Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Science ; 273(5278): 1096-100, 1996 Aug 23.
Article in English | MEDLINE | ID: mdl-8688094

ABSTRACT

Bruton's tyrosine kinase (BTK) is a member of the SRC-related TEC family of protein tyrosine kinases (PTKs). DT-40 lymphoma B cells, rendered BTK-deficient through targeted disruption of the btk gene by homologous recombination knockout, did not undergo radiation-induced apoptosis, but cells with disrupted lyn or syk genes did. Introduction of the wild-type, or a SRC homology 2 domain or a plecstrin homology domain mutant (but not a kinase domain mutant), human btk gene into BTK-deficient cells restored the apoptotic response to radiation. Thus, BTK is the PTK responsible for triggering radiation-induced apoptosis of lymphoma B cells, and its kinase domain is indispensable for the apoptotic response.


Subject(s)
Apoptosis , B-Lymphocytes/radiation effects , Lymphoma, B-Cell/pathology , Protein-Tyrosine Kinases/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , Chickens , Gamma Rays , Gene Targeting , Humans , Immunoglobulin M/immunology , Lymphoma, B-Cell/enzymology , Phosphorylation , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/physiology , Tumor Cells, Cultured , src Homology Domains
2.
Science ; 267(5199): 886-91, 1995 Feb 10.
Article in English | MEDLINE | ID: mdl-7531365

ABSTRACT

B-cell precursor (BCP) leukemia is the most common form of childhood cancer and the second most common form of acute leukemia in adults. Human BCP leukemia was treated in a severe combined immunodeficient mouse model by targeting of the tyrosine kinase inhibitor Genistein (Gen) to the B cell-specific receptor CD19 with the monoclonal antibody B43. The B43-Gen immunoconjugate bound with high affinity to BCP leukemia cells, selectively inhibited CD19-associated tyrosine kinases, and triggered rapid apoptotic cell death. At less than one-tenth the maximum tolerated dose more than 99.999 percent of human BCP leukemia cells were killed, which led to 100 percent long-term event-free survival from an otherwise invariably fatal leukemia. The B43-Gen immuno-conjugate might be useful in eliminating leukemia cells in patients who have failed conventional therapy.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Immunoconjugates/therapeutic use , Isoflavones/therapeutic use , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antibodies, Monoclonal , Antigens, CD19 , Apoptosis , DNA Damage , DNA, Neoplasm/metabolism , Genistein , Immunoconjugates/administration & dosage , Immunoconjugates/pharmacokinetics , Isoflavones/administration & dosage , Isoflavones/pharmacokinetics , Leukemic Infiltration , Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Mice , Mice, SCID , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tissue Distribution , Tumor Cells, Cultured
3.
J Clin Invest ; 91(3): 1044-51, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8450034

ABSTRACT

The radiation sensitivity of primary clonogenic blasts from 44 children with newly diagnosed B-cell precursor acute lymphoblastic leukemia (ALL) was analyzed using leukemic progenitor cell (LPC) colony assays. The derived values for SF2 (surviving fraction at 200 cGy) and alpha (initial slope of radiation survival curves constructed according to the linear quadratic model) indicated a marked interpatient heterogeneity in intrinsic radiation sensitivity of LPC populations. The SF2 values ranged from 0.01 to 1.00 (median = 0.430; mean +/- SE = 0.47 +/- 0.04), and the alpha values ranged from 0.000 to 3.272 Gy-1 (median = 0.280 Gy-1; mean +/- SE = 0.430 +/- 0.093 Gy-1). When CD19+ CD34+ versus CD19+ CD34- immunophenotypes were compared, a trend toward higher SF2 and lower alpha values were observed in LPC from CD34+ patients, consistent with greater radiation resistance. When patients were divided into three approximately equal groups based on increasing levels of CD34 expression, a clear ordering effect was observed indicating that increased CD34 expression levels are associated with significantly higher radiation resistance at the level of B-lineage LPC. The highest CD34 expression group (> or = 75% positivity) had 1.4-fold higher SF2 (P = 0.05) and twofold lower alpha values (P = 0.06) than the lowest group (< 30% positivity). Furthermore, the CD34 positivity of radiation resistant (alpha < or = 0.2 and SF2 > or = 0.5) B-cell precursor ALL cases was greater than the CD34 positivity of radiation sensitive (alpha > 0.2 and/or SF2 < 0.5) cases (56 +/- 9% versus 34 +/- 9%, P = 0.09). Whereas only 6 of 16 (38%) of radiation sensitive cases were CD34+, 11 of 15 (73%) of radiation resistant cases expressed CD34 (P = 0.04). Our results offer new insights into the inherent and/or acquired radiation resistance of primary clonogenic blasts from B-cell precursor ALL patients.


Subject(s)
B-Lymphocytes/radiation effects , Blast Crisis/pathology , Bone Marrow/radiation effects , Burkitt Lymphoma/pathology , Hematopoietic Stem Cells/radiation effects , Age Factors , Antigens, CD/analysis , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Blast Crisis/immunology , Bone Marrow/pathology , Burkitt Lymphoma/immunology , Child , Child, Preschool , Female , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Immunophenotyping , Male , Tumor Stem Cell Assay
4.
Clin Cancer Res ; 3(3): 347-55, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9815691

ABSTRACT

Multiagent chemotherapy regimens fail to cure more than one-half of the patients with acute myeloid leukemia (AML) because of the emergence of dominant multidrug-resistant subclones of leukemia cells. We have developed a recombinant diphtheria toxin-human granulocyte macrophage colony-stimulating factor chimeric fusion protein (DTctGMCSF) that specifically targets GMCSF receptor-positive AML cells. This novel biotherapeutic agent induced rapid apoptotic cell death of chemotherapy-resistant AML cell lines and primary leukemic cells from treatment-refractory AML patients. Our results suggest that DTctGMCSF may be useful in the treatment of AML patients whose leukemia has recurred and developed resistance to contemporary chemotherapy programs.


Subject(s)
Apoptosis/physiology , Diphtheria Toxin/toxicity , Drug Resistance, Multiple , Granulocyte-Macrophage Colony-Stimulating Factor/toxicity , Immunotoxins/toxicity , Radiation Tolerance , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Recombinant Fusion Proteins/toxicity , Acute Disease , Apoptosis/drug effects , DNA Fragmentation/drug effects , HL-60 Cells , Humans , Leukemia, Myeloid , Leukemia, Myeloid, Acute , Tumor Cells, Cultured
5.
Clin Cancer Res ; 4(4): 901-12, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9563884

ABSTRACT

The receptor (R) for epidermal growth factor (EGF) is expressed at high levels on human breast cancer cells and associates with ErbB2, ErbB3, and Src proto-oncogene family protein tyrosine kinases (PTKs) to form membrane-associated PTK complexes with pivotal signaling functions. Recombinant human EGF was conjugated to the soybean-derived PTK inhibitor genistein (Gen) to construct an EGF-R-directed cytotoxic agent with PTK inhibitory activity. The EGF-Gen conjugate was capable of binding to and entering EGF-R-positive MDA-MB-231 and BT-20 breast cancer cells (but not EGF-R-negative NALM-6 or HL-60 leukemia cells) via its EGF moiety, and it effectively competed with unconjugated EGF for target EGF-R molecules in ligand binding assays. EGF-Gen inhibited the EGF-R tyrosine kinase in breast cancer cells at nanomolar concentrations, whereas the IC50 for unconjugated Gen was >10 microM. Notably, EGF-Gen triggered a rapid apoptotic cell death in MDA-MB-231 as well as BT-20 breast cancer cells at nanomolar concentrations. The EGF-Gen-induced apoptosis was EGF-R-specific because cells treated with the control granulocyte-colony stimulating factor-Gen conjugate did not become apoptotic. Apoptosis was dependent both on the PTK inhibitory function of Gen and the targeting function of EGF, because cells treated with unconjugated Gen plus unconjugated EGF did not undergo apoptosis. The IC50s of EGF-Gen versus unconjugated Gen against MDA-MB-231 and BT-20 cells in clonogenic assays were 30 +/- 3 nM versus 120 +/- 18 microM (P < 0.001) and 30 +/- 10 nM versus 112 +/- 17 microM (P < 0.001), respectively. Thus, the EGF-Gen conjugate is a >100-fold more potent inhibitor of EGF-R tyrosine kinase activity in intact breast cancer cells than unconjugated Gen and a >100-fold more potent cytotoxic agent against EGF-R+ human breast cancer cells than unconjugated Gen. Taken together, these results indicate that the EGF-R-associated PTK complexes have vital antiapoptotic functions in human breast cancer cells and may therefore be used as therapeutic targets.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/therapy , Epidermal Growth Factor/pharmacology , Genistein/pharmacology , Protein-Tyrosine Kinases/drug effects , Aged , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , DNA Fragmentation/drug effects , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/metabolism , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Female , Genistein/chemistry , Genistein/metabolism , Humans , Middle Aged , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Mas , Tumor Cells, Cultured/drug effects
6.
Exp Hematol ; 21(6): 791-8, 1993 Jun.
Article in English | MEDLINE | ID: mdl-7684704

ABSTRACT

We examined bone marrow and peripheral blood specimens from pediatric acute lymphoblastic leukemia (ALL) patients after autologous bone marrow transplantation (BMT) as well as fetal lymphohematopoietic tissues for the presence of CD5+ B lymphocytes. CD5+ B lymphocytes represented 23.6 +/- 0.7% of CD19+ fetal spleen B lineage lymphoid cells, 24.2 +/- 2.5% of CD19+ fetal bone marrow B lineage lymphoid cells and 18.1 +/- 1.7% of CD19+ fetal liver B lineage lymphoid cells. By comparison, in normal pediatric bone marrow samples, only 1.5 +/- 0.3% of lymphoid cells and 4.3 +/- 0.2% of CD19+ B lineage lymphoid cells expressed CD5 antigen. Similarly, very few CD5+CD19+ cells (< or = 2% of lymphoid cells) were found in day 30 and day 100 post-BMT bone marrow or peripheral blood specimens from B lineage ALL patients undergoing autologous BMT using autografts purged ex vivo with B43(anti-CD19)-PAP immunotoxin plus 4-hydroperoxycyclophosphamide (4-HC). Two bone marrow samples that were obtained and analyzed 1 year post-BMT contained only 2 to 4% CD5+CD19+ cells, accounting for 5 to 7% of the total CD19+ population. The fraction of CD5+ B lymphocytes in post-BMT bone marrow samples was not greater than the fraction of CD5+ B lymphocytes in normal healthy bone marrow.


Subject(s)
Antigens, CD/analysis , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , Embryonic and Fetal Development/immunology , Antibodies, Monoclonal , Antigens, CD19 , Antigens, Differentiation, B-Lymphocyte/analysis , B-Lymphocytes/physiology , Bone Marrow/embryology , Bone Marrow/immunology , Bone Marrow Cells , CD5 Antigens , Cell Differentiation/physiology , Fetus/cytology , Fetus/immunology , Flow Cytometry , Hematopoiesis/physiology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/surgery , Spleen/cytology , Spleen/embryology , Spleen/immunology , Transplantation, Autologous
7.
Biochem Pharmacol ; 56(11): 1411-26, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9827573

ABSTRACT

Modulation of Ras function may provide a novel means by which cancer cells with oncogenic mutations can be sensitized to chemotherapeutic or radiotherapeutic regimens. Moreover, cancer cells without ras oncogene mutations can also be eliminated by compounds that interfere with the mevalonate pathway, which is more fundamental to mitogenesis because it allows the synthesis of sterol and nonsterol lipids and without which many Ras-related proteins and nuclear lamins would not be prenylated and functional.


Subject(s)
Genes, ras , Neoplasms/genetics , Neoplasms/therapy , ras Proteins/metabolism , Animals , Cell Death , Humans , Models, Biological , Mutation , Neoplasms/pathology , Signal Transduction
8.
Biochem Pharmacol ; 57(1): 9-17, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9920280

ABSTRACT

Nuclear factor-kappaB (NF-kappaB) activity affects cell survival and determines the sensitivity of cancer cells to cytotoxic agents as well as to ionizing radiation. Preventing the protective function of NF-kappaB may result in chemo- and radio-sensitization of cancer cells. Therefore, NF-kappaB has emerged as one of the most promising molecular targets in rational drug design efforts of translational cancer research programs.


Subject(s)
Gene Expression Regulation , NF-kappa B/metabolism , Neoplasms/therapy , Animals , Cell Survival/drug effects , Cell Survival/radiation effects , Drug Design , Gene Expression Regulation, Viral , Humans , Models, Biological , Radiation, Ionizing , Signal Transduction , Transforming Growth Factor beta/physiology
9.
Radiat Res ; 136(3): 313-9, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7506428

ABSTRACT

As presently reported, both ionizing radiation and engagement of the CD19 receptor are capable of inducing apoptosis in B-lineage acute lymphoblastic leukemia (ALL) cells. In both instances, activation of tyrosine kinases appears to be a proximal and mandatory step, since it can be prevented by the tyrosine kinase inhibitor genistein. This common biochemical signaling pathway involves the rapid activation of the Src family tyrosine kinase LCK (p56lck), which is physically associated with the CD19 receptor, and enhanced tyrosine phosphorylation of multiple substrates leading to stimulation of phosphoinositide turnover, and activation of protein kinase C. Importantly, engagement of the CD19 receptor promoted radiation-induced apoptosis in radiation-resistant B-lineage ALL cells in a cell type-specific fashion. Our results prompt the hypothesis that clonogenic B-lineage ALL blasts with an inherent or acquired resistance to radiation could be radiosensitized in clinical settings using anti-CD19 MoAb B43 or its homoconjugate as adjuncts.


Subject(s)
Antigens, CD/physiology , Antigens, Differentiation, B-Lymphocyte/physiology , Apoptosis/radiation effects , Burkitt Lymphoma/pathology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Antigens, CD19 , Burkitt Lymphoma/enzymology , Enzyme Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Radiation Tolerance , Tumor Cells, Cultured
10.
Leuk Lymphoma ; 25(1-2): 77-90, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9130616

ABSTRACT

We examined the effects of recombinant human granulocyte colony-stimulating factor (rhG-CSF), interleukin 3 (rhIL-3) and interleukin 6 (rhIL-6) on the radiation sensitivity of normal and leukemic bone marrow progenitor cell populations. Conditioning of leukemic progenitor cells (LPC) from acute lymphoblastic leukemia (ALL) patients with rhG-CSF enhanced their radiation sensitivity, whereas conditioning with rhIL-3 or rhIL-6 had the opposite effect. In contrast to its effects on LPC derived from ALL patients, rhG-CSF reduced the radiation sensitivity of normal myeloid progenitor cells as well as LPC from acute myeloblastic leukemia (AML) patients. Differential modulation of the radiation sensitivity of LPC by rhG-CSF may provide the basis for better total body irradiation (TBI) regimens for ALL patients undergoing autologous bone marrow transplantation (BMT).


Subject(s)
Bone Marrow/radiation effects , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cells/radiation effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Bone Marrow/drug effects , Cell Survival/radiation effects , Cells, Cultured , Combined Modality Therapy , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/radiotherapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Radiation-Protective Agents/pharmacology , Recombinant Proteins/therapeutic use , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects
11.
Leuk Lymphoma ; 19(1-2): 121-8, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8574156

ABSTRACT

The ability of total body irradiation (TBI) to eradicate clonogenic leukemia cells from B-lineage acute lymphoblastic leukemia patients prior to bone marrow transplantation (BMT) is greatly hampered by their inherent or acquired radiation resistance. The radiorefractory nature of these cells is believed to contribute to the high relapse rate subsequent to TBI and BMT in patients with B-lineage acute lymphoblastic leukemia (ALL). A method by which clonogenic leukemia cells could be radiosensitized in vivo could be clinically beneficial. In the present study, we used a highly radiation resistant subclone of the murine B-lineage leukemia cell line BCL-1 in a syngeneic BMT model to investigate if any of the B-cell stimulatory cytokines interleukin 2, interleukin 4, interleukin 5, or interleukin 6 could have radiosensitizing effects. All untreated BALB/c mice (N = 33) inoculated with 1 x 10(6) BCL-1 cells died of disseminated leukemia within 24 days with a median survival of 13.3 days. TBI (700 cGy = LD100/30 for BALB/c mice) followed by syngeneic BMT (N = 70) extended the median survival to 23.6 days (P < 0.001 by log-rank test). A single intraperitoneal bolus injection of 100 ng, 500 ng, or 2500 ng recombinant murine interleukin 6(rmIL-6) 2-4 hours before TBI extended the median survival to 32.5 days, 31.0 days, and 30.5 days, respectively (P < 0.01 by log-rank test for all dose groups). The improved survival was not due to any direct anti-leukemic activity of rmIL-6 and all control BALB/c mice (N = 15) that received the same doses of rmIL-6 but did not undergo TBI and BMT died of BCL-1 leukemia within 28 days with a median survival of 13.6 days. In contrast to rmIL-6, recombinant murine interleukin 5 (rmIL-5) had minimal radiosensitizing effects.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Apoptosis , Bone Marrow Transplantation , Interleukin-6/pharmacology , Leukemia, B-Cell/therapy , Leukemia, Experimental/therapy , Radiation-Sensitizing Agents/pharmacology , Animals , Female , Humans , Leukemia, B-Cell/pathology , Leukemia, B-Cell/physiopathology , Mice , Mice, Inbred BALB C , Recombinant Proteins/pharmacology , Time Factors , Transplantation, Isogeneic , Whole-Body Irradiation
12.
Leuk Lymphoma ; 20(5-6): 417-26, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8833397

ABSTRACT

Here we provide experimental evidence that ionizing radiation induces inhibitory tyrosine phosphorylation of the p34cdc2 kinase in human leukemic B-cell precursors. Herbimycin A markedly reduced tyrosine phosphorylation of p34cdc2 in irradiated leukemic B-cell precursors, thereby preventing radiation-induced cell cycle arrest at the G2-M transition checkpoint. Thus, tyrosine phosphorylation is directly responsible for the inactivation of p34cdc2 in irradiated human leukemic B-cell precursors and activation of protein tyrosine kinases is a proximal and mandatory step in radiation-induced G2-arrest arrest at the G2-M checkpoint. Human WEE1 kinase isolated from unirradiated or irradiated leukemic B-cell precursors had minimal tyrosine kinase activity towards p34cdc2. We detected no increase of human WEE1 kinase activity after radiation of leukemic B-cell precursors, as measured by (a) autophosphorylation, (b) tyrosine phosphorylation of a synthetic peptide derived from the p34cdc2 amino-terminal region or (c) recombinant human p34cdc2-cyclin B complex. Thus the signaling pathway leading to inhibitory tyrosine phosphorylation of p34cdc2 and G2-arrest in irradiated human leukemic B-cell precursors functions independent of p49 WEE1 HU and enzymes which augment the tyrosine kinase activity of p49 WEE 1HU.


Subject(s)
B-Lymphocytes/radiation effects , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins , G2 Phase/radiation effects , Hematopoietic Stem Cells/radiation effects , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/radiation effects , Nuclear Proteins , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Processing, Post-Translational/radiation effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Benzoquinones , CDC2 Protein Kinase/antagonists & inhibitors , Enzyme Activation/radiation effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Lactams, Macrocyclic , Macromolecular Substances , Maturation-Promoting Factor/physiology , Neoplasm Proteins/antagonists & inhibitors , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Peptide Fragments/metabolism , Phosphorylation/radiation effects , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Protein-Tyrosine Kinases/physiology , Quinones/pharmacology , Recombinant Fusion Proteins/metabolism , Rifabutin/analogs & derivatives , Tumor Cells, Cultured , Tyrosine/metabolism
13.
Leuk Lymphoma ; 25(3-4): 247-56, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9168435

ABSTRACT

Contemporary therapies for acute myeloid leukemia (AML) commonly fail to cure patients because of the emergence of drug resistance. Drug resistance in AML is multifactorial but can be associated with the overexpression of transmembrane transporter molecules, including P-glycoprotein (Pgp) or the multidrug resistance-associated protein (MRP), or associated with inactivation of the p53 tumor suppressor gene, as well as overexpression of the anti-apoptotic protein bcl-2. We are investigating if novel recombinant biotherapeutics can circumvent these resistance mechanisms to effectively treat refractory AML. To target the lethal action of diphtheria toxin (DT) to high affinity granulocyte-macrophage colony-stimulating factor (GMCSF) receptors on AML blasts, we have produced a recombinant chimeric fusion toxin, DTctGMCSF. Since DTctGMCSF enters and kills its target cells by unique mechanisms (GMCSF-receptor binding and protein synthesis inhibition) and is not similar in structure to Pgp or MRP substrates, we postulated that it would be an active agent against therapy-resistant AML. DTctGMCSF was selectively cytotoxic (IC50 1-10ng/ml) to GMCSF-receptor positive AML cells expressing the Pgp- or MRP-associated multi-drug resistant phenotypes, despite high level resistance to conventional chemotherapeutic agents. DTctGMCSF also efficiently killed AML cells deficient in p53 expression, as well as radiation-resistant AML cells and mixed lineage leukemia cells expressing high levels of bcl-2. In addition, DTctGMCSF killed > 99% of primary leukemic progenitor cells from therapy-refractory AML patients under conditions that we have previously found to not adversely affect the proliferative capacity or differentiation of pluripotent normal hematopoietic progenitor cells. DTctGMCSF may prove useful in treating myeloid leukemias that are otherwise resistant to a wide range of conventional therapies.


Subject(s)
Diphtheria Toxin/pharmacology , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/radiotherapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , Recombinant Fusion Proteins/pharmacology , Antineoplastic Agents/pharmacology , Diphtheria Toxin/genetics , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Leukemia, Myeloid/pathology , Radiation Tolerance , Recombinant Fusion Proteins/genetics , Stem Cells/drug effects , Tumor Cells, Cultured
14.
Leuk Lymphoma ; 25(3-4): 257-70, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9168436

ABSTRACT

Human granulocyte-macrophage colony stimulating factor (GMCSF) and its high affinity receptor function to regulate the proliferation and differentiation of myeloid lineage hematopoietic cells, and may participate in the pathogenesis of many malignant myeloid diseases. We have used genetic engineering based on the elucidated molecular structures of human granulocyte-macrophage colony-stimulating factor and diphtheria toxin (DT) to produce a recombinant fusion toxin, DTctGMCSF, that targets diphtheria toxin to high affinity GMCSF receptors expressed on the surface of blast cells from a large fraction of patients with acute myeloid leukemia (AML). DTctGMCSF was specifically immunoreactive with antidiphtheria toxin and anti-GMCSF antiseras, and exhibited the characteristic catalytic activity of diphtheria toxin, catalyzing the in vitro ADP-ribosylation of purified elongation factor 2. The cytotoxic effects of DTctGMCSF were examined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-tetrazolium (MTT) bromide assay of cell viability and in vivo assays of protein synthesis inhibition. DTctGMCSF were specifically cytotoxic to human leukemia cell lines bearing high affinity receptors for human GMCSF with IC50 of 10(-9) to 10(-11) M. It was not toxic to mammalian hematopoietic cell lines lacking human GMCSF (hGMCSF) receptors. In receptor positive cells, cytotoxicity can be specifically blocked by a large excess of hGMCSF, confirming that its cytotoxicity is mediated through the hGMCSF receptor. THough DTctGMCSF inhibited granulocyte-macrophage colony formation by committed myeloid progenitor cells (CFU-GM), it did not significantly affect erythroid burst formation by committed erythroid progenitor cells (BFU-E), or mixed granulocyte-erythroid-macrophage-megakaryocyte colony formation by pluripotent multilineage progenitor cells (CFU-GEMM). DTctGMCSF holds promise for the treatment of myeloid lineage malignancies, and is a useful reagent to study hematopoiesis.


Subject(s)
Diphtheria Toxin/pharmacology , Leukemia/drug therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Antineoplastic Agents/pharmacology , Bone Marrow/drug effects , Bone Marrow Cells , Cell Division/drug effects , Diphtheria Toxin/genetics , Diphtheria Toxin/immunology , Drug Screening Assays, Antitumor , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Immune Sera , Leukemia/genetics , Leukemia/pathology , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/pathology , Mice , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Recombinant Fusion Proteins/genetics , Stem Cells/drug effects , Tumor Cells, Cultured
16.
Blood ; 77(11): 2364-71, 1991 Jun 01.
Article in English | MEDLINE | ID: mdl-1710151

ABSTRACT

The purpose of the present study was to evaluate and compare the in vivo radioprotective effects of pre-total body irradiation (TBI) conditioning with recombinant granulocyte colony-stimulating factor (rG-CSF) and recombinant granulocyte-macrophage CSF (rGM-CSF) in a large series of lethally and supralethally irradiated mice. Also analyzed were the radioprotective effects of simultaneous as well as sequential combinations of rG-CSF and rGM-CSF. Our findings in 1,180 mice provide direct evidence that in vivo administration of rG-CSF or rGM-CSF before TBI protects a significant fraction of mice from the lethal effects of LD100/30 TBI. At equivalent doses, rG-CSF displayed a more potent radioprotective activity than rGM-CSF. Not only was rG-CSF radioprotective at much smaller doses than rGM-CSF, the survival rate after lethal TBI was also significantly higher in mice receiving optimally radioprotective doses of rG-CSF as compared with mice receiving optimally radioprotective doses of rGM-CSF. Pretreatment of mice with rGM-CSF markedly attenuated the radioprotective affects of rG-CSF in lethally as well as supralethally irradiated mice. Pretreatment with rG-CSF followed by rGM-CSF was slightly more effective than rG-CSF alone in supralethally irradiated mice but not in lethally irradiated mice. Notably, marked differences among different strains of mice were noted regarding the optimally radioprotective doses of rG-CSF or rGM-CSF as well as probability of survival and median survival time after lethal or supralethal TBI. This report confirms and extends previous studies concerning the potential of cytokines in prevention or therapy of lethal radiation injury.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Radiation-Protective Agents , Whole-Body Irradiation , Animals , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Interactions , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Recombinant Proteins/pharmacology , Time Factors
17.
Blood ; 75(3): 638-45, 1990 Feb 01.
Article in English | MEDLINE | ID: mdl-1688721

ABSTRACT

The purpose of this study was to investigate the in vivo radioprotective effects of recombinant human granulocyte colony stimulating factor (rhG-CSF) in lethally irradiated BALB/c mice. We initially analyzed the effects of increasing doses of rhG-CSF on survival of mice receiving 700 cGy (LD100/30) single dose total body irradiation (TBI). While 1 microgram/kg to 100 micrograms/kg doses of rhG-CSF were not radioprotective, a dose-dependent radioprotection was observed at 200 micrograms/kg to 4,000 micrograms/kg rhG-CSF. We next compared four different rhG-CSF treatment regimens side by side for their radioprotective effects in LD100/30 irradiated mice. One hundred percent of control mice receiving phosphate buffered saline died within 21 days after TBI with a median survival of 14 days. The median survival was prolonged to 20 days and the actuarial 60-day survival rate was increased to 27% when mice received 2,000 micrograms/kg rhG-CSF 24 hours before TBI (P = .0002; Mantel-Peto-Cox). Similarly, the median survival time was prolonged to 24 days and the actuarial 60-day survival rate was increased to 33%, when mice were given 2,000 micrograms/kg rhG-CSF 30 minutes before TBI. Optimal radioprotection was achieved when 2,000 micrograms/kg rhG-CSF was administered in two divided doses of 1,000 micrograms/kg given 24 hours before and 1,000 micrograms/kg given 30 minutes before TBI. This regimen prolonged the median survival time of LD100/30 irradiated mice to more than 60 days and increased the actuarial 60-day survival rate to 62% (P = .0001; Mantel-Peto-Cox). By comparison, no survival advantage was observed when mice received rhG-CSF 24 hours post-TBI. Similar radioprotective effects were observed when mice were irradiated with 650 cGy (LD80/30). The presented findings provide conclusive evidence that rhG-CSF has significant in vivo radioprotective effects for mice receiving LD100/30 or LD80/30 TBI.


Subject(s)
Colony-Stimulating Factors/pharmacology , Hematopoiesis/drug effects , Radiation-Protective Agents , Animals , Bone Marrow/pathology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Granulocyte Colony-Stimulating Factor , Mice , Recombinant Proteins , Survival Analysis , Time Factors
18.
J Biol Chem ; 271(11): 6389-97, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8626437

ABSTRACT

Exposure of human B-cell precursors (BCP) to ionizing radiation results in cell cycle arrest at the G2-M checkpoint as a result of inhibitory tyrosine phosphorylation of p34cdc2 . Here, we show that ionizing radiation promotes physical interactions between p34cdc2 and the Src family protein-tyrosine kinase Lyn in the cytoplasm of human BCP leading to tyrosine phosphorylation of p34cdc2. Lyn kinase immunoprecipitated from lysates of irradiated BCP as well as a full-length glutathione S-transferase (GST)-Lyn fusion protein-phosphorylated recombinant human p34cdc2 on tyrosine 15. Furthermore, Lyn kinase physically associated with and tyrosine-phosphorylated p34cdc2 kinase in vivo when co-expressed in COS-7 cells. Binding experiments with truncated GST-Lyn fusion proteins suggested a functional role for the SH3 rather than the SH2 domain of Lyn in Lyn-p34cdc2 interactions in BCP. The first 27 residues of the unique amino-terminal domain of Lyn were also essential for the ability of GST-Lyn fusion proteins to bind to p34cdc2 from BCP lysates. Ionizing radiation failed to cause tyrosine phosphorylation of p34cdc2 or G2 arrest in Lyn kinase-deficient BCP, supporting an important role of Lyn kinase in radiation-induced G2 phase-specific cell cycle arrest. Our findings implicate Lyn as an important cytoplasmic suppressor of p34cdc2 function.


Subject(s)
B-Lymphocytes/enzymology , CDC2 Protein Kinase/metabolism , src-Family Kinases/metabolism , Amino Acid Sequence , Animals , B-Lymphocytes/cytology , B-Lymphocytes/radiation effects , Binding Sites/genetics , CDC2 Protein Kinase/chemistry , CDC2 Protein Kinase/genetics , DNA Repair , G2 Phase/radiation effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/radiation effects , Humans , In Vitro Techniques , Molecular Sequence Data , Phosphorylation , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tyrosine/chemistry , Tyrosine/radiation effects , src-Family Kinases/chemistry , src-Family Kinases/genetics
19.
Proc Natl Acad Sci U S A ; 92(21): 9575-9, 1995 Oct 10.
Article in English | MEDLINE | ID: mdl-7568175

ABSTRACT

CD19 receptor is expressed at high levels on human B-lineage lymphoid cells and is physically associated with the Src protooncogene family protein-tyrosine kinase Lyn. Recent studies indicate that the membrane-associated CD19-Lyn receptor-enzyme complex plays a pivotal role for survival and clonogenicity of immature B-cell precursors from acute lymphoblastic leukemia patients, but its significance for mature B-lineage lymphoid cells (e.g., B-lineage lymphoma cells) is unknown. CD19-associated Lyn kinase can be selectively targeted and inhibited with B43-Gen, a CD19 receptor-specific immunoconjugate containing the naturally occurring protein-tyrosine kinase inhibitor genistein (Gen). We now present experimental evidence that targeting the membrane-associated CD19-Lyn complex in vitro with B43-Gen triggers rapid apoptotic cell death in highly radiation-resistant p53-Bax- Ramos-BT B-lineage lymphoma cells expressing high levels of Bcl-2 protein without affecting the Bcl-2 expression level. The therapeutic potential of this membrane-directed apoptosis induction strategy was examined in a scid mouse xenograft model of radiation-resistant high-grade human B-lineage lymphoma. Remarkably, in vivo treatment of scid mice challenged with an invariably fatal number of Ramos-BT cells with B43-Gen at a dose level < 1/10 the maximum tolerated dose resulted in 70% long-term event-free survival. Taken together, these results provide unprecedented evidence that the membrane-associated anti-apoptotic CD19-Lyn complex may be at least as important as Bcl-2/Bax ratio for survival of lymphoma cells.


Subject(s)
Antigens, CD19/metabolism , Apoptosis , Burkitt Lymphoma/metabolism , src-Family Kinases/metabolism , Animals , Antigens, CD19/immunology , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/immunology , Burkitt Lymphoma/pathology , Enzyme Inhibitors/therapeutic use , Genistein , Humans , Immunoconjugates/therapeutic use , Immunotoxins/therapeutic use , Isoflavones/therapeutic use , Mice , Mice, SCID , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Binding , Protein Conformation , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53
20.
Blood ; 85(10): 2817-28, 1995 May 15.
Article in English | MEDLINE | ID: mdl-7742543

ABSTRACT

Topotecan [(S)-9-dimethylaminomethyl-10-hydroxycamptothecin hydrochloride; SK&F 104864-A, NSC 609699], a water soluble semisynthetic analogue of the alkaloid camptothecin, is a potent topoisomerase I inhibitor. Here we show that topotecan stabilizes topoisomerase I/DNA cleavable complexes in radiation-resistant human B-lineage acute lymphoblastic leukemia (ALL) cells, causes rapid apoptotic cell death despite high-level expression of bcl-2 protein, and inhibits ALL cell in vitro clonogenic growth in a dose-dependent fashion. Furthermore, topotecan elicited potent antileukemic activity in three different severe combined immunodeficiency (SCID) mouse models of human poor prognosis ALL and markedly improved event-free survival of SCID mice challenged with otherwise fatal doses of human leukemia cells at systemic drug exposure levels that can be easily achieved in children with leukemia.


Subject(s)
Burkitt Lymphoma/drug therapy , Camptothecin/analogs & derivatives , Adolescent , Adult , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Apoptosis , Camptothecin/pharmacokinetics , Camptothecin/therapeutic use , Child , Child, Preschool , DNA Damage , Female , Humans , In Vitro Techniques , Male , Mice , Mice, SCID , Neoplasm Transplantation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Survival Analysis , Topoisomerase I Inhibitors , Topotecan
SELECTION OF CITATIONS
SEARCH DETAIL