Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...