The Co-expression of HER Family Members and CD109 is common in Pancreatic Cancer.

Main Article Content

Tanzeel Khan Alan M. Seddon Said A. Khelwatty Angus Dalgleish Izhar Bagwan Satvinder Mudan Helmout Modjtahedi

Abstract

In the absence of biomarkers for the early detection of pancreatic cancer and the development of more effective therapeutic interventions, pancreatic cancer is projected to become the second leading cause of death from cancer in the Western world. In the past four decades, aberrant expression and activation of human epidermal growth factor receptor (EGFR, also called HER) family members have been reported in a wide range of human cancers. Currently, of the various types of drugs targeting one or more members of the HER family, only the EGFR-specific tyrosine kinase inhibitor erlotinib and more recently Zenocutuzumab, a HER-2/HER3 bispecific antibody, were granted breakthrough therapy designation by the FDA for the treatment of patients with pancreatic cancer. However, the therapeutic benefit may be modest in some patients. Hence there is an urgent need for the identification of biomarkers as prognostic indicators, therapeutic targets, and for the response to therapy and the development of more effective therapeutic agents for this highly heterogeneous cancer. In this study, for the first time, we investigated the relative expression and prognostic significance of all members of the HER family, the type-III EGFR mutant (EGFRvIII), and CD109 in tissue microarrays (TMAs) and whole tumour specimens (WTS) from pancreatic cancer patients by immunohistochemistry. We found the positive expression of wild-type EGFR (wt-EGFR) (63%, 4.7%), HER2 (75%, 14%), HER3 (none, 14%), HER4 (45%, 21%), EGFRvIII (5%, 3%), and CD109 (67%, 55%) in TMAs (USA) and WTS (UK) from pancreatic cancer patients respectively. Our results also show that the co-expression of HER family members with CD109 occurs frequently in patients with pancreatic cancer. In addition, the co-expression of HER4/CD109 may also be associated with poorer overall survival in such patients. In this article, we shall discuss these findings and their implications and future opportunities for more effective targeting of HER positive pancreatic cancer using the HER inhibitors in combination with other drugs and therapeutic interventions.

Keywords: Pancreatic Cancer

Article Details

How to Cite
KHAN, Tanzeel et al. The Co-expression of HER Family Members and CD109 is common in Pancreatic Cancer.. Medical Research Archives, [S.l.], v. 11, n. 11, nov. 2023. ISSN 2375-1924. Available at: <https://esmed.org/MRA/mra/article/view/4689>. Date accessed: 16 may 2024. doi: https://doi.org/10.18103/mra.v11i11.4689.
Section
Research Articles

References

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394-424.

2. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17-48.

3. Sohn TA, Yeo CJ, Cameron JL, et al. Resected adenocarcinoma of the pancreas-616 patients: results, outcomes, and prognostic indicators. J Gastrointest Surg. 2000;4(6):567-579.

4. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7-33.

5. Rawla P, Sunkara T, Gaduputi V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J Oncol. 2019;10(1):10-27.

6. Arias-Pinilla GA, Modjtahedi H. Therapeutic Application of Monoclonal Antibodies in Pancreatic Cancer: Advances, Challenges and Future Opportunities. Cancers (Basel). 2021;13(8).

7. Li Q, Zhang L, Li X, et al. The prognostic significance of human epidermal growth factor receptor family protein expression in operable pancreatic cancer : HER1-4 protein expression and prognosis in pancreatic cancer. BMC Cancer. 2016;16(1):910.

8. Perini MV, Montagnini AL, Coudry R, et al. Prognostic significance of epidermal growth factor receptor overexpression in pancreas cancer and nodal metastasis. ANZ J Surg. 2015;85(3):174-178.

9. Mahipal A, McDonald MJ, Witkiewicz A, Carr BI. Cell membrane and cytoplasmic epidermal growth factor receptor expression in pancreatic ductal adenocarcinoma. Med Oncol. 2012;29(1):134-139.

10. Einama T, Ueda S, Tsuda H, et al. Membranous and cytoplasmic expression of epidermal growth factor receptor in metastatic pancreatic ductal adenocarcinoma. Exp Ther Med. 2012;3(6):931-936.

11. Ioannou N, Seddon AM, Dalgleish A, Mackintosh D, Modjtahedi H. Expression pattern and targeting of HER family members and IGF-IR in pancreatic cancer. Front Biosci (Landmark Ed). 2012;17:2698-2724.

12. Seshacharyulu P, Ponnusamy MP, Haridas D, Jain M, Ganti AK, Batra SK. Targeting the EGFR signaling pathway in cancer therapy. Expert Opin Ther Targets. 2012;16(1):15-31.

13. Tebbutt N, Pedersen MW, Johns TG. Targeting the ERBB family in cancer: couples therapy. Nat Rev Cancer. 2013;13(9):663-673.

14. Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25(15):1960-1966.

15. Schram AM, Goto K, Kim D-W, et al. Efficacy and safety of zenocutuzumab, a HER2 x HER3 bispecific antibody, across advanced NRG1 fusion (NRG1+) cancers. Journal of Clinical Oncology. 2022;40(16_suppl):105-105.

16. Lin M, Sutherland DR, Horsfall W, et al. Cell surface antigen CD109 is a novel member of the alpha(2) macroglobulin/C3, C4, C5 family of thioester-containing proteins. Blood. 2002;99(5):1683-1691.

17. Solomon KR, Sharma P, Chan M, Morrison PT, Finberg RW. CD109 represents a novel branch of the alpha2-macroglobulin/ complement gene family. Gene. 2004;327 (2): 171-183.

18. Arias-Pinilla GA, Dalgleish AG, Mudan S, Bagwan I, Walker AJ, Modjtahedi H. Development of novel monoclonal antibodies against CD109 overexpressed in human pancreatic cancer. Oncotarget. 2018;9(28): 19994-20007.

19. Zhang JM, Hashimoto M, Kawai K, et al. CD109 expression in squamous cell carcinoma of the uterine cervix. Pathol Int. 2005;55(4):165-169.

20. Sato T, Murakumo Y, Hagiwara S, et al. High-level expression of CD109 is frequently detected in lung squamous cell carcinomas. Pathol Int. 2007;57(11):719-724.

21. Hagiwara S, Murakumo Y, Sato T, et al. Up-regulation of CD109 expression is associated with carcinogenesis of the squamous epithelium of the oral cavity. Cancer Science. 2008;99(10):1916-1923.

22. Hagikura M, Murakumo Y, Hasegawa M, et al. Correlation of pathological grade and tumor stage of urothelial carcinomas with CD109 expression. Pathol Int. 2010;60(11): 735-743.

23. Ohshima Y, Yajima I, Kumasaka MY, et al. CD109 expression levels in malignant melanoma. J Dermatol Sci. 2010;57(2):140-142.

24. Tao J, Li H, Li Q, Yang Y. CD109 is a potential target for triple-negative breast cancer. Tumour Biol. 2014;35(12):12083-12090.

25. Emori M, Tsukahara T, Murata K, et al. Prognostic impact of CD109 expression in myxofibrosarcoma. J Surg Oncol. 2015;111 (8):975-979.

26. Hatsuzawa Y, Yamaguchi K, Takanashi T, et al. CD109 promotes the tumorigenic ability and metastatic motility of pancreatic ductal adenocarcinoma cells. Pancreatology. 2020; 20(3):493-500.

27. Dong F, Lu C, Chen X, Guo Y, Liu J. CD109 is a novel marker for squamous cell/adenosquamous carcinomas of the gallbladder. Diagn Pathol. 2015;10:137.

28. Ozbay P, Ekinci T, Yiǧit S, et al. Investigation of prognostic significance of CD109 expression in women with vulvar squamous cell carcinoma. Onco Targets Ther. 2013;6:621-627.

29. Kim SY, Choi KU, Hwang C, et al. Prognostic Significance of CD109 Expression in Patients with Ovarian Epithelial Cancer. J Pathol Transl Med. 2019;53(4):244-252.

30. Yokoyama M, Ichinoe M, Okina S, et al. CD109, a negative regulator of TGF-β signaling, is a putative risk marker in diffuse large B-cell lymphoma. Int J Hematol. 2017; 105(5):614-622.

31. Taki T, Shiraki Y, Enomoto A, et al. CD109 regulates in vivo tumor invasion in lung adenocarcinoma through TGF-β signaling. Cancer Sci. 2020;111(12):4616-4628.

32. Hagiwara S, Murakumo Y, Mii S, et al. Processing of CD109 by furin and its role in the regulation of TGF-beta signaling. Oncogene. 2010;29(15):2181-2191.

33. Li C, Hancock MA, Sehgal P, Zhou S, Reinhardt DP, Philip A. Soluble CD109 binds TGF-β and antagonizes TGF-β signalling and responses. Biochem J. 2016;473(5):537-547.

34. Zhang JM, Murakumo Y, Hagiwara S, et al. CD109 attenuates TGF-β1 signaling and enhances EGF signaling in SK-MG-1 human glioblastoma cells. Biochem Biophys Res Commun. 2015;459(2):252-258.

35. Gan HK, Cvrljevic AN, Johns TG. The epidermal growth factor receptor variant III (EGFRvIII): where wild things are altered. FEBS J. 2013;280(21):5350-5370.

36. Arias-Pinilla GA, Dalgleish AG, Mudan S, Bagwan I, Walker AJ, Modjtahedi H. Development and application of two novel monoclonal antibodies against overexpressed CD26 and integrin α3 in human pancreatic cancer. Scientific Reports. 2020;10(1):537.

37. Khelwatty SA, Puvanenthiran S, Essapen S, Bagwan I, Seddon AM, Modjtahedi H. HER2 Expression Is Predictive of Survival in Cetuximab Treated Patients with RAS Wild Type Metastatic Colorectal Cancer. Cancers (Basel). 2021;13(4).

38. Puvanenthiran S, Essapen S, Haagsma B, et al. Co-expression and prognostic significance of the HER family members, EGFRvIII, c-MET, CD44 in patients with ovarian cancer. Oncotarget. 2018;9(28):19662 -19674.

39. Saxby AJ, Nielsen A, Scarlett CJ, et al. Assessment of HER-2 status in pancreatic adenocarcinoma: correlation of immunohistochemistry, quantitative real-time RT-PCR, and FISH with aneuploidy and survival. Am J Surg Pathol. 2005;29(9):1125-1134.

40. Komoto M, Nakata B, Amano R, et al. HER2 overexpression correlates with survival after curative resection of pancreatic cancer. Cancer Sci. 2009;100(7):1243-1247.

41. Hirakawa T, Nakata B, Amano R, et al. HER3 overexpression as an independent indicator of poor prognosis for patients with curatively resected pancreatic cancer. Oncology. 2011;81(3-4):192-198.

42. Valsecchi M, McDonald M, Brody J, et al. Epidermal growth factor receptor and insulinlike growth factor 1 receptor expression predict poor survival in pancreatic ductal adenocarcinoma. Cancer. 2012;118:3484-3493.

43. Fagman JB, Ljungman D, Falk P, et al. EGFR, but not COX-2, protein in resected pancreatic ductal adenocarcinoma is associated with poor survival. Oncol Lett. 2019;17(6):5361-5368.

44. Han SH, Ryu KH, Kwon AY. The Prognostic Impact of HER2 Genetic and Protein Expression in Pancreatic Carcinoma-HER2 Protein and Gene in Pancreatic Cancer. Diagnostics (Basel). 2021;11(4).

45. Khan T, Seddon AM, Dalgleish AG, et al. Synergistic activity of agents targeting growth factor receptors, CDKs and downstream signaling molecules in a panel of pancreatic cancer cell lines and the identification of antagonistic combinations: Implications for future clinical trials in pancreatic cancer. Oncol Rep. 2020;44(6):2581-2594.

46. Ioannou N, Dalgleish AG, Seddon AM, et al. Anti-tumour activity of afatinib, an irreversible ErbB family blocker, in human pancreatic tumour cells. Br J Cancer. 2011;105(10):1554-1562.

47. Mo XT, Leung TH, Tang HW, et al. CD109 mediates tumorigenicity and cancer aggressiveness via regulation of EGFR and STAT3 signalling in cervical squamous cell carcinoma. Br J Cancer. 2020;123(5):833-843.

48. Zhou S, Hassan A, Kungyal T, et al. CD109 Is a Critical Determinant of EGFR Expression and Signaling, and Tumorigenicity in Squamous Cell Carcinoma Cells. Cancers (Basel). 2022;14(15).

49. te Velde EA, Franke AC, van Hillegersberg R, et al. HER-family gene amplification and expression in resected pancreatic cancer. Eur J Surg Oncol. 2009;35(10):1098-1104.

50. Bittoni A, Mandolesi A, Andrikou K, et al. HER family receptor expression and prognosis in pancreatic cancer. Int J Biol Markers. 2015;30(3):e327-332.

51. Penault-Llorca F, Cayre A, Arnould L, et al. Is there an immunohistochemical technique definitively valid in epidermal growth factor receptor assessment? Oncol Rep. 2006;16(6):1173-1179.

52. Bloomston M, Bhardwaj A, Ellison EC, Frankel WL. Epidermal growth factor receptor expression in pancreatic carcinoma using tissue microarray technique. Dig Surg. 2006;23(1-2):74-79.

53. Tsiambas E, Karameris A, Lazaris AC, et al. EGFR alterations in pancreatic ductal adenocarcinoma: a chromogenic in situ hybridization analysis based on tissue microarrays. Hepatogastroenterology. 2006; 53(69):452-457.

54. Funel N, Vasile E, Del Chiaro M, et al. Correlation of basal EGFR expression with pancreatic cancer grading but not with clinical outcome after gemcitabine-based treatment. Ann Oncol. 2011;22(2):482-484.

55. Handra-Luca A, Hammel P, Sauvanet A, Lesty C, Ruszniewski P, Couvelard A. EGFR expression in pancreatic adenocarcinoma. Relationship to tumour morphology and cell adhesion proteins. J Clin Pathol. 2014;67 (4):295-300.

56. Griffin MC, Robinson RA, Trask DK. Validation of tissue microarrays using p53 immunohistochemical studies of squamous cell carcinoma of the larynx. Mod Pathol. 2003;16(12):1181-1188.

57. Cros J, Raffenne J, Couvelard A, Pote N. Tumor Heterogeneity in Pancreatic Adenocarcinoma. Pathobiology. 2018;85(1-2) :64-71.

58. Juiz NA, Iovanna J, Dusetti N. Pancreatic Cancer Heterogeneity Can Be Explained Beyond the Genome. Front Oncol. 2019; 9:246.

59. Tobita K, Kijima H, Dowaki S, et al. Epidermal growth factor receptor expression in human pancreatic cancer: Significance for liver metastasis. Int J Mol Med. 2003;11 (3):305-309.

60. Ueda S, Ogata S, Tsuda H, et al. The correlation between cytoplasmic overexpression of epidermal growth factor receptor and tumor aggressiveness: poor prognosis in patients with pancreatic ductal adenocarcinoma. Pancreas. 2004;29(1):e1-8.

61. Chadha KS, Khoury T, Yu J, et al. Activated Akt and Erk expression and survival after surgery in pancreatic carcinoma. Ann Surg Oncol. 2006;13(7):933-939.

62. Ueda S, Hatsuse K, Tsuda H, et al. Potential crosstalk between insulin-like growth factor receptor type 1 and epidermal growth factor receptor in progression and metastasis of pancreatic cancer. Mod Pathol. 2006;19(6):788-796.

63. Dancer J, Takei H, Ro JY, Lowery-Nordberg M. Coexpression of EGFR and HER-2 in pancreatic ductal adenocarcinoma: a comparative study using immunohistochemistry correlated with gene amplification by fluorescencent in situ hybridization. Oncol Rep. 2007;18(1):151-155.

64. Pryczynicz A, Guzinska-Ustymowicz K, Kemona A, Czyzewska J. Expression of EGF and EGFR strongly correlates with metastasis of pancreatic ductal carcinoma. Anticancer Res. 2008;28(2B):1399-1404.

65. Komoto M, Nakata B, Nishii T, et al. In vitro and in vivo evidence that a combination of lapatinib plus S-1 is a promising treatment for pancreatic cancer. Cancer Sci. 2010; 101(2):468-473.

66. Lozano-Leon A, Perez-Quintela BV, Iglesias-Garcia J, et al. Clinical relevance of epidermal growth factor receptor (EGFR) alterations in human pancreatic tumors. Oncol Rep. 2011;26(2):315-320.

67. Walsh N, Kennedy S, Larkin A, et al. EGFR and HER2 inhibition in pancreatic cancer. Invest New Drugs. 2013;31(3):558-566.

68. Boeck S, Jung A, Laubender RP, et al. EGFR pathway biomarkers in erlotinib-treated patients with advanced pancreatic cancer: translational results from the randomised, crossover phase 3 trial AIO-PK0104. Br J Cancer. 2013;108(2):469-476.

69. Wu H, Zhu L, Zhang H, et al. Coexpression of EGFR and CXCR4 predicts poor prognosis in resected pancreatic ductal adenocarcinoma. PLoS One. 2015;10(2): e0116803.

70. Park SJ, Gu MJ, Lee DS, Yun SS, Kim HJ, Choi JH. EGFR expression in pancreatic intraepithelial neoplasia and ductal adenocarcinoma. Int J Clin Exp Pathol. 2015;8 (7):8298-8304.

71. Guo M, Luo G, Liu C, et al. The Prognostic and Predictive Role of Epidermal Growth Factor Receptor in Surgical Resected Pancreatic Cancer. Int J Mol Sci. 2016;17(7).

72. de Geus SW, Boogerd LS, Swijnenburg RJ, et al. Selecting Tumor-Specific Molecular Targets in Pancreatic Adenocarcinoma: Paving the Way for Image-Guided Pancreatic Surgery. Mol Imaging Biol. 2016;18(6):807-819.

73. Wang L, Wu H, Wang L, et al. Expression of amphiregulin predicts poor outcome in patients with pancreatic ductal adenocarcinoma. Diagn Pathol. 2016;11(1):60.

74. Sun J, Chen L, Dong M. MiR-338-5p Inhibits EGF-Induced EMT in Pancreatic Cancer Cells by Targeting EGFR/ERK Signaling. Front Oncol. 2021;11:616481.

75. Harder J, Ihorst G, Heinemann V, et al. Multicentre phase II trial of trastuzumab and capecitabine in patients with HER2 overexpressing metastatic pancreatic cancer. Br J Cancer. 2012;106(6):1033-1038.

76. Chou A, Waddell N, Cowley MJ, et al. Clinical and molecular characterization of HER2 amplified-pancreatic cancer. Genome Med. 2013;5(8):78.

77. Koperek O, Aumayr K, Schindl M, et al. Phosphorylation of STAT3 correlates with HER2 status, but not with survival in pancreatic ductal adenocarcinoma. Apmis. 2014;122(6):476-481.

78. Thomas G, Chardes T, Gaborit N, et al. HER3 as biomarker and therapeutic target in pancreatic cancer: new insights in pertuzumab therapy in preclinical models. Oncotarget. 2014;5(16):7138-7148.

79. Yan M, Schwaederle M, Arguello D, Millis SZ, Gatalica Z, Kurzrock R. HER2 expression status in diverse cancers: review of results from 37,992 patients. Cancer Metastasis Rev. 2015;34(1):157-164.

80. Ata A, Polat A, Serinsoz E, Sungur MA, Arican A. Prognostic value of increased HER2 expression in cancers of pancreas and biliary tree. Pathol Oncol Res. 2015;21(3):831-838.

81. Shibata W, Kinoshita H, Hikiba Y, et al. Overexpression of HER2 in the pancreas promotes development of intraductal papillary mucinous neoplasms in mice. Sci Rep. 2018;8(1):6150.

82. Adachi K, Sakurai Y, Ichinoe M, et al. CD109 expression in tumor cells and stroma correlates with progression and prognosis in pancreatic cancer. Virchows Arch. 2022;480(4):819-829.

83. Koh H, Lee H, Kim D. Usefulness of CD109 expression as a prognostic biomarker in patients with cancer: A systematic review and meta-analysis. Medicine. 2021;100 : e25006.

84. Haun RS, Fan CY, Mackintosh SG, Zhao H, Tackett AJ. CD109 Overexpression in Pancreatic Cancer Identified by Cell-Surface Glycoprotein Capture. J Proteomics Bioinform. 2014;Suppl 10:S10003.

85. Modi S, Jacot W, Yamashita T, Sohn J, Vidal M, Tokunaga E, Tsurutani J, Ueno NT, Prat A, Chae YS, Lee KS, Niikura N, Park YH, Xu B, Wang X, Gil-Gil M, Li W, Pierga JY, Im SA, Moore HCF, Rugo HS, Yerushalmi R, Zagouri F, Gombos A, Kim SB, Liu Q, Luo T, Saura C, Schmid P, Sun T, Gambhire D, Yung L, Wang Y, Singh J, Vitazka P, Meinhardt G, Harbeck N, Cameron DA; DESTINY-Breast04 Trial Investigators. Trastuzumab Deruxtecan in Previously Treated HER2-Low Advanced Breast Cancer. N Engl J Med. 2022 Jul 7;387(1):9-20. doi: 10.1056/NEJMoa2203690. Epub 2022 Jun 5. PMID: 35665782; PMCID: PMC10561652.

86. Corti C, Giugliano F, Nicolò E, Tarantino P, Criscitiello C, Curigliano G. HER2-Low Breast Cancer: a New Subtype? Curr Treat Options Oncol. 2023 May;24(5):468-478. doi: 10.1007/s11864-023-01068-1. Epub 2023 Mar 27. PMID: 36971965.

87. Cardin DB, Goff LW, Chan E, et al. Dual Src and EGFR inhibition in combination with gemcitabine in advanced pancreatic cancer: phase I results : A phase I clinical trial. Invest New Drugs. 2018;36(3):442-450.

88. Nagaraj NS, Washington MK, Merchant NB. Combined blockade of Src kinase and epidermal growth factor receptor with gemcitabine overcomes STAT3-mediated resistance of inhibition of pancreatic tumor growth. Clin Cancer Res. 2011;17(3):483-493.