Rescue of p53 functions by in vitro-transcribed mRNA impedes the growth of high-grade serous ovarian cancer

Monika Raab, Izabela Kostova, Samuel Peña-Llopis, Daniela Fietz, Monika Kressin, Seyed Mohsen Aberoumandi, Evelyn Ullrich, Sven Becker, Mourad Sanhaji, Klaus Strebhardt

PDF
Cancer Communications ›› 2024, Vol. 44 ›› Issue (1) : 101-126. DOI: 10.1002/cac2.12511
ORIGINAL ARTICLE

Rescue of p53 functions by in vitro-transcribed mRNA impedes the growth of high-grade serous ovarian cancer

Author information +
History +

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

Cite this article

Download citation ▾
Monika Raab, Izabela Kostova, Samuel Peña-Llopis, Daniela Fietz, Monika Kressin, Seyed Mohsen Aberoumandi, Evelyn Ullrich, Sven Becker, Mourad Sanhaji, Klaus Strebhardt. Rescue of p53 functions by in vitro-transcribed mRNA impedes the growth of high-grade serous ovarian cancer. Cancer Communications, 2024, 44(1): 101‒126 https://doi.org/10.1002/cac2.12511

References

[1]
Luo Z, Wang Q, Lau WB, Lau B, Xu L, Zhao L, et al. Tumor microenvironment: The culprit for ovarian cancer metastasis? Cancer Lett. 2016;377(2):174–182.
[2]
Yang Z, Wang W, Zhao L, Wang X, Gimple RC, Xu L, et al. Plasma cells shape the mesenchymal identity of ovarian cancers through transfer of exosome-derived microRNAs. Sci Adv. 2021;7(9):eabb0737.
[3]
Bristow RE, del Carmen MG, Kaufman HS, Montz FJ. Radical oophorectomy with primary stapled colorectal anastomosis for resection of locally advanced epithelial ovarian cancer. J Am Coll Surg. 2003;197(4):565–574.
[4]
Guppy AE, Nathan PD, Rustin GJ. Epithelial ovarian cancer: a review of current management. Clin Oncol (R Coll Radiol). 2005;17(6):399–411.
[5]
Wimberger P, Lehmann N, Kimmig R, Burges A, Meier W, Du Bois A, et al. Prognostic factors for complete debulking in advanced ovarian cancer and its impact on survival. An exploratory analysis of a prospectively randomized phase III study of the Arbeitsgemeinschaft Gynaekologische Onkologie Ovarian Cancer Study Group (AGO-OVAR). Gynecol Oncol. 2007;106(1):69–74.
[6]
Garcia A, Singh H. Bevacizumab and ovarian cancer. Ther Adv Med Oncol. 2013;5(2):133–141.
[7]
Lee JM, Ledermann JA, Kohn EC. PARP Inhibitors for BRCA1/2 mutation-associated and BRCA-like malignancies. Ann Oncol. 2014;25(1):32–40.
[8]
Romero I, Bast RC, Minireview: human ovarian cancer: biology, current management, and paths to personalizing therapy. Endocrinology. 2012;153(4):1593–1602.
[9]
Lane DP, Crawford LV. T antigen is bound to a host protein in SV40-transformed cells. Nature. 1979;278(5701):261–263.
[10]
Alexandrova EM, Yallowitz AR, Li D, Xu S, Schulz R, Proia DA, et al. Improving survival by exploiting tumour dependence on stabilized mutant p53 for treatment. Nature. 2015;523(7560):352–356.
[11]
Levine AJ, Oren M. The first 30 years of p53: growing ever more complex. Nat Rev Cancer. 2009;9(10):749–758.
[12]
Bieging KT, Mello SS, Attardi LD. Unravelling mechanisms of p53-mediated tumour suppression. Nat Rev Cancer. 2014;14(5):359–370.
[13]
Cancer Genome Atlas Research N. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474(7353):609–615.
[14]
Bernardini MQ, Baba T, Lee PS, Barnett JC, Sfakianos GP, Secord AA, et al. Expression signatures of TP53 mutations in serous ovarian cancers. BMC Cancer. 2010;10:237.
[15]
Bykov VJN, Eriksson SE, Bianchi J, Wiman KG. Targeting mutant p53 for efficient cancer therapy. Nat Rev Cancer. 2018;18(2):89–102.
[16]
Sriraman A, Radovanovic M, Wienken M, Najafova Z, Li Y, Dobbelstein M. Cooperation of Nutlin-3a and a Wip1 inhibitor to induce p53 activity. Oncotarget. 2016;7(22):31623–31638.
[17]
Lehmann S, Bykov VJ, Ali D, Andren O, Cherif H, Tidefelt U, et al. Targeting p53 in vivo: a first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J Clin Oncol. 2012;30(29):3633–3639.
[18]
Kreis NN, Sanhaji M, Kramer A, Sommer K, Rodel F, Strebhardt K, et al. Restoration of the tumor suppressor p53 by downregulating cyclin B1 in human papillomavirus 16/18-infected cancer cells. Oncogene. 2010;29(41):5591–5603.
[19]
Joerger AC, Bauer MR, Wilcken R, Baud MGJ, Harbrecht H, Exner TE, et al. Exploiting Transient Protein States for the Design of Small-Molecule Stabilizers of Mutant p53. Structure. 2015;23(12):2246–2255.
[20]
Yu X, Vazquez A, Levine AJ, Carpizo DR. Allele-specific p53 mutant reactivation. Cancer Cell. 2012;21(5):614–625.
[21]
Floquet C, Deforges J, Rousset JP, Bidou L. Rescue of non-sense mutated p53 tumor suppressor gene by aminoglycosides. Nucleic Acids Res. 2011;39(8):3350–3362.
[22]
Joerger AC, Fersht AR. The p53 Pathway: Origins, Inactivation in Cancer, and Emerging Therapeutic Approaches. Annu Rev Biochem. 2016;85:375–404.
[23]
Liu DS, Duong CP, Haupt S, Montgomery KG, House CM, Azar WJ, et al. Inhibiting the system xC(-)/glutathione axis selectively targets cancers with mutant-p53 accumulation. Nat Commun. 2017;8:14844.
[24]
Peng X, Zhang MQ, Conserva F, Hosny G, Selivanova G, Bykov VJ, et al. APR-246/PRIMA-1(MET) inhibits thioredoxin reductase 1 and converts the enzyme to a dedicated NADPH oxidase. Cell Death Dis. 2017;8(4):e2751.
[25]
Qin S, Tang X, Chen Y, Chen K, Fan N, Xiao W, et al. mRNA-based therapeutics: powerful and versatile tools to combat diseases. Signal Transduct Target Ther. 2022;7(1):166.
[26]
Sahin U, Kariko K, Tureci O. mRNA-based therapeutics–developing a new class of drugs. Nat Rev Drug Discov. 2014;13(10):759–780.
[27]
Uchida S, Kinoh H, Ishii T, Matsui A, Tockary TA, Takeda KM, et al. Systemic delivery of messenger RNA for the treatment of pancreatic cancer using polyplex nanomicelles with a cholesterol moiety. Biomaterials. 2016;82:221–228.
[28]
Zhang R, Men K, Zhang X, Huang R, Tian Y, Zhou B, et al. Delivery of a Modified mRNA Encoding IL-22 Binding Protein (IL-22BP) for Colon Cancer Gene Therapy. J Biomed Nanotechnol. 2018;14(7):1239–1251.
[29]
Bronder D, Tighe A, Wangsa D, Zong D, Meyer TJ, Wardenaar R, et al. TP53 loss initiates chromosomal instability in fallopian tube epithelial cells. Dis Model Mech. 2021;14(11):dmm049001.
[30]
McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM, et al. REporting recommendations for tumor MARKer prognostic studies (REMARK). Nat Clin Pract Urol. 2005;2(8):416–422.
[31]
Matthess Y, Kappel S, Spankuch B, Zimmer B, Kaufmann M, Strebhardt K. Conditional inhibition of cancer cell proliferation by tetracycline-responsive, H1 promoter-driven silencing of PLK1. Oncogene. 2005;24(18):2973–2980.
[32]
Raab M, Kappel S, Kramer A, Sanhaji M, Matthess Y, Kurunci-Csacsko E, et al. Toxicity modelling of Plk1-targeted therapies in genetically engineered mice and cultured primary mammalian cells. Nat Commun. 2011;2:395.
[33]
Mergener S, Siveke JT, Pena-Llopis S. Monosomy 3 Is Linked to Resistance to MEK Inhibitors in Uveal Melanoma. Int J Mol Sci. 2021;22(13):6727.
[34]
Tseng J-C, Vasquez KO, Peterson JD, editors. Optical Imaging on the IVIS SpectrumCT System: General and Technical Considerations for 2 D and 3 D Imaging, 2015.
[35]
Bialkowska AB, Ghaleb AM, Nandan MO, Yang VW. Improved Swiss-rolling Technique for Intestinal Tissue Preparation for Immunohistochemical and Immunofluorescent Analyses. J Vis Exp. 2016(113):54161.
[36]
Spankuch-Schmitt B, Bereiter-Hahn J, Kaufmann M, Strebhardt K. Effect of RNA silencing of polo-like kinase-1 (PLK1) on apoptosis and spindle formation in human cancer cells. J Natl Cancer Inst. 2002;94(24):1863–1877.
[37]
Petitjean A, Mathe E, Kato S, Ishioka C, Tavtigian SV, Hainaut P, et al. Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat. 2007;28(6):622–629.
[38]
Leroy B, Girard L, Hollestelle A, Minna JD, Gazdar AF, Soussi T. Analysis of TP53 mutation status in human cancer cell lines: a reassessment. Hum Mutat. 2014;35(6):756–765.
[39]
Mullany LK, Wong KK, Marciano DC, Katsonis P, King-Crane ER, Ren YA, et al. Specific TP53 Mutants Overrepresented in Ovarian Cancer Impact CNV, TP53 Activity, Responses to Nutlin-3a, and Cell Survival. Neoplasia. 2015;17(10):789–803.
[40]
Ikediobi ON, Davies H, Bignell G, Edkins S, Stevens C, O'Meara S, et al. Mutation analysis of 24 known cancer genes in the NCI-60 cell line set. Mol Cancer Ther. 2006;5(11):2606–2612.
[41]
Henningsen KM, Manzini V, Magerhans A, Gerber S, Dobbelstein M. MDM2-Driven Ubiquitination Rapidly Removes p53 from Its Cognate Promoters. Biomolecules. 2021;12(1):22.
[42]
Mitra AK, Davis DA, Tomar S, Roy L, Gurler H, Xie J, et al. In vivo tumor growth of high-grade serous ovarian cancer cell lines. Gynecol Oncol. 2015;138(2):372–377.
[43]
Bhowmik S, Bhowmick S, Maiti K, Chakra A, Shahi P, Rajamannar T. Phase-I randomized trial of doxorubicin hydrochloride liposome injection versus Caelyx((R)) in multiple myeloma. Leuk Lymphoma. 2018;59(6):1478–1481.
[44]
Tang Z, Feng W, Yang Y, Wang Q. Gemcitabine-loaded RGD modified liposome for ovarian cancer: preparation, characterization and pharmacodynamic studies. Drug Des Devel Ther. 2019;13:3281–3290.
[45]
Du M, Zhang S, Liu X, Xu C, Zhang X. Ploidy Status of Ovarian Cancer Cell Lines and Their Association with Gene Expression Profiles. Biomolecules. 2023;13(1):92.
[46]
Graf RP, Eskander R, Brueggeman L, Stupack DG. Association of Copy Number Variation Signature and Survival in Patients With Serous Ovarian Cancer. JAMA Netw Open. 2021;4(6):e2114162.
[47]
Delaney JR, Patel CB, Bapat J, Jones CM, Ramos-Zapatero M, Ortell KK, et al. Autophagy gene haploinsufficiency drives chromosome instability, increases migration, and promotes early ovarian tumors. PLoS Genet. 2020;16(1):e1008558.
[48]
McDermott JE, Arshad OA, Petyuk VA, Fu Y, Gritsenko MA, Clauss TR, et al. Proteogenomic Characterization of Ovarian HGSC Implicates Mitotic Kinases, Replication Stress in Observed Chromosomal Instability. Cell Rep Med. 2020;1(1):100004.
[49]
Roy S, Tomaszowski KH, Luzwick JW, Park S, Li J, Murphy M, et al. p53 orchestrates DNA replication restart homeostasis by suppressing mutagenic RAD52 and POLtheta pathways. Elife. 2018;7:e31723.
[50]
Liu Y, Parry JA, Chin A, Duensing S, Duensing A. Soluble histone H2AX is induced by DNA replication stress and sensitizes cells to undergo apoptosis. Mol Cancer. 2008;7:61.
[51]
Kreis NN, Sommer K, Sanhaji M, Kramer A, Matthess Y, Kaufmann M, et al. Long-term downregulation of Polo-like kinase 1 increases the cyclin-dependent kinase inhibitor p21(WAF1/CIP1). Cell Cycle. 2009;8(3):460–472.
[52]
Liu XS, Song B, Liu X. The substrates of Plk1, beyond the functions in mitosis. Protein Cell. 2010;1(11):999–1010.
[53]
Liu XS, Li H, Song B, Liu X. Polo-like kinase 1 phosphorylation of G2 and S-phase-expressed 1 protein is essential for p53 inactivation during G2 checkpoint recovery. EMBO Rep. 2010;11(8):626–632.
[54]
Arend RC, Londono-Joshi AI, Straughn JM,, Buchsbaum DJ. The Wnt/beta-catenin pathway in ovarian cancer: a review. Gynecol Oncol. 2013;131(3):772–779.
[55]
Morgan RJ,, Alvarez RD, Armstrong DK, Burger RA, Chen LM, Copeland L, et al. Ovarian cancer, version 2.2013. J Natl Compr Canc Netw. 2013;11(10):1199–1209.
[56]
Zeimet AG, Marth C. Why did p53 gene therapy fail in ovarian cancer? Lancet Oncol. 2003;4(7):415–422.
[57]
Lang F, Schrors B, Lower M, Tureci O, Sahin U. Identification of neoantigens for individualized therapeutic cancer vaccines. Nat Rev Drug Discov. 2022;21(4):261–282.
[58]
Tusup M, French LE, De Matos M, Gatfield D, Kundig T, Pascolo S. Design of in vitro Transcribed mRNA Vectors for Research and Therapy. Chimia (Aarau). 2019;73(6):391–394.
[59]
Mantovani F, Collavin L, Del Sal G. Mutant p53 as a guardian of the cancer cell. Cell Death Differ. 2019;26(2):199–212.
[60]
Rodriguez OC, Choudhury S, Kolukula V, Vietsch EE, Catania J, Preet A, et al. Dietary downregulation of mutant p53 levels via glucose restriction: mechanisms and implications for tumor therapy. Cell Cycle. 2012;11(23):4436–4446.
[61]
Ventura A, Kirsch DG, McLaughlin ME, Tuveson DA, Grimm J, Lintault L, et al. Restoration of p53 function leads to tumour regression in vivo. Nature. 2007;445(7128):661–665.
[62]
Lin ZP, Zhu YL, Ratner ES. Targeting Cyclin-Dependent Kinases for Treatment of Gynecologic Cancers. Front Oncol. 2018;8:303.
[63]
Tomasini R, Mak TW, Melino G. The impact of p53 and p73 on aneuploidy and cancer. Trends Cell Biol. 2008;18(5):244–252.
[64]
Tarapore P, Fukasawa K. p53 mutation and mitotic infidelity. Cancer Invest. 2000;18(2):148–155.
[65]
Zhang M, Zhuang G, Sun X, Shen Y, Wang W, Li Q, et al. TP53 mutation-mediated genomic instability induces the evolution of chemoresistance and recurrence in epithelial ovarian cancer. Diagn Pathol. 2017;12(1):16.
[66]
Bunz F, Fauth C, Speicher MR, Dutriaux A, Sedivy JM, Kinzler KW, et al. Targeted inactivation of p53 in human cells does not result in aneuploidy. Cancer Res. 2002;62(4):1129–1133.
[67]
Ertych N, Stolz A, Stenzinger A, Weichert W, Kaulfuss S, Burfeind P, et al. Increased microtubule assembly rates influence chromosomal instability in colorectal cancer cells. Nat Cell Biol. 2014;16(8):779–791.
[68]
Melino G, De Laurenzi V, Vousden KH. p73: Friend or foe in tumorigenesis. Nat Rev Cancer. 2002;2(8):605–615.
[69]
Warfel NA, El-Deiry WS. p21WAF1 and tumourigenesis: 20 years after. Curr Opin Oncol. 2013;25(1):52–58.
[70]
Medema RH, Klompmaker R, Smits VA, Rijksen G. p21waf1 can block cells at two points in the cell cycle, but does not interfere with processive DNA-replication or stress-activated kinases. Oncogene. 1998;16(4):431–441.
[71]
Schmidt AK, Pudelko K, Boekenkamp JE, Berger K, Kschischo M, Bastians H. The p53/p73 - p21(CIP1) tumor suppressor axis guards against chromosomal instability by restraining CDK1 in human cancer cells. Oncogene. 2021;40(2):436–451.
[72]
Klusmann I, Rodewald S, Muller L, Friedrich M, Wienken M, Li Y, et al. p53 Activity Results in DNA Replication Fork Processivity. Cell Rep. 2016;17(7):1845–1857.
[73]
Al-Bahlani S, Fraser M, Wong AY, Sayan BS, Bergeron R, Melino G, et al. P73 regulates cisplatin-induced apoptosis in ovarian cancer cells via a calcium/calpain-dependent mechanism. Oncogene. 2011;30(41):4219–4230.
[74]
Kim KC, Jung CS, Choi KH. Overexpression of p73 enhances cisplatin-induced apoptosis in HeLa cells. Arch Pharm Res. 2006;29(2):152–158.
[75]
du Bois A, Luck HJ, Meier W, Adams HP, Mobus V, Costa S, et al. A randomized clinical trial of cisplatin/paclitaxel versus carboplatin/paclitaxel as first-line treatment of ovarian cancer. J Natl Cancer Inst. 2003;95(17):1320–1329.
[76]
Yeung TL, Leung CS, Yip KP, Au Yeung CL, Wong ST, Mok SC. Cellular and molecular processes in ovarian cancer metastasis. A Review in the Theme: Cell and Molecular Processes in Cancer Metastasis. Am J Physiol Cell Physiol. 2015;309(7):C444–C456.

RIGHTS & PERMISSIONS

2023 2023 The Authors. Cancer Communications published by John Wiley & Sons Australia, Ltd. on behalf of Sun Yat-sen University Cancer Center.
PDF

Accesses

Citations

Detail

Sections
Recommended

/