Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 37: 116093, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33773393

ABSTRACT

We discovered 6-substituted thieno[2,3-d]pyrimidine compounds (3-9) with 3-4 bridge carbons and side-chain thiophene or furan rings for dual targeting one-carbon (C1) metabolism in folate receptor- (FR) expressing cancers. Synthesis involved nine steps starting from the bromo-aryl carboxylate. From patterns of growth inhibition toward Chinese hamster ovary cells expressing FRα or FRß, the proton-coupled folate transporter or reduced folate carrier, specificity for uptake by FRs was confirmed. Anti-proliferative activities were demonstrated toward FRα-expressing KB tumor cells and NCI-IGROV1 ovarian cancer cells. Inhibition of de novo purine biosynthesis at both 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and glycinamide ribonucleotide formyltransferase (GARFTase) was confirmed by metabolite rescue, metabolomics and enzyme assays. X-ray crystallographic structures were obtained with compounds 3-5 and human GARFTase. Our studies identify first-in-class C1 inhibitors with selective uptake by FRs and dual inhibition of enzyme targets in de novo purine biosynthesis, resulting in anti-tumor activity. This series affords an exciting new platform for selective multi-targeted anti-tumor agents.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Pyrimidines/pharmacology , Thiophenes/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetulus , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Folate Receptors, GPI-Anchored/metabolism , Humans , Molecular Docking Simulation , Molecular Structure , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Phosphoribosylglycinamide Formyltransferase/metabolism , Protein Binding , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/metabolism
2.
Cell Rep ; 21(5): 1331-1346, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-29091770

ABSTRACT

Mechanistic (or mammalian) target of rapamycin complex 1 (mTORC1) integrates signals from growth factors and nutrients to control biosynthetic processes, including protein, lipid, and nucleic acid synthesis. We find that the mTORC1 pathway is responsive to changes in purine nucleotides in a manner analogous to its sensing of amino acids. Depletion of cellular purines, but not pyrimidines, inhibits mTORC1, and restoration of intracellular adenine nucleotides via addition of exogenous purine nucleobases or nucleosides acutely reactivates mTORC1. Adenylate sensing by mTORC1 is dependent on the tuberous sclerosis complex (TSC) protein complex and its regulation of Rheb upstream of mTORC1, but independent of energy stress and AMP-activated protein kinase (AMPK). Even though mTORC1 signaling is not acutely sensitive to changes in intracellular guanylates, long-term depletion of guanylates decreases Rheb protein levels. Our findings suggest that nucleotide sensing, like amino acid sensing, enables mTORC1 to tightly coordinate nutrient availability with the synthesis of macromolecules, such as protein and nucleic acids, produced from those nutrients.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Purine Nucleotides/metabolism , A549 Cells , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/pharmacology , Fluorouracil/pharmacology , HeLa Cells , Humans , Mercaptopurine/pharmacology , Methotrexate/pharmacology , Mice , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/genetics , Phosphoribosylglycinamide Formyltransferase/metabolism , RNA Interference , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
Eur J Med Chem ; 139: 531-541, 2017 Oct 20.
Article in English | MEDLINE | ID: mdl-28830032

ABSTRACT

A novel series of 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines were designed and synthesized as potential antitumor agents targeting both thymidylate and purine nucleotide biosynthesis. Starting from the key intermediate 2-amino-4-oxo-pyrrolo[2,3-d]pyrimidin-6-yl-acetic acid, target compounds 1-6 were successfully obtained through two sequential condensation and saponification reactions in decent yield. The newly synthesized compounds showed antiproliferative potencies against a panel of tumor cell lines including KB, SW620 and MCF7. In particular, most compounds of this series exhibited nanomolar to subnanomolar inhibitory activities toward KB tumor cells, significantly more potent than the positive control methotrexate (MTX) and pemetrexed (PMX). Along with the results of nucleoside protection assays, molecular modeling studies suggested that the antitumor activity of compound 6 could be attributed to multitargeted inhibition of folate-dependent enzymes thymidylate synthase (TS), glycinamide ribonucleotide formyltransferase (GARFTase) and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase (AICARFTase). Growth inhibition by compound 6 also induced distinct early apoptosis and cell cycle arrest at S-phase, which resulted in cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Phosphoribosylglycinamide Formyltransferase/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship , Thymidylate Synthase/metabolism
4.
Biochemistry ; 55(32): 4574-82, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27439469

ABSTRACT

Pemetrexed and methotrexate are antifolates used for cancer chemotherapy and inflammatory diseases. These agents have toxic side effects resulting, in part, from nonspecific cellular transport by the reduced folate carrier (RFC), a ubiquitously expressed facilitative transporter. We previously described 2-amino-4-oxo-6-substituted pyrrolo[2,3-d]pyrimidine antifolates with modifications of the side chain linker and aromatic ring that are poor substrates for RFC but are efficiently transported via folate receptors (FRs) and the proton-coupled folate transporter (PCFT). These targeted antifolates are cytotoxic in vitro toward FR- and PCFT-expressing tumor cells and in vivo with human tumor xenografts in immune-compromised mice, reflecting selective cellular uptake. Antitumor efficacy is due to inhibition of glycinamide ribonucleotide (GAR) formyltransferase (GARFTase) activity in de novo synthesis of purine nucleotides. This study used purified human GARFTase (formyltransferase domain) to assess in vitro inhibition by eight novel thieno- and pyrrolo[2,3-d]pyrimidine antifolates. Seven analogues (AGF23, AGF71, AGF94, AGF117, AGF118, AGF145, and AGF147) inhibited GARFTase with Ki values in the low- to mid-nanomolar concentration range, whereas AGF50 inhibited GARFTase with micromolar potency similar to that of PMX. On the basis of crystal structures of ternary complexes with GARFTase, ß-GAR, and the monoglutamyl antifolates, differences in inhibitory potencies correlated well with antifolate binding and the positions of the terminal carboxylates. Our data provide a mechanistic basis for differences in inhibitory potencies between these novel antifolates and a framework for future structure-based drug design. These analogues could be more efficacious than clinically used antifolates, reflecting their selective cellular uptake by FRs and PCFT and potent GARFTase inhibition.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/metabolism , Animals , Humans , KB Cells , Mice , Models, Molecular , Phosphoribosylglycinamide Formyltransferase/chemistry , Protein Conformation , Xenograft Model Antitumor Assays
5.
J Med Chem ; 59(17): 7856-76, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27458733

ABSTRACT

Targeted antifolates with heteroatom replacements of the carbon vicinal to the phenyl ring in 1 by N (4), O (8), or S (9), or with N-substituted formyl (5), acetyl (6), or trifluoroacetyl (7) moieties, were synthesized and tested for selective cellular uptake by folate receptor (FR) α and ß or the proton-coupled folate transporter. Results show increased in vitro antiproliferative activity toward engineered Chinese hamster ovary cells expressing FRs by 4-9 over the CH2 analogue 1. Compounds 4-9 inhibited de novo purine biosynthesis and glycinamide ribonucleotide formyltransferase (GARFTase). X-ray crystal structures for 4 with FRα and GARFTase showed that the bound conformations of 4 required flexibility for attachment to both FRα and GARFTase. In mice bearing IGROV1 ovarian tumor xenografts, 4 was highly efficacious. Our results establish that heteroatom substitutions in the 3-atom bridge region of 6-substituted pyrrolo[2,3-d]pyrimidines related to 1 provide targeted antifolates that warrant further evaluation as anticancer agents.


Subject(s)
Antineoplastic Agents/chemistry , Folate Receptor 1/metabolism , Folic Acid Antagonists/chemistry , Proton-Coupled Folate Transporter/metabolism , Purine Nucleotides/antagonists & inhibitors , Pyrimidines/chemistry , Pyrroles/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Female , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/pharmacology , Heterografts , Humans , Mice, SCID , Molecular Docking Simulation , Neoplasm Transplantation , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Purine Nucleotides/biosynthesis , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Structure-Activity Relationship
6.
J Med Chem ; 58(17): 6938-59, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26317331

ABSTRACT

2-Amino-4-oxo-6-substituted-pyrrolo[2,3-d]pyrimidine antifolate thiophene regioisomers of AGF94 (4) with a thienoyl side chain and three-carbon bridge lengths [AGF150 (5) and AGF154 (7)] were synthesized as potential antitumor agents. These analogues inhibited proliferation of Chinese hamster ovary (CHO) sublines expressing folate receptors (FRs) α or ß (IC50s < 1 nM) or the proton-coupled folate transporter (PCFT) (IC50 < 7 nM). Compounds 5 and 7 inhibited KB, IGROV1, and SKOV3 human tumor cells at subnanomolar concentrations, reflecting both FRα and PCFT uptake. AGF152 (6) and AGF163 (8), 2,4-diamino-5-substituted-furo[2,3-d]pyrimidine thiophene regioisomers, also inhibited growth of FR-expressing CHO and KB cells. All four analogues inhibited glycinamide ribonucleotide formyltransferase (GARFTase). Crystal structures of human GARFTase complexed with 5 and 7 were reported. In severe combined immunodeficient mice bearing SKOV3 tumors, 7 was efficacious. The selectivity of these compounds for PCFT and for FRα and ß over the ubiquitously expressed reduced folate carrier is a paradigm for selective tumor targeting.


Subject(s)
Antineoplastic Agents/chemistry , Folate Receptor 1/antagonists & inhibitors , Folic Acid Antagonists/chemistry , Proton-Coupled Folate Transporter/antagonists & inhibitors , Pyrimidines/chemistry , Pyrroles/chemistry , Thiophenes/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Biological Transport , CHO Cells , Cell Line, Tumor , Cricetulus , Crystallography, X-Ray , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Folate Receptor 1/chemistry , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/pharmacology , Heterografts , Humans , Mice, SCID , Models, Molecular , Neoplasm Transplantation , Pemetrexed/pharmacology , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/chemistry , Proton-Coupled Folate Transporter/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Stereoisomerism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/pharmacology
7.
J Med Chem ; 58(3): 1479-93, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25602637

ABSTRACT

A new series of 5-substituted thiopheneyl pyrrolo[2,3-d]pyrimidines 6-11 with varying chain lengths (n = 1-6) were designed and synthesized as hybrids of the clinically used anticancer drug pemetrexed (PMX) and our 6-substituted thiopheneyl pyrrolo[2,3-d]pyrimidines 2c and 2d with folate receptor (FR) α and proton-coupled folate transporter (PCFT) uptake specificity over the reduced folate carrier (RFC) and inhibition of de novo purine nucleotide biosynthesis at glycinamide ribonucleotide formyltransferase (GARFTase). Compounds 6-11 inhibited KB human tumor cells in the order 9 = 10 > 8 > 7 > 6 = 11. Compounds 8-10 were variously transported by FRα, PCFT, and RFC and, unlike PMX, inhibited de novo purine nucleotide rather than thymidylate biosynthesis. The antiproliferative effects of 8 and 9 appeared to be due to their dual inhibitions of both GARFTase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase. Our studies identify a unique structure-activity relationship for transport and dual target inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , CHO Cells , Cell Proliferation/drug effects , Cells, Cultured , Cricetulus , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , KB Cells , Molecular Structure , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Phosphoribosylglycinamide Formyltransferase/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship
8.
Biochemistry ; 52(30): 5133-44, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23869564

ABSTRACT

Glycinamide ribonucleotide transformylase (GAR Tfase) is a folate-dependent enzyme in the de novo purine biosynthesis pathway, which has long been considered a potential target for development of anti-neoplastic therapeutics. Here we report the biological and X-ray crystallographic evaluations of both independent C10 diastereomers, 10S- and 10R-methylthio-DDACTHF, bound to human GAR Tfase, including the highest-resolution apo GAR Tfase structure to date (1.52 Å). Both diastereomers are potent inhibitors (Ki = 210 nM for 10R, and Ki = 180 nM for 10S) of GAR Tfase and exhibit effective inhibition of human leukemia cell growth (IC50 = 80 and 50 nM, respectively). Their inhibitory activity was surprisingly high, and these lipophilic C10-substituted analogues show distinct advantages over their hydrophilic counterparts, most strikingly in retaining potency in mutant human leukemia cell lines that lack reduced folate carrier protein activity (IC50 = 70 and 60 nM, respectively). Structural characterization reveals a new binding mode for these diastereoisomers, in which the lipophilic thiomethyl groups penetrate deeper into a hydrophobic pocket within the folate-binding site. In silico docking simulations of three other sulfur-containing folate analogues also indicate that this hydrophobic cleft represents a favorable region for binding lipophilic substituents. Overall, these results suggest sulfur and its substitutions play an important role in not only the binding of anti-folates to GAR Tfase but also the selectivity and cellular activity (growth inhibition), thereby presenting new possibilities for the future design of potent and selective anti-folate drugs that target GAR Tfase.


Subject(s)
Antimetabolites, Antineoplastic/chemistry , Carbon-Nitrogen Ligases/chemistry , Enzyme Inhibitors/chemistry , Models, Molecular , Phosphoribosylglycinamide Formyltransferase/chemistry , Tetrahydrofolates/chemistry , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacology , Apoproteins/antagonists & inhibitors , Apoproteins/chemistry , Apoproteins/metabolism , Binding Sites , Carbon-Nitrogen Ligases/antagonists & inhibitors , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Leukemia/drug therapy , Leukemia/enzymology , Molecular Conformation , Molecular Docking Simulation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/antagonists & inhibitors , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/chemistry , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/genetics , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/genetics , Phosphoribosylglycinamide Formyltransferase/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Stereoisomerism , Structure-Activity Relationship , Tetrahydrofolates/metabolism , Tetrahydrofolates/pharmacology
9.
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
10.
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
11.
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
12.
J Med Chem ; 51(17): 5441-8, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18686942

ABSTRACT

Glycinamide ribonucleotide transformylase (GAR Tfase) catalyzes the first of two formyl transfer steps in the de novo purine biosynthetic pathway that require folate cofactors and has emerged as a productive target for antineoplastic therapeutic intervention. The asymmetric synthesis and evaluation of the two diastereomers of 10-methylthio-DDACTHF (10R-3 and 10S-3) and related analogues as potential inhibitors of GAR Tfase are reported. This work, which defines the importance of the C10 stereochemistry for this class of inhibitors of GAR Tfase, revealed that both diastereomers are potent inhibitors of rhGAR Tfase (10R-3 Ki = 210 nM, 10S-3 Ki = 180 nM) that exhibit effective cell growth inhibition (CCRF-CEM IC50 = 80 and 50 nM, respectively), which is dependent on intracellular polyglutamation by folylpolyglutamate synthetase (FPGS) but not intracellular transport by the reduced folate carrier.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Inhibitory Concentration 50 , Peptide Synthases/metabolism , Stereoisomerism , Structure-Activity Relationship
13.
Cancer Chemother Pharmacol ; 62(2): 215-26, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17896107

ABSTRACT

PURPOSE: 4-[2-(2-Amino-4-oxo-4,6,7,8-tetrahydro-3H-pyrimidino[5,4,6][1,4] thiazin-6-yl)-(S)-ethyl]-2,5-thienoylamino-L: -glutamic acid (AG2034), is a classical antifolate, an analog of folic acid that has been shown to be an excellent inhibitor of glycinamide ribonucleotide formyltransferase (GARFT), ultimately inhibiting the de novo synthesis of purines. We examined the effect of this drug on cell proliferation, steady-state ATP levels, de novo and hypoxanthine salvage ATP synthesis, and on the phosphorylation of AMP kinase, in two different androgen independent prostate cancer cell lines, DU145 and PC-3. METHODS: Cells were maintained in culture medium containing 10 nM 5-methyl tetrahydrofolate supplemented with or without 1.7 microM hypoxanthine and 1.5 microM thymidine. Cytotoxicity of AG2034 was determined by clonogenic assays. AG2034-induced inhibition of cell proliferation was determined by electronic counting of cells over varying periods of time. Total cellular AMP and ATP pre- and post-drug treatment was quantified by reverse-phase HPLC. [(14)C]-Glycine incorporation and [(3)H]-hypoxanthine conversion into ATP were determined by liquid scintillation counting of HPLC isolated ATP fractions. The phosphorylation of AMP kinase (AMPK) was detected by western blotting. RESULTS: In the absence of 1.7 muM hypoxanthine, AG2034 was cytotoxic to both DU145 and PC-3 cells. In its presence, the cells remained cytostatic for 14 days after which time DU145 but not PC-3 re-initiated growth that was maintained for 35 days even though steady-state levels of ATP in both cell lines remained depleted and [(14)C]-glycine incorporation into ATP was inhibited by >95%. Salvage purine synthesis as measured by incorporation of [(3)H]-hypoxanthine into ATP was maintained in both cell lines albeit to different levels. When AG2034 was added to the culture medium in the presence or absence of 1.7 microM hypoxanthine, cellular ATP levels were reduced by 80% within 24 h in both the cell lines. In the absence of hypoxanthine, the AMP/ATP ratio in PC-3 cells increased by 38% and was accompanied by a modest increase in the level of phosphorylated AMPK; no increase was observed in the presence of hypoxanthine where the AMP/ATP ratio increased by approximately 10%. Under these same culture conditions, the AMP/ATP ratio in DU145 cells in the absence of hypoxanthine increased by 60% and was accompanied by a large increase in phosphorylated AMPK. In the presence of hypoxanthine however, even though the AMP/ATP ratio increased 2.5-fold, phosphorylated AMPK levels did not increase. CONCLUSIONS: The cytostatic versus the cytotoxic effect of AG2034 on PC-3 and DU145 cells is mediated by the presence or absence, respectively, of physiological levels of hypoxanthine (1.7 muM) in the media. The ability of DU145 as opposed to PC-3 cells to proliferate in the presence of AG2034 is independent of the intracellular concentration of ATP. Activation of the AMPK signaling pathway in drug-treated PC-3 and DU145 cells is cell line dependent and independent of the AMP/ATP ratio.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Glutamates/pharmacology , Hypoxanthine/metabolism , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Prostatic Neoplasms , Pyrimidines/pharmacology , Adenosine Triphosphate/biosynthesis , Adenylate Kinase/metabolism , Blotting, Western , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
14.
Mol Cancer Ther ; 5(10): 2549-55, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17041099

ABSTRACT

Pemetrexed is a new-generation antifolate inhibitor of thymidylate synthase (TS) and a weaker inhibitor of glycinamide ribonucleotide transformylase (GARFT) required for de novo purine synthesis. Methylthioadenosine phosphorylase (MTAP) salvages purines by releasing adenine from methylthioadenosine and is often deleted in mesothelioma. The current study addresses the effect of MTAP on pemetrexed activity using a highly potent transition state inhibitor of MTAP, MT-DADMe-Immucillin A (ImmA; K(i) = 86 pmol/L) in the MTAP(+) NCI-H28 and MTAP(-) NCI-H2052 mesothelioma cell lines. Based on selective nucleoside protection, TS was found to be the primary pemetrexed target in both cell lines with GARFT inhibition requiring 20- to 30-fold higher pemetrexed concentrations. ImmA had no effect on pemetrexed activity but, when thymidine was added, the pemetrexed IC(50) decreased by a factor of approximately 3 in MTAP(+) H28 cells with no effect in MTAP(-) H2052 cells. Conversely, the transfection of MTAP into H2052 cells increased the pemetrexed IC(50) by nearly 3-fold but only in the presence of thymidine; this was reversed by ImmA. An MTAP-specific short interfering RNA produced a 2-fold decrease in pemetrexed IC(50) in MTAP(+) HeLa cells in the presence of thymidine. These data indicate that suppression of constitutive MTAP has no effect on pemetrexed activity when the primary target is TS. There is a modest salutary effect when the pemetrexed target is GARFT alone.


Subject(s)
Glutamates/pharmacology , Guanine/analogs & derivatives , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Cell Line, Tumor , Guanine/pharmacology , Humans , Pemetrexed , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/biosynthesis , Purine-Nucleoside Phosphorylase/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Stem Cell Assay
15.
Mol Cancer Ther ; 5(9): 2211-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16985054

ABSTRACT

Because of its ability to mimic a low energy status of the cell, the cell-permeable nucleoside 5-aminoimidazole-4-carboxamide (AICA) riboside was proposed as an antineoplastic agent switching off major energy-consuming processes associated with the malignant phenotype (lipid production, DNA synthesis, cell proliferation, cell migration, etc.). Key to the antineoplastic action of AICA riboside is its conversion to ZMP, an AMP mimetic that at high concentrations activates the AMP-activated protein kinase (AMPK). Here, in an attempt to increase the efficacy of AICA riboside, we pretreated cancer cells with methotrexate, an antimetabolite blocking the metabolism of ZMP. Methotrexate enhanced the AICA riboside-induced accumulation of ZMP and led to a decrease in the levels of ATP, which functions as an intrasteric inhibitor of AMPK. Consequently, methotrexate markedly sensitized AMPK for activation by AICA riboside and potentiated the inhibitory effects of AICA riboside on tumor-associated processes. As cotreatment elicited antiproliferative effects already at concentrations of compounds that were only marginally effective when used alone, our findings on the cooperation between methotrexate and AICA riboside provide new opportunities both for the application of classic antimetabolic chemotherapeutics, such as methotrexate, and for the exploitation of the energy-sensing machinery as a target for cancer intervention.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Breast Neoplasms/drug therapy , Carcinoma, Squamous Cell/drug therapy , Methotrexate/pharmacology , Ribonucleosides/pharmacology , AMP-Activated Protein Kinases , Adenosine Triphosphate/metabolism , Aminoimidazole Carboxamide/antagonists & inhibitors , Aminoimidazole Carboxamide/metabolism , Aminoimidazole Carboxamide/pharmacokinetics , Aminoimidazole Carboxamide/pharmacology , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , DNA, Neoplasm/antagonists & inhibitors , DNA, Neoplasm/biosynthesis , Drug Synergism , Enzyme Activation/drug effects , Humans , Lipids/biosynthesis , Multienzyme Complexes/metabolism , Nucleotide Deaminases/antagonists & inhibitors , Nucleotide Deaminases/genetics , Nucleotide Deaminases/metabolism , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/antagonists & inhibitors , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/genetics , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/genetics , Phosphoribosylglycinamide Formyltransferase/metabolism , Protein Serine-Threonine Kinases/metabolism , Purines/antagonists & inhibitors , Purines/biosynthesis , RNA Interference , Ribonucleosides/pharmacokinetics , Ribonucleotides/antagonists & inhibitors , Ribonucleotides/metabolism
16.
Br J Cancer ; 95(3): 289-97, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16868547

ABSTRACT

The present study was performed to investigate the capability of gemcitabine and pemetrexed to synergistically interact with respect to cytotoxicity and apoptosis in T24 and J82 bladder cancer cells, and to establish a correlation between drug activity and gene expression of selected genes in tumour samples. The interaction between gemcitabine and pemetrexed was synergistic; indeed, pemetrexed favoured gemcitabine cytotoxicity by increasing cellular population in S-phase, reducing Akt phosphorylation as well as by inducing the expression of a major gemcitabine uptake system, the human equilibrative nucleoside transporter-1 (hENT1), and the key activating enzyme deoxycytidine kinase (dCK) in both cell lines. Bladder tumour specimens showed an heterogeneous gene expression pattern and patients with higher levels of dCK and hENT1 had better response. Moreover, human nucleoside concentrative transporter-1 was detectable only in 3/12 patients, two of whom presented a complete response to gemcitabine. These data provide evidence that the chemotherapeutic activity of the combination of gemcitabine and pemetrexed is synergistic against bladder cancer cells in vitro and that the assessment of the expression of genes involved in gemcitabine uptake and activation might be a possible determinant of bladder cancer response and may represent a new tool for treatment optimization.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Glutamates/administration & dosage , Guanine/analogs & derivatives , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Administration, Intravesical , Adult , Aged , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/antagonists & inhibitors , Deoxycytidine/pharmacology , Deoxycytidine Kinase/antagonists & inhibitors , Deoxycytidine Kinase/genetics , Dipyridamole/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative Nucleoside Transporter 1/genetics , Excitatory Amino Acid Antagonists , Female , Gene Expression Regulation, Neoplastic/drug effects , Glutamates/pharmacology , Guanine/administration & dosage , Guanine/antagonists & inhibitors , Guanine/pharmacology , Humans , Male , Middle Aged , Neoplasm Staging , Pemetrexed , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , S Phase/drug effects , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/genetics , Treatment Outcome , Gemcitabine
17.
J Med Chem ; 49(10): 2998-3002, 2006 May 18.
Article in English | MEDLINE | ID: mdl-16686541

ABSTRACT

Glycinamide ribonucleotide transformylase (GAR Tfase) catalyzes the first of two formyl transfer steps in the de novo purine biosynthetic pathway that require folate cofactors. Herein we report the discovery of a potent, nonpolyglutamatable, and selective inhibitor of GAR Tfase. Compound 12, which possesses a tetrazole in place of the gamma-carboxylic acid in the l-glutamate subunit of the potent GAR Tfase inhibitor 1, was active in cellular-based functional assays exhibiting purine-sensitive cytotoxic activity (IC(50) = 40 nM, CCRF-CEM) and was selective for inhibition of rhGAR Tfase (K(i) = 130 nM). Notably, 12 was only 2.5-fold less potent than 1 in cellular assays and 4-fold less potent against rhGAR Tfase. Like 1, this functional activity of 12 in the cell-based assay benefits from and requires transport into the cell by the reduced folate carrier but, unlike 1, is independent of folyl polyglutamate synthase (FPGS) expression levels and polyglutamation.


Subject(s)
Aminobutyrates/chemical synthesis , Antineoplastic Agents/chemical synthesis , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Pyrimidinones/chemical synthesis , Tetrazoles/chemical synthesis , Aminobutyrates/chemistry , Aminobutyrates/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Peptide Synthases/metabolism , Phosphoribosylglycinamide Formyltransferase/chemistry , Polyglutamic Acid/metabolism , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Structure-Activity Relationship , Substrate Specificity , Tetrahydrofolates/chemistry , Tetrazoles/chemistry , Tetrazoles/pharmacology
18.
Org Lett ; 8(8): 1653-5, 2006 Apr 13.
Article in English | MEDLINE | ID: mdl-16597133

ABSTRACT

[structure: see text] An efficient chemoenzymatic process is described for the synthesis of pelitrexol, a novel GARFT inhibitor. The remoteness of this molecule's stereocenter in the tetrahydropterin moiety from the terminal carbonyl group provided a significant challenge in synthesis. The introduction of an oxalamic ester adjacent to the stereocenter dramatically enhanced an enzyme's enantioselectivity for hydrolysis at the terminal ester, producing the desired S-acid with high optical purity and yield. The recycling of the "wrong" enantiomer is achieved via a dehydrogenation/hydrogenation strategy.


Subject(s)
Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Thiophenes/chemical synthesis , Catalysis , Fungal Proteins , Lipase/metabolism , Molecular Structure , Stereoisomerism
19.
Cancer Res ; 66(7): 3836-44, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16585211

ABSTRACT

Transport is required before reduced folates and anticancer antifolates [e.g., methotrexate (MTX)] exert their physiologic functions or cytotoxic effects. The folate/antifolate transporter with the widest tissue distribution and greatest activity is the reduced folate carrier (RFC). There is little evidence that RFC-mediated influx is posttranscriptionally regulated. We show that [(3)H]MTX influx in CCRF-CEM human childhood T-leukemia cells is potentiated up to 6-fold by exogenous 5-amino-4-imidazolecarboxamide riboside (AICAr) in a AICAr and MTX concentration-dependent manner. Metabolism to more biologically active polyglutamate forms is also potentiated for MTX and other antifolates. That potentiation of influx by AICAr is mediated by effects on the RFC is supported by analyses +/-AICAr showing (a) similarity and magnitude of kinetic constants for [(3)H]MTX influx; (b) similarity of inhibitory potency of known RFC substrates; (c) lack of potentiation in a CCRF-CEM subline that does not express the RFC; and (d) similarity of time and temperature dependence. Potentiation occurs rapidly and does not require new protein synthesis. Effects of specific inhibitors of folate metabolism and the time and sequence of AICAr incubation with cells suggest that both dihydrofolate reductase inhibition and metabolism of AICAr are essential for potentiation. Acute folate deficiency or incubation of CCRF-CEM with AICAr-related metabolites (e.g., adenosine) does not initiate potentiation. AICAr increases growth inhibitory potency of MTX and aminopterin against CCRF-CEM cells when both AICAr and antifolate are present for the first 24 hours of a 120-hour growth period. AICAr is the first small molecule that regulates RFC activity.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Leukemia, T-Cell/metabolism , Membrane Transport Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Ribonucleotides/pharmacology , Aminoimidazole Carboxamide/pharmacology , Biological Transport/drug effects , Cell Line, Tumor , Child , Dose-Response Relationship, Drug , Drug Synergism , Folic Acid Antagonists/metabolism , Folic Acid Antagonists/pharmacokinetics , Humans , Kinetics , Leukemia, T-Cell/drug therapy , Methotrexate/analogs & derivatives , Methotrexate/metabolism , Methotrexate/pharmacokinetics , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/metabolism , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/biosynthesis , Polyglutamic Acid/pharmacokinetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Reduced Folate Carrier Protein , Subcellular Fractions , Temperature , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/metabolism , Tritium
20.
Mol Cancer Ther ; 5(2): 438-49, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16505119

ABSTRACT

Pemetrexed, a new generation antifolate recently approved for the treatment of mesothelioma and non-small cell lung cancer, is an excellent substrate for the reduced folate carrier (RFC). To explore the carrier's effect on pemetrexed activity, RFC was inactivated in HCT-15 colon cancer cells by mutagenesis and PT632 selective pressure. A clone (PT1) was obtained with a glycine to arginine substitution at amino acid 401, resulting in the loss of RFC function. PT1 cells were resistant to PT632 (178-fold), methotrexate (4-fold), and ZD1694 (Tomudex, raltitrexed; 20-fold), but were 3-fold collaterally sensitive to pemetrexed when grown in 25 nmol/L of 5-formyltetrahydrofolate. PT1 cells transfected with wild-type RFC had antifolate sensitivities comparable to that of wild-type HCT-15 cells, indicating that the RFC mutation was the sole basis for resistance. Folate pools were contracted in PT1 cells by 32% or 60%, as measured by radiolabeling intracellular folates or by an enzyme binding assay, respectively. This was reflected in marked (6.5-fold) collateral sensitivity to trimetrexate. The initial uptake of pemetrexed in PT1 cells was markedly reduced ( approximately 85%) but intracellular pemetrexed levels increased to approximately 60% and approximately 70% to that of wild-type cells after 2 hours and 6 days, respectively. There was increased pemetrexed inhibition of glycinamide ribonucleotide transformylase and, to a lesser extent, thymidylate synthase in PT1 cells growing in 5-formyltetrahydrofolate based on nucleoside protection analyses. Hence, loss of RFC function leads to collateral sensitivity to pemetrexed in HCT-15 cells, likely due to cellular folate pool contraction resulting in partial preservation of pemetrexed polyglutamylation and increased target enzyme inhibition. micro


Subject(s)
Antineoplastic Agents/metabolism , Carrier Proteins/metabolism , Colonic Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Glutamates/metabolism , Guanine/analogs & derivatives , Receptors, Cell Surface/metabolism , Amino Acid Substitution , Arginine/chemistry , Arginine/genetics , Carrier Proteins/genetics , Cell Line, Tumor , Colonic Neoplasms/genetics , Folate Receptors, GPI-Anchored , Folic Acid Antagonists/pharmacology , Glycine/chemistry , Glycine/genetics , Guanine/agonists , Guanine/metabolism , Humans , Leucovorin/metabolism , Methotrexate/metabolism , Mutation , Pemetrexed , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Purines/pharmacology , Pyrimidines/pharmacology , Quinazolines/metabolism , Receptors, Cell Surface/genetics , Thiophenes/metabolism , Thymidylate Synthase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...