Journal List > J Korean Med Sci > v.33(24) > 1108415

Kim, Yoo, Chang, Kim, Chung, Kim, Park, Lee, and Yang: Procarbazine and CCNU Chemotherapy for Recurrent Glioblastoma with MGMT Promoter Methylation

Abstract

Background

While procarbazine, CCNU (lomustine), and vincristine (PCV) has been an alternative chemotherapy option for malignant gliomas, it is worth investigating whether the combination of only procarbazine and CCNU is comparable because vincristine adds toxicity with uncertain benefit. The purpose of this study was to evaluate the feasibility of procarbazine and CCNU chemotherapy for recurrent glioblastoma multiforme (GBM) with O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation.

Methods

Eight patients with recurrent GBM following concurrent chemoradiotherapy and temozolomide (TMZ) adjuvant therapy were enrolled in this trial; they received no other chemotherapeutic agents or target therapy. They received CCNU (75 mg/m2) on day 1 and procarbazine (60 mg/m2) through days 11 and 24 every 4 weeks. The median cycle of CCNU and procarbazine was 3.5 (range: 2–6).

Results

One patient achieved stable disease. The median progression-free survival (PFS) with procarbazine and CCNU chemotherapy was eight weeks (range: 5–73), and the PFS rates were 25% and 12.5% at 16 and 30 weeks, respectively. The median overall survival (OS) from the initial diagnosis to death was 40 months, and the median OS from the administration of procarbazine and CCNU chemotherapy to death was 9.7 months (95% confidence interval: 6.7–12.7). Serious adverse events were found at six visits, and two cases were considered to be grade 3 toxicities.

Conclusion

The efficacy of procarbazine and CCNU chemotherapy is not satisfactory. This study suggests the need to develop other treatment strategies for recurrent and TMZ-refractory GBM.

Trial Registration

ClinicalTrials.gov Identifier: NCT017337346

Graphical Abstract

jkms-33-e167-abf001

INTRODUCTION

The prognosis for glioblastoma multiforme (GBM) patients remains poor despite advances in surgical techniques, radiotherapy, and chemotherapy. The median overall survival (OS) is expected to be only 14.6 months after maximum safe resection and irradiation with concurrent and adjuvant temozolomide (TMZ) chemotherapy. In spite of multimodal therapies, most patients suffer recurrence and die within 40 weeks,12 and there is no consensus on treating recurrent and TMZ-refractory GBM. Bevacizumab, a humanized monoclonal antibody against vascular endothelial growth factor A, has shown significant biological activity in patients with recurrent GBM and has been under investigation with other target agents.3 Re-challenge with alternative-dose TMZ for recurrent GBM can be recommended even if the patient has a history of standard TMZ chemotherapy.45
Nitrosourea has become a second choice after TMZ for malignant gliomas.6 Retrospective and subgroup analyses suggest higher efficacy for procarbazine, CCNU (lomustine), and vincristine (PCV) chemotherapy than for TMZ in patients with anaplastic glioma, with good prognosis as well.78 With the PCV triple-drug regimen, CCNU (110 mg/m2, day 1) and procarbazine (60 mg/m2, days 8–21) are administered orally but vincristine is administered intravenously (maximum 2 mg, days 8 and 29). The molecular weight of vincristine is 825 daltons, so that it has less permeability through the blood-brain barrier.9 It is well-known that vincristine is associated with neurotoxicity and could impair the quality of life in brain cancer patients, and the number of outpatient visits will decrease by omitting intravenous administration of vincristine. Authors of one study reported that reducing CCNU dose by 30% reduced the hematological toxicity (grade 3/4) from 25.6% to 13%.10 Based on these findings, we designed a modified procarbazine and CCNU therapy protocol for treating recurrent GBM with O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation. The purpose of this study was to evaluate the feasibility of the procarbazine and CCNU chemotherapy for recurrent GBM with MGMT promoter methylation.

METHODS

Study population

We enrolled patients at least 20 years of age who had been diagnosed with pathologically confirmed GBM with MGMT promoter methylation, which we assessed by methylation-specific polymerase chain reaction (PCR); we allocated patients with recurrent GBM with an unmethylated MGMT promoter to another trial. Radiographically confirmed tumor progression by magnetic resonance imaging (MRI) following standard external beam fractionated radiotherapy and TMZ chemotherapy was required for enrollment, along with a minimum Karnofsky performance status of 60, adequate hematologic, hepatic, and renal function, and at least a two-week period from any prior surgery or other chemotherapy. The following patient groups were excluded: 1) those with other cancer history, 2) those who had been treated with CCNU or procarbazine, and 3) those with leptomeningeal seeding.

Treatment schedule

The patients had previously been treated with concurrent chemoradiotherapy (CCRT) with adjuvant TMZ following the initial diagnosis of GBM. We determined recurrence by MRI scans assessed using the Response Assessment in Neuro-Oncology criteria. CCNU (75 mg/m2) was administered on day 1 and procarbazine (60 mg/m2) was administered through days 11 and 24 every 4 weeks. Administration of CCNU and procarbazine was repeated every 28 days for up to 6 months. We evaluated all toxicities according to the Common Terminology Criteria for Adverse Events version 4.0. All patients underwent brain MRI at the baseline within two weeks of starting CCNU, and they were evaluated by laboratory examinations of blood and neurological evaluation every four weeks; they also underwent gadolinium-enhanced brain MRI after every two cycles of chemotherapy (i.e., every eight weeks). Specifically, we required stable or decreasing corticosteroid dose and stable or improved fluid-attenuated inversion recovery abnormality for a rating of complete response, partial response, or stable disease based on complete, partial, or no decrease in the enhancing tumor burden on standard post-gadolinium T1-weighted sequences.

Study design and statistical analysis

We calculated the sample size using the following formula:
n=pqzα/2d2jkms-33-e167-e001
where n is the required sample size, p is the known response rate, 0.15, q is 0.85 (1−p), α is 0.05, and d is the drop rate, 0.05. Approximately 52 patients needed to be enrolled for the study to achieve a difference from the historical control.
Progression-free survival (PFS) was the primary end point of this study. Secondary end points were OS using Kaplan-Meier estimation and safety of combining CCNU and procarbazine in patients with recurrent GBM.
We estimated PFS from the start of CCNU and procarbazine administration to tumor progression or dropout from the study and OS from the start of CCNU and procarbazine administration to the date of death, irrespective of its cause. We assessed PFS and OS by the Kaplan-Meier product-limit method for all patients using SPSS software version 18.0 (IBM Corp., Chicago, IL, USA).

Ethics statement

The prospective, multicenter clinical trial was approved by the Institutional Review Board of Korean Ministry of Food and Drug Safety (No. 12096) and recorded with ClinicalTrials.gov Identifier NCT017337346.

RESULTS

The patients were four males and four females with a median age of 56.5 years (range: 23–67), and their median Karnofsky performance scale at enrollment was 80% (range: 60–100). All patients had previously been treated with CCRT and adjuvant TMZ chemotherapy but no other chemotherapy or targeted agents. The median time between the initial GBM diagnosis and enrollment in the study was 8.75 months (range: 5.5–57).
The median cycle of CCNU and procarbazine was 3.5 (range: 2–6). Seven patients and one patient, respectively, achieved disease progression and stable disease following the administration of CCNU and procarbazine (Table 1).
Table 1

Patient characteristics

jkms-33-e167-i001
No. Age Sex Initial site Surgery CCRT Adjuvant TMZ, cycle KPS at recurrence CCNU and procarbazine, cycle Response F/U, mon Status
1 67 M Corpus callosum Biopsy Y 3 100 3 PD 35.3 Alive
2 52 F Thalamus Biopsy Y 6 60 6 SD 36.7 Alive
3 60 M Frontal Craniotomy Y 6 60 2 PD 64.0 Dead
4 23 F Parietal Craniotomy Y 5 90 6 PD 18.7 Dead
5 60 F Corpus callosum Biopsy Y 4 70 3 PD 13.0 Dead
6 50 F Temporal Craniotomy Y 2 100 2 PD 12.3 Alive
7 31 M Frontal Craniotomy Y 1 80 4 PD 9.0 Dead
8 66 M Parietal Craniotomy Y 5 70 4 PD 12.5 Dead
CCRT = concurrent chemoradiotherapy, TMZ = temozolomide, KPS = Karnofsky performance scale, F/U = follow-up, PD = progression of disease, SD = stable disease.
The median PFS for CCNU and procarbazine therapy to treat recurrent GBM was eight weeks (range: 5–73). PFS rates were 25% and 12.5% at 16 and 30 weeks, respectively, and the median OS from the start of CCNU and procarbazine administration to death was 9.7 months (95% confidence interval: 6.7–12.7, Fig. 1). The median OS from the initial diagnosis was 40 months. Serious adverse events are summarized in Table 2. Grade 3 toxicities including elevated hepatic enzyme and leukopenia developed in two patients, but they recovered following the delay of CCNU and procarbazine administration. In one patient, generalized muscle weakness developed and recovered without delay of the chemotherapy schedule. One patient had disseminated intravenous coagulation three times during follow-up period and was given conservative care.
Fig. 1

Kaplan-Meier analysis. (A) Progression-free survival. (B) Overall survival.

jkms-33-e167-g001
Table 2

Serious adverse events

jkms-33-e167-i002
Adverse events All Grade
1 2 3 4
Increased alanine aminotransferase 1 - - 1 -
Leukopenia 1 - - 1 -
Disseminated intravenous coagulation 3 - 3 - -
Generalized muscle weakness 1 - 1 - -
Total 6 - 4 2 -

DISCUSSION

In this study, the median PFS was two months in recurrent and TMZ-refractory GBM patients treated with modified CCNU and procarbazine chemotherapy; all patients had received radiotherapy with concomitant TMZ and cyclic TMZ therapy but no other cytotoxic agents or target therapies. CCNU is increasingly considered an alternative chemotherapeutic agent for GBM treatment because no other treatment has yielded better results in a controlled clinical trial, but PFS rates at six months are only in the range of 15%–25% with CCNU therapy.1112 The first randomized trial of PCV versus TMZ in chemotherapy-naïve patients with recurrent malignant glioma found that median PFS was 3.6 months with PCV and 4.7 months with TMZ (P = 0.229). In one study,13 the proportions of patients with at least one grade 3/4 adverse event were 9.2% and 12.2% in the PCV and TMZ arms (P = 0.40), respectively, and in a single-institution analysis, grade 3/4 hematological toxicity occurred in 25.6% of patients with recurrent GBM during traditional PCV chemotherapy.10
The characteristics of our protocol were that we omitted vincristine administration and we reduced the CCNU dose to increase the treatment's tolerability. Vincristine adds toxicity and CNS penetration is suboptimal,141516 and a retrospective analysis of CCNU and procarbazine versus PCV for grades 2 and 3 oligodendrogliomas presented no differences in PFS or OS.1718 A greater proportion of patients experienced neutropenia with PCV, but only those who received vincristine experienced neurotoxicity (14% vs. 0%). The NOA-05 multicenter trial analyzed the efficacy of traditional CCNU and procarbazine chemotherapy in patients with gliomatosis cerebri, and these patients achieved median PFS of 14 months. During 124 cycles of chemotherapy, authors observed grade 3/4 hematological toxicity in 15% of patients.19
There are no clinical publications that address whether CCNU and procarbazine can be substituted for PCV for newly diagnosed or recurrent GBM. We here attempted to assess the feasibility of modified CCNU and procarbazine chemotherapy for recurrent, TMZ-refractory GBM. We searched for clinical articles using the keywords “recurrent,” “glioblastoma,” “chemotherapy,” and “Korea” in PubMed (Table 3).20212223 We excluded articles about radiation therapy or radiosurgery. TMZ and the combination of ACNU and cisplatin were analyzed in three reports and one report, respectively. ACNU and cisplatin regimen had myelosuppression issues.20 Two reports analyzed the efficacy of continuous low-dose TMZ administration for recurrent and TMZ-refractory GBM. They showed different outcomes in terms of PFS and OS.2223 In the phase II RESCUE study (continuous TMZ 50 mg/m2/d), six-month PFS was 23.9% in patients with recurrent GBM.24 The efficacy of modified CCNU and procarbazine chemotherapy could be comparable with that of continuous low-dose TMZ therapy as a salvage therapy for recurrent GBM.
Table 3

Summary of clinical trials for recurrent glioblastoma treated with chemotherapeutic agents in Korea

jkms-33-e167-i003
Year No. of patients Type of study No. of institution Chemotherapy regimen Median progression free survival Median overall survival Adverse effects
2005 37 Retrospective 1 ACNU, cisplatin 6 mon 9 mon Grade 3/4 leukopenia: 41%
2006 16 Retrospective 1 Temozolomide (5 days every 28 days) 8 wk 17 wk Grade 3/4 leukopenia: 0%
2010 38 Retrospective 1 Temozolomide (daily) 17 wk 41 wk Grade 3 lymphopenia: 3 patients
2015 30 Retrospective 1 Temozolomide (daily) 8 wk 6 mon Grade 3/4 leukopenia: 0%
The present study 8 Prospective 6 Procarbazine, CCNU 8 wk 9.7 mon Grade 3 leukopenia: 1 patient
It cannot yet be determined whether the dose reduction decreased hematological toxicities because of the small number of patients and the short follow-up. Although PCV increased survival among selected patients, its toxic effects led to many dose delays and reductions during the course of treatment, whereas during a total of 30 cycles of modified CCNU and procarbazine chemotherapy, only one cycle was delayed due to leukopenia.
This is the first prospective, multicenter clinical trial for GBM approved by the Korean Food Institute Institutional Review Board. The limitation of the study is the small number of patients. Following the statistic calculation, 52 patients needed to be enrolled to achieve a difference from the historical control; however, this trial terminated before the expected end date because of the progressive deterioration of patients with recurrent GBM and slow candidate enrollment. These experiences and information should encourage clinicians and clinical researchers to suggest more challenging clinical trials in the future.
In conclusion, our findings show marginal efficacy of modified CCNU and procarbazine chemotherapy. This clinical trial suggests the need to develop treatment strategies beyond CCNU and procarbazine for recurrent, TMZ-refractory GBM.

ACKNOWLEDGMENTS

Three neuropathologists (Drs. Kim SH, Park SH, and Lee YS) independently conducted the pathology reviews.

Notes

Funding This work was supported by a fund from the National Cancer Center of Korea (No. 1120320).

Disclosure The authors have no potential conflicts of interest to disclose.

Author Contributions

  • Conceptualization: Yang SH, Kim SH, Chung DS.

  • Data curation: Kim SH, Yoo H, Chang JH, Kim CY, Chung DS.

  • Formal analysis: Yang SH, Kim SH.

  • Investigation: Kim SH, Park SH, Lee YS.

  • Resources: Kim SH, Yoo H, Chang JH, Kim CY, Chung DS, Yang SH.

  • Writing - original draft: Yang SH.

  • Writing - review & editing: Kim SH, Yoo H, Chang JH, Kim CY, Chung DS.

References

1. Kyritsis AP, Levin VA. An algorithm for chemotherapy treatment of recurrent glioma patients after temozolomide failure in the general oncology setting. Cancer Chemother Pharmacol. 2011; 67(5):971–983.
2. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005; 352(10):987–996.
3. Kreisl TN, Kim L, Moore K, Duic P, Royce C, Stroud I, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol. 2009; 27(5):740–745.
4. Franceschi E, Omuro AM, Lassman AB, Demopoulos A, Nolan C, Abrey LE. Salvage temozolomide for prior temozolomide responders. Cancer. 2005; 104(11):2473–2476.
5. Wick W, Platten M, Weller M. New (alternative) temozolomide regimens for the treatment of glioma. Neuro-oncol. 2009; 11(1):69–79.
6. Weller M, van den Bent M, Tonn JC, Stupp R, Preusser M, Cohen-Jonathan-Moyal E, et al. European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol. 2017; 18(6):e315–e329.
7. Lassman AB, Iwamoto FM, Cloughesy TF, Aldape KD, Rivera AL, Eichler AF, et al. International retrospective study of over 1000 adults with anaplastic oligodendroglial tumors. Neuro-oncol. 2011; 13(6):649–659.
8. Wick W, Roth P, Hartmann C, Hau P, Nakamura M, Stockhammer F, et al. Long-term analysis of the NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with PCV or temozolomide. Neuro-oncol. 2016; 18(11):1529–1537.
9. Wang F, Zhou F, Kruh GD, Gallo JM. Influence of blood-brain barrier efflux pumps on the distribution of vincristine in brain and brain tumors. Neuro-oncol. 2010; 12(10):1043–1049.
crossref
10. Yang SH, Hong YK, Yoon SC, Kim BS, Lee YS, Lee TK, et al. Radiotherapy plus concurrent and adjuvant procarbazine, lomustine, and vincristine chemotherapy for patients with malignant glioma. Oncol Rep. 2007; 17(6):1359–1364.
crossref
11. Batchelor TT, Mulholland P, Neyns B, Nabors LB, Campone M, Wick A, et al. Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma. J Clin Oncol. 2013; 31(26):3212–3218.
crossref
12. Weller M, Cloughesy T, Perry JR, Wick W. Standards of care for treatment of recurrent glioblastoma--are we there yet? Neuro-oncol. 2013; 15(1):4–27.
crossref
13. Brada M, Stenning S, Gabe R, Thompson LC, Levy D, Rampling R, et al. Temozolomide versus procarbazine, lomustine, and vincristine in recurrent high-grade glioma. J Clin Oncol. 2010; 28(30):4601–4608.
crossref
14. Boyle FM, Eller SL, Grossman SA. Penetration of intra-arterially administered vincristine in experimental brain tumor. Neuro-oncol. 2004; 6(4):300–305.
15. Greig NH, Soncrant TT, Shetty HU, Momma S, Smith QR, Rapoport SI. Brain uptake and anticancer activities of vincristine and vinblastine are restricted by their low cerebrovascular permeability and binding to plasma constituents in rat. Cancer Chemother Pharmacol. 1990; 26(4):263–268.
crossref
16. Levin VA. Relationship of octanol/water partition coefficient and molecular weight to rat brain capillary permeability. J Med Chem. 1980; 23(6):682–684.
crossref
17. Vesper J, Graf E, Wille C, Tilgner J, Trippel M, Nikkhah G, et al. Retrospective analysis of treatment outcome in 315 patients with oligodendroglial brain tumors. BMC Neurol. 2009; 9(1):33.
crossref
18. Webre C, Shonka N, Smith L, Liu D, De Groot J. PC or PCV, that is the question: primary anaplastic oligodendroglial tumors treated with procarbazine and CCNU with and without vincristine. Anticancer Res. 2015; 35(10):5467–5472.
19. Glas M, Bähr O, Felsberg J, Rasch K, Wiewrodt D, Schabet M, et al. NOA-05 phase 2 trial of procarbazine and lomustine therapy in gliomatosis cerebri. Ann Neurol. 2011; 70(3):445–453.
crossref
20. Gwak HS, Youn SM, Kwon AH, Lee SH, Kim JH, Rhee CH. ACNU-cisplatin continuous infusion chemotherapy as salvage therapy for recurrent glioblastomas: phase II study. J Neurooncol. 2005; 75(2):173–180.
crossref
21. Yang SH, Kim MK, Lee TK, Lee KS, Jeun SS, Park CK, et al. Temozolomide chemotherapy in patients with recurrent malignant gliomas. J Korean Med Sci. 2006; 21(4):739–744.
crossref
22. Kong DS, Lee JI, Kim JH, Kim ST, Kim WS, Suh YL, et al. Phase II trial of low-dose continuous (metronomic) treatment of temozolomide for recurrent glioblastoma. Neuro-oncol. 2010; 12(3):289–296.
crossref
23. Woo JY, Yang SH, Lee YS, Lee SY, Kim J, Hong YK. Continuous low-dose temozolomide chemotherapy and microvessel density in recurrent glioblastoma. J Korean Neurosurg Soc. 2015; 58(5):426–431.
crossref
24. Perry JR, Bélanger K, Mason WP, Fulton D, Kavan P, Easaw J, et al. Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. J Clin Oncol. 2010; 28(12):2051–2057.
crossref
TOOLS
ORCID iDs

Se-Hyuk Kim
https://orcid.org/0000-0002-7528-4007

Heon Yoo
https://orcid.org/0000-0002-9223-4300

Jong Hee Chang
https://orcid.org/0000-0003-1509-9800

Chae-Yong Kim
https://orcid.org/0000-0001-9773-5553

Dong Sup Chung
https://orcid.org/0000-0003-0690-4388

Se Hoon Kim
https://orcid.org/0000-0001-7516-7372

Sung-Hae Park
https://orcid.org/0000-0002-8681-1597

Youn Soo Lee
https://orcid.org/0000-0002-1653-6315

Seung Ho Yang
https://orcid.org/0000-0002-3490-1064

Similar articles