GP88/PGRN Serum Levels Are Associated with Prognosis for Oral Squamous Cell Carcinoma Patients

Thomas Greither, Tina Steiner, Matthias Bache, Ginette Serrero, Sven Otto, Helge Taubert, Alexander W Eckert, Matthias Kappler, Thomas Greither, Tina Steiner, Matthias Bache, Ginette Serrero, Sven Otto, Helge Taubert, Alexander W Eckert, Matthias Kappler

Abstract

Progranulin (PGRN)/GP88 is a growth factor that is expressed in a wide range of tumor tissues. The secreted form is involved in various biological processes including proliferation and inflammation. In several tumor types, the serum GP88 level is associated with a patient's prognosis; however, data for oral squamous cell carcinomas (OSCCs) have not yet been reported. We measured the serum GP88 levels in 96 OSCC patients by an enzyme immunosorbent assay (EIA) and correlated these data with clinicopathological parameters and patient outcomes. The GP88 levels in the serum of OSCC patients and healthy volunteers were comparable. In OSCC patients, the levels did not correlate with age, sex, or TNM status. In a Kaplan-Meier survival analysis, a serum GP88 level < 68 ng/mL was significantly associated with worsened survival (p = 0.0005, log-rank-test) as well as in uni- and multivariate Cox regression analyses (RR = 4.6 [1.6-12.9], p = 0.004 and RR = 4.2 [1.2-12.0], p = 0.008). This effect was predominant in OSCC patients older than 60.5 years (p = 0.027), while in younger patients no significant association between serum GP88 levels and prognosis could be observed. Altogether, lower serum GP88 levels are significantly associated with a worsened outcome for an OSCC and may be an interesting candidate for risk stratification during OSCC therapy.

Keywords: GP88; OSCC; prognosis; progranulin; serum.

Conflict of interest statement

GS is co-founder and CEO of A&G Pharmaceutical Inc., Columbia, Maryland, USA. The other authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Serum GP88 level distribution in OSCC patients and healthy volunteers.
Figure 2
Figure 2
Distribution of serum GP88 levels according to (a) sex, (b) age median, (c) T stage or (d) nodal metastasis.
Figure 3
Figure 3
ROC analysis of serum GP88 levels according to overall survival of the OSCC patients used for Youden index calculation. AUC of the ROC curve was 0.684 (95% CI: 0.577–0.792), p = 0.002.
Figure 4
Figure 4
Survival analyses according to (a) Kaplan-Meier and (b) univariate Cox regression survival analyses estimating the influence of the serum GP88 level on the OSCC patients prognosis.
Figure 5
Figure 5
Multivariate Cox regression analysis modelling the survival of OSCC patients according to their serum GP88 levels.

References

    1. Fitzmaurice C., Allen C., Barber R.M., Barregard L., Bhutta Z.A., Brenner H., Dicker D.J., Chimed-Orchir O., Dandona R., Dandona L., et al. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived with Disability, and Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to 2015: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2017;3:524–548. doi: 10.1001/jamaoncol.2016.5688.
    1. Zizzari V.L., Zara S., Tetè G., Vinci R., Gherlone E., Cataldi A. Biologic and clinical aspects of integration of different bone substitutes in oral surgery: A literature review. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2016;122:392–402. doi: 10.1016/j.oooo.2016.04.010.
    1. Probst F.A., Fliefel R., Burian E., Probst M., Eddicks M., Cornelsen M., Riedl C., Seitz H., Aszódi A., Schieker M., et al. Bone regeneration of minipig mandibular defect by adipose derived mesenchymal stem cells seeded tri-calcium phosphate- poly(D,L-lactide-co-glycolide) scaffolds. Sci. Rep. 2020;10:2062. doi: 10.1038/s41598-020-59038-8.
    1. Gherlone E.F., Capparé P., Tecco S., Polizzi E., Pantaleo G., Gastaldi G., Grusovin M.G. Implant Prosthetic Rehabilitation in Controlled HIV-Positive Patients: A Prospective Longitudinal Study with 1-Year Follow-Up. Clin. Implant Dent. Relat. Res. 2016;18:725–734. doi: 10.1111/cid.12353.
    1. Fliefel R., Ehrenfeld M., Otto S. Induced pluripotent stem cells (iPSCs) as a new source of bone in reconstructive surgery: A systematic review and meta-analysis of preclinical studies. J. Tissue Eng. Regen. Med. 2018;12:1780–1797. doi: 10.1002/term.2697.
    1. Capparè P., Tetè G., Sberna M.T., Panina-Bordignon P. The Emerging Role of Stem Cells in Regenerative Dentistry. Curr. Gene Ther. 2020;20:259–268. doi: 10.2174/1566523220999200818115803.
    1. Eckert A.W., Wickenhauser C., Salins P.C., Kappler M., Bukur J., Seliger B. Clinical relevance of the tumor microenvironment and immune escape of oral squamous cell carcinoma. J. Transl. Med. 2016;14:85. doi: 10.1186/s12967-016-0828-6.
    1. Serrero G., Hicks D. Measurement of Circulating Progranulin (PGRN/GP88/GEP) by Enzyme-Linked Immunosorbent Assay and Application in Human Diseases. Methods Mol. Biol. 2018;1806:95–105. doi: 10.1007/978-1-4939-8559-3_7.
    1. Tkaczuk K.R., Yue B., Zhan M., Tait N., Yarlagadda L., Dai H., Serrero G. Increased Circulating Level of the Survival Factor GP88 (Progranulin) in the Serum of Breast Cancer Patients When Compared to Healthy Subjects. Breast Cancer. 2011;5:155–162. doi: 10.4137/BCBCR.S7224.
    1. Tkaczuk K.H.R., Hawkins D., Yue B., Hicks D., Tait N., Serrero G. Association of Serum Progranulin Levels with Disease Progression, Therapy Response and Survival in Patients with Metastatic Breast Cancer. Clin. Breast Cancer. 2020;20:220–227. doi: 10.1016/j.clbc.2019.11.010.
    1. Koo D.H., Park C.-Y., Lee E.S., Ro J., Oh S.W. Progranulin as a prognostic biomarker for breast cancer recurrence in patients who had hormone receptor-positive tumors: A cohort study. PLoS ONE. 2012;7:e39880. doi: 10.1371/journal.pone.0039880.
    1. Edelman M.J., Feliciano J., Yue B., Bejarano P., Ioffe O., Reisman D., Hawkins D., Gai Q., Hicks D., Serrero G. GP88 (progranulin): A novel tissue and circulating biomarker for non-small cell lung carcinoma. Hum. Pathol. 2014;45:1893–1899. doi: 10.1016/j.humpath.2014.05.011.
    1. Greither T., Fischer K., Theil G., Marcou M., Holzhausen H.-J., Weigelt K., Serrero G., Hicks D., Yue B., Fornara P., et al. Expression of GP88 (progranulin) in serum of prostate cancer patients is associated with Gleason scores and overall survival. Cancer Manag. Res. 2018;10:4173–4180. doi: 10.2147/CMAR.S172069.
    1. Pan C.-X., Kinch M.S., Kiener P.A., Langermann S., Serrero G., Le S., Corvera J., Sweeney C.J., Li L., Zhang S., et al. PC cell-derived growth factor expression in prostatic intraepithelial neoplasia and prostatic adenocarcinoma. Clin. Cancer Res. 2004;10:1333–1337. doi: 10.1158/1078-0432.CCR-1123-03.
    1. Sleegers K., Brouwers N., van Damme P., Engelborghs S., Gijselinck I., van der Zee J., Peeters K., Mattheijssens M., Cruts M., Vandenberghe R., et al. Serum biomarker for progranulin-associated frontotemporal lobar degeneration. Ann. Neurol. 2009;65:603–609. doi: 10.1002/ana.21621.
    1. Schofield E.C., Halliday G.M., Kwok J., Loy C., Double K.L., Hodges J.R. Low serum progranulin predicts the presence of mutations: A prospective study. J. Alzheimers Dis. 2010;22:981–984. doi: 10.3233/JAD-2010-101032.
    1. Youn B.-S., Bang S.-I., Klöting N., Park J.W., Lee N., Oh J.-E., Pi K.-B., Lee T.H., Ruschke K., Fasshauer M., et al. Serum progranulin concentrations may be associated with macrophage infiltration into omental adipose tissue. Diabetes. 2009;58:627–636. doi: 10.2337/db08-1147.
    1. Matsubara T., Mita A., Minami K., Hosooka T., Kitazawa S., Takahashi K., Tamori Y., Yokoi N., Watanabe M., Matsuo E.-I., et al. PGRN is a key adipokine mediating high fat diet-induced insulin resistance and obesity through IL-6 in adipose tissue. Cell Metab. 2012;15:38–50. doi: 10.1016/j.cmet.2011.12.002.
    1. Tanaka A., Tsukamoto H., Mitoma H., Kiyohara C., Ueda N., Ayano M., Ohta S.-i., Inoue Y., Arinobu Y., Niiro H., et al. Serum progranulin levels are elevated in patients with systemic lupus erythematosus, reflecting disease activity. Arthritis Res. Ther. 2012;14:R244. doi: 10.1186/ar4087.
    1. Qiu F., Song L., Ding F., Liu H., Shu Q., Yang N., Liu W., Li X. Expression level of the growth factor progranulin is related with development of systemic lupus erythematosus. Diagn. Pathol. 2013;8:88. doi: 10.1186/1746-1596-8-88.
    1. Yamamoto Y., Takemura M., Serrero G., Hayashi J., Yue B., Tsuboi A., Kubo H., Mitsuhashi T., Mannami K., Sato M., et al. Increased serum GP88 (Progranulin) concentrations in rheumatoid arthritis. Inflammation. 2014;37:1806–1813. doi: 10.1007/s10753-014-9911-4.
    1. Cerezo L.A., Kuklová M., Hulejová H., Vernerová Z., Kaspříková N., Veigl D., Pavelka K., Vencovský J., Šenolt L. Progranulin Is Associated with Disease Activity in Patients with Rheumatoid Arthritis. Mediat. Inflamm. 2015;2015:740357. doi: 10.1155/2015/740357.
    1. Zhou J., Gao G., Crabb J.W., Serrero G. Purification of an autocrine growth factor homologous with mouse epithelin precursor from a highly tumorigenic cell line. J. Biol. Chem. 1993;268:10863–10869. doi: 10.1016/S0021-9258(18)82064-6.
    1. He Z., Bateman A. Progranulin gene expression regulates epithelial cell growth and promotes tumor growth in vivo. Cancer Res. 1999;59:3222–3229.
    1. Lu R., Serrero G. Inhibition of PC cell-derived growth factor (PCDGF, epithelin/granulin precursor) expression by antisense PCDGF cDNA transfection inhibits tumorigenicity of the human breast carcinoma cell line MDA-MB-468. Proc. Natl. Acad. Sci. USA. 2000;97:3993–3998. doi: 10.1073/pnas.97.8.3993.
    1. Cheung S.T., Wong S.Y., Leung K.L., Chen X., So S., Ng I.O., Fan S.T. Granulin-epithelin precursor overexpression promotes growth and invasion of hepatocellular carcinoma. Clin. Cancer Res. 2004;10:7629–7636. doi: 10.1158/1078-0432.CCR-04-0960.
    1. Abrhale T., Brodie A., Sabnis G., Macedo L., Tian C., Yue B., Serrero G. GP88 (PC-Cell Derived Growth Factor, progranulin) stimulates proliferation and confers letrozole resistance to aromatase overexpressing breast cancer cells. BMC Cancer. 2011;11:231. doi: 10.1186/1471-2407-11-231.
    1. Zhao J., Li X., Liu J., Jiang W., Wen D., Xue H. Effect of Progranulin on Migration and Invasion of Human Colon Cancer Cells. J. Coll. Physicians Surg. Pak. 2018;28:607–611. doi: 10.29271/jcpsp.2018.08.607.
    1. Yang D., Wang L.-L., Dong T.-T., Shen Y.-H., Guo X.-S., Liu C.-Y., Liu J., Zhang P., Li J., Sun Y.-P. Progranulin promotes colorectal cancer proliferation and angiogenesis through TNFR2/Akt and ERK signaling pathways. Am. J. Cancer Res. 2015;5:3085–3097.
    1. Kojima Y., Ono K., Inoue K., Takagi Y., Kikuta K.-i., Nishimura M., Yoshida Y., Nakashima Y., Matsumae H., Furukawa Y., et al. Progranulin expression in advanced human atherosclerotic plaque. Atherosclerosis. 2009;206:102–108. doi: 10.1016/j.atherosclerosis.2009.02.017.
    1. Yang D., Li R., Wang H., Wang J., Han L., Pan L., Li X., Kong Q., Wang G., Su X. Clinical implications of progranulin in gastric cancer and its regulation via a positive feedback loop involving AKT and ERK signaling pathways. Mol. Med. Rep. 2017;16:9685–9691. doi: 10.3892/mmr.2017.7796.
    1. Ong C.H.P., Bateman A. Progranulin (granulin-epithelin precursor, PC-cell derived growth factor, acrogranin) in proliferation and tumorigenesis. Histol. Histopathol. 2003;18:1275–1288. doi: 10.14670/HH-18.1275.
    1. Ding D., Li C., Zhao T., Li D., Yang L., Zhang B. LncRNA H19/miR-29b-3p/PGRN Axis Promoted Epithelial-Mesenchymal Transition of Colorectal Cancer Cells by Acting on Wnt Signaling. Mol. Cells. 2018;41:423–435. doi: 10.14348/molcells.2018.2258.
    1. Tangkeangsirisin W., Serrero G. PC cell-derived growth factor (PCDGF/GP88, progranulin) stimulates migration, invasiveness and VEGF expression in breast cancer cells. Carcinogenesis. 2004;25:1587–1592. doi: 10.1093/carcin/bgh171.
    1. Bandey I., Chiou S.-H., Huang A.-P., Tsai J.-C., Tu P.-H. Progranulin promotes Temozolomide resistance of glioblastoma by orchestrating DNA repair and tumor stemness. Oncogene. 2015;34:1853–1864. doi: 10.1038/onc.2014.92.
    1. Cheung P.F.Y., Cheng C.K.C., Wong N.C.L., Ho J.C.Y., Yip C.W., Lui V.C.H., Cheung A.N.Y., Fan S.T., Cheung S.T. Granulin-epithelin precursor is an oncofetal protein defining hepatic cancer stem cells. PLoS ONE. 2011;6:e28246. doi: 10.1371/journal.pone.0028246.
    1. Wang W., Hayashi J., Serrero G. PC cell-derived growth factor confers resistance to dexamethasone and promotes tumorigenesis in human multiple myeloma. Clin. Cancer Res. 2006;12:49–56. doi: 10.1158/1078-0432.CCR-05-0929.
    1. Pizarro G.O., Zhou X.C., Koch A., Gharib M., Raval S., Bible K., Jones M.B. Prosurvival function of the granulin-epithelin precursor is important in tumor progression and chemoresponse. Int. J. Cancer. 2007;120:2339–2343. doi: 10.1002/ijc.22559.
    1. Zhu J., Nathan C., Jin W., Sim D., Ashcroft G.S., Wahl S.M., Lacomis L., Erdjument-Bromage H., Tempst P., Wright C.D., et al. Conversion of proepithelin to epithelins: Roles of SLPI and elastase in host defense and wound repair. Cell. 2002;111:867–878. doi: 10.1016/S0092-8674(02)01141-8.
    1. Yin F., Banerjee R., Thomas B., Zhou P., Qian L., Jia T., Ma X., Ma Y., Iadecola C., Beal M.F., et al. Exaggerated inflammation, impaired host defense, and neuropathology in progranulin-deficient mice. J. Exp. Med. 2010;207:117–128. doi: 10.1084/jem.20091568.
    1. Eckert A.W., Horter S., Bethmann D., Kotrba J., Kaune T., Rot S., Bache M., Bilkenroth U., Reich W., Greither T., et al. Investigation of the Prognostic Role of Carbonic Anhydrase 9 (CAIX) of the Cellular mRNA/Protein Level or Soluble CAIX Protein in Patients with Oral Squamous Cell Carcinoma. Int. J. Mol. Sci. 2019;20:375. doi: 10.3390/ijms20020375.
    1. Rot S., Kaune T., Taubert H., Greither T., Kotrba J., Güttler A., Wichmann H., Bilkenroth U., Wienke A., Al-Nawas B., et al. Prognostic impact of mRNA levels of LGR5 transcript variants in OSCC patients. BMC Cancer. 2019;19:155. doi: 10.1186/s12885-019-5327-8.
    1. Yamamoto Y., Goto N., Takemura M., Yamasuge W., Yabe K., Takami T., Miyazaki T., Takeuchi T., Shiraki M., Shimizu M., et al. Association between increased serum GP88 (progranulin) concentrations and prognosis in patients with malignant lymphomas. Clin. Chim. Acta. 2017;473:139–146. doi: 10.1016/j.cca.2017.07.024.
    1. Wang M., Li G., Yin J., Lin T., Zhang J. Progranulin overexpression predicts overall survival in patients with glioblastoma. Med. Oncol. 2012;29:2423–2431. doi: 10.1007/s12032-011-0131-6.
    1. Zhang N., Yang N., Chen Q., Qiu F., Li X. Upregulated expression level of the growth factor, progranulin, is associated with the development of primary Sjögren’s syndrome. Exp. Ther. Med. 2014;8:1643–1647. doi: 10.3892/etm.2014.1981.
    1. Qi X., Guo H., Sun C., Tian Y., Ding M., Yang Y., Jin H. Clinical significance of progranulin correlated with serum soluble Oxford 40 ligand in primary Sjögren’s syndrome. Medicine. 2020;99:e19967. doi: 10.1097/MD.0000000000019967.
    1. Han J.J., Yu M., Houston N., Steinberg S.M., Kohn E.C. Progranulin is a potential prognostic biomarker in advanced epithelial ovarian cancers. Gynecol. Oncol. 2011;120:5–10. doi: 10.1016/j.ygyno.2010.09.006.
    1. Carlson A.M., Maurer M.J., Goergen K.M., Kalli K.R., Erskine C.L., Behrens M.D., Knutson K.L., Block M.S. Utility of progranulin and serum leukocyte protease inhibitor as diagnostic and prognostic biomarkers in ovarian cancer. Cancer Epidemiol. Biomark. Prev. 2013;22:1730–1735. doi: 10.1158/1055-9965.EPI-12-1368.
    1. Göbel M., Eisele L., Möllmann M., Hüttmann A., Johansson P., Scholtysik R., Bergmann M., Busch R., Döhner H., Hallek M., et al. Progranulin is a novel independent predictor of disease progression and overall survival in chronic lymphocytic leukemia. PLoS ONE. 2013;8:e72107. doi: 10.1371/journal.pone.0072107.
    1. El-Ghammaz A.M.S., Azzazi M.O., Mostafa N., Hegab H.M., Mahmoud A.A. Prognostic significance of serum progranulin level in de novo adult acute lymphoblastic leukemia patients. Clin. Exp. Med. 2020;20:269–276. doi: 10.1007/s10238-020-00610-x.
    1. Luo Q., He X., Zheng Y., Ning P., Xu Y., Yang D., Shang Y., Gao Z. Elevated progranulin as a novel biomarker to predict poor prognosis in community-acquired pneumonia. J. Infect. 2020;80:167–173. doi: 10.1016/j.jinf.2019.12.004.
    1. Li G., Dong T., Yang D., Gao A., Luo J., Yang H., Wang L. Progranulin promotes lymphangiogenesis through VEGF-C and is an independent risk factor in human esophageal cancers. Hum. Pathol. 2018;75:116–124. doi: 10.1016/j.humpath.2018.02.008.
    1. Taghavi N., Yazdi I. Prognostic factors of survival rate in oral squamous cell carcinoma: Clinical, histologic, genetic and molecular concepts. Arch. Iran. Med. 2015;18:314–319.
    1. Ong W., Zhao R., Lui B., Tan W., Ebrahimi A., Clark J.R., Soo K.-C., Tan N.-C., Tan H.-K., Iyer N.G. Prognostic significance of lymph node density in squamous cell carcinoma of the tongue. Head Neck. 2016;38(Suppl. 1):E859–E866. doi: 10.1002/hed.24113.
    1. Tang W., Lu Y., Tian Q.-Y., Zhang Y., Guo F.-J., Liu G.-Y., Syed N.M., Lai Y., Lin E.A., Kong L., et al. The growth factor progranulin binds to TNF receptors and is therapeutic against inflammatory arthritis in mice. Science. 2011;332:478–484. doi: 10.1126/science.1199214.
    1. Li X., Zuo Z., Chen Q., Li J., Tang W., Yang P. Progranulin is highly expressed in patients with chronic periodontitis and protects against experimental periodontitis in rats. J. Periodontol. 2018;89:1418–1427. doi: 10.1002/JPER.18-0132.
    1. Lee L.T., Wong Y.K., Hsiao H.Y., Wang Y.W., Chan M.Y., Chang K.W. Evaluation of saliva and plasma cytokine biomarkers in patients with oral squamous cell carcinoma. Int. J. Oral Maxillofac. Surg. 2018;47:699–707. doi: 10.1016/j.ijom.2017.09.016.
    1. Deepthi G., Nandan S.R.K., Kulkarni P.G. Salivary Tumour Necrosis Factor-α as a Biomarker in Oral Leukoplakia and Oral Squamous Cell Carcinoma. Asian Pac. J. Cancer Prev. 2019;20:2087–2093. doi: 10.31557/APJCP.2019.20.7.2087.
    1. Chen Y.-Q., Wang C.-J., Xie K., Lei M., Chai Y.-S., Xu F., Lin S.-H. Progranulin Improves Acute Lung Injury through Regulating the Differentiation of Regulatory T Cells and Interleukin-10 Immunomodulation to Promote Macrophage Polarization. Mediat. Inflamm. 2020;2020:9704327. doi: 10.1155/2020/9704327.
    1. Jian J., Konopka J., Liu C. Insights into the role of progranulin in immunity, infection, and inflammation. J. Leukoc. Biol. 2013;93:199–208. doi: 10.1189/jlb.0812429.
    1. Hanahan D., Weinberg R.A. Hallmarks of cancer: The next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Yan C., Grimm W.A., Garner W.L., Qin L., Travis T., Tan N., Han Y.-P. Epithelial to mesenchymal transition in human skin wound healing is induced by tumor necrosis factor-alpha through bone morphogenic protein-2. Am. J. Pathol. 2010;176:2247–2258. doi: 10.2353/ajpath.2010.090048.
    1. Zhou C., Liu J., Tang Y., Liang X. Inflammation linking EMT and cancer stem cells. Oral Oncol. 2012;48:1068–1075. doi: 10.1016/j.oraloncology.2012.06.005.
    1. Dikova V.R., Principe S., Bagan J.V. Salivary inflammatory proteins in patients with oral potentially malignant disorders. J. Clin. Exp. Dent. 2019;11:e659–e664. doi: 10.4317/jced.55917.
    1. Liu L., Guo H., Song A., Huang J., Zhang Y., Jin S., Li S., Zhang L., Yang C., Yang P. Progranulin inhibits LPS-induced macrophage M1 polarization via NF-кB and MAPK pathways. BMC Immunol. 2020;21:32. doi: 10.1186/s12865-020-00355-y.

Source: PubMed

3
Abonner