner. The loss of p21 unleashes CDK1 activity, leading to abnormal rates of microtubule assembly and subsequent CIN in cancer cells, which typically maintain CIN. Interestingly, our transcriptomic data revealed that restoring functional p53 led to significant upregulation of p73 and CDKN1A gene expression. Restoration of the p53/p73 and p21 axes proved sufficient to mitigate or at least partially reverse the observed CIN in surviving parental OVCAR‐8 cancer cells and highlights the connection between these factors and CIN. Moreover, a recent report indicated that p53 activation enhances DNA replication efficiency, whereas its elimination impedes replication fork progression [[223]72]. Interestingly, MDM2, a gene product induced by p53, also supports DNA replication even in p53‐deficient cells, suggesting that sustained MDM2 expression is one of the mechanisms by which p53 prevents replicative stress [[224]72]. Thus, in addition to its well‐known role in eliminating damaged cells, p53 contributes to genome protection during S‐phase by preventing the appearance of stalled or collapsed replication forks by preventing DNA damage during replication. Furthermore, cisplatin‐induced apoptosis is enhanced by p73 overexpression [[225]73, [226]74]. Therefore, IVT‐p53‐mRNA delivery can be used with carboplatin, a standard therapeutic agent for HGSOC patients [[227]75]. Given the high rate of p53 mutations in HGSOC, restoring p53 function may make the cells more susceptible to carboplatin, providing an important treatment alternative for further investigation. We tested 2 xenograft models and demonstrated a dose‐dependent reduction in p53‐mRNA during tumor growth. Dissemination of mock‐transfected cells in our orthotopic model mirrored the dissemination in HGSOC cancer patients [[228]76]. The p53‐mRNA treatment prior to orthotopic injection hampered tumor cell growth in a dose‐dependent manner within the ovaries and spread to the organs of the peritoneal cavity, demonstrating the efficacy of p53‐mRNA under orthotopic conditions. The usefulness of p53‐mRNA treatment was also corroborated in a second xenograft experiment in which HGSOC cells were directly administered to the peritoneal cavity. A significant advantage of intraperitoneal chemotherapy is that it prevents hepatotoxicity and allows the metabolism of cancer drugs. Upon intraperitoneal administration of liposomal p53‐mRNA, a massive dose‐dependent reduction in tumor cell growth and dissemination was observed. The absence of weight loss and detailed necropsy revealed no tissue damage, supporting the nontoxic behavior of p53‐mRNA. No approved drugs exist to restore functional p53 in cancer cells expressing mutant p53. Overcoming this challenge is crucial, particularly for HGSOC. However, p53 gene therapy failed in OC clinical trials [[229]56] probably because of neutralizing antibodies against viral delivery systems and the dominant‐negative effect of the endogenous mutant p53. To explore this aspect, a detailed analysis of p53 tetramers in cells treated with IVT‐p53‐mRNA should be the subject of future biochemical investigations. New‐generation IVT‐mRNAs with optimized translational efficacy, delivered as liposomal IVT‐mRNA, show promise for bypassing these obstacles. The IVT‐mRNA offers several advantages over DNA‐based drugs, including a high transfection efficiency, no risk of insertional mutagenesis, and functional activity without nuclear entry. However, the reactivation of p53 in cancer therapy faces certain hurdles. Off‐target effects and immune responses pose risks requiring further detailed research. Long‐term safety concerns, potential resistance, and ethical considerations must be addressed. Combating the evolution of cancer cell resistance remains challenging. Additionally, the costs and clinical efficacy across diverse cancer types and stages are complex. Overcoming these hurdles demands ongoing research, technological advancements, and rigorous clinical trials to ensure the safe and effective reactivation of tumor suppressor genes in cancer treatment. Nevertheless, this study supports IVT‐mRNA as an attractive tool for reactivating p53 in susceptible cancers for effective cancer therapy. 5. CONCLUSION This study demonstrated the effectiveness of the reactivation of the TP53 tumor suppressor gene in HGSOC by IVT mRNA. Liposomal IVT mRNA resulted in efficient and sustained expression of WT p53 and precise rescue of p53 function; thus, inhibiting cell proliferation in HGSOC cells, reducing chromosomal instability, and triggering cell death. In different mouse models, p53‐mRNA treatment led to a dose‐dependent reduction in tumor growth and dissemination within the peritoneal cavity. These findings suggested that IVT‐mRNA‐based approaches hold promise for reactivating p53 in cancer cells and may offer a valuable therapeutic option for HGSOC. DECLARATIONS AUTHOR CONTRIBUTIONS Monika Raab and Klaus Strebhardt conceptualized the study. Monika Raab, Samuel Peña‐Llopis, Mourad Sanhaji, Daniela Fitz, Monika Kressin, Sven Becker, and Klaus Strebhardt coordinated the study and wrote the manuscript. Monika Raab, Izabela Kostova, Samuel Peña‐Llopis, Daniela Fitz, Monika Kressin, Seyed Mohsen Aberoumandi, Evelyn Ullrich, Mourad Sanhaji, and Klaus Strebhardt designed and performed experiments, interpreted results, and prepared figures. All the authors have read and approved the final version of the manuscript. CONFLICT OF INTEREST STATEMENT The authors declare no potential conflict of interest. CONSENT FOR PUBLICATION Not applicable. ETHICS APPROVAL AND CONSENT TO PARTICIPATE All tissues were obtained from patients who had undergone surgery at the University Hospital of Goethe University, with the consent of the patients and approval of the Goethe University Committee for Ethical Review of Research involving Human Subjects (approval number: SGO‐1‐2017). All the patients signed an informed consent form. All animal experiments were approved by the regional council of Darmstadt (V 54‐19 c 18‐FK/1128). Supporting information Supporting Information [230]Click here for additional data file.^ (12.8MB, docx) ACKNOWLEDGEMENTS