Home Folate receptor: a potential target in ovarian cancer
Article Open Access

Folate receptor: a potential target in ovarian cancer

  • Marie Bartouskova , Bohuslav Melichar and Beatrice Mohelnikova-Duchonova EMAIL logo
Published/Copyright: February 6, 2015
Become an author with De Gruyter Brill

Abstract

Ovarian cancer is the most frequent cause of gynecological cancer-related death. Unfortunately, many patients are diagnosed at an advanced stage and have a poor prognosis. The standard treatment for advanced disease involves maximal cytoreductive surgery and chemotherapy based on platinum compounds and taxanes. Patients presenting at an advanced stage have a higher risk of recurrence. The development of drug resistance currently represents a major obstacle in the systematic treatment and, therefore, the discovery of new anticancer agents and approaches should improve the poor prognosis of these patients. Folate receptor α is overexpressed in epithelial ovarian cancer (EOC), but has limited expression in nonmalignant human tissues. The degree of folate receptor expression corresponds with the stage and grade of the disease. Because of this, folate receptor α seems to be a potential therapeutic target for the treatment of ovarian cancer. Currently, several approaches have been studied to target this protein in ovarian cancer treatment. This review summarizes current knowledge about the potential usage of folate receptors as prognostic and predictive biomarkers as well as their role in the management and targeted therapy of ovarian cancer.

Introduction

Folic acid antagonists have been used in the treatment of cancer for more than six decades, since 1948 [1]. Folic acid is a vitamin required for cell metabolism, DNA synthesis, and repair [2]. Two cellular folate uptake pathways have been characterized. The first is mediated by transmembrane-reduced folate carriers, which represent the predominant pathway for folate uptake by normal human cells. Whereas in epithelial cancer cells, membrane proteins called folate receptors are often highly expressed and mediate the folate uptake by tumors. Epithelial ovarian carcinoma (EOC) has the highest expression of these proteins, making the folate receptor an attractive candidate for targeted therapy and targeted drug delivery therapy in several types of cancers, specifically in EOC.

EOC had an age-standardised rates (ASRs) 13.1 per 100,000 inhabitants (2012) [3], with the highest mortality rate of all gynecologic tumors. Prognostic factors in EOC are stage, grade, age, histological subtype, performance status, volume of ascites, extent of residual disease following debulking surgery, and findings at second-look laparotomy [4]. Parameters of the host response, e.g., the presence of tumor-infiltrating lymphocytes [5] or the production of neopterin, a biomarker of immune system activation [6], have also been identified as independent prognostic factors. More research into the prognostic factors is clearly needed as prognostic factors can help to identify patients with poor prognosis and streamline the development of new approaches and drugs.

Currently, the standard treatment for advanced ovarian carcinoma is surgical tumor debulking followed by platinum and paclitaxel-based combination chemotherapy. However, despite the potentially high response rate to the initial chemotherapy ranging approximately between 60% and 80%, the majority of patients will ultimately experience recurrence [7], and only about 44% survive 5 years after the diagnosis. The development of drug resistance, either de novo or induced resistance, significantly limits the long-term efficacy of systemic chemotherapy [8]. For platinum therapy resistant recurrent EOC topotecan, liposomal doxorubicin, etoposide, gemcitabine, vinorelbine, cyclophosphamide, and/or other drugs may be used [9]. Therapeutic approaches that target the host response to neoplasia have so far been limited to the experimental setting [10, 11].

Despite the fact that anti-folic acid chemotherapy has been used for more than six decades, a new area for the exploration of anti-folate therapy has been opened with the advent of targeted therapy and with the development of new targeted approaches in medical oncology. Moreover, folate receptors may be not only the target for anticancer therapy, but also a potential predictive and prognostic biomarker and a target for tumor imaging.

This review summarizes published data concerning the folate receptors, their potential usage as prognostic and predictive biomarker as well as their role in the detection and targeted therapy of EOC, and seeks to define future research directions.

Folate receptor – structure, expression, and function

Folate receptors (FRs) are glycosylphosphatidylinositol-anchored membrane proteins of 38–40 kDa that bind folic acid or folate-drug conjugates with high affinity [12]. FRs are encoded by a multigene family FOLR (namely, FOLR1, FOLR2, and FOLR3), which is localized to chromosome 11q13.3–q14.1 [13]. FRs are membrane proteins that play an important role mainly in the cellular accumulation of folates and antifolates. Folate (pteroyl-l-glutamic acid, vitamin B9) is an essential vitamin that is important for DNA replication and cell division. Three types of folate receptors (FRα, FRβ, and FRγ) have been distinguished [14]. Each of folate receptors has tissue specific distribution and different folate-binding potential [15]. FRα binds folic acid with high affinity and transports folate by receptor-mediated endocytosis [14].

FR has been discovered as a tumor marker of EOC cell line in 1991 [16], and FR expression has been reported in 90% of EOC cases. Subsequently, different levels of FR expression were observed in various human tumors and normal tissues. In contrast to cancer cells, most of the nonmalignant tissues express low to negligible levels of FR, with the exception of the kidney and the lung. FR is limited to the apical membrane of the proximal tubule cells of human kidney and lung pneumocytes [17, 18]. However, in lung tissue, FR seems not to be able to capture folates from the circulation as folate-drug conjugates fail to accumulate in the lungs of patients [19]. Besides EOC, a number of other cancers have the high levels of FR, namely, renal cell carcinomas, lung cancer, and endometrial carcinoma [20]. Moderate expression has been found in the brain, breast, bladder, and pancreatic cancers [20]. In the above-mentioned quantitative radioligand-binding study, more than 50% of pancreatic cancer samples were positive for FR expression, but the study using immunohistochemistry demonstrated only 13% of pancreatic cancer samples as positively staining [21]. This discrepancy could be explained by the antibody specificity, thin slicing of samples, where antigen retrieval is compromised, or by the expression of isoforms other than α-isoforms, as in the immunohistochemical study, anti-FRα antibody has been used. Low FR expression has been reported in the liver, colorectal, prostate, lymphoma, and head and neck carcinomas [20]. Despite the fact that among all other human tissue types ovarian malignancies have the highest FR expression, the level of expression is highly dependent on tumor histology. Almost 100% of nonmucinous primary and metastatic ovarian tumors were found to have high FR expression, whereas mucinous tumors express very low or nondetectable FR levels [20]. Moreover, it seems that the metastatic tumors generally express higher levels of FR compared to the primary tumors [20, 21]. FR expression seems to provide a growth advantage to the tumor by greater folate uptake resulting in rapid cellular growth and division [14]. Consequently, FR protein expression has been found to be associated with tumor progression, dedifferentiation (grade), decreased survival, and platinum resistance [22–24]. Toffoli et al. formulated a hypothesis that FRα might increase the repair of DNA damage caused by platinum via increased folate cellular uptake [23]. Patients with suboptimal debulking surgery had significantly higher tissue FRα expression levels in contrast with patients with optimal surgery. Moreover, a higher expression of FRα was an independent prognostic factor associated with inferior disease-free survival and overall survival of patients [25].

There is evidence that FR might be involved in malignant transformation, tumor progression, and treatment outcomes. In addition to the modulation of treatment outcomes of standard platinum-based chemotherapy, FR is currently studied as a promising potential target for the targeted therapy. Moreover, because FR has been confirmed to be a tumor-associated antigen in ovarian cancer, it may be used for selective targeted delivery of drugs to tumor cells.

Folate receptor as a target for anticancer therapy

The FR represents a potential target for targeted therapy in some human cancers [26]. As the above-mentioned studies have shown a significant association of FR expression in ovarian cancer cells with biological aggressiveness and tumor phenotype, it is clear that FR plays an important role in tumor development and progression. The role of FRα in carcinogenesis may be to regulate folate uptake from serum [27] or generate regulatory signals for tumor growth [28], and therefore, FRα overexpression phenotype is associated with ovarian cancer cell proliferation [29]. The high expression of FRα in ovarian cancer, together with the effect on tumor biology, predisposes FRα for a potential target for the ovarian cancer treatment. Moreover, FRα could suppress the cytotoxic drug-induced apoptosis through the caspase pathway by regulating the apoptosis-related molecule Bcl-2 and Bax [25], and therefore, the combination of FRα target therapy and chemotherapy has been currently intensively studied.

Farletuzumab

Farletuzumab (MORAb-003) is a 145-kDa humanized monoclonal antibody against FRα produced in Chinese hamster ovary cells [30]. The therapeutic potential of farletuzumab in EOC has been shown both in in vitro and in vivo studies (Table 1).

Table 1

Overview of trials with farletuzumab.

Preclinical data
In vitroFarletuzumab inhibited FRα-dependent cell growth and mediated tumor cytotoxicity through complement-dependent cytotoxicity and antibody-dependent cytotoxicity [31].
In vivoPrecursor of farletuzumab reduced tumor growth in nude mice SKOV-3 ovarian tumor xenografts [31].
Farletuzumab exhibited low toxicity in non-human primates [31].
Farletuzumab in combination with docetaxel demonstrated synergistic activity in murine xenograft model [29].
Clinical data
 Phase IPrimary endpointNumber of patientsPrevious chemotherapy regimensMTDAdverse events grade 3/4Results (RECIST)Reference
 Platinum resistant EOCSafety, MTD255 (median)400 mg/m20SD (9)

PD (15)
[32]
 Phase IIPrimary endpointArm A FarletuzumabArm B combination therapy Farletuzumab+chemotherapyDose, mg/m2Adverse eventsResults (RECIST)Reference
 First platinum-sensitive relapseNormalized CA125, overall response rate (ORR)2847100Febrile neutropenia, thrombocytopenia, abdominal pain, diarrhea, large intestinal obstruction, subileus, urosepsis, and hyperglycemiaCR (3)

PR (30)

SD (9)

PD (2)
[33]
 Phase IIIPrimary endpointNumber of patientsArm AArm BArm CResultsReference
 Platinum-resistant EOCPFS, OS417Farletuzumab 2.5 mg/kg+ paclitaxelPlacebo 2.5 mg/kg+ paclitaxelNoPrematurely terminated
 Platinum sensitive EOCPFS1100Farletuzumab 1.25 mg/kg+ CBDCA+taxaneFarletuzumab 2.5 mg/kg+ CBDCA+taxaneArm C placebo+CBDCA+taxaneFailed to meet the primary endpoint[34]

MTD, maximum tolerated dose; EOC, epithelial ovarian carcinoma; PFS, progression-free survival; OS, overall survival; CR, complete response; PR, partial response; SD, stable disease; PD, progression disease; CBDCA, carboplatin; NA, not available.

Preclinical data

Preclinical in vitro studies have shown that farletuzumab inhibits FRα-dependent cell growth [31]. Moreover, farletuzumab has been shown to mediate immune antitumor activity via the antibody-dependent fixation of complement and cell lysis induction (complement-dependent cytotoxicity), as well as via antibody opsonization of tumor cells followed by the recruitment of immune killer cells (antibody-dependent cell-mediated cytotoxicity). In addition, farletuzumab has reduced tumor growth through inhibition of FRα-mediated lyn kinase phosphorylation [31].

In vivo, nude mice SKOV-3 ovarian tumor xenografts were treated with murine LK 26 antibody, which is the precursor of farletuzumab. A significant reduction of tumor growth was demonstrated in 4 weeks [31]. In accordance with these results, farletuzumab in combination with docetaxel has showed synergistic activity in murine xenografts bearing FRα-expressing ovarian tumors [29]. In relation to side effects, farletuzumab exhibited a low toxicity in nonhuman primates because of the absence of FR expression in normal tissues [31]. These encouraging preclinical data led to initiation of clinical trials with farletuzumab.

Clinical data

In a phase I study, a total of 25 pretreated patients with platinum-resistant epithelial ovarian cancer received farletuzumab administered intravenously on days 1, 8, 15, and 22 in a 5-week cycle. The maximal tolerable dose of farletuzumab of 400 mg/m2 has been established. There was no evidence of serious or severe drug-related adverse events of grade 3/4, and there were no anaphylactic reactions. The treatment-related grade 1/2 adverse events were hypersensitivity reactions (15 patients, 60%), fatigue (12 patients, 48%), and diarrhea (four patients, 16%). The most common hypersensitivity reactions, including pyrexia (eight patients, 32%) and chills (five patients, 20%), were easily controlled with antipyretics and/or antihistamines. Rare samples exhibited human anti-human antibodies (HAHA). Farletuzumab was not associated with clinically significant changes in cardiac, pulmonary, or renal function. There were no objective responses. Nine patients demonstrated a stable disease, and 15 patients had a progression of disease based on RECIST criteria [32]. In conclusion, farletuzumab in the phase I study has been proven to be safe and well tolerated.

Another open-label phase I clinical trial that enrolled a total of 15 platinum-sensitive ovarian cancer patients in first or second relapse received weekly 2.5 mg/kg farletuzumab intravenously in combination with six cycles of carboplatin (AUC5-6)/pegylated liposomal doxorubicin (30 mg/m2). We still await for the full report from this trial, avaliable from: http://www.clinicaltrial.gov/ct2/show/study/NCT01004380?term=farletuzumab&rank=1.

The phase II study was performed in platinum-sensitive ovarian cancer patients in first relapse in order to determine the efficacy of farletuzumab as a single agent or in combination with platinum and taxane. Patients with nonsymptomatic relapse (elevated CA125 only) received weekly farletuzumab in monotherapy until progression, whereas patients with symptomatic relapse of the disease or those who progressed on the single agent farletuzumab had a combination of six cycles of chemotherapy consisting of carboplatin (AUC 5-6) and taxane (paclitaxel 175 mg/m2 or docetaxel 75 mg/m2) with farletuzumab (37.5 mg/m2, 67.5 mg/m2, or 100 mg/m2). The primary endpoint was normalization of CA125.

In the arm with the single agent farletuzumab, 28 patients were enrolled and 25 patients completed at least 9 weeks of single agent treatment. The concentration of CA125 decreased in two patients and was stable in 18 patients. Twenty-one patients were treated with the single agent farletuzumab; 21 patients crossed over after progression to combination therapy. Thus, a total of 47 patients were treated with combination therapy. CA125 normalized in 38 of these patients after six cycles of combined therapy, and treatment continued only with farletuzumab in monotherapy until progression. According to the RECIST criteria, complete response was observed in 7% (3), partial response in 68% (30), stable disease in 21% (9), and 5% (2) of patients had progression. Farletuzumab alone as well as in combination with chemotherapy was well tolerated. The most common adverse events were febrile neutropenia, thrombocytopenia, abdominal pain, diarrhea, large intestinal obstruction, subileus, urosepsis, and hyperglycemia [33]. Phase II indicated that the combination of farletuzumab with carboplatin and taxane might enhance the response rate and duration of response in platinum-sensitive EOC patients in first relapse and served as a basis for subsequent trials.

Based on promising results of early phase I and phase II clinical trials, randomized, double-blind, placebo-controlled phase III studies have been conducted. The final results have so far not been published, and only limited preliminary data have been reported. The phase III (NCT00849667) trial aimed to evaluate the efficacy and safety of the weekly farletuzumab in combination with carboplatin and taxane in patients with platinum-sensitive ovarian cancer in first relapse. Patients were randomized to one of three arms, including placebo plus carboplatin/taxane, farletuzumab at 1.25 mg/kg intravenously plus carboplatin/taxane, or farletuzumab at 2.5 mg/kg intravenously plus carboplatin/taxane. The primary endpoints were progression-free survival; the secondary endpoints were safety and tolerability of weekly doses 1.25 mg/kg or 2.5 mg/kg of farletuzumab in combination with chemotherapy (carboplatin/taxane), overall survival, CA125-defined progression-free survival, duration of second compared to first remission, and patient quality of life. The median progression-free survival was 9.0 months in the placebo group, 9.5 months in the 1.25-mg/kg farletuzumab arm, and 9.7 months in the 2.5-mg/kg farletuzumab arm. With no statistically significant differences between the arms, the study did not meet its primary endpoints [34]. A similar phase III study that randomized platinum-resistant or refractory-relapsed ovarian cancer patients into two arms (paclitaxel with farletuzumab 2.5 mg/kg and paclitaxel with placebo) also failed in the primary outcome focused on progression-free survival and overall survival. The study has been prematurely terminated as it did not meet the pre-specified criteria for continuation after interim futility analysis, available from: http://www.clinicaltrial.gov/ct2/show/NCT00738699?term=NCT00738699&rank=1.

Unfortunately, the phase III randomized trials in both platinum-sensitive and platinum-resistant EOC failed to confirm the promising results of the invitro and in vivo studies. Nevertheless, the phase II trials demonstrated the safety of farletuzumab. The question remains whether the design of the randomized trials took into account the proper selection of patients that could profit from anti-FR therapy. It would be interesting to divide patients to subgroups according to FR expression status. From phase II trials, it seems to be possible that the subgroup of patients who may have benefit from anti-FR therapy exists, but the biomarkers that would predict treatment response are currently unknown.

Folate receptor and targeted drug delivery in anticancer therapy

Targeted delivery of anticancer therapy exclusively to cancer cells is certainly an attractive treatment approach in medical oncology. This concept brings several advantages, mainly improvements in efficacy of anticancer therapy and reduction of drug toxicity as high toxicity and insufficient activity are currently the biggest limitations of anticancer therapy [35]. The frequent overexpression of folate receptors in cancer cells may represent a target for tumor-selective drug delivery [36]. In general, two strategies have been shown to be effective for targeted delivery of drugs to FR-positive cancer cells [36]. First is the coupling of an anticancer drug to monoclonal antibody against the receptor. However, in this case, the targeting ligand is highly immunogenic with a range of side effects and with the inability for repeated administrations in the case of development of secondary antibodies. The second approach is coupling of anticancer drugs to folic acid, a high affinity FR ligand. Currently, there are five folic acid-based small-molecule drug conjugates (SMDCs) [26].

Folate-desacetylvinblastine monohydrazide (EC140; DAVLBH)

EC140 was produced by coupling a peptidic analog of the folic acid to DAVLBH via an acylhydrazone bond [37].

Preclinical data

EC140 is a water-soluble conjugate with a high affinity for FR-positive cells. EC140 has been shown to have a specific and dose-responsive activity in vitro [37]. Subsequently, the activity has been described in vivo, in both syngeneic and xenograft models with minimal to moderate toxicity [37]. However, immediately after that, it was demonstrated that EC145 (vintafolide) was more active and less toxic than EC140. Vintafolide was shown to induce more durable complete responses in tumor-bearing animals [38]. Furthermore, EC145 was shown to be FR specific as it was not active against a FR-negative tumor model [39]. Therefore, subsequent research aiming to evaluate clinical use of folate-vinca alkaloids has focused mainly on vintafolide (EC145).

Vintafolide (EC145)

Vintafolide (EC145) is a folate-vinca alkaloid (folate-desacetylvinblastine monohydrazide) conjugate with a microtubule-destabilizing effect targeting FR-expressing cells [40] and a disulfide bond-containing analog EC140 [38]. The therapeutic potential of vintafolide in EOC has been shown both in in vitro and in vivo studies (Table 2).

Table 2

Overview of trials with vintafolide.

Preclinical data
In vitroVintafolide displayed strong synergistic activity against nasopharyngeal KB cells when combined with doxorubicin [41].
In vivoVintafolide plus chemotherapy produced far greater antitumor effect compared to the single agents alone [41].
Vintafolide treatment led to CR in 5 out 5 mice xenografts with tolerable toxicity [42].
Clinical data
 Phase IPrimary endpointNumber of patientsPrevious chemotherapy regimensMTDAdverse events 3/4Results (RECIST) EOC onlyReference
 Advanced solid tumorsSafety, MTD3282.5 mgConstipation, fatiguePR (1)

SD (1)
[42]
 Phase IIPrimary endpointArm AArm BDose, mgAdverse eventsResultsReference
PLD alonePLD+vintafolide
 Precedent study

 Platinum-resistant EOC
PFS491002.5Abdominal pain, leukopenia, neutropenia, peripheral sensory neuropathyPFS 5.0 months (B) vs. 2.7 months (A)[41]
 Phase IIIPrimary endpointNumber of patientsArm AArm BArm CResultsReference
 Proceed studyPFS640Vintafolide 2.5 mg+PLDPlacebo+PLDNoPrematurely terminatedUnpublished data
 Platinum-resistant EOC

MTD, maximum tolerated dose; EOC, epithelial ovarian carcinoma; PFS, progression-free survival; CR, complete response; PR, partial response; SD, stable disease; PD, progression disease; PLD, pegylated liposomal doxorubicin.

Preclinical data

In vitro, vintafolide displayed a strong synergistic activity against nasopharyngeal KB cell line when combined with doxorubicin. In vivo, all vintafolide drug combinations, namely, pegylated liposomal doxorubicin, cisplatin, carboplatin, paclitaxel, docetaxel, topotecan, and irinotecan produced far greater antitumor effect compared to single agents alone, without significantly increasing overall toxicity [43]. Moreover, these results were not observed with combinations of EC140 or vindesine [43].

In vivo, nude mice with nasopharyngeal carcinoma KB cells or J6456 murine lymphoma cell xenografts were treated by vintafolide. The treatment of FR-positive nude mice bearing human xenografts led to complete response in five out of five mice, and four out of five mice have been cured (i.e., remission without a relapse for >90 days post-tumor implantation). Vintafolide treatment was not accompanied by noticeable weight loss or major organ tissue degeneration. Furthermore, the enhanced therapeutic index due to folate conjugation was also indicated by the fact that the unconjugated drug (desacetylvinblastine monohydrazide) was found to be completely inactive when administered at nontoxic dose levels and only marginally active when given at highly toxic dose levels [40]. Taken together, vintafolide had significant antiproliferative activity, tolerable toxicity, and seemed to be a potent, folate-targeted anticancer agent for phase I clinical trials.

Clinical data

Phase I study has been conducted in cohorts of three to six patients with a wide range of refractory solid tumors to determine the maximum-tolerated dose of vintafolide administered as a bolus intravenous injection or 1-h infusion on days 1, 3, 5 and days 15, 17, and 19 of each 28-day cycle with dose escalation. The maximal tolerated dose was 2.5 mg regardless of the type of administration. The dose-limiting toxicity was constipation. The other reported adverse events were nausea, fatigue, and vomiting, mostly grade 2 toxicities [42]. Vintafolide has been proven to have an acceptable safety profile in patients with advanced cancer.

The phase II PRECEDENT trial compared vintafolide (2.5 mg intravenously three times per week during weeks 1 and 3) combined with pegylated liposomal doxorubicin (PLD, 50 mg/m2 intravenously once every 28 days) with PLD monotherapy in a total of 149 pretreated patients with platinum-resistant EOC. The primary aim was to compare progression-free survival. Median progression-free survival of 5.0 months for vintafolide plus PLD was significantly longer compared to median progression-free survival of only 2.7 months for PLD monotherapy. The best efficacy was observed in patients with lesions 100% positive for FR with median progression-free survival of 5.5 months compared with 1.5 months for PLD alone (p=0.013). FR negative patients had no benefit from the addition of vintafolide [41]. Overall survival was evaluated as a secondary endpoint. There were no statistically significant differences between the two arms in overall survival. In general, the drug combination was well tolerated, although higher frequency of neutropenia, leukopenia, abdominal pain, and peripheral neuropathy was observed in the vintafolide plus PLD arm. On the other hand, this arm had a higher total median cumulative PLD dose per patient and a higher frequency of safety evaluations [41]. Thus, vintafolide plus PLD has been the first combination demonstrating an improvement over standard therapy, with marginally increased toxicity, in a randomized trial in patients with platinum-resistant EOC.

A phase III study (PROCEED) started in 2010 as a double-blind trial evaluating PLD in combination with placebo or PLD with vintafolide. The trial planned to enroll approximately 500 FR-positive patients, and all participants were scheduled to undergo imaging with the FR-targeting investigational diagnostic agent EC20 during the screening period. However, the study has been suspended, and the results have so far not been published. Nevertheless, vintafolide is also tested in other types of human cancer. A multicenter trial of vintafolide in advanced FR-positive adenocarcinoma of the lung brought also inconclusive results [44]. The patients with FR-positive lesions had a trend of better overall survival (47.2 weeks vs. 14.9 weeks) compared to patients with at least one, but not all, FR-positive target lesions, although the statistical significance was not reached (p=0.101) [44]. To investigate the clinical benefit, a randomized phase II trial evaluating the activity of vintafolide plus docetaxel vs. docetaxel alone in highly FR positive lung tumors has been initiated. It is evident from the data that vintafolide has been shown to be a promising candidate for the treatment of ovarian cancer, and the ongoing trials will address the clinical use of this drug.

Folate-maytansinoid conjugate (EC131)

Folate-maytansinoid conjugate (EC131) is a new folate receptor targeted drug-conjugate. Maytansinoid is a strong microtubule-inhibiting agent. Folate-maytansinoid conjugate was prepared by covalently attaching the folic acid to maytansinoid DM1 with use as an intramolecular disulfide bond [45].

Preclinical data

In vitro, EC131 has been highly cytotoxic toward FR-positive nasopharyngeal carcinoma KB cells.

In vivo, the antitumor activity of EC131 was explored against BALB/c mice bearing subcutaneously injected FR-positive lung adenocarcinoma cells (M109), which express similar numbers of folate receptors as human ovarian carcinoma [20]. Tumors in the untreated animals attained a size of approximately 1500 mm3 by day 42, whereas tumors in the EC131-treated group of animals attained the same volume by day 45. The treatment led to complete response in one out of five mice and partial response in three out of five mice. The treated group and the untreated group differed significantly on day 18 post-tumor implantation. EC131 treatment was not observed to lead to weight loss or organ tissue degeneration. The antitumor activity of EC131 was reduced by application of folic acid leading to no response to EC131 treatment in a group of mice with a diet containing folic acid. Thus, the antitumor activity was proven to be dependent on the binding of EC131 to tumor-associated FR [45]. Furthermore, EC131 also showed marked antitumor activity in FR-positive human KB tumor xenografts. The EC131 treatment led to complete response in 4/5 of the mice and partial response in 1/5 of the mice. No signs of drug toxicity were detected during and after the therapy. Taken together, these results indicate that EC131 has a significant activity in xenograft models and seemed to be a potent folate-targeted anticancer agent [45] waiting for further validation studies and clinical trials (Table 3).

Table 3

FR-targeted delivery drugs in in vivo and in vitro studies.

Drug conjugated to folateIn vitroIn vivo on xenograftsaClinical trialsReferences
EC72Mitomycin CSpecific and dose-responsive activity on FR-positive cellsMinimal toxicity

Doubled OS
No[46, 47]
EC131MyatansinoidSpecific activity on FR-positive cellsCR in 4/5 miceNo[45]
PR in 1/5 mice
Minimal toxicity
EC140Desacetylvinblastine monohydrazideSpecific and dose-responsive activity on FR-positive cellsCR in 1/5 mice

PR in 4/5 mice

Minimal to moderate toxicity
No[37, 38]
EC145DesacetylvinblastineSpecific and dose-responsive activity on FR-positive cells.CR in 5/5 mice

Minimal toxicity
Phase I–III[38–44]
More active and less toxic than EC140.

aNude mice bearing FR-positive human KB cells. CR, complete response; PR, partial response; OS, overall survival.

Folate-mitomycin C conjugate (EC72)

EC72 is a folate drug conjugate, which was prepared by covalently attaching folate to the potent DNA crosslinker mitomycin C, a cytotoxic drug with a high antitumor and antibiotic activity [46].

Preclinical data

In vitro, EC72 exhibits dose-dependent activity against a panel of FR-positive cell lines (KB, ID8, and M109 cell lines). Similarly to EC131, the activity of EC72 is also blocked by the presence of excess free folic acid and does not affect normal tissues with low levels of FR [46].

In vivo therapy experiments were performed in both syngeneic and xenograft models. In a syngeneic model, BALB/c mice bearing subcutaneously injected FR-positive lung adenocarcinoma cells (M109) have been treated by intraperitoneal injection of EC72 or mitomycin C starting 4 days after implantation of M109 tumor cells. The EC72-treated animals had a significant 185% increase in median overall survival time in comparison with the untreated control group (p=0.006). No noticeable toxicity was found in the context of weight loss or physical change in behavior. The EC72 treatment led to the complete response in one out of four mice and partial response in three of four mice. Mitomycin C-treated animals did not survive significantly longer than the controls, presumably due to mitomycin C-related toxicity [47]. The antitumor activity of EC72 has been shown also in xenograft models using xenografts with human KB carcinoma cells. The survival of EC72-treated mice was doubled compared to untreated controls. There was no evidence of toxicity [47]. In conclusion, both tumor models proved the antitumor activity of EC72 in a dose-dependent manner and the lack of serious toxicity. EC72 seems to be a promising candidate for further validation studies and future clinical trials (Table 3).

Imaging methods

In light of the development of FR-targeted anticancer therapy, the need for quantification of functional FRs has occurred in clinical cancer samples. Several methodical approaches have been reported to analyze the FR-positive cells, namely, polymerase chain reaction detecting mRNA expression levels [48], immunohistochemistry, and radioligand binding assay detecting protein expression levels [20–25]. Quantitative radioligand binding assay has an advantage of sensitively measuring only functional receptors which is very important for FR-targeted therapies. On the other hand, immunohistochemistry can avoid specificity concerns regarding nonmalignant cells as activated macrophages that may contribute to radioligand signaling. Thus, both methods are currently used to established FR expression status in trials.

As several folic acid-based radiopharmaceuticals targeting FR have been developed, some of these agents have been tested in imaging FR via single-photon emission computed tomography (SPECT) or via positron emission tomography (PET) [49].

With regard to PET imaging, the first 18F-labeled folate has been synthetized for application in 2006 by amide coupling of the prosthetic group 4-[18F]fluorobenzylamine and native folic acid [50]. Despite the good visualization of FR-positive tumors, the preparation of the radiopharmaceutical was complicated and time consuming and provided a low region-selective product. Therefore, another radiofolate was developed using the copper-catalyzed azide-alkyne cycloaddition (CuAAC; click reaction) resulting in the first 18F-click-labeled folate in high radiochemical yields, but in vivo animal PET imaging revealed an unfavorable biological distribution profile with a poor signal-to-noise ratio caused by the polarity of the radiofolate [51]. Fischer et al. used the efficacy of CuAAC for 18F-radiolabeling in combination with the inevitable polarity of [18F]FDG to enhance pharmacodynamics and significantly enhanced tumor-to-background ratio [52]. Another approach is 18F-fluorination, which led to the development of 2′-[18F]fluorofolic acid with promising results in vivo, as a clear-cut visualization of FR-positive KB tumors, and healthy tissues have been shown [53, 54]. Recently, several new 18F-folate was developed, being available in very high radiochemical yields via a fast and convenient two-step radiosynthesis and showed good in vivo behavior waiting for testing in clinical trials [49, 55].

Radionuclides showing promising results in preclinical in vivo SPECT are 111In-diethylenetriamine pentaacetic acid (DTPA)-folates, 99mTc-folates, and 67Ga-folates [56–58]. The first radionuclide tested in clinical trials was 111In-DTPA-folate [59]. However, 99mTc-based imaging agents showed more rapid pharmacokinetics and lower cost [19]. In vivo, 99mTc-etarfolatide evaluation of FRs correlated to the immunohistochemistry staining results in 61% of 154 patients [60]. The authors suggest that the discrepancy may reflect a different FR expression in the primary tumor and metastatic lesions. The administration of 99mTc-etarfolatide was safe as none of the serious adverse events were related to the administration of etarfolatide. 99mTc-etarfolatide imaging has been tested in two clinical phase II trials with vintafolide to identify the presence of FR in recurrent/refractory ovarian or endometrial tumors and the correlation of FR expression with response to FR-targeted therapy [9, 61]. In both studies, etarfolatide has been able to identify patients likely to benefit from vintafolide. In addition to histological verification of FRs by immunohistochemistry, SPECT with radiofolates seems to be another approach to verify FR-positive tumors that, in addition, brings the possibility to evaluate the treatment response.

Conclusion

FRα has been shown to be a biomarker of ovarian cancer as it is overexpressed in EOC, but has limited expression in nonmalignant human tissues. The degree of FR expressions corresponds with the stage and grade of the disease. Because of this, FRα seems to be an ideal target for EOC treatment. Moreover, the FR expression levels correlated with the response to platinum-based chemotherapy, and it seems that FR may increase the repair of DNA damage caused by platinum. Together, it is hypothesized that the addition of anti-FR therapy to conventional chemotherapy may be synergistic rather than additive.

Currently, two principal approaches have been studied to use this protein in ovarian cancer treatment. The first is the development of antibodies against FR (farletuzumab) and the second is the use of FR for targeted delivery of anticancer drugs selectively to cancer cells (vintafolide/EC145, EC140, EC131, and EC72). Despite the fact that all these drugs demonstrated antitumor activity in in vitro and in in vivo models as well as a lack of severe adverse events, none of them showed so far a benefit for EOC patients in phase III clinical trials. Based on promising results of phase I and phase II, farletuzumab and vintafolide have been tested in phase III trials recently. However, both of these trials failed, and the questions regarding the efficacy of this approach remain. It is not clear how to design randomized trials to ensure a proper selection of patients that could benefit from anti-FR therapy. Several approaches have been tested, including immunohistochemistry, radioligand-labeled folates in PET and SPECT. However, all these methods are semiquantitative approaches and have limitations with regard to the specificity and sensitivity. From the phase II trial, it seems to be possible that the subgroup(s) of patients who may have benefit from anti-FR therapy exists, but these subgroups still remain to be identified. Moreover, FR has been used for specific drug delivery of anticancer agents as vinca-alkaloids and mitomycin C. However, in clinical practice, the most effective drugs for EOC are still paclitaxel and platinum compounds.

More research is needed to answer the question regarding FR isoform selectivity and the role of FR isoforms in ovarian cancer development and progression. The next point is the role of the patient’s diet as there is a lack of data concerning the interference of anti-FR drugs with the circulating folates. Last, but not the least, is the impact of other potential critical points downstream in FR-signaling pathway that may influence the final effect on the cancer cells. An example is the role of RAS mutations in anti-EGFR therapy in colorectal cancer treatment strategy, where the expression of EGFR is not determinative for the effect of the treatment, but the downstream mutations in the pathway are the most important for the final benefit of the patients in a clinical practice.

In conclusion, the recent shift of farletuzumab and vintafolide into phase III trials had suggested that FR targeting therapy is reaching a final point of the research, and the strategy of identifying high FR expression subgroups of cancer patients would be used in a clinical practice to improve the prognosis of EOC patients. Against the expectations, the phase III trials did not confirm the superiority of anti-FR therapy to chemotherapy alone and even opened up more questions.


Corresponding author: Beatrice Mohelnikova-Duchonova, Department of Oncology, Palacky University Medical School and Teaching Hospital, Olomouc, IP Pavlova 6, 775 25 Olomouc, Czech Republic, E-mail:

Acknowledgments

Supported by the Internal Grant Agency of the Czech Ministry of Health (grant no. NT14056-3) and research projects LO1304 and RVO61989592.

  1. Conflict of interest statement: The authors have no conflicts of interest to disclose.

References

1. Farber S, Diamond LK. Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid. N Engl J Med 1948;238:787–93.10.1056/NEJM194806032382301Search in Google Scholar

2. Choi SW, Mason JB. Folate and carcinogenesis: an integrated scheme. J Nutr 2000;130:129–32.10.1093/jn/130.2.129Search in Google Scholar

3. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 2013;49:1374–403.10.1016/j.ejca.2012.12.027Search in Google Scholar

4. Berman ML. Future directions in the surgical management of ovarian cancer. Gynecol Oncol 2003;90(2 Pt 2):S33–9.10.1016/S0090-8258(03)00342-1Search in Google Scholar

5. Tomsova M, Melichar B, Sedlakova I, Steiner I. Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma. Gynecol Oncol 2008;108:415–20.10.1016/j.ygyno.2007.10.016Search in Google Scholar PubMed

6. Melichar B, Freedman RS, Solichova D. Neopterin as an indicator of immune activation and prognosis in patients with gynecological malignancies. Int J Gynecol Cancer 2006;16:240–52.10.1111/j.1525-1438.2006.00294.xSearch in Google Scholar PubMed

7. Markman M. Pharmaceutical management of ovarian cancer: current status. Drugs 2008;68:771–89.10.2165/00003495-200868060-00004Search in Google Scholar PubMed

8. Mohelnikova-Duchonova B, Melichar B. Human equilibrative nucleoside transporter 1 (hENT1): do we really have a new predictive biomarker of chemotherapy outcome in pancreatic cancer patients? Pancreatology 2013;13:558–63.10.1016/j.pan.2013.09.005Search in Google Scholar PubMed

9. Luvero D, Milani A, Ledermann JA. Treatment options in recurrent ovarian cancer: latest evidence and clinical potential. Ther Adv Med Oncol 2014;6:229–39.10.1177/1758834014544121Search in Google Scholar PubMed PubMed Central

10. Melichar B, Freedman RS. Immunology of the peritoneal cavity: relevance for host-tumor relation. Int J Gynecol Cancer 2002;12:3–17.10.1046/j.1525-1438.2002.01093.xSearch in Google Scholar PubMed

11. Freedman RS, Vadhan-Raj S, Butts C, Savary C, Melichar B, Verschraegen C, et al. Pilot study of Flt3 ligand comparing intraperitoneal with subcutaneous routes on hematologic and immunologic responses in patients with peritoneal carcinomatosis and mesoteliomas. Clin Can Res 2003;9:5228–37.Search in Google Scholar

12. Leamon CP, Low PS. Folate-mediated targeting: from diagnostics to drug and gene delivery. Drug Discov Today 2001;6:44–51.10.1016/S1359-6446(00)01594-4Search in Google Scholar

13. Saikawa Y, Price K, Hance KW, Chen TY, Elwood PC. Structural and functional analysis of the human KB cell folate receptor gene P4 promoter: cooperation of three clustered Sp1-binding sites with initiator region for basal promoter activity. Biochemistry 1995;34:9951–61.10.1021/bi00031a018Search in Google Scholar

14. Kelemen LE. The role of folate receptor alpha in cancer development, progression and treatment: cause, consequence or innocent bystander? Int J Cancer 2006;119:243–50.10.1002/ijc.21712Search in Google Scholar

15. Antony C. Folate receptors. Annu Rev Nutr 1996;16:501–21.10.1146/annurev.nu.16.070196.002441Search in Google Scholar

16. Coney LR, Tomassetti A, Carayannopoulos L, Frasca V, Kamen BA, Colnaghi MI, et al. Cloning of a tumor-associated antigen: MOv18 and MOv19 antibodies recognize a folate-binding protein. Cancer Res 1991;51:6125–32.Search in Google Scholar

17. Holm J, Hansen SI, Høier-Madsen M, Bostad L. A high-affinity folate binding protein in proximal tubule cells of human kidney. Kidney Int. 1992;41:50–5.10.1038/ki.1992.7Search in Google Scholar

18. Franklin WA, Waintrub M, Edwards D, Christensen K, Prendegrast P, Woods J, et al. New anti-lung-cancer antibody cluster 12 reacts with human folate receptors present on adenocarcinoma. Int J Cancer Suppl 1994;8:89–95.10.1002/ijc.2910570719Search in Google Scholar

19. Leamon CP, Parker MA, Vlahov IR, Xu LC, Reddy JA, Vetzel M, et al. Synthesis and biological evaluation of EC20: a new folate-derived, 99mTc-based radiopharmaceutical. Bioconjug Chem 2002;13:1200–10.10.1021/bc0200430Search in Google Scholar

20. Parker N, Turk MJ, Westrick E, Lewis JD, Low PS, Leamon CP. Folate receptor expression in carcinomas and normal tissues determined by a quantitative radioligand binding assay. Anal Biochem 2005;338:284–93.10.1016/j.ab.2004.12.026Search in Google Scholar

21. Garin-Chesa P, Campbell I, Saigo PE, Lewis JL Jr, Old LJ, Rettig WJ. Trophoblast and ovarian cancer antigen LK26. Sensitivity and specificity in immunopathology and molecular identification as a folate-binding protein. Am J Pathol 1993;142:557–67.Search in Google Scholar

22. Toffoli G, Cernigoi C, Russo A, Gallo A, Bagnoli M, Boiocchi M. Overexpression of folate binding protein in ovarian cancers. Int J Cancer 1997;74:193–8.10.1002/(SICI)1097-0215(19970422)74:2<193::AID-IJC10>3.0.CO;2-FSearch in Google Scholar

23. Toffoli G, Russo A, Gallo A, Cernigoi C, Miotti S, Sorio R, et al. Expression of folate binding protein as a prognostic factor for response to platinum-containing chemotherapy and survival in human ovarian cancer. Int J Cancer 1998;79:121–6.10.1002/(SICI)1097-0215(19980417)79:2<121::AID-IJC4>3.0.CO;2-VSearch in Google Scholar

24. Kalli KR, Oberg AL, Keeney GL, Christianson TJ, Low PS, Knutson KL, et al. Folate receptor alpha as a tumor target in epithelial ovarian cancer. Gynecol Oncol 2008;108:619–26.10.1016/j.ygyno.2007.11.020Search in Google Scholar

25. Chen YL, Chang MC, Huang CY, Chiang YC, Lin HW, Chen CA, et al. Serous ovarian carcinoma patients with high alpha-folate receptor had reducing survival and cytotoxic chemo-response. Mol Oncol 2012;6:360–9.10.1016/j.molonc.2011.11.010Search in Google Scholar

26. Vlahov IR, Leamon CP. Engineering folate-drug conjugates to target cancer: from chemistry to clinic. Bioconjug Chem 2012;23:1357–69.10.1021/bc2005522Search in Google Scholar

27. Kane MA, Elwood PC, Portillo RM, Antony AC, Najfeld V, Finley A, et al. Influence on immunoreactive folate-binding proteins of extracellular folate concentration in cultured human cells. J Clin Invest 1988;81:1398–406.10.1172/JCI113469Search in Google Scholar

28. Anthony M, McLeay AC. A unique case of derangement of vitamin B12 metabolism. Proc Aust Assoc Neurol 1976;13:61–5.Search in Google Scholar

29. Spannuth WA, Sood AK, Coleman RL. Farletuzumab in epithelial ovarian carcinoma. Expert Opin Biol Ther 2010;10:431–43.10.1517/14712591003592069Search in Google Scholar

30. Jelovac D, Armstrong DK. Role of farletuzumab in epithelial ovarian carcinoma. Curr Pharm Des 2012;18:3812–5.10.2174/138161212802002698Search in Google Scholar

31. Ebel W, Routhier EL, Foley B, Jacob S, McDonough JM, Patel RK, et al. Preclinical evaluation of MORAb-003, a humanized monoclonal antibody antagonizing folate receptor-alpha. Cancer Immun 2007;7:6.Search in Google Scholar

32. Konner JA, Bell-McGuinn KM, Sabbatini P, Hensley ML, Tew WP, Pandit-Taskar N, et al. Farletuzumab, a humanized monoclonal antibody against folate receptor alpha, in epithelial ovarian cancer: a phase I study. Clin Cancer Res 2010;16:5288–95.10.1158/1078-0432.CCR-10-0700Search in Google Scholar

33. Armstrong DK, White AJ, Weil SC, Phillips M, Coleman RL. Farletuzumab (a monoclonal antibody against folate receptor alpha) in relapsed platinum-sensitive ovarian cancer. Gynecol Oncol 2013;129:452–8.10.1016/j.ygyno.2013.03.002Search in Google Scholar

34. Vergote I, Armstrong D, Scambia G, Fujiwara K, Gorbunova V, Schweizer C, et al. Phase 3 double-blind, placebo-controlled study of weekly farletuzumab with carboplatin/taxane in subjects with platinum-sensitive ovarian cancer in first relapse. Int J Gynecol Cancer 2013;23:11.Search in Google Scholar

35. Mohelnikova-Duchonova B, Melichar B, Soucek P. FOLFOX/FOLFIRI pharmacogenetics: the call for a personalized approach in colorectal cancer therapy. World J Gastroenterol 2014;20:10316–30.10.3748/wjg.v20.i30.10316Search in Google Scholar

36. Sudimack J, Lee RJ. Targeted drug delivery via the folate receptor. Adv Drug Deliv Rev 2000;41:147–62.10.1016/S0169-409X(99)00062-9Search in Google Scholar

37. Leamon CP, Reddy JA, Vlahov IR, Kleindl PJ, Vetzel M, Westrick E. Synthesis and biological evaluation of EC140: a novel folate-targeted vinca alkaloid conjugate. Bioconjug Chem 2006;17:1226–32.10.1021/bc060145gSearch in Google Scholar PubMed

38. Leamon CP, Reddy JA, Vlahov IR, Westrick E, Parker N, Nicoson JS, et al. Comparative preclinical activity of the folate-targeted Vinca alkaloid conjugates EC140 and EC145. Int J Cancer 2007;121:1585–92.10.1002/ijc.22853Search in Google Scholar PubMed

39. Leamon CP, Reddy JA, Vlahov IR, Westrick E, Dawson A, Dorton R, et al. Preclinical antitumor activity of a novel folate-targeted dual drug conjugate. Mol Pharm 2007;4:659–67.10.1021/mp070049cSearch in Google Scholar PubMed

40. Reddy JA, Dorton R, Westrick E, Dawson A, Smith T, Xu LC, et al. Preclinical evaluation of EC145, a folate-vinca alkaloid conjugate. Cancer Res 2007;67:4434–42.10.1158/0008-5472.CAN-07-0033Search in Google Scholar PubMed

41. Naumann RW, Coleman RL, Burger RA, Sausville EA, Kutarska E, Ghamande SA, et al. PRECEDENT: a randomized phase II trial comparing vintafolide (EC145) and pegylated liposomal doxorubicin (PLD) in combination versus PLD alone in patients with platinum-resistant ovarian cancer. J Clin Oncol 2013;31:4400–6.10.1200/JCO.2013.49.7685Search in Google Scholar PubMed

42. Lorusso PM, Edelman MJ, Bever SL, Forman KM, Pilat M, Quinn MF, et al. Phase I study of folate conjugate EC145 (Vintafolide) in patients with refractory solid tumors. J Clin Oncol 2012;30:4011–6.10.1200/JCO.2011.41.4946Search in Google Scholar PubMed PubMed Central

43. Reddy JA, Dorton R, Bloomfield A, Nelson M, Vetzel M, Guan J, et al. Rational combination therapy of vintafolide (EC145) with commonly used chemotherapeutic drugs. Clin Cancer Res 2014;20:2104–14.10.1158/1078-0432.CCR-13-2423Search in Google Scholar PubMed

44. Edelman MJ, Harb WA, Pal SE, Boccia RV, Kraut MJ, BonomiP, et al. Multicenter trial of EC145 in advanced, folate-receptor positive adenocarcinoma of the lung. J Thorac Oncol 2012;7:1618–21.10.1097/JTO.0b013e318267d051Search in Google Scholar PubMed

45. Reddy JA, Westrick E, Santhapuram HK, Howard SJ, Miller ML, Vetzel M, et al. Folate receptor-specific antitumor activity of EC131, a folate-maytansinoid conjugate. Cancer Res 2007;67:6376–82.10.1158/0008-5472.CAN-06-3894Search in Google Scholar PubMed

46. Reddy JA, Westrick E, Vlahov I, Howard SJ, Santhapuram HK, Leamon CP. Folate receptor specific anti-tumor activity of folate-mitomycin conjugates. Cancer Chemother Pharmacol 2006;58:229–36.10.1007/s00280-005-0151-zSearch in Google Scholar PubMed

47. Leamon CP, Reddy JA, Vlahov IR, Vetzel M, Parker N, Nicoson JS, et al. Synthesis and biological evaluation of EC72: a new folate-targeted chemotherapeutic. Bioconjug Chem 2005;16:803–11.10.1021/bc049709bSearch in Google Scholar PubMed

48. Campbell IG, Jones TA, Foulkes WD, Trowsdale J. Folate-binding protein is a marker for ovarian cancer. Cancer Res 1991;51:5329–38.Search in Google Scholar

49. Schieferstein H, Ross TL. (18) F-labeled folic acid derivatives for imaging of the folate receptor via positron emission tomography. J Labelled Comp Radiopharm 2013;56:432–40.10.1002/jlcr.3104Search in Google Scholar PubMed

50. Bettio A, Honer M, Müller C, Brühlmeier M, Müller U, Schibli R, et al. Synthesis and preclinical evaluation of a folic acid derivative labeled with 18F for PET imaging of folate receptor-positive tumors. J Nucl Med 2006;3:1153–60.Search in Google Scholar

51. Ross TL, Honer M, Lam PY, Mindt TL, Groehn V, Schibli R, et al. Fluorine-18 click radiosynthesis and preclinical evaluation of a new 18F-labeled folic acid derivative. Bioconjug Chem 2008;3:2462–70.10.1021/bc800356rSearch in Google Scholar PubMed

52. Schieferstein H, Betzel T, Fischer CR, Ross TL. 18F-click labeling and preclinical evaluation of a new 18F-folate for PET imaging. EJNMMI Res 2013;3:68.10.1186/2191-219X-3-68Search in Google Scholar PubMed PubMed Central

53. Ross TL, Honer M, Müller C, Groehn V, Schibli R, Ametamey SM. A new 18F-labeled folic acid derivative with improved properties for the PET imaging of folate receptor-positive tumors. J Nucl Med 2010;3:1756–62.10.2967/jnumed.110.079756Search in Google Scholar PubMed

54. Betzel T, Müller C, Groehn V, Müller A, Reber J, Fischer CR, et al. Radiosynthesis and preclinical evaluation of 3′-Aza-2′-[18F]fluorofolic acid: a novel PET radiotracer for folate receptor targeting. Bioconjug Chem 2013;3:205–14.10.1021/bc300483aSearch in Google Scholar PubMed

55. Gent YY, Weijers K, Molthoff CF, Windhorst AD, Huisman MC, Smith DE, et al. Evaluation of the novel folate receptor ligand [18F]fluoro-PEG-folate for macrophage targeting in a rat model of arthritis. Arthritis Res Ther 2013;3:R37.10.1186/ar4191Search in Google Scholar PubMed PubMed Central

56. Wang S, Lee RJ, Mathias CJ, Green MA, Low PS. Synthesis, purification, and tumor cell uptake of67Ga-deferoxamine-folate, a potential radiopharmaceutical for tumor imaging. Bioconjug Chem 1996;3:56–62.10.1021/bc9500709Search in Google Scholar PubMed

57. Mindt TL, Müller C, Melis M, De-Jong M, Schibli R. “Click-to-chelate”: in vitro and in vivo comparison of a 99mTc(CO)3-labeled N(tau)-histidine folate derivative with its isostructural, clicked 1,2,3-triazole analogue. Bioconjug Chem 2008;3:1689–95.10.1021/bc800183rSearch in Google Scholar PubMed

58. Müller C, Vlahov IR, Santhapuram HK, Leamon CP, Schibli R. Tumor targeting using 67Ga-DOTA-Bz-folate-investigations of methods to improve the tissue distribution of radiofolates. Nucl Med Biol 2011;3:715–23.10.1016/j.nucmedbio.2010.12.013Search in Google Scholar PubMed

59. Siegel BA, Dehdashti F, Mutch DG, Podoloff DA, Wendt R, Sutton GP, et al. Evaluation of 111In-DTPA-folate as a receptor-targeted diagnostic agent for ovarian cancer: initial clinical results. J Nucl Med 2003;44:700–7.Search in Google Scholar

60. Fisher RE, Siegel BA, Edell SL, Oyesiku NM, Morgenstern DE, Messmann RA, et al. Exploratory study of 99mTc-EC20 imaging for identifying patients with folate receptor-positive solid tumors. J Nucl Med 2008;49:899–90.10.2967/jnumed.107.049478Search in Google Scholar PubMed

61. Morris RT, Joyrich RN, Naumann RW, Shah NP, Maurer AH, Strauss HW, et al. Phase II study of treatment of advanced ovarian cancer with folate-receptor-targeted therapeutic (vintafolide) and companion SPECT-based imaging agent (99mTc-etarfolatide). Ann Oncol 2014; 25:852–8.10.1093/annonc/mdu024Search in Google Scholar PubMed

Received: 2014-10-28
Accepted: 2014-11-25
Published Online: 2015-2-6
Published in Print: 2015-3-1

©2015 by De Gruyter

This article is distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Downloaded on 3.10.2025 from https://www.degruyterbrill.com/document/doi/10.1515/pterid-2014-0013/html
Scroll to top button