Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jan 1.
Published in final edited form as: J Thorac Oncol. 2013 Jan;8(1):19–30. doi: 10.1097/JTO.0b013e31827628ff

Significance of Folate Receptor alpha and Thymidylate Synthase Protein Expression in Patients with Non-Small Cell Lung Cancer treated with Pemetrexed

Daniel C Christoph 1,4, Bernadette Reyna Asuncion 1, Biftu Hassan 1, Cindy Tran 1, Julia D Maltzman 3, Daniel J O’Shannessy 3, Murry W Wynes 1, Thomas C Gauler 4,6, Jeremias Wohlschlaeger 5, Mathias Hoiczyk 4, Martin Schuler 4,6, Wilfried E Eberhardt 4,6, Fred R Hirsch 1,2
PMCID: PMC3645936  NIHMSID: NIHMS460563  PMID: 23242435

Abstract

Introduction

Folate receptor alpha (FRA) regulates cellular uptake of folates and antifolates. Information about FRA protein expression in metastatic Non-Small Cell Lung Cancer (NSCLC) is limited. We investigated FRA as a biomarker for pemetrexed-based chemotherapy and compared it to thymidylate synthase (TS), the main target of pemetrexed.

Materials and Methods

Pre-treatment tumor specimens from 207 NSCLC patients with advanced disease were assessed for FRA and TS protein expression by immunohistochemistry using the H-score (range: 0–300) and correlated to patients’ clinicopathological data, radiographic response (RECIST), progression-free survival (PFS) and overall survival (OS).

Results

Low total (cytoplasmic and nuclear) TS protein expression (H-score <210) was associated with improved PFS (median: 5.6 vs. 3.5 months; HR=0.6379, P=0.0131) and prolonged OS (median: 22.5 vs. 11.5 months; HR: 0.5680, P=0.0107). An association between lower TS levels and response to pemetrexed-based therapy was found: mean H-score 187±5, median 180 for responders vs. 201±4, median 210, P=0.0244. High intracellular FRA expression (H-score ≥ 110) was associated with prolonged OS (28.9 vs. 11.7 months, HR=0.5316, P=0.0040) and a trend for association with PFS (5.6 vs. 4.1 months, HR=0.7395, P=0.0801) was noted. Membranous FRA expression was seen in 83% of patients, moreover, high membranous expression (H-score ≥20) was associated with improved PFS (5.6 vs. 3.7 months, HR=0.6445, P=0.0306) and OS (22.1 vs. 11.5 months, HR=0.5378, P=0.0131).

Conclusions

A large number of NSCLC patients have high expression of FRA and/or low level of TS expression. Expression levels of FRA and TS were associated with clinical benefit from pemetrexed-therapy.

Keywords: Non-Small Cell Lung Cancer, Biomarker, Folate Receptor alpha, FRA, Thymidylate Synthase, TS, Pemetrexed

INTRODUCTION

Lung cancer is the leading cause of cancer deaths worldwide and Non-Small Cell Lung Cancer (NSCLC) accounts for 80% of all lung cancer cases1. The standard first-line therapy for patients with advanced NSCLC is a platinum-based doublet combination chemotherapy with or without bevacizumab2. In clinical practice, pemetrexed, a multitarget antifolate drug, is used in combination with cisplatin3 or carboplatin4 in non-squamous cell carcinomas. Furthermore, it is administered as continuous maintenance therapy after platinum-based chemotherapy, as single agent after progression of first-line therapy5,6 or it is given to patients who are medically unfit for platinum-based combination chemotherapy7. Pemetrexed primarily inhibits thymidylate synthase (TS). But at higher concentrations it also blocks folate-dependent enzymes such as dihydrofolate reductase (DHFR) and glycinamide ribonucleotide formyl transferase (GARFT), all of which are involved in the de novo biosynthesis of thymidine and purine nucleotides8.

Predictive biomarkers have proven to be effective for decisions regarding treatment with specific molecular targeted agents. However, for pemetrexed few studies have investigated predictive biomarkers for treatment efficacy and the majority of these have focused on TS itself. Previous studies have shown that low TS protein levels are associated with prolonged progression-free survival (PFS) of NSCLC patients undergoing pemetrexed-based treatment9,10. However, inconsistent results have been reported regarding the association between objective response (OR), overall survival (OS) and TS expression levels9,10. An association between improved OS and low TS levels was found in only one subgroup of adenocarcinoma patients9, but information about Caucasian patients is very limited.

For the transport of folates and antifolates into eukaryotic cells, three transporters have been identified: the ubiquitous reduced folate carrier (RFC), the proton-coupled folate transporter (PCFT), and a family of folate receptors of which folate receptor α (FRA) is the most widely studied11,12. However, FRA-mediated transport contributes little to the uptake of either folates or antifolates unless the receptor is highly expressed relative to RFC, and/or transport mediated by RFC and other routes is impaired13. FRA is a glycosylphosphatidylinositol (GPI) anchored cell surface protein that is able to bind free folate with high affinity14 and assists in the uptake of antifolates via a classical mechanism of receptor-mediated endocytosis. Although FRA has a very limited expression profile in normal adult tissues, it is expressed in lung tissues15, in type I and II pneumocytes1620, basilar and mucociliary bronchiolar cells and bronchial epithelium1820.

High expression levels of FRA have been found in certain cancer cells of epithelial origin including certain lung tumors19,21,22. Lung tumors that retain alveolar epithelial cell characteristics, such as adenocarcinoma, bronchioloalveolar carcinoma and large cell carcinoma, usually express detectable amounts of FRA protein21,23.

Novel therapeutic agents directed against FRA have been developed within the last few years and are currently under clinical investigation for the treatment of lung cancer. These agents are either conjugates of a cytotoxic agent with folic acid (e.g. EC-145, Endocyte, Inc., West Lafayette, IN)24 or antibodies eliciting robust antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) (e.g. farletuzumab, Morphotek, Inc., Exton, PA)25. In vitro experiments showed that neither loss of constitutive FRA expression by RNA interference26 nor the addition of a cytotoxic antibody (e.g. farletuzumab) significantly inhibited the transport of pemetrexed into NSCLC cells or interfered with the efficacy of pemetrexed27.

Very limited information about FRA expression in metastatic NSCLC is available. This limits the development of potential novel drugs targeting this receptor. Early and locally advanced adenocarcinomas, large cell carcinomas and carcinoid tumors have been shown to express FRA protein, but protein expression of FRA in squamous cell carcinomas and small-cell carcinomas has rarely been observed18,23,28. FRA gene expression as measured by real-time polymerase chain reaction (RT-PCR) in NSCLC patients with well-differentiated, early p-stage, pT1, pN0 cancers was shown to be significantly higher than those in poorly differentiated, advanced p-stage, pT2-4, pN1-3 patients29. The disease-free survival (DFS) and 3-year survival rates of surgically treated patients with tumors expressing high FRA mRNA levels were significantly higher than those of patients with low FRA mRNA levels29. In contrast, no associations between intracellular or membranous expression of FRA and DFS or OS were found in a study of 320 patients with early or locally advanced NSCLC23. Recently, a significant association between improved OS and higher levels of membranous FRA expression in adenocarcinomas of the lung was reported28.

Currently, there are no clinically validated biomarkers to predict response or outcome following pemetrexed-based chemotherapy. Therefore, we investigated the association of FRA and TS expression detected by immunohistochemistry (IHC) and clinical outcome following pemetrexed-based chemotherapy in a cohort of Caucasian patients with advanced NSCLC. Furthermore, we investigated the expression patterns of FRA and TS in advanced NSCLC and its correlation with clinicopathological parameters.

MATERIALS AND METHODS

Study Population

The study included all patients with NSCLC, who received pemetrexed-based chemotherapy at any chemotherapy line at the West German Cancer Center between November 2004 and October 2011 for the treatment of their metastatic disease. Analysis of tumor samples, which were available following routine diagnostic histopathological workup, provided adequate pre-treatment biopsies from 207 patients (Table 1). Clinicopathological data included age, gender, histology, complete history and radiographic findings (detailed information about the distribution of N-stages for the different histological subtypes is shown in Supp. Table 1). Tumor staging was based on the tumor, node, and metastasis (TNM) staging system (7th edition) as proposed by the International Association for the Study of Lung Cancer (IASLC)30. Response Evaluation Criteria in Solid Tumors (RECIST) version 1.131 was applied for the evaluation of response of NSCLC patients to treatment. PFS was calculated from the first day of chemotherapy treatment until progression or the last visit where a patient was alive without progression. OS was defined as the time between the start of pemetrexed-based chemotherapy until the date of death, or the date of last follow-up. Patients were censored at the last follow-up if still alive or lost to follow-up. Surveillance of PFS and OS for this study was stopped on February 1, 2012. Living patients provided written informed consent for the tissue used in this study. The study was approved by the Ethics Committee of the Medical Faculty of the University Duisburg-Essen (no. 10-4404).

Table 1.

Clinicopathological data of NSCLC patients: CR: complete remission; IASLC: International Association for the Study of Lung Cancer; OS: overall survival; PD: progressive disease; PFS: progression-free survival; PR: partial remission; and SD: stable disease. Note that percentages may not total 100 because of rounding.

Epidemiologic and clinicopathological data Number of patients (%)
207 (100%)
Age
Median age at diagnosis in years (range) 59 (23–90)
Gender
Female 94 (45%)
Male 113 (55%)
T-stage
T1 24 (12%)
T2 79 (38%)
T3 37 (18%)
T4 67 (32%)
N-stage
N0 38 (18%)
N1 18 (9%)
N2 81 (39%)
N3 70 (34%)
M-stage
M0 80 (39%)
M1 127 (61%)
IASLC-stage
I 8 (4%)
II 14 (7%)
III 58 (28%)
IV 127 (61%)
Histologic subtype
Adenocarcinoma 156 (75%)
Large cell carcinoma 12 (6%)
Squamous cell carcinoma 12 (6%)
Adenocarcinoma with large cell carcinoma 5 (2%)
Adenocarcinoma with squamous cell carcinoma 5 (2%)
Other combined tumors 10 (5%)
Other carcinomas 4 (2%)
Carcinoma not otherwise specified 3 (1%)
Histological grading
G1 8 (4%)
G2 63 (30%)
G3 130 (63%)
G4 3 (1%)
Unspecified 3 (1%)
Characteristics Number of patients (%)
207 (100%)
Chemotherapy
Pemetrexed plus cisplatin 69 (33%)
Pemetrexed plus carboplatin 31 (15%)
Pemetrexed plus cisplatin and carboplatin 2 (<1%)
Pemetrexed plus other compounds 5 (2%)
Pemetrexed single-agent 100 (48%)
Median number of cycles (range) 4 (1–23)
Median cumulative pemetrexed dose in mg (range) 3,235 (600–26,080)
Median duration of pemetrexed-based therapy in months (range) 2.8 (0.7–18.9)
Chemotherapy line
1st line 77 (37%)
2nd line 59 (29%)
3rd line 37 (18%)
4th line 20 (10%)
>4th line 14 (7%)
Response to pemetrexed-based treatment
CR 8 (4%)
PR 68 (33%)
SD 72 (35%)
PD 39 (19%)
Not evaluable 20 (10%)
Survival time from start of treatment to progression or death
Median PFS time in months (95% CI) 5.3 (4.7–6.0)
Median OS time in months (95% CI) 16.7 (10.9–22.4)

Tissue micro array (TMA) preparation

Sections were prepared from each formalin-fixed, paraffin-embedded (FFPE) tissue block and stained with hematoxylin and eosin for histological examination to identify 3 tumor-rich foci, which were marked with a cytology pen. A tissue arrayer (Beecher Instruments, Inc., Sun Prairie, WI, USA) was used to construct the TMA blocks. Three core samples of 1 mm size from marked tumor-rich regions of each donor FFPE block were sampled and transferred to a recipient paraffin block.

Immunohistochemistry (IHC) for FRA and TS

The TMA blocks were sectioned at 4 μm thick and mounted on PlusGOLD® slides (Thermo Scientific Fisher, Inc., Pittsburgh, PA, USA) for IHC staining. In cases of cores lost from TMA sections or inadequate tumor samples for TMA preparation, whole tissue sections were used.

The IHC detection of FRA protein has been recently described elsewhere28. Briefly, after deparaffinization, rehydration and heat-induced antigen-retrieval, IHC staining process was done using Dako Autostainer Universal Staining System (Dako, Carpentaria, CA, USA). Tissue was blocked for endogenous peroxidase activity. Unconjugated antibody, mouse monoclonal FRA (clone 26B314, dilution 2.5 μg/ml, Morphotek, Inc., Exton, PA, USA), was applied and incubated for 60 minutes at room temperature (RT). For negative control slide non-immunized unconjugated antibody, universal negative control mouse antibody (Dako), was applied and incubated under the same conditions. MACH4 Mouse Probe Primary Antibody Enhancer (Biocare Medical, Concord, CA, USA) was applied and incubated for 15 minutes followed by Polymer-HRP reagent (Biocare) for 20 minutes at RT. Visual color was developed with 3-3′-diaminobenzidine (DAB) tetra-hydrochloride (Dako). Slides were counterstained with Hematoxylin (Dako).

As FRA is expressed in the proximal tubules of the kidney, human kidney sections were used as a positive control for FRA IHC23,28. Figure 1A shows highly specific staining of the FRA antibody, since FRA expression is restricted to the luminal surface of proximal tubule cells17,19.

Figure 1.

Figure 1

IHC for folate receptor alpha (FRA) and thymidylate synthase (TS) in Non-Small Cell Lung Cancer (NSCLC) samples with different protein expression levels. A–E: IHC with an antibody against FRA (dilution 2.5 μg/ml). A) renal tissue was used as positive control, B) negatively, C) weakly, D) moderately, and E) strongly stained NSCLC sample. F–I: IHC with an antibody against TS (dilution 50 μg/ml). F) colorectal carcinoma was used as positive control; G) weakly, H) moderately, and I) strongly stained NSCLC sample. Magnification 1:400.

We have recently published the IHC procedure for TS32. Briefly, after baking the slides in dry oven for an hour, heat-induced antigen retrieval, and deparaffinization, the IHC staining procedure was performed using the Benchmark XT autostainer (Ventana, Tucson, AZ, USA). Endogenous peroxidase activity was blocked and unconjugated mouse anti-TS antibody (clone 4H4B1, dilution 50 μg/ml, Invitrogen, Frederick, MD, USA) was manually applied and incubated for 1 hour at RT. Tissue sections of colorectal carcinoma were included as controls, and lymphocytes were used as an intra-specimen positive control. For negative control, unconjugated non-immunized ready-to-use mouse antibody (Ventana) was applied as the primary antibody. Staining visualization was accomplished with ultraView Universal DAB detection kit (Ventana), post counterstaining with Bluing Reagent (Ventana).

IHC evaluations were performed independently by a pathologist (B.R.A) and a trained reader (D.C.C.). Ambiguous cases were reevaluated jointly until a consensus was reached. Furthermore, we performed a round-robin test on a subset of patients with the Laboratory Corporation of America (Los Angeles, CA, USA) revealing highly similar H-scores. Immunoreactivity levels in each section were assessed under a light microscope and images were captured at 400-fold magnification. Tumor staining intensity was graded on a scale from 0+ to 3+ and within each intensity category the percentage of tumor cells was noted. The percentage score was then multiplied by its intensity category to obtain a final “H-score”, ranging from 0 to 30023,33. The highest H-score of the triplicate scores per patient was used for further analyses32. Tumor samples representing different H-scores for either FRA or TS are shown in Figure 1.

Evaluation of total TS protein expression was based on nuclear and cytoplasmic staining9,33,34. For FRA, varied staining intensities and patterns of both the intracellular and membranous compartments were observed between cases22,23. While FRA is a GPI-anchored receptor that cycles into the cell via an endocytic mechanism, remaining membrane associated, we decided to evaluate FRA staining for both the intracellular and membrane compartments separately.

Statistics

The objective of this study was to assess the impact of the two selected biomarkers (FRA and TS) on treatment response, and the outcome of patients with NSCLC. Another goal was to evaluate the intracellular and membranous FRA expression in metastatic NSCLC specimens to determine their expression pattern in different histological subtypes. Descriptive analyses were performed on these H-scores and correlated with patient’s clinical data: age at diagnosis, gender, histological grade or subtype (adenocarcinoma vs. large cell carcinoma vs. squamous cell carcinoma), and TNM/IASLC stage. A Spearman’s test35 was used to further correlate as continuous variables intracellular and membranous FRA H-scores, total TS H-score, and age at diagnosis. Distributional differences of H-scores between different levels of categorical variables (response, gender, histological grade, subtype, or stage) were analyzed using either the Kruskal-Wallis test or Wilcoxon ranked sum test (equivalent to Kruskal-Wallis test when comparing two groups)36. Receiver operating characteristic (ROC) curves37 were constructed by calculating the sensitivity and specificity at several cutoff-values. The optimal cutoff-value was then given by the maximum of the Youden index38.

For the Kaplan-Meier (KM) curves of PFS and OS, we tested binary H-score cutoff-values of biomarkers and applied the maximum chi-square method28,39,40. Univariate and multivariate analyses of both PFS and OS were performed using the Cox proportional hazards model. All variables with P-values less than 0.10 at the time of univariate regression analysis were entered into the multivariate analysis model. For the assessment between survival and combined expression of FRA and TS, the H-score of total FRA expression (intracellular plus membranous expression) was determined by calculating the mean of intracellular and membranous expression. Furthermore, subgroup analyses for patients with first-line treatment and for patients with adenocarcinomas were calculated. A P-value less than 0.05 was considered statistically significant. Nevertheless, as this is a retrospective study all P-values or results of statistical tests should be regarded as exploratory. Statistical analyses were performed by using GraphPad Prism (Version 5.00 for Windows, GraphPad Software, SanDiego, CA, USA) or SPSS software package (Version 20, SPSS Inc., Chicago, IL, USA).

RESULTS

Patient cohort

Demographic and clinicopathologic data of the patients are shown in Table 1. Median PFS and OS of the entire cohort were 5.3 months (95%-CI: 4.7–6.0) and 16.7 months (95%-CI: 10.9–22.4), respectively. The main histologic subtype in the present cohort was adenocarcinoma, occurring in 156 patients (75%); large cell carcinoma and squamous cell carcinoma subtypes were each found in 12 patients (6%). Median PFS and OS of adenocarcinoma patients were 5.4 months (95%-CI: 4.6–6.2) and 16.7 months (95%-CI: 10.1–23.2), respectively. The majority of patients were males (n=113, 55%). 77 (37%) patients received pemetrexed-based first-line chemotherapy and in this subgroup median PFS was 7.3 months (95%-CI: 5.4–9.2) and median OS was 39.6 months (95%-CI: 19.4–59.9).

Intratumoral comparison of FRA and TS expression

Due to technical reasons, IHC and H-scoring was only successful for FRA in 207 cases, and 196 cases for TS. Intracellular expression of FRA was strongly correlated with membranous expression (Figure 2A, Spearman’s correlation; r=0.8460; P<0.0001). Similarly, strong correlations in the subgroups of adenocarcinoma or large cell carcinoma patients were found (Suppl. Figures 1A/B). A weaker, but still significant correlation in squamous cell carcinomas was observed (r=0.6597, P=0.0196; Suppl. Figure 1C). There were no correlations between TS protein expression and intracellular (r=−0.1029, P=0.1513) or membranous FRA expression (r=−0.1227; P=0.0868).

Figure 2.

Figure 2

A) Intratumoral correlation of intracellular and membranous protein expression of FRA. Scatter-plots of (B) TS, (C) intracellular, and (D) membranous FRA protein expression in different histological subtypes (vertical line: median). ADC: adenocarcinoma, LCC: large cell carcinoma, SCC: squamous cell carcinoma.

Since data on FRA expression in metastatic NSCLC are limited, we were particularly interested in the incidence of intracellular and membranous FRA expression. Intracellular or membranous FRA expression (H-score higher than 5) were detected in 192 patients (93%) or 171 patients (83%), respectively. As an H-score of ≥ 5 is a low cutoff-value, we tested higher cutoff-values as well. Considering an H-score of ≥ 100, 140 patients (68%) or 99 patients (48%) had tumors with high intracellular or membranous expression. Using an H-score of ≥ 200 as cutoff-value to distinguish between low and high FRA expressing tumors 42 patients (20%) or 30 patients (15%) showed tumors with strong intracellular or membranous expression. To summarize, the incidence of FRA membrane expression observed in the present study compares favorably with a recent report28.

Association between clinicopathological data and TS expression

The median H-score for total TS expression was 200 (range: 60–290). There was no statistically significant correlation between age, sex, histological grades or subtypes (Figure 2B), T- or M-stages and total TS protein levels (Table 2A). N-stages were associated with total TS protein expression (PKW=0.0003). Interestingly, lower total TS protein expression was associated with early N-stages at diagnosis (mean for N0/N1: 174±6, median 160 vs. mean for N2/N3: 203±3, median 207.5) (P<0.0001). Lower total TS protein levels were also found in early IASLC stages (mean for IASLC stages I+II: 176±11, median 160) compared to advanced IASLC stages (mean for IASLC stages III + IV: 198±3, median 200; P=0.0331).

Table 2.

Associations between clinicopathological data and (A) total TS protein expression, (B) intracellular or (C) membranous protein expression of FRA. IASLC: International Association for the Study of Lung Cancer.

Table 2A
Characteristic Number of patients (%) Mean total TS H-score ± SEM, median P-value
Histological subtype PKW=0.9902
Adenocarcinoma 147 (75%) 196 ± 3, 190
Large cell carcinoma 12 (6%) 184 ± 17, 202.5
Squamous cell carcinoma 10 (5%) 191 ± 16, 205
Histological subtype P=0.9856
Adenocarcinoma 147 (75%) 196 ± 3, 190
Squamous cell carcinoma 10 (5%) 191 ± 16, 205
Gender P=0.5982
Female 90 (46%) 196 ± 5, 200
Male 106 (54%) 196 ± 4, 200
Grading PKW=0.5688
G1 7 (4%) 213 ± 12, 210
G2 58 (30%) 192 ± 5, 190
G3 126 (65%) 196 ± 4, 200
G4 3 (2%) 207 ± 7, 200
Grading P=0.4339
G2 58 (30%) 192 ± 5, 190
G3 126 (65%) 196 ± 4, 200
T-stage PKW=0.3635
T1 24 (12%) 190 ± 8, 190
T2 74 (38%) 200 ± 5, 200
T3 35 (18%) 187 ± 7, 190
T4 63 (32%) 196 ± 5, 200
N-stage PKW=0.0003
N0 37 (19%) 173 ± 7, 160
N1 17 (9%) 176 ± 12, 160
N2 75 (38%) 205 ± 4, 220
N3 67 (34%) 201 ± 5, 205
N-stage P<0.0001
N0 + N1 54 (28%) 174 ± 6, 160
N2 + N3 142 (72%) 203 ± 3, 207.5
M-stage
M0 76 (39%) 193 ± 5, 200 P=0.7362
M1 120 (61%) 197 ± 4, 200
IASLC-Stage PKW=0.1586
IASLC I 8 (4%) 174 ± 15, 160
IASLC II 13 (7%) 178 ± 16, 160
IASLC III 55 (28%) 199 ± 6, 200
IASLC IV 120 (61%) 197 ± 4, 200
IASLC-Stage P=0.0331
IASLC I + II 22 (11%) 176 ± 11, 160
IASLC III + IV 185 (89%) 198 ± 3, 200
Table 2B
Characteristic Number of patients (%) Mean intracellular FRA H-score ± SEM, median P-value
Histological subtype PKW=0.0368
Adenocarcinoma 156 (77%) 134 ± 6, 130
Large cell carcinoma 12 (6%) 92 ± 23, 67.5
Squamous cell carcinoma 12 (6%) 87 ± 19, 90
Histological subtype P=0.0386
Adenocarcinoma 156 (77%) 134 ± 6, 130
Squamous cell carcinoma 12 (6%) 87 ± 19, 90
Gender P=0.2520
Female 94 (45%) 134 ± 8, 135
Male 113 (55%) 122 ± 7, 110
Grading PKW=0.0318
G1 8 (4%) 105 ± 25, 105
G2 63 (30%) 151 ± 9, 155
G3 130 (63%) 121 ± 7, 110
G4 3 (1%) 57 ± 32, 30
Grading P=0.0173
G2 63 (30%) 151 ± 9, 155
G3 130 (63%) 121 ± 7, 110
T-stage PKW=0.3841
T1 24 (12%) 147 ± 13, 157.5
T2 79 (38%) 129 ± 9, 125
T3 37 (18%) 130 ± 12, 120
T4 67 (33%) 118 ± 10, 110
N-stage PKW=0.0120
N0 38 (18%) 148 ± 11, 152.5
N1 18 (9%) 167 ± 16, 172.5
N2 81 (39%) 114 ± 9, 100
N3 70 (34%) 123 ± 9, 110
N-stage P=0.0014
N0 + N1 56 (27%) 154 ± 9, 110
N2 + N3 151 (73%) 118 ± 6, 70
M-stage
M0 80 (39%) 135 ± 8, 132.5 P=0.1916
M1 127 (61%) 123 ± 7, 110
IASLC-Stage PKW=0.3385
IASLC I 8 (4%) 153 ± 16, 150
IASLC II 14 (7%) 146 ± 22, 150
IASLC III 58 (28%) 131 ± 10, 125
IASLC IV 127 (61%) 123 ± 7, 110
IASLC-Stage P=0.0848
IASLC I + II 22 (11%) 153 ± 14, 150
IASLC III + IV 185 (89%) 125 ± 6, 120
Table 2C
Characteristic Number of patients (%) Mean membranous FRA H-score ± SEM, median P-value
Histological subtype PKW=0.0059
Adenocarcinoma 156 (77%) 103 ± 7, 97.5
Large cell carcinoma 12 (6%) 59 ± 20, 30
Squamous cell carcinoma 12 (6%) 46 ± 24, 0
Histological subtype P=0.0066
Adenocarcinoma 156 (77%) 103 ± 7, 97.5
Squamous cell carcinoma 12 (6%) 46 ± 24, 0
Gender P=0.0325
Female 94 (45%) 108 ± 8, 120
Male 113 (55%) 86 ± 8, 60
Grading PKW=0.0375
G1 8 (4%) 66 ± 25, 55
G2 63 (30%) 116 ± 11, 130
G3 130 (63%) 91 ± 7, 70
G4 3 (1%) 20 ± 20, 0
Grading P=0.0674
G2 63 (30%) 116 ± 11, 130
G3 130 (63%) 91 ± 7, 70
T-stage PKW=0.3585
T1 24 (12%) 118 ± 15, 135
T2 79 (38%) 98 ± 9, 90
T3 37 (18%) 96 ± 13, 105
T4 67 (33%) 86 ± 10, 60
N-stage PKW=0.1770
N0 38 (18%) 106 ± 12, 120
N1 18 (9%) 129 ± 19, 140
N2 81 (39%) 89 ± 9, 70
N3 70 (34%) 90 ± 9, 67.5
N-stage P=0.0398
N0 + N1 56 (27%) 114 ± 10, 130
N2 + N3 151 (73%) 89 ± 7, 70
M-stage
M0 80 (39%) 105 ± 9, 105 P=0.1916
M1 127 (61%) 90 ± 7, 70
IASLC-Stage PKW=0.4508
IASLC I 8 (4%) 78 ± 27, 80
IASLC II 14 (7%) 111 ± 23, 135
IASLC III 58 (28%) 107 ± 11, 100
IASLC IV 127 (61%) 90 ± 7, 70
IASLC-Stage P=0.9624
IASLC I + II 22 (11%) 100 ± 17, 125
IASLC III + IV 185 (89%) 95 ± 6, 90

TS expression is associated with treatment response

Low total TS expression was associated with objective response (OR) (mean 187±5, median 180 for responders (CR+PR) vs. 201±4, median 210 for non-responders (SD+PD), P=0.0244) (Suppl. Table 2A), but not disease control (DC) meaning objective response or disease stabilization (mean 194±4, median 190 for CR/PR/SD vs. 201±7, median 210 for PD, P=0.1375) (Suppl. Table 2A). As we observed a significant association between low total TS protein levels and OR, we calculated ROC curves analyzing TS and response. We discovered an area-under-curve (AUC) value of 0.5997 (P=0.0247). We used the optimal cutoff of 197.5 before estimating the sensitivity, specificity, positive and negative predictive value for response, which were 62%, 60%, 70%, and 52%, respectively.

Association between survival and TS expression

In the univariate analysis considering TS, low total TS expressers (H-score <210, χ2=6.151) had a significantly longer PFS (5.6 vs. 3.5 months) as compared to high expressers (P=0.0131; HR=0.6379, 95%-CI: 0.4472–0.9100) (Figure 3A). A similar association between TS and OS was observed: low TS expressers (H-score <210, χ2=6.516) had a prolonged OS (22.5 vs. 11.5 months) as compared to high expressers (P=0.0107; HR=0.5680, 95%-CI: 0.3678–0.8769) (Figure 3B). We performed a Cox proportional hazard model for PFS/OS and total TS protein levels. In the univariate analysis the association between PFS and total TS protein levels remained significant (PCOX=0.047). No significant associations between clinicopathological parameters and PFS were observed. The univariate analysis considering continuous TS protein levels showed a trend for an association between OS and TS (PCOX=0.077), but no association was observed after adjustment for histological grades and N-stages (PCOX=0.152). We performed survival analyses for the subgroups of patients who received first-line treatment or suffered from adenocarcinomas (Suppl. Figures 2A/B and 3A/B). Both revealed similarly significant results.

Figure 3.

Figure 3

Analyses of total thymidylate synthase (TS) protein expression and (A) progression-free survival (PFS) or (B) overall survival (OS); analyses of intracellular protein expression of folate receptor alpha (FRA) and (C) PFS or (D) OS; analyses of membranous FRA protein expression and (E) PFS or (F) OS.

Association between clinicopathological data, response and intracellular FRA expression

The median H-score for intracellular FRA expression was 120 (range: 0–290). Intracellular expression was significantly higher in adenocarcinomas (mean 134±6, median 130) compared to large cell carcinomas (mean 92±23, median 67.5) or squamous cell carcinomas (mean 87±19, median 90) (PKW=0.0368) (Figure 2C). Differentiation of the tumors was significantly associated with intracellular expression (PKW=0.0318) (Table 2B). Higher intracellular levels were found in moderately differentiated tumors (G2: mean 151±9; median 155) compared to poorly differentiated tumors (G3: mean 121±7, median 110; P=0.0173). T- or M-stages were not associated with intracellular expression. But an association between N-stages and intracellular expression was found (PKW=0.0120). Of note, higher intracellular levels were observed in N0/N1-stages (mean H-score: 154±9, median 110) compared to N2/N3-stages (mean H-score 118±6, median 70, P=0.0014). No association between response and intracellular FRA expression levels was noted.

Association between survival and intracellular FRA expression

A trend for a significant association between high intracellular FRA expression (H-score ≥ 110, χ2=3.063) and prolonged PFS (5.6 vs. 4.1 months; P=0.0801; HR=0.7395, 95%-CI: 0.5275–1.037) was observed (Figure 3C). High expressers of intracellular FRA (H-score ≥ 110, χ2=8.272) were associated with an improved OS (28.9 vs. 11.7 months) as compared to low expressers (P=0.0040; HR=0.5316, 95%-CI: 0.3456–0.8177) (Figure 3D). We performed a Cox proportional hazard model for OS and intracellular FRA protein levels. In the univariate analysis the association between OS and intracellular protein levels was significant (PCOX=0.021), but when adjusting for histological grades and N-stages in the multivariate analysis, there was a trend for an association (PCOX=0.056). Survival analyses for the subgroups of first-line treatment or adenocarcinoma patients (Suppl. Figures 2C/D and 3C/D) showed that high expressers had an improved OS (P=0.0203 or P=0.0028, respectively). However, no significant associations between intracellular FRA expression and PFS were found in theses subgroups (P=0.1224 or P=0.1759, respectively).

Association between clinicopathological data, response and membranous FRA expression

The median H-score for membranous FRA expression was 90 (range: 0–290). FRA was significantly more expressed in adenocarcinomas (mean 103±7, median 97.5) compared to large cell carcinomas (mean 59±20, median 30) or squamous cell carcinomas (mean 46±20, median 0) (PKW=0.0059) (Figure 2D). Membranous expression was significantly higher in females (mean 108±8, median 120) than in males (mean 86±8, median 60) (P=0.0325) and significantly associated with differentiation of the tumors (PKW=0.0375). The difference between membranous levels of moderately differentiated carcinomas (G2: mean H-score 116±11; median 130) and poorly differentiated carcinomas (G3: mean H-score 91±7, median 70) showed a trend for a significant association (P=0.0674). T- or M-stages were not associated with membranous expression (Table 2C). Higher membranous levels were observed in N0/N1-stages (mean 114±10, median 130) compared to N2/N3-stages (mean 89±7, median 70, P=0.0398). No association between response and membranous FRA expression levels was observed.

Association between survival and membranous FRA expression

High membranous FRA expression (H-score ≥ 20, χ2=4.676) was associated with a prolonged PFS (5.6 vs. 3.7 months, P=0.0306, HR=0.6445; 95%-CI: 0.4328–0.9597; Figure 3E). Similarly, high expressers (H-score ≥ 20, χ2=6.156) showed an improved OS (22.1 vs. 11.5 months, P=0.0131, HR=0.5378, 95%-CI: 0.3294–0.8778; Figure 3F). Again, we performed survival analyses for the subgroups of first-line treatment or adenocarcinoma patients (Suppl. Figures 2E/F and 3E/F). Strong associations between prolonged OS and high membranous FRA expression were seen in both of these subgroups (P<0.0001 or P=0.0064, respectively). There was a trend for an association between high membranous expression and improved PFS in the subgroup of first-line treatment patients (P=0.0514), and the association was significant in the subgroup of adenocarcinoma patients (P=0.0423).

Analyses of survival and combined TS/FRA expression

We performed survival analyses considering the combined total FRA and TS expression (Figure 4). PFS of patients with tumors expressing high levels of total FRA and low total TS levels was prolonged (5.8 months) compared to patients with low total FRA and high total TS levels expressing tumors (3.1 months; P=0.0033; HR=0.4071, 95%-CI: 0.2235–0.7414; Figure 4C). Similarly, patients with tumors expressing high levels of total FRA and low total TS levels had a significant improvement to OS of 28.9 months compared to patients with tumors showing low levels of total FRA and high total TS levels (6.9 months; P=0.0045; HR=0.2133, 95%-CI: 0.07341–0.6198; Figure 4D) or compared to patients with tumors containing high levels of both total FRA and total TS (15.2 months; P=0.0205, HR= 0.5650, 95%-CI: 0.3486–0.9157; Figure 4F).

Figure 4.

Figure 4

Analysis of the combination of total folate receptor alpha (FRA) and thymidylate synthase (TS) protein expression and (A) progression-free survival (PFS) or (B) overall survival (OS); comparison of survival times between high total FRA plus low total TS expressers and low total FRA plus high total TS-expressers (C/D) or high total FRA plus high total TS-expressers (E/F). For the categorization of low expressers or high expressers the following H-scores were used: total TS: 210 (PFS χ2=6.151, OS χ2: 6.516), total FRA: 60 (PFS χ2=4.032) or 20 (OS χ 2=5.541).

DISCUSSION

Pemetrexed is a multitarget antifolate drug approved for the treatment of advanced NSCLC. While the mechanism for the drug activity is known, no predictive marker has so far been validated for better selection of patients, in contrast to other targeted treatments. The identification of predictive markers for effective therapy would be helpful for maximizing therapeutic efficacy and minimizing non-effective treatment in patients with cancer. Therefore, novel strategies for the prediction of clinical response for known antitumor agents, such as pemetrexed, are urgently required for individualized therapy.

In the current study we confirmed an association between OR and low total TS protein expression (P=0.0244), which was shown recently10. In our study, low TS-expressers had a median PFS time of 5.6 months compared to 3.5 months of high TS-expressers (P=0.0131). Similarly, median OS time was improved in low TS-expressers (22.5 months) compared to high TS-expressers (11.5 months, P=0.0107). Our results are partly in accordance with other reports9,10, as we observed an association between median PFS or OS times and TS expression levels. However, in the aforementioned studies, OS was only associated with TS levels in patients with adenocarcinomas9. Surprisingly, we did not find any association between TS protein levels and the histological subtypes of adenocarcinoma, large cell carcinoma, and squamous cell carcinoma. Some studies reported higher TS mRNA levels in squamous cell carcinomas compared to adenocarcinomas of the lung41,42. However, our results are consistent with another study using automated protein in situ quantification (AQUA)43 and our recently published study32, although these studies32,43 investigated TS protein expression in early and locally-advanced NSCLC. Furthermore, it should be mentioned that the percentages of large cell or squamous cell carcinomas in our study is quite low (6% each, respectively), thus limiting the comparison of FRA and TS expression between the different histological subtypes. These patients received pemetrexed until this drug was exclusively approved for metastatic non-squamous NSCLC patients in 2009. Although we found significant associations between OR, PFS, OS, and TS protein expression, it should be noted that the AUC value of the ROC curve predicting response (0.5997) does not appear to support the idea that TS is a good predictive biomarker.

No associations between response and intracellular or membranous FRA levels were found, neither intracellular nor membranous FRA expression are robust enough for a predictive biomarker. These results may not be unexpected as a study investigating FRA in Malignant Pleural Mesothelioma did not reveal an association between total FRA protein expression and response to pemetrexed-based chemotherapy44. Additionally, in the latter study, no associations between time-to-treatment failure or OS and total FRA expression were noted. However, in our study we investigated FRA expression in metastatic NSCLC in the intracellular and membrane compartments separately and found associations between high intracellular or membranous FRA expression and improved OS (P=0.0040 or P=0.0131, respectively). Only high membranous FRA expression was associated with a prolonged PFS (5.6 vs. 3.7 months, P=0.0306). Yet due to the lack of a control group (cohort of patients with pulmonary adenocarcinomas without pemetrexed-based treatment) we were unable to evaluate the prognostic value of intracellular or membranous FRA expression.

In our study of NSCLC patients with advanced disease, higher intracellular or membranous FRA levels were observed in more differentiated tumors. Assessment of FRA gene expression in early and locally advanced stages of NSCLC (n=117) revealed similar results29. No associations between intracellular or membranous FRA expression and T- or M-stages were observed. But early N-stages were associated with higher expression levels of this protein in our series of metastatic NSCLC, consistent with results from another study with early and locally advanced NSCLC patients29.

We examined the incidence of membranous FRA expression in metastatic NSCLC. Levels of membranous FRA expression (H-score ≥ 5) was detected in 171 patients (83%). But this H-score is a low cutoff-value, and we tested higher cutoff-values in addition such as an H-score of ≥ 100 revealing that about half of the patients (n=99, 48%) had tumors with higher membranous FRA levels. These results indicate that FRA is a potential target for novel therapeutics in metastatic NSCLC and cytotoxic-conjugated antibodies45,46 as well as antibodies inducing ADCC and CDCC25 are being tested in clinical trials. We found significant differences in FRA expression between adenocarcinomas and squamous cell carcinomas. Our results are consistent with other studies, in which FRA was predominantly expressed in adenocarcinomas, relative to squamous cell carcinomas18,28. However, it should be mentioned that in one of these studies the M-score was used, which is another semi-quantitative assessment of FRA protein expression28. To summarize, novel FRA-directed therapeutics should primarily be investigated in adenocarcinomas of the lung with greater differentiation.

We noted that the median H-score of intracellular FRA expression was higher than the median H-score of membranous expression, which is consistent with a recently published study23. However, the significance of intracellular FRA expression is unclear. The intracellular staining is most likely a combination of several things: a) some background staining in the assay; b) a result of the process of tissue preparation for IHC (as GPI-anchored protein, FRA is susceptible to leaching or stripping from the membrane during alcohol washes); c) FRA expression in the cytoplasm where FRA is synthesized in the endoplasmic reticulum47; and d) membrane-bound FRA undergoes an endocytic pathway48, in which clathrin-coated buds and vesicles and endosomal vacuoles are loaded with the receptor and transported intracellularly. However, only the membranous form of FRA is biologically active/relevant and transports folates and antifolates. Therefore, the associations between intracellular FRA expression and OS should be interpreted with caution.

While the current study demonstrates some encouraging results, there are several limitations. Firstly, this is a retrospective study. Only patients with available pre-treatment tumor specimens were included in our study. Secondly, the clinical response and survival data are based on a retrospective analysis of medical records and image readings and are not collected from a prospective clinical trial. Various quantitative and semi-quantitative methods have been employed in different studies for measuring FRA expression in tissue biopsies, including IHC, radioimmunoassays, quantitative autoradiography, and cytofluorimetric analysis (all of which use anti-FR antibodies) as well as quantitative RT-PCR, fluorescence in situ hybridization, and radioligand binding assays19. However, the applicability of many of those methods to paraffin-embedded tissues is limited49. Unfortunately, results from antibody-based methods can be hampered by factors such as non-specific binding to tissues, inaccessibility to the antigen, heterogenous antigen expression and/or poor tissue sampling, and the possibility that the antibody might not distinguish between functional FRA and non-functional FRA, both of which have been demonstrated in human cells19.

In conclusion, in NSCLC patients treated with pemetrexed-based chemotherapy, low total TS protein expression is associated with a better OR and an improved PFS/OS. For FRA, associations between prolonged PFS/OS and membranous expression were observed. Membrane-bound FRA was detected in a large percentage of NSCLC-patients indicating a clear target of FRA by novel therapeutics as a favorable goal. Future prospective studies regarding FRA and TS are needed for further validations.

Supplementary Material

2

Suppl. Table 1: Distribution of N-stages for the different histological subtypes of adenocarcinoma, large cell carcinoma, squamous cell carcinoma, exclusive of other histological subtypes. In general, the majority of the patients had higher N-stages with no significant differences between the histological subtypes being observed.

Suppl. Table 2: Association between protein expression of total TS and objective response (OR) or clinical disease control (DC). (A) Responding and non-responding patients of the entire cohort were compared and significant differences in the mean or median TS H-score for OR were found. (B) Comparison of patients who received first-line treatment revealed a trend for associations between the mean or median TS H-scores and OR or DC. (C) Patients with adenocarcinomas were compared and significant differences in the mean or median TS H-score for OR were observed.

3. Suppl. Figure 1.

Correlation between intracellular and membranous protein expression of folate receptor alpha (FRA) in different histological subtypes: (A) in adenocarcinomas (ADC), (B) in large cell carcinomas (LCC), (C) in squamous cell carcinomas (SCC), and (D) in all other histological subtypes (OTH).

4. Suppl. Figure 2.

Analysis of a subgroup of patients with first-line pemetrexed-based treatment. Analyses of total thymidylate synthase (TS) protein expression and (A) progression-free survival (PFS) or (B) overall survival (OS); analyses of intracellular protein expression of folate receptor alpha (FRA) and (C) PFS or (D) OS; analyses of membranous FRA protein expression and (E) PFS or (F) OS.

5. Suppl. Figure 3.

Analysis of a subgroup of patients with adenocarcinomas of the lung. Analyses of total thymidylate synthase (TS) protein expression and (A) progression-free survival (PFS) or (B) overall survival (OS); analyses of intracellular protein expression of folate receptor alpha (FRA) and (C) PFS or (D) OS; analyses of membranous FRA protein expression and (E) PFS or (F) OS.

Acknowledgments

Source of Funding:

This work was supported by a research grant from Morphotek, Inc., a subsidiary of Eisai Corporation of North America. D.C.C. received an IASLC Fellowship Award and travel support from Lilly Germany. J.D.M. and D.J.O. are employees of Morphotek, Inc., a subsidiary of Eisai, Inc.. T.C.G. received consulting fee or honorarium and travel support from Lilly Germany. W.E.E. received research funding, consulting fee and honoraria from Lilly Germany. F.R.H. received consulting fee from Eli Lilly.

This work was supported by an IASLC Fellowship Award (D.C.C.) and a research grant from Morphotek, Inc., a subsidiary of Eisai Corporation of North America. The authors would like to acknowledge A. Peglow, S. Fox, H. Loewendick, I. Perelmuter, and R. Daniels for collecting and preparing clinical data of the patients, B. Kamen for scientific advises and critical discussions, and S. Robinson for critical reading of the manuscript.

Footnotes

Conflicts of Interest

For the remaining authors no conflicts of interest were identified for this paper.

References

  • 1.Jemal A, Thun MJ, Ries LAG, et al. Annual report to the nation on the status of cancer, 1975–2005, featuring trends in lung cancer, tobacco use, and tobacco control. J Natl Cancer Inst. 2008;100:1672–1694. doi: 10.1093/jnci/djn389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Chang MH, Ahn JS, Lee J, et al. The efficacy of pemetrexed as a third- or fourth-line therapy and the significance of thymidylate synthase expression in patients with advanced non-small cell lung cancer. Lung Cancer. 2010;69:323–329. doi: 10.1016/j.lungcan.2009.12.002. [DOI] [PubMed] [Google Scholar]
  • 3.Scagliotti GV, Parikh P, von Pawel J, 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;26:3543–3551. doi: 10.1200/JCO.2007.15.0375. [DOI] [PubMed] [Google Scholar]
  • 4.Gronberg BH, Bremnes RM, Flotten O, et al. Phase III study by the Norwegian lung cancer study group: pemetrexed plus carboplatin compared with gemcitabine plus carboplatin as first-line chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol. 2009;27:3217–3224. doi: 10.1200/JCO.2008.20.9114. [DOI] [PubMed] [Google Scholar]
  • 5.Ciuleanu T, Brodowicz T, Zielinski C, 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;374:1432–1440. doi: 10.1016/S0140-6736(09)61497-5. [DOI] [PubMed] [Google Scholar]
  • 6.Belani CP, Wu Y-L, Chen Y-M, et al. Efficacy and Safety of Pemetrexed Maintenance Therapy versus Best Supportive Care in Patients from East Asia with Advanced, Nonsquamous Non-small Cell Lung Cancer: An Exploratory Subgroup Analysis of a Global, Randomized, Phase 3 Clinical Trial. J Thorac Oncol. 2012;7:567–573. doi: 10.1097/JTO.0b013e31823d4f9d. [DOI] [PubMed] [Google Scholar]
  • 7.Bearz A, Garassino I, Cavina R, et al. Pemetrexed single agent in previously treated non-small cell lung cancer: a multi-institutional observational study. Lung Cancer. 2008;60:240–245. doi: 10.1016/j.lungcan.2007.10.008. [DOI] [PubMed] [Google Scholar]
  • 8.Adjei AA. Pharmacology and mechanism of action of pemetrexed. Clin Lung Cancer. 2004;5:S51–S55. doi: 10.3816/clc.2004.s.003. [DOI] [PubMed] [Google Scholar]
  • 9.Chen C-Y, Chang Y-L, Shih J-Y, et al. Thymidylate synthase and dihydrofolate reductase expression in non-small cell lung carcinoma: the association with treatment efficacy of pemetrexed. Lung Cancer. 2011;74:132–138. doi: 10.1016/j.lungcan.2011.01.024. [DOI] [PubMed] [Google Scholar]
  • 10.Sun J-M, Han J, Ahn JS. Significance of Thymidylate Synthase and Thyroid Transcription Factor 1 Expression in Patients with Nonsquamous Non-small Cell Lung Cancer Treated with Pemetrexed-Based Chemotherapy. J Thorac Oncol. 2011;6:1392–1399. doi: 10.1097/JTO.0b013e3182208ea8. [DOI] [PubMed] [Google Scholar]
  • 11.Assaraf YG. Molecular basis of antifolate resistance. Cancer Metastasis Rev. 2007;26:153–181. doi: 10.1007/s10555-007-9049-z. [DOI] [PubMed] [Google Scholar]
  • 12.Zhao R, Goldman ID. Resistance to antifolates. Oncogene. 2003;22:7431–7457. doi: 10.1038/sj.onc.1206946. [DOI] [PubMed] [Google Scholar]
  • 13.Chattopadhyay S, Moran RG, Goldman ID. Pemetrexed: biochemical and cellular pharmacology, mechanisms, and clinical applications. Mol Cancer Ther. 2007;6:404–417. doi: 10.1158/1535-7163.MCT-06-0343. [DOI] [PubMed] [Google Scholar]
  • 14.O’Shannessy DJ, Somers EB, Albone E, et al. Characterization of the human folate receptor alpha via novel antibody-based probes. Oncotarget. 2011;2:1227–1243. doi: 10.18632/oncotarget.412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Leamon CP, Low PS. Folate-mediated targeting: from diagnostics to drug and gene delivery. Drug Discov Today. 2001;6:44–51. doi: 10.1016/s1359-6446(00)01594-4. [DOI] [PubMed] [Google Scholar]
  • 16.Antony AC. Folate receptors. Annu Rev Nutr. 1996;16:501–521. doi: 10.1146/annurev.nu.16.070196.002441. [DOI] [PubMed] [Google Scholar]
  • 17.Elnakat H, Ratnam M. Distribution, functionality and gene regulation of folate receptor isoforms: implications in targeted therapy. Adv Drug Deliv Rev. 2004;56:1067–1084. doi: 10.1016/j.addr.2004.01.001. [DOI] [PubMed] [Google Scholar]
  • 18.Franklin WA, Waintrub M, Edwards D, et al. New anti-lung-cancer antibody cluster 12 reacts with human folate receptors present on adenocarcinoma. Int J Cancer. 1994;57:89–95. doi: 10.1002/ijc.2910570719. [DOI] [PubMed] [Google Scholar]
  • 19.Parker N, Turk MJ, Westrick E, et al. Folate receptor expression in carcinomas and normal tissues determined by a quantitative radioligand binding assay. Anal Biochem. 2005;338:284–293. doi: 10.1016/j.ab.2004.12.026. [DOI] [PubMed] [Google Scholar]
  • 20.Weitman SD, Weinberg AG, Coney LR, et al. Cellular localization of the folate receptor: potential role in drug toxicity and folate homeostasis. Cancer Res. 1992:6708–6711. 52, 23. [PubMed] [Google Scholar]
  • 21.Gruner BA, Weitman SD. The folate receptor as a potential therapeutic anticancer target. Invest New Drugs. 1999;16:205–219. doi: 10.1023/a:1006147932159. [DOI] [PubMed] [Google Scholar]
  • 22.Smith AE, Pinkney M, Piggitt NH, et al. A novel monoclonal antibody for detection of folate receptor alpha in paraffin-embedded tissues. Hybridoma (Larchmt) 2007;26:281–288. doi: 10.1089/hyb.2007.0512. [DOI] [PubMed] [Google Scholar]
  • 23.Nunez MI, Behrens C, Woods DM, et al. High expression of folate receptor alpha in lung cancer correlates with adenocarcinoma histology and mutation. J Thorac Oncol. 2012;7:833–840. doi: 10.1097/JTO.0b013e31824de09c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pribble B, Edelman MJ. EC145 : a novel targeted agent for adenocarcinoma of the lung. Expert Opin Investig Drugs. 2012;21:755–761. doi: 10.1517/13543784.2012.671294. [DOI] [PubMed] [Google Scholar]
  • 25.Ebel W, Routhier EL, Foley B, et al. Preclinical evaluation of MORAb-003, a humanized monoclonal antibody antagonizing folate receptor-alpha. Cancer Immunity. 2007;7:1–8. [PMC free article] [PubMed] [Google Scholar]
  • 26.Chattopadhyay S, Wang Y, Zhao R, et al. Lack of impact of the loss of constitutive folate receptor alpha expression, achieved by RNA Interference, on the activity of the new generation antifolate pemetrexed in HeLa cells. Clin Cancer Res. 2004;10:7986–7993. doi: 10.1158/1078-0432.CCR-04-1225. [DOI] [PubMed] [Google Scholar]
  • 27.Kamen BA, Smith AK. Farletuzumab, an anti-folate receptor a antibody, does not block binding of folate or anti-folates to receptor nor does it alter the potency of anti-folates in vitro. Cancer Chemother Pharmacol. 2012;70:113–120. doi: 10.1007/s00280-012-1890-2. [DOI] [PubMed] [Google Scholar]
  • 28.O’Shannessy DJ, Yu G, Smale R, et al. Folate receptor alpha expression in lung cancer: diagnostic and prognostic significance. Oncotarget. 2012 doi: 10.18632/oncotarget.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Iwakiri S, Sonobe M, Nagai S, et al. Expression status of folate receptor alpha is significantly correlated with prognosis in non-small-cell lung cancers. Ann Surg Oncol. 2008;15:889–899. doi: 10.1245/s10434-007-9755-3. [DOI] [PubMed] [Google Scholar]
  • 30.Detterbeck FC, Boffa DJ, Tanoue LT. The new lung cancer staging system. Chest. 2009;136:260–271. doi: 10.1378/chest.08-0978. [DOI] [PubMed] [Google Scholar]
  • 31.Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026. [DOI] [PubMed] [Google Scholar]
  • 32.Wynes MW, Konopa K, Singh S, et al. Thymidylate Synthase Protein Expression by IHC and Gene Copy Number by SISH Correlate and Show Great Variability in Non-Small Cell Lung Cancer. J Thorac Oncol. 2012;7:982–992. doi: 10.1097/JTO.0b013e31824fe95a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Christoph DC, Reyna-Asuncion B, Mascaux C, et al. Folylpoly-glutamate synthetase expression is associated with tumor response and outcome from pemetrexed-based chemotherapy in malignant pleural mesothelioma. J Thorac Oncol. 2012;7:1440–1448. doi: 10.1097/JTO.0b013e318260deaa. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Monica V, Scagliotti GV, Ceppi P, et al. Differential Thymidylate Synthase Expression in Different Variants of Large-Cell Carcinoma of the Lung. Clin Cancer Res. 2009;15:7547–7552. doi: 10.1158/1078-0432.CCR-09-1641. [DOI] [PubMed] [Google Scholar]
  • 35.Spearman C. The proof and measurement of association between two things. Int J Epidemiol. 2010;39:1137–1150. doi: 10.1093/ije/dyq191. [DOI] [PubMed] [Google Scholar]
  • 36.Kruskal WH, Wallis WA. Use of ranks in one-criterion variance analysis. Am Stat. 1952;47:583–621. [Google Scholar]
  • 37.Fawcett T. ROC graphs: notes and practical considerations for researchers. ReCALL. 2004;31:1–38. [Google Scholar]
  • 38.Ruopp MD, Perkins NJ, Whitcomb BW, et al. Youden Index and optimal cut-point estimated from observations affected by a lower limit of detection. Biom J. 2008;50:419–430. doi: 10.1002/bimj.200710415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Halpern J. Maximally selected chi square statistics for small samples. Biometrics. 1982;38:1017–1023. [Google Scholar]
  • 40.Miller R, Siegmund D. Maximally selected chi square statistics. Biometrics. 1982;38:1011–1016. [Google Scholar]
  • 41.Ceppi P, Volante M, Saviozzi S, et al. Squamous cell carcinoma of the lung compared with other histotypes shows higher messenger RNA and protein levels for thymidylate synthase. Cancer. 2006;107:1589–1596. doi: 10.1002/cncr.22208. [DOI] [PubMed] [Google Scholar]
  • 42.Gandara DR, Mack PC, Omori A, et al. Thymidylate synthase (TS) RNA expression in non-small cell lung cancer (NSCLC): implications for personalizing pemetrexed therapy. J Thorac Oncol. 2009;4(Suppl 1):D7.1. [Google Scholar]
  • 43.Zheng Z, Li X, Schell MJ, et al. Thymidylate synthase in situ protein expression and survival in stage I nonsmall-cell lung cancer. Cancer. 2008;112:2765–2773. doi: 10.1002/cncr.23491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Nutt JE, Razak ARA, O’Toole K, et al. The role of folate receptor alpha (FRalpha) in the response of malignant pleural mesothelioma to pemetrexed-containing chemotherapy. Br J Cancer. 2010;102:553–560. doi: 10.1038/sj.bjc.6605501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dosio F, Milla P, Cattel L. EC-145, a folate-targeted Vinca alkaloid conjugate for the potential treatment of folate receptor-expressing cancers. Curr Opin Investig Drugs. 2010;11:1424–1433. [PubMed] [Google Scholar]
  • 46.Ghamande S, Gilbert L, Penson R, et al. The co-development of a folate receptor molecular diagnostic imaging agent (99mTc-EC20) and folate receptor targeted drug conjugate (EC145) in the treatment of ovarian cancer patients. Eur J Cancer. 2011;47:OP93. [Google Scholar]
  • 47.Bonnon C, Wendeler MW, Paccaud J-P, et al. Selective export of human GPI-anchored proteins from the endoplasmic reticulum. J Cell Sci. 2010;123:1705–1715. doi: 10.1242/jcs.062950. [DOI] [PubMed] [Google Scholar]
  • 48.Rijnboutt S, Jansen G, Posthuma G, et al. Endocytosis of GPI-linked membrane folate receptor-alpha. J Cell Biol. 1996;132:35–47. doi: 10.1083/jcb.132.1.35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Shia J, Klimstra DS, Nitzkorski JR, et al. Immunohistochemical expression of folate receptor alpha in colorectal carcinoma: patterns and biological significance. Hum Pathol. 2008;9:498–505. doi: 10.1016/j.humpath.2007.09.013. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

2

Suppl. Table 1: Distribution of N-stages for the different histological subtypes of adenocarcinoma, large cell carcinoma, squamous cell carcinoma, exclusive of other histological subtypes. In general, the majority of the patients had higher N-stages with no significant differences between the histological subtypes being observed.

Suppl. Table 2: Association between protein expression of total TS and objective response (OR) or clinical disease control (DC). (A) Responding and non-responding patients of the entire cohort were compared and significant differences in the mean or median TS H-score for OR were found. (B) Comparison of patients who received first-line treatment revealed a trend for associations between the mean or median TS H-scores and OR or DC. (C) Patients with adenocarcinomas were compared and significant differences in the mean or median TS H-score for OR were observed.

3. Suppl. Figure 1.

Correlation between intracellular and membranous protein expression of folate receptor alpha (FRA) in different histological subtypes: (A) in adenocarcinomas (ADC), (B) in large cell carcinomas (LCC), (C) in squamous cell carcinomas (SCC), and (D) in all other histological subtypes (OTH).

4. Suppl. Figure 2.

Analysis of a subgroup of patients with first-line pemetrexed-based treatment. Analyses of total thymidylate synthase (TS) protein expression and (A) progression-free survival (PFS) or (B) overall survival (OS); analyses of intracellular protein expression of folate receptor alpha (FRA) and (C) PFS or (D) OS; analyses of membranous FRA protein expression and (E) PFS or (F) OS.

5. Suppl. Figure 3.

Analysis of a subgroup of patients with adenocarcinomas of the lung. Analyses of total thymidylate synthase (TS) protein expression and (A) progression-free survival (PFS) or (B) overall survival (OS); analyses of intracellular protein expression of folate receptor alpha (FRA) and (C) PFS or (D) OS; analyses of membranous FRA protein expression and (E) PFS or (F) OS.

RESOURCES