Skip to main content
Top
Published in: Targeted Oncology 3/2016

01-06-2016 | Original Research Article

Dose-Dense Temozolomide in Patients with MGMT-Silenced Chemorefractory Colorectal Cancer

Authors: Filippo Pietrantonio, Filippo de Braud, Massimo Milione, Claudia Maggi, Roberto Iacovelli, Katia Fiorella Dotti, Federica Perrone, Elena Tamborini, Marta Caporale, Rosa Berenato, Giorgia Leone, Alessio Pellegrinelli, Ilaria Bossi, Fabrizio Festinese, Stefano Federici, Maria Di Bartolomeo

Published in: Targeted Oncology | Issue 3/2016

Login to get access

Abstract

Background

In a phase II study, we showed that temozolomide (TMZ) was tolerable and active in heavily pre-treated patients with advanced colorectal cancer (CRC) and MGMT methylation. A schedule of dose-dense TMZ may have enhanced activity due to the higher cumulative dose and induction of MGMT depletion, even in resistant tumors.

Methods

Thirty-two patients with chemorefractory MGMT-methylated CRC were treated with TMZ at a daily dose of 75 mg/m2 for 21 consecutive days every 4 weeks, for up to six cycles or until the occurrence of progressive disease/unacceptable toxicity. The primary endpoint was treatment activity in terms of objective response rate (ORR). MGMT protein expression was tested by immunohistochemistry (IHC) on two pooled cohorts: patients from the previous study of standard-dose TMZ and those from the current investigation.

Results

From November 2013 to December 2014, 32 patients were treated at Fondazione IRCCS Istituto Nazionale dei Tumori. We observed only three episodes of grade 3 asthenia and no significant myelotoxicity. The ORR was 16 % (all partial responses occurring in RAS-BRAF-mutated tumors). Median progression-free survival (PFS) and overall survival (OS) were 2.3 and 6.7 months, respectively. Patients with MGMT-low expression by IHC had a significantly higher ORR (p < 0.0001) and PFS (p = 0.001) compared to those with MGMT-high expression, while no difference was observed in OS.

Conclusions

Our data confirm the encouraging activity of TMZ in chemorefractory CRC patients selected for MGMT silencing, even in the RAS-BRAF-mutated population. The role of MGMT IHC as a biomarker for improving patient selection warrants further prospective confirmation.
https://static-content.springer.com/image/art%3A10.1007%2Fs11523-015-0397-2/MediaObjects/11523_2015_397_Figa_HTML.gif
Literature
1.
go back to reference Shen L, Kondo Y, Rosner GL, Xiao L, Hernandez NS, Vilaythong J et al (2005) MGMT promoter methylation and field defect in sporadic colorectal cancer. J Natl Cancer Inst 97:1330–8CrossRefPubMed Shen L, Kondo Y, Rosner GL, Xiao L, Hernandez NS, Vilaythong J et al (2005) MGMT promoter methylation and field defect in sporadic colorectal cancer. J Natl Cancer Inst 97:1330–8CrossRefPubMed
2.
go back to reference Esteller M, Risques RA, Toyota M, Capella G, Moreno V, Peinado MA et al (2001) Promoter hypermethylation of the DNA repair gene O(6)-methylguanine-DNA methyltransferase is associated with the presence of G: C to A: T transition mutations in p53 in human colorectal tumorigenesis. Cancer Res 61:4689–92PubMed Esteller M, Risques RA, Toyota M, Capella G, Moreno V, Peinado MA et al (2001) Promoter hypermethylation of the DNA repair gene O(6)-methylguanine-DNA methyltransferase is associated with the presence of G: C to A: T transition mutations in p53 in human colorectal tumorigenesis. Cancer Res 61:4689–92PubMed
3.
go back to reference Esteller M, Herman JG (2004) Generating mutations but providing chemosensitivity: the role of O6-methylguanine DNA methyltransferase in human cancer. Oncogene 23:1–8CrossRefPubMed Esteller M, Herman JG (2004) Generating mutations but providing chemosensitivity: the role of O6-methylguanine DNA methyltransferase in human cancer. Oncogene 23:1–8CrossRefPubMed
4.
go back to reference Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M et al (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 352:997–1003CrossRefPubMed Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M et al (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 352:997–1003CrossRefPubMed
5.
go back to reference Amatu A, Sartore-Bianchi A, Moutinho C, Belotti A, Bencardino K, Chirico G et al (2013) Promoter CpG island hypermethylation of the DNA repair enzyme MGMT predicts clinical response to dacarbazine in a phase II study for metastatic colorectal cancer. Clin Cancer Res 19:2265–72CrossRefPubMed Amatu A, Sartore-Bianchi A, Moutinho C, Belotti A, Bencardino K, Chirico G et al (2013) Promoter CpG island hypermethylation of the DNA repair enzyme MGMT predicts clinical response to dacarbazine in a phase II study for metastatic colorectal cancer. Clin Cancer Res 19:2265–72CrossRefPubMed
6.
go back to reference Hochhauser D, Glynne-Jones R, Potter V, Grávalos C, Doyle TJ, Pathiraja K et al (2013) A phase II study of temozolomide in patients with advanced aerodigestive tract and colorectal cancers and methylation of the O6-methylguanine-DNA methyltransferase promoter. Mol Cancer Ther 12:809–18CrossRefPubMed Hochhauser D, Glynne-Jones R, Potter V, Grávalos C, Doyle TJ, Pathiraja K et al (2013) A phase II study of temozolomide in patients with advanced aerodigestive tract and colorectal cancers and methylation of the O6-methylguanine-DNA methyltransferase promoter. Mol Cancer Ther 12:809–18CrossRefPubMed
7.
go back to reference Inno A, Fanetti G, Di Bartolomeo M, Gori S, Maggi C, Cirillo M et al (2014) Role of MGMT as biomarker in colorectal cancer. World J Clin Cases 2:835–9CrossRefPubMedPubMedCentral Inno A, Fanetti G, Di Bartolomeo M, Gori S, Maggi C, Cirillo M et al (2014) Role of MGMT as biomarker in colorectal cancer. World J Clin Cases 2:835–9CrossRefPubMedPubMedCentral
8.
go back to reference Pietrantonio F, Perrone F, de Braud F, Castano A, Maggi C, Bossi I et al (2014) Activity of temozolomide in patients with advanced chemorefractory colorectal cancer and MGMT promoter methylation. Ann Oncol 25:404–8CrossRefPubMed Pietrantonio F, Perrone F, de Braud F, Castano A, Maggi C, Bossi I et al (2014) Activity of temozolomide in patients with advanced chemorefractory colorectal cancer and MGMT promoter methylation. Ann Oncol 25:404–8CrossRefPubMed
9.
go back to reference Tolcher AW, Gerson SL, Denis L, Geyer C, Hammond LA, Patnaik A et al (2003) Marked inactivation of O6-alkylguanine-DNA alkyl-transferase activity with protracted temozolomide schedules. Br J Cancer 88:1004–11CrossRefPubMedPubMedCentral Tolcher AW, Gerson SL, Denis L, Geyer C, Hammond LA, Patnaik A et al (2003) Marked inactivation of O6-alkylguanine-DNA alkyl-transferase activity with protracted temozolomide schedules. Br J Cancer 88:1004–11CrossRefPubMedPubMedCentral
10.
go back to reference Chinot OL, Barrié M, Fuentes S, Eudes N, Lancelot S, Metellus P et al (2007) Correlation between O6-methylguanine-DNA methyltransferase and survival in inoperable newly diagnosed glioblastoma patients treated with neoadjuvant temozolomide. J Clin Oncol 25:1470–5CrossRefPubMed Chinot OL, Barrié M, Fuentes S, Eudes N, Lancelot S, Metellus P et al (2007) Correlation between O6-methylguanine-DNA methyltransferase and survival in inoperable newly diagnosed glioblastoma patients treated with neoadjuvant temozolomide. J Clin Oncol 25:1470–5CrossRefPubMed
11.
go back to reference Di Bartolomeo M, Pietrantonio F, Perrone F, Dotti KF, Lampis A, Bertan C, Italian Trials in Medical Oncology Group et al (2014) Lack of KRAS, NRAS, BRAF and TP53 mutations improves outcome of elderly metastatic colorectal cancer patients treated with cetuximab, oxaliplatin and UFT. Target Oncol 9:155–62CrossRefPubMed Di Bartolomeo M, Pietrantonio F, Perrone F, Dotti KF, Lampis A, Bertan C, Italian Trials in Medical Oncology Group et al (2014) Lack of KRAS, NRAS, BRAF and TP53 mutations improves outcome of elderly metastatic colorectal cancer patients treated with cetuximab, oxaliplatin and UFT. Target Oncol 9:155–62CrossRefPubMed
12.
go back to reference Simon R (1989) Optimal two-stage design for phase II clinical trials. Control Clin Trials 10:1–10CrossRefPubMed Simon R (1989) Optimal two-stage design for phase II clinical trials. Control Clin Trials 10:1–10CrossRefPubMed
14.
go back to reference Hsu CY, Lin SC, Ho HL, Chang-Chien YC, Hsu SP, Yen YS et al (2013) Exclusion of histiocytes/endothelial cells and using endothelial cells as internal reference are crucial for interpretation of MGMT immunohistochemistry in glioblastoma. Am J Surg Pathol 37:264–71CrossRefPubMed Hsu CY, Lin SC, Ho HL, Chang-Chien YC, Hsu SP, Yen YS et al (2013) Exclusion of histiocytes/endothelial cells and using endothelial cells as internal reference are crucial for interpretation of MGMT immunohistochemistry in glioblastoma. Am J Surg Pathol 37:264–71CrossRefPubMed
15.
go back to reference Preusser M, Charles Janzer R, Felsberg J, Reifenberger G, Hamou MF et al (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 18:520–32PubMed Preusser M, Charles Janzer R, Felsberg J, Reifenberger G, Hamou MF et al (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 18:520–32PubMed
16.
go back to reference Capper D, Mittelbronn M, Meyermann R, Schittenhelm J (2008) Pitfalls in the assessment of MGMT expression and in its correlation with survival in diffuse astrocytomas: proposal of a feasible immunohistochemical approach. Acta Neuropathol 115:249–59CrossRefPubMed Capper D, Mittelbronn M, Meyermann R, Schittenhelm J (2008) Pitfalls in the assessment of MGMT expression and in its correlation with survival in diffuse astrocytomas: proposal of a feasible immunohistochemical approach. Acta Neuropathol 115:249–59CrossRefPubMed
17.
go back to reference Brell M, Tortosa A, Verger E, Gil JM, Viñolas N, Villá S et al (2005) Prognostic significance of O6-methylguanine-DNA methyltransferase determined by promoter hypermethylation and immunohistochemical expression in anaplastic gliomas. Clin Cancer Res 11:5167–74CrossRefPubMed Brell M, Tortosa A, Verger E, Gil JM, Viñolas N, Villá S et al (2005) Prognostic significance of O6-methylguanine-DNA methyltransferase determined by promoter hypermethylation and immunohistochemical expression in anaplastic gliomas. Clin Cancer Res 11:5167–74CrossRefPubMed
18.
go back to reference Amatu A, Bencardino K, Barault L, Cassingena A, Bonazzina E, Ghezzi S et al (2015) Phase II study of temozolomide (TMZ) in metastatic colorectal cancer (mCRC) patients molecularly selected by MGMT promoter hypermethylation [abstract]. J Clin Oncol 33(Suppl 3):583 Amatu A, Bencardino K, Barault L, Cassingena A, Bonazzina E, Ghezzi S et al (2015) Phase II study of temozolomide (TMZ) in metastatic colorectal cancer (mCRC) patients molecularly selected by MGMT promoter hypermethylation [abstract]. J Clin Oncol 33(Suppl 3):583
19.
go back to reference Esteller M, Toyota M, Sanchez-Cespedes M, Capella G, Peinado MA, Watkins DN et al (2000) Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is associated with G to A mutations in K-ras in colorectal tumorigenesis. Cancer Res 60:2368–71PubMed Esteller M, Toyota M, Sanchez-Cespedes M, Capella G, Peinado MA, Watkins DN et al (2000) Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is associated with G to A mutations in K-ras in colorectal tumorigenesis. Cancer Res 60:2368–71PubMed
20.
go back to reference Sato A, Sunayama J, Matsuda K, Seino S, Suzuki K, Watanabe E et al (2011) MEK-ERK signaling dictates DNA-repair gene MGMT expression and temozolomide resistance of stem-like glioblastoma cells via the MDM2-p53 axis. Stem Cells 29:1942–51CrossRefPubMed Sato A, Sunayama J, Matsuda K, Seino S, Suzuki K, Watanabe E et al (2011) MEK-ERK signaling dictates DNA-repair gene MGMT expression and temozolomide resistance of stem-like glioblastoma cells via the MDM2-p53 axis. Stem Cells 29:1942–51CrossRefPubMed
21.
go back to reference Tosoni A, Franceschi E, Ermani M, Bertorelle R, Bonaldi L, Blatt V et al (2008) Temozolomide three weeks on and one week off as first line therapy for patients with recurrent or progressive low grade gliomas. J Neuro-Oncol 89:179–85CrossRef Tosoni A, Franceschi E, Ermani M, Bertorelle R, Bonaldi L, Blatt V et al (2008) Temozolomide three weeks on and one week off as first line therapy for patients with recurrent or progressive low grade gliomas. J Neuro-Oncol 89:179–85CrossRef
22.
go back to reference Miyazaki M, Nishihara H, Terasaka S, Kobayashi H, Yamaguchi S, Ito T et al (2014) Immunohistochemical evaluation of O6 -methylguanine DNA methyltransferase (MGMT) expression in 117 cases of glioblastoma. Neuropathology 34:268–76CrossRefPubMed Miyazaki M, Nishihara H, Terasaka S, Kobayashi H, Yamaguchi S, Ito T et al (2014) Immunohistochemical evaluation of O6 -methylguanine DNA methyltransferase (MGMT) expression in 117 cases of glioblastoma. Neuropathology 34:268–76CrossRefPubMed
23.
go back to reference Brell M, Ibáñez J, Tortosa A (2011) O6-Methylguanine-DNA methyltransferase protein expression by immunohistochemistry in brain and non-brain systemic tumours: systematic review and meta-analysis of correlation with methylation-specific polymerase chain reaction. BMC Cancer 11:35CrossRefPubMedPubMedCentral Brell M, Ibáñez J, Tortosa A (2011) O6-Methylguanine-DNA methyltransferase protein expression by immunohistochemistry in brain and non-brain systemic tumours: systematic review and meta-analysis of correlation with methylation-specific polymerase chain reaction. BMC Cancer 11:35CrossRefPubMedPubMedCentral
24.
go back to reference Barault L, Amatu A, Bleeker FE, Moutinho C, Falcomatà C, Fiano V, et al. Digital PCR quantification of MGMT methylation refines prediction of clinical benefit from alkylating agents in glioblastoma and metastatic colorectal cancer. Ann Oncol. 2015. doi:10.1093/annonc/mdv272 Barault L, Amatu A, Bleeker FE, Moutinho C, Falcomatà C, Fiano V, et al. Digital PCR quantification of MGMT methylation refines prediction of clinical benefit from alkylating agents in glioblastoma and metastatic colorectal cancer. Ann Oncol. 2015. doi:10.​1093/​annonc/​mdv272
25.
go back to reference Fine RL, Gulati AP, Krantz BA, Moss RA, Schreibman S, Tsushima DA et al (2013) Capecitabine and temozolomide (CAPTEM) for metastatic, well-differentiated neuroendocrine cancers: The Pancreas Center at Columbia University experience. Cancer Chemother Pharmacol 71:663–70CrossRefPubMed Fine RL, Gulati AP, Krantz BA, Moss RA, Schreibman S, Tsushima DA et al (2013) Capecitabine and temozolomide (CAPTEM) for metastatic, well-differentiated neuroendocrine cancers: The Pancreas Center at Columbia University experience. Cancer Chemother Pharmacol 71:663–70CrossRefPubMed
26.
go back to reference Reynés G, Balañá C, Gallego O, Iglesias L, Pérez P, García JL (2014) A phase I study of irinotecan in combination with metronomic temozolomide in patients with recurrent glioblastoma. Anti-Cancer Drugs 25:717–22PubMed Reynés G, Balañá C, Gallego O, Iglesias L, Pérez P, García JL (2014) A phase I study of irinotecan in combination with metronomic temozolomide in patients with recurrent glioblastoma. Anti-Cancer Drugs 25:717–22PubMed
27.
go back to reference Ku GY, Krol G, Ilson DH (2007) Successful treatment of leptomeningeal disease in colorectal cancer with a regimen of bevacizumab, temozolomide, and irinotecan. J Clin Oncol 25:e14–6CrossRefPubMed Ku GY, Krol G, Ilson DH (2007) Successful treatment of leptomeningeal disease in colorectal cancer with a regimen of bevacizumab, temozolomide, and irinotecan. J Clin Oncol 25:e14–6CrossRefPubMed
Metadata
Title
Dose-Dense Temozolomide in Patients with MGMT-Silenced Chemorefractory Colorectal Cancer
Authors
Filippo Pietrantonio
Filippo de Braud
Massimo Milione
Claudia Maggi
Roberto Iacovelli
Katia Fiorella Dotti
Federica Perrone
Elena Tamborini
Marta Caporale
Rosa Berenato
Giorgia Leone
Alessio Pellegrinelli
Ilaria Bossi
Fabrizio Festinese
Stefano Federici
Maria Di Bartolomeo
Publication date
01-06-2016
Publisher
Springer International Publishing
Published in
Targeted Oncology / Issue 3/2016
Print ISSN: 1776-2596
Electronic ISSN: 1776-260X
DOI
https://doi.org/10.1007/s11523-015-0397-2

Other articles of this Issue 3/2016

Targeted Oncology 3/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine