Hostname: page-component-848d4c4894-sjtt6 Total loading time: 0 Render date: 2024-06-28T10:46:32.815Z Has data issue: false hasContentIssue false

When Temozolomide Alone Fails: Adding Procarbazine in Salvage Therapy of Glioma

Published online by Cambridge University Press:  02 December 2014

Fleur Huang
Affiliation:
Division of Radiation Oncology, McGill University Health Centre, Montreal Neurological Hospital, Montreal, Quebec, Canada
Petr Kavan
Affiliation:
Department of Oncology, McGill University Health Centre, Montreal Neurological Hospital, Montreal, Quebec, Canada
Marie-Christine Guiot
Affiliation:
Department of Pathology, McGill University Health Centre, Montreal Neurological Hospital, Montreal, Quebec, Canada
Yelena Markovic
Affiliation:
Department of Pathology, McGill University Health Centre, Montreal Neurological Hospital, Montreal, Quebec, Canada
David Roberge
Affiliation:
Division of Radiation Oncology, McGill University Health Centre, Montreal Neurological Hospital, Montreal, Quebec, Canada
Rights & Permissions [Opens in a new window]

Abstract

Core share and HTML view are not available for this content. However, as you have access to this content, a full PDF is available via the ‘Save PDF’ action button.
Background:

Since temozolomide (TMZ) entry into routine practice in the first-line management of glial tumors, post-TMZ recurrences present a growing challenge. Without standard chemotherapy for TMZ failure, care in such palliative settings requires consideration not only of efficacy but of toxicity and convenience.

Methods:

At our institution, a combination regimen has been used: oral alkylating agents procarbazine (PCB) (100-150 mg/m2/day) and TMZ (150-200 mg/m2/day) administered on days 1-5 of a 28-day cycle. This treatment has been initiated upon radiological and/or clinical disease progression, and continued until evidence of further progression or toxicity. We retrospectively reviewed our experence with this regimen.

Results:

Since November 2004, 17 patients (median age 53) were treated for histologically confirmed glioma (glioblastoma multiforme (GBM), N=12; Grade 3 glioma, N=3; Grade 2 glioma, N=2) after a median of 2 recurrences. TMZ was previously given either as adjuvant therapy (post-chemoradiotherapy maintenance in 8 of 13 cases) or as salvage monotherapy (4 cases). Of 16 evaluable cases, 14 (13 high grade tumors) showed O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. Two patients achieved partial response and one had complete response by RECIST criteria. Disease progressed after a median of 4 cycles (range 1 to 11+), with an actuarial progression-free survival of 42% after 6 cycles. Grade 3/4 toxicity was rare, and no dose reductions were needed. One patient discontinued treatment due to procarbazine hypersensitivity.

Conclusion:

Combination PCB-TMZ is well-tolerated, with modest activity in TMZ-exposed glioma.

Résumé:

<span class='bold'>RÉSUMÉ:</span><span class='bold'><span class='italic'>Contexte:</span></span>

Depuis que le témozolomide (TMZ) est utilisé de routine comme traitement majeur des tumeurs gliales, les rechutes après le traitement par le TMZ presentment un défi de plus en plus grand. Quand on fait face à un échec du traitement par le TMZ et en l’absence de chimiothérapie standard, on doit considerer non seulement l’efficacité mais aussi la toxicité et les aspects pratiques du traitement dans ce contexte palliatif.

<span class='bold'><span class='italic'>Méthodes:</span></span>

Dans notre institution, nous avons utilisé un traitement d’association : deux agents alkylants oraux, la procarbazine (PCB) (100–150 mg/m2/ j) et le TMZ (150–200 mg/m2/ j), administrés aux jours 1–5 d’un cycle de 28 jours. Ce traitement était commencé lorsqu’on observait une progression radiologique et/ou clinique de la tumeur et il était maintenu jusqu’à ce qu’on observe une progression plus marquée ou de la toxicité. Nous avons fait une revue rétrospective de ce mode de traitement.

<span class='bold'><span class='italic'>Résultats:</span></span>

Depuis novembre 2004, 17 patients dont l’âge médian était de 53 ans ont été traités pour un gliome confirmé par l’histologie (glioblastome muliforme (GBM) N = 12; gliome de grade 3, N = 3; gliome de grade 2, N = 2) après un nombre de rechutes médian de 2. Le TMZ avait été donné antérieurement soit comme traitement adjuvant (traitement d’entretien post–chimioradiothérapie chez 8 patients sur 13) ou en monothérapie comme traitement de rattrapage (chez 4 patients). Chez 14 (13 cas de tumeurs de haut grade) des 16 patients évaluables, on a observé une méthylation du promoteur de l’O–6–méthylguanine–DNA méthyltransférase (MGMT). Deux patients ont eu une réponse partielle et un a eu une réponse complete selon les critères RECIST. La maladie a progressé après 1 à 11 cycles ou plus, médiane de 4 cycles, avec une survie actuarielle sans progression de 42% après 6 cycles. Une toxicité de grade 3/4 était rare et aucune réduction de dose n’a été nécessaire. Un patient a cessé le traitement à cause d’une hypersensibilité à la procarbazine.

<span class='bold'><span class='italic'>Conclusion:</span></span>

La combinaison PCB–TMZ est bien tolérée et son efficacité sur les gliomes qui ont déjà été exposés au TMZ est modeste.

Type
Original Articles
Copyright
Copyright © The Canadian Journal of Neurological 2008

References

Stupp, R, Hegi, ME, van den Bent, MJ, Mason, WP, Weller, M, Mirimanoff, RO, et al. Changing paradigms—an update on the multidisciplinary management of malignant glioma. Oncologist. 2006 Feb; 11(2):16580.Google Scholar
Brandes, AA, Tosoni, A, Basso, U, Reni, M, Valduga, F, Monfardini, S, et al. Second-line chemotherapy with irinotecan plus carmustine in glioblastoma recurrent or progressive after first-line temozolomide chemotherapy: a phase II study of the Gruppo Italiano Cooperativo di Neuro-Oncologia (GICNO). J Clin Oncol. 2004 Dec 1; 22(23):477986.CrossRefGoogle ScholarPubMed
Rabik, CA, Njoku, MC, Dolan, ME. Inactivation of O6-alkylguanine DNA alkyltransferase as a means to enhance chemotherapy. Cancer Treat Rev. 2006 Jun; 32(4):26176.CrossRefGoogle ScholarPubMed
Brandes, AA, Reni, M. Treatment options for recurrent glioblastoma. PCNSM2- Perspectives in central nervous system malignancies. Budapest, Hungary; 2006.Google Scholar
Khan, RB, Raizer, JJ, Malkin, MG, Bazylewicz, KA, Abrey, LE. A phase II study of extended low-dose temozolomide in recurrent malignant gliomas. Neuro-oncol. 2002 Jan; 4(1):3943.CrossRefGoogle ScholarPubMed
Tosoni, A, Cavallo, G, Ermani, M, Scopece, L, Franceschi, E, Ghimenton, C, et al. Is protracted low-dose temozolomide feasible in glioma patients? Neurology. 2006 Feb 14; 66(3):4279.Google Scholar
Berrocal, A, Perez-Segura, P, Gil, M, Balaña, C, Yaya-Tur, R, Yaya-Tur, R, et al. Phase II study of extended schedule temozolomide in refractory gliomas. J Clin Oncol. 2006; 24(18S):1516.CrossRefGoogle Scholar
Vera, K, Djafari, L, Faivre, S, Guillamo, JS, Djazouli, K, Osorio, M, et al. Dose-dense regimen of temozolomide given every other week in patients with primary central nervous system tumors. Ann Oncol. 2004 Jan; 15(1):16171.Google Scholar
Koch, D, Wick, W. Higher dosing of temozolomide? Regression of an anaplastic oligoastrocytoma over more than three years. J Neurooncol. 2006 Apr; 77(2):21920.Google Scholar
Spence, AM, Kiem, HP, Partap, S, Schuetze, S, Silber, JR, Peterson, RA. Complications of a temozolomide overdose: a case report. J Neurooncol. 2006 Oct; 80(1):5761.Google Scholar
Franceschi, E, Omuro, AM, Lassman, AB, Demopoulos, A, Nolan, C, Abrey, LE. Salvage temozolomide for prior temozolomide responders. Cancer. 2005 Dec 1; 104(11):24736.Google Scholar
Groves, MD, Puduvalli, VK, Conrad, CA, Gilbert, MR, Yung, WK, Jaeckle, K, et al. Phase II trial of temozolomide plus marimastat for recurrent anaplastic gliomas: a relationship among efficacy, joint toxicity and anticonvulsant status. J Neurooncol. 2006 Oct; 80(1):8390.Google Scholar
Raizer, JJ, Abrey, LE, Wen, P, Cloughesy, T, Robins, IA, Fine, HA, et al. A phase II trial of erlotinib (OSI-774) in patients with recurrent malignant gliomas not on EIAEDs. 2004 ASCO Annual Meeting; 2004; New-Orleans, Louisiana: J Clin Oncol. 2004; p. 1502.Google Scholar
Esteller, M, Garcia-Foncillas, J, Andion, E, Goodman, SN, Hidalgo, OF, Vanaclocha, V, et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000 Nov 9; 343(19):13504.Google Scholar
Bollag, W. The tumor-inhibitory effects of the methylhydrazine derivative Ro 4-6467/1 (Nsc-77213). Cancer Chemother Rep. 1963 Dec; 33:14.Google ScholarPubMed
Goodman & Gilman’s The pharmacological basis of therapeutics. 11th ed. New York: McGraw-Hill; 2006.Google Scholar
European Organization for Research on Treatment of Cancer (EORTC) Brain Tumor Group. Evaluation of CCNU, VM-26 plus CCNU, and procarbazine in supratentorial brain gliomas. Final evaluation of a randomized study. J Neurosurg. 1981 Jul; 55(1):2731.Google Scholar
Rodriguez, LA, Prados, M, Silver, P, Levin, VA. Reevaluation of procarbazine for the treatment of recurrent malignant central nervous system tumors. Cancer. 1989 Dec 15; 64(12):24203.Google Scholar
Newton, HB, Junck, L, Bromberg, J, Page, MA, Greenberg, HS. Procarbazine chemotherapy in the treatment of recurrent malignant astrocytomas after radiation and nitrosourea failure. Neurology. 1990 Nov; 40(11):17436.Google Scholar
Grossman, SA, Carson, KA, Batchelor, TT, Lesser, G, Mikkelsen, T, Alavi, JB, et al. The effect of enzyme-inducing antiseizure drugs on the pharmacokinetics and tolerability of procarbazine hydrochloride. Clin Cancer Res. 2006 Sep 1; 12(17):517481.Google Scholar
Newlands, ES, Foster, T, Zaknoen, S. Phase I study of temozolamide (TMZ) combined with procarbazine (PCB) in patients with gliomas. Br J Cancer. 2003 Jul 21; 89(2):24851.Google Scholar
DeVita, VT Jr, Hellman, S, Rosenberg, S, editors. Cancer principles & practice of oncology. 7th ed. Philadelphia: Lippincott Williams & Wilkins; 2005.Google Scholar