527
Views
4
CrossRef citations to date
0
Altmetric
Review

Proteomics advances for precision therapy in ovarian cancer

, , , , , & show all
Pages 841-850 | Received 25 Jul 2019, Accepted 06 Sep 2019, Published online: 13 Sep 2019

References

  • Jayson GC, Kohn EC, Kitchener HC, et al. Ovarian cancer. Lancet. 2014;384(9951):1376–1388.
  • Labidi-Galy SI, Papp E, Hallberg D, et al. High grade serous ovarian carcinomas originate in the fallopian tube. Nat Commun. 2017;8(1):1093.
  • Cancer Genome Atlas Research, N. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474(7353):609–615.
  • Berger AC, Korkut A, Kanchi RS, et al. A comprehensive pan-cancer molecular study of gynecologic and breast cancers. Cancer Cell. 2018;33(4):690–705 e9.
  • Peng G, Mills GB. Surviving ovarian cancer: an affair between defective DNA repair and RB1. Clin Cancer Res. 2018;24(3):508–510.
  • Dimitrakopoulos L, Prassas I, Diamandis EP, et al. Onco-proteogenomics: multi-omics level data integration for accurate phenotype prediction. Crit Rev Clin Lab Sci. 2017;54(6):414–432.
  • Dimitrakopoulos L, Prassas I, Diamandis EP, et al. Proteogenomics: opportunities and caveats. Clin Chem. 2016;62(4):551–557.
  • Yarchoan M, Albacker LA, Hopkins AC, et al. PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers. JCI Insight. 2019;4(6):e126908.
  • Hennessy BT, Timms KM, Carey MS, et al. Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly (ADP ribose) polymerase inhibitors in ovarian cancer. J Clin Oncol. 2010;28(22):3570–3576.
  • Suzuki S, Moore DH 2nd, Ginzinger DG, et al. An approach to analysis of large-scale correlations between genome changes and clinical endpoints in ovarian cancer. Cancer Res. 2000;60(19):5382–5385.
  • Shayesteh L, Lu Y, Kuo WL, et al. PIK3CA is implicated as an oncogene in ovarian cancer. Nat Genet. 1999;21(1):99–102.
  • Eder AM, Sui X, Rosen DG, et al. Atypical PKCiota contributes to poor prognosis through loss of apical-basal polarity and cyclin E overexpression in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102(35):12519–12524.
  • Nanjundan M, Nakayama Y, Cheng KW, et al. Amplification of MDS1/EVI1 and EVI1, located in the 3q26.2 amplicon, is associated with favorable patient prognosis in ovarian cancer. Cancer Res. 2007;67(7):3074–3084.
  • Chaluvally-Raghavan P, Jeong KJ, Pradeep S, et al. Direct upregulation of STAT3 by microRNA-551b-3p deregulates growth and metastasis of ovarian cancer. Cell Rep. 2016;15(7):1493–1504.
  • Nanjundan M, Cheng KW, Zhang F, et al. Overexpression of SnoN/SkiL, amplified at the 3q26.2 locus, in ovarian cancers: a role in ovarian pathogenesis. Mol Oncol. 2008;2(2):164–181.
  • Yang JY, Yoshihara K, Tanaka K, et al. Predicting time to ovarian carcinoma recurrence using protein markers. J Clin Invest. 2013;123(9):3740–3750.
  • Chao A, Lai CH, Wang TH, et al. Genomic scar signatures associated with homologous recombination deficiency predict adverse clinical outcomes in patients with ovarian clear cell carcinoma. J Mol Med (Berl). 2018;96(6):527–536.
  • Ashley CW, Da Cruz Paula A, Kumar R, et al. Analysis of mutational signatures in primary and metastatic endometrial cancer reveals distinct patterns of DNA repair defects and shifts during tumor progression. Gynecol Oncol. 2019;152(1):11–19.
  • Peng G, Chun-Jen Lin C, Mo W, et al. Genome-wide transcriptome profiling of homologous recombination DNA repair. Nat Commun. 2014;5:3361.
  • Knijnenburg TA, Wang L, Zimmermann MT, et al. Genomic and molecular landscape of DNA damage repair deficiency across The Cancer Genome Atlas. Cell Rep. 2018;23(1):239–254 e6.
  • Abkevich V, Timms KM, Hennessy BT, et al. Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer. Br J Cancer. 2012;107(10):1776–1782.
  • Vogel C, Marcotte EM. Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet. 2012;13(4):227–232.
  • Goncalves E, Fragoulis A, Garcia-Alonso L, et al. Widespread post-transcriptional attenuation of genomic copy-number variation in cancer. Cell Syst. 2017;5(4):386–398 e4.
  • Zhang H, Liu T, Zhang Z, et al. Integrated proteogenomic characterization of human high-grade serous ovarian cancer. Cell. 2016;166(3):755–765.
  • Akbani R, Ng PK, Werner HM, et al. A pan-cancer proteomic perspective on The Cancer Genome Atlas. Nat Commun. 2014;5:3887.
  • Zhao W, Li J, Akbani R, et al. Credentialing individual samples for proteogenomic analysis. Mol Cell Proteomics. 2018;17(8):1515–1530.
  • Mertins P, Yang F, Liu T, et al. Ischemia in tumors induces early and sustained phosphorylation changes in stress kinase pathways but does not affect global protein levels. Mol Cell Proteomics. 2014;13(7):1690–1704.
  • Tanhaei S, Nikpour P, Ghaedi K, et al. RNA/protein discordant expression of Fndc5 in central nervous system is likely to be mediated through microRNAs. DNA Cell Biol. 2018;37(4):373–380.
  • Johansson HJ, Socciarelli F, Vacanti NM, et al. Breast cancer quantitative proteome and proteogenomic landscape. Nat Commun. 2019;10(1):1600.
  • Carey MS, Agarwal R, Gilks B, et al. Functional proteomic analysis of advanced serous ovarian cancer using reverse phase protein array: TGF-beta pathway signaling indicates response to primary chemotherapy. Clin Cancer Res. 2010;16(10):2852–2860.
  • Akbani R, Becker KF, Carragher N, et al. Realizing the promise of reverse phase protein arrays for clinical, translational, and basic research: a workshop report: the RPPA (Reverse Phase Protein Array) society. Mol Cell Proteomics. 2014;13(7):1625–1643.
  • Lu Y, Ling S, Hegde AM, et al. Using reverse-phase protein arrays as pharmacodynamic assays for functional proteomics, biomarker discovery, and drug development in cancer. Semin Oncol. 2016;43(4):476–483.
  • Labrie M, Kim TB, Ju Z, et al. Adaptive responses in a PARP inhibitor window of opportunity trial illustrate limited functional interlesional heterogeneity and potential combination therapy options. Oncotarget. 2019;10(37):3533–3546.
  • Le T, Faught W, Hopkins L, et al. Importance of CA125 normalization during neoadjuvant chemotherapy followed by planned delayed surgical debulking in patients with epithelial ovarian cancer. J Obstet Gynaecol Can. 2008;30(8):665–670.
  • Skaznik-Wikiel ME, Sukumvanich P, Beriwal S, et al. Possible use of CA-125 level normalization after the third chemotherapy cycle in deciding on chemotherapy regimen in patients with epithelial ovarian cancer: brief report. Int J Gynecol Cancer. 2011;21(6):1013–1017.
  • Cai RL, Yan-Neale Y, Cueto MA, et al. HDAC1, a histone deacetylase, forms a complex with Hus1 and Rad9, two G2/M checkpoint Rad proteins. J Biol Chem. 2000;275(36):27909–27916.
  • Yu KH, Levine DA, Zhang H, et al. Predicting ovarian cancer patients’ clinical response to platinum-based chemotherapy by their tumor proteomic signatures. J Proteome Res. 2016;15(8):2455–2465.
  • Peng X, Xu X, Wang Y, et al. A-to-I RNA editing contributes to proteomic diversity in cancer. Cancer Cell. 2018;33(4):817–828 e7.
  • Gonzalez VD, Samusik N, Chen TJ, et al. Commonly occurring cell subsets in high-grade serous ovarian tumors identified by single-cell mass cytometry. Cell Rep. 2018;22(7):1875–1888.
  • McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168(4):613–628.
  • Hammerlindl H, Schaider H. Tumor cell-intrinsic phenotypic plasticity facilitates adaptive cellular reprogramming driving acquired drug resistance. J Cell Commun Signal. 2018;12(1):133–141.
  • Emmons MF, Faiao-Flores F, Smalley KSM. The role of phenotypic plasticity in the escape of cancer cells from targeted therapy. Biochem Pharmacol. 2016;122:1–9.
  • Holohan C, Van Schaeybroeck S, Longley DB, et al. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13(10):714–726.
  • Sun C, Fang Y, Yin J, et al. Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers. Sci Transl Med. 2017;9(392).
  • Choi JI, Park SH, Lee HJ, et al. Inhibition of phospho-S6 kinase, a protein involved in the compensatory adaptive response, increases the efficacy of paclitaxel in reducing the viability of matrix-attached ovarian cancer cells. PLoS One. 2016;11(5):e0155052.
  • Fang P, Madden JA, Neums L, et al. Olaparib-induced adaptive response is disrupted by FOXM1 targeting that enhances sensitivity to PARP inhibition. Mol Cancer Res. 2018;16(6):961–973.
  • Fang Y, McGrail DJ, Sun C, et al. Sequential therapy with PARP and WEE1 inhibitors minimizes toxicity while maintaining efficacy. Cancer Cell. 2019;35(6):851–867 e7.
  • Hew KE, Miller PC, El-Ashry D, et al. MAPK activation predicts poor outcome and the MEK inhibitor, selumetinib, reverses antiestrogen resistance in ER-positive high-grade serous ovarian cancer. Clin Cancer Res. 2016;22(4):935–947.
  • Iavarone C, Zervantonakis IK, Selfors LM, et al. Combined MEK and BCL-2/XL inhibition is effective in high-grade serous ovarian cancer patient-derived xenograft models and BIM levels are predictive of responsiveness. Mol Cancer Ther. 2019;18(3):642–655.
  • Konstantinopoulos PA, Barry WT, Birrer M, et al. Olaparib and alpha-specific PI3K inhibitor alpelisib for patients with epithelial ovarian cancer: a dose-escalation and dose-expansion phase 1b trial. Lancet Oncol. 2019;20(4):570–580.
  • Liu JF, Tolaney SM, Birrer M, et al. A phase 1 trial of the poly(ADP-ribose) polymerase inhibitor olaparib (AZD2281) in combination with the anti-angiogenic cediranib (AZD2171) in recurrent epithelial ovarian or triple-negative breast cancer. Eur J Cancer. 2013;49(14):2972–2978.
  • Matulonis UA, Wulf GM, Barry WT, et al. Phase I dose escalation study of the PI3kinase pathway inhibitor BKM120 and the oral poly (ADP ribose) polymerase (PARP) inhibitor olaparib for the treatment of high-grade serous ovarian and breast cancer. Ann Oncol. 2017;28(3):512–518.
  • Sun C, Yin J, Fang Y, et al. BRD4 inhibition is synthetic lethal with PARP inhibitors through the induction of homologous recombination deficiency. Cancer Cell. 2018;33(3):401–416 e8.
  • Stewart C, Ralyea C, Lockwood S. Ovarian cancer: an integrated review. Semin Oncol Nurs. 2019;35(2):151–156.
  • Lee HH, Bellat V, Law B. Chemotherapy induces adaptive drug resistance and metastatic potentials via phenotypic CXCR4-expressing cell state transition in ovarian cancer. PLoS One. 2017;12(2):e0171044.
  • Shen L, Sun B, Sheng J, et al. PGC1alpha promotes cisplatin resistance in human ovarian carcinoma cells through upregulation of mitochondrial biogenesis. Int J Oncol. 2018;53(1):404–416.
  • Amoroso MR, Matassa DS, Agliarulo I, et al. Stress-adaptive response in ovarian cancer drug resistance: role of TRAP1 in oxidative metabolism-driven inflammation. Adv Protein Chem Struct Biol. 2017;108:163–198.
  • Reinthaller A. Antiangiogenic therapies in ovarian cancer. Memo. 2016;9(3):139–143.
  • Previs RA, Armaiz-Pena GN, Ivan C, et al. Role of YAP1 as a marker of sensitivity to dual AKT and P70S6K inhibition in ovarian and uterine malignancies. J Natl Cancer Inst. 2017;109(7). Online ID: djw296.
  • Yang X, Shen F, Hu W, et al. New ways to successfully target tumor vasculature in ovarian cancer. Curr Opin Obstet Gynecol. 2015;27(1):58–65.
  • Lyons YA, Pradeep S, Wu SY, et al. Macrophage depletion through colony stimulating factor 1 receptor pathway blockade overcomes adaptive resistance to anti-VEGF therapy. Oncotarget. 2017;8(57):96496–96505.
  • Kurimchak AM, Shelton C, Duncan KE, et al. Resistance to BET bromodomain inhibitors is mediated by kinome reprogramming in ovarian cancer. Cell Rep. 2016;16(5):1273–1286.
  • Ohmoto A, Yachida S. Current status of poly(ADP-ribose) polymerase inhibitors and future directions. Onco Targets Ther. 2017;10:5195–5208.
  • Lord CJ, Ashworth A. PARP inhibitors: synthetic lethality in the clinic. Science. 2017;355(6330):1152–1158.
  • Pilie PG, Tang C, Mills GB, et al. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol. 2019;16(2):81–104.
  • Vanderstichele A, Busschaert P, Olbrecht S, et al. Genomic signatures as predictive biomarkers of homologous recombination deficiency in ovarian cancer. Eur J Cancer. 2017;86:5–14.
  • Murai J, Huang SY, Das BB, et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 2012;72(21):5588–5599.
  • Shen Y, Aoyagi-Scharber M, Wang B. Trapping poly(ADP-ribose) polymerase. J Pharmacol Exp Ther. 2015;353(3):446–457.
  • Murai J, Huang SY, Renaud A, et al. Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Mol Cancer Ther. 2014;13(2):433–443.
  • Ibrahim YH, Garcia-Garcia C, Serra V, et al. PI3K inhibition impairs BRCA1/2 expression and sensitizes BRCA-proficient triple-negative breast cancer to PARP inhibition. Cancer Discov. 2012;2(11):1036–1047.
  • Vena F, Jia R, Esfandiari A, et al. MEK inhibition leads to BRCA2 downregulation and sensitization to DNA damaging agents in pancreas and ovarian cancer models. Oncotarget. 2018;9(14):11592–11603.
  • Chen M-J, Li J, Akbani R, et al. TCPA v6.0: an integrative platform for pan-cancer analysis of functional proteomic data. Mol Cell Proteomics. 2019. In press.
  • Chen MM, Li J, Wang Y, et al. TCPA v3.0: an integrative platform to explore the pan-cancer analysis of functional proteomic data. Mol Cell Proteomics. 2019;18:S15–S25.
  • Grote T, Siwak DR, Fritsche HA, et al. Validation of reverse phase protein array for practical screening of potential biomarkers in serum and plasma: accurate detection of CA19-9 levels in pancreatic cancer. Proteomics. 2008;8(15):3051–3060.
  • Lee J, Geiss GK, Demirkan G, et al. Implementation of a multiplex and quantitative proteomics platform for assessing protein lysates Using DNA-barcoded antibodies. Mol Cell Proteomics. 2018;17(6):1245–1258.
  • Li J, Lu Y, Akbani R, et al. TCPA: a resource for cancer functional proteomics data. Nat Methods. 2013;10(11):1046–1047.
  • Cohen AA, Geva-Zatorsky N, Eden E, et al. Dynamic proteomics of individual cancer cells in response to a drug. Science. 2008;322(5907):1511–1516.
  • Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420–1428.
  • Gascoigne KE, Taylor SS. Cancer cells display profound intra- and interline variation following prolonged exposure to antimitotic drugs. Cancer Cell. 2008;14(2):111–122.
  • Sharma SV, Lee DY, Li B, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell. 2010;141(1):69–80.
  • Lin JR, Fallahi-Sichani M, Chen JY, et al. Cyclic Immunofluorescence (CycIF), a highly multiplexed method for single-cell imaging. Curr Protoc Chem Biol. 2016;8(4):251–264.
  • Tsujikawa T, Kumar S, Borkar RN, et al. Quantitative multiplex immunohistochemistry reveals myeloid-inflamed tumor-immune complexity associated with poor prognosis. Cell Rep. 2017;19(1):203–217.
  • Keren L, Bosse M, Marquez D, et al. A structured tumor-immune microenvironment in triple negative breast cancer revealed by multiplexed ion beam imaging. Cell. 2018;174(6):1373–1387 e19.
  • Lin JR, Fallahi-Sichani M, Sorger PK. Highly multiplexed imaging of single cells using a high-throughput cyclic immunofluorescence method. Nat Commun. 2015;6:8390.
  • Eng JG, Luoh S, Gray J, et al. Cyclic multiplexed-immunofluorescence (cmIF), a highly multiplexed method for single-cell analysis, in biomarkers for immunotherapy of cancer. Methods Mol Biol. 2019;2055. [In press].
  • Faria SS, Morris CF, Silva AR, et al. A timely shift from shotgun to targeted proteomics and how it can be groundbreaking for cancer research. Front Oncol. 2017;7:13.
  • Zhang B, Whiteaker JR, Hoofnagle AN, et al. Clinical potential of mass spectrometry-based proteogenomics. Nat Rev Clin Oncol. 2019;16(4):256–268.
  • Chang CY, Picotti P, Huttenhain R, et al. Protein significance analysis in selected reaction monitoring (SRM) measurements. Mol Cell Proteomics. 2012;11(4):M111 014662.
  • Gillet LC, Navarro P, Tate S, et al. Targeted data extraction of the MS/MS spectra generated by data-independent acquisition: a new concept for consistent and accurate proteome analysis. Mol Cell Proteomics. 2012;11(6):O111 016717.
  • Ludwig C, Gillet L, Rosenberger G, et al. Data-independent acquisition-based SWATH-MS for quantitative proteomics: a tutorial. Mol Syst Biol. 2018;14(8):e8126.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.