Methotrexate and gemcitabine combination chemotherapy for the treatment of malignant pleural mesothelioma

  • Authors:
    • Kozo Kuribayashi
    • Shigeru Miyata
    • Kazuya Fukuoka
    • Aki Murakami
    • Syusai Yamada
    • Kunihiro Tamura
    • Noriko Hirayama
    • Takayuki Terada
    • Chiharu Tabata
    • Yoshihiro Fujimori
    • Takashi Nakano
  • View Affiliations

  • Published online on: May 10, 2013     https://doi.org/10.3892/mco.2013.118
  • Pages: 639-642
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Malignant pleural mesothelioma (MPM) is an aggressive tumor of serosal surfaces with a poor prognosis. Methotrexate and gemcitabine have exhibited single-agent activity in MPM. We evaluated the feasibility of sequential administration of these agents in the treatment of MPM. A total of 21 patients with MPM received a 30‑min infusion of 100 mg/m2 methotrexate and, 30 min later, a 30‑min infusion of 800 mg/m2 gemcitabine. Twenty-four hours following the administration of methotrexate, leucovorin rescue therapy was initiated (10 mg̸m2 leucovorin administered 4 times at 6‑h intervals). These treatments were administered weekly, with 4 weekly administrations constituting a cycle of therapy. A total of 88 cycles were administered to the 21 patients, with each patient receiving 1‑10 cycles (median, 4.2 cycles). Eight patients (38.1%) exhibited a partial response, 10 patients (47.6%) had stable disease and 3 patients (14.3%) had progressive disease. The median overall survival was 19.4 months (range, 02-41 months). One-year and 2‑year survival rates were 61.9 and 38.1%, respectively. Hematological toxicity was considered acceptable, with grade 3‑4 toxicities occurring in 3 (14.3%) patients. Non‑hematologic toxicity was generally mild. There was no treatment‑related mortality. Our results suggest that methotrexate and gemcitabine combination therapy is feasible and effective in the treatment of MPM. This regimen may offer an alternative to platinum-based chemotherapy and a prospective trial including a larger cohort of patients is recommended to confirm these results.

Introduction

Malignant pleural mesothelioma (MPM) arises from the mesothelial surface of the pleural cavity and is a locally invasive tumor with poor prognosis (1,2). In >70% of patients, the tumor is associated with exposure to asbestos fibers following a long latent period of 20–50 years (3). The incidence of mesothelioma is rare in the general population; however, it is expected to increase in the next 20 years in industrialized countries as a result of past asbestos use (4,5).

MPM is refractory to the currently available treatment options. The efficacy of surgical therapy has not been precisely defined (6) and radiotherapy may be palliative but does not prolong survival (7). For the majority of patients with MPM, systemic chemotherapy remains the standard of care (8). Prior to 2003, the majority of studies on chemotherapy for MPM were conducted using either single agents or combination regimens in the setting of small phase II trials. The results demonstrated <20% of tumor regression with no significant effect on patient survival, which was 6–9 months (8,9). Since 2003, the combination of cisplatin and pemetrexed (PTX) has been used as standard chemotherapy for MPM (10). This was based on a randomized phase III study in which PTX plus cisplatin achieved a response rate of 41.3% and a median survival of 12.1 months, compared to 16.7% response rate and 9.3-month median survival achieved by cisplatin alone (10).

In this study, a non-platinum-based combination therapy with two anti-metabolites (methotrexate and gemcitabine) was devised. Methotrexate is an analogue of folic acid known to be effective against breast cancer, lymphoblastic leukemia and osteosarcoma (11,12). Gemcitabine is a pyrimidine analogue, effective against a wide range of solid tumors, including pancreatic carcinoma and non-small cell lung carcinoma (13). Methotrexate and gemcitabine have been reported to exhibit single-agent activity in MPM (8,9); however, the combined administration of these agents has not yet been investigated.

In the present study, we evaluated the feasibility and efficacy of methotrexate and gemcitabine combination therapy in the treatment of MPM, through the analysis of toxicity, response and survival data.

Patients and methods

Patients

Patients with histologically confirmed MPM who had previously received 0–1 chemotherapy cycles, not including gemcitabine and methotrexate, were considered eligible for this single-center study. Tumor extension was classified according to the tumor-node-metastasis (TNM) staging system developed by the International Mesothelioma Interest Group (IMIG) (14). Patients were 18–75 years of age, with an Eastern Cooperative Oncology Group (ECOG) performance status of 0–2, had adequate bone marrow function (hemoglobin concentration ≥10 g/dl, total leukocyte count ≥3.0×109/l, granulocyte count ≥1.5×109/l and platelet count ≥100×109/l), adequate renal function (serum creatinine level <1.5 mg/dl) and adequate hepatic function (total bilirubin level ≤1.5 times the upper limit of normal and serum alanine transferase and alkaline phosphatase levels ≤3 times the upper limit of normal). Patients with a concurrent malignancy of another type or symptoms and/or signs of metastases in the central nervous system were excluded. Patients with prior surgery were considered eligible. This study was approved by the Institutional Review Board of Hyogo College of Medicine and informed consent was obtained from each patient.

Treatment

Patients received a 30-min intravenous (i.v.) infusion of 100 mg/m2 methotrexate and, 30 min later, a 30-min i.v. infusion of 800 mg/m2 gemcitabine. For leucovorin rescue, calcium leucovorin (10 mg/m2, p.o. or i.v.) was administered 4 times at 6-h intervals, initiated 24 h after the administration of methotrexate. These treatments were administered weekly, with 4 treatments constituting a cycle of therapy. A maximum of 6 cycles were administered, unless therapy was terminated due to tumor progression, patient death or wish of treatment discontinuation, or in the presence of convincing evidence that further treatment was not beneficial. Antiemetic and symptomatic treatments were permitted. Analyses of blood cell count and chemistry were performed weekly. Treatment was delayed in the case of i) absolute neutrophil count <1.5×109/l and/or platelet count <100×109/l; ii) any grade 3 or 4 non-hematological toxicity (except for nausea/vomiting) that did not resolve to grade 1 or less. If these toxicities were not resolved within the cycle, the dose was reduced to 75% of the previous dose level for the next cycle.

Response and toxicity criteria

Chest imaging by computed tomography (CT) was performed at baseline, following completion of every other treatment cycle and every 8weeks following completion of therapy. Objective response was evaluated and calculated using the modified Response Evaluation Criteria in Solid Tumors (RESIST) criteria for MPM (15). Treatment-related toxicities were evaluated according to the National Cancer Institute Common Toxicity Criteria version 3.0 (16).

Statistical analysis

Survival was calculated as the time period from treatment initiation to death, using the Kaplan-Meier method (17).

Results

Patient characteristics

The characteristics of the 21 eligible patients are listed in Table I. There were 16 males and 5 females, with a median age of 63 years (range, 51–75 years). The histological pattern of MPM was epithelial in 17 cases, sarcomatous in 3 cases and biphasic in 1 case. Nineteen patients (90.4%) had stage III and IV disease according to the IMIG staging system at the time of enrollment. Thirteen patients (61.9%) had an ECOG performance status of 0 or 1.

Table I

Patient characteristics.

Table I

Patient characteristics.

CharacteristicsNo. (%)
Gender
  Male16 (76.2)
  Female5 (23.8)
Age, years
  Median63
  Range51–75
Performance status
  01 (4.8)
  112 (57.1)
  28 (38.1)
IMIG stage
  Ib1 (4.8)
  II1 (4.8)
  III4 (19.0)
  IV15 (71.4)
Histological subtype
  Epithelial17 (81.0)
  Sarcomatous3 (14.3)
  Biphasic1 (4.7)
Previous treatment
  None10 (47.6)
  Surgery2 (9.5)
  Chemotherapy9 (42.9)
Asbestos exposure
  Yes13 (61.9)
  No8 (38.1)

[i] IMIG, International Mesothelioma Interest Group.

Responses to treatment

A total of 88 cycles were administered to the 21 patients. Each patient received a median 4.2 cycles (range, 2–10 cycles). Response to chemotherapy is shown in Table II. No patients exhibited a complete response. Eight patients (38.1%) exhibited a partial response. According to the histological pattern, a PR was observed in 6 out of the 17 patients with epithelial type and in 2 out of the 3 patients with sarcomatous type MPM. Out of the total 21 patients, 10 (47.6%) had stable disease and 3 (14.3%) had progressive disease with no period of stabilization.

Table II

Response to chemotherapy and histologic subtype.

Table II

Response to chemotherapy and histologic subtype.

Histologic subtype
ResponseOverall no. (%)Epithelial no. (n)Sarcomatous no. (n)Biphasic no. (n)
Complete response0000
Partial response8 (38.1)620
Stable disease10 (47.6)811
Progressive disease3 (14.3)300
Toxicity

The toxicity observed in each patient is shown in Table III. There was no treatment-related mortality. The most frequently observed hematological side effects were neutropenia and thrombocytopenia. Grade 3–4 hematologic toxicities were observed in 3 patients (14.3%), which, however, were manageable and did not result in life-threatening complications. Six patients (28.6%) experienced grade 1–2 gastrointestinal toxicities (nausea, vomiting and anorexia) and 7 patients (33.3%) developed grade 1–2 liver dysfunction. Two patients developed interstitial pneumonitis (grade 2) and were administered glucocorticosteroid therapy.

Table III

Chemotherapy-related toxicity in eligible patients.

Table III

Chemotherapy-related toxicity in eligible patients.

ToxicityGrade 1 no. (%)Grade 2 no. (%)Grade 3 no. (%)Grade 4 no. (%)
Hematologic52 (9.5)2 (9.5)1 (4.8)
Gastrointestinal5 (23.8)1 (4.8)0 (0)0 (0)
Hepatobiliary4 (19.0)3 (14.3)0 (0)0 (0)
Pulmonary0 (0)2 (9.5)0 (0)0 (0)
Survival

The median overall survival was 19.4 months (range, 2–41 months), with a 1- and 2-year survival rates of 61.9 and 38.1%, respectively (Fig. 1). As regards the histological pattern, the median survival was 19.6 months for the epithelial, 22.6 months for the sarcomatous and 7.1 months for the biphasic type of MPM.

Discussion

MPM is notoriously refractory to the majority of treatments and the standard first-line treatment is currently cisplatin and PTX chemotherapy (10). In the present study, we evaluated the feasibility of a non-platimum regimen for MPM, involving the sequential administration of the anti-metabolites, methotrexate and gemcitabine.

Methotrexate, an antifolate, has long been used as an anticancer agent and exerts its action through the inhibition of dihydrofolate reductase (DHFR) (12). High-dose methotrexate (1500 mg/m2) has been reported to be effective in the treatment of MPM, with a response rate of 37% (18). However, high-dose methotrexate was associated with severe toxicity and this method of treatment has been abandoned. The efficacy of low- or medium-dose methotrexate has not been assessed in MPM. In the treatment of gastric cancer (19) and head and neck cancer (20), weekly administration of medium-dose methotrexate (100–200 mg/m2) combined with sequential administration of 5-fluorouracil (5-FU) (600 mg/m2) has been reported to be effective and of low toxicity. In this study, a moderate dose of methotrexate (100 mg/m2) was administered weekly in combination with gemcitabine.

PTX is a newly developed antifolate that targets multiple enzymes involved in DNA synthesis and folate metabolism. Single use of PTX has been reported to be moderately effective against MPM (21). Following combination therapy with 1,250 mg/m2 gemcitabine administered on days 1 and 8 and 500 mg/m2 PTX administered on day 8 or 1, chemotherapy-naïve MPM patients exhibited a response rate of 17–26%, with a median survival of 8–10 months (22). Hematologic toxicities included grade 3–4 neutropenia (60%) and febrile neutropenia (10%). These results indicated that the combination of PTX and gemcitabine was moderately effective in MPM patients but was associated with a notably high incidence of neutropenia (22).

In this study on the methotrexate and gemcitabine doublet regimen, 3 patients (14.3%) exhibited grade 3–4 hematologic toxicity, with no sepsis or hemorrhage. There was no observed grade 3–4 non-hematological toxicity. Two patients developed interstitial pneumonitits (grade 2) which responded well to steroid therapy. Thus, the tolerability and toxicity profiles were considered acceptable.

The response rate with the methotrexate and gemcitabine combination chemotherapy was 38.1%, which is within the range of 20–50% observed with other ‘active’ agents for MPM (8,10). Median survival was 19.4 months. Antifolates may be one of the key agents for MPM, since the majority of mesothelioma cells of all histological MPM subtypes express high-affinity α folate receptor (23). In our combination regimen, we observed that methotrexate, an old-type antifolate, exhibited desirable efficacy. Methotrexate has also been reported to be more efficient compared to PTX, a newly developed antifolate, against osteosarcoma cells (24), indicating that methotrexate possesses a therapeutic potential.

In the present study, 10 out of the 21 patients were chemotherapy-naïve and their response rate to this regimen was similar to the overall response rate described above. This suggests that methotrexate plus gemcitabine may be beneficial as the first-line treatment for MPM. Eleven patients who had been previously treated also exhibited a response rate similar to the overall response rate. Although the optimal regimen constituting the second-line chemotherapy remains to be determined, results of the present study suggest that methotrexate plus gemcitabine may also be beneficial as a second-line treatment.

In conclusion, the present study demonstrated that the methotrexate and gemcitabine combination therapy is feasible, with a more favorable toxicity profile and efficient in the treatment of MPM. Further clinical evaluation is required, with prospective trials including a larger cohort of patients.

References

1. 

Robinson BW, Musk AW and Lake RA: Malignant mesothelioma. Lancet. 366:397–408. 2005. View Article : Google Scholar : PubMed/NCBI

2. 

Nakano T: Current therapies for malignant pleural mesothelioma. Environ Health Prev Med. 13:75–83. 2008. View Article : Google Scholar : PubMed/NCBI

3. 

Wagner JC, Slegg CA and Marchand P: Diffuse pleural mesothelioma and asbestos exposure in North western Cape Province. Br J Ind Med. 17:260–271. 1960.PubMed/NCBI

4. 

Kaufman AJ and Pass HI: Current concepts in malignant pleural mesothelioma. Expert Rev Anticancer Ther. 8:293–303. 2008. View Article : Google Scholar : PubMed/NCBI

5. 

Yang H, Testa JR and Carbone M: Mesothelioma epidemiology, carcinogenesis, and pathogenesis. Curr Treat Options Oncol. 9:147–57. 2008. View Article : Google Scholar : PubMed/NCBI

6. 

Maziak DE, Gagliardi A, Haynes AE, Mackay JA and Evans WK: Surgical management of malignant pleural mesothelioma: a systematic review and evidence summary. Lung Cancer. 48:157–169. 2005. View Article : Google Scholar : PubMed/NCBI

7. 

Ung YC, Yu E, Falkson C, Haynes AE, Stys-Norman D and Evans WK: The role of radiation therapy in malignant pleural mesothelioma: a systematic review. Radiother Oncol. 80:13–18. 2006. View Article : Google Scholar : PubMed/NCBI

8. 

Steele JPC and Klabatsa A: Chemotherapy options and new advances in malignant pleural mesothelioma. Ann Oncol. 16:345–351. 2005. View Article : Google Scholar : PubMed/NCBI

9. 

Fennell DA, Gaudino G, O’Byrne KJ, Mutti L and van Meerbeeck J: Advances in the systemic therapy of malignant pleural mesothelioma. Nat Clin Pract Oncol. 5:136–147. 2008. View Article : Google Scholar : PubMed/NCBI

10. 

Vogelzang NJ, Rusthoven JJ, Symanowski J, et al: Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma. J Clin Oncol. 21:2636–2644. 2003. View Article : Google Scholar : PubMed/NCBI

11. 

Huennekens FM: The methotrexate story: a paradigm for development of cancer chemotherapeutic agents. Adv Enzyme Regul. 34:397–419. 1994. View Article : Google Scholar : PubMed/NCBI

12. 

McGuire JJ: Anticancer antifolates: current status and future directions. Curr Pharm Des. 9:2593–2613. 2003. View Article : Google Scholar : PubMed/NCBI

13. 

Lund B, Kristjansen P and Hansen H: Clinical and preclinical activity of 2′,2′-difluorodesoxycitidine (Gemcitabine). Cancer Treat Rev. 19:45–55. 1993.

14. 

Rusch VW: A proposed new international TNM staging system for malignant pleural mesothelioma. From the International Mesothelioma Interest Group Chest. 108:1122–1128. 1995.PubMed/NCBI

15. 

Byrne MJ and Nowak AK: Modified RECIST criteria for assessment of response in malignant pleural mesothelioma. Ann Oncol. 15:257–60. 2004. View Article : Google Scholar : PubMed/NCBI

16. 

National Cancer Institute Common Toxicity Criteria for Adverse Events version 3.0. http://ctep.cancer.gov/reporting/ctc.html Accessed August 9, 2003.

17. 

Kaplan M and Meier P: Nonparametric estimation from incomplete observations. J Am Stat Assoc. 53:457–481. 1958. View Article : Google Scholar

18. 

Solheim OP, Saeter G, Finnanger AM and Stenwig AE: High-dose methotrexate in the treatment of malignant mesothelioma of the pleura: a phase II study. Br J Cancer. 65:956–960. 1992. View Article : Google Scholar : PubMed/NCBI

19. 

Imazawa M, Kojima T, Boku N, et al: Efficacy of sequential methotrexate and 5-fluorouracil (MTX/5FU) in improving oral intake in patients with advanced gastric cancer with severe peritoneal dissemination. Gastric Cancer. 12:153–157. 2009. View Article : Google Scholar : PubMed/NCBI

20. 

Ringborg U, Ewert G, Kinnman J, Lundquist PG and Strander H: Methotrexate and 5-fluorouracil in head and neck cancer. Semin Oncol 10. (Suppl 2): 20–22. 1983.

21. 

Scagliotti GV, Shin DM, Kindler HL, et al: Phase II study of pemetrexed with and without folic acid and vitamin B12 as front-line therapy in malignant pleural mesothelioma. J Clin Oncol. 21:1556–1561. 2003. View Article : Google Scholar : PubMed/NCBI

22. 

Jänne PA, Simon GR, Langer CJ, et al: Phase II trial of pemetrexed and gemcitabine in chemotherapy-naive malignant pleural mesothelioma. J Clin Oncol. 26:1465–1471. 2008.PubMed/NCBI

23. 

Bueno R, Appasani K, Mercer H, Lester S and Sugarbaker D: The alpha folate receptor is highly activated in malignant pleural mesothelioma. J Thorac Cardiovasc Surg. 121:225–233. 2001. View Article : Google Scholar : PubMed/NCBI

24. 

Bodmer N, Walters DK and Fuchs B: Pemetrexed, a multitargeted antifolate drug, demonstrates lower efficacy in comparison to methotrexate against osteosarcoma cell lines. Pediatr Blood Cancer. 50:905–908. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-August 2013
Volume 1 Issue 4

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kuribayashi K, Miyata S, Fukuoka K, Murakami A, Yamada S, Tamura K, Hirayama N, Terada T, Tabata C, Fujimori Y, Fujimori Y, et al: Methotrexate and gemcitabine combination chemotherapy for the treatment of malignant pleural mesothelioma. Mol Clin Oncol 1: 639-642, 2013
APA
Kuribayashi, K., Miyata, S., Fukuoka, K., Murakami, A., Yamada, S., Tamura, K. ... Nakano, T. (2013). Methotrexate and gemcitabine combination chemotherapy for the treatment of malignant pleural mesothelioma. Molecular and Clinical Oncology, 1, 639-642. https://doi.org/10.3892/mco.2013.118
MLA
Kuribayashi, K., Miyata, S., Fukuoka, K., Murakami, A., Yamada, S., Tamura, K., Hirayama, N., Terada, T., Tabata, C., Fujimori, Y., Nakano, T."Methotrexate and gemcitabine combination chemotherapy for the treatment of malignant pleural mesothelioma". Molecular and Clinical Oncology 1.4 (2013): 639-642.
Chicago
Kuribayashi, K., Miyata, S., Fukuoka, K., Murakami, A., Yamada, S., Tamura, K., Hirayama, N., Terada, T., Tabata, C., Fujimori, Y., Nakano, T."Methotrexate and gemcitabine combination chemotherapy for the treatment of malignant pleural mesothelioma". Molecular and Clinical Oncology 1, no. 4 (2013): 639-642. https://doi.org/10.3892/mco.2013.118