International Journal of Molecular Sciences the Potential of Targeting Ribosome Biogenesis in High-grade Serous Ovarian Cancer

Overall survival for patients with ovarian cancer (OC) has shown little improvement for decades meaning new therapeutic options are critical. OC comprises multiple histological subtypes, of which the most common and aggressive subtype is high-grade serous ovarian cancer (HGSOC). HGSOC is characterized by genomic structural variations with relatively few recurrent somatic mutations or dominantly acting oncogenes that can be targeted for the development of novel therapies. However, deregulation of pathways controlling homologous recombination (HR) and ribosome biogenesis has been observed in a high proportion of HGSOC, raising the possibility that targeting these basic cellular processes may provide improved patient outcomes. The poly (ADP-ribose) polymerase (PARP) inhibitor olaparib has been approved to treat women with defects in HR due to germline BRCA mutations. Recent evidence demonstrated the efficacy of targeting ribosome biogenesis with the specific inhibitor of ribosomal RNA synthesis, CX-5461 in v-myc avian myelocytomatosis viral oncogene homolog (MYC)-driven haematological and prostate cancers. CX-5461 has now progressed to a phase I clinical trial in patients with haematological malignancies and phase I/II trial in breast cancer. Here we review the currently available targeted therapies for HGSOC and discuss the potential of targeting ribosome biogenesis as a novel therapeutic approach against HGSOC.

[1]  Ignace Vergote,et al.  Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. , 2016, The New England journal of medicine.

[2]  K. Khanna,et al.  Inhibition of RNA polymerase I transcription initiation by CX-5461 activates non-canonical ATM/ATR signaling , 2016, Oncotarget.

[3]  R. Pearson,et al.  The Dual Inhibition of RNA Pol I Transcription and PIM Kinase as a New Therapeutic Approach to Treat Advanced Prostate Cancer , 2016, Clinical Cancer Research.

[4]  Y. Drew,et al.  Homologous recombination deficiency and ovarian cancer. , 2016, European journal of cancer.

[5]  K. Wiman,et al.  Strong synergy with APR-246 and DNA-damaging drugs in primary cancer cells from patients with TP53 mutant High-Grade Serous ovarian cancer , 2016, Journal of Ovarian Research.

[6]  G. Narayanan,et al.  Targeted agents in epithelial ovarian cancer: review on emerging therapies and future developments , 2016, Ecancermedicalscience.

[7]  L. Montanaro,et al.  Direct relationship between the level of p53 stabilization induced by rRNA synthesis-inhibiting drugs and the cell ribosome biogenesis rate , 2016, Oncogene.

[8]  G. Shapiro,et al.  Homologous Recombination Deficiency: Exploiting the Fundamental Vulnerability of Ovarian Cancer. , 2015, Cancer discovery.

[9]  V. Beral,et al.  Rethinking ovarian cancer II: reducing mortality from high-grade serous ovarian cancer , 2015, Nature Reviews Cancer.

[10]  P. Brown,et al.  Transient rRNA synthesis inhibition with CX-5461 is sufficient to elicit growth arrest and cell death in acute lymphoblastic leukemia cells , 2015, Oncotarget.

[11]  Kylie L. Gorringe,et al.  Mutational landscape of mucinous ovarian carcinoma and its neoplastic precursors , 2015, Genome Medicine.

[12]  Jacobus Pfisterer,et al.  Standard chemotherapy with or without bevacizumab for women with newly diagnosed ovarian cancer (ICON7): overall survival results of a phase 3 randomised trial , 2015, The Lancet. Oncology.

[13]  R. Pearson,et al.  The nucleolus as a fundamental regulator of the p53 response and a new target for cancer therapy. , 2015, Biochimica et biophysica acta.

[14]  G. Thomas,et al.  A liaison between mTOR signaling, ribosome biogenesis and cancer. , 2015, Biochimica et biophysica acta.

[15]  P. Thaker,et al.  A phase II evaluation of selumetinib (AZD6244, ARRY-142886), a selective MEK-1/2 inhibitor in the treatment of recurrent or persistent endometrial cancer: an NRG Oncology/Gynecologic Oncology Group study. , 2015, Gynecologic oncology.

[16]  P. Brown,et al.  rRNA synthesis inhibitor, CX-5461, activates ATM/ATR pathway in acute lymphoblastic leukemia, arrests cells in G2 phase and induces apoptosis , 2015, Oncotarget.

[17]  Joshy George,et al.  Whole–genome characterization of chemoresistant ovarian cancer , 2015, Nature.

[18]  G. McArthur,et al.  A phase 1, open-label, dose escalation, safety, PK and PD study of a first in class Pol1 inhibitor (CX-5461) in patients with advanced hematologic malignancies (HM). , 2015 .

[19]  C. Scott,et al.  Poly (ADP-ribose) polymerase inhibitors: recent advances and future development. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[20]  S. Mabuchi,et al.  The PI3K/AKT/mTOR pathway as a therapeutic target in ovarian cancer. , 2015, Gynecologic oncology.

[21]  C. Mathers,et al.  Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012 , 2015, International journal of cancer.

[22]  A. Leary,et al.  The PI3K/Akt/mTOR pathway in ovarian cancer: therapeutic opportunities and challenges , 2015, Chinese journal of cancer.

[23]  R. Hannan,et al.  Perturbations at the ribosomal genes loci are at the centre of cellular dysfunction and human disease , 2014, Cell & Bioscience.

[24]  J. Ledermann,et al.  Treatment options in recurrent ovarian cancer: latest evidence and clinical potential , 2014, Therapeutic advances in medical oncology.

[25]  D. Matei,et al.  Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. , 2014, The Lancet. Oncology.

[26]  R. Pearson,et al.  Targeting the nucleolus for cancer intervention. , 2014, Biochimica et biophysica acta.

[27]  A. Reuss,et al.  Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: The AURELIA open-label randomized phase III trial. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[28]  R. Hannan,et al.  Targeting RNA polymerase I to treat MYC-driven cancer , 2014, Oncogene.

[29]  C. Rubbi,et al.  Nucleolar control of p53: a cellular Achilles’ heel and a target for cancer therapy , 2014, Cellular and Molecular Life Sciences.

[30]  M. Burgess,et al.  BRCA 1/2-Mutation Related and Sporadic Breast and Ovarian Cancers: More Alike than Different , 2014, Front. Oncol..

[31]  C. Rommel,et al.  PI3K and cancer: lessons, challenges and opportunities , 2014, Nature Reviews Drug Discovery.

[32]  C. Bieberich,et al.  A targeting modality for destruction of RNA polymerase I that possesses anticancer activity. , 2014, Cancer cell.

[33]  D. Bowtell,et al.  Synergistic inhibition of ovarian cancer cell growth by combining selective PI3K/mTOR and RAS/ERK pathway inhibitors. , 2013, European journal of cancer.

[34]  R. Hannan,et al.  The nucleolus: an emerging target for cancer therapy. , 2013, Trends in molecular medicine.

[35]  C. Landen,et al.  The role of the fallopian tube in the origin of ovarian cancer. , 2013, American journal of obstetrics and gynecology.

[36]  R. Pearson,et al.  AKT signalling is required for ribosomal RNA synthesis and progression of Eμ‐Myc B‐cell lymphoma in vivo , 2013, The FEBS journal.

[37]  D. Eick,et al.  Functional ribosome biogenesis is a prerequisite for p53 destabilization: impact of chemotherapy on nucleolar functions and RNA metabolism , 2013, Biological chemistry.

[38]  Ling-Zhi Wang,et al.  Functional genomics identifies five distinct molecular subtypes with clinical relevance and pathways for growth control in epithelial ovarian cancer , 2013, EMBO molecular medicine.

[39]  R. Pearson,et al.  Dysregulation of the basal RNA polymerase transcription apparatus in cancer , 2013, Nature Reviews Cancer.

[40]  I. Willis,et al.  Regulation of pol III transcription by nutrient and stress signaling pathways. , 2013, Biochimica et biophysica acta.

[41]  R. Pearson,et al.  Dysregulation of RNA polymerase I transcription during disease. , 2013, Biochimica et biophysica acta.

[42]  M. Morgan,et al.  Selumetinib in women with recurrent low-grade serous carcinoma of the ovary or peritoneum: an open-label, single-arm, phase 2 study. , 2013, The Lancet. Oncology.

[43]  D. Ruggero Revisiting the Nucleolus: From Marker to Dynamic Integrator of Cancer Signaling , 2012, Science Signaling.

[44]  J. George,et al.  BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[45]  Carleen Cullinane,et al.  Inhibition of RNA polymerase I as a therapeutic strategy to promote cancer-specific activation of p53. , 2012, Cancer cell.

[46]  H. Hurwitz,et al.  A phase I study of bevacizumab, everolimus and panitumumab in advanced solid tumors , 2012, Cancer Chemotherapy and Pharmacology.

[47]  G. Thomas,et al.  Suprainduction of p53 by disruption of 40S and 60S ribosome biogenesis leads to the activation of a novel G2/M checkpoint. , 2012, Genes & development.

[48]  D. Matei,et al.  Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. , 2012, The New England journal of medicine.

[49]  Jacobus Pfisterer,et al.  A phase 3 trial of bevacizumab in ovarian cancer. , 2011, The New England journal of medicine.

[50]  D. Spandidos,et al.  Gene alterations in the PI3K/PTEN/AKT pathway as a mechanism of drug-resistance (review). , 2011, International journal of oncology.

[51]  J. Brenton,et al.  Evolution of platinum resistance in high-grade serous ovarian cancer. , 2011, The Lancet. Oncology.

[52]  S. Mok,et al.  The insulin-like growth factor 1 pathway is a potential therapeutic target for low-grade serous ovarian carcinoma. , 2011, Gynecologic oncology.

[53]  Ricky W Johnstone,et al.  AKT Promotes rRNA Synthesis and Cooperates with c-MYC to Stimulate Ribosome Biogenesis in Cancer , 2011, Science Signaling.

[54]  I. Shih,et al.  Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer--shifting the paradigm. , 2011, Human pathology.

[55]  Benjamin J. Raphael,et al.  Integrated Genomic Analyses of Ovarian Carcinoma , 2011, Nature.

[56]  Rodney J Hicks,et al.  In Vivo Activity of Combined PI3K/mTOR and MEK Inhibition in a KrasG12D;Pten Deletion Mouse Model of Ovarian Cancer , 2011, Molecular Cancer Therapeutics.

[57]  S. Pyndiah,et al.  c-MYC Suppresses BIN1 to Release Poly(ADP-Ribose) Polymerase 1: A Mechanism by Which Cancer Cells Acquire Cisplatin Resistance , 2011, Science Signaling.

[58]  R. Hannan,et al.  Targeting RNA polymerase I with an oral small molecule CX-5461 inhibits ribosomal RNA synthesis and solid tumor growth. , 2011, Cancer research.

[59]  R. Pearson,et al.  Signaling to the ribosome in cancer—It is more than just mTORC1 , 2011, IUBMB life.

[60]  G. McArthur,et al.  c-MYC coordinately regulates ribosomal gene chromatin remodeling and Pol I availability during granulocyte differentiation , 2010, Nucleic acids research.

[61]  David D. L. Bowtell,et al.  The genesis and evolution of high-grade serous ovarian cancer , 2010, Nature Reviews Cancer.

[62]  F. Boisvert,et al.  The Nucleolus under Stress , 2010, Molecular Cell.

[63]  Maria P. Pavlou,et al.  Integrating high-throughput technologies in the quest for effective biomarkers for ovarian cancer , 2010, Nature Reviews Cancer.

[64]  Carlos Caldas,et al.  Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary , 2010, The Journal of pathology.

[65]  D. Felsher,et al.  MYC as a regulator of ribosome biogenesis and protein synthesis , 2010, Nature Reviews Cancer.

[66]  P. Hainaut,et al.  Mutant p53 reactivation by PRIMA-1MET induces multiple signaling pathways converging on apoptosis , 2010, Oncogene.

[67]  E. Kremmer,et al.  Chemotherapeutic Drugs Inhibit Ribosome Biogenesis at Various Levels* , 2010, The Journal of Biological Chemistry.

[68]  I. Grummt,et al.  The RNA polymerase I transcription machinery: an emerging target for the treatment of cancer. , 2010, Annual review of pharmacology and toxicology.

[69]  D. Ruggero The role of Myc-induced protein synthesis in cancer. , 2009, Cancer research.

[70]  Yanping Zhang,et al.  Signaling to p53: ribosomal proteins find their way. , 2009, Cancer cell.

[71]  I. Shih,et al.  Ovarian Low-grade and High-grade Serous Carcinoma: Pathogenesis, Clinicopathologic and Molecular Biologic Features, and Diagnostic Problems , 2009, Advances in anatomic pathology.

[72]  Robert C. Bast,et al.  The biology of ovarian cancer: new opportunities for translation , 2009, Nature Reviews Cancer.

[73]  Jan Bergman,et al.  PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. , 2009, Cancer cell.

[74]  V. Velculescu,et al.  Frequent activating mutations of PIK3CA in ovarian clear cell carcinoma. , 2009, The American journal of pathology.

[75]  R. Memmott,et al.  Akt-dependent and -independent mechanisms of mTOR regulation in cancer. , 2009, Cellular signalling.

[76]  A. Wright,et al.  c-Myc induces changes in higher order rDNA structure on stimulation of quiescent cells , 2009, Oncogene.

[77]  R. Penson,et al.  Ovarian stromal and germ cell tumors. , 2009, Seminars in oncology.

[78]  Matthew Burnell,et al.  Sensitivity and specificity of multimodal and ultrasound screening for ovarian cancer, and stage distribution of detected cancers: results of the prevalence screen of the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) , 2009, Journal of Family Planning and Reproductive Health Care.

[79]  Timothy A. Yap,et al.  Beyond chemotherapy: targeted therapies in ovarian cancer , 2009, Nature Reviews Cancer.

[80]  E. Raymond,et al.  Safety and pharmacokinetics of paclitaxel and the oral mTOR inhibitor everolimus in advanced solid tumours , 2009, British Journal of Cancer.

[81]  K. Vousden,et al.  Cooperation between the ribosomal proteins L5 and L11 in the p53 pathway , 2008, Oncogene.

[82]  Pier Paolo Pandolfi,et al.  The PTEN–PI3K pathway: of feedbacks and cross-talks , 2008, Oncogene.

[83]  R. Tothill,et al.  Novel Molecular Subtypes of Serous and Endometrioid Ovarian Cancer Linked to Clinical Outcome , 2008, Clinical Cancer Research.

[84]  B. Karlan,et al.  Secondary BRCA1 mutations in BRCA1-mutated ovarian carcinomas with platinum resistance. , 2008, Cancer research.

[85]  F. Couch,et al.  Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers , 2008, Nature.

[86]  Anil K Sood,et al.  Early events in the pathogenesis of epithelial ovarian cancer. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[87]  K. Vousden,et al.  Coping with stress: multiple ways to activate p53 , 2007, Oncogene.

[88]  J. Krischer,et al.  BRCA1 and BRCA2 mutations account for a large proportion of ovarian carcinoma cases , 2005, Cancer.

[89]  J. Testa,et al.  Perturbations of the AKT signaling pathway in human cancer , 2005, Oncogene.

[90]  Alan Ashworth,et al.  Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy , 2005, Nature.

[91]  Carla Grandori,et al.  c-Myc binds to human ribosomal DNA and stimulates transcription of rRNA genes by RNA polymerase I , 2005, Nature Cell Biology.

[92]  Lars-Gunnar Larsson,et al.  c-Myc associates with ribosomal DNA and activates RNA polymerase I transcription , 2005, Nature Cell Biology.

[93]  R. Pearson,et al.  MAD1 and c‐MYC regulate UBF and rDNA transcription during granulocyte differentiation , 2004, The EMBO journal.

[94]  Andrew K Godwin,et al.  AKT and mTOR phosphorylation is frequently detected in ovarian cancer and can be targeted to disrupt ovarian tumor cell growth , 2004, Oncogene.

[95]  E. Schmidt The role of c-myc in regulation of translation initiation , 2004, Oncogene.

[96]  M. Jordan,et al.  Microtubules as a target for anticancer drugs , 2004, Nature Reviews Cancer.

[97]  Brigitte M. Ronnett,et al.  The Histologic Type and Stage Distribution of Ovarian Carcinomas of Surface Epithelial Origin , 2004, International journal of gynecological pathology : official journal of the International Society of Gynecological Pathologists.

[98]  Z. Siddik,et al.  Cisplatin: mode of cytotoxic action and molecular basis of resistance , 2003, Oncogene.

[99]  Hys Ngan,et al.  Carcinoma of the Ovary , 2003, International journal of gynaecology and obstetrics: the official organ of the International Federation of Gynaecology and Obstetrics.

[100]  R. Agarwal,et al.  Ovarian cancer: strategies for overcoming resistance to chemotherapy , 2003, Nature Reviews Cancer.

[101]  Ed Hurt,et al.  Pre-ribosomes on the road from the nucleolus to the cytoplasm. , 2003, Trends in cell biology.

[102]  P. Pandolfi,et al.  Does the ribosome translate cancer? , 2003, Nature Reviews Cancer.

[103]  I. Grummt,et al.  ERK-dependent phosphorylation of the transcription initiation factor TIF-IA is required for RNA polymerase I transcription and cell growth. , 2003, Molecular cell.

[104]  Mef Nilbert,et al.  Distinct sets of gene alterations in endometrial carcinoma implicate alternate modes of tumorigenesis , 2002, Cancer.

[105]  D. Leary,et al.  Regulation of ribosome biogenesis within the nucleolus , 2001, FEBS letters.

[106]  R. Hannan,et al.  An immediate response of ribosomal transcription to growth factor stimulation in mammals is mediated by ERK phosphorylation of UBF. , 2001, Molecular cell.

[107]  J. Levine,et al.  Surfing the p53 network , 2000, Nature.

[108]  D. Treré,et al.  Nucleolar size indicates the rapidity of cell proliferation in cancer tissues , 2000, The Journal of pathology.

[109]  M. Bookman,et al.  Second-line treatment of ovarian cancer. , 2000, The oncologist.

[110]  Emmett V Schmidt,et al.  The role of c-myc in cellular growth control , 1999, Oncogene.

[111]  L. Montanaro,et al.  Nucleolar function and size in cancer cells. , 1998, The American journal of pathology.

[112]  R. Bast,et al.  A radioimmunoassay using a monoclonal antibody to monitor the course of epithelial ovarian cancer. , 1983, The New England journal of medicine.

[113]  R. Pearson,et al.  Combination Therapy Targeting Ribosome Biogenesis and mRNA Translation Synergistically Extends Survival in MYC-Driven Lymphoma. , 2016, Cancer discovery.

[114]  J. Coward,et al.  New perspectives on targeted therapy in ovarian cancer , 2015 .

[115]  D. Gershenson The life and times of low-grade serous carcinoma of the ovary. , 2013, American Society of Clinical Oncology educational book. American Society of Clinical Oncology. Annual Meeting.

[116]  J. Thigpen Olaparib Maintenance Therapy in Platinum-Sensitive Relapsed Ovarian Cancer , 2012 .

[117]  J. Dungan Association Between BRCA1 and BRCA2 Mutations and Survival in Women With Invasive Epithelial Ovarian Cancer , 2012 .

[118]  E. Friedman,et al.  Effect of BRCA1/2 mutations on long-term survival of patients with invasive ovarian cancer: the national Israeli study of ovarian cancer. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[119]  I. Wierstra,et al.  The c-myc promoter: still MysterY and challenge. , 2008, Advances in cancer research.

[120]  Celia Quevedo,et al.  Biochemical mechanisms of cisplatin cytotoxicity. , 2007, Anti-cancer agents in medicinal chemistry.

[121]  T. Moss,et al.  A housekeeper with power of attorney: the rRNA genes in ribosome biogenesis , 2006, Cellular and Molecular Life Sciences.

[122]  P Maisonneuve,et al.  Carcinoma of the ovary. FIGO 26th Annual Report on the Results of Treatment in Gynecological Cancer. , 2006, International journal of gynaecology and obstetrics: the official organ of the International Federation of Gynaecology and Obstetrics.