Open Access

Clinical feasibility of modified procarbazine and lomustine chemotherapy without vincristine as a salvage treatment for recurrent adult glioma

  • Authors:
    • Stephen Ahn
    • Young Il Kim
    • Ja Young Shin
    • Jae-Sung Park
    • Changyoung Yoo
    • Youn Soo Lee
    • Yong-Kil Hong
    • Sin-Soo Jeun
    • Seung Ho Yang
  • View Affiliations

  • Published online on: February 9, 2022     https://doi.org/10.3892/ol.2022.13234
  • Article Number: 114
  • Copyright: © Ahn et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Procarbazine, lomustine and vincristine (PCV) chemotherapy is considered a salvage option for adult glioma; however, its significant toxicities frequently lead to dose reduction or discontinuation in patients with recurrent glioma. The current study evaluated the safety and efficacy of modified procarbazine and lomustine (PC) chemotherapy that omits vincristine and reduces the lomustine dose compared with those of conventional PCV chemotherapy. Using electronic medical records, all patients with adult recurrent glioma who received PC or PCV chemotherapy between 2009 and 2020 at Seoul St. Mary's Hospital or St. Vincent's Hospital were examined retrospectively. A total of 59 patients met the eligibility criteria. Among them, 15 patients received modified PC chemotherapy (PC group) and 44 patients received PCV chemotherapy (PCV group). The PC group presented a significantly lower hematology toxicity (anemia, 6.7 vs. 45.5%, P=0.02; thrombocytopenia 20.0 vs. 70.4%, P<0.001). Additionally, the clinical impacts of PC chemotherapy, including delay of a cycle, dose reduction, discontinuation of drug(s) or total cessation of chemotherapy, were significantly less frequent compared with the PCV group (26.7 vs. 68.2%, P=0.012). The overall survival of the PC group was also significantly longer than that of PCV group (396 vs. 232 days, P=0.042), while there was no significant difference in progression‑free survival between the two groups (284.5 vs. 131 days, P=0.077). The results suggested that modified PC chemotherapy may be an alternative chemotherapeutic regimen with tolerable toxicity and without loss of clinical efficacy in patients with recurrent adult glioma. Further prospective and larger studies are required to validate our findings.

Introduction

Glioma is the most common and most malignant brain tumor in adults, composing most of all brain malignancy diagnoses in this population (1,2). Typically, the clinical outcomes of this condition are devastating, although aggressive multimodal treatments, including surgery, radiotherapy, and chemotherapy-which mainly is composed of drugs in the temozolomide (TMZ) and nitrosourea classes-can have some effect (3,4). For cases of recurrent glioblastoma, which is the most common and most malignant type of glioma, almost all patients eventually experience a recurrence and die within six months after diagnosis of recurrence (5,6).

There is no consensus regarding salvageable options for recurrent and TMZ-resistant adult glioma (710). Among the current treatments, procarbazine, lomustine, and vincristine (PCV) chemotherapy is one of the representative salvageable options for recurrent adult glioma (1114). However, the various and severe toxicities of this treatment, including hematologic toxicity from lomustine and peripheral neurotoxicity from vincristine, often result in its reduction or discontinuation in glioma patients (1517). In addition to its toxicity, vincristine is composed of relatively heavy molecules (825 Daltons), and concerns exist about its successful crossing of the blood-brain barrier (18).

In this context, a few studies have suggested procarbazine and lomustine (PC) chemotherapy without vincristine as an alternative to PCV chemotherapy with lesser toxicity and no loss of efficacy (1922). Vesper et al first suggested that PC chemotherapy protocols might be as effective as PCV chemotherapy while avoiding the toxicity of vincristine (20). In addition, Webre et al suggested that PC chemotherapy can achieve comparable clinical outcomes to PCV chemotherapy with lesser neurotoxicity in anaplastic oligodendroglioma (21). We have also tested the modified PC chemotherapy protocol of reduced dose of lomustine (75 mg/m2, day 1) and procarbazine (60 mg/m2, days 11–24) every four weeks in recurrent glioblastoma patients with expectation of lesser toxicity and non-inferior efficacy (19).

In this study, we retrospectively analyzed the efficacy and safety of modified PC chemotherapy in recurrent glioma patients compared with those of conventional PCV chemotherapy. This study tried to validate that the modified PC chemotherapy is less toxic than PCV chemotherapy, and that the survival outcomes of PC chemotherapy are noninferior to those of PCV chemotherapy in patients with recurrent adult glioma.

Materials and methods

Study population

This retrospective study was approved by the institutional review board of our institution. The electronic medical records of adult glioma patients treated at our institution between 2010 and 2020 were examined. The study inclusion criteria were i) glioma pathologically confirmed by craniotomy or biopsy, ii) glioma recurrence confirmed radiologically and/or pathologically, iii) receipt of PC or PCV chemotherapy following recurrence diagnosis, and iv) accessible baseline clinical variables and survival data. The study exclusion criteria were i) received PC or PCV chemotherapy as adjuvant therapy after initial diagnosis, ii) a medical history of hematologic or rheumatologic disease, iii) failure to complete the first cycle of PC or PCV chemotherapy. A flow of the study design is presented in Fig. 1.

Treatment protocols

After maximal safe resection at initial surgery, we performed adjuvant therapy following the best treatment protocol(s) by glioma subtype. If the diagnosis was glioblastoma, we performed concomitant chemoradiation (TMZ dose: 75 mg/m2) and six cycles of adjuvant TMZ chemotherapy (TMZ dose: 150–200 mg/m2). If the diagnosis was grade II or III glioma, we conducted adjuvant radiotherapy, in which the dosage was either 5,940 cGy for 33 fractions or 6,000 cGy for 30 fractions. In the case of grade III glioma, we added adjuvant chemotherapy of PCV or PC chemotherapy. When recurrence occurred, the first chemotherapy considered was TMZ; when the use of TMZ chemotherapy was not possible due to various reasons, such as prior history of TMZ administration within 6 months, and swallowing difficulty, then bevacizumab or nitrosourea-based chemotherapy including the modified PC chemotherapy or the conventional PC chemotherapy, was considered according to clinician preference.

PC chemotherapy was composed of lomustine (75 mg/m2, day 1) and procarbazine (60 mg/m2, days 11–24) administered orally every four weeks. This modified protocol was discussed in a previous study of one of our authors (19). PCV chemotherapy was administered to recurrent glioma patients, with lomustine (110 mg/m2, day 1) and procarbazine (60 mg/m2, days 8–21) administered orally but vincristine administered intravenously [1.4 mg/m2 (maximum of 2 mg), days 8 and 29] every six weeks.

Clinical variables

The clinical variables of sex; age; pathological diagnosis, including molecular features; prior history of surgery, radiation, or chemotherapy; radiological findings; performance status; and survival status and/or death date were collected. Diagnosis of recurrent glioma was performed by two neuropathologists according to the 2016 World Health Organization classification of the central nervous system. IDH mutation was evaluated by immunohistochemistry or directing sequencing. If necessary, IDH 2 mutation was evaluated by directing sequencing. The presence of a 1p19q co-deletion was examined using fluorescence in situ hybridization. The O6-methylguanine-DNA-methyltransferase (MGMT) gene methylation status was evaluated by polymerase chain reaction. Performance status was estimated according to the scale of the Eastern Cooperative Oncology Group (ECOG). All kinds of toxicities were evaluated according to the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. Also, the related impact of the toxicity of PC or PCV chemotherapy on the course of the treatment schedule was classified into four categories: delay of a cycle, dose reduction, discontinuation of drug(s), or total cessation of chemotherapy. Radiographic responses on magnetic resonance imaging (MRI) were determined by specialized neuroradiologists according to the response assessment in neuro-oncology (RANO) criteria. The date of recurrence was defined as the date of MRI showing recurrence. Survival status and/or death date were collected from the Korea Central Cancer Registry database.

Statistical analysis

The overall survival (OS) after recurrence was defined as days from the starting date of PC or PCV chemotherapy to death, while progression-free survival (PFS) was defined as days from the starting date of PC or PCV chemotherapy to disease progression was confirmed by MRI. Patients who were confirmed to be alive on March 31, 2021, were censored. The mean duration of follow-up was 424.6 days (range: 55–2,491 days). All clinical variables were considered with descriptive statistics. The differences of clinical variables between the two treatment groups were compared using Fisher's exact test or the Chi-square test. The normality test was performed for continuous variables. Kaplan-Meier survival analysis and the log-rank test were used to calculate the median OS and PFS values of the groups. Univariate and multivariate analyses were conducted using a Cox proportional regression model. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated. Multivariate analysis was performed on the variables with P-values <0.2, and P-values <0.05 were considered to indicate statistical significance. All statistical analyses were conducted using the R version 4.0.5 software program (R Foundation for Statistical Computing, Vienna, Austria).

Results

Patient characteristics

Among a total of 59 patients enrolled in this study, 15 received PC chemotherapy (PC group) and 44 patients received PCV chemotherapy (PCV group) as salvage treatment for recurrent gliomas. Clinical characteristics, including sex, age, initial diagnosis, IDH mutation, 1p19q co-deletion, MGMT promoter methylation, and prior history of radiotherapy were not statistically different between the two groups. However, the median interval from radiation to chemotherapy in the PC group was shorter than that of the PCV group [4.0 (range: 0–42) months vs. 22.0 (range: 0–167) months, P=0.004]. Also, fewer patients in the PC group had prior history of any chemotherapy (33.3 vs. 84.7%; P=0.003), while there was no difference in the median interval from the last chemotherapy session to initiation of PC or PCV chemotherapy between the two groups. The summarized baseline characteristics of these patients are described in Table I.

Table I.

Baseline characteristics of patients with recurrent adult glioma who received PC or PCV chemotherapy.

Table I.

Baseline characteristics of patients with recurrent adult glioma who received PC or PCV chemotherapy.

CharacteristicPC group (n=15)PCV group (n=44)P-value
Male sex, n (%)11 (73.3)27 (61.4)0.600
Age at chemotherapy (years), n (range)52.2 (20–79)49.6 (21–73)0.528
Initial diagnosis, n (%) >0.999
  GBM5 (33.3)14 (31.8)
  Non-GBM10 (66.7)30 (68.2)
IDH mutation, n (%) 0.103
  Yes3 (20.0)13 (29.5)
  No10 (66.7)31 (70.5)
  Unknown2 (13.3)0 (0.0)
1p19q co-deletion, n (%) 0.392
  Yes2 (13.3)7 (15.9)
  No11 (73.3)24 (54.5)
  Unknown2 (13.3)13 (29.5)
MGMT methylation, n (%) 0.311
  Yes8 (53.4)16 (36.4)
  No4 (26.7)16 (36.4)
  Unknown3 (20.0)12 (27.3)
Prior radiation therapy, n (%) >0.999
  Yes15 (100.0)43 (97.7)
  No0 (0.0)1 (2.3)
Median interval from radiation to PC or PCV (months),4.0 (0–42)22.0 (0–167)0.004
n (range)
Prior chemotherapy, n (%) <0.003
  Never10 (66.7)7 (16.3)
  TMZ5 (33.3)30 (69.8)
  TMZ, bevacizumab0 (0.0)1 (2.3)
  TMZ, PCV0 (0.0)5 (11.6)
Median interval from last chemotherapy to PC or PCV1.0 (0–13)2.0 (0–61)0.610
(months), n (range)
ECOG score, n (%) 0.311
  0-17 (46.7)29 (65.9)
  ≥28 (53.3)15 (34.1)

[i] ECOG, Eastern Cooperative Oncology Group; GBM, glioblastoma; MGMT, O6-methylguanine-DNA-methyltransferase; PC, procarbazine and lomustine; PCV, procarbazine, lomustine and vincristine; TMZ, temozolomide.

Toxicity experiences and the impact of toxicity on chemotherapy schedule

The PC group presented a significantly lower hematology toxicity profile (anemia: 6.7 vs. 45.5%; P=0.017 and thrombocytopenia: 20.0 vs. 70.4%; P<0.001) and liver function (elevated liver enzymes: 0 vs. 25.0%; P=0.078). CTCAE grade III or IV toxicities were less frequently observed in the PC group, although there was no statistically significant difference. Rates of other toxicities, including neutropenia, kidney injury, allergic skin reactions, and peripheral neurotoxicity, were not significantly different between the two groups. Detailed information about toxicities is presented in Table II.

Table II.

Toxicity in patients with recurrent adult glioma who received PC or PCV chemotherapy.

Table II.

Toxicity in patients with recurrent adult glioma who received PC or PCV chemotherapy.

All gradesaGrades III and IVa


Toxic indicatorPC group (n=15)PCV group (n=44)P-valuePC group (n=15)PCV group (n=44)P-value
Anemia, n (%)1 (6.7)20 (45.5)0.0171 (6.7)6 (13.6)0.796
Neutropenia, n (%)3 (20.0)17 (38.6)0.3173 (20.0)8 (18.2)>0.999
Thrombocytopenia, n (%)3 (20.0)31 (70.4)<0.0013 (20.0)14 (31.8)0.587
Elevated liver enzymes, n (%)0 (0.0)11 (25.0)0.0780 (0.0)6 (13.6)0.310
Elevated creatinine, n (%)0 (0.0)2 (4.6)0.9890 (0.0)1 (2.3)>0.999
Allergic reaction, n (%)0 (0.0)1 (2.3)>0.9990 (0.0)0 (0.0)>0.999
Peripheral neurotoxicity, n (%)0 (0.0)5 (11.4)0.4080 (0.0)0 (0.0)>0.999

a Toxicities were graded according to CTCAE version 5.0. PC, procarbazine and lomustine; PCV, procarbazine, lomustine and vincristine.

We describe the adverse impacts of toxicity on chemotherapy schedule in Table III. The PC group significantly less frequently experienced any of delay of cycle, dose reduction, discontinuation of one of the chemotherapeutic drugs, or cessation of the entire chemotherapy regimen than did the PCV group (26.7 vs. 68.2%; P=0.012). Each type of toxicity was less frequently observed in the PC group, although this result failed to show statistical significance.

Table III.

Adverse impacts of the toxicity of PC or PCV on the course of chemotherapy for recurrent adult glioma.

Table III.

Adverse impacts of the toxicity of PC or PCV on the course of chemotherapy for recurrent adult glioma.

Adverse impactPC group (n=15)PCV group (n=44)P-value
Total, n (%)4 (26.7)30 (68.2)0.012
Delay of a cycle, n (%)3 (20.0)14 (31.8)0.587
Dose reduction, n (%)1 (6.7)6 (13.6)0.796
Drug discontinuation, n (%)0 (0.0)8 (18.2)0.180
Chemotherapy cessation, n (%)0 (0.0)2 (4.5)0.989

[i] PC, procarbazine and lomustine; PCV, procarbazine, lomustine and vincristine.

Comparison of clinical outcomes between the PC and PCV groups

The OS of the PC group was significantly longer than that of the PCV group (396 vs. 232 days; P=0.042), while there was no significant difference in PFS between the two groups (284.5 vs. 131 days; P=0.077). The Kaplan-Meier survival curves of OS and PFS in the two groups are illustrated in Fig. 2. Univariate and multivariate Cox analyses for OS were performed and are described in Table IV. In multivariate analysis for OS, the PCV group (HR 2.09 CI, 1.07–4.25, P=0.023) and older age (≥65) (HR 3.12 CI, 1.12–8.66, P=0.029) were associated with inferior OS, while presence of 1p19q co-deletion (HR 0.34 CI, 0.13–50.87, P=0.024) was associated with superior OS.

Table IV.

Univariate and multivariate Cox regression analysis for overall survival.

Table IV.

Univariate and multivariate Cox regression analysis for overall survival.

Univariate analysisMultivariate analysisa


VariablesHazard ratio (95% CI)P-valueHazard ratio (95% CI)P-value
Male vs. female1.04 (0.58, 1.86)0.903
Age ≥65 vs. <65 (years)3.45 (1.49, 8.00)0.0043.12 (1.12, 8.66)0.029
Non-glioblastoma vs. glioblastoma0.60 (0.33, 1.07)0.0810.66 (0.32, 1.35)0.251
IDH mutated vs. non-mutated0.45 (0.22, 0.93)0.032  0.73 (0.58, 3.26)0.472
1p19q co-deleted vs. not-deleted0.35 (0.13, 0.90)0.030  0.34 (0.13, 0.87)0.024
MGMT methylated vs. non-methylated0.45 (0.21, 0.98)0.0450.26 (0.08, 0.86)0.028
Prior history of chemotherapy vs. no1.61 (0.84, 3.11)0.1520.78 (0.35, 1.76)0.557
history of chemotherapy
Prior history of radiotherapy2.67 (0.36, 19.58)0.330
vs. no history of radiotherapy
PCV group vs. PC group2.06 (1.01, 4.18)0.0462.09 (1.07, 4.25)0.023

a Multivariable analysis was performed on the variables with P-values threshold <0.2. CI, confidence interval; IDH, isocitrate dehydrogenase 1; PC, procarbazine and lomustine; PCV, procarbazine, lomustine, and vincristine.

Discussion

In our retrospective and comparative study, we tried to compare the safety and the efficacy of our modified PC chemotherapy, compared to those of PCV chemotherapy. We also tried to evaluate how the toxicity of these chemotherapies affected the course of chemotherapy in patients with recurrent adult glioma. Our findings showed that anemia and thrombocytopenia were significantly more frequent in PCV groups than in the PC groups (anemia: 45.5 vs. 6.7%; P=0.017 and thrombocytopenia: 70.4 vs. 20.0%; P<0.001, respectively). Anemia of higher than CTCAE grade III was also more frequent in the PCV group than the PC group. Peripheral neurotoxicity, which is a major concern with vincristine, was not observed in the PC group, while it was observed in 11.4% of patients in the PCV group. In addition, frequent and severe adverse events in the PCV group also resulted in greater disruption to the course of chemotherapy, such as delay of a cycle, dose reduction, discontinuation of vincristine, and cessation of salvage chemotherapy (68.2 vs. 26.7%; P=0.012). In contrast, regarding concerns about inferior efficacy when omitting vincristine, our findings suggest that survival outcomes were not different between the two groups. Interestingly, the OS of the PC group was significantly superior to that of the PCV group (396 vs. 232 days; P=0.042), while the PFS of the PC group was not different from that of the PCV group (284.5 vs. 131 days; P=0.077). This may be explained by numerous studies showing that the occurrence of less toxicity after chemotherapy correlates with better prognosis (23,24). In summary, our modified PC chemotherapy, which omitted vincristine and reduced the dose of lomustine, showed lower toxicity and non-inferior efficacy for adult recurrent glioma patients compared to those of conventional PCV chemotherapy.

There were significant differences regarding prior history of chemotherapy and the interval from radiation to chemotherapy between the two groups, although several baseline characteristics, including initial diagnosis, molecular features, and prior history of radiotherapy, were not significantly different between the two groups. In detail, significantly fewer patients in the PC group had a prior history of any chemotherapy and in the PC group, 5 patients were diagnosed with primary glioblastoma at initial diagnosis, and these patients received concomitant chemoradiation therapy followed by TMZ. Regarding to the difference of the median interval from radiation to chemotherapy, we thought that the PCV group had a longer period of stable state after radiation compared that of the PC group. Another explanation is that the PC group received earlier chemotherapy as salvage treatment than did the PCV group.

When considering chemotherapeutic drugs for recurrent glioma, there have been options identified to date, including TMZ rechallenged or continuously administered with low-dose, bevacizumab, and PCV-based chemotherapy (4,2528). As a salvage therapy after TMZ for recurrent glioma, numerous clinical trials have assessed the efficacy of PCV-based chemotherapy (11,12,14). However, in clinics, toxicities including hematologic, neurologic, liver, kidney, and skin problems were diagnosed frequently and sometimes very severe, which is a major hindrance when choosing PCV chemotherapy as salvage therapy for recurrent glioma patients, especially in those who are elderly or with a lower performance status (15,29). In addition, there have been concerns about the efficacy of vincristine because its molecular weight (825 Daltons) might be too high to penetrate the blood-brain barrier (18). In this context, a few studies have put forth the idea of adopting a modified PC-based chemotherapy regimen without vincristine for glioma patients (1922). Vesper et al retrospectively analyzed clinical outcomes and toxicities of 315 patients with oligodendroglial brain tumors who received PCV or PC chemotherapy as adjuvant treatment after surgical resection and radiation. Their study showed that the PFS of patients who received PC chemotherapy was not different from that of patients who received PCV chemotherapy, with significantly fewer hematologic and neurological toxicities (20). Webre et al also evaluated 97 patients with primary anaplastic oligodendroglioma who received PCV or PC chemotherapy as adjuvant treatment, reporting that the clinical outcomes of PC chemotherapy for primary anaplastic oligodendroglial tumors were not different from those of patients who received PCV chemotherapy, with lower hematologic toxicities (21).

In accordance with two previous studies exploring the use of PC chemotherapy in primary oligodendroglial patients (20,21), we added evidence that our modified PC chemotherapy is as beneficial as PCV chemotherapy but with significantly less toxicity due to omission of vincristine and reduction of the dose of lomustine. Taken together, we suggest that PC chemotherapy can be an alternative option to PCV chemotherapy, especially for use in patients expected to be intolerable to PCV chemotherapy, including elderly patients or those with lower performance.

Our study should be considered within the scope of several limitations. First, our study included heterogeneous recurrent gliomas, and the unknown molecular status of 1p19q co-deletion in about 30% of these patients could cause severe bias. Second, although several baseline characteristics, including initial diagnosis, molecular features, and prior history of radiotherapy, were not significantly different between the two groups, there were significant differences regarding prior history of chemotherapy and the interval from radiation to chemotherapy between the two groups, which can cause several biases in both toxicity profile and clinical outcomes. Third, this is not a randomized study, and selection bias about treatment group have affected the results. Although our institution tried to minimize clinician biases through multi-disciplinary discussion, treatment characteristics could not be identical between the two groups. Fourth, due to the retrospective nature of this study, not all adverse reactions were considered. Fifth, the number of patients is insufficient to draw a strong conclusion. Therefore, further prospective and larger studies are needed to validate whether PC chemotherapy could be an alternative to PCV chemotherapy as a secondary salvage option for recurrent glioma patients.

In conclusion, this study showed significantly fewer toxicities after our modified PC chemotherapy than after PCV chemotherapy in recurrent glioma patients. The OS and PFS of the modified PC chemotherapy were noninferior to those of PCV chemotherapy. Further prospective and larger studies are needed to validate the modified PC chemotherapy without vincristine as an alternative option for the conventional PCV chemotherapy for recurrent glioma patients.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datsets used and/or analyzed are available from the corresponding author on reasonable request.

Authors' contributions

SA and YIK wrote the manuscript. SA, YIK, JSP, JYS, CY, and YSL collected and analyzed the data. JSP, CY, YSL, YKH, SSJ and SHY supervised the current study. JSP, CY, YSL, YKH, SSJ and SHY wrote, reviewed and edited the manuscript. YKH, SSJ and SHY conceptualized the present study. SA, YIK, and JSP confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The Institutional Review Board of Seoul St. Mary's Hospital approved the current study (approval no. XC21RIDI0089). Due to the retrospective manner of the study, the requirement for informed consent to patriciate was waived.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ostrom QT, Cioffi G, Gittleman H, Patil N, Waite K, Kruchko C and Barnholtz-Sloan JS: CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2012–2016. Neuro Oncol. 21 (Suppl 5):v1–v100. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Lapointe S, Perry A and Butowski NA: Primary brain tumours in adults. Lancet. 392:432–446. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Molinaro AM, Taylor JW, Wiencke JK and Wrensch MR: Genetic and molecular epidemiology of adult diffuse glioma. Nat Rev Neurol. 15:405–417. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, Bendszus M, Balana C, Chinot O, Dirven L, et al: EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol. 18:1702021. View Article : Google Scholar : PubMed/NCBI

5 

Stupp R, Hegi ME, Mason WP, Van Den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al: Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-Year analysis of the EORTC-NCIC trial. Lancet Oncol. 10:459–466. 2009. View Article : Google Scholar : PubMed/NCBI

6 

van Linde ME, Brahm CG, de Witt Hamer PC, Reijneveld JC, Bruynzeel AME, Vandertop WP, van de Ven PM, Wagemakers M, van der Weide HL, Enting RH, et al: Treatment outcome of patients with recurrent glioblastoma multiforme: A retrospective multicenter analysis. J Neurooncol. 135:183–192. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Wen PY, Weller M, Lee EQ, Alexander BM, Barnholtz-Sloan JS, Barthel FP, Batchelor TT, Bindra RS, Chang SM, Chiocca EA, et al: Glioblastoma in adults: A society for neuro-oncology (SNO) and European society of neuro-oncology (EANO) consensus review on current management and future directions. Neuro Oncol. 22:1073–1113. 2020. View Article : Google Scholar : PubMed/NCBI

8 

Alexander BM and Cloughesy TF: Adult glioblastoma. J Clin Oncol. 35:2402–2409. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Weller M and Le Rhun E: How did lomustine become standard of care in recurrent glioblastoma? Cancer Treat Rev. 87:1020292020. View Article : Google Scholar : PubMed/NCBI

10 

McDuff SGR, Dietrich J, Atkins KM, Oh KS, Loeffler JS and Shih HA: Radiation and chemotherapy for high-risk lower grade gliomas: Choosing between temozolomide and PCV. Cancer Med. 9:3–11. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Schmidt F, Fischer J, Herrlinger U, Dietz K, Dichgans J and Weller M: PCV chemotherapy for recurrent glioblastoma. Neurology. 66:587–589. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Happold C, Roth P, Wick W, Steinbach JP, Linnebank M, Weller M and Eisele G: ACNU-based chemotherapy for recurrent glioma in the temozolomide era. J Neurooncol. 92:45–48. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Brada M, Stenning S, Gabe R, Thompson LC, Levy D, Rampling R, Erridge S, Saran F, Gattamaneni R, Hopkins K, et al: Temozolomide versus procarbazine, lomustine, and vincristine in recurrent high-grade glioma. J Clin Oncol. 28:4601–4608. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Parasramka S, Talari G, Rosenfeld M, Guo J and Villano JL: Procarbazine, lomustine and vincristine for recurrent high-grade glioma. Cochrane Database Syst Rev. 7:CD0117732017.PubMed/NCBI

15 

Jutras G, Bélanger K, Letarte N, Adam JP, Roberge D, Lemieux B, Lemieux-Blanchard É, Masucci L, Ménard C, Bahary JP, et al: Procarbazine, lomustine and vincristine toxicity in low-grade gliomas. Curr Oncol. 25:e33–e39. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Keogh RJ, Aslam R, Hennessy MA, Coyne Z, Hennessy BT, Breathnach OS, Grogan L and Morris PG: One year of procarbazine lomustine and vincristine is poorly tolerated in low grade glioma: A real world experience in a national neuro-oncology centre. BMC Cancer. 21:1402021. View Article : Google Scholar : PubMed/NCBI

17 

Park SB, Goldstein D, Krishnan AV, Lin CS, Friedlander ML, Cassidy J, Koltzenburg M and Kiernan MC: Chemotherapy-induced peripheral neurotoxicity: A critical analysis. CA Cancer J Clin. 63:419–437. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Wang F, Zhou F, Kruh GD and Gallo JM: Influence of blood-brain barrier efflux pumps on the distribution of vincristine in brain and brain tumors. Neuro Oncol. 12:1043–1049. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Kim SH, Yoo H, Chang JH, Kim CY, Chung DS, Kim SH, Park SH, Lee YS and Yang SH: Procarbazine and CCNU chemotherapy for recurrent glioblastoma with MGMT promoter methylation. J Korean Med Sci. 33:e1672018. View Article : Google Scholar : PubMed/NCBI

20 

Vesper J, Graf E, Wille C, Tilgner J, Trippel M, Nikkhah G and Ostertag C: Retrospective analysis of treatment outcome in 315 patients with oligodendroglial brain tumors. BMC Neurol. 9:332009. View Article : Google Scholar : PubMed/NCBI

21 

Webre C, Shonka N, Smith L, Liu D and 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. 35:5467–5472. 2015.PubMed/NCBI

22 

Yang SH, Hong YK, Yoon SC, Kim BS, Lee YS, Lee TK, Lee KS, Jeun SS, Kim MC and Park CK: Radiotherapy plus concurrent and adjuvant procarbazine, lomustine, and vincristine chemotherapy for patients with malignant glioma. Oncol Rep. 17:1359–1364. 2007.PubMed/NCBI

23 

Cha JY, Park JS, Hong YK, Jeun SS and Ahn S: Impact of body mass index on survival outcome in patients with newly diagnosed glioblastoma: A retrospective single-center study. Integr Cancer Ther. 20:15347354219912332021. View Article : Google Scholar : PubMed/NCBI

24 

Trestini I, Carbognin L, Bonaiuto C, Tortora G and Bria E: The obesity paradox in cancer: Clinical insights and perspectives. Eat Weight Disord. 23:185–193. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Seystahl K, Hentschel B, Loew S, Gramatzki D, Felsberg J, Herrlinger U, Westphal M, Schackert G, Thon N, Tatagiba M, et al: Bevacizumab versus alkylating chemotherapy in recurrent glioblastoma. J Cancer Res Clin Oncol. 146:659–670. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Cai Y, Jiang YG, Wang M, Jiang ZH and Tan ZG: A comparative study of the effectiveness and safety of combined procarbazine, lomustine, and vincristine as a therapeutic method for recurrent high-grade glioma: A protocol for systematic review and meta-analysis. Medicine (Baltimore). 99:e222382020. View Article : Google Scholar : PubMed/NCBI

27 

Toft A, Urup T, Christensen IJ, Michaelsen SR, Lukram B, Grunnet K, Kosteljanetz M, Larsen VA, Lassen U, Broholm H and Poulsen HS: Biomarkers in recurrent grade III glioma patients treated with bevacizumab and irinotecan. Cancer Invest. 36:165–174. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Wick W and Winkler F: Regimen of procarbazine, lomustine, and vincristine versus temozolomide for gliomas. Cancer. 124:2674–2676. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Tabouret E, Reyes-Botero G, Dehais C, Daros M, Barrie M, Matta M, Petrirena G, Autran D, Duran A, Bequet C, et al: Relationships between dose intensity, toxicity, and outcome in patients with oligodendroglial tumors treated with the PCV regimen. Anticancer Res. 35:2901–2908. 2015.PubMed/NCBI

Related Articles

Journal Cover

April-2022
Volume 23 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ahn S, Kim YI, Shin JY, Park J, Yoo C, Lee YS, Hong Y, Jeun S and Yang SH: Clinical feasibility of modified procarbazine and lomustine chemotherapy without vincristine as a salvage treatment for recurrent adult glioma. Oncol Lett 23: 114, 2022
APA
Ahn, S., Kim, Y.I., Shin, J.Y., Park, J., Yoo, C., Lee, Y.S. ... Yang, S.H. (2022). Clinical feasibility of modified procarbazine and lomustine chemotherapy without vincristine as a salvage treatment for recurrent adult glioma. Oncology Letters, 23, 114. https://doi.org/10.3892/ol.2022.13234
MLA
Ahn, S., Kim, Y. I., Shin, J. Y., Park, J., Yoo, C., Lee, Y. S., Hong, Y., Jeun, S., Yang, S. H."Clinical feasibility of modified procarbazine and lomustine chemotherapy without vincristine as a salvage treatment for recurrent adult glioma". Oncology Letters 23.4 (2022): 114.
Chicago
Ahn, S., Kim, Y. I., Shin, J. Y., Park, J., Yoo, C., Lee, Y. S., Hong, Y., Jeun, S., Yang, S. H."Clinical feasibility of modified procarbazine and lomustine chemotherapy without vincristine as a salvage treatment for recurrent adult glioma". Oncology Letters 23, no. 4 (2022): 114. https://doi.org/10.3892/ol.2022.13234