Applications of Proteomics in Ovarian Cancer: Dawn of a New Era
Abstract
:1. Introduction
2. Signaling Pathways in OC
3. Proteomic Biomarkers for OC
3.1. Tissue Proteomics
3.2. Proteomics of Post-Translational Modifications
3.3. Quantitative Proteomics
3.4. Biofluid Proteomics
4. Proteomic Techniques
4.1. Two-Dimensional Gel Electrophoresis (2DE)
4.2. MS-Based Techniques
4.2.1. MALDI-TOF
4.2.2. SELDI-TOF
4.3. Protein Microarrays
4.4. Mitochondrial Proteomics Methods
5. New Approaches in Proteomics
5.1. Targeted Proteomics
5.2. Peptidomics
5.3. Exosomes
6. Proteomics in the Treatment of Ovarian Cancer
6.1. Tackling Chemotherapy Resistance
6.2. Targeted Therapy Using Proteomics
7. PARP Inhibitors
8. VEGF/VEGF Receptor Inhibitors
9. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Acknowledgments
Conflicts of Interest
References
- Ovarian Cancer Survival Statistics. Available online: https://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/ovarian-cancer (accessed on 26 February 2022).
- Boussios, S.; Mikropoulos, C.; Samartzis, E.; Karihtala, P.; Moschetta, M.; Sheriff, M.; Karathanasi, A.; Sadauskaite, A.; Rassy, E.; Pavlidis, N. Wise Management of Ovarian Cancer: On the Cutting Edge. J. Pers. Med. 2020, 10, 41. [Google Scholar] [CrossRef]
- Boussios, S.; Zarkavelis, G.; Seraj, E.; Zerdes, I.; Tatsi, K.; Pentheroudakis, G. Non-epithelial Ovarian Cancer: Elucidating Uncommon Gynaecological Malignancies. AntiCancer Res. 2016, 36, 5031–5042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boussios, S.; Moschetta, M.; Zarkavelis, G.; Papadaki, A.; Kefas, A.; Tatsi, K. Ovarian sex-cord stromal tumours and small cell tumours: Pathological, genetic and management aspects. Crit. Rev. Oncol. Hematol. 2017, 120, 43–51. [Google Scholar] [CrossRef] [PubMed]
- Boussios, S.; Karathanasi, A.; Zakynthinakis-Kyriakou, N.; Tsiouris, A.K.; Chatziantoniou, A.A.; Kanellos, F.S.; Tatsi, K. Ovarian carcinosarcoma: Current developments and future perspectives. Crit. Rev. Oncol. Hematol. 2019, 134, 46–55. [Google Scholar] [CrossRef] [PubMed]
- Pavlidis, N.; Rassy, E.; Vermorken, J.B.; Assi, T.; Kattan, J.; Boussios, S.; Smith-Gagen, J. The outcome of patients with serous papillary peritoneal cancer, fallopian tube cancer, and epithelial ovarian cancer by treatment eras: 27 years data from the SEER registry. Cancer Epidemiol. 2021, 75, 102045. [Google Scholar] [CrossRef] [PubMed]
- Menon, U.; Gentry-Maharaj, A.; Burnell, M.; Singh, N.; Ryan, A.; Karpinskyj, C.; Carlino, G.; Taylor, J.; Massingham, S.K.; Raikou, M.; et al. Ovarian cancer population screening and mortality after long-term follow-up in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): A randomised controlled trial. Lancet 2021, 397, 2182–2193. [Google Scholar] [CrossRef]
- Buys, S.S.; Partridge, E.; Greene, M.H.; Prorok, P.C.; Reding, D.; Riley, T.L.; Hartge, P.; Fagerstrom, R.; Ragard, L.R.; Chia, D.; et al. Ovarian cancer screening in the Prostate, Lung, Colorectal and Ovarian (PLCO) cancer screening trial: Findings from the initial screen of a randomized trial. Am. J. Obstet. Gynecol. 2005, 193, 1630–1639. [Google Scholar] [CrossRef]
- Lu, K.H.; Skates, S.; Hernandez, M.A.; Bedi, D.; Bevers, T.; Leeds, L.; Moore, R.; Granai, C.; Harris, S.; Newland, W.; et al. A 2-stage ovarian cancer screening strategy using the Risk of Ovarian Cancer Algorithm (ROCA) identifies early-stage incident cancers and demonstrates high positive predictive value. Cancer 2013, 119, 3454–3461. [Google Scholar] [CrossRef]
- Jacobs, I.J.; Menon, U.; Ryan, A.; Gentry-Maharaj, A.; Burnell, M.; Kalsi, J.K.; Amso, N.N.; Apostolidou, S.; Benjamin, E.; Cruickshank, D.; et al. Ovarian cancer screening and mortality in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): A randomised controlled trial. Lancet 2016, 387, 945–956. [Google Scholar] [CrossRef] [Green Version]
- Boussios, S.; Karathanasi, A.; Cooke, D.; Neille, C.; Sadauskaite, A.; Moschetta, M.; Zakynthinakis-Kyriakou, N.; Pavlidis, N. PARP Inhibitors in Ovarian Cancer: The Route to “Ithaca”. Diagnostics 2019, 9, 55. [Google Scholar] [CrossRef] [Green Version]
- Boussios, S.; Abson, C.; Moschetta, M.; Rassy, E.; Karathanasi, A.; Bhat, T.; Ghumman, F.; Sheriff, M.; Pavlidis, N. Poly (ADP-Ribose) Polymerase Inhibitors: Talazoparib in Ovarian Cancer and Beyond. Drugs R&D 2020, 20, 55–73. [Google Scholar]
- Drescher, C.W.; Hawley, S.; Thorpe, J.D.; Marticke, S.; McIntosh, M.; Gambhir, S.S.; Urban, N. Impact of screening test performance and cost on mortality reduction and cost-effectiveness of multimodal ovarian cancer screening. Cancer Prev. Res. 2012, 5, 1015–1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, Z.; Wei, C.; Luo, Z.; Li, L. Clinical value of serum human epididymis protein 4 assay in the diagnosis of ovarian cancer: A meta-analysis. Onco. Targets. Ther. 2013, 6, 957–966. [Google Scholar] [PubMed] [Green Version]
- Shulman, L.P.; Francis, M.; Bullock, R.; Pappas, T. Clinical Performance Comparison of Two In-Vitro Diagnostic Multivariate Index Assays (IVDMIAs) for Presurgical Assessment for Ovarian Cancer Risk. Adv. Ther. 2019, 36, 2402–2413. [Google Scholar] [CrossRef] [Green Version]
- Goossens, N.; Nakagawa, S.; Sun, X.; Hoshida, Y. Cancer biomarker discovery and validation. Transl. Cancer Res. 2015, 4, 256–269. [Google Scholar]
- Bonifácio, V.D.B. Ovarian Cancer Biomarkers: Moving Forward in Early Detection. Adv. Exp. Med. Biol. 2020, 1219, 355–363. [Google Scholar]
- Feng, Y.; Xiao, M.; Zhang, Z.; Cui, R.; Jiang, X.; Wang, S.; Bai, H.; Liu, C.; Zhang, Z. Potential interaction between lysophosphatidic acid and tumor-associated macrophages in ovarian carcinoma. J. Inflamm. 2020, 17, 23. [Google Scholar] [CrossRef]
- Onallah, H.; Davidson, B.; Reich, R. Diverse Effects of Lysophosphatidic Acid Receptors on Ovarian Cancer Signaling Pathways. J. Oncol. 2019, 2019, 7547469. [Google Scholar] [CrossRef]
- Ghoneum, A.; Said, N. PI3K-AKT-mTOR and NFκB Pathways in Ovarian Cancer: Implications for Targeted Therapeutics. Cancers 2019, 11, 949. [Google Scholar] [CrossRef] [Green Version]
- Rinne, N.; Christie, E.L.; Ardasheva, A.; Kwok, C.H.; Demchenko, N.; Low, C.; Tralau-Stewart, C.; Fotopoulou, C.; Cunnea, P. Targeting the PI3K/AKT/mTOR pathway in epithelial ovarian cancer, therapeutic treatment options for platinum-resistant ovarian cancer. Cancer Drug. Resist. 2021, 4, 573–595. [Google Scholar] [CrossRef]
- Harrington, B.; Annunziata, C.M. NF-κB Signaling in Ovarian Cancer. Cancers 2019, 11, 1182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- House, C.D.; Jordan, E.; Hernandez, L.; Ozaki, M.; James, J.M.; Kim, M.; Kruhlak, M.J.; Batchelor, E.; Elloumi, F.; Cam, M.C.; et al. NFκB Promotes Ovarian Tumorigenesis via Classical Pathways That Support Proliferative Cancer Cells and Alternative Pathways That Support ALDH+ Cancer Stem-like Cells. Cancer Res. 2017, 77, 6927–6940. [Google Scholar] [CrossRef] [Green Version]
- Choi, K.C.; Auersperg, N.; Leung, P.C. Mitogen-activated protein kinases in normal and (pre)neoplastic ovarian surface epithelium. Reprod. Biol. Endocrinol. 2003, 1, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hew, K.E.; Miller, P.C.; El-Ashry, D.; Sun, J.; Besser, A.H.; Ince, T.A.; Gu, M.; Wei, Z.; Zhang, G.; Brafford, P.; 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, 935–947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wiener, J.R.; Windham, T.C.; Estrella, V.C.; Parikh, N.U.; Thall, P.F.; Deavers, M.T.; Bast, R.C.; Mills, G.B.; Gallick, G.E. Activated SRC protein tyrosine kinase is overexpressed in late-stage human ovarian cancers. Gynecol. Oncol. 2003, 88, 73–79. [Google Scholar] [CrossRef] [Green Version]
- Wang, Q.; Peng, H.; Qi, X.; Wu, M.; Zhao, X. Targeted therapies in gynecological cancers: A comprehensive review of clinical evidence. Signal. Transduct. Target. Ther. 2020, 5, 137. [Google Scholar] [CrossRef]
- Siwak, D.R.; Carey, M.; Hennessy, B.T.; Nguyen, C.T.; McGahren Murray, M.J.; Nolden, L.; Mills, G.B. Targeting the epidermal growth factor receptor in epithelial ovarian cancer: Current knowledge and future challenges. J. Oncol. 2010, 2010, 568938. [Google Scholar] [CrossRef] [Green Version]
- de Graeff, P.; Crijns, A.P.; Ten Hoor, K.A.; Klip, H.G.; Hollema, H.; Oien, K.; Bartlett, J.M.; Wisman, G.B.; de Bock, G.H.; de Vries, E.G.; et al. The ErbB signalling pathway: Protein expression and prognostic value in epithelial ovarian cancer. Br. J. Cancer 2008, 99, 341–349. [Google Scholar] [CrossRef] [Green Version]
- Mukherjee, S.; Pal, M.; Mukhopadhyay, S.; Das, I.; Hazra, R.; Ghosh, S.; Mondal, R.K.; Bal, R. VEGF Expression to Support Targeted Therapy in Ovarian Surface Epithelial Neoplasms. J. Clin. Diagn. Res. 2017, 11, 43–46. [Google Scholar] [CrossRef]
- Masoumi Moghaddam, S.; Amini, A.; Morris, D.L.; Pourgholami, M.H. Significance of vascular endothelial growth factor in growth and peritoneal dissemination of ovarian cancer. Cancer Metastasis Rev. 2012, 31, 143–162. [Google Scholar] [CrossRef] [Green Version]
- Liang, R.; Chen, X.; Chen, L.; Wan, F.; Chen, K.; Sun, Y.; Zhu, X. STAT3 signaling in ovarian cancer: A potential therapeutic target. J. Cancer 2020, 11, 837–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gritsina, G.; Xiao, F.; O’Brien, S.W.; Gabbasov, R.; Maglaty, M.A.; Xu, R.H.; Thapa, R.J.; Zhou, Y.; Nicolas, E.; Litwin, S.; et al. Targeted Blockade of JAK/STAT3 Signaling Inhibits Ovarian Carcinoma Growth. Mol. Cancer Ther. 2015, 14, 1035–1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Browning, L.; Patel, M.R.; Horvath, E.B.; Tawara, K.; Jorcyk, C.L. IL-6 and ovarian cancer: Inflammatory cytokines in promotion of metastasis. Cancer Manag. Res. 2018, 10, 6685–6693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, J.; Ward, A.C. Role of the interleukin 6 receptor family in epithelial ovarian cancer and its clinical implications. Biochim. Biophys. Acta 2014, 1845, 117–125. [Google Scholar] [CrossRef] [PubMed]
- Espina, V.; Wulfkuhle, J.D.; Calvert, V.S.; VanMeter, A.; Zhou, W.; Coukos, G.; Geho, D.H.; Petricoin, E.F., 3rd; Liotta, L.A. Laser-capture microdissection. Nat. Protoc. 2006, 1, 586–603. [Google Scholar] [CrossRef] [PubMed]
- Ariztia, E.V.; Lee, C.J.; Gogoi, R.; Fishman, D.A. The tumor microenvironment: Key to early detection. Crit. Rev. Clin. Lab. Sci. 2006, 43, 393–425. [Google Scholar] [CrossRef]
- Meani, F.; Pecorelli, S.; Liotta, L.; Petricoin, E.F. Clinical application of proteomics in ovarian cancer prevention and treatment. Mol. Diagn. Ther. 2009, 13, 297–311. [Google Scholar] [CrossRef]
- Köbel, M.; Kalloger, S.E.; Boyd, N.; McKinney, S.; Mehl, E.; Palmer, C.; Leung, S.; Bowen, N.J.; Ionescu, D.N.; Rajput, A.; et al. Ovarian carcinoma subtypes are different diseases: Implications for biomarker studies. PLoS Med. 2008, 5, e232. [Google Scholar] [CrossRef]
- Boussios, S.; Karihtala, P.; Moschetta, M.; Abson, C.; Karathanasi, A.; Zakynthinakis-Kyriakou, N.; Ryan, J.E.; Sheriff, M.; Rassy, E.; Pavlidis, N. Veliparib in ovarian cancer: A new synthetically lethal therapeutic approach. Investig. New Drugs 2020, 38, 181–193. [Google Scholar] [CrossRef]
- Lheureux, S.; Braunstein, M.; Oza, A.M. Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA Cancer J. Clin. 2019, 69, 280–304. [Google Scholar] [CrossRef] [Green Version]
- Wulfkuhle, J.D.; Speer, R.; Pierobon, M.; Laird, J.; Espina, V.; Deng, J.; Mammano, E.; Yang, S.X.; Swain, S.M.; Nitti, D.; et al. Multiplexed cell signaling analysis of human breast cancer applications for personalized therapy. J. Proteome Res. 2008, 7, 1508–1517. [Google Scholar] [CrossRef] [PubMed]
- Toss, A.; De Matteis, E.; Rossi, E.; Casa, L.D.; Iannone, A.; Federico, M.; Cortesi, L. Ovarian cancer: Can proteomics give new insights for therapy and diagnosis? Int. J. Mol. Sci. 2013, 14, 8271–8290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Swiatly, A.; Plewa, S.; Matysiak, J.; Kokot, Z.J. Mass spectrometry-based proteomics techniques and their application in ovarian cancer research. J. Ovarian Res. 2018, 11, 88. [Google Scholar] [CrossRef] [PubMed]
- Mechref, Y.; Hu, Y.; Garcia, A.; Zhou, S.; Desantos-Garcia, J.L.; Hussein, A. Defining putative glycan cancer biomarkers by MS. Bioanalysis 2012, 4, 2457–2469. [Google Scholar] [CrossRef] [Green Version]
- Elzek, M.A.; Rodland, K.D. Proteomics of ovarian cancer: Functional insights and clinical applications. Cancer Metastasis Rev. 2015, 34, 83–96. [Google Scholar] [CrossRef] [Green Version]
- Blom, N.; Sicheritz-Pontén, T.; Gupta, R.; Gammeltoft, S.; Brunak, S. Prediction of post-translational glycosylation and phosphorylation of proteins from the amino acid sequence. Proteomics 2004, 4, 1633–1649. [Google Scholar] [CrossRef]
- Tousi, F.; Hancock, W.S.; Hincapie, M. Technologies and strategies for glycoproteomics and glycomics and their application to clinical biomarker research. Anal. Methods 2011, 3, 20–32. [Google Scholar] [CrossRef]
- Abbott, K.L.; Lim, J.M.; Wells, L.; Benigno, B.B.; McDonald, J.F.; Pierce, M. Identification of candidate biomarkers with cancer-specific glycosylation in the tissue and serum of endometrioid ovarian cancer patients by glycoproteomic analysis. Proteomics 2010, 10, 470–481. [Google Scholar] [CrossRef] [Green Version]
- Shetty, V.; Hafner, J.; Shah, P.; Nickens, Z.; Philip, R. Investigation of ovarian cancer associated sialylation changes in N-linked glycopeptides by quantitative proteomics. Clin. Proteom. 2012, 9, 10. [Google Scholar] [CrossRef] [Green Version]
- Kuzmanov, U.; Musrap, N.; Kosanam, H.; Smith, C.R.; Batruch, I.; Dimitromanolakis, A.; Diamandis, E.P. Glycoproteomic identification of potential glycoprotein biomarkers in ovarian cancer proximal fluids. Clin. Chem. Lab. Med. 2013, 51, 1467–1476. [Google Scholar] [CrossRef]
- Saldova, R.; Struwe, W.B.; Wynne, K.; Elia, G.; Duffy, M.J.; Rudd, P.M. Exploring the glycosylation of serum CA125. Int. J. Mol. Sci. 2013, 14, 15636–15654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liau, B.; Tan, B.; Teo, G.; Zhang, P.; Choo, A.; Rudd, P.M. Shotgun Glycomics Identifies Tumor-Associated Glycan Ligands Bound by an Ovarian Carcinoma-Specific Monoclonal Antibody. Sci. Rep. 2017, 7, 14489. [Google Scholar] [CrossRef] [PubMed]
- Maes, E.; Tirez, K.; Baggerman, G.; Valkenborg, D.; Schoofs, L.; Encinar, J.R.; Mertens, I. The use of elemental mass spectrometry in phosphoproteomic applications. Mass. Spectrom. Rev. 2016, 35, 350–360. [Google Scholar] [CrossRef] [PubMed]
- Francavilla, C.; Lupia, M.; Tsafou, K.; Villa, A.; Kowalczyk, K.; Rakownikow Jersie-Christensen, R.; Bertalot, G.; Confalonieri, S.; Brunak, S.; Jensen, L.J.; et al. Phosphoproteomics of Primary Cells Reveals Druggable Kinase Signatures in Ovarian Cancer. Cell Rep. 2017, 18, 3242–3256. [Google Scholar] [CrossRef] [Green Version]
- Longuespée, R.; Boyon, C.; Desmons, A.; Vinatier, D.; Leblanc, E.; Farré, I.; Wisztorski, M.; Ly, K.; D’Anjou, F.; Day, R.; et al. Ovarian cancer molecular pathology. Cancer Metastasis Rev. 2012, 31, 713–732. [Google Scholar] [CrossRef]
- Ryu, J.; Thomas, S.N. Quantitative Mass Spectrometry-Based Proteomics for Biomarker Development in Ovarian Cancer. Molecules 2021, 26, 2674. [Google Scholar] [CrossRef]
- Li, N.; Zhan, X. Mass Spectrometry-based Mitochondrial Proteomics in Human Ovarian Cancers. Mass. Spectrom. Rev. 2020, 39, 471–498. [Google Scholar] [CrossRef]
- Li, K.W.; Gonzalez-Lozano, M.A.; Koopmans, F.; Smit, A.B. Recent Developments in Data Independent Acquisition (DIA) Mass Spectrometry: Application of Quantitative Analysis of the Brain Proteome. Front. Mol. Neurosci. 2020, 13, 564446. [Google Scholar] [CrossRef]
- Jurisicova, A.; Jurisica, I.; Kislinger, T. Advances in ovarian cancer proteomics: The quest for biomarkers and improved therapeutic interventions. Expert Rev. Proteom. 2008, 5, 551–560. [Google Scholar] [CrossRef]
- Ueland, F.R. A Perspective on Ovarian Cancer Biomarkers: Past, Present and Yet-To-Come. Diagnostics 2017, 7, 14. [Google Scholar] [CrossRef] [Green Version]
- Bast, R.C., Jr.; Feeney, M.; Lazarus, H.; Nadler, L.M.; Colvin, R.B.; Knapp, R.C. Reactivity of a monoclonal antibody with human ovarian carcinoma. J. Clin. Investig. 1981, 68, 1331–1337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bast, R.C., Jr.; Lu, Z.; Han, C.Y.; Lu, K.H.; Anderson, K.S.; Drescher, C.W.; Skates, S.J. Biomarkers and Strategies for Early Detection of Ovarian Cancer. Cancer Epidemiol. Biomark. Prev. 2020, 29, 2504–2512. [Google Scholar] [CrossRef] [PubMed]
- Schummer, M.; Ng, W.V.; Bumgarner, R.E.; Nelson, P.S.; Schummer, B.; Bednarski, D.W.; Hassell, L.; Baldwin, R.L.; Karlan, B.Y.; Hood, L. Comparative hybridization of an array of 21,500 ovarian cDNAs for the discovery of genes overexpressed in ovarian carcinomas. Gene 1999, 238, 375–385. [Google Scholar] [CrossRef]
- Hellström, I.; Raycraft, J.; Hayden-Ledbetter, M.; Ledbetter, J.A.; Schummer, M.; McIntosh, M.; Drescher, C.; Urban, N.; Hellström, K.E. The HE4 (WFDC2) protein is a biomarker for ovarian carcinoma. Cancer Res. 2003, 63, 3695–3700. [Google Scholar] [PubMed]
- Moore, R.G.; McMeekin, D.S.; Brown, A.K.; DiSilvestro, P.; Miller, M.C.; Allard, W.J.; Gajewski, W.; Kurman, R.; Bast, R.C., Jr.; Skates, S.J. A novel multiple marker bioassay utilizing HE4 and CA125 for the prediction of ovarian cancer in patients with a pelvic mass. Gynecol. Oncol. 2009, 112, 40–46. [Google Scholar] [CrossRef] [Green Version]
- Cohen, J.D.; Li, L.; Wang, Y.; Thoburn, C.; Afsari, B.; Danilova, L.; Douville, C.; Javed, A.A.; Wong, F.; Mattox, A.; et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018, 359, 926–930. [Google Scholar] [CrossRef] [Green Version]
- Van Gorp, T.; Cadron, I.; Despierre, E.; Daemen, A.; Leunen, K.; Amant, F.; Timmerman, D.; De Moor, B.; Vergote, I. HE4 and CA125 as a diagnostic test in ovarian cancer: Prospective validation of the Risk of Ovarian Malignancy Algorithm. Br. J. Cancer 2011, 104, 863–870. [Google Scholar] [CrossRef] [Green Version]
- Ueland, F.R.; Desimone, C.P.; Seamon, L.G.; Miller, R.A.; Goodrich, S.; Podzielinski, I.; Sokoll, L.; Smith, A.; van Nagell, J.R., Jr.; Zhang, Z. Effectiveness of a multivariate index assay in the preoperative assessment of ovarian tumors. Obstet. Gynecol. 2011, 117, 1289–1297. [Google Scholar] [CrossRef]
- Hurley, L.C.; Levin, N.K.; Chatterjee, M.; Coles, J.; Muszkat, S.; Howarth, Z.; Dyson, G.; Tainsky, M.A. Evaluation of paraneoplastic antigens reveals TRIM21 autoantibodies as biomarker for early detection of ovarian cancer in combination with autoantibodies to NY-ESO-1 and TP53. Cancer Biomark. 2020, 27, 407–421. [Google Scholar] [CrossRef]
- Yang, W.L.; Lu, Z.; Guo, J.; Fellman, B.M.; Ning, J.; Lu, K.H.; Menon, U.; Kobayashi, M.; Hanash, S.M.; Celestino, J.; et al. Human epididymis protein 4 antigen-autoantibody complexes complement cancer antigen 125 for detecting early-stage ovarian cancer. Cancer 2020, 126, 725–736. [Google Scholar] [CrossRef]
- Nakamura, K.; Sawada, K.; Yoshimura, A.; Kinose, Y.; Nakatsuka, E.; Kimura, T. Clinical relevance of circulating cell-free microRNAs in ovarian cancer. Mol. Cancer 2016, 15, 48. [Google Scholar] [CrossRef] [Green Version]
- Elias, K.M.; Fendler, W.; Stawiski, K.; Fiascone, S.J.; Vitonis, A.F.; Berkowitz, R.S.; Frendl, G.; Konstantinopoulos, P.; Crum, C.P.; Kedzierska, M.; et al. Diagnostic potential for a serum miRNA neural network for detection of ovarian cancer. Elife 2017, 6, e28932. [Google Scholar] [CrossRef] [PubMed]
- Dong, Y.; Kaushal, A.; Bui, L.; Chu, S.; Fuller, P.J.; Nicklin, J.; Samaratunga, H.; Clements, J.A. Human kallikrein 4 (KLK4) is highly expressed in serous ovarian carcinomas. Clin. Cancer Res. 2001, 7, 2363–2371. [Google Scholar] [PubMed]
- Zhang, Y.; Guo, B.; Bi, R. Ovarian cancer: Biomarker proteomic diagnosis in progress. Appl. Biochem. Biotechnol. 2012, 168, 910–916. [Google Scholar] [CrossRef] [PubMed]
- Guo, J.; Yang, W.L.; Pak, D.; Celestino, J.; Lu, K.H.; Ning, J.; Lokshin, A.E.; Cheng, Z.; Lu, Z.; Bast, R.C., Jr. Osteopontin, Macrophage Migration Inhibitory Factor and Anti-Interleukin-8 Autoantibodies Complement CA125 for Detection of Early Stage Ovarian Cancer. Cancers 2019, 11, 596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Z.; Bast, R.C., Jr.; Yu, Y.; Li, J.; Sokoll, L.J.; Rai, A.J.; Rosenzweig, J.M.; Cameron, B.; Wang, Y.Y.; Meng, X.Y.; et al. Three biomarkers identified from serum proteomic analysis for the detection of early stage ovarian cancer. Cancer Res. 2004, 64, 5882–5890. [Google Scholar] [CrossRef] [Green Version]
- Edgell, T.; Martin-Roussety, G.; Barker, G.; Autelitano, D.J.; Allen, D.; Grant, P.; Rice, G.E. Phase II biomarker trial of a multimarker diagnostic for ovarian cancer. J. Cancer Res. Clin. Oncol. 2010, 136, 1079–1088. [Google Scholar] [CrossRef] [Green Version]
- Buas, M.F.; Gu, H.; Djukovic, D.; Zhu, J.; Drescher, C.W.; Urban, N.; Raftery, D.; Li, C.I. Identification of novel candidate plasma metabolite biomarkers for distinguishing serous ovarian carcinoma and benign serous ovarian tumors. Gynecol. Oncol. 2016, 140, 138–144. [Google Scholar] [CrossRef] [Green Version]
- Terry, K.L.; Schock, H.; Fortner, R.T.; Hüsing, A.; Fichorova, R.N.; Yamamoto, H.S.; Vitonis, A.F.; Johnson, T.; Overvad, K.; Tjønneland, A.; et al. A Prospective Evaluation of Early Detection Biomarkers for Ovarian Cancer in the European EPIC Cohort. Clin. Cancer Res. 2016, 22, 4664–4675. [Google Scholar] [CrossRef] [Green Version]
- Pisanic, T.R., 2nd; Cope, L.M.; Lin, S.F.; Yen, T.T.; Athamanolap, P.; Asaka, R.; Nakayama, K.; Fader, A.N.; Wang, T.H.; Shih, I.M.; et al. Methylomic Analysis of Ovarian Cancers Identifies Tumor-Specific Alterations Readily Detectable in Early Precursor Lesions. Clin. Cancer Res. 2018, 24, 6536–6547. [Google Scholar] [CrossRef] [Green Version]
- Mukama, T.; Fortner, R.T.; Katzke, V.; Hynes, L.C.; Petrera, A.; Hauck, S.M.; Johnson, T.; Schulze, M.; Schiborn, C.; Rostgaard-Hansen, A.L.; et al. Prospective evaluation of 92 serum protein biomarkers for early detection of ovarian cancer. Br. J. Cancer 2022, 126, 1301–1309. [Google Scholar] [CrossRef] [PubMed]
- Ke, C.; Hou, Y.; Zhang, H.; Fan, L.; Ge, T.; Guo, B.; Zhang, F.; Yang, K.; Wang, J.; Lou, G.; et al. Large-scale profiling of metabolic dysregulation in ovarian cancer. Int. J. Cancer 2015, 136, 516–526. [Google Scholar] [CrossRef] [PubMed]
- Todeschini, P.; Salviato, E.; Paracchini, L.; Ferracin, M.; Petrillo, M.; Zanotti, L.; Tognon, G.; Gambino, A.; Calura, E.; Caratti, G.; et al. Circulating miRNA landscape identifies miR-1246 as promising diagnostic biomarker in high-grade serous ovarian carcinoma: A validation across two independent cohorts. Cancer Lett. 2017, 388, 320–327. [Google Scholar] [CrossRef] [PubMed]
- Yokoi, A.; Matsuzaki, J.; Yamamoto, Y.; Yoneoka, Y.; Takahashi, K.; Shimizu, H.; Uehara, T.; Ishikawa, M.; Ikeda, S.I.; Sonoda, T.; et al. Integrated extracellular microRNA profiling for ovarian cancer screening. Nat. Commun. 2018, 9, 4319. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Fu, Z.; Dai, C.; Cao, J.; Liu, X.; Xu, J.; Lv, M.; Gu, Y.; Zhang, J.; Hua, X.; et al. LncRNAs expression profiling in normal ovary, benign ovarian cyst and malignant epithelial ovarian cancer. Sci. Rep. 2016, 6, 38983. [Google Scholar] [CrossRef] [Green Version]
- Huang, L.; Wang, N.; Mitchell, C.E.; Brownlee, T.; Maple, S.R.; De Felippis, M.R. A Novel Sample Preparation for Shotgun Proteomics Characterization of HCPs in Antibodies. Anal. Chem. 2017, 89, 5436–5444. [Google Scholar] [CrossRef]
- Lee, J.Y.; Kim, H.S.; Suh, D.H.; Kim, M.K.; Chung, H.H.; Song, Y.S. Ovarian cancer biomarker discovery based on genomic approaches. J. Cancer Prev. 2013, 18, 298–312. [Google Scholar] [CrossRef] [Green Version]
- Meinhold-Heerlein, I.; Bauerschlag, D.; Zhou, Y.; Sapinoso, L.M.; Ching, K.; Frierson, H., Jr.; Bräutigam, K.; Sehouli, J.; Stickeler, E.; Könsgen, D.; et al. An integrated clinical-genomics approach identifies a candidate multi-analyte blood test for serous ovarian carcinoma. Clin. Cancer Res. 2007, 13, 458–466. [Google Scholar] [CrossRef] [Green Version]
- Huang, J.; Hu, W.; Sood, A.K. Prognostic biomarkers in ovarian cancer. Cancer Biomark. 2010, 8, 231–251. [Google Scholar] [CrossRef] [Green Version]
- Koehn, H.; Oehler, M.K. Proteins’ promise–progress and challenges in ovarian cancer proteomics. Menopause Int. 2007, 13, 148–153. [Google Scholar] [CrossRef]
- Ardekani, A.M.; Liotta, L.A.; Petricoin, E.F., 3rd. Clinical potential of proteomics in the diagnosis of ovarian cancer. Expert Rev. Mol. Diagn. 2002, 2, 312–320. [Google Scholar] [CrossRef] [PubMed]
- Boyce, E.A.; Kohn, E.C. Ovarian cancer in the proteomics era: Diagnosis, prognosis, and therapeutics targets. Int. J. Gynecol. Cancer 2005, 15, 266–273. [Google Scholar] [CrossRef] [PubMed]
- Tchabo, N.E.; Liel, M.S.; Kohn, E.C. Applying proteomics in clinical trials: Assessing the potential and practical limitations in ovarian cancer. Am. J. Pharm. 2005, 5, 141–148. [Google Scholar] [CrossRef] [PubMed]
- Conrads, T.P.; Fusaro, V.A.; Ross, S.; Johann, D.; Rajapakse, V.; Hitt, B.A.; Steinberg, S.M.; Kohn, E.C.; Fishman, D.A.; Whitely, G.; et al. High-resolution serum proteomic features for ovarian cancer detection. Endocr. Relat. Cancer 2004, 11, 163–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gong, N.; Ma, X.; Ye, X.; Zhou, Q.; Chen, X.; Tan, X.; Yao, S.; Huo, S.; Zhang, T.; Chen, S.; et al. Carbon-dot-supported atomically dispersed gold as a mitochondrial oxidative stress amplifier for cancer treatment. Nat. Nanotechnol. 2019, 14, 379–387. [Google Scholar] [CrossRef]
- Rabilloud, T.; Kieffer, S.; Procaccio, V.; Louwagie, M.; Courchesne, P.L.; Patterson, S.D.; Martinez, P.; Garin, J.; Lunardi, J. Two-dimensional electrophoresis of human placental mitochondria and protein identification by mass spectrometry: Toward a human mitochondrial proteome. Electrophoresis 1998, 19, 1006–1014. [Google Scholar] [CrossRef]
- Beadnell, T.C.; Scheid, A.D.; Vivian, C.J.; Welch, D.R. Roles of the mitochondrial genetics in cancer metastasis: Not to be ignored any longer. Cancer Metastasis Rev. 2018, 37, 615–632. [Google Scholar] [CrossRef]
- Hecker, D. Enzyme histochemical and electron microscopic studies on the problem of infiltrating (invasive) tumor growth. 2. Electron microscopic studies. Gegenbaurs. Morphol. Jahrb. 1977, 123, 51–64. [Google Scholar]
- Dier, U.; Shin, D.H.; Hemachandra, L.P.; Uusitalo, L.M.; Hempel, N. Bioenergetic analysis of ovarian cancer cell lines: Profiling of histological subtypes and identification of a mitochondria-defective cell line. PLoS ONE 2014, 9, e98479. [Google Scholar] [CrossRef]
- Hou, L.; Wang, R.; Wei, H.; Li, S.; Liu, L.; Lu, X.; Yu, H.; Liu, Z. ABT737 enhances ovarian cancer cells sensitivity to cisplatin through regulation of mitochondrial fission via Sirt3 activation. Life Sci. 2019, 232, 116561. [Google Scholar] [CrossRef]
- Lu, P.; Vander Mause, E.R.; Redd Bowman, K.E.; Brown, S.M.; Ahne, L.; Lim, C.S. Mitochondrially targeted p53 or DBD subdomain is superior to wild type p53 in ovarian cancer cells even with strong dominant negative mutant p53. J. Ovarian. Res. 2019, 12, 45. [Google Scholar] [CrossRef] [PubMed]
- Wang, D.; You, D.; Li, L. Galectin-3 regulates chemotherapy sensitivity in epithelial ovarian carcinoma via regulating mitochondrial function. J. Toxicol. Sci. 2019, 44, 47–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schirmer, E.C.; Yates, J.R., 3rd; Gerace, L. MudPIT: A powerful proteomics tool for discovery. Discov. Med. 2003, 3, 38–39. [Google Scholar] [PubMed]
- Chappell, N.P.; Teng, P.N.; Hood, B.; Wang, G.; Darcy, K.M.; Hamilton, C.A.; Maxwell, G.L.; Conrads, T.P. Mitochondrial proteomic analysis of cisplatin resistance in ovarian cancer. J. Proteome Res. 2012, 11, 4605–4614. [Google Scholar] [CrossRef]
- Li, N.; Li, H.; Cao, L.; Zhan, X. Quantitative analysis of the mitochondrial proteome in human ovarian carcinomas. Endocr. Relat. Cancer 2018, 25, 909–931. [Google Scholar] [CrossRef] [Green Version]
- Drake, J.M.; Paull, E.O.; Graham, N.A.; Lee, J.K.; Smith, B.A.; Titz, B.; Stoyanova, T.; Faltermeier, C.M.; Uzunangelov, V.; Carlin, D.E.; et al. Phosphoproteome Integration Reveals Patient-Specific Networks in Prostate Cancer. Cell 2016, 166, 1041–1054. [Google Scholar] [CrossRef] [Green Version]
- Thomas, S.N.; Zhang, H. Targeted proteomic assays for the verification of global proteomics insights. Expert Rev. Proteom. 2016, 13, 897–899. [Google Scholar] [CrossRef] [Green Version]
- Shi, T.; Su, D.; Liu, T.; Tang, K.; Camp, D.G., 2nd; Qian, W.J.; Smith, R.D. Advancing the sensitivity of selected reaction monitoring-based targeted quantitative proteomics. Proteomics 2012, 12, 1074–1092. [Google Scholar] [CrossRef] [Green Version]
- Gillet, L.C.; Leitner, A.; Aebersold, R. Mass Spectrometry Applied to Bottom-Up Proteomics: Entering the High-Throughput Era for Hypothesis Testing. Annu. Rev. Anal. Chem. 2016, 9, 449–472. [Google Scholar] [CrossRef]
- Peterson, A.C.; Russell, J.D.; Bailey, D.J.; Westphall, M.S.; Coon, J.J. Parallel reaction monitoring for high resolution and high mass accuracy quantitative, targeted proteomics. Mol. Cell. Proteom. 2012, 11, 475–488. [Google Scholar] [CrossRef] [Green Version]
- Villanueva, J.; Shaffer, D.R.; Philip, J.; Chaparro, C.A.; Erdjument-Bromage, H.; Olshen, A.B.; Fleisher, M.; Lilja, H.; Brogi, E.; Boyd, J.; et al. Differential exoprotease activities confer tumor-specific serum peptidome patterns. J. Clin. Investig. 2006, 116, 271–284. [Google Scholar] [CrossRef] [PubMed]
- Lopez, M.F.; Mikulskis, A.; Kuzdzal, S.; Golenko, E.; Petricoin, E.F., 3rd; Liotta, L.A.; Patton, W.F.; Whiteley, G.R.; Rosenblatt, K.; Gurnani, P.; et al. A novel, high-throughput workflow for discovery and identification of serum carrier protein-bound peptide biomarker candidates in ovarian cancer samples. Clin. Chem. 2007, 53, 1067–1074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fredolini, C.; Meani, F.; Luchini, A.; Zhou, W.; Russo, P.; Ross, M.; Patanarut, A.; Tamburro, D.; Gambara, G.; Ornstein, D.; et al. Investigation of the ovarian and prostate cancer peptidome for candidate early detection markers using a novel nanoparticle biomarker capture technology. AAPS J. 2010, 12, 504–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bery, A.; Leung, F.; Smith, C.R.; Diamandis, E.P.; Kulasingam, V. Deciphering the ovarian cancer ascites fluid peptidome. Clin. Proteom. 2014, 11, 13. [Google Scholar] [CrossRef] [Green Version]
- Sinha, A.; Ignatchenko, V.; Ignatchenko, A.; Mejia-Guerrero, S.; Kislinger, T. In-depth proteomic analyses of ovarian cancer cell line exosomes reveals differential enrichment of functional categories compared to the NCI 60 proteome. Biochem. Biophys. Res. Commun. 2014, 445, 694–701. [Google Scholar] [CrossRef]
- Zhang, W.; Ou, X.; Wu, X. Proteomics profiling of plasma exosomes in epithelial ovarian cancer: A potential role in the coagulation cascade, diagnosis and prognosis. Int. J. Oncol. 2019, 54, 1719–1733. [Google Scholar] [CrossRef] [Green Version]
- Kim, S.; Han, Y.; Kim, S.I.; Kim, H.S.; Kim, S.J.; Song, Y.S. Tumor evolution and chemoresistance in ovarian cancer. NPJ Precis. Oncol. 2018, 2, 20. [Google Scholar] [CrossRef] [Green Version]
- Ye, M.; Lin, Y.; Pan, S.; Wang, Z.W.; Zhu, X. Applications of Multi-omics Approaches for Exploring the Molecular Mechanism of Ovarian Carcinogenesis. Front. Oncol. 2021, 11, 745808. [Google Scholar] [CrossRef]
- Wantoch von Rekowski, K.; König, P.; Henze, S.; Schlesinger, M.; Zawierucha, P.; Januchowski, R.; Bendas, G. Insight into Cisplatin-Resistance Signaling of W1 Ovarian Cancer Cells Emerges mTOR and HSP27 as Targets for Sensitization Strategies. Int. J. Mol. Sci. 2020, 21, 9240. [Google Scholar] [CrossRef]
- Bradbury, M.; Borràs, E.; Pérez-Benavente, A.; Gil-Moreno, A.; Santamaria, A.; Sabidó, E. Proteomic Studies on the Management of High-Grade Serous Ovarian Cancer Patients: A Mini-Review. Cancers 2021, 13, 2067. [Google Scholar] [CrossRef]
- Zhang, Y.; Wei, H.; Zhou, Y.; Li, Z.; Gou, W.; Meng, Y.; Zheng, W.; Li, J.; Li, Y.; Zhu, W. Identification of potent SENP1 inhibitors that inactivate SENP1/JAK2/STAT signaling pathway and overcome platinum drug resistance in ovarian cancer. Clin. Transl. Med. 2021, 11, e649. [Google Scholar] [CrossRef] [PubMed]
- Hutter, C.; Zenklusen, J.C. The Cancer Genome Atlas: Creating Lasting Value beyond Its Data. Cell 2018, 173, 283–285. [Google Scholar] [CrossRef] [PubMed]
- Labrie, M.; Kendsersky, N.D.; Ma, H.; Campbell, L.; Eng, J.; Chin, K.; Mills, G.B. Proteomics advances for precision therapy in ovarian cancer. Expert Rev. Proteom. 2019, 16, 841–850. [Google Scholar] [CrossRef] [PubMed]
- Ohmoto, A.; Yachida, S. Current status of poly(ADP-ribose) polymerase inhibitors and future directions. OncoTargets Ther. 2017, 10, 5195–5208. [Google Scholar] [CrossRef] [Green Version]
- Radhakrishnan, S.K.; Jette, N.; Lees-Miller, S.P. Non-homologous end joining: Emerging themes and unanswered questions. DNA Repair 2014, 17, 2–8. [Google Scholar] [CrossRef] [Green Version]
- Luo, L.; Keyomarsi, K. PARP inhibitors as single agents and in combination therapy: The most promising treatment strategies in clinical trials for BRCA-mutant ovarian and triple-negative breast cancers. Expert Opin. Investig. Drugs 2022, in press. [CrossRef]
- Audeh, M.W.; Carmichael, J.; Penson, R.T.; Friedlander, M.; Powell, B.; Bell-McGuinn, K.M.; Scott, C.; Weitzel, J.N.; Oaknin, A.; Loman, N.; et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: A proof-of-concept trial. Lancet 2010, 376, 245–251. [Google Scholar] [CrossRef]
- Stark, D.; Nankivell, M.; Pujade-Lauraine, E.; Kristensen, G.; Elit, L.; Stockler, M.; Hilpert, F.; Cervantes, A.; Brown, J.; Lanceley, A.; et al. Standard chemotherapy with or without bevacizumab in advanced ovarian cancer: Quality-of-life outcomes from the International Collaboration on Ovarian Neoplasms (ICON7) phase 3 randomised trial. Lancet Oncol. 2013, 14, 236–243. [Google Scholar] [CrossRef] [Green Version]
Approval Status | Biomarker | Sample | Sensitivity | Specificity | References | Discovery |
---|---|---|---|---|---|---|
FDA approved biomarkers | CA125 | Serum/Plasma | 60–70% | 94% | [15,61,62,63] | Immunoassay-1981 |
HE4 | Serum/Plasma/Urine | 72.9% | 94% | [61,64,65,66] | ctDNA arrays, Immunoassay-2008 | |
CancerSEEK | Blood | 98% | 99% | [63,67] | ctDNA, Multiplex PCR assays, others | |
ROMA | Serum | 79% | 78% | [15,63,68] | Immunoassays, Menopausal status-2010 | |
OVA1 (Transthyretin, β-2 microglobulin, CA125, transferrin and apolipoprotein A1) | Blood | 94% | 54% | [61,69] | Multivariate Immunoassay-2009 | |
OVERA (HE4, FSH, CA125, transferrin and apolipoprotein A1) | Blood | 91–94% | 69% | [15,61,63] | Multivariate Immunoassay-2016 | |
Other biomarker candidates | Anti-TP53, TRIM-21, NY-ESO-1 (CTAG-1A) and PAX-8 | Serum | 46–67% | 94–98% | [63,70] | PCR, line BLOT, ctDNA, Western blot, ELISA |
HE4 antigen-autoantibody complexes with CA125 | Serum | 38% alone 60–80% when combined | 98% | [63,71] | Multiplexed Immunoassay | |
MiRNAs (multiple) | Tumour/Serum/Plasma | Negative predictive value 78.6% | Positive predictive value 91.3% | [63,72,73] | Microarrays, PCR | |
Kallikrein | Serum | 21–26% | 94% | [74] | PCR, Densitometry, DNA sequencing | |
APC, RASSF1A, CHDH1, RUNX3, TFP12, SRP5 and OPCML | Serum | 85% | 91% | [63,75] | DNA methylation, PCR | |
CA125, osteopontin, macrophage inhibitory factor and anti-IL8 autoantibodies | Serum | 82% | 98% | [63,76] | Multiplexed immunoassay | |
CA125, apolipoprotein B, transthyretin | Serum | 74% | 97% | [77] | SELDI TOF, immunoassay |
Biomarker | Discovery | Sample | No. of Patients | No. of Controls | Sensitivity | Specificity | References |
---|---|---|---|---|---|---|---|
CA125, C-Reactive protein, Serum amyloid A, IL-6, IL-8 | Multiplexed assay | Plasma | 150 | 212 | 94% | 91% | [78] |
Four lipid metabolites | LC-MS | Plasma | 50 | 50 | 95% | 35% | [79] |
CA125, HE4, CA72.4, and CA15.3 (European EPIC cohort) | Immunoassay | Blood | 810 | 1939 | 95% | 98% | [80] |
c17orf64, IRX2, TUBB6 | Genome-wide methylation analysis, qMSP assays | Tissue | 23 | 36 | 100% | 100% | [81] |
92 proteins (CA125, HE4, FOLR1, KLK11, WISP1, MDK, CXCL13, MSLN, ADAM8 were significant) | Multi assay | Blood | 91 | 180 | AUC > 0.70 for the 9 proteins | Not reported | [82] |
metabolites | UPLC-MS | Plasma | 140 | 308 | Not reported | Not reported | [83] |
TRIM21, NY-ESO-1, TP53, PAX8 | ELISA, Western Blot | Serum | 114 | 50 | 46–67% | 94–98% | [70] |
miR-1246, miR-595, miR-2278 | Microarray, RT-qPCR | Serum/Tissue | 168 | 65 | 87% | 77% | [84] |
10-miRNA profile (miR-320a, miR-665, miR-3184-5p, miR-6717-5p, miR-4459, miR-6076, miR-3195, miR-1275, miR-3185, miR-4640-5p) | Microarray | Serum | 428 | 2759 | 99% | 100% | [85] |
HE4 autoantibody | Immunoassay | Serum | 145 | 212 | 38% alone, 60–80% when combined | 98% | [71] |
lncRNAs | Microarray, qPCR | Tissue | 18 | 31 | Not reported | Not reported | [86] |
Biomarkers | References | |
---|---|---|
Protein antigen | CA125, HE4, CA72.4, CA15-3, CEA and V-CAM1 Glycodelin, E-cadherin and IL-639 or transthyretin | [63] |
Immune related-Cytokine, chemokine | IL-6, IL-7, IL-8, IL-12, B7-H3, B7-H4 interferon-γ, auto antibodies against TP53, TRIM-21, NY-ESO-1 (CTAG-1A), PAX-8 | [63,87] |
Signalling molecule | EGFR, HER2, p53 mutaion, cyclin D1, cyclin E, sFas | [87] |
Inherited gene mutations | BRCA1, BRCA2 MSH2, MLH1, MSH6, PMS2 RAD51C, RAD51D, BRIP1, BARD1, CHEK2, MPE11A, NBN, PALB2, RAD50, TP53 | [88] |
Gene expression | CA125, osteopontin, kallikrein 10, secretory leukoprostease inhibitor, matrix metalloproteinase-7 FOL3, survivin, MCM3, E2Fs, VTCN1, SYNE1, AKAP14, KNDC1, DLEC1 ovarian cancer prognostic profile (115 gene signature) ctDNA: APC, RASSF1A, CHDH1, RUNX3, TFP12, SRP5, OPCML | [89,90] |
Angiogenesis | VEGF, FGF-1, Claudin-3, claudin-7, EZH2, EphA2 | [87] |
Epigenetic changes | Hypermethylation: BRCA1, RASSF1A, APC, p14ARF, p16INK4a, DAPKinase59 ARMCH1, ICAM4, LOC134466, PEG3, PYCARD SGNE160 MiRNAs: miR-200a, miR-141, miR-199a, miR-140, miR-145, miR-125b163 miR-18266, miR-21, miR-92, miR-93, miR-126, miR-29a, miR-155, miR-127, miR-99b68 | [88] |
Protein antigen | CA125, HE4, CA72.4, CA15-3, CEA and V-CAM1 Glycodelin, E-cadherin, IL-639, or transthyretin. | [63] |
Biomarkers | Techniques | Reference |
---|---|---|
Annexin3, Destin | MALDI-TOF | [44] |
ERp57 | MALDI-TOF, ESI-Q-TOF | [44] |
Activated Leucocyte Cell Adhesion Molecule, Nestin | Orbitrap | [44] |
Pyruvate kinase isozymes M1/M2 Heat shock protein family D | ESI-Q-TOF | [44] |
Abbreviation: ESI-Q-TOF; Electrospray-ionisation quadrupole time-of-flight mass spectrometry |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ghose, A.; Gullapalli, S.V.N.; Chohan, N.; Bolina, A.; Moschetta, M.; Rassy, E.; Boussios, S. Applications of Proteomics in Ovarian Cancer: Dawn of a New Era. Proteomes 2022, 10, 16. https://doi.org/10.3390/proteomes10020016
Ghose A, Gullapalli SVN, Chohan N, Bolina A, Moschetta M, Rassy E, Boussios S. Applications of Proteomics in Ovarian Cancer: Dawn of a New Era. Proteomes. 2022; 10(2):16. https://doi.org/10.3390/proteomes10020016
Chicago/Turabian StyleGhose, Aruni, Sri Vidya Niharika Gullapalli, Naila Chohan, Anita Bolina, Michele Moschetta, Elie Rassy, and Stergios Boussios. 2022. "Applications of Proteomics in Ovarian Cancer: Dawn of a New Era" Proteomes 10, no. 2: 16. https://doi.org/10.3390/proteomes10020016
APA StyleGhose, A., Gullapalli, S. V. N., Chohan, N., Bolina, A., Moschetta, M., Rassy, E., & Boussios, S. (2022). Applications of Proteomics in Ovarian Cancer: Dawn of a New Era. Proteomes, 10(2), 16. https://doi.org/10.3390/proteomes10020016