Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cancer Chemother Pharmacol ; 43(3): 263-8, 1999.
Article in English | MEDLINE | ID: mdl-9923558

ABSTRACT

PURPOSE: The alkylating agent cyclophosphamide (CP) is a prodrug that is metabolized to both cytotoxic and inactive compounds. We have previously shown that following dose escalation from conventional-dose (CD) to high-dose (HD) levels; the fraction of the dose cleared by bioactivation is significantly decreased (66% versus 48.5%) in favor of inactivating elimination pathways when the HD is given as a single 1-h infusion. Based on the concept of bioactivating enzyme saturation with increasing doses, we investigated the influence of fractionated application of HD-CP on dose-dependent changes in metabolism. PATIENTS AND METHODS: Plasma concentrations of CP (measured by high-performance liquid chromatography, HPLC) and urinary concentrations of CP and its major metabolites (quantified by [31P]-nuclear magnetic resonance spectroscopy; [31P]-NMR spectroscopy), were determined in four patients with high-risk primary breast cancer who received adjuvant chemotherapy including both CD-CP (500 mg/ m2 infused over 1 h) and split HD-CP (50 mg/kg infused over 1 h on each of 2 consecutive days (d): d1 and d2. RESULTS: (Data are given as mean values for CD and d1/d2 of HD, respectively). Systemic clearance (CL) of CP was similar during CD and d1 of HD, but significantly increased on d2 of HD (CL: 83 and 78/115 ml/min; P < 0.01 for d1 versus d2). The latter was translated into an increase in formation CL of both active (+ 16.4 ml/min) and inactive metabolites (+ 17.6 ml/ min) and reflects autoinduction of metabolism. As compared with CD-CP, no statistically significant decrease was observed in the relative contribution of bioactivation CL to overall CL during both days of HD (63% versus 57%/53%). Recovery of intact CP in 24-h urine corresponded to 24%, 29%, 22% of the dose (P < 0.05 for d1 versus d2 of HD). CONCLUSIONS: Following dose escalation of CP, dividing the high dose over 2 days instead of one single infusion may favorably impact the metabolism of CP in terms of bioactivation. In addition, on day 2 of a split regimen, renal elimination of CP is decreased, which implies that more drug is available for metabolism.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Cyclophosphamide/pharmacokinetics , Adult , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/therapeutic use , Area Under Curve , Breast Neoplasms/drug therapy , Chromatography, High Pressure Liquid , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Male , Middle Aged
2.
J Clin Oncol ; 15(5): 1885-96, 1997 May.
Article in English | MEDLINE | ID: mdl-9164199

ABSTRACT

PURPOSE: The alkylating anticancer agent cyclophosphamide (CP) is a prodrug that undergoes a complex metabolism in humans producing both active and inactive metabolites. In parallel, unchanged CP is excreted via the kidneys. The aim of this study was to investigate the influence of dose escalation on CP pharmacokinetics and relative contribution of activating and inactivating elimination pathways. PATIENTS AND METHODS: Pharmacokinetics of CP were assessed in 12 patients with high-risk primary breast cancer who received an adjuvant chemotherapy regimen that included four courses of conventional-dose CP (500 mg/m2 over 1 hour every 3 weeks) followed by one final course of high-dose CP (100 mg/kg over 1 hour). Plasma concentrations of CP were analyzed by high-performance liquid chromatography (HPLC), 24-hour urinary concentrations of CP, and its inactive metabolites (carboxyphosphamide, dechloroethylcyclophosphamide [dechlorethylCP], ketocyclophosphamide [ketoCP]) were determined by 31-phosphorus-nuclear magnetic resonance (31P-NMR)-spectroscopy. RESULTS: There was no difference in dose-corrected area under the concentration-time curve (AUC) (216 v 223 [mumol.h/[mL.g]), elimination half-life (4.8 v 4.8 hours), systemic clearance (79 v 77 mL/min) and volume of distribution (0.49 v 0.45 L/kg) of CP between conventional- and high-dose therapy, respectively. However, during high-dose chemotherapy, we observed a significant increase in the renal clearance of CP (15 v 23 mL/min; P < .01) and in the formation clearance of carboxyphosphamide (7 v 12 mL/min; P < .05) and dechloroethylCP (3.2 v 4.2 mL/min; P < .05), whereas metabolic clearance to ketoCP remained unchanged (1.3 v 1.2 mL/min). Consequently, metabolic clearance to the remaining (reactive) metabolites decreased from 52 to 38 mL/min (P < .001). The relative contribution of the different elimination pathways to overall clearance of CP demonstrated wide interindividual variability. CONCLUSION: Overall pharmacokinetics of CP are apparently not affected during eightfold dose escalation. However, there is a shift in the relative contribution of different clearances to systemic CP clearance in favor of inactivating elimination pathways, thereby indicating saturation of bioactivating enzymes during dose escalation. Besides individual enzyme capacity, hydration and concomitant medication with dexamethasone modulated CP disposition.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/pharmacokinetics , Breast Neoplasms/metabolism , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacokinetics , Adult , Antineoplastic Agents, Alkylating/blood , Antineoplastic Agents, Alkylating/urine , Breast Neoplasms/drug therapy , Cyclophosphamide/blood , Cyclophosphamide/urine , Female , Humans , Middle Aged
3.
Bone Marrow Transplant ; 19(7): 653-9, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9156241

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is considered to play a pivotal role in hemopoietic regulation. Its pharmacological application is reported to shorten chemotherapy-induced neutropenia as well as time to engraftment in patients after bone marrow transplantation (BMT). In order possibly to establish further rationale for G-CSF treatment strategies in patients undergoing BMT, we evaluated G-CSF plasma levels of 89 patients after allogeneic BMT for chronic myeloid leukemia (CML). EDTA anti-coagulated plasma samples were collected starting on day -1 (before grafting) and thereafter twice weekly for four consecutive weeks. G-CSF levels were estimated by enzyme immunoassay. Patients with late (> 30 days) bone marrow engraftment had consistently higher G-CSF levels at day +1 (after grafting) compared to patients with early (< or = 30 days) engraftment, while all patients had low plasma levels on day -1/0. Mean G-CSF plasma levels and time to engraftment were correlated (r = 0.79). In univariate analyses, high G-CSF levels at days +1, +4, +7, +10 and several clinical variables (such as TBI, unrelated donor transplant, state of disease) were predictive of late engraftment. Further analysis by multivariate Cox regression resulted in the following predictive model: high G-CSF plasma levels at day +7 and +10 (after grafting), in combination with a blastic phase of the disease were highly predictive of late engraftment. The significantly higher G-CSF levels in patients with impaired engraftment may reflect early compensating mechanisms of the hemopoietic system, which should be investigated further.


Subject(s)
Bone Marrow Transplantation , Graft Survival , Granulocyte Colony-Stimulating Factor/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Adolescent , Adult , Age Factors , Child , Child, Preschool , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Predictive Value of Tests , Sex Factors , Transplantation, Homologous
4.
Blood ; 86(5): 1916-23, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7544646

ABSTRACT

The class I receptor tyrosine kinase (RTK) HER2 is an oncoprotein that is frequently involved in the pathogenesis of tumors of epithelial origin. Here we report mRNA expression in peripheral blood and bone marrow cells from healthy donors in hematopoietic cell lines and leukemic blasts from patients with acute lymphoblastic leukemia (ALL), acute myeloblastic leukemia (AML), chronic lymphoblastic leukemia (CLL), and chronic myeloid leukemia (CML). However, cell surface expression of HER2 protein (p185HER2) was found exclusively on a subset of leukemic cells of the B-lymphoblastic lineage. p185HER2 expression was found on blasts in 2 of 15 samples from infants, 9 of 19 samples from adult patients with C-ALL (CD19+CD10+), and 1 of 2 samples from patients with pro-B ALL (CD19+CD10-), whereas none of the leukemic cells from patients with AML (0/30), T-ALL (0/7), CLL (0/5) (CD19+CD5+), or CML in chronic and accelerated phase (0/5) or in blast crisis with myeloid differentiation (0/14) were positive for p185HER2. However, cells from 3 of 4 patients with CML in B-lymphoid blast crisis (CD19+CD10+) expressed high levels of p185HER2, which was also found on the surface of the CML-derived B-cell lines BV-173 and Nalm-1. Our study shows p185HER2 expression on malignant cells of hematopoietic origin for the first time. Aberrant expression of this oncogenic receptor tyrosine kinase in hematopoietic cell types may be an oncogenic event contributing to the development of a subset of B-lymphoblastic leukemias.


Subject(s)
B-Lymphocyte Subsets/enzymology , Blast Crisis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor, ErbB-2/biosynthesis , 3T3 Cells , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/analysis , Antigens, CD34 , Biomarkers, Tumor/analysis , Bone Marrow/pathology , Cell Line , Cell Separation/methods , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Phosphotyrosine , Receptor Protein-Tyrosine Kinases/analysis , Receptor, ErbB-2/analysis , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Transfection , Tumor Cells, Cultured , Tyrosine/analogs & derivatives , Tyrosine/analysis , Tyrosine/metabolism
5.
Blood ; 86(2): 444-50, 1995 Jul 15.
Article in English | MEDLINE | ID: mdl-7541660

ABSTRACT

This prospective multicenter study examined whether simultaneous administration of granulocyte colony-stimulating factor (G-CSF; Filgrastim) and induction chemotherapy for adult acute lymphoblastic leukemia (ALL) could prevent treatment-related neutropenia, infections, and resulting treatment delays. Seventy-six patients were randomly assigned to receive either G-CSF (n = 37) or no growth factor (n = 39) in conjunction with a uniform chemotherapy consisting of cyclophosphamide, cytarabine, mercaptopurine, intrathecal methotrexate, and cranial irradiation. The median duration of neutropenia (absolute neutrophil count < 1 x 10(9)/L) during chemotherapy was 8 days in patients receiving C-CSF, compared with 12.5 days in the control group (P < .002). A similar reduction from 11.5 to 7 days was observed in patients with T-ALL receiving additional mediastinal irradiation (P = .13). Infections occurred in 43% and 56% of patients in the G-CSF and control arm, respectively (P = .25); the incidence of nonviral infections was reduced by 50%, from 32 episodes in the control arm to 16 episodes in the G-CSF arm. Prolonged interruptions of chemotherapy administration were less frequent, with delays of 2 weeks or more occurring in only 24% of patients receiving G-CSF as opposed to 46% in the control arm (P = .01). Accordingly, chemotherapy was completed significantly earlier with the use of G-CSF (39 v 44 days, P = .008). With a median follow-up of 20 months, the probability of disease-free survival was 0.45 in the G-CSF group and 0.43 in the control group (P = .34). In conclusion, adult ALL patients appear to benefit by the simultaneous administration of G-CSF with induction chemotherapy because of a significant reduction in the duration of neutropenia, a trend to fewer infections, and a more rapid completion of chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Granulocyte Colony-Stimulating Factor/therapeutic use , Immunologic Factors/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Combined Modality Therapy , Cranial Irradiation , Cyclophosphamide/administration & dosage , Cytarabine/administration & dosage , Disease-Free Survival , Female , Filgrastim , Humans , Infection Control , Male , Mercaptopurine/administration & dosage , Methotrexate/administration & dosage , Middle Aged , Neutropenia/prevention & control , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Prospective Studies , Recombinant Proteins/therapeutic use , Remission Induction , Survival Analysis , Treatment Outcome
6.
Cancer Res ; 53(18): 4424-31, 1993 Sep 15.
Article in English | MEDLINE | ID: mdl-7689935

ABSTRACT

The function of the steel factor receptor, p145c-kit, in patient-derived acute myeloblastic leukemia (AML) cells was investigated. Steel factor stimulation of AML cells coexpressing p145c-kit and the progenitor cell antigen CD34 resulted in complete receptor down-regulation, a marked decrease of CD34 antigen expression, and the induction of the granulocyte lineage antigen CD15. These changes in surface marker expression paralleled morphological differentiation to granulated blasts and promyelocytes. Interestingly, the same phenotype was achieved by IL-3 stimulation of AML cells. p145c-kit extracellular domain-specific antibodies had either blocking or enhancing effects on ligand binding, receptor phosphorylation and down-regulation, and induction of cell proliferation. Correlations of these phenomena with distinct effects of antibody stimulation on cell substrate phosphorylation provide clues for the dissection of the p145c-kit signal and the analysis of its relevance for AML.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins/physiology , Animals , Antibodies, Monoclonal/immunology , Cell Differentiation , Down-Regulation , Hematopoietic Cell Growth Factors/physiology , Humans , Interleukin-3/pharmacology , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred BALB C , Molecular Weight , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-kit , Stem Cell Factor
7.
Intervirology ; 35(1-4): 133-9, 1993.
Article in English | MEDLINE | ID: mdl-8407240

ABSTRACT

Immunopathologic mechanisms leading to liver tissue injury in hepatitis caused by hepatitis A virus (HAV) were studied in an autologous in vitro model. Data show virus-specific killing by liver-infiltrating T lymphocytes in man and support the hypothesis that hepatocellular damage as well as efficient elimination of virus-infected hepatocytes is mediated by HLA-restricted, HAV-specific CD8+ T lymphocytes. Furthermore, experimental results demonstrate that human interferon-gamma produced by HAV-specific T cells may act as a key factor in T-cell-promoted clearance of HAV-infected hepatocytes. Besides the well-known hepatotropism, the myelotropic properties of HAV have some important clinical implications. Perturbations of hematopoietic regulation, ranging from transient granulocytopenia to rare cases of bone marrow failure, are associated with HAV infection. In an attempt to elucidate the pathogenetic mechanisms, we could show a direct suppressive effect of HAV on human bone marrow progenitors and a significant progressive decline in these cells in HAV-infected long-term bone marrow cultures.


Subject(s)
Bone Marrow/pathology , Hematopoietic Stem Cells/pathology , Hepatovirus/physiology , Liver/immunology , Bone Marrow/immunology , Bone Marrow/microbiology , Cells, Cultured , Hematopoiesis , Hematopoietic Stem Cells/microbiology , Humans , T-Lymphocytes/immunology , Virus Replication
8.
Ann Hematol ; 64 Suppl: A132-6, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1322183

ABSTRACT

Perturbations of hematopoietic regulation ranging from transient granulocytopenia to rare cases of bone marrow failure are associated with infections due to hepatitis A virus (HAV). In an attempt to elucidate the pathogenetic mechanisms we had previously established that HAV has a direct suppressive effect on human bone marrow progenitors (CFU-GM, -GEMM, BFU-E). These studies were extended to long-term bone marrow cultures (LTBMC): Inoculation of bone marrow mononuclear cells with HAV did not interfere with the establishment of an adherent stromal layer, nor did the inoculation of already established layers cause any morphologically recognizable changes to the stroma. In contrast, a significant and progressive decline of the CFU-GM content in the culture supernatants was demonstrated. HAV antigen was detected by APAAP stain in a subpopulation of stromal cells, and sequential estimations of virus titers in the supernatants provided evidence for viral replication in primary bone marrow cultures. Interferon-gamma and tumor necrosis factor-alpha levels of infected cultures did not differ from those of uninfected controls. These findings argue for a direct suppression of (pre-) CFU-GM by HAV in a model system (LTBMC) lacking an immune defense which would limit viral replication.


Subject(s)
Bone Marrow Cells , Hematopoiesis/physiology , Hepatovirus/physiology , Antigens, Viral , Bone Marrow/microbiology , Cell Transformation, Viral , Cells, Cultured/chemistry , Hepatovirus/immunology , Humans , Interferon-gamma/analysis , Tumor Necrosis Factor-alpha/analysis
9.
Leukemia ; 6(6): 582-7, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1602795

ABSTRACT

Cytosine arabinoside (ara-C) is one of the most active compounds in the treatment of acute leukemias. In the majority of current protocols ara-C is combined with other cytotoxic agents in an attempt to increase antileukemic activity. The present study investigated the impact of etoposide, teniposide, amsacrine, mitoxantrone, anthracyclines, and asparaginase on the cellular accumulation of ara-C and its intracellular metabolism in order to provide a better rationale for combination therapy. Intracellular accumulation and phosphorylation of ara-C were determined in peripheral blast cells from twenty patients with acute leukemias after exposure to 1 and 10 mumol/l ara-C alone and after preincubation with 1 and 10 micrograms/ml etoposide, 10 and 100 micrograms/ml teniposide, 10 mumol/l amsacrine, 500 ng/ml mitoxantrone (or daunorubicin or doxorubicin) or 10 mumol/l asparaginase. Ara-C accumulation at 10 mumol/l was decreased by 1 microgram/ml etoposide (67 +/- 18% of control), 10 micrograms/ml etoposide (30 +/- 22%), 10 micrograms/ml teniposide (12 +/- 23%), 100 micrograms/ml teniposide (10 +/- 18%), and amsacrine (51 +/- 21%). Intracellular ara-CTP formation was determined at an extracellular concentration of 10 mumol/l and preincubation with these drugs. The intracellular formation of ara-CTP was decreased by 1 microgram/ml etoposide (77 +/- 15% of control), 10 micrograms/ml etoposide (32 +/- 22%), 10 micrograms/ml teniposide (10 +/- 9%), 100 micrograms/ml teniposide (0 +/- 0%), but not by amsacrine. These data indicate that prior exposure to etoposide and teniposide influence ara-C metabolism and possibly cytotoxicity, and thus should not immediately precede ara-C administration in clinical trials.


Subject(s)
Arabinofuranosylcytosine Triphosphate/metabolism , Cytarabine/pharmacokinetics , Etoposide/pharmacology , Leukemia/metabolism , Teniposide/pharmacology , Acute Disease , Adult , Aged , Amsacrine/pharmacology , Cytarabine/metabolism , Drug Interactions , Humans , Leukemia/pathology , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphorylation , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
10.
Eur J Clin Pharmacol ; 42(6): 629-33, 1992.
Article in English | MEDLINE | ID: mdl-1623903

ABSTRACT

3,4-dihydro-6-[4-(3,4-dimethoxybenzoyl)-1-piperazinyl]-2(1H)-quinolinone (OPC-8212) is a quinolinone derivative with positive inotropic properties. In order to elucidate the effect of OPC-8212 on the haemopoietic system we studied its in vitro effect on bone-marrow progenitor cells (granulocyte/monocyte colony-forming units [CFU-GM] and erythroid burst-forming units [BFU-E]), on the proliferation and secretion of granulocyte/monocyte colony-stimulating factor (GM-CSF) and interferon-gamma (IFN-gamma) by peripheral lymphocytes, and on GM-CSF secretion by fibroblasts from healthy individuals. The dose-effect relations of OPC-8212 on CFU-GM proliferation and on lymphocytic GM-CSF secretion showed no effect at very low drug concentrations, with a threshold at the lower end of the therapeutic range and highly significant dose-dependent inhibition at concentrations above that threshold. BFU-E, peripheral lymphocyte proliferation and lymphocytic IFN-gamma secretion were depressed, although to a lesser extent, in a linear dose-dependent fashion. OPC-8212 did not affect GM-CSF secretion by one strain of fibroblasts but reduced it at higher concentrations in assays with another strain of cells. We conclude that direct toxic effects on bone-marrow progenitor cells, in combination with the inhibition of cytokines involved in the regulation of haemopoiesis in certain susceptible individuals, may be responsible for idiosyncratic reactions to OPC-8212.


Subject(s)
Bone Marrow Cells , Cardiotonic Agents/toxicity , Hematopoietic Stem Cells/drug effects , Lymphocytes/drug effects , Quinolines/toxicity , Bone Marrow/drug effects , Cells, Cultured , Fibroblasts/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , Humans , Interferon-gamma/drug effects , Lymphocyte Activation , Pyrazines , Skin/cytology
11.
Leukemia ; 5(10): 841-7, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1961019

ABSTRACT

A monoclonal antibody (1G2) was raised by immunization of a Balb/c mouse with the leukemic blasts from a patient suffering from chronic myelogenous leukemia blast crisis (CML-BC). Sequential immunoprecipitation of the protein from human umbilical vein endothelial cells with 1G2 and the endoglin-specific monoclonal antibody 44G4 indicated that both antibodies react with the same molecule, a homodimer of molecular mass 180,000. This protein was first identified on acute lymphoblastic leukemia and was shown to be primarily associated with endothelial cells. In addition, 1G2 and 44G4 identified the same subpopulation of human bone marrow mononuclear cells (BMMNC), as established by two colour immunofluorescence analysis. By cell sorting and colony assays it could be demonstrated that endoglin is not expressed on hemopoietic precursor cells (CFU-G, CFU-GM, CFU-GEMM, BFU-E). May-Grünwald-Giemsa staining of sorted BMMNC revealed that 1G2 recognized immature proerythroblasts and double-fluorescence analysis showed that endoglin is present on a subset of glycophorin A-positive BMMNC. 1G2 was not reactive on bone marrow B-cells (CD19, CD20), T-cells (CD3, CD7), natural killer cells (CD56), myeloid cells (CD13, CD14, CD15, CD33), and on CD34-positive cells. Endoglin contains an arginine-glycine-aspartic acid sequence, a feature generally associated with extracellular matrix proteins which interact with integrins. It is suggested that proerythroblasts may utilize endoglin to interact with integrins in cell-cell adhesion events in the stromal or hemopoietic compartment of the bone marrow.


Subject(s)
Bone Marrow/physiology , Erythroid Precursor Cells/physiology , Membrane Glycoproteins/physiology , Vascular Cell Adhesion Molecule-1 , Animals , Antibodies, Monoclonal/immunology , Antigens, CD , Antigens, Neoplasm/immunology , Blood Platelets/immunology , Bone Marrow/immunology , Bone Marrow Cells , Cell Adhesion , Endoglin , Endothelium, Vascular/immunology , Erythrocytes/immunology , Erythroid Precursor Cells/immunology , Granulocytes/immunology , Hematopoietic Stem Cells/immunology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Lymphocytes/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Phenotype , Receptors, Cell Surface
12.
Leukemia ; 5(10): 854-60, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1720490

ABSTRACT

A monoclonal antibody (17F11) was raised by immunization of a Balb/c mouse with leukemic blasts from a patient with acute non-lymphocytic leukemia (ANLL). This antibody recognizes most leukemic blasts of myeloid but not of lymphoid lineage and no peripheral blood cells. By screening NIH-3T3 fibroblasts transfected with the human proto-oncogene c-kit (NIH-3T3/hckit) it could be shown that 17F11 specifically recognizes the gene product P145c-kit. Immunofluorescence analysis on normal hemopoietic cells revealed that 17F11 weakly stains 1-3% of bone marrow mononuclear cells (BMMNC). By FACS sorting and colony assays it could be shown that granulocyte--macrophage progenitor cells could be enriched 10-20-fold, granulocyte progenitors 50-80-fold, and erythroid and multipotential progenitor cells 15-20-fold, in the 17F11 positive fraction. Double fluorescence analysis revealed that P145c-kit is co-expressed on 40-60% of the CD34 positive BMMNC. Finally, these data show that P145c-kit is expressed on blast cells from most patients with ANLL (26/30) and chronic myeloid leukemia in blast crisis (7/9), but is absent on blasts from patients with acute lymphoblastic leukemia expressing the T-, B-lineage, or common ALL phenotypes.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Surface/genetics , Hematopoietic Stem Cells/immunology , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogenes/genetics , Animals , Antibodies, Monoclonal/immunology , Antigens, CD/physiology , Antigens, CD34 , Antigens, Neoplasm/immunology , Antigens, Surface/immunology , Bone Marrow/immunology , Bone Marrow/physiology , Bone Marrow Cells , Cells, Cultured , Gene Expression , Hematopoietic Stem Cells/physiology , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred BALB C , Proto-Oncogene Mas , Proto-Oncogene Proteins c-kit , Tumor Cells, Cultured
13.
Pneumologie ; 45(5): 340-2, 1991 May.
Article in German | MEDLINE | ID: mdl-1857690

ABSTRACT

Pulmonary affections caused by atypical mycobacteria are an increasingly common problem particularly in patients with immune deficiency disorders. We here report a case of pulmonary infiltrates due to Mycobacterium xenopi in a patient after allogeneic bone marrow transplantation for acute myeloid leukemia in first complete remission and under immunosuppressive treatment with prednisolone and Cyclosporin A. While sputum cultures, serology as well as bronchial lavage and transbronchial biopsy remained inconclusive, diagnosis could only be established by open lung biopsy. We suggest that particularly in immunocompromised patients unclear pulmonary infiltrates require rapid and possibly invasive diagnostic procedures.


Subject(s)
Bone Marrow Transplantation/immunology , Mycobacterium Infections, Nontuberculous/microbiology , Respiratory Tract Infections/microbiology , Adult , Biopsy , Diagnosis, Differential , Humans , Leukemia, Myeloid, Acute/therapy , Lung/microbiology , Male , Mycobacterium Infections, Nontuberculous/immunology , Nontuberculous Mycobacteria/isolation & purification , Transplantation, Homologous , Tuberculosis, Pulmonary/diagnosis
15.
Hybridoma ; 10(1): 77-88, 1991 Feb.
Article in English | MEDLINE | ID: mdl-2032736

ABSTRACT

A monoclonal antibody (11G7) detecting a novel antigen on human hemopoietic progenitor cells (named 11G7R = 11G7 receptor) was raised by immunization of a Balb/c mouse with the leukemic blasts of a patient suffering from chronic myelogenous leukemia blast crisis (CML-BC). The antigen is expressed on most of MHC class II bearing peripheral blood leucocytes (PBL) and on a subpopulation of bone marrow mononuclear cells (BMMNC). By FACS-sorting and colony assays, it could be demonstrated that 11G7R is expressed on myelo-monocytic and myelo-granulocytic bone marrow precursor cells (GM-CFC, G-CFC, M-CFC) but is absent from erythroid precursor cells (BFU-E) and on cells exhibiting the capacity to form mixed colonies (GEMM-CFC). Double-fluorescence analysis on BMMNC revealed that 11G7R is expressed on a subset of B-cells, myeloid cells and cells carrying the HPCA-1 antigen (CD34). It has a similar distribution pattern to the myeloid antigens CD13 and CD33. However, in contrast to these antigens, 11G7R is also expressed on the blasts of several lymphoid leukemias (4/9 B-ALL, 1/2 T-ALL) and therefore it is not restricted to the myeloid lineage.


Subject(s)
Antibodies, Monoclonal , Antigens, Differentiation , Hematopoietic Stem Cells/immunology , Animals , Antigens, Differentiation/chemistry , Humans , Hybridomas/immunology , Immunochemistry , Immunoglobulin G , Leukemia/immunology , Leukocytes, Mononuclear/immunology , Mice , Tumor Cells, Cultured/immunology
16.
J Virol ; 65(2): 981-4, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1846211

ABSTRACT

The effect of murine cytomegalovirus on myelopoiesis was studied in long-term bone marrow culture to find an in vitro correlate for the lethal virus interference with bone marrow reconstitution (W. Mutter, M. J. Reddehase, F. W. Busch, H.-J. Bühring, and U. H. Koszinowski, J. Exp. Med. 167:1645-1658, 1988). The in vitro generation of granulocyte-monocyte progenitors (CFU-GM) discontinued after infection of the stromal cell layer, whereas the proliferation and differentiation of CFU-GM to granulocyte-monocyte colonies remained unaffected. A protocol was established to probe the functional integrity of earlier hematopoietic cells. Pre-CFU-GM (the progenitors of the CFU-GM) could be recovered from an infected bone marrow donor culture by transfer onto an inductive recipient stromal cell layer. Thus, at least in vitro, infection of bone marrow stroma appears to be the only cause of the defect in myelopoiesis.


Subject(s)
Bone Marrow/microbiology , Cytomegalovirus Infections/physiopathology , Cytomegalovirus/physiology , Hematopoietic Stem Cells/microbiology , Animals , Cells, Cultured , Colony-Forming Units Assay , Kinetics , Mice , Salivary Glands/physiopathology , Virus Replication
17.
Cancer Immunol Immunother ; 33(1): 54-60, 1991.
Article in English | MEDLINE | ID: mdl-1902397

ABSTRACT

T cell clones derived from a chronic myelogenous leukaemia (CML) patient during interferon alpha (IFN alpha, Wellferon) biotherapy preferentially lysed autologous rather than allogeneic CML target cells in an apparently MHC-unrestricted fashion, but also lysed bone marrow cells from certain normal donors regardless of whether or not they shared HLA antigens with the patient. Although T cell clones inhibited both CML and normal bone marrow in the colony-forming assay, they blocked proliferation of CML cells more efficiently than bone marrow cells. This inhibitory effect was mediated at least in part by the tumour necrosis factor alpha (TNF alpha) and IFN gamma secreted by the clones. Antisera to these cytokines partially prevented inhibition. Involvement of additional factors is also suggested in blocking CML cell proliferation because this was not 100% inhibited even by a combination of TNF alpha and IFN gamma. In addition, most clones failed strongly to block the proliferation of normal bone marrow cells, which were susceptible to inhibition by these cytokines.


Subject(s)
Interferon Type I/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , T-Lymphocytes/immunology , Bone Marrow Cells , Cell Division/immunology , Clone Cells , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Hematopoietic Stem Cells/immunology , Humans , Immunotherapy, Adoptive , Interferon-gamma/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/physiology
18.
Skin Pharmacol ; 4(3): 158-64, 1991.
Article in English | MEDLINE | ID: mdl-1768426

ABSTRACT

To investigate the secretion of granulocyte-macrophage colony-stimulating factor (GM-CSF) and of interleukin-3 (IL-3) by human keratinocytes in vitro, adult human keratinocytes (aHKc) from 3 different donors and a spontaneously transformed keratinocytic line (HaCaT) were cultured and exposed to various cytokines and to the protein kinase C-activating agent phorbol-12-myristate-13-acetate (PMA). GM-CSF and IL-3 were measured by highly specific and sensitive immunoassays. Our findings showed that long-term cultured aHKc and HaCaT cells are capable of secreting GM-CSF but not IL-3 upon cytokine and PMA stimulation. Both interleukin-1 and tumor necrosis factor alpha, which are known to be present in human epidermis, particularly during cutaneous inflammatory processes, were found to stimulate GM-CSF release. Therefore, we conclude that increased GM-CSF levels may play an important role in the interactions between epidermal keratinocytes and blood cells in vivo.


Subject(s)
Cytokines/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-3/metabolism , Keratinocytes/metabolism , Adult , Cell Line , Humans , Keratinocytes/drug effects , Tetradecanoylphorbol Acetate/pharmacology
19.
Blut ; 60(4): 219-22, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2337680

ABSTRACT

Drug-induced myelotoxicity is usually the dose-limiting factor of treatment of malignant tumors with cytostatic drugs. Suppression of in vitro myelopoiesis (CFU-GM) by cytostatics may be a suitable model reflecting the in vivo situation. Thus the inhibitory effects of the anthracyclines doxorubicin, theprubicin, idarubicin and cytorhodin S on CFU-GM were compared. Normal human bone marrow cells were incubated with these drugs for one hour and alternatively, for the whole culture period. For each substance and each incubation time a dose-response curve was established and the D50 determined. As certain calcium antagonists can increase the toxicity of some cytostatic drugs in various tumor models, the effect of the addition of verapamil (2 microM) was also investigated. It could be shown, that the myelotoxicity on CFU-GM of the drugs mentioned above was not increased after short-term or permanent exposure to this calcium antagonist.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Hematopoietic Stem Cells/drug effects , Verapamil/pharmacology , Bone Marrow Cells , Cell Survival/drug effects , Colony-Forming Units Assay , Hematopoietic Stem Cells/cytology , Humans , Kinetics
20.
Cancer Immunol Immunother ; 32(3): 167-72, 1990.
Article in English | MEDLINE | ID: mdl-2289210

ABSTRACT

Chronic myelogenous leukemia (CML) patients in chronic phase display compromised lymphokine-activated killer (LAK) cell induction, which is partly restored after therapy with interferon alpha. However, the relative resistance of the leukemic cells from these patients to autologous or allogeneic LAK lysis is not affected by this treatment. In an attempt to render CML cells more susceptible to lysis or cytostasis, they were precultured in serum-free medium with or without recombinant growth factors. In eight patients studied, interleukin-3 (IL-3) significantly enhanced the spontaneous short-term (6-day) proliferation of CML cells, with retention of ability to form colonies in methylcellulose. Culture in either medium alone or IL-3 led to a significant enrichment of CD14+ and CD33+ cells but to a reduction in CD34+ cells. In contrast, culture of the same cells in IL-2 (to generate autologous LAK activity) resulted in a loss of CD14+ and CD33+ as well as CD34+ cells but in a significant increase in CD3+ and CD56+ cells. Despite similarities in their phenotypes, IL-3 cultured cells but not those cultured in medium alone acquired susceptibility to lysis by the IL-2-cultured autologous LAK cells. These results may have significance for the design of novel combination immunotherapy in CML.


Subject(s)
Interferon Type I/pharmacology , Killer Cells, Lymphokine-Activated/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid, Chronic-Phase/drug therapy , Antigens, Surface/immunology , Cell Division/drug effects , Cell Survival/drug effects , Cytokines/pharmacology , Cytotoxicity, Immunologic/immunology , Humans , Interferon Type I/therapeutic use , Interleukin-3/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Chronic-Phase/pathology , Phenotype , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...