DOI QR코드

DOI QR Code

Objective Quantitation of EGFR Protein Levels using Quantitative Dot Blot Method for the Prognosis of Gastric Cancer Patients

  • Xin, Lei (Department of Gastrointestinal Surgery, Yantaishan Hospital) ;
  • Tang, Fangrong (Yantai Quanticision Diagnostics, Inc. (Division of Quanticision Diagnostics, Inc. of USA)) ;
  • Song, Bo (Department of Gastroenterology, Yantaishan Hospital) ;
  • Yang, Maozhou (Yantai Quanticision Diagnostics, Inc. (Division of Quanticision Diagnostics, Inc. of USA)) ;
  • Zhang, Jiandi (Yantai Quanticision Diagnostics, Inc. (Division of Quanticision Diagnostics, Inc. of USA))
  • Received : 2021.08.03
  • Accepted : 2021.10.28
  • Published : 2021.12.31

Abstract

Purpose: An underlying factor for the failure of several clinical trials of anti-epidermal growth factor receptor (EGFR) therapies is the lack of an effective method to identify patients who overexpress EGFR protein. The quantitative dot blot method (QDB) was used to measure EGFR protein levels objectively, absolutely, and quantitatively. Its feasibility was evaluated for the prognosis of overall survival (OS) of patients with gastric cancer. Materials and Methods: Slices of 2×5 ㎛ from formalin-fixed paraffin-embedded gastric cancer specimens were used to extract total tissue lysates for QDB measurement. Absolutely quantitated EGFR protein levels were used for the Kaplan-Meier OS analysis. Results: EGFR protein levels ranged from 0 to 772.6 pmol/g (n=246) for all gastric cancer patients. A poor correlation was observed between quantitated EGFR levels and immunohistochemistry scores with ρ=0.024 and P=0.717 in Spearman's correlation analysis. EGFR was identified as an independent negative prognostic biomarker for gastric cancer patients only through absolute quantitation, with a hazard ratio of 1.92 (95% confidence interval, 1.05-3.53; P=0.034) in multivariate Cox regression OS analysis. A cutoff of 208 pmol/g was proposed to stratify patients with a 3-year survival probability of 44% for patients with EGFR levels above the cutoff versus 68% for those below the cutoff based on Kaplan-Meier OS analysis (log rank test, P=0.002). Conclusions: A QDB-based assay was developed for gastric cancer specimens to measure EGFR protein levels absolutely, quantitatively, and objectively. This assay should facilitate clinical trials aimed at evaluation of anti-EGFR therapies retrospectively and prospectively for gastric cancer.

Keywords

Acknowledgement

The authors wish to thank Ms. Wenfeng Zhang, Ms. Yan Lyu, and Ms. Yunyun Zhang for their excellent technical support.

References

  1. Diaz-Serrano A, Sanchez-Torre A, Paz-Ares L. Necitumumab for the treatment of advanced non-small-cell lung cancer. Future Oncol 2019;15:705-716. https://doi.org/10.2217/fon-2018-0594
  2. Pirker R, Pereira JR, von Pawel J, Krzakowski M, Ramlau R, Park K, et al. EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: analysis of data from the phase 3 FLEX study. Lancet Oncol 2012;13:33-42. https://doi.org/10.1016/S1470-2045(11)70318-7
  3. Paz-Ares L, Mezger J, Ciuleanu TE, Fischer JR, von Pawel J, Provencio M, et al. Necitumumab plus pemetrexed and cisplatin as first-line therapy in patients with stage IV non-squamous non-small-cell lung cancer (INSPIRE): an open-label, randomised, controlled phase 3 study. Lancet Oncol 2015;16:328-337. https://doi.org/10.1016/S1470-2045(15)70046-X
  4. Apicella M, Corso S, Giordano S. Targeted therapies for gastric cancer: failures and hopes from clinical trials. Oncotarget 2017;8:57654-57669. https://doi.org/10.18632/oncotarget.14825
  5. Li K, Li J. Current molecular targeted therapy in advanced gastric cancer: a comprehensive review of therapeutic mechanism, clinical trials, and practical application. Gastroenterol Res Pract 2016;2016:4105615.
  6. Lynch TJ, Patel T, Dreisbach L, McCleod M, Heim WJ, Hermann RC, et al. Cetuximab and first-line taxane/carboplatin chemotherapy in advanced non-small-cell lung cancer: results of the randomized multicenter phase III trial BMS099. J Clin Oncol 2010;28:911-917. https://doi.org/10.1200/JCO.2009.21.9618
  7. Chang S, Hur JY, Choi YL, Lee CH, Kim WS. Current status and future perspectives of liquid biopsy in non-small cell lung cancer. J Pathol Transl Med 2020;54:204-212. https://doi.org/10.4132/jptm.2020.02.27
  8. Pirker R, Pereira JR, Szczesna A, von Pawel J, Krzakowski M, Ramlau R, et al. Cetuximab plus chemotherapy in patients with advanced non-small-cell lung cancer (FLEX): an open-label randomised phase III trial. Lancet 2009;373:1525-1531. https://doi.org/10.1016/S0140-6736(09)60569-9
  9. Thatcher N, Hirsch FR, Luft AV, Szczesna A, Ciuleanu TE, Dediu M, et al. Necitumumab plus gemcitabine and cisplatin versus gemcitabine and cisplatin alone as first-line therapy in patients with stage IV squamous non-small-cell lung cancer (SQUIRE): an open-label, randomised, controlled phase 3 trial. Lancet Oncol 2015;16:763-774. https://doi.org/10.1016/S1470-2045(15)00021-2
  10. Hirsch FR, Herbst RS, Olsen C, Chansky K, Crowley J, Kelly K, et al. Increased EGFR gene copy number detected by fluorescent in situ hybridization predicts outcome in non-small-cell lung cancer patients treated with cetuximab and chemotherapy. J Clin Oncol 2008;26:3351-3357. https://doi.org/10.1200/JCO.2007.14.0111
  11. Lordick F, Kang YK, Chung HC, Salman P, Oh SC, Bodoky G, et al. Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer (EXPAND): a randomised, open-label phase 3 trial. Lancet Oncol 2013;14:490-499. https://doi.org/10.1016/S1470-2045(13)70102-5
  12. Burtness B, Goldwasser MA, Flood W, Mattar B, Forastiere AA; Eastern Cooperative Oncology Group. Phase III randomized trial of cisplatin plus placebo compared with cisplatin plus cetuximab in metastatic/recurrent head and neck cancer: an Eastern Cooperative Oncology Group study. J Clin Oncol 2005;23:8646-8654. https://doi.org/10.1200/JCO.2005.02.4646
  13. Lordick F, Luber B, Lorenzen S, Hegewisch-Becker S, Folprecht G, Woll E, et al. Cetuximab plus oxaliplatin/leucovorin/5-fluorouracil in first-line metastatic gastric cancer: a phase II study of the Arbeitsgemeinschaft Internistische Onkologie (AIO). Br J Cancer 2010;102:500-505. https://doi.org/10.1038/sj.bjc.6605521
  14. Luber B, Deplazes J, Keller G, Walch A, Rauser S, Eichmann M, et al. Biomarker analysis of cetuximab plus oxaliplatin/leucovorin/5-fluorouracil in first-line metastatic gastric and oesophago-gastric junction cancer: results from a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie (AIO). BMC Cancer 2011;11:509. https://doi.org/10.1186/1471-2407-11-509
  15. Tol J, Dijkstra JR, Klomp M, Teerenstra S, Dommerholt M, Vink-Borger ME, et al. Markers for EGFR pathway activation as predictor of outcome in metastatic colorectal cancer patients treated with or without cetuximab. Eur J Cancer 2010;46:1997-2009. https://doi.org/10.1016/j.ejca.2010.03.036
  16. Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen RB, et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 2006;354:567-578. https://doi.org/10.1056/NEJMoa053422
  17. Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med 2008;359:1116-1127. https://doi.org/10.1056/NEJMoa0802656
  18. Chae YK, Arya A, Chiec L, Shah H, Rosenberg A, Patel S, et al. Challenges and future of biomarker tests in the era of precision oncology: Can we rely on immunohistochemistry (IHC) or fluorescence in situ hybridization (FISH) to select the optimal patients for matched therapy? Oncotarget 2017;8:100863-100898. https://doi.org/10.18632/oncotarget.19809
  19. Kim ST, Banks KC, Lee SH, Kim K, Park JO, Park SH, et al. Prospective feasibility study for using cell-free circulating tumor DNA-guided therapy in refractory metastatic solid cancers: an interim analysis. JCO Precis Oncol 2017;1:1-15.
  20. Nakamura Y, Shitara K, Lee J. The right treatment of the right patient: integrating genetic profiling into clinical decision making in advanced gastric cancer in Asia. Am Soc Clin Oncol Educ Book 2021;41:e166-e173. https://doi.org/10.1200/EDBK_321247
  21. Gown AM. Diagnostic Immunohistochemistry: What can go wrong and how to prevent it. Arch Pathol Lab Med 2016;140:893-898. https://doi.org/10.5858/arpa.2016-0119-RA
  22. Han SW, Oh DY, Im SA, Park SR, Lee KW, Song HS, et al. Phase II study and biomarker analysis of cetuximab combined with modified FOLFOX6 in advanced gastric cancer. Br J Cancer 2009;100:298-304. https://doi.org/10.1038/sj.bjc.6604861
  23. Licitra L, Storkel S, Kerr KM, Van Cutsem E, Pirker R, Hirsch FR, et al. Predictive value of epidermal growth factor receptor expression for first-line chemotherapy plus cetuximab in patients with head and neck and colorectal cancer: analysis of data from the EXTREME and CRYSTAL studies. Eur J Cancer 2013;49:1161-1168. https://doi.org/10.1016/j.ejca.2012.11.018
  24. Rudbeck L. Adding quality to your qualitative IHC. MLO Med Lab Obs 2015;47:18-19, 21.
  25. Nielsen TO, Leung SCY, Rimm DL, Dodson A, Acs B, Badve S, et al. Assessment of Ki67 in breast cancer: updated recommendations from the international Ki67 in breast cancer working group. J Natl Cancer Inst 2021;113:808-819. https://doi.org/10.1093/jnci/djaa201
  26. Tian G, Tang F, Yang C, Zhang W, Bergquist J, Wang B, et al. Quantitative dot blot analysis (QDB), a versatile high throughput immunoblot method. Oncotarget 2017;8:58553-58562. https://doi.org/10.18632/oncotarget.17236
  27. Zhang W, Yu G, Zhang Y, Tang F, Lv J, Tian G, et al. Quantitative dot blot (QDB) as a universal platform for absolute quantification of tissue biomarkers. Anal Biochem 2019;576:42-47. https://doi.org/10.1016/j.ab.2019.04.003
  28. Yu G, Zhang W, Zhang Y, Lv J, Wu S, Sui X, et al. Developing a routine lab test for absolute quantification of HER2 in FFPE breast cancer tissues using quantitative dot blot (QDB) method. Sci Rep 2020;10:12502. https://doi.org/10.1038/s41598-020-69471-4
  29. Hao J, Lyu Y, Zou J, Zhang Y, Xie S, Jing L, et al. Improving prognosis of surrogate assay for breast cancer patients by absolute quantitation of Ki67 protein levels using quantitative dot blot (QDB) method. Front Oncol 2021;11:737781. https://doi.org/10.3389/fonc.2021.737781
  30. Sun G, Yang L, Dong C, Ma B, Shan M, Zheng Y, et al. Expression and characterization analysis of EGFR, PI3K and its phosphorylated protein in different subtypes of breast cancer. Chin J Endocr Surg 2014;8:207-212.
  31. Yarden Y, Pines G. The ERBB network: at last, cancer therapy meets systems biology. Nat Rev Cancer 2012;12:553-563. https://doi.org/10.1038/nrc3309
  32. Higaki E, Kuwata T, Nagatsuma AK, Nishida Y, Kinoshita T, Aizawa M, et al. Gene copy number gain of EGFR is a poor prognostic biomarker in gastric cancer: evaluation of 855 patients with bright-field dual in situ hybridization (DISH) method. Gastric Cancer 2016;19:63-73. https://doi.org/10.1007/s10120-014-0449-9
  33. Kim MA, Lee HS, Lee HE, Jeon YK, Yang HK, Kim WH. EGFR in gastric carcinomas: prognostic significance of protein overexpression and high gene copy number. Histopathology 2008;52:738-746. https://doi.org/10.1111/j.1365-2559.2008.03021.x
  34. Terashima M, Kitada K, Ochiai A, Ichikawa W, Kurahashi I, Sakuramoto S, et al. Impact of expression of human epidermal growth factor receptors EGFR and ERBB2 on survival in stage II/III gastric cancer. Clin Cancer Res 2012;18:5992-6000. https://doi.org/10.1158/1078-0432.CCR-12-1318
  35. Birkman EM, Algars A, Lintunen M, Ristamaki R, Sundstrom J, Carpen O. EGFR gene amplification is relatively common and associates with outcome in intestinal adenocarcinoma of the stomach, gastro-oesophageal junction and distal oesophagus. BMC Cancer 2016;16:406. https://doi.org/10.1186/s12885-016-2456-1
  36. Nagatsuma AK, Aizawa M, Kuwata T, Doi T, Ohtsu A, Fujii H, et al. Expression profiles of HER2, EGFR, MET and FGFR2 in a large cohort of patients with gastric adenocarcinoma. Gastric Cancer 2015;18:227-238. https://doi.org/10.1007/s10120-014-0360-4
  37. Zhang F, Yang X, Li L, Sun L, Wang BO, Yu X. Epidermal growth factor receptor expression and gene copy number analysis in gastric carcinoma samples from Chinese patients. Oncol Lett 2016;11:173-181. https://doi.org/10.3892/ol.2015.3875
  38. Ruschoff J, Hanna W, Bilous M, Hofmann M, Osamura RY, Penault-Llorca F, et al. HER2 testing in gastric cancer: a practical approach. Mod Pathol 2012;25:637-650. https://doi.org/10.1038/modpathol.2011.198
  39. Stahl P, Seeschaaf C, Lebok P, Kutup A, Bockhorn M, Izbicki JR, et al. Heterogeneity of amplification of HER2, EGFR, CCND1 and MYC in gastric cancer. BMC Gastroenterol 2015;15:7. https://doi.org/10.1186/s12876-015-0231-4
  40. Fuse N, Kuboki Y, Kuwata T, Nishina T, Kadowaki S, Shinozaki E, et al. Prognostic impact of HER2, EGFR, and c-MET status on overall survival of advanced gastric cancer patients. Gastric Cancer 2016;19:183-191. https://doi.org/10.1007/s10120-015-0471-6
  41. Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst 2009;101:1446-1452. https://doi.org/10.1093/jnci/djp335
  42. Zhang J, Yang M. Developing a growing cancer profile database based on quantitative analysis of protein biomarkers in formalin-fixed paraffin-embedded specimens. Future Oncol 2020;16:2471-2474. https://doi.org/10.2217/fon-2020-0480
  43. Genova C, Socinski MA, Hozak RR, Mi G, Kurek R, Shahidi J, et al. EGFR gene copy number by FISH may predict outcome of necitumumab in squamous lung carcinomas: analysis from the SQUIRE study. J Thorac Oncol 2018;13:228-236. https://doi.org/10.1016/j.jtho.2017.11.109
  44. Herbst RS, Redman MW, Kim ES, Semrad TJ, Bazhenova L, Masters G, et al. Cetuximab plus carboplatin and paclitaxel with or without bevacizumab versus carboplatin and paclitaxel with or without bevacizumab in advanced NSCLC (SWOG S0819): a randomised, phase 3 study. Lancet Oncol 2018;19:101-114. https://doi.org/10.1016/S1470-2045(17)30694-0
  45. Hembrough T, Thyparambil S, Liao WL, Darfler MM, Abdo J, Bengali KM, et al. Selected reaction monitoring (SRM) analysis of epidermal growth factor receptor (EGFR) in formalin fixed tumor tissue. Clin Proteomics 2012;9:5. https://doi.org/10.1186/1559-0275-9-5
  46. Lievre A, Bachet JB, Le Corre D, Boige V, Landi B, Emile JF, et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 2006;66:3992-3995. https://doi.org/10.1158/0008-5472.CAN-06-0191
  47. Queiros P, Pinheiro H, Carvalho J, Oliveira P, Gullo I, Carneiro F, et al. KRAS mutations in microsatellite instable gastric tumours: impact of targeted treatment and intratumoural heterogeneity. Virchows Arch 2015;467:383-392. https://doi.org/10.1007/s00428-015-1823-7
  48. Park JE, Jin MH, Hur M, Nam AR, Bang JH, Won J, et al. GC1118, a novel anti-EGFR antibody, has potent KRAS mutation-independent antitumor activity compared with cetuximab in gastric cancer. Gastric Cancer 2019;22:932-940. https://doi.org/10.1007/s10120-019-00943-x
  49. Oh DY, Lee KW, Han SW, Kim JW, Shin JW, Jo SJ, et al. A first-in-human phase I study of GC1118, a novel anti-epidermal growth factor receptor antibody, in patients with advanced solid tumors. Oncologist 2019;24:1037-e636. https://doi.org/10.1634/theoncologist.2019-0294