Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Genet ; 14(1): 33-41, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8782817

ABSTRACT

The recurrent translocation t(8;16)(p11;p13) is a cytogenetic hallmark for the M4/M5 subtype of acute myeloid leukaemia. Here we identify the breakpoint-associated genes. Positional cloning on chromosome 16 implicates the CREB-binding protein (CBP), a transcriptional adaptor/coactivator protein. At the chromosome 8 breakpoint we identify a novel gene, MOZ, which encodes a 2,004-amino-acid protein characterized by two C4HC3 zinc fingers and a single C2HC zinc finger in conjunction with a putative acetyltransferase signature. In-frame MOZ-CBP fusion transcripts combine the MOZ finger motifs and putative acetyltransferase domain with a largely intact CBP. We suggest that MOZ may represent a chromatin-associated acetyltransferase, and raise the possibility that a dominant MOZ-CBP fusion protein could mediate leukaemogenesis via aberrant chromatin acetylation.


Subject(s)
Acetyltransferases/genetics , Chromosomes, Human, Pair 16 , Chromosomes, Human, Pair 8 , Leukemia, Monocytic, Acute/genetics , Leukemia, Myelomonocytic, Acute/genetics , Nuclear Proteins/genetics , Trans-Activators , Transcription Factors/genetics , Translocation, Genetic , Amino Acid Sequence , Animals , Base Sequence , CREB-Binding Protein , Chromosome Mapping , Cloning, Molecular , Cricetinae , Gene Expression , Histone Acetyltransferases , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Homology, Amino Acid , Zinc Fingers/genetics
2.
Science ; 216(4541): 75-7, 1982 Apr 02.
Article in English | MEDLINE | ID: mdl-6950518

ABSTRACT

The ornithine decarboxylase inhibitor DL-alpha-difluoromethyl ornithine inhibited a proliferation-associated increase in ornithine decarboxylase activity in cultured human promyelocytic leukemia cells, resulting in a marked suppression of cell proliferation and subsequent cell loss. It also inhibited increases in ornithine decarboxylase activity associated with the phorbol ester-induced conversion of promyelocytic HL-60 cells to monocyte-like cells and the retinoic acid-induced conversion to granulocyte-like cells. However, the inhibition of ornithine decarboxylase activity did not prevent cellular differentiation. These results suggest that polyamine biosynthesis has a specific role in cell proliferation rather than in inducing differentiation that is not accompanied by proliferation. The data also demonstrate that cessation of proliferation in HL-60 cells is not necessarily associated with differentiation.


Subject(s)
Carboxy-Lyases/physiology , Leukemia, Myeloid, Acute/enzymology , Ornithine Decarboxylase/physiology , Cell Adhesion , Cell Differentiation , Cell Division , Cells, Cultured , Female , Granulocytes/pathology , Humans , Leukemia, Myeloid, Acute/pathology , Macrophages/pathology , Polyamines/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology
3.
Oncogene ; 25(55): 7289-96, 2006 Nov 23.
Article in English | MEDLINE | ID: mdl-16767164

ABSTRACT

In the core binding factor (CBF)beta-smooth muscle myosin heavy chain (SMMHC) acute myeloid leukemia (AML) oncoprotein, CBFbeta lies N-terminal to the alpha-helical rod domain of SMMHC. Deletion of the SMMHC assembly competence domain (ACD), conserved among skeletal, smooth and nonmuscle myosins, prevents multimerization, inhibition of CBF and inhibition of cell proliferation. To define the amino acids critical for ACD function, three outer surface residues of ACD helices A-D, the subsequent helices E-H or the more N-terminal X or Z helices were now mutated. Variants were assessed for multimerization in low ionic strength in vitro and for nuclear localization as a measure of in vivo multimerization. Mutation of individual helices C-H reduced multimerization, with alteration of the outer surface of helices D or E having the greatest effect. The ability of these SMMHC variants to slow murine myeloid progenitor proliferation largely paralleled their effects on multimerization. Divergence at the boundaries of the ACD may reflect quantitative differences between in vitro and in vivo filament assembly. Each helix mutant retained the ability to bind the mSin3A corepressor. Agents interacting with the outer surface of the CBFbeta-SMMHC ACD that prevent multimerization may be effective as novel therapeutics in AML.


Subject(s)
Oncogene Proteins, Fusion/metabolism , Amino Acid Sequence , Biopolymers , Cell Line , Humans , Molecular Sequence Data , Mutation , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics
4.
J Clin Invest ; 74(6): 2193-7, 1984 Dec.
Article in English | MEDLINE | ID: mdl-6511923

ABSTRACT

In this paper, we report analysis of differentiation in human hemopoietic colonies derived from a single cell. Cord blood mononulear cells and panned My-10 antigen-positive bone marrow and cord blood cells were plated in methylcellulose medium containing erythropoietin and conditioned medium. Initially, we performed mapping studies to identify candidate colony-forming cells. Subsequently, using a micromanipulator, we transferred single cells individually to 35-mm dishes for analysis of colony formation. Cellular composition of the colony was determined by identifying all of the cells in the May-Grunwald-Giemsa stained preparation. Of 150 single candidate cells replated, 63 produced colonies. The incidences of single lineage colonies included 19 erythroid, 17 monocyte-macrophage, and 9 eosinophil colonies. There were 18 mixed hemopoietic colonies consisting of cells in two, three, four, and five lineages in varying combinations. In some instances, we noted the predominance of one lineage and the presence of very small populations of cells in a second or third lineage. These results provide evidence for the single-cell origin of human multilineage hemopoietic colonies, and are consistent with the stochastic model of stem cell differentiation in man. They also indicate that restriction of the proliferative potential of committed progenitors is a stochastic process.


Subject(s)
Hematopoietic Stem Cells/cytology , Bone Marrow Cells , Cell Count , Cell Differentiation , Cells, Cultured , Colony-Forming Units Assay , Female , Fetal Blood/cytology , Humans , Pregnancy
5.
J Clin Invest ; 80(2): 357-66, 1987 Aug.
Article in English | MEDLINE | ID: mdl-3038955

ABSTRACT

Morphological and biochemical studies of human colony-forming units-erythroid (CFU-E) have been hindered by their extreme rarity. Since burst-forming units-erythroid (BFU-E) develop into CFU-E, we used normal human blood BFU-E to generate large numbers of highly purified CFU-E in vitro. Using density centrifugation, sheep erythrocyte rosetting, surface immunoglobulin-positive cell depletion, adherence to plastic, and negative panning with monoclonal antibodies, human blood BFU-E were purified from 0.017 to 0.368%, a 22-fold purification with a 43% yield. The panned cells were cultured in methylcellulose with recombinant erythropoietin (rEp) and conditioned medium for 9 d. These cells were then collected and CFU-E were further purified using adherence and density centrifugation. This yielded almost 10(7) erythroid colony forming cells with a purity of 70 +/- 18%. Analysis of these cells by light and electron microscopy showed 94% erythroid cells. The prominent cell was a primitive blast with high nuclear/cytoplasmic ratio, dispersed nuclear chromatin and a distinct large nucleolus. The relation between the number of erythroid colonies and the number of day 9 cells plated in plasma clots was a straight line through the origin with a maximum number of erythroid colonies at 1 U/ml of rEp and no erythroid colonies without rEp. Specific binding with 125I-rEp showed that 60% of the binding was inhibited by excess pure erythropoietin (Ep), but not by albumin, fetal calf serum, and a variety of growth factors or glycoproteins. By days 12-13 of cell culture, when the progenitor cells matured to late erythroblasts, specific binding markedly declined. In this study, human CFU-E have been isolated in sufficient purity to characterize the morphology of these rare cells and in sufficient numbers to measure specific binding of Ep.


Subject(s)
Erythroblasts/cytology , Erythropoiesis , Erythropoietin/metabolism , Antibodies, Monoclonal , Cells, Cultured , Colony-Forming Units Assay , Erythroblasts/metabolism , Humans , In Vitro Techniques , Methylcellulose , Microscopy, Electron , Receptors, Cell Surface/metabolism , Receptors, Erythropoietin , Time Factors
6.
Leukemia ; 19(6): 921-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15815715

ABSTRACT

CBFbeta-SMMHC is expressed in 8% of acute myeloid leukemias and inhibits AML1/RUNX1. In this study, murine marrow or human CD34(+) cells were transduced with retroviral or lentiviral vectors expressing CBFbeta-SMMHC or two mutant variants. CBFbeta-SMMHC reduced murine or human myeloid cell proliferation three- to four-fold in liquid culture relative to empty vector-transduced cells, during a period when vector-transduced cells accumulated five-fold and human cells 20-fold. CBFbeta-SMMHC decreased the formation of myeloid, but not erythroid, colonies two- to four-fold, and myeloid colonies expressing CBFbeta-SMMHC were markedly reduced in size. However, CBFbeta-SMMHC did not slow differentiation to granulocytes or monocytes. Neither CBFbeta-SMMHC(Delta2-11), which does not bind AML1, nor CBFbeta-SMMHC(DeltaACD), which does not multimerize or efficiently bind corepressors, slowed proliferation or reduced myeloid colonies. CBFbeta-SMMHC increased the G1/S ratio 1.4-fold. AML1 had an effect opposite to CBFbeta-SMMHC, stimulating proliferation of murine myeloid progenitors 2.0-fold in liquid culture. Thus, CBFbeta-SMMHC directly inhibits the proliferation of normal myeloid progenitors via inhibition of AML1 and dependent upon the integrity of its assembly competence domain. These findings support the development of therapeutics that target the ability of CBFbeta-SMMHC to interact with AML1 or to multimerize via its assembly competence domain.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid/pathology , Leukemia, Myeloid/therapy , Oncogene Proteins, Fusion/genetics , Acute Disease , Animals , Cell Differentiation , Cell Division , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/genetics , Female , Fetal Blood/cytology , Humans , Lentivirus/genetics , Leukemia, Myeloid/physiopathology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/genetics , Retroviridae/genetics , Transcription Factors/genetics , Transduction, Genetic
7.
Leukemia ; 19(12): 2101-16, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16136167

ABSTRACT

From 1981 to 2000, a total of 1823 children with acute myeloid leukemia (AML) enrolled on four consecutive Pediatric Oncology Group (POG) clinical trials. POG 8101 demonstrated that the induction rate associated with the 3+7+7 combination of daunorubicin, Ara-C, and 6-thioguanine (DAT) was greater than that associated with an induction regimen used to treat acute lymphoblastic leukemia (82 vs 61%; P=0.02). Designed as a pilot study to determine the feasibility of administration of noncross-resistant drug pairs and later modified to assess the effect of dose intensification of Ara-C during the second induction course, POG 8498 confirmed the high initial rate of response to DAT (84.2%) and showed that dose intensification of Ara-C during the second induction course resulted in a trend toward higher event-free survival (EFS) estimates than did standard-dose DAT (2+5) during the second induction course (5 year EFS estimates, 22 vs 27%; P=0.33). Age <2 years and leukocyte count <100 000/mm3 emerged as significantly good prognostic factors. The most significant observation made in the POG 8498 study was the markedly superior outcome of children with Down's syndrome who were treated on the high-dose Ara-C regimen. POG 8821 compared the efficacy of autologous bone marrow transplantation (BMT) with that of intensive consolidation chemotherapy. Intent-to-treat analysis revealed similar 5-year EFS estimates for the group that underwent autologous BMT (36+/-4.7%) and for the group that received only intensive chemotherapy (35+/-4.5%) (P=0.25). There was a high rate of treatment-related mortality in the autologous transplantation group. The study demonstrated superior results of allogeneic BMT for patients with histocompatible related donors (5-year EFS estimate 63+/-5.4%) and of children with Down's syndrome (5-year EFS estimate, 66+/-8.6%). The POG 9421 AML study evaluated high-dose Ara-C as part of the first induction course and the use of the multidrug resistance modulator cyclosporine. Preliminary results showed that patients receiving both high-dose Ara-C for remission induction and the MDR modulator for consolidation had a superior outcome (5-year EFS estimate, 42+/-8.2%) than did patients receiving other treatment; however, the difference was not statistically significant. These four studies demonstrate the importance of dose intensification of Ara-C in the treatment of childhood AML; cytogenetics as the single most prognostic factor and the unique curability of AML in children with Down's syndrome.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Protocols/standards , Leukemia, Myeloid/therapy , Acute Disease , Adolescent , Bone Marrow Transplantation , Child , Child, Preschool , Cytarabine/therapeutic use , Dose-Response Relationship, Drug , Down Syndrome/complications , Down Syndrome/drug therapy , Follow-Up Studies , Humans , Infant , Infant, Newborn , Leukemia, Myeloid/complications , Leukemia, Myeloid/mortality , Prognosis , Remission Induction/methods , Survival Analysis , Treatment Outcome
8.
Cancer Res ; 51(16): 4279-86, 1991 Aug 15.
Article in English | MEDLINE | ID: mdl-1868448

ABSTRACT

Transfection of the wild-type p53 gene into malignant cell lines usually results in an inhibition of proliferation. However, the physiological function of the endogenous p53 gene product has been difficult to ascertain. In order to examine whether p53 is involved in the regulation of proliferation and/or differentiation of hematopoietic tissue, we modified a recently developed flow cytometric assay to assess p53 protein expression in normal human hematopoietic cells, primary leukemias, and selected leukemia cell lines. In normal human bone marrow, p53 protein was not detected in the proliferative, progenitor cell populations identified by the cell surface antigens CD34 (progenitor cells of multiple lineages) or glycophorin (erythroid precursors). In contrast, low but detectable levels of p53 protein were observed in the nonproliferative, mature lymphoid, granulocytic, and monocytic cell populations. Similarly, p53 levels increased and DNA synthesis decreased during 12-O-tetradecanoylphorbol-13-acetate-induced differentiation of ML-1 myeloblastic leukemia cells. Both of these results suggest that endogenous, wild-type p53 protein may play a role in hematopoietic cell maturation, possibly by contributing to the inhibition of proliferation that occurs during terminal differentiation. Leukemia cells deviated from this pattern of expression: (a) in contrast to the normal, proliferative bone marrow progenitor cells, a significant percentage of patient leukemia samples expressed detectable levels of p53 protein; and (b) leukemia cell lines exhibited lineage-specific abnormalities in p53 expression, with overexpression in lymphoid cell lines and lack of expression in myeloid cell lines.


Subject(s)
Cell Division , Hematopoietic Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics , Acute Disease , Adult , Amino Acid Sequence , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Cells , Cell Line , Cells, Cultured , Flow Cytometry , Gene Expression/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Immunoblotting , Kinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Molecular Sequence Data , Oligopeptides/chemical synthesis , Oligopeptides/immunology , RNA, Messenger/analysis , RNA, Messenger/genetics , Tetradecanoylphorbol Acetate/pharmacology , Tumor Suppressor Protein p53/analysis
9.
Cancer Res ; 46(9): 4751-5, 1986 Sep.
Article in English | MEDLINE | ID: mdl-2425958

ABSTRACT

Several mouse monoclonal antibodies which recognize carbohydrate sequences distinguish between different types of human lung cancer immunohistologically. These antibodies bind to glycolipid antigens produced by the cancer cells. When these glycolipids are separated by thin-layer chromatography, immunostaining of the chromatograms yields complex patterns of antigen-positive bands. To determine whether glycolipid patterns are useful in the classification of lung cancer, 16 human lung cancer cell lines comprising the major histological types of primary lung cancer were studied. Neutral glycolipids and gangliosides were isolated and separated by thin-layer chromatography. Six anti-carbohydrate antibodies which recognize structurally related antigens were used for immunostaining. Neuraminidase treatment of the chromatograms was used to detect "cryptic" sialylated antigens. All the cell lines were unique with regard to the type, amount, and chromatography pattern of the glycolipid antigens produced. Small cell lung cancer cell lines synthesized the greatest variety of antigens, whereas cell lines with large cell cytology synthesized the least. Interestingly, there was an inverse relationship between expression of some glycolipid antigens and DNA amplification of the c-myc oncogene. This suggests that enhanced c-myc expression may influence the types of glycolipids expressed at the surface of lung tumor cells.


Subject(s)
Antigens, Neoplasm/immunology , Glycolipids/immunology , Lung Neoplasms/immunology , ABO Blood-Group System/immunology , Carbohydrate Sequence , Epitopes , Gangliosides/immunology , Glycolipids/metabolism , Humans , Lung Neoplasms/metabolism , P Blood-Group System/immunology
10.
Cancer Res ; 57(5): 837-41, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9041182

ABSTRACT

Inactivation of the cyclin-dependent kinase inhibitors p16INK4A and p15INK4B are frequent alterations in neoplasia, often resulting from homozygous deletion or promoter region hypermethylation. We have analyzed both modes of inactivation of p15INK4B and p16INK4A in the major types of adult and pediatric hematological malignancies. Hypermethylation of p15INK4B, without alteration of p16INK4A, was an almost universal finding in adult acute myelogenous leukemia, and occurred very frequently in adult acute lymphocytic leukemia and pediatric acute myelogenous leukemia and acute lymphocytic leukemia. In contrast, neither p15INK4B nor p16INK4A were inactivated in any stage of chronic myelogenous leukemia. Hypermethylation of p16INK4A, often without alterations of p15INK4B, was found in non-Hodgkin's lymphoma and was much more frequent in cases with high-grade than low-grade histology. Enriched normal bone marrow stem cells had no detectable promoter region methylation of these genes, as analyzed by a newly developed PCR method. Remarkably distinct patterns of inactivation of p15INK4B and p16INK4A characterize different types of hematological malignancy, and alterations in these tumor suppressor genes are one of the most common alterations in hematological malignancies.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins , Leukemia/genetics , Lymphoma/genetics , Tumor Suppressor Proteins , Adult , Biomarkers, Tumor , Child , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16 , DNA Methylation , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Prognosis , Sequence Deletion
11.
Cancer Res ; 56(5): 973-77, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8640788

ABSTRACT

Estrogen appears to be a negative regulator of normal hematopoiesis. Chromosome 6q, which contains the estrogen receptor (ER) gene, is frequently altered in human hematopoietic neoplasms. The ER gene, which has growth and metastasis suppressor activity in many different cell types, is inactivated by promoter methylation in some ER-negative breast tumors and 100% of colorectal tumors. We now report that the promoter region of the ER gene is aberrantly methylated in 86% of human hematopoietic tumors, including 8 of 9 pediatric acute lymphocytic leukemia, 17 of 18 adult acute lymphocytic leukemia, 21 of 23 adult acute myelogenous leukemia, 3 of 6 chronic phase chronic myelogenous leukemia, 9 of 9 blast crisis chronic myelogenous leukemia and 5 of 8 lymphomas. This methylation event was also present in all nine leukemia cell lines examined, where it was associated with very low or absent ER expression. In addition, rat and mouse leukemia cell line also exhibited this change, indicating that ER CpG island methylation in leukemias is conserved among species. Our results suggest that ER CpG island methylation could be an important step in the genesis of human hematopoietic neoplasms and might be a useful molecular marker for monitoring the clinical status of these diseases.


Subject(s)
Chromosomes, Human, Pair 6 , Leukemia/genetics , Lymphoma/genetics , Receptors, Estrogen/genetics , Adult , Animals , Biomarkers, Tumor , Child , Child, Preschool , CpG Islands , Humans , Infant , Infant, Newborn , Leukemia/metabolism , Lymphoma/metabolism , Methylation , Mice , Rats , Tumor Cells, Cultured
12.
Cancer Res ; 43(10): 4569-74, 1983 Oct.
Article in English | MEDLINE | ID: mdl-6349795

ABSTRACT

The expression of myeloid and megakaryocytic markers of differentiation has been studied in one K-562 cell subline, in its clones, and in the original cell line. Cytotoxicity, electron microscopy, immunofluorescence studies with a panel of polyclonal and monoclonal antibodies, and radioimmunoassays were performed on K-562 cells before and after induction with hemin, sodium butyrate, and 12-O-tetradecanoylphorbol-13-acetate. Myeloid membrane markers were present in all K-562 cell lines. Only the early granulopoietic cell surface markers were expressed in 75 to 95% of the cells, while none of the late membrane markers was detected. In contrast, neither the early (myeloperoxidase) nor late (lactoferrin) cytoplasmic markers were present. Thus, K-562 cells showed a membrane phenotype similar to that of a normal or leukemic promyelocyte but lacking myeloperoxidase. Membrane megakaryocytic markers, such as platelet glycoprotein IIIa and platelet peroxidase, were also detected in K-562 cells. However, some other early megakaryocytic markers, such as platelet glycoprotein lb, Factor VIII-R-Ag, and platelet Factor 4, could not be detected by fluorescent labeling. Cloning of the cell line did not result in the selection of a unipotential cell line. These results could be explained by the expression of multilineage markers in a single cell. In all of the cell lines and clones, hemin slightly increased the expression of the myeloid membrane markers without any modification of the megakaryocytic markers. Sodium butyrate and 12-O-tetradecanoylphorbol-13-acetate diminished most of the myeloid markers and very significantly increased the expression of the megakaryocytic markers.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Antibodies, Monoclonal , Cell Differentiation , Cell Line , Flow Cytometry , Fluorescent Antibody Technique , Humans , Leukemia, Myeloid, Acute/ultrastructure , Radioimmunoassay
13.
Leukemia ; 30(6): 1327-34, 2016 06.
Article in English | MEDLINE | ID: mdl-26965284

ABSTRACT

Missing in metastasis (MIM) is abundantly expressed in hematopoietic cells. Here we characterized the impact of MIM deficiency on murine bone marrow (BM) cells. Although MIM(-/-) cells proliferated similarly to wild type (WT), they exhibited stronger response to chemokine stromal-derived factor 1 (SDF-1), increase in surface expression of CXCR4, impaired CXCR4 internalization and constitutive activation of Rac, Cdc42 and p38. Transplantation of MIM(-/-) BM cells into lethally irradiated mice showed enhanced homing to BM, which was abolished when mice were pretreated with a p38 antagonist. Interestingly, MIM(-/-) BM cells, including hematopoietic stem and progenitor cells (HSPCs), showed two- to fivefold increase in mobilization into the peripheral blood upon treatment with AMD3100. In vitro, MIM(-/-) leukocytes were susceptible to AMD3100 and maintained increased response to AMD3100 for mobilization even after transfer into WT mice. MIM(-/-) mice had also a higher level of SDF-1 in the circulation. Our data highlighted an unprecedented role of MIM in the homeostasis of BM cells, including HSPCs, through modulation of the CXCR4/SDF-1 axis and interactions of BM leukocytes with their microenvironments.


Subject(s)
Bone Marrow Cells/metabolism , Microfilament Proteins/physiology , Neoplasm Proteins/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Cell Movement , Chemokine CXCL12/metabolism , Hematopoietic Stem Cells/metabolism , Homeostasis , Leukocytes , Mice , Receptors, CXCR4/metabolism
14.
Oncogene ; 12(4): 903-13, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8632913

ABSTRACT

Degenerate PCR was employed to identify novel tyrosine kinase genes from an enriched population of human umbilical cord blood hematopoietic stem/progenitor cells. One novel tyrosine kinase gene, designated Tnk1, was cloned. The sequence of the complete Tnk1 coding region predicts a 72 kD protein. Comparison of Tnk1 to available sequences in protein databases reveals that it is most homologous to Ack, an intracellular tyrosine kinase which associates with the GTP-bound form of p21cdc42Hs. Like Ack, Tnk1 consists of an N-terminal kinase domain, a putative SH3 domain immediately C-terminal to the kinase domain, and a proline-rich C-terminal region. Analysis of Tnk1 mRNA expression demonstrates that Tnk1 is expressed in all cord blood, bone marrow and adult blood sub-populations, as well as in most of the leukemia cell lines examined (16 of 20). Hybridization to fetal multi-tissue Northern blots detected several different Tnk1 transcripts in all fetal tissues examined. In contrast, a single Tnk1 transcript was detected in only five of 16 adult tissues examined (prostate, testis, ovary, small intestine and colon). Fluorescence in situ hybridization (FISH) analysis of metaphase chromosomes localized the Tnk1 gene to the short arm of chromosome 17 (17p13.1), near the p53 locus. Thus, Tnk1 is a novel tyrosine kinase that may be involved in signalling pathways utilized broadly during fetal development, more selectively in adult tissues and in cell of the lymphohematopoietic system.


Subject(s)
Chromosomes, Human, Pair 17 , Fetal Blood/cytology , Fetal Proteins/biosynthesis , Fetal Proteins/genetics , Hematopoietic Stem Cells/enzymology , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Amino Acid Sequence , Antigens, CD/blood , Antigens, CD34/blood , Antigens, Differentiation/blood , Base Sequence , Blotting, Northern , Cell Line , Chromosome Mapping , DNA Primers , Female , Fetus , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Leukemia , Membrane Glycoproteins , Molecular Sequence Data , N-Glycosyl Hydrolases/blood , Organ Specificity , Polymerase Chain Reaction , Pregnancy , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Restriction Mapping , Sequence Homology, Amino Acid , Tumor Cells, Cultured
15.
J Clin Oncol ; 9(2): 247-58, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1988573

ABSTRACT

Two hundred fifty-six children with previously untreated acute nonlymphocytic leukemia (ANLL) were evaluated on a Pediatric Oncology Group (POG) phase III randomized trial of both induction and continuation chemotherapies. Induction therapy compared vincristine, cytarabine, and dexamethasone (VADx) with daunorubicin, cytarabine, and thioguanine (DAT). The complete remission (CR) rate using DAT was superior (82% v 61%, P = .02). Postremission therapy consisted of either "standard" two-cycle therapy or a more intensive four-cycle regimen given for 2 years. Overall, there was no difference in outcome for patients randomized to either continuation regimen. The overall complete continuous remission rate (CCR) for the "best" induction/continuation therapy combination at 2 years was .50 (SE = .06), at 3 years was .35 (.04), and at 4 years was .34 (.05). Analysis of selected clinical and laboratory parameters demonstrated differences in induction responses favoring DAT induction but did not impact eventual disease-free survival. There were two subgroups of patients who responded better to four-cycle continuation therapy. These were patients with French-American-British (FAB) M1/M2 (2-year CCR was .20 v .44, P = .01) and patients older than 10 years at diagnosis (.32 v .62, P = .004).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Agranulocytosis/chemically induced , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bone Marrow/pathology , Child , Child, Preschool , Cytarabine/administration & dosage , DNA Nucleotidylexotransferase/metabolism , Daunorubicin/administration & dosage , Dexamethasone/administration & dosage , Female , Fever/chemically induced , Humans , Infant , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Multivariate Analysis , Random Allocation , Receptors, Glucocorticoid/analysis , Recurrence , Remission Induction , Retrospective Studies , Survival Rate , Thioguanine/administration & dosage , Tumor Cells, Cultured , Vincristine/administration & dosage
16.
J Clin Oncol ; 12(7): 1383-9, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8021728

ABSTRACT

PURPOSE: To determine the potential efficacy and toxicity of intravenous (i.v.) methotrexate (MTX) and mercaptopurine (MP) as postremission intensification treatment for children with B-lineage acute lymphoblastic leukemia (ALL) at higher risk to relapse. PATIENTS AND METHODS: Eighty-three patients (age 1 to 20 years) with higher-risk B-lineage ALL were entered onto this protocol. Following standard four-drug remission induction, 80 patients received 12 intensive 2-week cycles of MTX/MP: MTX 200 mg/m2 i.v. push, then 800 mg/m2 i.v. 24-hour infusion on day 1; MP 200 mg/m2 i.v. in 20 minutes, then 800 mg/m2 i.v. 8-hour infusion day 2; MTX 20 mg/m2 intramuscularly day 8; and MP 50 mg/m2 by mouth days 8 to 14. Age-based triple intrathecal therapy (MTX, hydrocortisone, and cytarabine) was administered for CNS prophylaxis. Continuation therapy was weekly MTX/MP (as on days 8 to 14) for 2 years. RESULTS: Eighty-one patients (98%) entered remission. There were 28 relapses (marrow, n = 11; marrow and CNS, n = 2; isolated CNS, n = 9; testes, n = 5; ovaries, n = 1). No overt relapse occurred during the intensive phase of therapy. The event-free survival (EFS) rate at 4 years is 57.4% +/- 9.1% (SE). Hematologic, mucosal, and infectious toxicities were seen in 12%, 9%, and 5% of intensive MTX/MP courses, but were generally mild. CONCLUSION: Combined data from this and our previous trial suggest that intensive MTX/MP may produce long-term disease-free survival in 70 to 75% of children with B-lineage ALL. In comparison to other intensive regimens, intensive MTX/MP is easy to administer, effective, and relatively nontoxic. If patients at risk for failure of MTX/MP can be identified prospectively, more aggressive regimens could be restricted to this smaller (25% to 30%) cohort.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B-Lymphocytes , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Child, Preschool , Female , Humans , Infant , Infusions, Intravenous , Male , Mercaptopurine/administration & dosage , Methotrexate/administration & dosage , Prognosis , Survival Analysis , Treatment Outcome
17.
J Clin Oncol ; 9(4): 572-80, 1991 Apr.
Article in English | MEDLINE | ID: mdl-2066754

ABSTRACT

In June 1984, the Pediatric Oncology Group (POG) initiated a pilot study (8498) using high-dose cytarabine (HdA; 3 g/m2) for intensification of early therapy in childhood acute myelogenous leukemia (AML) (group I). Remission induction therapy consisted of two courses of daunorubicin, cytarabine (Ara-C), and thioguanine (DAT). Postremission therapy consisted of four sequential courses, each consisting of (1) four doses of HdA (HdA4) followed by asparaginase (L-Asp), (2) etoposide (VP) plus azacytidine (Az), (3) prednisone, vincristine, methotrexate, and mercaptopurine (POMP), and (4) Ara-C daily for 5 days by continuous infusion. Six doses of intrathecal Ara-C were given for CNS prophylaxis. In December 1986, the protocol was amended (group II) to substitute six doses of HdA (HdA6) for the second DAT (two + five) induction course; postinduction, a single course of HdA6 was given instead of four HdA/L-Asp courses, and the remainder of the therapy was unchanged. One hundred forty group I patients and 145 group II patients were assessable. The two groups were similar with regard to clinical prognostic groups. No significant differences were noted in the two groups with regard to remission induction (85% [SE = 2%] in each group), induction deaths (6.5% v 7.0%), or deaths in remission (one in each group). Cerebellar toxicity was reported in three patients in group II (with HdA6) but none in group I (HdA4). At present, patients who received HdA6 (group II) had higher event-free survival than patients in group I (EFS at 3 years, 34% [SE = 11%] v 29% [SE = 4%]), and disease-free survival (DFS at 3 years, 42% [SE = 14%] v 34% [SE = 4%]), but the differences were not statistically significant. In both groups, children less than 2 years and those with WBCs less than 100,000/microL had significantly better outcome (EFS of 55% [SE = 10%] and 36% [SE = 5%] at 3 years, respectively) than children greater than or equal to 2 years and those with WBCs greater than or equal to 100,000/microL (EFS of 27% [SE = 5%] and 20% [SE = 9%] at 3 years, respectively.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytarabine/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Adolescent , Adult , Child , Child, Preschool , Humans , Infant , Leukemia, Myeloid, Acute/mortality , Pilot Projects , Prognosis , Recurrence , Remission Induction , Survival Rate
18.
J Clin Oncol ; 5(12): 1900-11, 1987 Dec.
Article in English | MEDLINE | ID: mdl-3500279

ABSTRACT

2'Deoxycoformycin (dCF) specifically inhibits adenosine deaminase (ADA) and causes selective cytotoxicity of normal and malignant T cells. In clinical trials, dCF caused rapid lysis of malignant T lymphoblasts. Although dCF has been associated with dose-limiting nonhematopoietic toxicities, myelosuppression has not been observed. Since dCF is relatively nontoxic to hematopoietic stem cells, we tested dCF for utility in the ex vivo purging of malignant T lymphoblasts from remission leukemic bone marrow for autologous bone marrow transplantation. We found that T lymphoblast cell lines were sensitive to dCF (plus deoxyadenosine [dAdo]) under conditions that did not ablate human hematopoietic colony-forming cells. Moreover, combined pharmacologic (dCF plus dAdo) and immunologic (anti-T cell monoclonal antibodies [McAb] plus complement) purging resulted in additive reduction in clonogenic T lymphoblasts. These results provide the basis for a clinical trial of bone marrow transplantation using combined pharmacologic/immunologic purging of T lymphoblasts from patients' harvested autologous marrow.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Agents/pharmacology , Coformycin/pharmacology , Colony-Forming Units Assay , Lymphoma/pathology , Ribonucleosides/pharmacology , T-Lymphocytes/drug effects , Cell Line , Coformycin/analogs & derivatives , Complement System Proteins/physiology , Deoxyadenosines/pharmacology , Humans , Lymphocyte Depletion , Pentostatin , T-Lymphocytes/immunology
19.
J Clin Oncol ; 8(8): 1389-98, 1990 Aug.
Article in English | MEDLINE | ID: mdl-1696310

ABSTRACT

We studied the blasts from 795 children greater than 1 year of age with newly diagnosed, untreated B-precursor acute lymphoblastic leukemia (ALL) for expression of the hematopoietic stem cell-associated antigen CD34. All cases were confirmed as B-lineage lymphoblastic leukemia by virtue of expression of CD19 and/or CD22, lack of T-cell antigens, and lack of surface-membrane immunoglobulin (Ig). The CD34 antigen was present in at least 10% of blast cells in 587 (73.8%) of the patients. There was no significant difference in presenting clinical characteristics between CD34+ and CD34- patients save for an increased incidence of CNS involvement at diagnosis in the latter. Patients with CD34+ leukemia were more likely to have blasts expressing CD22, CD9, and CD13 antigens but were less likely to coexpress CD20. Patients with pre-B (cytoplasmic mu) ALL were significantly more likely to lack CD34 on their blasts, while children with hyperdiploid ALL were more likely to be CD34+. Although remission induction rates were not significantly different between patients with CD34+ and CD34-ALL (P = .23), event-free survival was shorter for patients with CD34- leukemia (P = .0014). Even though CD34 expression was associated with certain other known prognostically favorable variables including hyperdiploidy and lack of cytoplasmic Ig, it had an independent favorable effect on treatment outcome, even after adjusting for competing prognostic factors.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation/metabolism , Burkitt Lymphoma/immunology , Antigens, CD34 , Burkitt Lymphoma/genetics , Burkitt Lymphoma/mortality , Child , Child, Preschool , Cytoplasm/immunology , Diploidy , Female , Follow-Up Studies , Humans , Immunoglobulin mu-Chains/metabolism , Infant , Male , Multicenter Studies as Topic , Prognosis , Survival Rate , United States
20.
J Clin Oncol ; 7(10): 1539-44, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2778483

ABSTRACT

Methotrexate (MTX) and mercaptopurine (MP) are the mainstays of continuation therapy for acute lymphocytic leukemia (ALL). These drugs are stored in tissues as active metabolites. Relapse in ALL might reflect failure to achieve adequate intracellular drug levels. Assured (parenteral) delivery of higher doses of MTX and MP should maximize tissue levels of these drugs by overcoming individual variations in absorption, metabolism, clearance, and compliance. Fifty-nine children with ALL at lower risk of relapse received 12 intensive MTX/MP courses immediately after 4 weeks of standard vincristine, prednisone, and asparaginase induction. Each 2-week intensive course included: MTX, 200 mg/m2 intravenous (IV) push then 800 mg/m2 IV over 24 hours on day 1; MP, 200 mg/m2 IV push then 800 mg/m2 IV over 8 hours on day 2; MTX, 20 mg/m2 intramuscularly on day 8; and MP, 50 mg/m2 orally daily on days 8 to 14. After the 6 months of intensive therapy, continuation therapy was weekly MTX/MP (as on days 8 to 14) for 1 or 2 years. Age-based MTX was given intrathecally (IT) for CNS prophylaxis. All patients entered remission. Three patients relapsed: bone marrow (at 24 and 37 months), and bone marrow and CNS (at 34 months). There were no isolated CNS relapses or deaths in remission. Event-free survival at 4 years is 94% (SE, 7%) by Kaplan-Meier analysis. Toxicities (infection, mucositis) occurred in less than 10% of intensive MTX/MP courses. However, a child with Down's syndrome withdrew after three courses because of recurrent severe mucositis. Further studies of this regimen are in progress.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Infusions, Intravenous , Male , Mercaptopurine/administration & dosage , Methotrexate/administration & dosage , Neoplasm Recurrence, Local , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Remission Induction , Seizures/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL