J Pathol Transl Med.  2022 Nov;56(6):309-318. 10.4132/jptm.2022.08.30.

The application of high-throughput proteomics in cytopathology

Affiliations
  • 1School of Medicine, European University Cyprus, Nicosia, Cyprus
  • 2Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
  • 3Department of Pathology, Seoul National University Hospital, Seoul, Korea

Abstract

High-throughput genomics and transcriptomics are often applied in routine pathology practice to facilitate cancer diagnosis, assess prognosis, and predict response to therapy. However, the proteins rather than nucleic acids are the functional molecules defining the cellular phenotype in health and disease, whereas genomic profiling cannot evaluate processes such as the RNA splicing or posttranslational modifications and gene expression does not necessarily correlate with protein expression. Proteomic applications have recently advanced, overcoming the issue of low depth, inconsistency, and suboptimal accuracy, also enabling the use of minimal patient-derived specimens. This review aims to present the recent evidence regarding the use of high-throughput proteomics in both exfoliative and fine-needle aspiration cytology. Most studies used mass spectrometry, as this is associated with high depth, sensitivity, and specificity, and aimed to complement the traditional cytomorphologic diagnosis, in addition to identify novel cancer biomarkers. Examples of diagnostic dilemmas subjected to proteomic analysis included the evaluation of indeterminate thyroid nodules or prediction of lymph node metastasis from thyroid cancer, also the differentiation between benign and malignant serous effusions, pancreatic cancer from autoimmune pancreatitis, non-neoplastic from malignant biliary strictures, and benign from malignant salivary gland tumors. A few cancer biomarkers—related to diverse cancers involving the breast, thyroid, bladder, lung, serous cavities, salivary glands, and bone marrow—were also discovered. Notably, residual liquid-based cytology samples were suitable for satisfactory and reproducible proteomic analysis. Proteomics could become another routine pathology platform in the near future, potentially by using validated multi-omics protocols.

Keyword

Cytology; Fine-needle aspiration; Mass spectrometry; Cancer biomarker; Proteomics

Figure

  • Fig. 1. Example of a proteomic analysis workflow utilizing cytology specimens. HPLC, high-performance liquid chromatography; ESI, electrospray ionization.


Reference

References

1. Mardis ER. Next-generation sequencing platforms. Annu Rev Anal Chem (Palo Alto Calif). 2013; 6:287–303.
Article
2. Hou YC, Neidich JA, Duncavage EJ, Spencer DH, Schroeder MC. Clinical whole-genome sequencing in cancer diagnosis. Hum Mutat. 2022; 43:1519–30.
Article
3. Hu T, Chitnis N, Monos D, Dinh A. Next-generation sequencing technologies: an overview. Hum Immunol. 2021; 82:801–11.
Article
4. Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med. 2015; 372:793–5.
Article
5. Esagian SM, Grigoriadou G, Nikas IP, et al. Comparison of liquidbased to tissue-based biopsy analysis by targeted next generation sequencing in advanced non-small cell lung cancer: a comprehensive systematic review. J Cancer Res Clin Oncol. 2020; 146:2051–66.
Article
6. Grigoriadou G, Esagian SM, Ryu HS, Nikas IP. Molecular profiling of malignant pleural effusions with next generation sequencing (NGS): evidence that supports its role in cancer management. J Pers Med. 2020; 10:206.
Article
7. Nikas IP, Mountzios G, Sydney GI, Ioakim KJ, Won JK, Papageorgis P. Evaluating pancreatic and biliary neoplasms with small biopsy-based next generation sequencing (NGS): doing more with less. Cancers (Basel). 2022; 14:397.
Article
8. Tsamis KI, Sakkas H, Giannakis A, Ryu HS, Gartzonika C, Nikas IP. Evaluating infectious, neoplastic, immunological, and degenerative diseases of the central nervous system with cerebrospinal fluidbased next-generation sequencing. Mol Diagn Ther. 2021; 25:207–29.
Article
9. Roy-Chowdhuri S, Pisapia P, Salto-Tellez M, et al. Invited reviewnext-generation sequencing: a modern tool in cytopathology. Virchows Arch. 2019; 475:3–11.
Article
10. Liu Y, Beyer A, Aebersold R. On the dependency of cellular protein levels on mRNA abundance. Cell. 2016; 165:535–50.
Article
11. Nagaraj N, Wisniewski JR, Geiger T, et al. Deep proteome and transcriptome mapping of a human cancer cell line. Mol Syst Biol. 2011; 7:548.
Article
12. Kageyama S, Isono T, Iwaki H, et al. Proteome research in urothelial carcinoma. Int J Urol. 2015; 22:621–8.
Article
13. Banks RE, Dunn MJ, Hochstrasser DF, et al. Proteomics: new perspectives, new biomedical opportunities. Lancet. 2000; 356:1749–56.
Article
14. Ramazi S, Allahverdi A, Zahiri J. Evaluation of post-translational modifications in histone proteins: a review on histone modification defects in developmental and neurological disorders. J Biosci. 2020; 45:135.
Article
15. Doll S, Gnad F, Mann M. The case for proteomics and phosphoproteomics in personalized cancer medicine. Proteomics Clin Appl. 2019; 13:e1800113.
Article
16. Menyhart O, Gyorffy B. Multi-omics approaches in cancer research with applications in tumor subtyping, prognosis, and diagnosis. Comput Struct Biotechnol J. 2021; 19:949–60.
Article
17. Singla D, Sangha MK. Multi-omic approaches to improve cancer diagnosis, prognosis, and therapeutics. In: Raza K, ed. Computational intelligence in oncology: applications in diagnosis, prognosis and therapeutics of cancers. Singapore: Springer Singapore;2022. p. 411–33.
18. Chandramouli K, Qian PY. Proteomics: challenges, techniques and possibilities to overcome biological sample complexity. Hum Genomics Proteomics. 2009; 2009:239204.
19. Aebersold R, Mann M. Mass-spectrometric exploration of proteome structure and function. Nature. 2016; 537:347–55.
Article
20. Choudhary C, Weinert BT, Nishida Y, Verdin E, Mann M. The growing landscape of lysine acetylation links metabolism and cell signalling. Nat Rev Mol Cell Biol. 2014; 15:536–50.
Article
21. Bekker-Jensen DB, Kelstrup CD, Batth TS, et al. An optimized shotgun strategy for the rapid generation of comprehensive human proteomes. Cell Syst. 2017; 4:587–99.
Article
22. Zhu Y, Piehowski PD, Zhao R, et al. Nanodroplet processing platform for deep and quantitative proteome profiling of 10-100 mammalian cells. Nat Commun. 2018; 9:882.
Article
23. Altelaar AF, Heck AJ. Trends in ultrasensitive proteomics. Curr Opin Chem Biol. 2012; 16:206–13.
Article
24. Anderson NL, Anderson NG. Proteome and proteomics: new technologies, new concepts, and new words. Electrophoresis. 1998; 19:1853–61.
Article
25. Blackstock WP, Weir MP. Proteomics: quantitative and physical mapping of cellular proteins. Trends Biotechnol. 1999; 17:121–7.
Article
26. Melton L. Protein arrays: proteomics in multiplex. Nature. 2004; 429:101–7.
27. Mueller LN, Rinner O, Schmidt A, et al. SuperHirn-a novel tool for high resolution LC-MS-based peptide/protein profiling. Proteomics. 2007; 7:3470–80.
Article
28. Ghose A, Gullapalli SV, Chohan N, et al. Applications of proteomics in ovarian cancer: dawn of a new era. Proteomes. 2022; 10:16.
Article
29. Hall DA, Ptacek J, Snyder M. Protein microarray technology. Mech Ageing Dev. 2007; 128:161–7.
Article
30. Sreekumar A, Nyati MK, Varambally S, et al. Profiling of cancer cells using protein microarrays: discovery of novel radiation-regulated proteins. Cancer Res. 2001; 61:7585–93.
31. Domon B, Aebersold R. Mass spectrometry and protein analysis. Science. 2006; 312:212–7.
Article
32. Mann M, Hendrickson RC, Pandey A. Analysis of proteins and proteomes by mass spectrometry. Annu Rev Biochem. 2001; 70:437–73.
Article
33. Alexander H, Stegner AL, Wagner-Mann C, Du Bois GC, Alexander S, Sauter ER. Proteomic analysis to identify breast cancer biomarkers in nipple aspirate fluid. Clin Cancer Res. 2004; 10:7500–10.
Article
34. Gygi SP, Corthals GL, Zhang Y, Rochon Y, Aebersold R. Evaluation of two-dimensional gel electrophoresis-based proteome analysis technology. Proc Natl Acad Sci U S A. 2000; 97:9390–5.
Article
35. Krause K, Jessnitzer B, Fuhrer D. Proteomics in thyroid tumor research. J Clin Endocrinol Metab. 2009; 94:2717–24.
Article
36. Duncan M, DeMarco ML. MALDI-MS: Emerging roles in pathology and laboratory medicine. Clin Mass Spectrom. 2019; 13:1–4.
Article
37. Kriegsmann J, Kriegsmann M, Casadonte R. MALDI TOF imaging mass spectrometry in clinical pathology: a valuable tool for cancer diagnostics (review). Int J Oncol. 2015; 46:893–906.
Article
38. Wright GL Jr. SELDI proteinchip MS: a platform for biomarker discovery and cancer diagnosis. Expert Rev Mol Diagn. 2002; 2:549–63.
Article
39. Sauter ER, Shan S, Hewett JE, Speckman P, Du Bois GC. Proteomic analysis of nipple aspirate fluid using SELDI-TOF-MS. Int J Cancer. 2005; 114:791–6.
Article
40. Leito I, Herodes K, Huopolainen M, et al. Towards the electrospray ionization mass spectrometry ionization efficiency scale of organic compounds. Rapid Commun Mass Spectrom. 2008; 22:379–84.
Article
41. Chen Y, Quan L, Jia C, et al. Proteomics-based approach reveals the involvement of SERPINB9 in recurrent and relapsed multiple myeloma. J Proteome Res. 2021; 20:2673–86.
Article
42. Pawlik TM, Fritsche H, Coombes KR, et al. Significant differences in nipple aspirate fluid protein expression between healthy women and those with breast cancer demonstrated by time-of-flight mass spectrometry. Breast Cancer Res Treat. 2005; 89:149–57.
Article
43. Sauter ER, Zhu W, Fan XJ, Wassell RP, Chervoneva I, Du Bois GC. Proteomic analysis of nipple aspirate fluid to detect biologic markers of breast cancer. Br J Cancer. 2002; 86:1440–3.
Article
44. George AL, Shaheed SU, Sutton CW. High-throughput proteomic profiling of nipple aspirate fluid from breast cancer patients compared with non-cancer controls: a step closer to clinical feasibility. J Clin Med. 2021; 10:2243.
Article
45. Franzen B, Alexeyenko A, Kamali-Moghaddam M, et al. Protein profiling of fine-needle aspirates reveals subtype-associated immune signatures and involvement of chemokines in breast cancer. Mol Oncol. 2019; 13:376–91.
Article
46. Fowler LJ, Lovell MO, Izbicka E. Fine-needle aspiration in PreservCyt: a novel and reproducible method for possible ancillary proteomic pattern expression of breast neoplasms by SELDI-TOF. Mod Pathol. 2004; 17:1012–20.
Article
47. Rapkiewicz A, Espina V, Zujewski JA, et al. The needle in the haystack: application of breast fine-needle aspirate samples to quantitative protein microarray technology. Cancer. 2007; 111:173–84.
Article
48. Pagni F, Mainini V, Garancini M, et al. Proteomics for the diagnosis of thyroid lesions: preliminary report. Cytopathology. 2015; 26:318–24.
Article
49. Mainini V, Pagni F, Garancini M, et al. An alternative approach in endocrine pathology research: MALDI-IMS in papillary thyroid carcinoma. Endocr Pathol. 2013; 24:250–3.
Article
50. Capitoli G, Piga I, Clerici F, et al. Analysis of Hashimoto’s thyroiditis on fine needle aspiration samples by MALDI-imaging. Biochim Biophys Acta Proteins Proteom. 2020; 1868:140481.
Article
51. Pagni F, De Sio G, Garancini M, et al. Proteomics in thyroid cytopathology: relevance of MALDI-imaging in distinguishing malignant from benign lesions. Proteomics. 2016; 16:1775–84.
Article
52. Capitoli G, Piga I, L'Imperio V, et al. Cytomolecular classification of thyroid nodules using fine-needle washes aspiration biopsies. Int J Mol Sci. 2022; 23:4156.
Article
53. Capitoli G, Piga I, Galimberti S, et al. MALDI-MSI as a Complementary diagnostic tool in cytopathology: a pilot study for the characterization of thyroid nodules. Cancers (Basel). 2019; 11:1377.
Article
54. Schwamborn K, Krieg RC, Uhlig S, Ikenberg H, Wellmann A. MALDI imaging as a specific diagnostic tool for routine cervical cytology specimens. Int J Mol Med. 2011; 27:417–21.
Article
55. Schwamborn K, Weirich G, Steiger K, et al. Discerning the primary carcinoma in malignant peritoneal and pleural effusions using imaging mass spectrometry: a deasibility study. Proteomics Clin Appl. 2019; 13:e1800064.
56. Perzanowska A, Fatalska A, Wojtas G, et al. An MRM-based cytokeratin marker assay as a tool for cancer studies: application to lung cancer pleural effusions. Proteomics Clin Appl. 2018; 12:170084.
Article
57. Li H, Tang Z, Zhu H, Ge H, Cui S, Jiang W. Proteomic study of benign and malignant pleural effusion. J Cancer Res Clin Oncol. 2016; 142:1191–200.
Article
58. Inoue H, Eguchi A, Kobayashi Y, et al. Extracellular vesicles from pancreatic ductal adenocarcinoma endoscopic ultrasound-fine needle aspiration samples contain a protein barcode. J Hepatobiliary Pancreat Sci. 2022; 29:394–403.
Article
59. Lee LS, Banks PA, Bellizzi AM, et al. Inflammatory protein profiling of pancreatic cyst fluid using EUS-FNA in tandem with cytokine microarray differentiates between branch duct IPMN and inflammatory cysts. J Immunol Methods. 2012; 382:142–9.
Article
60. Navaneethan U, Lourdusamy V, Gk Venkatesh P, Willard B, Sanaka MR, Parsi MA. Bile proteomics for differentiation of malignant from benign biliary strictures: a pilot study. Gastroenterol Rep (Oxf). 2015; 3:136–43.
Article
61. Seccia V, Navari E, Donadio E, et al. Proteomic investigation of malignant major salivary gland tumors. Head Neck Pathol. 2020; 14:362–73.
Article
62. Pawlik TM, Hawke DH, Liu Y, et al. Proteomic analysis of nipple aspirate fluid from women with early-stage breast cancer using isotopecoded affinity tags and tandem mass spectrometry reveals differential expression of vitamin D binding protein. BMC Cancer. 2006; 6:68.
Article
63. Pavlou MP, Kulasingam V, Sauter ER, Kliethermes B, Diamandis EP. Nipple aspirate fluid proteome of healthy females and patients with breast cancer. Clin Chem. 2010; 56:848–55.
Article
64. Noble JL, Dua RS, Coulton GR, Isacke CM, Gui GP. A comparative proteinomic analysis of nipple aspiration fluid from healthy women and women with breast cancer. Eur J Cancer. 2007; 43:2315–20.
Article
65. Giusti L, Iacconi P, Ciregia F, et al. Proteomic analysis of human thyroid fine needle aspiration fluid. J Endocrinol Invest. 2007; 30:865–9.
Article
66. Giusti L, Iacconi P, Ciregia F, et al. Fine-needle aspiration of thyroid nodules: proteomic analysis to identify cancer biomarkers. J Proteome Res. 2008; 7:4079–88.
Article
67. Ciregia F, Giusti L, Molinaro A, et al. Proteomic analysis of fineneedle aspiration in differential diagnosis of thyroid nodules. Transl Res. 2016; 176:81–94.
Article
68. Ucal Y, Eravci M, Tokat F, Duren M, Ince U, Ozpinar A. Proteomic analysis reveals differential protein expression in variants of papillary thyroid carcinoma. EuPA Open Proteom. 2017; 17:1–6.
Article
69. Lin P, Yao Z, Sun Y, et al. Deciphering novel biomarkers of lymph node metastasis of thyroid papillary microcarcinoma using proteomic analysis of ultrasound-guided fine-needle aspiration biopsy samples. J Proteomics. 2019; 204:103414.
Article
70. Park JH, Lee C, Han D, et al. Moesin (MSN) as a novel proteomebased diagnostic marker for early detection of invasive bladder urothelial carcinoma in liquid-based cytology. Cancers (Basel). 2020; 12:1018.
Article
71. Yang N, Feng S, Shedden K, et al. Urinary glycoprotein biomarker discovery for bladder cancer detection using LC/MS-MS and labelfree quantification. Clin Cancer Res. 2011; 17:3349–59.
Article
72. Theodorescu D, Wittke S, Ross MM, et al. Discovery and validation of new protein biomarkers for urothelial cancer: a prospective analysis. Lancet Oncol. 2006; 7:230–40.
Article
73. Lee H, Kim K, Woo J, et al. Quantitative proteomic analysis identifies AHNAK (neuroblast differentiation-associated protein AHNAK) as a novel candidate biomarker for bladder urothelial carcinoma diagnosis by liquid-based cytology. Mol Cell Proteomics. 2018; 17:1788–802.
Article
74. Boylan KL, Afiuni-Zadeh S, Geller MA, et al. A feasibility study to identify proteins in the residual Pap test fluid of women with normal cytology by mass spectrometry-based proteomics. Clin Proteomics. 2014; 11:30.
Article
75. Boylan KL, Afiuni-Zadeh S, Geller MA, Argenta PA, Griffin TJ, Skubitz APN. Evaluation of the potential of Pap test fluid and cervical swabs to serve as clinical diagnostic biospecimens for the detection of ovarian cancer by mass spectrometry-based proteomics. Clin Proteomics. 2021; 18:4.
Article
76. Liu PJ, Chen CD, Wang CL, et al. In-depth proteomic analysis of six types of exudative pleural effusions for nonsmall cell lung cancer biomarker discovery. Mol Cell Proteomics. 2015; 14:917–32.
Article
77. Li Y, Lian H, Jia Q, Wan Y. Proteome screening of pleural effusions identifies IL1A as a diagnostic biomarker for non-small cell lung cancer. Biochem Biophys Res Commun. 2015; 457:177–82.
Article
78. Hegmans JP, Veltman JD, Fung ET, et al. Protein profiling of pleural effusions to identify malignant pleural mesothelioma using SELDITOF MS. Technol Cancer Res Treat. 2009; 8:323–32.
Article
79. Ucal Y, Tokat F, Duren M, Ince U, Ozpinar A. Peptide profile differences of noninvasive follicular thyroid neoplasm with papillary-like nuclear features, encapsulated follicular variant, and classical papillary thyroid carcinoma: an application of matrix-assisted laser desorption/ionization mass spectrometry imaging. Thyroid. 2019; 29:1125–37.
Article
80. Torres-Cabala C, Bibbo M, Panizo-Santos A, et al. Proteomic identification of new biomarkers and application in thyroid cytology. Acta Cytol. 2006; 50:518–28.
Article
Full Text Links
  • JPTM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr