Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Biol Chem ; 287(7): 4982-95, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22179615

ABSTRACT

The proton-coupled folate transporter (PCFT; SLC46A1) is a proton-folate symporter that is abundantly expressed in solid tumors and normal tissues, such as duodenum. The acidic pH optimum for PCFT is relevant to intestinal absorption of folates and could afford a means of selectively targeting tumors with novel cytotoxic antifolates. PCFT is a member of the major facilitator superfamily of transporters. Because major facilitator superfamily members exist as homo-oligomers, we tested this for PCFT because such structures could be significant to PCFT mechanism and regulation. By transiently expressing PCFT in reduced folate carrier- and PCFT-null HeLa (R1-11) cells and chemical cross-linking with 1,1-methanediyl bismethanethiosulfonate and Western blotting, PCFT species with molecular masses approximating those of the PCFT dimer and higher order oligomers were detected. Blue native polyacrylamide gel electrophoresis identified PCFT dimer, trimer, and tetramer forms. PCFT monomers with hemagglutinin and His(10) epitope tags were co-expressed in R1-11 cells, solubilized, and bound to nickel affinity columns, establishing their physical associations. Co-expressing YPet and ECFP*-tagged PCFT monomers enabled transport and fluorescence resonance energy transfer in plasma membranes of R1-11 cells. Combined wild-type (WT) and inactive mutant P425R PCFTs were targeted to the cell surface by surface biotinylation/Western blots and confocal microscopy and functionally exhibited a "dominant-positive" phenotype, implying positive cooperativity between PCFT monomers and functional rescue of mutant by WT PCFT. Our results demonstrate the existence of PCFT homo-oligomers and imply their functional and regulatory impact. Better understanding of these higher order PCFT structures may lead to therapeutic applications related to folate uptake in hereditary folate malabsorption, and delivery of PCFT-targeted chemotherapy drugs for cancer.


Subject(s)
Cell Membrane/metabolism , Folic Acid/metabolism , Protein Multimerization/physiology , Proton-Coupled Folate Transporter/metabolism , Biological Transport , Cell Membrane/genetics , Folic Acid/genetics , HeLa Cells , Humans , Hydrogen-Ion Concentration , Proton-Coupled Folate Transporter/chemistry , Proton-Coupled Folate Transporter/genetics
2.
Mol Pharmacol ; 82(4): 591-600, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22740639

ABSTRACT

Uptake of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with four or three bridge carbons [compound 1 (C1) and compound 2 (C2), respectively] into solid tumors by the proton-coupled folate transporter (PCFT) represents a novel therapeutic strategy that harnesses the acidic tumor microenvironment. Although these compounds are not substrates for the reduced folate carrier (RFC), the major facilitative folate transporter, RFC expression may alter drug efficacies by affecting cellular tetrahydrofolate (THF) cofactor pools that can compete for polyglutamylation and/or binding to intracellular enzyme targets. Human tumor cells including wild-type (WT) and R5 (RFC-null) HeLa cells express high levels of PCFT protein. C1 and C2 inhibited proliferation of R5 cells 3 to 4 times more potently than WT cells or R5 cells transfected with RFC. Transport of C1 and C2 was virtually identical between WT and R5 cells, establishing that differences in drug sensitivities between sublines were independent of PCFT transport. Steady-state intracellular [³H]THF cofactors derived from [³H]5-formyl-THF were depleted in R5 cells compared with those in WT cells, an effect exacerbated by C1 and C2. Whereas C1 and C2 polyglutamates accumulated to similar levels in WT and R5 cells, there were differences in polyglutamyl distributions in favor of the longest chain length forms. In severe combined immunodeficient mice, the antitumor efficacies of C1 and C2 were greater toward subcutaneous R5 tumors than toward WT tumors, confirming the collateral drug sensitivities observed in vitro. Thus, solid tumor-targeted antifolates with PCFT-selective cellular uptake should have enhanced activities toward tumors lacking RFC function, reflecting contraction of THF cofactor pools.


Subject(s)
Antineoplastic Agents/pharmacology , Folic Acid Antagonists/pharmacology , Proton-Coupled Folate Transporter/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , Reduced Folate Carrier Protein/metabolism , Thiophenes/pharmacology , Animals , Antineoplastic Agents/metabolism , Biological Transport , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Extracellular Space/metabolism , Female , Folic Acid Antagonists/metabolism , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Pteroylpolyglutamic Acids/metabolism , Pyrimidines/metabolism , Pyrroles/metabolism , Structure-Activity Relationship , Tetrahydrofolates/metabolism , Thiophenes/metabolism , Tumor Microenvironment
3.
J Pharmacol Exp Ther ; 342(3): 696-708, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22653877

ABSTRACT

N-[4-[[(2,4-diamino-6-pterdinyl)methyl]amino]benzoyl]-L/D-glutamic acid (L/D-AMT) is an investigational drug in phase 1 clinical development that consists of the L-and D-enantiomers of aminopterin (AMT). L/D-AMT is obtained from a novel process for making the L-enantiomer (L-AMT), a potent oral antiinflammatory agent. The purpose of these studies was to characterize oral uptake and safety in the dog and human of each enantiomer alone and in combination and provide in vitro evidence for a mechanism of intestinal absorption. This is the first report of L /D-AMT in humans. In dogs (n = 40) orally dosed with L-AMT or D-AMT absorption was stereoselective for the L-enantiomer (6- to 12-fold larger peak plasma concentration after oral administration and area under the plasma concentration-time curve at 0-4 h; p < 0.001). D-AMT was not toxic at the maximal dose tested (82.5 mg/kg), which was 100-fold larger than the maximal nonlethal L-AMT dose (0.8 mg/kg). Dogs (n = 10) and humans with psoriasis (n = 21) orally administered L-AMT and L /D-AMT at the same L-enantiomer dose resulted in stereoselective absorption (absent D-enantiomer in plasma), bioequivalent L-enantiomer pharmacokinetics, and equivalent safety. Thus, the D-enantiomer in L/D-AMT did not perturb L-enantiomer absorption or alter the safety of L-AMT. In vitro uptake by the human proton-coupled folate transporter (PCFT) demonstrated minimal transport of D-AMT compared with L-AMT, mirroring the in vivo findings. Enantiomer selectivity by PCFT was attributable almost entirely to decreased binding affinity rather than changes in transport rate. Collectively, our results demonstrate a strong in vitro-in vivo correlation implicating stereoselective transport by PCFT as the mechanism underlying stereoselective absorption observed in vivo.


Subject(s)
Aminopterin/adverse effects , Aminopterin/pharmacokinetics , Intestinal Absorption/physiology , Proton-Coupled Folate Transporter/metabolism , Psoriasis/metabolism , Administration, Oral , Adult , Aminopterin/administration & dosage , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Biological Transport/drug effects , CHO Cells , Cells, Cultured , Cricetinae , Cross-Over Studies , Dogs , Female , Humans , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Male , Membrane Transport Proteins/metabolism , Middle Aged , Stereoisomerism , Young Adult
4.
J Neurol ; 269(4): 2162-2171, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34537872

ABSTRACT

BACKGROUND AND OBJECTIVE: Pathogenic variants in KCNT1 have been associated with severe forms of epilepsy, typically sleep-related hypermotor epilepsy or epilepsy of infancy with migrating focal seizures. To show that pathogenic variants in KCNT1 can be associated with mild extra-frontal epilepsy, we report a KCNT1 family with a wide spectrum of phenotypes ranging from developmental and epileptic encephalopathy to mild focal epilepsy without cognitive regression and not consistent with sleep-related hypermotor epilepsy. METHODS: A large Canadian family of Caucasian descent including 9 affected family members was recruited. Family members were phenotyped by direct interview and review of existing medical records. Clinical epilepsy gene panel analysis and exome sequencing were performed. RESULTS: Phenotypic information was available for five family members of which two had developmental and epileptic encephalopathy and three had normal development and focal epilepsy with presumed extra-frontal onset. All three had predominantly nocturnal seizures that did not show hyperkinetic features. All three reported clusters of seizures at night with a feeling of being unable to breathe associated with gasping for air, choking and/or repetitive swallowing possibly suggesting insular or opercular involvement. Genetic analysis identified a heterozygous KCNT1 c.2882G > A, p.Arg961His variant that was predicted to be deleterious. DISCUSSION: This family demonstrates that the phenotypic spectrum associated with KCNT1 pathogenic variants is broader than previously assumed. Our findings indicate that variants in KCNT1 can be associated with mild focal epilepsy and should not be excluded during variant interpretation in such patients based solely on gene-disease validity.


Subject(s)
Epilepsies, Partial , Epileptic Syndromes , Nerve Tissue Proteins , Potassium Channels, Sodium-Activated , Canada , Epilepsies, Partial/genetics , Epileptic Syndromes/genetics , Humans , Mutation , Nerve Tissue Proteins/genetics , Phenotype , Potassium Channels, Sodium-Activated/genetics
5.
Mol Pharmacol ; 80(6): 1096-107, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21940787

ABSTRACT

The proton-coupled folate transporter (PCFT) is a proton-folate symporter with an acidic pH optimum. By real-time reverse transcription-polymerase chain reaction, PCFT was expressed in the majority of 53 human tumor cell lines, with the highest levels in Caco-2 (colorectal adenocarcinoma), SKOV3 (ovarian), and HepG2 (hepatoma) cells. A novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate (compound 1) was used to establish whether PCFT can deliver cytotoxic drug under pH conditions that mimic the tumor microenvironment. Both 1 and pemetrexed (Pmx) inhibited proliferation of R1-11-PCFT4 HeLa cells engineered to express PCFT without the reduced folate carrier (RFC) and of HepG2 cells expressing both PCFT and RFC. Unlike Pmx, 1 did not inhibit proliferation of R1-11-RFC6 HeLa cells, which express RFC without PCFT. Treatment of R1-11-PCFT4 cells at pH 6.8 with 1 or Pmx inhibited colony formation with dose and time dependence. Transport of [(3)H]compound 1 into R1-11-PCFT4 and HepG2 cells was optimal at pH 5.5 but appreciable at pH 6.8. At pH 6.8, [(3)H]compound 1 was metabolized to (3)H-labeled polyglutamates. Glycinamide ribonucleotide formyltransferase (GARFTase) in R1-11-PCFT4 cells was inhibited by 1 at pH 6.8, as measured by an in situ GARFTase assay, and was accompanied by substantially reduced ATP levels. Compound 1 caused S-phase accumulation and a modest level of apoptosis. An in vivo efficacy trial with severe combined immunodeficient mice implanted with subcutaneous HepG2 tumors showed that compound 1 was active. Our findings suggest exciting new therapeutic possibilities to selectively deliver novel antifolate drugs via transport by PCFT over RFC by exploiting the acidic tumor microenvironment.


Subject(s)
Drug Delivery Systems/methods , Folic Acid Antagonists/metabolism , Neoplasms/metabolism , Proton-Coupled Folate Transporter/metabolism , Pyrimidines/metabolism , Animals , Dose-Response Relationship, Drug , Female , Folic Acid Antagonists/administration & dosage , HeLa Cells , Hep G2 Cells , Humans , Mice , Mice, Inbred ICR , Mice, SCID , Neoplasms/drug therapy , Pyrimidines/administration & dosage , Xenograft Model Antitumor Assays/methods
6.
J Biol Chem ; 285(7): 4732-40, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20018840

ABSTRACT

The reduced folate carrier (RFC) is the major transport system for folates in mammals. We previously demonstrated the existence of human RFC (hRFC) homo-oligomers and established the importance of these higher order structures to intracellular trafficking and carrier function. In this report, we examined the operational significance of hRFC oligomerization and the minimal functional unit for transport. In negative dominance experiments, multimeric transporters composed of different ratios of active (either wild type (WT) or cysteine-less (CLFL)) and inactive (either inherently inactive (Y281L and R373A) due to mutation, or resulting from inactivation of the Y126C mutant by (2-sulfonatoethyl) methanethiosulfonate (MTSES)) hRFC monomers were expressed in hRFC-null HeLa (R5) cells, and residual WT or CLFL activity was measured. In either case, residual transport activity with increasing levels of inactive mutant correlated linearly with the fraction of WT or CLFL hRFC in plasma membranes. When active covalent hRFC dimers, generated by fusing CLFL and Y126C monomers, were expressed in R5 cells and treated with MTSES, transport activity of the CLFL-CLFL dimer was unaffected, whereas Y126C-Y126C was potently (64%) inhibited; heterodimeric CLFL-Y126C and Y126C-CLFL were only partly (27 and 23%, respectively) inhibited by MTSES. In contrast to Y126C-Y126C, trans-stimulation of methotrexate uptake by intracellular folates for Y126C-CLFL and CLFL-Y126C was nominally affected by MTSES. Collectively, these results strongly support the notion that each hRFC monomer comprises a single translocation pathway for anionic folate substrates and functions independently of other monomers (i.e. despite an oligomeric structure, hRFC functions as a monomer).


Subject(s)
Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Blotting, Western , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Immunoprecipitation , Membrane Transport Proteins/genetics , Mesylates/pharmacology , Microscopy, Confocal , Mutagenesis, Site-Directed , Mutation , Protein Multimerization/genetics , Protein Multimerization/physiology , Reduced Folate Carrier Protein , Signal Transduction/drug effects , Signal Transduction/genetics , Structure-Activity Relationship
7.
Biochem J ; 430(2): 265-74, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20557288

ABSTRACT

RFC (reduced folate carrier) is the major transporter for reduced folates and antifolates [e.g. MTX (methotrexate)]. RFC is characterized by two halves, each with six TMD (transmembrane domain) alpha helices connected by a hydrophilic loop, and cytoplasmic N- and C-termini. We previously identified TMDs 4, 5, 7, 8, 10 and 11 as forming the hydrophilic cavity for translocation of (anti)folates. The proximal end of TMD8 (positions 311-314) was implicated in substrate binding from scanning-cysteine accessibility methods; cysteine replacement of Ser313 resulted in loss of transport. In the present study, Ser313 was mutated to alanine, cysteine, phenylalanine and threonine. Mutant RFCs were expressed in RFC-null R5 HeLa cells. Replacement of Ser313 with cysteine or phenylalanine abolished MTX transport, whereas residual activity was preserved for the alanine and threonine mutants. In stable K562 transfectants, S313A and S313T RFCs showed substantially decreased Vmax values without changes in Kt values for MTX compared with wild-type RFC. S313A and S313T RFCs differentially impacted binding of ten diverse (anti)folate substrates. Cross-linking between TMD8 and TMD5 was studied by expressing cysteine-less TMD1-6 (N6) and TMD7-12 (C6) half-molecules with cysteine insertions spanning these helices in R5 cells, followed by treatment with thiol-reactive homobifunctional cross-linkers. C6-C6 and N6-N6 cross-links were seen for all cysteine pairs. From the N6 and C6 cysteine pairs, Cys175/Cys311 was cross-linked; cross-linking increased in the presence of transport substrates. The results of the present study indicate that the proximal end of TMD8 is juxtaposed to TMD5 and is conformationally active in the presence of transport substrates, and TMD8, including Ser313, probably contributes to the RFC substrate-binding domain.


Subject(s)
Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Amino Acid Sequence , Cross-Linking Reagents/chemistry , Humans , Kinetics , Membrane Transport Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Secondary , Reduced Folate Carrier Protein , Substrate Specificity
8.
Mol Pharmacol ; 78(4): 577-87, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20601456

ABSTRACT

The proton-coupled folate transporter (PCFT) is a folate-proton symporter with an acidic pH optimum, approximating the microenvironments of solid tumors. We tested 6-substituted pyrrolo[2,3-d]pyrimidine antifolates with one to six carbons in the bridge region for inhibition of proliferation in isogenic Chinese hamster ovary (CHO) and HeLa cells expressing PCFT or reduced folate carrier (RFC). Only analogs with three and four bridge carbons (N-{4-[3-2-amino-4-oxo-4,7-dihydro-3H-pyrrolo[2,3-d]-pyrimidin-6-yl)propyl]benzoyl}-L-glutamic acid (compound 2) and N-{4-[4-2-amino-4-oxo-4,7-dihydro-3H-pyrrolo[2,3-d]-pyrimidin-6-yl)butyl]benzoyl}*-L-glutamic acid (compound 3), respectively) were inhibitory, with 2 ≫ 3. Activity toward RFC-expressing cells was negligible. Compound 2 and pemetrexed (Pmx) competed with [(3)H]methotrexate for PCFT transport in PCFT-expressing CHO (R2/hPCFT4) cells from pH 5.5 to 7.2; inhibition increased with decreasing pH. In Xenopus laevis oocytes microinjected with PCFT cRNA, uptake of 2, like that of Pmx, was electrogenic. Cytotoxicity of 2 toward R2/hPCFT4 cells was abolished in the presence of adenosine or 5-amino-4-imidazolecarboxamide, suggesting that glycinamide ribonucleotide formyltransferase (GARFTase) in de novo purine biosynthesis was the primary target. Compound 2 decreased GTP and ATP pools by ∼50 and 75%, respectively. By an in situ GARFTase assay, 2 was ∼20-fold more inhibitory toward intracellular GARFTase than toward cell growth or colony formation. Compound 2 irreversibly inhibited clonogenicity, although this required at least 4 h of exposure. Our results document the potent antiproliferative activity of compound 2, attributable to its efficient cellular uptake by PCFT, resulting in inhibition of GARFTase and de novo purine biosynthesis. Furthermore, they establish the feasibility of selective chemotherapy drug delivery via PCFT over RFC, a process that takes advantage of a unique biological feature of solid tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Folic Acid Antagonists/metabolism , Membrane Transport Proteins/metabolism , Neoplasms/drug therapy , Purines/biosynthesis , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , CHO Cells , Cricetinae , Cricetulus , Female , Growth Inhibitors/administration & dosage , Growth Inhibitors/chemistry , Growth Inhibitors/metabolism , HeLa Cells , Humans , Neoplasms/metabolism , Proton-Coupled Folate Transporter , Purines/antagonists & inhibitors , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrroles/chemistry , Pyrroles/metabolism , Xenopus laevis
9.
Mol Pharmacol ; 73(4): 1274-81, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18182479

ABSTRACT

Reduced folate carrier (RFC) is the major membrane transporter for folates and antifolates in mammalian tissues. Recent studies used radioaffinity labeling with N-hydroxysuccinimide (NHS)-[(3)H]methotrexate (MTX) to localize substrate binding to residues in transmembrane domain (TMD) 11 of human RFC. To identify the modified residue(s), seven nucleophilic residues in TMD11 were mutated to Val or Ala and mutant constructs expressed in RFC-null HeLa cells. Only K411A RFC was not inhibited by NHS-MTX. By radioaffinity labeling with NHS-[(3)H]MTX, wild-type (wt) RFC was labeled; for K411A RFC, radiolabeling was abolished. When Lys411 was replaced with Ala, Arg, Gln, Glu, Leu, and Met, only K411E RFC showed substantially decreased transport. Nine classic diamino furo[2,3-d]pyrimidine antifolates with unsubstituted alpha- and gamma-carboxylates (1), hydrogen- or methyl-substituted alpha-(2,3) or gamma-(4,5) carboxylates, or substitutions of both alpha- and gamma-carboxylates (6-9) were used to inhibit [(3)H]MTX transport with RFC-null K562 cells expressing wt and K411A RFCs. For wt and K411A RFCs, inhibitory potencies were in the order 4 > 5 > 1 > 3 > 2; 6 to 9 were poor inhibitors. Inhibitions decreased in the presence of physiologic anions. When NHS esters of 1, 2, and 4 were used to covalently modify wt RFC, inhibitory potencies were in the order 2 > 1 > 4; inhibition was abolished for K411A RFC. These results establish that Lys411 participates in substrate binding via an ionic association with the substrate gamma-carboxylate; however, this is not essential for transport. An unmodified alpha-carboxylate is required for high-affinity substrate binding to RFC, whereas the gamma-carboxyl is not essential.


Subject(s)
Affinity Labels , Lysine/metabolism , Membrane Transport Proteins/metabolism , Mutagenesis, Site-Directed/methods , Biological Transport/drug effects , Esters/metabolism , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , HeLa Cells , Humans , Kinetics , Membrane Transport Proteins/chemistry , Methotrexate/metabolism , Mutant Proteins/metabolism , Protein Structure, Tertiary , Reduced Folate Carrier Protein , Structure-Activity Relationship , Succinimides/metabolism , Transfection
10.
Mol Genet Metab ; 93(2): 95-103, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17983788

ABSTRACT

The ubiquitously expressed reduced folate carrier (RFC) or SLC19A1 is recognized to be an essential transport system for folates in mammalian cells and tissues. In addition to its generalized role as a folate transporter, RFC provides specialized tissue functions including absorption across intestinal/colonic epithelia, transport across the basolateral membrane of renal proximal tubules, transplacental transport of folates, and folate transport across the blood-brain barrier. The human RFC (hRFC) gene is regulated by five major upstream non-coding regions (designated A1/A2, A, B, C, and D), each transcribed from a unique promoter. Altogether, at least 14 distinct hRFC transcripts can be envisaged in which different 5' untranslated regions (UTRs) are fused to a common splice acceptor region (positions -1 to -49) within the first coding exon with a common 1776bp coding sequence. The 5' non-coding regions are characterized by alternate transcription start sites, multiple splice forms, and selective tissue distributions. Alternate 5' UTRs impact mRNA stabilities and translation efficiencies, and result in synthesis of modified hRFC proteins translated from upstream AUGs. In this report, we describe production and characterization of transgenic mice (TghRFC1) containing a functional hRFC gene and of humanized mice in which the mRFC gene is inactivated and an active hRFC gene has been introduced. The mice appear to be healthy and to breed well. Analysis of tissue specificity of expression in both the TghRFC1 and humanized hRFC mice by real-time RT-PCR demonstrates that the hRFC gene is expressed with a specificity closely resembling that seen in human tissues. For the humanized hRFC mice, levels of B and A1/A2 5' UTRs predominated in all mice/tissues, thus resembling results in normal human tissues. Lower levels of A and C 5' UTRs were also detected. The availability of humanized mouse models for hRFC will permit investigators to address critical unanswered questions pertinent to human health and disease. These include the ability to analyze the hRFC gene in vivo, to control dietary and other environmental conditions that may impact levels of gene expression, and to control the genetics of the mice in order to assess the effects of hRFC gene alterations on tissue folate uptake and distribution, none of which can be easily achieved in human populations.


Subject(s)
Folic Acid/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , 5' Untranslated Regions , Animals , Base Sequence , DNA Primers/genetics , Female , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Transgenic , Models, Animal , Pregnancy , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reduced Folate Carrier Protein , Species Specificity , Tissue Distribution
11.
Cancer Chemother Pharmacol ; 71(4): 999-1011, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23412628

ABSTRACT

PURPOSE: We examined whether the novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate, compound 2, might be an effective treatment for malignant pleural mesothelioma (MPM), reflecting its selective membrane transport by the proton-coupled folate transport (PCFT) over the reduced folate carrier (RFC). METHODS: HeLa sublines expressing exclusively PCFT (R1-11-PCFT4) or RFC (R1-11-RFC6) and H2452 MPM cells were assayed for transport with [(3)H]compound 2. [(3)H]Polyglutamate metabolites of compound 2 were measured in R1-11-PCFT4 and H2452 cells. In vitro cell proliferation assays and colony formation assays were performed. Inhibition of glycinamide ribonucleotide formyltransferase (GARFTase) was assayed by nucleoside protection assays and in situ GARFTase assays with [(14)C]glycine. In vivo efficacy was established with early- and advanced-stage H2452 xenografts in severe-combined immunodeficient (SCID) mice administered intravenous compound 2. RESULTS: [(3)H]Compound 2 was selectively transported by PCFT and was metabolized to polyglutamates. Compound 2 selectively inhibited proliferation of R1-11-PCFT4 cells over R1-11-RFC6 cells. H2452 human MPM cells were sensitive to the antiproliferative effects of compound 2. By colony-forming assays with H2452 cells, compound 2 was cytotoxic. Compound 2 inhibited GARFTase in de novo purine biosynthesis. In vivo efficacy was confirmed toward early- and advanced-stage H2452 xenografts in SCID mice administered compound 2. CONCLUSIONS: Our results demonstrate potent antitumor efficacy of compound 2 toward H2452 MPM cells in vitro and in vivo, reflecting its efficient membrane transport by PCFT, synthesis of polyglutamates, and inhibition of GARFTase. Selectivity for non-RFC cellular uptake processes by tumor-targeted antifolates such as compound 2 presents an exciting new opportunity for treating solid tumors.


Subject(s)
Folic Acid Antagonists/therapeutic use , Mesothelioma/drug therapy , Proton-Coupled Folate Transporter/physiology , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Animals , Cell Proliferation/drug effects , Female , Folic Acid Antagonists/pharmacokinetics , Folic Acid Antagonists/pharmacology , HeLa Cells , Humans , Mice , Mice, SCID , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Polyglutamic Acid/metabolism , Pyrimidines/pharmacokinetics , Pyrroles/pharmacokinetics , Xenograft Model Antitumor Assays
12.
J Med Chem ; 56(21): 8684-95, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24111942

ABSTRACT

A new series of 6-substituted straight side chain pyrrolo[2,3-d]pyrimidines 3a-d with varying chain lengths (n = 5-8) was designed and synthesized as part of our program to provide targeted antitumor agents with folate receptor (FR) cellular uptake specificity and glycinamide ribonucleotide formyltransferase (GARFTase) inhibition. Carboxylic acids 4a-d were converted to the acid chlorides and reacted with diazomethane, followed by 48% HBr to generate the α-bromomethylketones 5a-d. Condensation of 2,4-diamino-6-hydroxypyrimidine 6 with 5a-d afforded the 6-substituted pyrrolo[2,3-d]pyrimidines 7a-d. Hydrolysis and subsequent coupling with diethyl l-glutamate and saponification afforded target compounds 3a-d. Compounds 3b-d showed selective cellular uptake via FRα and -ß, associated with high affinity binding and inhibition of de novo purine nucleotide biosynthesis via GARFTase, resulting in potent inhibition against FR-expressing Chinese hamster cells and human KB tumor cells in culture. Our studies establish, for the first time, that a side chain benzoyl group is not essential for tumor-selective drug uptake by FRα.


Subject(s)
Folate Receptor 1/antagonists & inhibitors , Folic Acid Antagonists/pharmacology , Folic Acid/metabolism , Purine Nucleotides/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , Animals , CHO Cells , Cell Proliferation/drug effects , Cells, Cultured , Cricetulus , Dose-Response Relationship, Drug , Folate Receptor 1/metabolism , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/chemistry , Humans , KB Cells , Models, Molecular , Molecular Structure , Purine Nucleotides/biosynthesis , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship
13.
J Med Chem ; 56(24): 10016-10032, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24256410

ABSTRACT

We synthesized 5-substituted pyrrolo[2,3-d]pyrimidine antifolates (compounds 5-10) with one-to-six bridge carbons and a benozyl ring in the side chain as antitumor agents. Compound 8 with a 4-carbon bridge was the most active analogue and potently inhibited proliferation of folate receptor (FR) α-expressing Chinese hamster ovary and KB human tumor cells. Growth inhibition was reversed completely or in part by excess folic acid, indicating that FRα is involved in cellular uptake, and resulted in S-phase accumulation and apoptosis. Antiproliferative effects of compound 8 toward KB cells were protected by excess adenosine but not thymidine, establishing de novo purine nucleotide biosynthesis as the targeted pathway. However, 5-aminoimidazole-4-carboxamide (AICA) protection was incomplete, suggesting inhibition of both AICA ribonucleotide formyltransferase (AICARFTase) and glycinamide ribonucleotide formyltransferase (GARFTase). Inhibition of GARFTase and AICARFTase by compound 8 was confirmed by cellular metabolic assays and resulted in ATP pool depletion. To our knowledge, this is the first example of an antifolate that acts as a dual inhibitor of GARFTase and AICARFTase as its principal mechanism of action.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Hydroxymethyl and Formyl Transferases/antagonists & inhibitors , Purine Nucleotides/biosynthesis , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , CHO Cells , Cell Proliferation/drug effects , Cricetulus , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Humans , Hydroxymethyl and Formyl Transferases/metabolism , KB Cells , Models, Molecular , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacology , Structure-Activity Relationship
14.
J Med Chem ; 55(4): 1758-70, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22243528

ABSTRACT

We previously reported the selective transport of classical 2-amino-4-oxo-6-substituted pyrrolo[2,3-d]pyrimidines with a thienoyl-for-benzoyl-substituted side chain and a three- (3a) and four-carbon (3b) bridge. Compound 3a was more potent than 3b against tumor cells. While 3b was completely selective for transport by folate receptors (FRs) and the proton-coupled folate transporter (PCFT) over the reduced folate carrier (RFC), 3a was not. To determine if decreasing the distance between the bicyclic scaffold and l-glutamate in 3b would preserve transport selectivity and potency against human tumor cells, 3b regioisomers with [1,3] (7 and 8) and [1,2] (4, 5, and 6) substitutions on the thienoyl ring and with acetylenic insertions in the four-atom bridge were synthesized and evaluated. Compounds 7 and 8 were potent nanomolar inhibitors of KB and IGROV1 human tumor cells with complete selectivity for FRα and PCFT over RFC.


Subject(s)
Antineoplastic Agents/chemical synthesis , Folate Receptor 1/metabolism , Proton-Coupled Folate Transporter/metabolism , Purines/antagonists & inhibitors , Pyrimidines/chemical synthesis , Pyrroles/chemical synthesis , Reduced Folate Carrier Protein/metabolism , Thiophenes/chemical synthesis , Alkynes/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Drug Screening Assays, Antitumor , Humans , Purines/biosynthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Stereoisomerism , Structure-Activity Relationship , Thiophenes/chemistry , Thiophenes/pharmacology
15.
J Med Chem ; 54(20): 7150-64, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21879757

ABSTRACT

2-Amino-4-oxo-6-substituted pyrrolo[2,3-d]pyrimidine antifolates with a thienoyl side chain (compounds 1-3, respectively) were synthesized for comparison with compound 4, the previous lead compound of this series. Conversion of hydroxyl acetylen-thiophene carboxylic esters to thiophenyl-α-bromomethylketones and condensation with 2,4-diamino-6-hydroxypyrimidine afforded the 6-substituted pyrrolo[2,3-d]pyrimidine compounds of type 18 and 19. Coupling with l-glutamate diethyl ester, followed by saponification, afforded 1-3. Compound 3 selectively inhibited the proliferation of cells expressing folate receptors (FRs) α or ß, or the proton-coupled folate transporter (PCFT), including KB and IGROV1 human tumor cells, much more potently than 4. Compound 3 was more inhibitory than 4 toward ß-glycinamide ribonucleotide formyltransferase (GARFTase). Both 3 and 4 depleted cellular ATP pools. In SCID mice with IGROV1 tumors, 3 was more efficacious than 4. Collectively, our results show potent antitumor activity for 3 in vitro and in vivo, associated with its selective membrane transport by FRs and PCFT over RFC and inhibition of GARFTase, clearly establishing the 3-atom bridge as superior to the 1-, 2-, and 4-atom bridge lengths for the activity of this series.


Subject(s)
Antineoplastic Agents/chemical synthesis , Folate Receptor 1/metabolism , Folate Receptor 2/metabolism , Folic Acid Antagonists/chemical synthesis , Glutamates/chemical synthesis , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Proton-Coupled Folate Transporter/metabolism , Pyrimidines/chemical synthesis , Pyrimidinones/chemical synthesis , Reduced Folate Carrier Protein/metabolism , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Glutamates/chemistry , Glutamates/pharmacology , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Oocytes/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous , Xenopus
16.
J Med Chem ; 53(3): 1306-18, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20085328

ABSTRACT

2-Amino-4-oxo-6-substituted pyrrolo[2,3-d]pyrimidines with a thienoyl side chain and four to six carbon bridge lengths (compounds 1-3) were synthesized as substrates for folate receptors (FRs) and the proton-coupled folate transporter (PCFT). Conversion of acetylene carboxylic acids to alpha-bromomethylketones and condensation with 2,4-diamino-6-hydroxypyrimidine afforded the 6-substituted pyrrolo[2,3-d]pyrimidines. Sonogashira coupling with (S)-2-[(5-bromo-thiophene-2-carbonyl)-amino]-pentanedioic acid diethyl ester, followed by hydrogenation and saponification, afforded 1-3. Compounds 1 and 2 potently inhibited KB and IGROV1 human tumor cells that express FR alpha, reduced folate carrier (RFC), and PCFT. The analogs were selective for FR and PCFT over RFC. Glycinamide ribonucleotide formyltransferase was the principal cellular target. In SCID mice with KB tumors, 1 was highly active against both early (3.5 log kill, 1/5 cures) and advanced (3.7 log kill, 4/5 complete remissions) stage tumors. Our results demonstrate potent in vitro and in vivo antitumor activity for 1 due to selective transport by FRs and PCFT over RFC.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Folic Acid Antagonists/chemical synthesis , Membrane Transport Proteins/metabolism , Purines/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Animals , Antineoplastic Agents/chemistry , CHO Cells , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Cells, Cultured , Cricetinae , Cricetulus , Female , Folate Receptors, GPI-Anchored , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Humans , Mice , Mice, Inbred ICR , Mice, SCID , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proton-Coupled Folate Transporter , Purines/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Reduced Folate Carrier Protein
17.
J Med Chem ; 52(9): 2940-51, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19371039

ABSTRACT

A series of seven 2-amino-4-oxo-6-substituted thieno[2,3-d]pyrimidines with bridge length variations (from 2 to 8 carbon atoms) were synthesized as selective folate receptor (FR) alpha and beta substrates and as antitumor agents. The syntheses were accomplished from appropriate allylalcohols and 4-iodobenzoate to afford the aldehydes, which were converted to the appropriate 2-amino-4-carbethoxy-5-substituted thiophenes 23-29. Cyclization with chloroformamidine afforded the thieno[2,3-d]pyrimidines 30-36, which were hydrolyzed and coupled with diethyl-L-glutamate, followed by saponification, to give the target compounds 2-8. Compounds 3-6 were potent growth inhibitors (IC(50) 4.7-334 nM) of human tumor cells (KB and IGROV1) that express FRs. In addition, compounds 3-6 inhibited the growth of Chinese hamster ovary (CHO) cells that expressed FRs but not the reduced folate carrier (RFC) or proton-coupled folate transporter (PCFT). However, the compounds were inactive toward CHO cells that lacked FRs but contained either the RFC or PCFT. By nucleoside and 5-amino-4-imidazole carboxamide (AICA) protection studies, along with in vitro and in situ enzyme activity assays, the mechanism of antitumor activity was identified as the dual inhibition of glycinamide ribonucleotide formyltransferase and, likely, AICA ribonucleotide formyltransferase. The dual inhibitory activity of the active thieno[2,3-d]pyrimidine antifolates and the FR specificity represent unique mechanistic features for these compounds distinct from all other known antifolates. The potent inhibitory effects of compounds 3-6 toward cells expressing FRs but not PCFT provide direct evidence that cellular uptake of this series of compounds by FRs does not depend on the presence of PCFT and argues that direct coupling between these transporters is not obligatory.


Subject(s)
Anion Transport Proteins/metabolism , Carrier Proteins/metabolism , Membrane Transport Proteins/metabolism , Protons , Purines/biosynthesis , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Receptors, Cell Surface/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , CHO Cells , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Folate Receptors, GPI-Anchored , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Gene Expression Regulation , Humans , Hydrogen Bonding , Nucleosides/pharmacology , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Pyrimidines/chemistry , Reduced Folate Carrier Protein , Substrate Specificity , Transfection
18.
J Med Chem ; 51(16): 5052-63, 2008 Aug 28.
Article in English | MEDLINE | ID: mdl-18680275

ABSTRACT

6-Substituted classical pyrrolo[2,3-d]pyrimidine antifolates with a three- to six-carbon bridge between the heterocycle and the benzoyl-L-glutamate (compounds 2-5, respectively) were synthesized starting from methyl 4-formylbenzoate and a Wittig reaction with the appropriate triphenylphosphonium bromide, followed by reduction and conversion to the alpha-bromomethylketones. Cyclocondensation of 2,4-diamino-4-oxopyrimidine with the alpha-bromoketones, coupling with diethyl-L-glutamate, and saponification afforded 2-5. Compounds 2-5 had negligible substrate activity for RFC but showed variably potent (nanomolar) and selective inhibitory activities toward Chinese hamster ovary cells that expressed FRalpha or FRbeta and toward FRalpha-expressing KB and IGROV1 human tumor cells. Inhibition of KB cell colony formation was also observed. Glycinamide ribonucleotide formyl transferase (GARFTase) was identified as the primary intracellular target of the pyrrolo[2,3-d]pyrimidines. The combined properties of selective FR targeting, lack of RFC transport, and GARFTase inhibition resulting in potent antitumor activity are unprecedented and warrant development of these analogues as antitumor agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Folic Acid Antagonists/chemical synthesis , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Pyrimidines/chemical synthesis , Pyrroles/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , CHO Cells , Carrier Proteins/antagonists & inhibitors , Cell Line , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Folate Receptors, GPI-Anchored , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Humans , KB Cells , Membrane Transport Proteins/drug effects , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Reduced Folate Carrier Protein
SELECTION OF CITATIONS
SEARCH DETAIL