Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters










Publication year range
1.
J Clin Oncol ; 19(11): 2804-11, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11387351

ABSTRACT

PURPOSE: To develop more effective chemotherapy regimens for childhood acute myelogenous leukemia (AML). PATIENTS AND METHODS: Between June 1991 and December 1996, we administered the nucleoside analog 2-chlorodeoxyadenosine (2-CDA) to 73 children with primary AML and 20 children with secondary AML or myelodysplastic syndrome (MDS). Patients received one or two 5-day courses of 2-CDA (8.9 mg/m(2)/d) given by continuous infusion. All patients then received one to three courses of daunomycin, cytarabine, and etoposide (DAV) remission induction therapy. RESULTS: Seventy-two patients with primary AML were assessable for response. Their rate of complete remission (CR) was 24% after one course of 2-CDA, 40% after two courses of 2-CDA, and 78% after DAV therapy. Of the 57 patients who entered CR, 11 subsequently underwent allogeneic bone marrow transplantation (BMT), and 40 underwent autologous BMT. Twenty-nine patients remain in continuous CR after BMT. Two patients remain in CR after chemotherapy only. The 5-year event-free survival (EFS) estimate was 40% (SE = 0.080%). Patients with French-American-British (FAB) M5 AML had a higher rate of CR after treatment with 2-CDA (45% after one course and 70.6% after two courses) than did others (P =.002). In contrast, no patient with FAB M7 AML (n = 10) entered CR after treatment with 2-CDA. Similarly, no patient with primary MDS (n = 6) responded to 2-CDA. Seven patients with secondary AML or MDS (n = 14) had a partial response to one course of 2-CDA. CONCLUSION: This agent was well tolerated, and its toxicity was acceptable. Future trials should examine the effectiveness of 2-CDA given in combination with other agents effective against AML.


Subject(s)
Antineoplastic Agents/pharmacology , Cladribine/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Adolescent , Antineoplastic Agents/administration & dosage , Bone Marrow Transplantation , Child , Child, Preschool , Cladribine/administration & dosage , Disease-Free Survival , Female , Humans , Infant , Infant, Newborn , Infusions, Intravenous , Male , Treatment Outcome
2.
Food Chem Toxicol ; 39(4): 401-6, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11295487

ABSTRACT

Evidence suggests that when compared with non-smokers, cigarette smokers are exposed to an increased burden of free radicals from both the vapor phase and particulate phase of the cigarette smoke aerosol. In this study, primary emphasis was placed on the free radicals found in the particulate phase. Published reports hypothesize that the particulate phase free radicals of cigarette mainstream smoke (MS) condensate consist of a hydroquinone/semiquinone/quinone shuttle. However, our results do not suggest that there is a positive correlation between the smoke yield of hydroquinone and the presence of particulate phase free radicals. First, 10-fold reductions in MS hydroquinone yield were obtained when KNO3 was applied to the surface of tobacco of an American blended cigarette. Surprisingly, there was no significant corresponding change in the yield of particulate phase radicals. Second, in experiments testing MS from low and high hydroquinone-yielding tobaccos there was no consistent corresponding relationship between hydroquinone and particulate phase radical yields. In one series of blends there was at best an inverse relationship between hydroquinone and particulate phase radical yields. In contrast with the published literature, we conclude that the particular compound or compounds driving particulate phase free radical formation are currently unknown. An additional experiment reported here suggested that components of the water soluble extract of burley tobacco may be driving the formation of particulate phase free radicals.


Subject(s)
Antioxidants/metabolism , Hydroquinones/metabolism , Nicotiana , Plants, Toxic , Smoke , Electron Spin Resonance Spectroscopy , Free Radicals , Nitrates/administration & dosage , Potassium Compounds/administration & dosage , Smoking
3.
Biochem Pharmacol ; 60(3): 325-38, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10856427

ABSTRACT

Tumors resistant to chemotherapeutic oxazaphosphorines such as cyclophosphamide often overexpress aldehyde dehydrogenase (ALDH), some isozymes of which catalyze the oxidization of aldophosphamide, an intermediate of cyclophosphamide activation, with formation of inert carboxyphosphamide. Since resistance to oxazaphosphorines can be produced in mammalian cells by transfecting them with the gene for human ALDH isozyme 3 (hALDH3), it seems possible that patients receiving therapy for solid tumors with cyclophosphamide might be protected from myelosuppression by their prior transplantation with autologous bone marrow that has been transduced with a retroviral vector causing overexpression of hALDH3. We investigated whether retroviral introduction of hALDH3 into a human leukemia cell line confers resistance to oxazaphosphorines. This was examined in the polyclonal transduced population, that is, without selecting out high expression clones. hALDH3 activity was 0.016 IU/mg protein in the transduced cells (compared with 2x10(-5) IU/mg in untransduced cells), but there was no detectable resistance to aldophosphamide-generating compounds (mafosfamide or 4-hydroperoxycyclophosphamide). The lack of protection was due, in part, to low catalytic activity of hALDH3 towards aldophosphamide, since, with NAD as cofactor, the catalytic efficiency of homogeneous, recombinant hALDH3 for aldophosphamide oxidation was shown to be about seven times lower than that of recombinant hALDH1. The two polymorphic forms of hALDH3 had identical kinetics with either benzaldehyde or aldophosphamide as substrate. Results of initial velocity measurements were consistent with an ordered sequential mechanism for ALDH1 but not for hALDH3; a kinetic mechanism for the latter is proposed, and the corresponding rate equation is presented.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Antineoplastic Agents/metabolism , Phosphoramide Mustards/metabolism , Aldehyde Dehydrogenase/chemistry , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Aldehydes/metabolism , Antineoplastic Agents/pharmacology , Benzaldehydes/metabolism , Cloning, Molecular , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/pharmacology , DNA, Complementary/analysis , Enzyme Inhibitors/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Oxidation-Reduction , Phosphoramide Mustards/pharmacology , Recombinant Proteins/chemistry , Retinal Dehydrogenase , Sequence Analysis , Substrate Specificity , Transfection , Tumor Cells, Cultured
5.
Nat Med ; 4(10): 1136-43, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9771746

ABSTRACT

One of the main impediments to effective gene therapy of blood disorders is the resistance of human hematopoietic stem cells to stable genetic modification. We show here that a small minority of retrovirally transduced stem cells can be selectively enriched in vivo, which might be a way to circumvent this obstacle. We constructed two retroviral vectors containing an antifolate-resistant dihydrofolate reductase cDNA transcriptionally linked to a reporter gene. Mice were transplanted with transduced bone marrow cells and then treated with an antifolate-based regimen that kills unmodified stem cells. Drug treatment significantly increased the percentage of vector-expressing peripheral blood erythrocytes, platelets, granulocytes, and T and B lymphocytes. Secondary transplant experiments demonstrated that selection occurred at the level of hematopoietic stem cells. This system for in vivo stem-cell selection provides a means to increase the number of genetically modified cells after transplant, and may circumvent an substantial obstacle to successful gene therapy for human blood diseases.


Subject(s)
Genetic Vectors , Hematopoietic Stem Cell Transplantation , Retroviridae/genetics , Selection, Genetic , Tetrahydrofolate Dehydrogenase/genetics , Animals , Blood Cells/enzymology , Drug Resistance , Female , Folic Acid Antagonists/pharmacology , Genes, Reporter , Genetic Therapy/methods , Mice , Mice, Inbred C57BL , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Thionucleotides/pharmacology , Transformation, Genetic , Trimetrexate/pharmacology
6.
Cancer Gene Ther ; 5(4): 225-35, 1998.
Article in English | MEDLINE | ID: mdl-9694074

ABSTRACT

Expression of certain variants of dihydrofolate reductase (DHFR) in mammalian cells protects them from methotrexate. Retroviral transfer of the gene for such a variant DHFR into hematopoietic cells might permit selection of modified cells in vivo by antifolate administration or alleviate antifolate-induced myelosuppression in patients receiving antifolate therapy. We examined protection of cells of the human lymphoblastoid line, CCRF-CEM, transduced with variants of mouse DHFR. In transduced cells selected with G418 but not with antifolate, the variant that had arginine substituted for leucine 22 did not protect against either methotrexate or trimetrexate; however, four other variants did offer protection, with the best having leucine 22 changed to tyrosine. Polyclonal cultures transduced with the different variants express DHFR at about the same level, but clones within each polyclonal population differ in DHFR expression levels and in resistance. These differences in expression were shown to reflect different integration sites for proviral DNA. Exposure to trimetrexate selects highly resistant clones, with high expression due to both high copy number and integration sites that are favorable for expression. Differences in the resistance of cultures expressing different variants at the same level are due to differences in the catalytic activity of the expressed DHFR, its affinity for antifolates, and its stability.


Subject(s)
Folic Acid Antagonists/pharmacology , Gene Transfer Techniques , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Tetrahydrofolate Dehydrogenase/genetics , Animals , Cloning, Molecular , Drug Resistance, Neoplasm , Enzyme Stability/genetics , Escherichia coli/genetics , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Methotrexate/pharmacology , Mice , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retroviridae/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Trimetrexate/pharmacology , Tumor Cells, Cultured
7.
Anticancer Drug Des ; 13(4): 307-15, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9627670

ABSTRACT

The novel furopyrimidine, N-[4-[(2,4-diaminofuro[2,3-d]pyrimidin-5-yl)-methyl]-methylamino] -benzoyl]-L-glutamate (MTXO), a classical antifolate with weak antitumor activity compared with methotrexate (MTX), has been studied as inhibitorcofactor ternary crystal complexes with recombinant Phe-31 to Ser (F31S) and Phe-31 to Gly (F31G) variant human dihydrofolate reductase (hDHFR). Kinetic data show that the binding affinity of MTXO is significantly weaker for the variant hDHFR enzyme than for the wild type enzyme. Structural data for the Phe-31 variants, along with wild type hDHFR, provide the first direct comparison of the binding interactions of a single antifolate in a family of variant hDHFR. These ternary hDHFR complexes crystallize in the rhombohedral space group R3, isomorphous to that reported for wild type hDHFR MTXO-NADPH ternary complex. MTXO binds with its 2,4-diaminofuropyrimidine ring interacting with Glu-30 in hDHFR. The greatest change on modification of the side chain at position 31 is loss of hydrophobic contacts to the inhibitor, which results in the significant decrease in binding affinity of MTXO for the Phe-31 variants. The presence of the 6-5 furopyrimidine ring instead of the 6-6 pteridine ring causes a different bridge conformation compared with MTX, and in the case of the wild type MTXO complex also results in weaker hydrophobic contacts to Phe-31 than observed for MTXT. For the design of antitumor agents related to MTXO, increasing the bridge of MTXO from two to three or four atoms should provide increased DHFR inhibitory potency and antitumor activity.


Subject(s)
Folic Acid Antagonists/chemistry , Folic Acid/analogs & derivatives , NADP/chemistry , Tetrahydrofolate Dehydrogenase/chemistry , Antimetabolites, Antineoplastic/chemistry , Binding Sites , Crystallography, X-Ray , Folic Acid/chemistry , Humans , Methotrexate/chemistry , Mutation , Tetrahydrofolate Dehydrogenase/genetics
8.
Hum Mutat ; 11(4): 259-63, 1998.
Article in English | MEDLINE | ID: mdl-9554740

ABSTRACT

Mammalian cells cultured in the presence of the chemotherapeutic agent, methotrexate, develop resistance to this drug. Sometimes this is due to mutations in the gene for dihydrofolate reductase, the primary target of methotrexate. However, it has not been possible to link such polymorphism to resistance of neoplastic disease to therapy with methotrexate. Nevertheless, interest in this possibility lead to the introduction of many mutations into the cDNA for human DHFR by mutagenesis. Most of the corresponding enzyme variants have been expressed in Escherichia coli and characterized. Many mutations in codons for hydrophobic residues at the active site greatly decrease inhibition by methotrexate, and by the related substrate analogue, trimetrexate, while allowing the retention of considerable catalytic efficiency. Introduction of some of these mutants into mammalian cells by retroviral transfer provides substantial protection from toxic effects of the inhibitors, and has promise for the myeloprotection of patients receiving therapy with methotrexate or trimetrexate. Another potential use is in therapy for inherited disorders of hematopoiesis, where genetic modification of enough cells is a perennial problem. After transplantation of bone marrow that has been transduced with a bicistronic vector encoding both the mutant DHFR and a therapeutic gene, subsequent administration of methotrexate or trimetrexate should permit selection and enrichment of genetically modified hematopoietic cells.


Subject(s)
Methotrexate/pharmacology , Mutation , Tetrahydrofolate Dehydrogenase/genetics , Bone Marrow Transplantation , Cell Line , Drug Resistance/genetics , Escherichia coli/genetics , Genetic Therapy , Genetic Variation , Hematopoiesis/drug effects , Hematopoiesis/genetics , Humans , Mutagenesis, Site-Directed
9.
Stem Cells ; 16 Suppl 1: 223-33, 1998.
Article in English | MEDLINE | ID: mdl-11012166

ABSTRACT

Transfer of drug resistance genes to hematopoietic cells is being studied as a means to protect against the myelosuppression associated with cancer chemotherapy and as a strategy for the in vivo selection and amplification of genetically modified cells. The goal of this study was to test if retroviral-mediated gene transfer of a dihydrofolate reductase (DHFR) variant (L22Y) could be used for in vivo selection of transduced myeloid cells and to determine what proportion of transduced cells was required for protection from myelosuppression. Based on previous work suggesting that selection with antifolates may also require inhibition of nucleoside transport mechanisms, mice transplanted with DHFR-transduced bone marrow cells were treated with trimetrexate and the nucleoside transport inhibitor prodrug nitrobenzylmercaptopurine riboside phosphate. In vivo selection of transduced myeloid progenitors was seen in the bone marrow and in circulating mature peripheral blood cells following drug treatment. These results show that the novel combination of the L22Y-DHFR cDNA, trimetrexate and nitrobenzylmercaptopurine riboside phosphate can be used to select for transduced myeloid cells, and that this approach warrants further study in large animal models. A bicistronic vector containing a human CD24 reporter gene was used to determine the number of modified cells needed for chemoprotection. Partial protection from neutropenia was seen when greater than 10% of myeloid cells expressed the vector, and high levels of protection were obtained when the proportion exceeded 30%. These results suggest that gene transfer may be useful for myeloprotection in certain pediatric cancers, but that more efficient gene transfer will be required to apply this approach to adult cancer patients.


Subject(s)
Genetic Therapy/methods , Genetic Variation , Genetic Vectors , Hematopoietic Stem Cells/cytology , Membrane Glycoproteins , Tetrahydrofolate Dehydrogenase/genetics , Transfection/methods , Adult , Amino Acid Substitution , Animals , Antigens, CD/genetics , Bone Marrow Cells/cytology , CD24 Antigen , Encephalomyocarditis virus , Female , Gene Transfer Techniques , Genes, Reporter , Harvey murine sarcoma virus , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Humans , Mice , Recombinant Proteins/metabolism , Tetrahydrofolate Dehydrogenase/metabolism , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Thionucleotides/pharmacology , Trimetrexate/pharmacology
10.
Biochemistry ; 36(45): 13897-903, 1997 Nov 11.
Article in English | MEDLINE | ID: mdl-9374868

ABSTRACT

Structural data for two independent crystal forms (monoclinic, C2, and orthorhombic, P2(1)2(1)2(1)) of the ternary complex of the potent antitumor agent PT523 [N alpha-(4-amino-4-deoxypteroyl)-N delta-hemiphthaloyl-L-ornithine], reduced nicotinamide adenine dinucleotide phosphate (NADPH), and recombinant human dihydrofolate reductase (hDHFR) reveals multiple binding orientations for the hemiphthaloyl group of the inhibitor. Analysis of these data shows that PT523 binds with its pteridine ring in the same orientation observed for methotrexate (MTX) analogues. However, in each structure, the hemiphthaloyl ring occupies three alternate conformations. In the C2 lattice, the phthaloyl moiety binds in two extended conformations, A and C, with each conformer having a 180 degrees flip of the o-carboxylate group, and a third, lower occupancy conformer B, with the phthaloyl group folded within contact of the active-site pocket. In the orthorhombic lattice, PT523 also has three conformers for the phthaloyl group; however, these differ from those observed in the monoclinic lattice. Two major conformers, A and C, are displaced on either side of the extended position observed in the C2 lattice, one near the folded B conformer of the C2 lattice and the other extended. These conformers form tighter intermolecular contacts than those in the C2 lattice. Conformer B is folded back away from the active site in a unique position. There are also significant differences in the conformation of the adenine-ribose moiety of NADPH in both complexes that differ from that observed for other inhibitor-NADPH-hDHFR ternary complexes. These data suggest that the added intermolecular contacts made by the hemiphaloyl group of PT523 contribute to its tighter binding to hDHFR than MTX, which does not extend as far from the active site and cannot make these contacts. These crystallographic observations of multiple conformations for the hemiphthaloyl group are in general agreement with solution NMR data for the binding of PT523 to hDHFR [Johnson et al. (1997) Biochemistry 36, 4399-4411], which show that the hemiphthaloyl group may adopt more than one conformation. However, the crystallographic data reveal more discretely occupied positions than can be interpreted from the solution data. These results suggest that crystal packing interactions may influence their stability.


Subject(s)
Folic Acid Antagonists/metabolism , NADP/metabolism , Ornithine/analogs & derivatives , Pterins/metabolism , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , Binding, Competitive , Crystallization , Crystallography, X-Ray , Folic Acid Antagonists/chemistry , Humans , NADP/chemistry , Ornithine/chemistry , Ornithine/metabolism , Protein Binding , Pterins/chemistry , Structure-Activity Relationship
11.
Hum Gene Ther ; 8(17): 2069-77, 1997 Nov 20.
Article in English | MEDLINE | ID: mdl-9414255

ABSTRACT

Retroviral transduction of antifolate-resistant variants of human dihydrofolate reductase (hDHFR) into cells can increase their resistance to the cytotoxic effects of these drugs. We evaluated the ability of wild-type hDHFR and 20 mutant enzymes (13 with single-amino acid substitutions, 7 with two substitutions) to prevent growth inhibition in antifolate-treated CCRF-CEM cells. The wild-type enzyme and all of the variants significantly protected transduced cells from trimetrexate (TMTX)-induced growth inhibition. However, only half of the variants conferred more protection than does the wild-type enzyme. For the variants tested, the observed protective effect was higher for TMTX than for methotrexate (< or =7.5-fold increased resistance), piritrexim (< or =16-fold), and edatrexate (negligible). Transduction of the variants L22Y-F31S and L22Y-F31R led to the greatest protection against TMTX (approximately 200-fold). Protection from loss of cell viability was similar to protection from growth inhibition. The protection associated with a particular mutant hDHFR did not result from the level of expression: Efficient protection resulted from low affinity of the variant for antifolates, reasonable catalytic activity, and good thermal stability. Clones isolated from a polyclonal population of transduced cells varied by as much as 30-fold in their resistance to TMTX, the resistance differences depending on hDHFR expression levels.


Subject(s)
Folic Acid Antagonists/pharmacology , Tetrahydrofolate Dehydrogenase/genetics , Aminopterin/analogs & derivatives , Aminopterin/antagonists & inhibitors , Animals , Cell Survival , Drug Resistance, Neoplasm/genetics , Gene Expression , Genetic Variation , Growth Inhibitors/antagonists & inhibitors , Humans , Kinetics , Methotrexate/antagonists & inhibitors , Pyrimidines/antagonists & inhibitors , Rabbits , Tetrahydrofolate Dehydrogenase/pharmacology , Thymidine/metabolism , Transfection , Trimetrexate/antagonists & inhibitors
12.
Blood ; 90(9): 3546-54, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9345038

ABSTRACT

Antifolates such as methotrexate (MTX) and trimetrexate (TMTX) are widely used in the treatment of cancer and nonmalignant disorders. Transient, yet sometimes severe myelosuppression is an important limitation to the use of these drugs. It has previously been shown that clonogenic myeloid progenitors and colony-forming units-spleen are resistant to antifolates, suggesting that myelotoxicity occurs late in hematopoietic development. The goal of this study was to define the mechanisms by which primitive hematopoietic cells resist the toxic effects of antifolate drugs. To test the hypothesis that myeloid progenitors may salvage extracellular nucleotide precursors to resist TMTX toxicity, a defined liquid culture system was developed to measure TMTX toxicity in expanding progenitor populations. These in vitro experiments showed that both human and murine progenitors can resist TMTX toxicity by importing thymidine and hypoxanthine from the serum. As predicted from these findings, several drugs that block thymidine transport sensitized progenitors to TMTX in vitro, although to differing degrees. These nucleoside transport inhibitors were used to test whether progenitors and hematopoietic stem cells (HSCs) could be sensitized to TMTX in vivo. Treatment of mice with TMTX and nitrobenzylmercaptopurineriboside phosphate (NBMPR-P), a potent transport inhibitor, caused significant depletions of clonogenic progenitors within the bone marrow (20-fold) and spleen (6-fold). Furthermore, NBMPR-P administration dramatically sensitized HSCs to TMTX, with dual-treated mice showing a greater than 90% reduction in bone marrow repopulating activity. These studies demonstrate that both myeloid progenitor cells and HSCs resist TMTX by using nucleotide salvage mechanisms and that these pathways can be pharmacologically blocked in vivo using nucleoside transport inhibitors. These results have important implications regarding the use of transport inhibitors for cancer therapy and for using variants of dihydrofolate reductase for in vivo selection of genetically modified HSCs.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Drug Resistance, Neoplasm/physiology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Thioinosine/analogs & derivatives , Thionucleotides/pharmacology , Thymidine/metabolism , Trimetrexate/toxicity , Animals , Biological Transport/drug effects , Biological Transport/physiology , Cell Death/drug effects , Female , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Thioinosine/pharmacology
13.
Hum Gene Ther ; 8(15): 1773-83, 1997 Oct 10.
Article in English | MEDLINE | ID: mdl-9358027

ABSTRACT

Chemoresistance gene transfer is an experimental method to protect hematopoietic cells from the toxicity of anticancer drugs. Because multiple drugs are usually given together in cancer therapy, this strategy will ultimately require vectors expressing multiple chemoresistance genes. For this reason, we designed a bicistronic retroviral vector (HaMID) containing a modified human multidrug resistance-1 cDNA and a mutant human dihydrofolate reductase cDNA bearing a leucine to tyrosine substitution at codon 22 (L22Y). To determine if this vector would confer dual drug resistance to hematopoietic cells, recombinant retrovirus was used to transduce the human CEM T lymphoblastic cell line as well as primary murine myeloid progenitors. Growth suppression assays, using polyclonal transduced CEM cells, demonstrated increased resistance to taxol (13-fold), trimetrexate (8.9-fold), vinblastine (5.6-fold), methotrexate (2.5-fold), and etoposide (1.5-fold) when used as single agents. HaMID-transduced cells also grew at a logarithmic rate in the simultaneous presence of 25 nM taxol and 100 nM trimetrexate while control cells were entirely growth inhibited by this drug combination. Similarly, HaMID-transduced murine myeloid progenitors acquired increased resistance to taxol (2.9-fold) and trimetrexate (140-fold), and were able to form colonies in the simultaneous presence of both drugs. Our results suggest that retroviral transfer of HaMID into primary hematopoietic cells should reduce the myelosuppression associated with the combined use of antifolates and P-glycoprotein-effluxed drugs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antineoplastic Agents, Phytogenic/adverse effects , Drug Resistance, Multiple , Folic Acid Antagonists/adverse effects , Genetic Vectors , Hematopoietic Stem Cells/drug effects , Retroviridae/genetics , Tetrahydrofolate Dehydrogenase/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Cells, Cultured , Cloning, Molecular , Etoposide/adverse effects , Gene Expression , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Methotrexate/adverse effects , Paclitaxel/adverse effects , Proviruses , Transformation, Genetic , Trimetrexate/adverse effects , Tumor Cells, Cultured , Vinblastine/adverse effects
14.
Hum Gene Ther ; 8(13): 1531-43, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9322086

ABSTRACT

Class 1 aldehyde dehydrogenases (ALDH-1) function as drug resistance gene products by catalyzing the irreversible conversion of aldophosphamide, an active metabolite of cyclophosphamide, to an inert compound. Because the dose-limiting toxicity of cyclophosphamide is myelosuppression, retrovirus-mediated transfer of ALDH-1 to bone marrow cells has been proposed as a protective strategy. Here we show that expression of ALDH-1 vectors was problematic due to low levels of ALDH-1 mRNA accumulation. A number of vectors containing several different ALDH-1 cDNAs were introduced into a variety of different cell lines either by transfection or transduction. Detectable ALDH-1 protein and enzyme activity was only seen in one transfected cell clone. Cells transduced with ALDH-1 retroviral vectors had no detectable protein expression and very low levels of ALDH-1 mRNA. Analogous vectors containing other drug resistance cDNAs led to much higher levels of steady-state mRNA. The mRNA half-life from ALDH-1 vectors was less than 2 hr suggesting that vector-derived mRNAs were destabilized by ALDH-1 coding sequences. These results suggest that methods which increase the stability of ALDH-1 mRNAs will be important for increased drug resistance in retrovirally transduced hematopoietic cells.


Subject(s)
Aldehyde Dehydrogenase/genetics , Genetic Vectors , RNA, Messenger/genetics , Retroviridae/genetics , Animals , DNA, Complementary , Drug Resistance/genetics , Half-Life , Mice , RNA, Messenger/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Transduction, Genetic , Transfection , Tumor Cells, Cultured
15.
Acta Crystallogr D Biol Crystallogr ; 53(Pt 6): 638-49, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-15299851

ABSTRACT

The novel furopyrimidine N-(4-{N-[(2,4-diaminofuro[2,3-d]pyrimidin-5-yl)methyl]methylamino}benzoyl)-L- glutamate (MTXO), a classical antifolate with antitumor activity comparable to that of methotrexate (MTX), has been studied as inhibitor-cofactor ternary crystal complexes with wild-type Pneumocystis carinii (pc) and recombinant human wild-type dihydrofolate reductase (hDHFR). These structural data provide the first direct comparison of the binding interactions of the same antifolate inhibitor in the active site for pc and human DHFR. The human ternary DHFR complex crystallizes in the rhombohedral space group R3 and is isomorphous to the ternary complex reported for a gamma-tetrazole methotrexate analogue, MTXT. The pcDHFR complex crystallizes in the monoclinic space group P2(1) and is isomorphous to that reported for a trimethoprim (TMP) complex. Interpretation of difference Fourier electron-density maps for these ternary complexes revealed that MTXO binds with its 2,4-diaminofuropyrimidine ring interacting with Glu32 in pc and Glu30 in human DHFR, as observed for MTXT. The presence of the 6-5 furopyrimidine ring instead of the 6-6 pteridine ring results in a different bridge conformation compared with that of MTXT. The bridge torsion angles for MTXO, i.e. C(4a)-C(5)-C(8)-N(9) and C(5)-C(8)-N(9)-C(1'), are -156.5/51.9 degrees and -162.6/51.8 degrees, respectively for h and pc, compared with -146.8/57.4 degrees for MTXT. In each case, the p-aminobenzoylglutamate conformation is similar to that observed for MTXT. In the pcDHFR complex, the active-site region is conserved and the additional 20 residues in the sequence compared with the human enzyme are located in external loop regions. There is a significant change in the nicotinamide ribose conformation of the cofactor which places the nicotinamide O atom close to the 4NH(2) group of MTXO (2.7 A), a shift not observed in hDHFR structures. As a consequence of this, there is a loss of a hydrogen bond between the nicotinamide carbonyl group and the backbone of Ala12 in pcDHFR. In the human ternary complexes, the cofactor NADPH is bound with a more extended conformation, and the nicotinamide O atom makes a 3.5 A contact with the 4NH(2) group of MTXO. Although the novel classical antifolate MTXO is not highly active against pcDHFR, there are correlations between its binding interactions consistent with its lower potency as an inhibitor of h and pcDHFR compared with MTX.

16.
Leukemia ; 10(3): 439-46, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8642859

ABSTRACT

Resistance to methotrexate (MTX) in some sublines of mammalian cells is reported to be due to one of the following amino acid substitutions in dihydrofolate reductase (DHFR) that lower inhibition by MTX: Gly15 to Trp, Leu22 to Arg or Phe or Phe31 to Trp or Ser. We have produced variants of human DHFR (hDHFR) with these substitutions by directed mutagenesis. Recombinant hDHFR variants expressed in Escherichia coli have greatly decreased inhibition by MTX, but decreased catalytic efficiency, and in one case decreased stability. When a retroviral vector encoding wild-type (wt) hDHFR or one of these variants was introduced into murine fibroblasts or bone marrow progenitors, modest protection from MTX was conferred, even by wt. Relapsed pediatric patients with acute lymphoblastic leukemia who have received multiple courses of high-dose MTX seem most likely to develop such MTX resistance. cDNA was reverse transcribed from blast mRNA from 17 of these patients. However, upon amplification and sequencing of DHFR cDNA, no resistance mutation was found. The explanation for this probably lies in the need for considerable gene amplification to offset lowered catalytic efficiency, and the need for two-base changes for most substitutions, both of which are probably infrequent events.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Genes , Methotrexate/therapeutic use , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Tetrahydrofolate Dehydrogenase/genetics , Adolescent , Animals , Base Sequence , Child , Child, Preschool , Drug Resistance, Neoplasm , Female , Genetic Vectors , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Recurrence , Retroviridae/genetics , Tetrahydrofolate Dehydrogenase/isolation & purification
17.
Blood ; 87(6): 2579-87, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8630426

ABSTRACT

Trimetrexate (TMTX) is an anticancer drug with potential advantages over the more commonly used antifolate, methotrexate (MTX); however, its use has been limited by severe myelosuppression. Retroviral vectors containing mutant dihydrofolate reductase (DHFR) genes have been used to protect bone marrow cells from MTX, suggesting a similar approach could be used for TMTX. We first screened six variants of human DHFR to determine which allowed maximal TMTX resistance in fibroblasts. A variant enzyme containing a Leu-to-Tyr mutation in the 22nd codon (L22Y) was best, allowing a 100-fold increase in resistance over controls. Murine hematopoietic progenitor cells transduced with an L22Y-containing retroviral vector also showed high-level TMTX resistance in vitro. Mice reconstituted with L22Y-transduced bone marrow cells were challenged with a 5-day course of TMTX to determine whether hematopoiesis could be protected in vivo. Transfer of the L22Y vector resulted in consistent protection from TMTX-induced neutropenia and reticulocytopenia at levels that correlated with the proviral copy number in circulating leukocytes. We conclude that the L22Y vector is highly effective in protecting hematopoiesis from TMTX toxicity and may provide a means for increasing the therapeutic utility of TMTX in certain cancers.


Subject(s)
Bone Marrow/drug effects , Drug Resistance, Neoplasm/genetics , Genetic Therapy , Genetic Vectors , Hematopoietic Stem Cells/drug effects , Retroviridae/genetics , Tetrahydrofolate Dehydrogenase/genetics , Transfection , Trimetrexate/pharmacology , Animals , Base Sequence , Bone Marrow Cells , Bone Marrow Transplantation , DNA, Complementary/genetics , Fibroblasts/drug effects , Fibroblasts/enzymology , Hematopoietic Stem Cells/enzymology , Humans , Kinetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Recombinant Fusion Proteins/metabolism , Tetrahydrofolate Dehydrogenase/metabolism
18.
J Biol Chem ; 270(10): 5057-64, 1995 Mar 10.
Article in English | MEDLINE | ID: mdl-7890613

ABSTRACT

Although substitution of tyrosine, phenylalanine, tryptophan, or arginine for leucine 22 in human dihydrofolate reductase greatly slows hydride transfer, there is little loss in overall activity (kcat) at pH 7.65 (except for the arginine 22 variant), but Km for dihydrofolate and NADPH are increased significantly. The greatest effect, decreased binding of methotrexate to the enzyme-NADPH complex by 740- to 28,000-fold due to a large increase in the rate of methotrexate dissociation, makes these variants suitable to act as selectable markers. Affinities for four other inhibitors are also greatly decreased. Binding of methotrexate to apoenzyme is decreased much less (decreases as much as 120-fold), binding of tetrahydrofolate is decreased as much as 23-fold, and binding of dihydrofolate is decreased little or increased. Crystal structures of ternary complexes of three of the variants show that the mutations cause little perturbation of the protein backbone, of side chains of other active site residues, or of bound inhibitor. The largest structural deviations occur in the ternary complex of the arginine variant at residues 21-27 and in the orientation of the methotrexate. Tyrosine 22 and arginine 22 relieve short contacts to methotrexate and NADPH by occupying low probability conformations, but this is unnecessary for phenylalanine 22 in the piritrexim complex.


Subject(s)
Genetic Variation , Leucine , Methotrexate/pharmacology , Protein Conformation , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Drug Resistance , Enzyme Stability , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , NADP/metabolism , Point Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
20.
Biochemistry ; 33(33): 9945-52, 1994 Aug 23.
Article in English | MEDLINE | ID: mdl-8061003

ABSTRACT

Directed mutagenesis has been used to construct five variants of human dihydrofolate reductase in which smaller residues are substituted for phenylalanine 34, a residue participating in the binding of substrate and methotrexate by interaction with their pteridine rings. The variant enzymes are stable and have decreased affinities for methotrexate (by factors of 2700-60000 at pH 7.65) due to a decreased rate of methotrexate association and a much larger increase in the rate constant for dissociation. However, the catalytic efficiencies of the variants are also lowered by factors of 160-5000, so that it is doubtful whether these enzymes are capable of conferring methotrexate resistance on the cells harboring them. High concentrations of dihydrofolate cause marked inhibition of all the variants, which complicates the determination of kinetic parameters. By the use of stopped-flow spectrophotometry and fluorimetry and other methods, it has been shown that, like the wild-type enzyme, the variants have a branched reaction pathway, but in contrast to the wild-type enzyme, the distribution of flux between alternate pathways is dependent on the concentration of dihydrofolate. This different branch point is a consequence of the very rapid dissociation of tetrahydrofolate from the ternary product complexes of the variant enzymes. Inhibition by dihydrofolate is due to its combination with the enzyme-NADP complex and the slow dissociation of NADP from the resulting abortive complex. When steady state kinetics for this model are simulated using the experimentally determined rate and dissociation constants for the alanine 34 variant, most steady state experimental results are closely approximated.


Subject(s)
Phenylalanine , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , Ammonium Sulfate , Catalysis , Chemical Precipitation , Enzyme Stability , Folic Acid/analogs & derivatives , Folic Acid/metabolism , Humans , Hydrogen-Ion Concentration , Kinetics , Methotrexate/metabolism , Methotrexate/pharmacology , Mutagenesis, Site-Directed , NADP/metabolism , NADP/pharmacology , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...