755
Views
13
CrossRef citations to date
0
Altmetric
Review

Cancer chemotherapy: targeting folic acid synthesis

&
Pages 293-301 | Published online: 30 Sep 2022

Abstract

Antifolates are structural analogs of folates, essential one-carbon donors in the synthesis of DNA in mammalian cells. Antifolates are inhibitors of key enzymes in folate metabolism, namely dihydrofolate reductase, β-glycinamide ribonucleotide transformylase, 5′-amino-4′-imidazolecarboxamide ribonucleotide transformylase, and thymidylate synthetase. Methotrexate is one of the earliest anticancer drugs and is extensively used in lymphoma, acute lymphoblastic leukemia, and osteosarcoma, among others. Pemetrexed has been approved in combination with cisplatin as first-line treatment for advanced non-squamous-cell lung cancer, as a single agent for relapsed non-small-cell lung cancer after platinum-containing chemotherapy, and in combination with cisplatin for the treatment of pleural mesothelioma. Raltitrexed is approved in many countries (except in the United States) for advanced colorectal cancer, but its utilization is mainly limited to patients intolerant to 5-fluorouracil. Pralatrexate has recently been approved in the United States for relapsed or refractory peripheral T-cell lymphoma. This article gives an overview of the cellular mechanism, pharmacology, and clinical use of classical and newer antifolates and discusses some of the main resistance mechanisms to antifolate drugs.

Introduction

Folates are essential, one-carbon donors in the synthesis of purines, pyrimidines, serine, and methionine, all critical to de novo synthesis of DNA in mammalian cells, as they lack the capacity to synthesize folates and require these anionic hydrophilic molecules to be transported into the cells via sophisticated transport systems (reduced folate carrier, RFC). After folate was discovered to be vital to many cellular processes, the antifolates aminopterin and methotrexate (MTX) were synthesized in the early 1940s.Citation1 In 1948, aminopterin was the first drug to induce temporary remissions in childhood leukemia.Citation1,Citation2 Only 10 years later, MTX was part of a therapy regimen that was first shown to cure some selected solid tumors, namely choriocarcinoma.Citation3 MTX is still used in the treatment of a variety of tumors, including acute lymphocytic leukemia,Citation4 breast cancer,Citation5 osteosarcoma,Citation6 primary central nervous system lymphoma,Citation7 and head and neck cancer.Citation8 Above all, it is also used in certain autoimmune diseases, such as rheumatoid arthritis or psoriasis. Recently, the newer antifolate pemetrexed or multitargeted antifolate (MTA) has been established in the first-line treatment of mesotheliomaCitation9 and non-squamous, non-small-cell lung cancer (NSCLC).Citation10 An important task for the future is treatment individualization, eg, by identifying genetic variations in drug pathway-associated genes with an important impact on clinical outcome in patients receiving antifolatesCitation11Citation13 or the use of therapeutic drug monitoring (TDM), eg, with MTX, enabling adequate drug exposure in all patients.Citation14,Citation15

Cellular folate metabolism

Folates (pteroylglutamates) belong to the family of B9 vitamins that are essential to mammalian cells. They form a family of cofactors based on the structure of folic acid (2-NH2-4-OH-pteroylglutamic acid). Folic acid undergoes intracellular reduction first to dihydrofolate and then to tetrahydrofolate (THF). Both reduction steps are mediated by dihydrofolate reductase (DHFR). The major dietary form of folates is 5′-methyl-THF (5′-MTHF). Together with homocysteine, MTHF is converted to methionine and THF, a vitamin-B12-dependent step that is mediated by methionine synthase. THF is the main substrate for folylpolyglutamate synthetase (FPGS), which progressively adds glutamates at the γ-carboxyl residues. The resulting folate polyglutamates are polyanionic molecules that can no longer be transported through the lipophilic cell membrane. These folate polyglutamates are the biologically active form of folates, as they serve as one-carbon donors in de novo synthesis of purines, thymidylate, and polyamines. Furthermore, folates are required for the synthesis of S-adenosyl methionine, which promotes methylation of DNA, histones, lipids, and neurotransmitters.Citation16

Antifolate drug metabolism

As structural analogs of folates, antifolates use the same transport systems to enter the cells, namely the reduced folate receptor (RFC), folate receptors (FR), and the recently discovered proton-coupled folate transporter (PCFT) or soluble carrier 46A1 (SLC46A1).Citation16 The RFC plays a dominant role in cellular uptake of antifolates, as it has low affinity to its main endogenous ligand MTHF. Its affinity to antifolates varies from low for MTX to high for talotrexin (PT-523).Citation17 The RFC works as an anion exchanger that utilizes the gradient built up by an asymmetrical distribution of organic phosphates across cell membranes.Citation18,Citation19 The RFC is expressed both in tumor cells and normal tissue,Citation20 limiting the tolerability of antifolates. On the contrary, the folate receptors FR-α and FR-β are overexpressed at the surface of some tumor cells, making these tumors vulnerable to antifolate drugs,Citation21,Citation22 The FR family consists of two cell-surface receptors (FR-α and FR-β) and a constitutively secreted isoform (FR-γ).Citation23,Citation24 FR-α is expressed in a number of normal epithelial cells as well as in a number of carcinomas, with the exception of carcinomas of the head and neck.Citation21 FR-β serves as a myeloid differentiation marker and is overexpressed in a variety of nonepithelial malignancies,Citation21,Citation25 whereas the expression of FR-γ is restricted to hematopoietic tissues.Citation24,Citation26 In contrast to the high-capacity and low-affinity RFC, transport by FR-α and FR-β is by low-capacity and high-affinity endocytosis.Citation27 After antifolate transport to the endosomal compartment, transport to the intracellular compartment involves the PCFT.Citation28,Citation29 Accordingly, mutations in the gene encoding for PCFT have been shown to cause rare hereditary folate malabsorption.Citation29 In addition to its role in folate endocytosis, PCFT also serves as a high-affinity folate-proton symporter that is important for the intestinal absorption in the proximal small intestine.Citation30 Besides these specific folate transporters, a number of other transport systems have been described to be involved in the efflux of antifolates, including the multidrug resistance-associated proteins MRP1–5 and the breast cancer resistance protein (BCRP or ABCG2).Citation31,Citation32

Intracellularly, the classical antifolates undergo polyglutamation by FPGS, resulting in effective intracellular retention and increased affinity of the antifolates to their target enzymes.Citation33Citation36 The nonclassical lipophilic antifolates such as talotrexin or trimetrexate (TMQ) are not substrates of FPGS and do not require activation by polyglutamation for anticancer activity.Citation37

Cellular targets: TYMS, DHFR, GARFT, and AICARFT

Antifolates are inhibitors of key enzymes in folate metabolism, namely DHFR, β-glycinamide ribonucleotide transformylase (GARFT), 5′-amino-4′-imidazolecarboxamide ribonucleotide transformylase (AICARFT), and thymidylate synthetase (TYMS). GARFT and AIRCARFT are two key enzymes of the de novo purine biosynthesis. In a first step, GARFT enables the formation of the purine imidazole ring of purines. The substrate for this reaction is 10′-formyl-THF, which is synthesized from THF and formate, a step mediated by 10′-formyl-THF synthetase. In a second step, AICARFT generates inosine monophosphate, which serves as the precursor for purine nucleotides adenylate (AMP) and guanylate (GMP). DHFR catalyzes the reduction of DHF to 5′,6′,7′,8′-THF, which is converted to 5′,10′-methyltetrahydrofolate (5′,10′-MTHF), the substrate for TYMS.Citation38 TYMS catalyzes the initial step in DNA synthesis, in which deoxythymidine monophosphate (dTMP), a precursor of DNA synthesis, is generated from deoxyuridine monophosphate (dUMP), resulting in the oxidation of MTHF to DHF. This metabolic step is essential for de novo synthesis of thymidine nucleotides for DNA synthesis. DHFR was the first enzyme to be identified as a cellular target for the antifolates aminopterin and MTX.Citation39 The latter exhibits its anticancer effect by almost irreversible inhibition of DHFR, with subsequent disruption of purine and pyrimidine synthesis.Citation40 Newer antifolates have been designed with a higher affinity toward DHFR, eg, talotrexin, with a 15-fold increased affinity when compared with MTX.Citation37

Mechanisms of resistance

Antifolate resistance might result from impaired cellular influx or increased efflux, impaired polyglutamation, increased expression, or mutation of cellular targets, or intra-cellular accumulation of THF cofactors. Various transport-resistant phenotypes have been described in MTX-resistant cell line models, some of them resulting from mutations of the RFC gene,Citation41Citation43 and others from RFC overexpression.Citation44Citation46 A genetic polymorphism within the RFC gene (80G > A) results in replacement of arginine in position 27 with histidine,Citation47 and is associated with a worse clinical outcome in children with acute lymphoblastic leukemia (ALL) receiving MTX.Citation48 In osteosarcoma, which is known for its intrinsic resistance to conventionally dosed MTX, mutations at the 3′-UTR and promoter methylation of the RFC were described.Citation49 The role of FR is less well characterized and more controversial. Although overexpression of FR-α was found to predict resistance to platinum-based chemotherapy in ovarian cancer patients,Citation50 suppression of FR expression by gene methylation was also found as a potential mechanism of resistance.Citation51 Similarly, hypermethylation of the PCFT gene (SLC46A1) was found in a resistant HeLa cell line.Citation52 Multidrug resistance-associated proteins (MRP or ABCC) 1–4 confer the efflux of MTX and have been shown to potentially confer resistance to MTX in cell line models.Citation53,Citation54 However, MTX polyglutamates have low affinity toward the ABCC transporters, which is why this type of resistance might not be clinically relevant. However, breast cancer resistance protein (BCRP or ABCG2) also transports polyglutamates out of the cell, and mutations within the ABCG2 gene (at amino acid position 482) have been shown to confer resistance to various antifolates.Citation55,Citation56 Overexpression of P-glycoprotein (MDR1) is suggested to be important for antifolate resistance in the presence of a defective RFC or in case high doses of MTX are administered.Citation57 Impaired polyglutamation is another mechanism that is of special importance for the classical antifolates that undergo extensive polyglutamation to be active. Finally, amplification of the gene encoding for DHFR has been identified in ALL,Citation58 ovarian cancer,Citation59 and soft-tissue sarcomaCitation60 as a potential mechanism of resistance to MTX, but the clinical relevance of such amplifications is unclear at present.Citation61

Specific substances

Classical antifolates

The classical antifolates have a similar structure to MTX, utilize the RFC for entering human cells, and are subject to intracellular polyglutamation.

MTX

MTX is one of the earliest anticancer drugs and is extensively used in lymphoma, acute lymphoblastic leukemia, and osteosarcoma. The drug competitively inhibits DHFR and, to a lesser extent, GARFT, AICARFT, and TYMS. Although thymidylate depletion is the main cytotoxic driver of MTX, inhibition of GARFT and AICARFT also results in impaired purine synthesis. As a result of their inability to synthesize DNA and RNA, the malignant cells are unable to proliferate and cause further damage, resulting in cell apoptosis.

Pharmacology

7-Hydroxymethotrexate (7-OH-MTX) is the main metabolite in serum following MTX infusion,Citation62 and it contributes to activity and toxicity. The concentrations of 7-OH-MTX exceed those of the parent compound in plasma shortly after the infusion.Citation63 Both MTX and 7-OH-MTX exhibit first-order pharmacokinetics.Citation62 MTX is eliminated by renal excretion involving passive glomerular filtration and active tubular reabsorption and secretion. 7-OH-MTX is also renally cleared but more slowly than MTX. Renal elimination is prolonged in patients with renal impairment or third-space fluid collections, due to slow redistribution of MTX from these extravascular compartments.Citation62 MTX is prone to drug–drug interactions, especially nonsteroidal antirheumatics (NSARs).Citation64 The uptake of MTX into the cell is primarily mediated by the RFC and, to a lesser amount, by the FR-α. Intracellularly, MTX undergoes extensive γ-polyglutamation by FPGS, and these negatively charged polyglutamates are retained intracellularly. Polyglutamates can also undergo hydrolation by γ-glutamyl hydrolase (GGH, also known as folylpolyglutamate hydrolase or FPGH) into short-chain polyglutamates.Citation65,Citation66 The MTX pentaglutamate moiety is most active, with Ki values 100 times below Ki values of the nonglutamated compound.

High-dose MTX

MTX at doses ≥1 g/m2 is the backbone for treating diseases such as primary central nervous system lymphoma (PCNSL), osteosarcoma, or ALL. Careful patient selection, adequate hydration and urinary alkalinization, avoidance of drug interactions, drainage of third-space fluids, and TDM with appropriate adjustment of leucovorin (LV) rescue make HDMTX a well-tolerated treatment option most of the time. LV rescue starts 24 h after the start of MTX infusion at a dose of 15 mg/m2 IV push every 6 h for 3 days and should be continued until serum MTX concentrations drop below 0.05 μmol/L. Despite supporting measures, acute renal failure is seen in ≤2% of patients receiving HDMTX, as a consequence of the precipitation of MTX and 7-OH-MTX in the kidney tubules.Citation67 Because of considerable interpatient variability, TDM is essential to identify patients at high risk for severe toxicity, and the need for increased hydration or prolonged LV rescue. Before TDM with supplemental LV rescue was incorporated into HDMTX regimens, toxicity was substantial, including a 6% fatality rate;Citation68 80% of these fatalities were attributed to severe myelosuppression, which resulted in either sepsis or hemorrhage, and 20% were attributed to renal failure. Conventional treatment for HDMTX-induced renal dysfunction includes prompt escalation of LV rescue and adequate hydration and urine alkalinization, provided adequate urine output can be maintained. MTX removal by hemodialysis is of potential value in refractory cases. Finally, carboxypeptidase-G2 (CPDG2), a recombinant bacterial enzyme that hydrolyzes MTX to the inactive metabolite 2,4-diamino-N10-methylpteroic acid (DAMPA), is another option in refractory cases of MTX-associated renal dysfunction. CPDG2 lowers plasma concentrations of MTX within 15 min of administration by roughly 99%.Citation69 More recent studies suggest individual exposure to MTX to be an important predictor of a favorable treatment response in patients with PCNSL,Citation14,Citation15 but this awaits prospective validation.

Raltitrexed

Raltitrexed is a selective and direct TYMS inhibitor. As an analog of the THF cofactor, it cannot be incorporated into DNA, and cellular accumulation of dUMP does not result in resistance to raltitrexed.Citation70 Its long-lasting inhibition of TYMS allows a convenient 3-weekly schedule of administration. Raltitrexed is approved in many countries (except the United States) for advanced colorectal cancer, but its utilization is mainly limited to patients who are intolerant to 5-fluorouracil (5-FU). Although raltitrexed proved to be equally active to 5-FU/LV in advanced colorectal cancer, there were some raltitrexed-associated deaths due to combined gastrointestinal and hematologic toxicity.Citation71 Combining the phase III MCRC trials, raltitrexed-related mortality (1.6%–4%) was greater than with 5-FU (1.2%–2.8%).Citation72 This occurred in spite of a significantly lower all-cause serious toxicity rate with raltitrexed and has been attributed to administration of raltitrexed after a toxic event or in the presence of renal impairment.Citation73 Patient education, monitoring of renal function, and supportive measures are essential in the management of patients receiving raltitrexed.Citation74

Pharmacology

Raltitrexed predominantly enters the cell by RFC and then undergoes polyglutamation, with the polyglutamated form again being more potent than the parent compound. With repeated administration at 3-weekly intervals, no clinically significant accumulation of raltitrexed was found in patients with normal renal function.Citation75 Raltitrexed is contraindicated in patients with severe renal or hepatic impairment and/or clinically significant cardiac arrhythmias requiring drug therapy. The importance of dose reductions in patients with reduced creatinine clearance is critical in preventing subsequent severe toxicity. In patients after accidental overdosing or those suffering from severe toxicity, LV rescue is of potential value.

Pralatrexate

Pralatrexate (PDX; 10′-propargyl 10′-deazaaminopterin) is a newer antifolate that was rationally designed to have a high affinity for the RFC, resulting in increased cellular internalization.Citation76 In a phase II lymphoma study, PDX demonstrated some activity against peripheral T-cell lymphoma (TCL).Citation77 Subsequently, the multicenter confirmatory phase II PROPEL (Pralatrexate in Relapsed or Refractory Peripheral T-cell Lymphoma) trial has led to the approval of PDX in the United States for relapsed or refractory TCL.Citation78,Citation79 Treatment response in a total of 109 evaluable patients in the PROPEL trial was 29%, with 12 patients (11%) achieving a complete response.Citation79 The median duration of response was 10.1 months. Mucosal inflammation was seen in >70% of patients but was usually mild to moderate. Hematological toxicity consists of severe thrombocytopenia in a third of patients and severe anemia in 16% of patients. Febrile neutropenia was noted in 5% of cases. Patients should receive supplementation with B12 and folic acid to avoid severe toxicity.

PDX was rationally designed to have high affinity for the RFC, which leads to better cellular internalization of the drug and a greater antitumor effect when compared with MTX.Citation80 The structural difference between PDX and MTX is based on the presence of a propargyl group substitution at carbon 10 instead of the methyl group in MTX. The basis of increased efficacy of PDX in vitro is based on its increased affinity for the RFC, but whether this is enough to overcome MTX resistance is unknown. PDX effectively binds to and inactivates DHFR, depleting intracellular reduced folate stores and blocking DNA synthesis.

Lometrexol

Lometrexol (LMTX) is a potent and selective inhibitor of GARFT, with broad antitumor spectrum. GARFT catalyzes the formation of purines from the reaction of 10′-formyltetrahydrofolate (10′-FTHF) to THF. Its inhibition results in a depletion of intracellular purine levels. LMTX enters the cell via the RFC and undergoes extensive polyglutamation, with a slow elimination of polyglutamates. Without folic acid supplementation, severe cumulative myelosuppression and mucositis are likely.Citation81 At present, LMTX is not approved as an anticancer agent.

Edatrexate

Edatrexate (EDX) is a classic antifolate with a more efficient intracellular polyglutamation compared with MTX.Citation82 EDX polyglutamates are potent inhibitors of DHFR but less potent inhibitors of TYMS.Citation83 EDX exhibits saturable, nonlinear Michaelis–Menten pharmacokinetics, with ≤55% of EDX undergoing renal excretion as unchanged parent compound.Citation84 As EDX was associated with severe stomatitis, toxic dermatitis, and even fatalities,Citation85 clinical development was halted.

Nonpolyglutamable classical antifolates

Talotrexin

Talotrexin (PT-523) is a newer antifolate and potent antagonist of DHFR. Talotrexin combines characteristics of both the classical and nonclassical antifolates. As talotrexin does not contain a glutamic acid side chain, it is not converted to intracellular polyglutamates, with a potential advantage for drug safety and less bone marrow toxicity.Citation86 The drug binds tightly to DHFR, with an inhibitory constant (Ki) of 0.35 pmol/L, 15-fold lower than for MTX. Talotrexin exhibits linear pharmacokinetics with a rapid initial disposition phase.Citation87 Patients with relapsed or refractory leukemia or MDS received talotrexin for five subsequent days, together with supplemental folic acid and vitamin B12.Citation87 Dose-limiting toxicity was stomatitis, and talotrexin 0.6 mg/m2/day for 5 days every 3 weeks was recommended for phase II studies, where evaluation in patients with refractory ALL is ongoing.Citation87

Nonclassical antifolates

Nonclassical antifolates do not contain glutamic acid and are not polyglutamable; they are more lipophilic than the classical antifolates and enter cells by passive diffusion.

TMQ

TMQ is a nonclassical, lipophilic quinazoline derivative with a high inhibitory potency toward DHFR.Citation88 Because of its lipophilicity, TMQ can rapidly enter cells by a non-energy-dependent process. Unlike most antifolate drugs, TMQ is mainly eliminated by hepatic metabolism instead of renal excretion, with a terminal elimination half-life of 15–20 h.Citation88 Cell lines resistant to MTX because of deficiencies in folate transport generally retain their sensitivity to TMQ. As TMQ lacks the glutamic acid side chain, it cannot be polyglutamated and is not retained within the cell for prolonged periods of time.Citation89 Although TMQ has undergone broad phase II testing in solid tumors,Citation90 results were disappointing and there is no current indication in oncology.

Piritrexim

Piritrexim (PTX) is an oral lipophilic antifolate that is not a substrate of the active folate transport systems but enters cells via an energy-independent process and is effective against cancer cells resistant to MTX because of transport defects.Citation91 PTX is not polyglutamated by FPGS, but it is a potent inhibitor of DHFR and TYMS. Oral absorption of PTX is rapid, with an overall bioavailability of 75%–95%.Citation92 The terminal half-life following oral administration is 4.5–5.6 h,Citation92 with hepatic metabolism being the primary route of drug clearance. Despite the potential as an oral antifolate, PTX did not show any therapeutic advantage over more established antifolates.

Nolatrexed

Nolatrexed (Thymitaq, TM) is a nonclassical, lipophilic antifolate and a noncompetitive, high-affinity TYMS inhibitor. TM causes extensive dTMP depletion and dUMP accumulation, causing thymineless cell death. TM is not dependent on the cell cycle, as high concentrations of TM failed to induce S-phase arrest but still resulted in apoptosis.Citation93 Although TM itself is lipophilic, it can be administered via intravenous infusion as a water-soluble dihydrochloride salt. Due to its lipophilicity, TM enters cells by passive diffusion and does not undergo polyglutamation.Citation94 TM was granted orphan drug status for the treatment of unresectable hepatocellular carcinoma by the US Food and Drug Administration (2001) and the European Medicines Agency (2007), based on a randomized phase III Asian study comparing TM with doxorubicin, and two North American phase II studies.Citation95 TM has never gained widespread use in oncology.

MTA

Pemetrexed

Pemetrexed has been approved in combination with cisplatin as a first-line treatment for advanced non-squamous-cell lung cancer,Citation10 as a single agent for relapsed NSCLC after platinum-containing chemotherapy,Citation96 and in combination with cisplatin for the treatment of pleural mesothelioma.Citation9 Important is histotype-selective activity of pemetrexed, with a significant benefit seen only in patients with non-squamous-cell lung cancer,Citation10 potentially as a consequence of increased TYMS expression in tumors of squamous histology.Citation97 Supportive treatment with oral folic acid 0.5 mg/day and intramuscular vitamin B12 1 mg every 9 weeks is routinely used, as it has been shown to reduce the incidence of potentially fatal myelosuppression.Citation98 Pemetrexed is a cell-cycle-dependent antifolate with a 6–5-fused pyrrolopyrimidine-based nucleus,Citation99 and it inhibits TYMS, DHFR, GARFT, AICARFT, and C1-THF synthase, which catalyzes the incorporation of a formyl group into 10′-FTHF for purine synthesis, and the incorporation of a methylene group into 5′,10′-MTHF for thymidylate synthesis. Mechanisms of resistance include diminished accumulation of pemetrexed polyglutamates due to decreased activity of FPGS,Citation100 increased enzymatic cleavage of pemetrexed poly-γ-glutamates by high intracellular GH activity, and TYMS amplification.Citation101 Inhibition of TYMS leads to intracellular accumulation of dUMP and subsequent efflux of deoxyuridine (dUrd) into the circulation, which can be used as a pharmacodynamic marker of in vivo TYMS inhibition following administration of pemetrexed.Citation102

Pharmacology

Pemetrexed is transported into cells mainly by the RFC and undergoes rapid intracellular transformation by FPGS to the more potent polyglutamate derivatives.Citation103 Pemetrexed has a small steady-state volume of distribution of about 15 L and is rapidly eliminated from plasma with a terminal elimination half-life of between 2 and 5 h at doses of 525–700 mg/m2. Pemetrexed undergoes mainly urinary excretion as an unchanged parent compound. Furthermore, pemetrexed exhibits dose-proportional increases in plasma concentration and no signs of accumulation in patients with normal renal function. Third-space accumulation seems not to play a clinically significant role.Citation104 As pemetrexed is often combined with potentially nephrotoxic cisplatin, adequate monitoring of renal function is important. Recommendations for the management of pemetrexed toxicity in the presence of renal failure have not been established, but treatment options with LV, folate, thymidine, carboxypeptidase, or hemodialysis are all possible.Citation105 Homocysteine is a marker for overall folate status in the body and predicted severe pemetrexed-associated toxicity in a clinical study.Citation106

Summary

The approval of pemetrexed for the first-line treatment of non-squamous-cell lung cancer, second-line treatment of NSCLC, and first-line treatment of malignant pleural mesothelioma has substantially added to the clinical importance of antifolates in oncology. Treatment individualization ever since has played an important role in the development of antifolate drugs. Although LV rescue and TDM are standard for HDMTX regimens, and folate and B12 supplementation is standard for pemetrexed to increase MTD, new strategies will include pharmacogenetic markers such as tumoral TYMS expression for further improvement of antifolate treatment.

Disclosure

The authors report no conflicts of interest in this work.

References

  • FarberSCutlerECHawkinsJWHarrisonJHPeirceEC2ndLenzGGThe action of pteroylglutamic conjugates on manScience1947106276461962117831847
  • FarberSDiamondLKTemporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acidN Engl J Med19482382378779318860765
  • LiMCHertzRBergenstalDMTherapy of choriocarcinoma and related trophoblastic tumors with folic acid and purine antagonistsN Engl J Med19582592667413566422
  • KantarjianHThomasDO’BrienSLong-term follow-up results of hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone (Hyper-CVAD), a dose-intensive regimen, in adult acute lymphocytic leukemiaCancer2004101122788280115481055
  • BonadonnaGValagussaPMoliterniAZambettiMBrambillaCAdjuvant cyclophosphamide, methotrexate, and fluorouracil in node-positive breast cancer: the results of 20 years of follow-upN Engl J Med1995332149019067877646
  • FuchsNBielackSSEplerDLong-term results of the co-operative German-Austrian-Swiss osteosarcoma study group’s protocol COSS-86 of intensive multidrug chemotherapy and surgery for osteosarcoma of the limbsAnn Oncol1998988938999789613
  • FerreriAJReniMDell’OroSCombined treatment with high-dose methotrexate, vincristine and procarbazine, without intrathecal chemotherapy, followed by consolidation radiotherapy for primary central nervous system lymphoma in immunocompetent patientsOncology200160213414011244328
  • GuardiolaEPeyradeFChaigneauLResults of a randomised phase II study comparing docetaxel with methotrexate in patients with recurrent head and neck cancerEur J Cancer200440142071207615341981
  • VogelzangNJRusthovenJJSymanowskiJPhase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesotheliomaJ Clin Oncol200321142636264412860938
  • ScagliottiGVParikhPvon PawelJPhase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancerJ Clin Oncol200826213543355118506025
  • BuikhuisenWABurgersJAVincentADPemetrexed pathway-associated germline polymorphisms: a useful tool for treatment individualization?J Clin Oncol20102827e482e48320606096
  • ChenJSChaoYBangYJA phase I/II and pharmacogenomic study of pemetrexed and cisplatin in patients with unresectable, advanced gastric carcinomaAnticancer Drugs201021877778420634689
  • SmitEFBurgersSABiesmaBRandomized phase II and pharmacogenetic study of pemetrexed compared with pemetrexed plus carboplatin in pretreated patients with advanced non-small-cell lung cancerJ Clin Oncol200927122038204519307503
  • FerreriAJGuerraERegazziMArea under the curve of methotrexate and creatinine clearance are outcome-determining factors in primary CNS lymphomasBr J Cancer200490235335814735176
  • JoergerMHuitemaADKrahenbuhlSMethotrexate area under the curve is an important outcome predictor in patients with primary CNS lymphoma: a pharmacokinetic-pharmacodynamic analysis from the IELSG no. 20 trialBr J Cancer2010102467367720125159
  • ZhaoRMatherlyLHGoldmanIDMembrane transporters and folate homeostasis: intestinal absorption and transport into systemic compartments and tissuesExpert Rev Mol Med200911e419173758
  • ChenGWrightJERosowskyADihydrofolate reductase binding and cellular uptake of nonpolyglutamatable antifolates: correlates of cytotoxicity toward methotrexate-sensitive and -resistant human head and neck squamous carcinoma cellsMol Pharmacol19954847587657476904
  • GoldmanIDThe characteristics of the membrane transport of amethopterin and the naturally occurring folatesAnn N Y Acad Sci19711864004225289428
  • GoldmanIDLichtensteinNSOliverioVTCarrier-mediated transport of the folic acid analogue, methotrexate, in the L1210 leukemia cellJ Biol Chem196824319500750175303004
  • WhetstineJRFlatleyRMMatherlyLHThe human reduced folate carrier gene is ubiquitously and differentially expressed in normal human tissues: identification of seven non-coding exons and characterization of a novel promoterBiochem J2002367Pt 362964012144527
  • RossJFChaudhuriPKRatnamMDifferential regulation of folate receptor isoforms in normal and malignant tissues in vivo and in established cell lines. Physiologic and clinical implicationsCancer1994739243224437513252
  • WuMGunningWRatnamMExpression of folate receptor type alpha in relation to cell type, malignancy, and differentiation in ovary, uterus, and cervixCancer Epidemiol Biomarkers Prev19998977578210498396
  • ShenFWangHZhengXRatnamMExpression levels of functional folate receptors alpha and beta are related to the number of N-glycosylated sitesBiochem J1997327Pt 37597649581553
  • ShenFWuMRossJFMillerDRatnamMFolate receptor type gamma is primarily a secretory protein due to lack of an efficient signal for glycosylphosphatidylinositol modification: protein characterization and cell type specificityBiochemistry19953416566056657727426
  • RossJFWangHBehmFGFolate receptor type beta is a neutrophilic lineage marker and is differentially expressed in myeloid leukemiaCancer199985234835710023702
  • ShenFRossJFWangXRatnamMIdentification of a novel folate receptor, a truncated receptor, and receptor type beta in hematopoietic cells: cDNA cloning, expression, immunoreactivity, and tissue specificityBiochemistry1994335120912158110752
  • WesterhofGRSchornagelJHKathmannICarrier- and receptor-mediated transport of folate antagonists targeting folate-dependent enzymes: correlates of molecular-structure and biological activityMol Pharmacol19954834594717565626
  • AndrewsNCWhen is a heme transporter not a heme transporter? When it’s a folate transporterCell Metab2007515617189201
  • QiuAJansenMSakarisAIdentification of an intestinal folate transporter and the molecular basis for hereditary folate malabsorptionCell2006127591792817129779
  • ShayeghiMLatunde-DadaGOOakhillJSIdentification of an intestinal heme transporterCell2005122578980116143108
  • KruhGDBelinskyMGThe MRP family of drug efflux pumpsOncogene200322477537755214576857
  • WielingaPHooijbergJHGunnarsdottirSThe human multidrug resistance protein MRP5 transports folates and can mediate cellular resistance against antifolatesCancer Res200565104425443015899835
  • AllegraCJHoangKYehGCDrakeJCBaramJEvidence for direct inhibition of de novo purine synthesis in human MCF-7 breast cells as a principal mode of metabolic inhibition by methotrexateJ Biol Chem19872622813520135262443493
  • BaughCMKrumdieckCLNairMGPolygammaglutamyl metabolites of methotrexateBiochem Biophys Res Commun197352127344197190
  • CurtGAJolivetJCarneyDNDeterminants of the sensitivity of human small-cell lung cancer cell lines to methotrexateJ Clin Invest1985764132313292414316
  • FabreIFabreGGoldmanIDPolyglutamylation, an important element in methotrexate cytotoxicity and selectivity in tumor versus murine granulocytic progenitor cells in vitroCancer Res1984448319031956204743
  • RosowskyAWrightJEVaidyaCMForschRAThe effect of side-chain, para-aminobenzoyl region, and B-ring modifications on dihydrofolate reductase binding, influx via the reduced folate carrier, and cytotoxicity of the potent nonpolyglutamatable antifolate N(alpha)-(4-amino-4-deoxypteroyl)-N(delta)-hemiphthaloyl-L-ornithinePharmacol Ther200085319120510739874
  • BanerjeeDMayer-KuckukPCapiauxGBudak-AlpdoganTGorlickRBertinoJRNovel aspects of resistance to drugs targeted to dihydrofolate reductase and thymidylate synthaseBiochim Biophys Acta200215872–316417312084458
  • OsbornMJFreemanMHuennekensFMInhibition of dihydrofolic reductase by aminopterin and amethopterinProc Soc Exp Biol Med195897242943113518295
  • JohnsonJMMeieringEMWrightJEPardoJRosowskyAWagnerGNMR solution structure of the antitumor compound PT523 and NADPH in the ternary complex with human dihydrofolate reductaseBiochemistry19973615439944119109647
  • BrigleKESpinellaMJSierraEEGoldmanIDCharacterization of a mutation in the reduced folate carrier in a transport defective L1210 murine leukemia cell lineJ Biol Chem19952703922974229797559435
  • DroriSJansenGMauritzRPetersGJAssarafYGClustering of mutations in the first transmembrane domain of the human reduced folate carrier in GW1843U89-resistant leukemia cells with impaired antifolate transport and augmented folate uptakeJ Biol Chem200027540308553086310899164
  • JansenGMauritzRDroriSA structurally altered human reduced folate carrier with increased folic acid transport mediates a novel mechanism of antifolate resistanceJ Biol Chem19982734630189301989804775
  • GorlickRGokerETrippettTDefective transport is a common mechanism of acquired methotrexate resistance in acute lymphocytic leukemia and is associated with decreased reduced folate carrier expressionBlood1997893101310189028333
  • MatherlyLHTaubJWRavindranathYElevated dihydrofolate reductase and impaired methotrexate transport as elements in methotrexate resistance in childhood acute lymphoblastic leukemiaBlood19958525005097812005
  • RothemLAronheimAAssarafYGAlterations in the expression of transcription factors and the reduced folate carrier as a novel mechanism of antifolate resistance in human leukemia cellsJ Biol Chem2003278118935894112519783
  • ChangoAEmery-FillonNde CourcyGPA polymorphism (80G->A) in the reduced folate carrier gene and its associations with folate status and homocysteinemiaMol Genet Metab200070431031510993718
  • LaverdiereCChiassonSCosteaIMoghrabiAKrajinovicMPolymorphism G80A in the reduced folate carrier gene and its relationship to methotrexate plasma levels and outcome of childhood acute lymphoblastic leukemiaBlood2002100103832383412411325
  • YangRQinJHoangBHHealeyJHGorlickRPolymorphisms and methylation of the reduced folate carrier in osteosarcomaClin Orthop Relat Res200846692046205118528741
  • ToffoliGRussoAGalloAExpression of folate binding protein as a prognostic factor for response to platinum-containing chemotherapy and survival in human ovarian cancerInt J Cancer19987921211269583724
  • HsuehCTDolnickBJRegulation of folate-binding protein gene expression by DNA methylation in methotrexate-resistant KB cellsBiochem Pharmacol1994476101910277511899
  • Diop-BoveNKWuJZhaoRLockerJGoldmanIDHypermethylation of the human proton-coupled folate transporter (SLC46A1) minimal transcriptional regulatory region in an antifolate-resistant HeLa cell lineMol Cancer Ther2009882424243119671745
  • ChenZSLeeKWaltherSAnalysis of methotrexate and folate transport by multidrug resistance protein 4 (ABCC4): MRP4 is a component of the methotrexate efflux systemCancer Res200262113144315012036927
  • SirotnakFMWendelHGBornmannWGCo-administration of probenecid, an inhibitor of a cMOAT/MRP-like plasma membrane ATPase, greatly enhanced the efficacy of a new 10-deazaaminopterin against human solid tumors in vivoClin Cancer Res2000693705371210999764
  • BramEIferganIShafranABermanBJansenGAssarafYGMutant Gly482 and Thr482 ABCG2 mediate high-level resistance to lipophilic antifolatesCancer Chemother Pharmacol200658682683416612649
  • ShafranAIferganIBramEABCG2 harboring the Gly482 mutation confers high-level resistance to various hydrophilic antifolatesCancer Res200565188414842216166320
  • GiffordAJKavallarisMMadafiglioJP-glycoprotein-mediated methotrexate resistance in CCRF-CEM sublines deficient in methotrexate accumulation due to a point mutation in the reduced folate carrier geneInt J Cancer19987821761819754649
  • HornsRCJrDowerWJSchimkeRTGene amplification in a leukemic patient treated with methotrexateJ Clin Oncol198421276583327
  • TrentJMBuickRNOlsonSHornsRCJrSchimkeRTCytologic evidence for gene amplification in methotrexate-resistant cells obtained from a patient with ovarian adenocarcinomaJ Clin Oncol1984218156699660
  • LiWWLinJTTongWPTrippettTMBrennanMFBertinoJRMechanisms of natural resistance to antifolates in human soft tissue sarcomasCancer Res1992526143414381371715
  • SpencerHTSorrentinoBPPuiCHChunduruSKSleepSEBlakleyRLMutations in the gene for human dihydrofolate reductase: an unlikely cause of clinical relapse in pediatric leukemia after therapy with methotrexateLeukemia19961034394468642859
  • JolivetJCowanKHCurtGAClendeninnNJChabnerBAThe pharmacology and clinical use of methotrexateN Engl J Med198330918109411046353235
  • ErttmannRBielackSLandbeckGKinetics of 7-hydroxy-methotrexate after high-dose methotrexate therapyCancer Chemother Pharmacol19851521011043874719
  • JoergerMHuitemaADvan den BongardHJDeterminants of the elimination of methotrexate and 7-hydroxy-methotrexate following high-dose infusional therapy to cancer patientsBr J Clin Pharmacol2006621718016842380
  • MoranRGRoles of folylpoly-gamma-glutamate synthetase in therapeutics with tetrahydrofolate antimetabolites: an overviewSemin Oncol1999262 Suppl 6243210598551
  • GalivanJRyanTRheeMYaoRChaveKGlutamyl hydrolase: properties and pharmacologic impactSemin Oncol1999262 Suppl 6333710598552
  • WidemannBCAdamsonPCUnderstanding and managing methotrexate nephrotoxicityOncologist200611669470316794248
  • von HoffDDPentaJSHelmanLJSlavikMIncidence of drug-related deaths secondary to high-dose methotrexate and citrovorum factor administrationCancer Treat Rep1977614745748301783
  • GreenMRChamberlainMCRenal dysfunction during and after high-dose methotrexateCancer Chemother Pharmacol200963459960418504579
  • SalongaDDanenbergKDJohnsonMColorectal tumors responding to 5-fluorouracil have low gene expression levels of dihydropyrimidine dehydrogenase, thymidylate synthase, and thymidine phosphorylaseClin Cancer Res2000641322132710778957
  • MaughanTJamesRKerrDExcess treatment related deaths and impaired quality of life show raltitrexed is inferior to infusional 5FU regimen in the palliative chemotherapy of advanced colorectal cancer (CRC): final results of MRC CRO6 [abstract]Ann Oncol200011Suppl 443
  • CunninghamDZalcbergJMarounJEfficacy, tolerability and management of raltitrexed (Tomudex) monotherapy in patients with advanced colorectal cancer: a review of phase II/III trialsEur J Cancer200238447848611872339
  • ThomasRJWilliamsMGarcia-VargasJLessons learned from raltitrexed–quality assurance, patient education and intensive supportive drugs to optimise tolerabilityClin Oncol (R Coll Radiol)200315522723212924450
  • FordHECunninghamDSafety of raltitrexedLancet199935491921824182510577674
  • ClarkeSJBealePJRivoryLPClinical and preclinical pharmacokinetics of raltitrexedClin Pharmacokinet200039642944311192475
  • SirotnakFMDeGrawJIMoccioDMSamuelsLLGoutasLJNew folate analogs of the 10-deaza-aminopterin series. Basis for structural design and biochemical and pharmacologic propertiesCancer Chemother Pharmacol198412118256690069
  • O’ConnorOAHorwitzSHamlinPPhase II-I-II study of two different doses and schedules of pralatrexate, a high-affinity substrate for the reduced folate carrier, in patients with relapsed or refractory lymphoma reveals marked activity in T-cell malignanciesJ Clin Oncol200927264357436419652067
  • ThompsonCAFDA approves pralatrexate for treatment of rare lymphomaAm J Health Syst Pharm20096621189019850775
  • O’ConnorOProBPinter-BrownLResults of the pivotal, multicenter, phase II study of pralatrexate in patients with relapsed or refractory peripheral T-cell lymphoma (PTCL)J Clin Oncol200927Suppl 15449s Abstract 8561.
  • MatherlyLHHouZDengYHuman reduced folate carrier: translation of basic biology to cancer etiology and therapyCancer Metastasis Rev200726111112817334909
  • RayMSMuggiaFMLeichmanCGPhase I study of (6R)-5, 10-dideazatetrahydrofolate: a folate antimetabolite inhibitory to de novo purine synthesisJ Natl Cancer Inst19938514115411598320744
  • JolivetJJansenGPetersGJPinardMFSchornagelJHLeucovorin rescue of human cancer and bone marrow cells following edatrexate or methotrexateBiochem Pharmacol19944746596657510479
  • GrantSCKrisMGYoungCWSirotnakFMEdatrexate, an antifolate with antitumor activity: a reviewCancer Invest199311136458422595
  • KrisMGKinahanJJGrallaRJPhase I trial and clinical pharmacological evaluation of 10-ethyl-10-deazaaminopterin in adult patients with advanced cancerCancer Res19884819557355793416310
  • SchornagelJHVerweijJde MulderPHRandomized phase III trial of edatrexate versus methotrexate in patients with metastatic and/or recurrent squamous cell carcinoma of the head and neck: a European Organization for Research and Treatment of Cancer Head and Neck Cancer Cooperative Group studyJ Clin Oncol1995137164916557602354
  • RosowskyAPT523 and other aminopterin analogs with a hemiphthaloyl-L-ornithine side chain: exceptionally tight-binding inhibitors of dihydrofolate reductase which are transported by the reduced folate carrier but cannot form polyglutamatesCurr Med Chem19996432935210101216
  • GilesFRizzieriDAGeorgeSA phase I study of talvesta (talotrexin) in relapsed or refractory leukemia or myelodysplastic syndromeBlood (ASH Annual Meeting Abstracts)2006108111968
  • MarshallJLDeLapRJClinical pharmacokinetics and pharmacology of trimetrexateClin Pharmacokinet19942631902008194282
  • FlemingGFSchilskyRLAntifolates: the next generationSemin Oncol19921967077191462168
  • TakimotoCHNew antifolates: pharmacology and clinical applicationsOncologist199611 and 2688110387971
  • TaylorIWSlowiaczekPFriedlanderMLTattersallMHSelective toxicity of a new lipophilic antifolate, BW301U, for methotrexate-resistant cells with reduced drug uptakeCancer Res19854539789823971388
  • AdamsonPCBalisFMMiserJPediatric phase I trial, pharmacokinetic study, and limited sampling strategy for piritrexim administered on a low-dose, intermittent scheduleCancer Res19925235215241732038
  • SakoffJAAcklandSPThymidylate synthase inhibition induces S-phase arrest, biphasic mitochondrial alterations and caspase-dependent apoptosis in leukaemia cellsCancer Chemother Pharmacol200046647748711138461
  • van der WiltCLSmidKPetersGJEffects of antifolates on the binding of 5-fluoro-2′-deoxyuridine monophosphate to thymidylate synthaseBiochem Pharmacol200264466967512167486
  • JhawerMRosenLDanceyJPhase II trial of nolatrexed dihydrochloride [Thymitaq, AG 337] in patients with advanced hepatocellular carcinomaInvest New Drugs2007251859416957834
  • HannaNShepherdFAFossellaFVRandomized phase III trial of pemetrexed versus docetaxel in patients with non-small-cell lung cancer previously treated with chemotherapyJ Clin Oncol20042291589159715117980
  • SelvaggiGScagliottiGVHistologic subtype in NSCLC: does it matter?Oncology (Williston Park)200923131133114020043461
  • NiyikizaCHanauskeARRusthovenJJPemetrexed safety and dosing strategySemin Oncol2002296 Suppl 18242912571807
  • GoldmanIDZhaoRMolecular, biochemical, and cellular pharmacology of pemetrexedSemin Oncol2002296 Suppl 1831712571805
  • PizzornoGMorosonBACashmoreARMultifactorial resistance to 5,10-dideazatetrahydrofolic acid in cell lines derived from human lymphoblastic leukemia CCRF-CEMCancer Res19955535665737834626
  • RheeMSRyanTJGalivanJGlutamyl hydrolase and the multitargeted antifolate LY231514Cancer Chemother Pharmacol199944542743210501918
  • de JongeMJPuntCJSparreboomAPhase I and pharmacologic study of oral ZD9331, a novel nonpolyglutamated thymidylate synthase inhibitor, in adult patients with solid tumorsJ Clin Oncol20022071923193111919253
  • CalvertAHBiochemical pharmacology of pemetrexedOncology (Williston Park)20041813 Suppl 8131715655931
  • DickgreberNJSorensenJBPaz-AresLGPemetrexed safety and pharmacokinetics in patients with third-space fluidClin Cancer Res201016102872288020460481
  • BrandesJCGrossmanSAAhmadHAlteration of pemetrexed excretion in the presence of acute renal failure and effusions: presentation of a case and review of the literatureCancer Invest200624328328716809156
  • NiyikizaCBakerSDSeitzDEHomocysteine and methylmalonic acid: markers to predict and avoid toxicity from pemetrexed therapyMol Cancer Ther20021754555212479273