Skip to main content
Log in

Clinical and in vitro studies of the correlation between MGMT and the effect of streptozocin in pancreatic NET

  • Original Article
  • Published:
Cancer Chemotherapy and Pharmacology Aims and scope Submit manuscript

Abstract

Purpose

This study aimed to determine the correlation between DNA repair enzyme O6-methylguanine DNA methyltransferase (MGMT) status and the response to streptozocin in advanced well-differentiated pancreatic neuroendocrine tumors (WD panNETs).

Methods

To test the hypothesis that MGMT deficiency was required for an alkylating drug response, we retrospectively reviewed the response of 13 patients with WD panNETs to alkylating agents in relation to MGMT status. We also studied MGMT expression in streptozocin resistance using panNET cell lines.

Results

The cohort included 54% of patients with and 46% without MGMT expression. Among these, 83.3% (5/6) of MGMT-negative cases showed a partial response to streptozocin. In contrast, only 14.2% (1/7) of MGMT-positive cases showed a partial response (P = 0.013). Induced expression of MGMT in BON1 cells (a panNET cell line with undetectable endogenous MGMT) produced streptozocin resistance. Knockdown of MGMT in QGP1 cells, which express MGMT endogenously, did not alter the response to streptozocin.

Conclusions

We observed a relationship between MGMT status and streptozocin response in both patients and cell culture. Despite limited cases examined, high concordance of negative expression of MGMT and response to streptozocin treatment suggest that MGMT expression can be a potential biomarker for this treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, Shih T, Yao JC (2017) Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol 3:1335–1342. https://doi.org/10.1001/jamaoncol.2017.0589

    Article  PubMed  PubMed Central  Google Scholar 

  2. Panzuto F, Boninsegna L, Fazio N, Campana D, Pia Brizzi M, Capurso G, Scarpa A, De Braud F, Dogliotti L, Tomassetti P, Delle Fave G, Falconi M (2011) Metastatic and locally advanced pancreatic endocrine carcinomas: analysis of factors associated with disease progression. J Clin Oncol 29:2372–2377. https://doi.org/10.1200/jco.2010.33.0688

    Article  PubMed  Google Scholar 

  3. Caplin ME, Pavel M, Cwikla JB, Phan AT, Raderer M, Sedlackova E, Cadiot G, Wolin EM, Capdevila J, Wall L, Rindi G, Langley A, Martinez S, Blumberg J, Ruszniewski P (2014) Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med 371:224–233. https://doi.org/10.1056/NEJMoa1316158

    Article  CAS  PubMed  Google Scholar 

  4. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, Hobday TJ, Okusaka T, Capdevila J, de Vries EG, Tomassetti P, Pavel ME, Hoosen S, Haas T, Lincy J, Lebwohl D, Oberg K (2011) Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 364:514–523. https://doi.org/10.1056/NEJMoa1009290

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, Valle J, Metrakos P, Smith D, Vinik A, Chen JS, Horsch D, Hammel P, Wiedenmann B, Van Cutsem E, Patyna S, Lu DR, Blanckmeister C, Chao R, Ruszniewski P (2011) Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 364:501–513. https://doi.org/10.1056/NEJMoa1003825

    Article  CAS  PubMed  Google Scholar 

  6. Rinke A, Muller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M, Mayer C, Aminossadati B, Pape UF, Blaker M, Harder J, Arnold C, Gress T, Arnold R (2009) Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol 27:4656–4663. https://doi.org/10.1200/jco.2009.22.8510

    Article  CAS  PubMed  Google Scholar 

  7. Pavel M, Baudin E, Couvelard A, Krenning E, Oberg K, Steinmuller T, Anlauf M, Wiedenmann B, Salazar R (2012) ENETS Consensus Guidelines for the management of patients with liver and other distant metastases from neuroendocrine neoplasms of foregut, midgut, hindgut, and unknown primary. Neuroendocrinology 95:157–176. https://doi.org/10.1159/000335597

    Article  CAS  PubMed  Google Scholar 

  8. Ito T, Hijioka S, Masui T, Kasajima A, Nakamoto Y, Kobayashi N, Komoto I, Hijioka M, Lee L, Igarashi H, Jensen RT, Imamura M (2017) Advances in the diagnosis and treatment of pancreatic neuroendocrine neoplasms in Japan. J Gastroenterol 52:9–18. https://doi.org/10.1007/s00535-016-1250-9

    Article  CAS  PubMed  Google Scholar 

  9. Clewemar Antonodimitrakis P, Sundin A, Wassberg C, Granberg D, Skogseid B, Eriksson B (2016) Streptozocin and 5-fluorouracil for the treatment of pancreatic neuroendocrine tumors: efficacy, prognostic factors and toxicity. Neuroendocrinology 103:345–353. https://doi.org/10.1159/000439086

    Article  CAS  PubMed  Google Scholar 

  10. Krug S, Boch M, Daniel H, Nimphius W, Muller D, Michl P, Rinke A, Gress TM (2015) Streptozocin-based chemotherapy in patients with advanced neuroendocrine neoplasms–predictive and prognostic markers for treatment stratification. PLoS One 10:e0143822. https://doi.org/10.1371/journal.pone.0143822

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Dilz LM, Denecke T, Steffen IG, Prasad V, von Weikersthal LF, Pape UF, Wiedenmann B, Pavel M (2015) Streptozocin/5-fluorouracil chemotherapy is associated with durable response in patients with advanced pancreatic neuroendocrine tumours. Eur J Cancer (Oxf) 51:1253–1262. https://doi.org/10.1016/j.ejca.2015.04.005

    Article  CAS  Google Scholar 

  12. Aoki T, Kokudo N, Komoto I, Takaori K, Kimura W, Sano K, Takamoto T, Hashimoto T, Okusaka T, Morizane C, Ito T, Imamura M (2015) Streptozocin chemotherapy for advanced/metastatic well-differentiated neuroendocrine tumors: an analysis of a multi-center survey in Japan. J Gastroenterol 50:769–775. https://doi.org/10.1007/s00535-014-1006-3

    Article  CAS  PubMed  Google Scholar 

  13. Strosberg JR, Fine RL, Choi J, Nasir A, Coppola D, Chen DT, Helm J, Kvols L (2011) First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer 117:268–275. https://doi.org/10.1002/cncr.25425

    Article  CAS  PubMed  Google Scholar 

  14. Liu L, Gerson SL (2006) Targeted modulation of MGMT: clinical implications. Clin Cancer Res 12:328–331. https://doi.org/10.1158/1078-0432.ccr-05-2543

    Article  CAS  PubMed  Google Scholar 

  15. Zhang J, Stevens MF, Bradshaw TD (2012) Temozolomide: mechanisms of action, repair and resistance. Curr Mol Pharmacol 5:102–114

    Article  CAS  PubMed  Google Scholar 

  16. Christmann M, Verbeek B, Roos WP, Kaina B (2011) O(6)-Methylguanine-DNA methyltransferase (MGMT) in normal tissues and tumors: enzyme activity, promoter methylation and immunohistochemistry. Biochim Biophys Acta 1816:179–190. https://doi.org/10.1016/j.bbcan.2011.06.002

    Article  CAS  PubMed  Google Scholar 

  17. Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG (1999) Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res 59:793–797

    CAS  PubMed  Google Scholar 

  18. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, Bromberg JE, Hau P, Mirimanoff RO, Cairncross JG, Janzer RC, Stupp R (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 352:997–1003. https://doi.org/10.1056/NEJMoa043331

    Article  CAS  PubMed  Google Scholar 

  19. Weller M, Stupp R, Reifenberger G, Brandes AA, van den Bent MJ, Wick W, Hegi ME (2010) MGMT promoter methylation in malignant gliomas: ready for personalized medicine? Nat Rev Neurol 6:39–51. https://doi.org/10.1038/nrneurol.2009.197

    Article  CAS  PubMed  Google Scholar 

  20. Gorlia T, van den Bent MJ, Hegi ME, Mirimanoff RO, Weller M, Cairncross JG, Eisenhauer E, Belanger K, Brandes AA, Allgeier A, Lacombe D, Stupp R (2008) Nomograms for predicting survival of patients with newly diagnosed glioblastoma: prognostic factor analysis of EORTC and NCIC trial 26981-22981/CE.3. Lancet Oncol 9:29–38. https://doi.org/10.1016/S1470-2045(07)70384-4

    Article  PubMed  Google Scholar 

  21. Karayan-Tapon L, Quillien V, Guilhot J, Wager M, Fromont G, Saikali S, Etcheverry A, Hamlat A, Loussouarn D, Campion L, Campone M, Vallette FM, Gratas-Rabbia-Re C (2010) Prognostic value of O6-methylguanine-DNA methyltransferase status in glioblastoma patients, assessed by five different methods. J Neurooncol 97:311–322. https://doi.org/10.1007/s11060-009-0031-1

    Article  CAS  PubMed  Google Scholar 

  22. Raj N, Klimstra DS, Horvat N, Zhang L, Chou JF, Capanu M, Basturk O, Do RKG, Allen PJ, Reidy-Lagunes D (2017) O6-Methylguanine DNA methyltransferase status does not predict response or resistance to alkylating agents in well-differentiated pancreatic neuroendocrine tumors. Pancreas 46:758–763. https://doi.org/10.1097/mpa.0000000000000842

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Girot P, Dumars C, Mosnier JF, Muzellec L, Senellart H, Foubert F, Caroli-Bosc FX, Cauchin E, Regenet N, Matysiak-Budnik T, Touchefeu Y (2017) Short article: Evaluation of O6-methylguanine-DNA methyltransferase as a predicting factor of response to temozolomide-based chemotherapy in well-differentiated metastatic pancreatic neuroendocrine tumors. Eur J Gastroenterol Hepatol 29:826–830. https://doi.org/10.1097/meg.0000000000000874

    Article  CAS  PubMed  Google Scholar 

  24. Cives M, Ghayouri M, Morse B, Brelsford M, Black M, Rizzo A, Meeker A, Strosberg J (2016) Analysis of potential response predictors to capecitabine/temozolomide in metastatic pancreatic neuroendocrine tumors. Endocr Relat Cancer 23:759–767. https://doi.org/10.1530/erc-16-0147

    Article  CAS  PubMed  Google Scholar 

  25. Walter T, van Brakel B, Vercherat C, Hervieu V, Forestier J, Chayvialle JA, Molin Y, Lombard-Bohas C, Joly MO, Scoazec JY (2015) O6-Methylguanine-DNA methyltransferase status in neuroendocrine tumours: prognostic relevance and association with response to alkylating agents. Br J Cancer 112:523–531. https://doi.org/10.1038/bjc.2014.660

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Schmitt AM, Pavel M, Rudolph T, Dawson H, Blank A, Komminoth P, Vassella E, Perren A (2014) Prognostic and predictive roles of MGMT protein expression and promoter methylation in sporadic pancreatic neuroendocrine neoplasms. Neuroendocrinology 100:35–44. https://doi.org/10.1159/000365514

    Article  CAS  PubMed  Google Scholar 

  27. Kulke MH, Hornick JL, Frauenhoffer C, Hooshmand S, Ryan DP, Enzinger PC, Meyerhardt JA, Clark JW, Stuart K, Fuchs CS, Redston MS (2009) O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clin Cancer Res 15:338–345. https://doi.org/10.1158/1078-0432.ccr-08-1476

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ekeblad S, Sundin A, Janson ET, Welin S, Granberg D, Kindmark H, Dunder K, Kozlovacki G, Orlefors H, Sigurd M, Oberg K, Eriksson B, Skogseid B (2007) Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clin Cancer Res 13:2986–2991. https://doi.org/10.1158/1078-0432.ccr-06-2053

    Article  CAS  PubMed  Google Scholar 

  29. Cros J, Hentic O, Rebours V, Zappa M, Gille N, Theou-Anton N, Vernerey D, Maire F, Levy P, Bedossa P, Paradis V, Hammel P, Ruszniewski P, Couvelard A (2016) MGMT expression predicts response to temozolomide in pancreatic neuroendocrine tumors. Endocr Relat Cancer 23:625–633. https://doi.org/10.1530/erc-16-0117

    Article  CAS  PubMed  Google Scholar 

  30. Owen DH, Alexander AJ, Konda B, Wei L, Hemminger JA, Schmidt CR, Abdel-Misih SRZ, Dillhoff ME, Sipos JA, Kirschner LS, Shah MH (2017) Combination therapy with capecitabine and temozolomide in patients with low and high grade neuroendocrine tumors, with an exploratory analysis of O(6)-methylguanine DNA methyltransferase as a biomarker for response. Oncotarget 8:104046–104056. https://doi.org/10.18632/oncotarget.22001

    Article  PubMed  PubMed Central  Google Scholar 

  31. Campana D, Walter T, Pusceddu S, Gelsomino F, Graillot E, Prinzi N, Spallanzani A, Fiorentino M, Barritault M, Dall’Olio F, Brighi N, Biasco G (2018) Correlation between MGMT promoter methylation and response to temozolomide-based therapy in neuroendocrine neoplasms: an observational retrospective multicenter study. Endocrine 60:490–498. https://doi.org/10.1007/s12020-017-1474-3

    Article  CAS  PubMed  Google Scholar 

  32. Townsend CM Jr, Ishizuka J, Thompson JC (1993) Studies of growth regulation in a neuroendocrine cell line. Acta Oncol (Stockh) 32:125–130

    Article  Google Scholar 

  33. Fine RL, Gulati AP, Krantz BA, Moss RA, Schreibman S, Tsushima DA, Mowatt KB, Dinnen RD, Mao Y, Stevens PD, Schrope B, Allendorf J, Lee JA, Sherman WH, Chabot JA (2013) Capecitabine and temozolomide (CAPTEM) for metastatic, well-differentiated neuroendocrine cancers: the Pancreas Center at Columbia University experience. Cancer Chemother Pharmacol 71:663–670. https://doi.org/10.1007/s00280-012-2055-z

    Article  CAS  PubMed  Google Scholar 

  34. Krug S, Boch M, Nimphius W, Gress TM, Michl P, Rinke A (2017) Relevance of dihydropyrimidine-dehydrogenase and thymidylate-synthase in patients with pancreatic neuroendocrine neoplasms treated with 5-FU-based chemotherapy. Pancreatology 17:139–145. https://doi.org/10.1016/j.pan.2016.12.006

    Article  CAS  PubMed  Google Scholar 

  35. Maxwell JA, Johnson SP, Quinn JA, McLendon RE, Ali-Osman F, Friedman AH, Herndon JE IInd, Bierau K, Bigley J, Bigner DD, Friedman HS (2006) Quantitative analysis of O6-alkylguanine-DNA alkyltransferase in malignant glioma. Mol Cancer Ther 5:2531–2539. https://doi.org/10.1158/1535-7163.mct-06-0106

    Article  CAS  PubMed  Google Scholar 

  36. Preusser M, Charles Janzer R, Felsberg J, Reifenberger G, Hamou MF, Diserens AC, Stupp R, Gorlia T, Marosi C, Heinzl H, Hainfellner JA, Hegi M (2008) Anti-O6-methylguanine-methyltransferase (MGMT) immunohistochemistry in glioblastoma multiforme: observer variability and lack of association with patient survival impede its use as clinical biomarker. Brain Pathol (Zurich) 18:520–532. https://doi.org/10.1111/j.1750-3639.2008.00153.x

    Article  CAS  Google Scholar 

  37. Mollemann M, Wolter M, Felsberg J, Collins VP, Reifenberger G (2005) Frequent promoter hypermethylation and low expression of the MGMT gene in oligodendroglial tumors. Int J Cancer 113:379–385. https://doi.org/10.1002/ijc.20575

    Article  CAS  PubMed  Google Scholar 

  38. Sato K, Kitajima Y, Kohya N, Miyoshi A, Koga Y, Miyazaki K (2005) Deficient MGMT and proficient hMLH1 expression renders gallbladder carcinoma cells sensitive to alkylating agents through G2-M cell cycle arrest. Int J Oncol 26:1653–1661

    CAS  PubMed  Google Scholar 

  39. Sato K, Kitajima Y, Nakagawachi T, Soejima H, Miyoshi A, Koga Y, Miyazaki K (2005) Cisplatin represses transcriptional activity from the minimal promoter of the O6-methylguanine methyltransferase gene and increases sensitivity of human gallbladder cancer cells to 1-(4-amino-2-methyl-5-pyrimidinyl) methyl-3-2-chloroethyl)-3-nitrosourea. Oncol Rep 13:899–906

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors extend our sincere appreciation to Dr. Hironori Koga (Kurume University, Kurume, Japan).

Funding

This study was funded by the Aichi Cancer Research Foundation, Japan (K.S.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Susumu Hijioka.

Ethics declarations

Conflict of interest

Susumu Hijioka has received speaker honoraria from Novelphama, Novartis, and Teijin Pharma. Nobumasa Mizuno has received research funding from AstraZeneca, Zeria Pharmaceutical, Taiho Pharmaceutical, Merck Serono, Eisai, NanoCarrier, MSD, Dainippon Sumitomo Pharma, and Novartis. Keiichiro Sakuma, Masahiro Aoki, Takamichi Kuwahara, Nozomi Okuno, Kazuo Hara, and Yasushi Yatabe have no conflicts of interest.

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Informed consent

Informed consent was obtained from all individual participants included in the study.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 22 KB)

Online Resource 1. OS for pancreatic neuroendocrine tumor patients treated with streptozocin therapy

. The solid line is MGMT- negative (deficient) as determined by IHC, and the dotted line is MGMT- positive (intact) as determined by IHC. The median OS of the MGMT negative and MGMT positive groups was 827 days (range,163-1296), and 447 days (range, 81–1218 days), respectively (P = 0.339). (TIF 66 KB)

Online Resource 2

. MTT assay of MGMT-knockdown QGP1 cells treated with streptozocin. MTT activity was assayed in MGMT-knockdown QGP1 subclones treated with streptozocin. P = 0.022 (*) compared with Luc shRNA-transduced QGP1 cells treated with the same concentration of streptozocin. (TIF 167 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hijioka, S., Sakuma, K., Aoki, M. et al. Clinical and in vitro studies of the correlation between MGMT and the effect of streptozocin in pancreatic NET. Cancer Chemother Pharmacol 83, 43–52 (2019). https://doi.org/10.1007/s00280-018-3700-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00280-018-3700-y

Keywords

Navigation