Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Cancer Therapy: Clinical

Pemetrexed Safety and Pharmacokinetics in Patients with Third-Space Fluid

Nicolas J. Dickgreber, Jens Benn Sorensen, Luis G. Paz-Ares, Tine Kjestrup Schytte, Jane E. Latz, Karen B. Schneck, Zheng Yuan and José Miguel Sanchez-Torres
Nicolas J. Dickgreber
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jens Benn Sorensen
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Luis G. Paz-Ares
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tine Kjestrup Schytte
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jane E. Latz
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Karen B. Schneck
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Zheng Yuan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
José Miguel Sanchez-Torres
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1078-0432.CCR-09-3324 Published May 2010
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Purpose: Pemetrexed is established as first-line treatment with cisplatin for malignant pleural mesothelioma and advanced nonsquamous non–small-cell lung cancer (NSCLC) and as single-agent second-line treatment for nonsquamous NSCLC. Because the structure and pharmacokinetics of pemetrexed are similar to those of methotrexate, and methotrexate is associated with severe toxicity in patients with third-space fluid (TSF), the safety of pemetrexed in patients with TSF was evaluated.

Experimental Design: Patients with TSF (pleural effusions, ascites) and relapsed, stage III/IV NSCLC or malignant pleural/peritoneal mesothelioma were treated with pemetrexed (500 mg/m2) on day 1 of each 21-day cycle. TSF was drained at any time only if clinically indicated. Plasma samples were collected during cycles 1 and 2 to compare pemetrexed concentrations with reference data from patients without TSF.

Results: Thirty-one patients with TSF received 123 pemetrexed doses (median, 4 cycles per patient; range, 1-11; mean dose intensity, 97.5%). Seven grade 3/4 drug-related toxicities, including four hematologic, were reported; there were no treatment-related deaths. There was no correlation between TSF amount and type, number, and sequelae of toxicities. Pemetrexed plasma concentrations were within the range of those in patients without TSF. Pemetrexed clearance and central volume of distribution were not statistically different between patients with and without TSF.

Conclusions: No clinically relevant alterations of pemetrexed pharmacokinetics occurred in patients with TSF. Pemetrexed was well tolerated; toxicities were expected and manageable. The standard pemetrexed dose recommendations were adequate for patients with TSF in this study. These data suggest that draining TSF before administering pemetrexed is unnecessary. Clin Cancer Res; 16(10); 2872–80. ©2010 AACR.

Translational Relevance

This study examined the pharmacokinetics and safety of pemetrexed in patients with third-space fluid (TSF). The practice of draining TSF before pemetrexed administration was enacted based on structural and pharmacokinetic similarities between pemetrexed and methotrexate, which requires draining TSF, and without actual evidence of safety issues associated with administering pemetrexed to patients with TSF. Standard pemetrexed dose recommendations (starting dose and dose reductions) were adequate for patients with TSF treated in this study, suggesting that these patients can be treated with pemetrexed and obtain benefit, and that draining TSF before administering pemetrexed may not be necessary. Determining the necessity of draining TSF is important because the procedure is inconvenient, often painful, and introduces clinical risk due to potential procedural complications (e.g., pneumothorax, infection). These findings raise a broader question to consider in drug development: when and whether to draw analogies from prior observations versus limited information.

Patients with thoracic tumors at diagnosis often present with imaging evidence of third-space fluid (TSF) such as malignant pleural effusions or ascites (1). TSF may alter drug pharmacokinetics because it serves as an additional compartment into which the drug can be distributed and from which elimination can be delayed (2).

The pharmacokinetics of methotrexate are altered in patients with TSF. Methotrexate is significantly distributed into TSF and only slowly released, resulting in prolongation of the terminal half-life of methotrexate in the plasma (3–5) and potentially increased toxicity (6). The toxicity of methotrexate for normal tissues is believed to depend on the duration of exposure to the drug above a threshold rather than on the peak level of drug achieved or overall exposure [i.e., area under the curve (AUC); refs. 7, 8]. The current clinical practice in patients with significant TSF accumulations is to drain the fluid before methotrexate treatment and to monitor plasma methotrexate levels.

Pemetrexed, a folate analogue metabolic inhibitor, has a chemical structure similar to methotrexate. It is indicated for locally advanced or metastatic nonsquamous non–small-cell lung cancer (NSCLC) as first-line therapy in combination with cisplatin (9), or as maintenance treatment for patients whose disease has not progressed after four cycles of platinum-based first-line chemotherapy (10), or after prior chemotherapy as a single agent (11). Additionally, pemetrexed in combination with cisplatin is indicated for treatment of mesothelioma (12).

Similar to methotrexate, the current practice in patients with TSF accumulations is to drain the fluid before pemetrexed administration. This practice is based on the structural and pharmacokinetic similarities between pemetrexed and methotrexate. The primary route of elimination for both pemetrexed and methotrexate is renal excretion of unchanged drug in the urine (13, 14). Like methotrexate, the clearance of pemetrexed is decreased in patients with renal insufficiency (15, 16). For both drugs, decreased clearance results in greater systemic exposure, which may be associated with increased toxicity.

Although there are similarities between pemetrexed and methotrexate, there are sufficient differences in the pharmacokinetics and safety profiles of the two drugs to raise the question whether draining TSF before pemetrexed administration is necessary. Whereas methotrexate toxicity correlates with time above a concentration threshold (7), pemetrexed toxicity seems to correlate with total systemic exposure (AUC; refs. 14, 17–20). In addition, the terminal elimination half-life (t1/2) of methotrexate has been correlated with higher toxicity (7), whereas a similar correlation with pemetrexed has not been shown.

The maximum tolerated dose (MTD) for pemetrexed administered in combination with standard folic acid and vitamin B12 supplementation is a further indication that it may not be necessary to drain TSF before pemetrexed administration. The MTD for pemetrexed administered in combination with standard supplementation is 925 mg/m2 or higher, depending on patient pretreatment status (lightly or heavily pretreated; ref. 21). This MTD corresponds to an 85% increase in drug exposure relative to the currently approved 500 mg/m2 dose; thus, patients treated at 500 mg/m2 could potentially tolerate an 85% increase in AUC without difficulty. This conclusion is supported by other studies which have shown that patients experience minimal additional toxicity with higher (900 and 1,000 mg/m2) pemetrexed doses (22, 23).

The differences in the pharmacokinetic/pharmacodynamic relationships for methotrexate and pemetrexed, coupled with information on pemetrexed MTD, suggest that draining TSF before pemetrexed administration may not be needed. Because draining TSF is inconvenient, often painful, and introduces clinical risk due to potential complications of the procedure (e.g., pneumothorax, infection), determining the necessity of drainage is clinically important.

To define the dosing and/or clinical practices required for patients with significant TSF (pleural effusions or ascites), a single-arm, open-label, phase II trial of single-agent pemetrexed in patients with TSF (pleural effusions or ascites) was conducted. Specifically, the safety and pharmacokinetics of pemetrexed were evaluated to determine (a) whether existing pemetrexed dosing recommendations (starting dose and dose reductions) were suitable for patients with TSF, and (b) whether draining TSF is necessary for patients before pemetrexed administration. A formal evaluation of efficacy was not an objective for this study.

Materials and Methods

Patient selection

Patients ≥18 years old with clinically detectable and stable-appearing TSF accumulations and either locally advanced or metastatic (stage III or IV) NSCLC or malignant pleural or peritoneal mesothelioma were eligible. In addition, patients must have had an Eastern Cooperative Oncology Group (24) performance status of 0 or 1, adequate bone marrow reserve and organ function, including calculated creatinine clearance ≥45 mL/min, and life expectancy ≥8 weeks. Previous treatment with one platinum-containing chemotherapy regimen in the locally advanced or metastatic setting was required for patients with NSCLC. Patients with mesothelioma could have received one previous chemotherapy regimen and could be enrolled if they were clinical candidates for treatment with single-agent pemetrexed in the investigator's opinion. Among reasons for exclusion were brain metastases, a concomitant serious systemic disorder, and inability or unwillingness to interrupt use of aspirin and other nonsteroidal anti-inflammatory drugs or to take folic acid, vitamin B12, or corticosteroids.

Institutional ethics review boards approved the protocol, and the trial was conducted according to Good Clinical Practice guidelines and the Declaration of Helsinki. All patients provided written informed consent before treatment.

Treatment plan

This was a multicenter, open-label phase II study of single-agent pemetrexed with a target enrollment of 30 patients. Pemetrexed (500 mg/m2) was administered i.v. over 10 minutes on day 1 of each 21-day cycle. Chemotherapy was continued for a total of six cycles unless the investigator or patient opted to discontinue treatment. Patients could receive more than six cycles at the discretion of the treating physician and study sponsor. Patients received 4 mg of prophylactic dexamethasone orally twice per day on the day before, the day of, and the day after the first day of each cycle. Patients also received oral folic acid (350-1,000 μg) daily and a vitamin B12 injection (1,000 μg) every 9 weeks, beginning 1 to 2 weeks before the first dose and continuing until 3 weeks after the last dose of study treatment.

Dose adjustments at the start of a subsequent cycle of study treatment were based on platelet and neutrophil nadir counts from the preceding cycle and on grade 3/4 nonhematologic toxicities from the preceding cycle. Patients requiring a dose reduction received the reduced dose for the remainder of the study. Patients who had two dose reductions and who experienced toxicity requiring a third dose reduction were discontinued from study therapy. Cycles were delayed for up to 42 days to allow neutrophil levels to return to ≥1.5 × 109/L and platelet levels to return to ≥100 × 109/L. For grade 3/4 nonhematologic toxicities (except for grade 3 transaminase elevation, nausea, and vomiting), treatment was delayed until resolution to at least the patient's baseline. Patients received full supportive care therapies concomitantly during the study. Colony-stimulating factors were administered according to the American Society of Clinical Oncology guidelines (25). A safety evaluation was done after 10 patients were enrolled and treated with at least two cycles of pemetrexed.

Baseline and treatment assessments

Before entering the study, patients underwent a medical history and physical examination, and baseline tumor measurements were taken for patients with measurable lesions. Measurable lesions were not required for study participation because efficacy was not a primary or secondary end point. During the study, lesion measurements were done at the discretion of the investigator for patients showing evidence of a response. Tumor response was assessed using the Response Evaluation Criteria in Solid Tumors (26) and summarized for all patients with available data.

All patients who received at least one dose of pemetrexed were considered evaluable for safety. Toxicity was assessed before each cycle according to the Common Terminology Criteria for Adverse Events, version 3.0 (27).

Plasma samples for pemetrexed pharmacokinetic determinations were collected from all patients during cycles 1 and 2 immediately before the end of infusion (9.5 minutes), 2 hours postinfusion, 9 to 10 hours postinfusion, 24 to 48 hours postinfusion, and 20 to 22 days postinfusion.

The extent of TSF was estimated immediately before each cycle. Mild TSF was defined as a small amount of fluid, less than moderate, and only detectable by radiological imaging. Moderate TSF was defined as a medium amount of fluid, with pleural effusions less than one third of the way up the thorax on one side by imaging, and with ascites detectable by physical exam. Severe TSF was defined as a large amount of fluid, with pleural effusion more than one third of the way up the thorax on imaging, and with ascites detectable by visual appearance of the abdomen. TSF was drained at any time during the study if clinically indicated. If drainage was done, it was accomplished as completely as possible, the volume measured, and a sample of the fluid collected to determine the pemetrexed concentration. Additionally, a single plasma sample was obtained at the time of drainage to determine the corresponding plasma pemetrexed concentration.

Pharmacokinetic evaluations

Plasma and TSF samples were analyzed for pemetrexed concentrations (Taylor Technology, Inc.) using a validated liquid chromatography/electrospray ionization-tandem mass spectrometry method to generate a linear response over the concentration ranges of 10 to 2,000 ng/mL and 1,000 to 200,000 ng/mL (28). Pharmacokinetic analyses were done on a pooled data set containing pemetrexed concentration-time results from the current study combined with a reference data set (21) containing concentration-time results from patients without TSF. The data were analyzed by a population pharmacokinetics approach in the nonlinear mixed-effects modeling program NONMEM version VI with PREDPP (version V), using the first-order conditional estimation method with interaction (29–31). The pharmacokinetics of pemetrexed were described by a three-compartment model parameterized in terms of clearance (CL), central volume of distribution (V1), peripheral volume of distribution (V2 and V3), and intercompartmental clearances (Q1 and Q2), incorporating between-patient variability with respect to CL, V1, and V2, and a combined additive and proportional residual error term. The model included previously established covariates—Cockcroft-Gault creatinine clearance (32) with respect to CL and body surface area with respect to V1 and V2.

Potential differences in pemetrexed pharmacokinetics due to the presence of TSF were examined by adding this factor to the model as a covariate (dichotomous variable) with respect to CL, V1, and V2; nested models were compared based on the minimum objective function (MOF). Potentially significant alterations in pharmacokinetics were identified based on maximum likelihood criteria as those resulting in a decrease in the MOF of ≥3.841 points (P < 0.05 based on χ2 distribution with 1 degree of freedom) when added to the model. Parameter sensitivity analysis was used to define the 90% confidence interval for the ratio comparing pharmacokinetic parameters in patients with and without TSF. The criteria for clinically relevant changes based on a log-normal distribution were ±20% (ratio of 0.8 to 1.25) for CL and ±30% (ratio of 0.7 to 1.43) for V1 and V2. Changes within these ranges result in pemetrexed exposures [AUC and maximum concentration (Cmax)] that remain lower than the exposures from the MTD (925 mg/m2) established for pemetrexed with vitamin supplementation (21) and are shown to be tolerable (22, 23). The criteria for clinical relevance were based on AUC having been identified as the primary determinant of response in patients given pemetrexed (14, 17–20).

Results

Patients

The study was conducted at five study sites in Europe between December 26, 2006 and March 11, 2009. Thirty-one patients were enrolled in the study and received at least one dose of study drug (Fig. 1). Baseline and disease characteristics are summarized in Table 1. Twenty-seven men and four women were enrolled and treated in this study: 23 (74%) patients had a diagnosis of NSCLC and 8 (26%) had mesothelioma. Most patients (58%) had stage IV disease and mild (48%) or moderate (45%) amounts of TSF at baseline. For the majority of the patients (25 of the 31), the amount of TSF did not differ throughout the study, although 4 of the 25 had drainage done before a cycle. Three patients had a decrease in TSF while on study, and three additional patients had either an increase or both an increase and a decrease in TSF. The reference patient population (21) to which the pharmacokinetic data are compared had a median age approximately 5 years younger, but with a similar age range. The reference population also had a similar PS 0/1 ratio, with most patients reporting prior chemotherapy and metastatic disease.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

CONSORT flow diagram of patient disposition.

View this table:
  • View inline
  • View popup
Table 1.

Patient and disease characteristics

Treatment

The 31 patients received a median of four cycles (range, 1-11; 123 total cycles) of pemetrexed treatment. Among the 10 patients (32.3%) completing at least six cycles, 2 patients completed seven cycles and 1 patient completed 11 cycles; 15 patients (48.4%) discontinued the study before completing six cycles due to progressive disease (Fig. 1). The delivered dose intensity for pemetrexed was 97.5% (162.5 mg/m2 per patient per week), with no dose omissions; however, one patient required a dose reduction due to asthenia not related to study drug, and there were four cycle delays due to adverse events (one each of asthenia, pain, pyrexia, and post-thoracentesis pneumothorax).

Safety

Seven grade 3 and 4 drug-related toxicities were reported; these included four hematologic toxicities, including one incident each of leukopenia, thrombocytopenia, neutropenia, and febrile neutropenia, and three nonhematologic toxicities, including one incident each of pulmonary pain, pleural effusion, and ascites. The patient who experienced grade 3/4 febrile neutropenia withdrew from the study due to this toxicity. The most frequently occurring grade 1/2 toxicities (>10% of patients) were nausea (9 patients, 29%), fatigue (7 patients, 22.6%), and anorexia (5 patients, 16.1%). There were no treatment-related deaths during the study or within 30 days of study discontinuation. Two patients died during the study from reasons judged by the investigators as non–treatment related: respiratory failure following atrial fibrillation and dyspnea (day 22 of cycle 4 in a patient with NSCLC) and pneumonia (day 18 of cycle 1 in a patient with malignant pleural mesothelioma). One patient required a platelet transfusion during the study, and 5 (16%) patients required RBC transfusions due to grade 2 anemia, including 1 patient who had grade 2 anemia at baseline. Toxicities resulted in the hospitalization of 12 (39%) patients during the study and 2 (6%) patients within 30 days of study discontinuation. No correlation was observed between the volume of TSF (mild, moderate, severe) and the type, number, severity, or sequelae of toxicities (Table 2).

View this table:
  • View inline
  • View popup
Table 2.

Incidence of safety parameters by relative amount of TSF

Pharmacokinetic analyses

Observed pemetrexed plasma concentrations

Figure 2 compares dose-normalized plasma pemetrexed concentration-versus-time data for patients with TSF treated in this study and a reference population consisting of patients without TSF (21). The concentration-versus-time data for the two studies were similar, indicating that pemetrexed pharmacokinetics were similar for the two groups of patients. All but one of the plasma samples scheduled to be collected during the 24- to 48-hour postinfusion interval were collected at approximately 24 hours; one sample collected at 48 hours was below the limit of detection. All 26 plasma samples collected at the end of cycle 1 and all but one of the 24 samples collected at the end of cycle 2 had pemetrexed concentrations below the quantification limit of <10 ng/mL. Likewise, in the reference study of patients without TSF, no patient had a quantifiable plasma concentration of pemetrexed at the end of a cycle. The single patient with TSF who had quantifiable pemetrexed concentrations at the end of cycle 2 did not have toxicities remarkably different from the other study participants.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Comparison of dose-normalized pemetrexed concentration-versus-time from dose for individuals with TSF (TSF+; current study) and without TSF [TSF−; reference data (21)].

Evaluation of TSF status on pemetrexed pharmacokinetics

Table 3 summarizes the evaluation of potential differences in pemetrexed pharmacokinetics based on TSF status. The three-compartment model did not support an interindividual variability term for V3, and thus a statistical comparison was not done and lack of a statistically significant difference was presumed. Pemetrexed CL and V2 were not significantly different between the two populations (ΔMOF <3.841 points; P < 0.05). Although the difference in V1 was statistically significant between patients with and without TSF, the 90% confidence intervals were within the predefined acceptance ranges of ±30%. A change in pemetrexed CL of 20% corresponds to an alteration in AUC of 25%. The Cmax of patients in the two studies was similar due to the small difference (∼4%) in steady-state volume of distribution (Vss = V1 + V2 + V3), where the median (range) was 16.2 liters (10.3-25.7 liters) for patients in the reference study (21) and 16.8 liters (11.4-25.5 liters) for patients in this study.

View this table:
  • View inline
  • View popup
Table 3.

Evaluation of potential differences in pemetrexed pharmacokinetics by presence of TSF

There were no systematic changes in pemetrexed exposure as shown by model-predicted estimates of Cmax (Fig. 3A) and AUC (Fig. 3B) over the course of two cycles, and no evident trend between pemetrexed exposure and the amount of TSF (mild to severe pleural effusions and moderate ascites). Only four patients had fluid drained before pemetrexed administration in cycle 2. These patients, all with adequate renal function, did not show a marked difference in pemetrexed pharmacokinetics in the presence or absence of TSF (Fig. 3). Pemetrexed concentrations in the four TSF samples were consistent with time-matched plasma samples, suggesting that pemetrexed does not accumulate in TSF relative to plasma.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Model-predicted Cmax (A) and AUC (B) by TSF status and cycle.

Efficacy

Although efficacy was not an objective of this study and measurable disease was not required for study participation, baseline and postbaseline lesion measurements, if available, were reported by the investigator. Among the 22 patients with response data, there were no complete responses, 2 patients had a partial response, 8 patients had stable disease, and 12 patients had a best response of progressive disease. The other 9 patients had unknown response (including 1 patient with no baseline assessment, 7 patients with no postbaseline assessments, and 1 patient with unconfirmed stable disease).

Discussion

Because mesothelioma and advanced nonsquamous NSCLC often present with TSF accumulation, and its drainage introduces additional clinical risks, this single-arm, open-label, phase II trial of single-agent pemetrexed was undertaken to define the dosing and clinical practices required for patients with NSCLC or malignant pleural or peritoneal mesothelioma with TSF accumulation. A median of four cycles of the label-recommended dose of single-agent pemetrexed, 500 mg/m2, administered as an i.v. infusion over 10 minutes on day 1 of each 21-day cycle, was safely given to patients with adequate renal clearance and standard vitamin supplementation without first draining the accumulated fluid. The toxicities that occurred were expected, manageable, and consistent with the known pemetrexed safety profile (33). This included four grade 3/4 hematologic toxicities and approximately 15–30% grade 1/2 fatigue, nausea, and anorexia. The dose delay and reduction recommendations provided in the pemetrexed label were adequate for patients with TSF who were treated in this study.

Plasma samples for measurement of pemetrexed concentrations were obtained after cycles 1 and 2, and pemetrexed concentration-versus-time results from this study were compared with those from a previously completed study in which single-agent pemetrexed was administered with vitamin supplementation to patients without TSF (21). The similar concentration-versus-time plots suggest that pemetrexed pharmacokinetics are comparable between patients with and without TSF. In addition, pemetrexed Cmax and AUC were consistent between cycles and were not affected by the draining of TSF. The relative amounts of TSF did not systematically correlate with any differences in pemetrexed pharmacokinetic parameters. Observations near the end of a cycle (19 days after pemetrexed dosing) were generally below the quantifiable limit, a finding that is consistent with the disposition of pemetrexed in patients without TSF. These results suggest that pemetrexed is not sequestered in third-space compartments and that the extent of exposure to pemetrexed is similar in patients with and without TSF.

The absence of overt differences in the safety profile or pharmacokinetics of pemetrexed administered to patients with TSF suggests that pemetrexed can be safely given to patients without draining the TSF. Furthermore, these results indicated that the dosing guidelines previously established for patients without TSF are also applicable for patients with TSF. However, given that pemetrexed clearance is primarily through the kidneys, patients with marginal renal function might still require drainage. Indeed, a case report by Brandes and colleagues (34) found elevated levels of pemetrexed in plasma and ascites in a patient with renal failure following chemotherapy with cisplatin and pemetrexed. Although efficacy was not an objective of this study, lesion measurements were done at baseline, with subsequent measurements done at the investigator's discretion. Among the 31 evaluable patients in this study, 2 patients showed a partial response and 8 patients, stable disease; 9 responses were unknown. Although the control of TSF by systemic chemotherapy has not been routinely described, chemotherapy is considered to potentially reduce TSF (and by extension, improve quality of life parameters associated with TSF accumulation; ref. 35).

Although this investigation examined pemetrexed administered as a single agent, it is coadministered with cisplatin for mesothelioma and advanced nonsquamous NSCLC (9, 12). Earlier work (33, 36, 37) examined possible pharmacokinetic interactions between pemetrexed and cisplatin and concluded that the two agents did not influence each other's distribution or elimination. The pemetrexed pharmacokinetic results from this study were consistent with previous results for pemetrexed in combination with cisplatin, as well as those from single-agent pemetrexed studies (21, 28, 33, 36, 37). Thus, the results from the present study can likely be extended to patients receiving pemetrexed-cisplatin combination therapy (9, 12).

In addition to malignant pleural mesothelioma and NSCLC, pemetrexed has shown antitumor activity in a range of other solid malignancies including bladder, head and neck, breast, cervical, colorectal, pancreatic, and gastric cancers (38, 39). Some of these tumors may also present with TSF accumulations; for example, ascites is common in patients with gastrointestinal and gynecologic cancers, and pleural effusions may occur with breast cancer. Because the pharmacokinetics of pemetrexed have been characterized in patients with a variety of solid tumors and shown to be comparable across tumor types, the results from this study can likely be extended to patients with other tumors types and TSF accumulations.

Given the structural similarity of pemetrexed and methotrexate and the known alterations in pharmacokinetics and increased toxicity associated with methotrexate treatment of patients with TSF, this study examined pemetrexed treatment of patients with pleural effusions or ascites associated with NSCLC or mesothelioma. No clinically relevant differences in pemetrexed pharmacokinetics in patients with TSF were identified relative to a reference population of patients without TSF. The established dose of pemetrexed given with vitamin supplementation was well tolerated in patients with TSF, and the toxicities were consistent with the known pemetrexed safety profile. The standard dose recommendations (starting dose and dose reductions) for pemetrexed were adequate for patients with TSF who were treated in this study. Data from this study suggest that draining the TSF accumulations before pemetrexed treatment is not necessary. Further studies are warranted to confirm this finding.

Disclosure of Potential Conflicts of Interest

N.J. Dickgreber and J.B. Sorensen: commercial research support, honoraria from speakers bureau, and consultant/advisory board, Eli Lilly; L.G. Paz-Ares: consultant/advisory board, Eli Lilly. The other authors declare no conflicts.

Acknowledgments

We thank all the patients, investigators, and institutions involved in this study, and Mary Dugan Wood and Angela Lorio for writing and editorial assistance.

Grant Support: This study (H3E-MC-JMHX) was sponsored and funded by Eli Lilly and Company, Indianapolis, Indiana.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Footnotes

  • Note: Interim results of this study were disclosed as a poster presentation at the 2008 ASCO Annual Meeting, May 31 to June 3, 2008, Chicago, Illinois, and some of the final results of this study were disclosed as a poster presentation at ECCO 15—34th ESMO Congress, September 20-24, 2009, Berlin, Germany.

  • Received December 18, 2009.
  • Revision received March 19, 2010.
  • Accepted March 27, 2010.

References

  1. ↵
    1. Fenton KN,
    2. Richardson JD
    . Diagnosis and management of malignant pleural effusions. Am J Surg 1995;170:69–74.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Abeloff M,
    2. Armitage J,
    3. Niederhuber J,
    4. et al.
    1. Spira A,
    2. Brahmer J
    . Effusions. In: Abeloff M, Armitage J, Niederhuber J, et al., editors. Clinical oncology. Third ed. Philadelphia (PA): Elsevier Churchill Livingstone; 2004, p. 1179–12.
  3. ↵
    1. Wan SH,
    2. Huffman DH,
    3. Azarnoff DL,
    4. Stephens R,
    5. Hoogstraten B
    . Effect of route of administration and effusions on methotrexate pharmacokinetics. Cancer Res 1974;34:3487–91.
    OpenUrlAbstract/FREE Full Text
    1. Chabner BA,
    2. Stoller RG,
    3. Hande K,
    4. Jacobs S,
    5. Young RC
    . Methotrexate disposition in humans: case studies in ovarian cancer and following high-dose infusion. Drug Metab Rev 1978;8:107–17.
    OpenUrlPubMed
  4. ↵
    1. Evans WE,
    2. Pratt CB
    . Effect of pleural effusion on high-dose methotrexate kinetics. Clin Pharmacol Ther 1978;23:68–72.
    OpenUrlPubMed
  5. ↵
    1. Li J,
    2. Gwilt P
    . The effect of malignant effusions on methotrexate disposition. Cancer Chemother Pharmacol 2002;50:373–82.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Chabner BA,
    2. Young RC
    . Threshold methotrexate concentration for in vivo inhibition of DNA synthesis in normal and tumorous target tissues. J Clin Invest 1973;52:1804–11.
    OpenUrlCrossRefPubMed
  7. ↵
    Methotrexate [package insert]. Bedford (OH): Ben Venue Laboratories, Inc.; 2009.
  8. ↵
    1. Scagliotti GV,
    2. Parikh P,
    3. von Pawel J,
    4. et al
    . Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol 2008;26:3543–51.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    1. Ciuleanu T,
    2. Brodowicz T,
    3. Zielinski C,
    4. et al
    . Maintenance pemetrexed plus best supportive care versus placebo plus best supportive care for non-small-cell lung cancer: a randomised, double-blind, phase 3 study. Lancet 2009;374:1432–40.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Hanna N,
    2. Shepherd FA,
    3. Fossella FV,
    4. et al
    . Randomized phase III trial of pemetrexed versus docetaxel in patients with non-small-cell lung cancer previously treated with chemotherapy. J Clin Oncol 2004;22:1589–97.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Vogelzang NJ,
    2. Rusthoven JJ,
    3. Symanowski J,
    4. et al
    . Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma. J Clin Oncol 2003;21:2636–44.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Jolivet J,
    2. Cowan KH,
    3. Curt GA,
    4. Clendeninn NJ,
    5. Chabner BA
    . The pharmacology and clinical use of methotrexate. N Engl J Med 1983;309:1094–104.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Rinaldi DA,
    2. Kuhn JG,
    3. Burris HA,
    4. et al
    . A phase I evaluation of multitargeted antifolate (MTA, LY231514), administered every 21 days, utilizing the modified continual reassessment method for dose escalation. Cancer Chemother Pharmacol 1999;44:372–80.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Bressolle F,
    2. Bologna C,
    3. Kinowski JM,
    4. Sany J,
    5. Combe B
    . Effects of moderate renal insufficiency on pharmacokinetics of methotrexate in rheumatoid arthritis patients. Ann Rheum Dis 1998;57:110–3.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Mita AC,
    2. Sweeney CJ,
    3. Baker SD,
    4. et al
    . Phase I and pharmacokinetic study of pemetrexed administered every 3 weeks to advanced cancer patients with normal and impaired renal function. J Clin Oncol 2006;24:552–62.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. Latz JE,
    2. Karlsson MO,
    3. Rusthoven JJ,
    4. Ghosh A,
    5. Johnson RD
    . A semimechanistic-physiologic population pharmacokinetic/pharmacodynamic model for neutropenia following pemetrexed therapy. Cancer Chemother Pharmacol 2006;57:412–26.
    OpenUrlCrossRefPubMed
    1. Latz JE,
    2. Rusthoven JJ,
    3. Karlsson MO,
    4. Ghosh A,
    5. Johnson RD
    . Clinical application of a semimechanistic-physiologic population PK/PD model for neutropenia following pemetrexed therapy. Cancer Chemother Pharmacol 2006;57:427–35.
    OpenUrlCrossRefPubMed
    1. Latz J,
    2. Claret L,
    3. Symanowski J,
    4. et al
    . Evaluation of pemetrexed (PEM) dosing paradigms using exposure-response relationships (ERRs). J Clin Oncol ASCO Annual Meeting Proceedings Part I 2007;25:abstract 2530.
    OpenUrl
  17. ↵
    1. Latz J,
    2. Adachi S,
    3. Symanowski J,
    4. et al
    . Correlation of pemetrexed (PEM) NSCLC exposure-response relationships (ERRs) to clinical study results from western and Japanese patient populations: PD44-3-4. J Thorac Oncol 2007;2:S455–6.
    OpenUrl
  18. ↵
    1. Takimoto CH,
    2. Hammond-Thelin LA,
    3. Latz JE,
    4. et al
    . Phase I and pharmacokinetic study of pemetrexed with high-dose folic acid supplementation or multivitamin supplementation in patients with locally advanced or metastatic cancer. Clin Cancer Res 2007;13:2675–83.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Cullen MH,
    2. Zatloukal P,
    3. Sörenson S,
    4. et al
    . A randomized phase III trial comparing standard and high-dose pemetrexed as second-line treatment in patients with locally advanced or metastatic non-small-cell lung cancer. Ann Oncol 2008;19:939–45.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    1. Ohe Y,
    2. Ichinose Y,
    3. Nakagawa K,
    4. et al
    . Efficacy and safety of two doses of pemetrexed supplemented with folic acid and vitamin B12 in previously treated patients with non-small cell lung cancer. Clin Cancer Res 2008;14:4206–12.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Oken MM,
    2. Creech RH,
    3. Tormey DC,
    4. et al
    . Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol 1982;5:649–55.
    OpenUrlCrossRefPubMed
  22. ↵
    1. Smith TJ,
    2. Khatcheressian J,
    3. Lyman GH,
    4. et al
    . 2006 update of recommendations for the use of white blood cell growth factors: an evidence-based clinical practice guideline. J Clin Oncol 2006;24:3187–205.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Therasse P,
    2. Arbuck SG,
    3. Eisenhauer EA,
    4. et al
    . European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst 2000;92:205–16.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    U.S. Department of Health and Human Services, National Institutes of Health, National Cancer Institute, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis. Common Terminology Criteria for Adverse Events v3.0 (CTCAE). 2006.
  25. ↵
    1. Latz JE,
    2. Chaudhary A,
    3. Ghosh A,
    4. Johnson RD
    . Population pharmacokinetic analysis of ten phase II clinical trials of pemetrexed in cancer patients. Cancer Chemother Pharmacol 2006;57:401–11.
    OpenUrlCrossRefPubMed
  26. ↵
    1. Ludden TM
    . Population pharmacokinetics. J Clin Pharmacol 1988;28:1059–63.
    OpenUrlPubMed
    1. Sheiner LB,
    2. Steimer JL
    . Pharmacokinetic/pharmacodynamic modeling in drug development. Ann Rev Pharmacol Toxicol 2000;40:67–95.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Beal SL,
    2. Sheiner LB,
    3. Boeckmann AJ
    . NONMEM user's guide. Ellicott City (MD): Icon Development Solutions; 1989–2006.
  28. ↵
    1. Cockcroft DW,
    2. Gault MH
    . Prediction of creatinine clearance from serum creatinine. Nephron 1976;16:31–41.
    OpenUrlCrossRefPubMed
  29. ↵
    ALIMTA® pemetrexed for injections (USA package insert). Indianapolis (IN): Eli Lilly and Company; 2009.
  30. ↵
    1. Brandes JC,
    2. Grossman SA,
    3. Ahmad H
    . Alteration of pemetrexed excretion in the presence of acute renal failure and effusions: presentation of a case and review of the literature. Cancer Invest 2006;24:283–7.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Sekine I,
    2. Sumi M,
    3. Saijo N
    . Local control of regional and metastatic lesions and indication for systemic chemotherapy in patients with non-small cell lung cancer. Oncologist 2008;13:21–7.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    1. Specenier PM,
    2. Ciuleanu T,
    3. Latz JE,
    4. Musib LC,
    5. Darstein CL,
    6. Vermorken JB
    . Pharmacokinetic evaluation of platinum derived from cisplatin administered alone and with pemetrexed in head and neck cancer patients. Cancer Chemother Pharmacol 2009;64:233–41.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Dickgreber NJ,
    2. Fink TH,
    3. Latz JE,
    4. Hossain AM,
    5. Musib LC,
    6. Thomas M
    . Phase I and pharmacokinetic study of pemetrexed plus cisplatin in chemonaive patients with locally advanced or metastatic malignant pleural mesothelioma or non-small cell lung cancer. Clin Cancer Res 2009;15:382–9.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    1. Hanauske AR,
    2. Chen V,
    3. Paoletti P,
    4. Niyikiza C
    . Pemetrexed disodium: a novel antifolate clinically active against multiple solid tumors. Oncologist 2001;6:363–73.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Adjei AA
    . Pemetrexed (ALIMTA), a novel multitargeted antineoplastic agent. Clin Cancer Res 2004;10:4276–80s.
    OpenUrlCrossRef
PreviousNext
Back to top
Clinical Cancer Research: 16 (10)
May 2010
Volume 16, Issue 10
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Clinical Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Pemetrexed Safety and Pharmacokinetics in Patients with Third-Space Fluid
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Pemetrexed Safety and Pharmacokinetics in Patients with Third-Space Fluid
Nicolas J. Dickgreber, Jens Benn Sorensen, Luis G. Paz-Ares, Tine Kjestrup Schytte, Jane E. Latz, Karen B. Schneck, Zheng Yuan and José Miguel Sanchez-Torres
Clin Cancer Res May 15 2010 (16) (10) 2872-2880; DOI: 10.1158/1078-0432.CCR-09-3324

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Pemetrexed Safety and Pharmacokinetics in Patients with Third-Space Fluid
Nicolas J. Dickgreber, Jens Benn Sorensen, Luis G. Paz-Ares, Tine Kjestrup Schytte, Jane E. Latz, Karen B. Schneck, Zheng Yuan and José Miguel Sanchez-Torres
Clin Cancer Res May 15 2010 (16) (10) 2872-2880; DOI: 10.1158/1078-0432.CCR-09-3324
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Disclosure of Potential Conflicts of Interest
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Radiation and TGFβ Blockade in Metastatic Breast Cancer
  • Biomarker Analysis from the BERIL-1 Study
  • Novel Intermediate Endpoint in Immunotherapy Studies
Show more Cancer Therapy: Clinical
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Clinical Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement