Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Leukemia ; 30(4): 889-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26442612

ABSTRACT

CC-486, the oral formulation of azacitidine (AZA), is an epigenetic modifier and DNA methyltransferase inhibitor in clinical development for treatment of hematologic malignancies. CC-486 administered for 7 days per 28-day treatment cycle was evaluated in a phase 1 dose-finding study. AZA has a short plasma half-life and DNA incorporation is S-phase-restricted; extending CC-486 exposure may increase the number of AZA-affected diseased target cells and maximize therapeutic effects. Patients with lower-risk myelodysplastic syndromes (MDS) received 300 mg CC-486 once daily for 14 days (n=28) or 21 days (n=27) of repeated 28-day cycles. Median patient age was 72 years (range 31-87) and 75% of patients had International Prognostic Scoring System Intermediate-1 risk MDS. Median number of CC-486 treatment cycles was 7 (range 2-24) for the 14-day dosing schedule and 6 (1-24) for the 21-day schedule. Overall response (complete or partial remission, red blood cell (RBC) or platelet transfusion independence (TI), or hematologic improvement) (International Working Group 2006) was attained by 36% of patients receiving 14-day dosing and 41% receiving 21-day dosing. RBC TI rates were similar with both dosing schedules (31% and 38%, respectively). CC-486 was generally well-tolerated. Extended dosing schedules of oral CC-486 may provide effective long-term treatment for patients with lower-risk MDS.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , Myelodysplastic Syndromes/drug therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/pharmacokinetics , Azacitidine/pharmacokinetics , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Male , Middle Aged , Myelodysplastic Syndromes/pathology , Neoplasm Staging , Prognosis , Risk Factors , Safety , Tissue Distribution
2.
Leukemia ; 30(3): 649-57, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26464171

ABSTRACT

Established prognostic tools in patients with myelodysplastic syndromes (MDS) were largely derived from untreated patient cohorts. Although azanucleosides are standard therapies for higher-risk (HR)-MDS, the relative prognostic performance of existing prognostic tools among patients with HR-MDS receiving azanucleoside therapy is unknown. In the MDS Clinical Research Consortium database, we compared the prognostic utility of the International Prognostic Scoring System (IPSS), revised IPSS (IPSS-R), MD Anderson Prognostic Scoring System (MDAPSS), World Health Organization-based Prognostic Scoring System (WPSS) and the French Prognostic Scoring System (FPSS) among 632 patients who presented with HR-MDS and were treated with azanucleosides as the first-line therapy. Median follow-up from diagnosis was 15.7 months. No prognostic tool predicted the probability of achieving an objective response. Nonetheless, all five tools were associated with overall survival (OS, P=0.025 for the IPSS, P=0.011 for WPSS and P<0.001 for the other three tools). The corrected Akaike Information Criteria, which were used to compare OS with the different prognostic scoring systems as covariates (lower is better) were 4138 (MDAPSS), 4156 (FPSS), 4196 (IPSS-R), 4186 (WPSS) and 4196 (IPSS). Patients in the highest-risk groups of the prognostic tools had a median OS from diagnosis of 11-16 months and should be considered for up-front transplantation or experimental approaches.


Subject(s)
Antineoplastic Agents/therapeutic use , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/drug therapy , Aged , Databases, Factual , Decitabine , Female , Follow-Up Studies , Humans , Male , Middle Aged , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/pathology , Prognosis , Research Design , Risk Factors , Survival Analysis
3.
Leukemia ; 24(8): 1437-44, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20535150

ABSTRACT

We report the results of a phase I dose escalation trial of the multikinase inhibitor sorafenib in relapsed and refractory acute leukemia patients using an intermittent dosing regimen. Fifteen patients with advanced leukemia (12 with acute myeloid leukemia, 2 with acute lymphoblastic leukemia, 1 with biphenotypic) and a median age of 63 (range 37-85) years were enrolled and treated on a dose escalation trial. Toxicities >or=grade 3 were present in 55% of cycles and the maximum tolerated dose (MTD) was determined to be 400 mg b.i.d. x 21 days in a 28-day cycle. Plasma inhibitory assays of kinase targets extracellular signal-regulated kinase (ERK) and FLT3-internal tandem duplication (ITD) showed excellent target inhibition, with FLT3-ITD silencing occurring below the MTD. The N-oxide metabolite of sorafenib seemed to be a more potent inhibitor of FLT3-ITD than the parent compound. Despite marked ex vivo FLT-3 ITD inhibition, no patients met the criteria for complete or partial response in this monotherapy study. Out of 15 patients, 11 experienced stable disease as best response. Although sorafenib showed only modest clinical activity as a single agent in this heavily treated population, robust inhibition of FLT3 and ERK suggests that there may be a potential important role in combination therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzenesulfonates/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Benzenesulfonates/pharmacokinetics , Benzenesulfonates/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Humans , Leukemia, Myeloid, Acute/blood , Male , Middle Aged , Niacinamide/analogs & derivatives , Phenylurea Compounds , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacokinetics , Pyridines/pharmacology , Recurrence , Sorafenib , fms-Like Tyrosine Kinase 3/antagonists & inhibitors
4.
Oncogene ; 27(25): 3615-23, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18223691

ABSTRACT

Decitabine (DAC) and 5-azacitidine have recently been approved for the treatment of myelodysplastic syndrome. The pharmacodynamic effects of DAC and 5-azacitidine outside their known activity as inhibitors of DNA methyltransferases (DNMTs) require further investigation. The purpose of this study was to investigate the effect of DAC on the expression of p21(WAF1/CIP1), a gene with a putative CpG island surrounding its promoter region. Promoter methylation analysis of p21(WAF1/CIP1) in leukemia cells revealed the absence of CpG methylation. However, DAC upregulated p21(WAF1/CIP1) expression in a dose-dependent manner (ED(50)=103.34 nM) and induced G2/M cell cycle arrest in leukemia cells. Sequential application of DAC followed by different histone deacetylase inhibitors induced expression of p21(WAF1/CIP1) synergistically. Upregulation of p21(WAF1/CIP1) paralleled DAC-induced apoptosis (ED(50)=153 nM). Low doses of DAC induced gamma-H2AX expression (ED(50)=16.5 nM) and upregulated p21(WAF1/CIP1) in congenic HCT 116 colon cancer cells in a DNMT-independent and p53-dependent fashion. Inhibition of p53 transactivation by pifithrin-alpha or the kinase activity of ATM by either the specific ATM inhibitor KU-5593 or caffeine abrogated p21(WAF1/CIP1) upregulation, indicating that DAC upregulation of p21(WAF1/CIP1) was p53- and ATM-dependent in leukemia cells. In conclusion, DAC upregulates p21(WAF1/CIP1) in DNMT-independent manner via the DNA damage/ATM/p53 axis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytosine/analogs & derivatives , DNA Damage , Gene Expression Regulation, Leukemic , Leukemia/drug therapy , Leukemia/metabolism , Caffeine/pharmacology , Cell Line, Tumor , CpG Islands , Cytosine/pharmacology , DNA Repair , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Humans , Models, Biological , Models, Genetic , Transcriptional Activation
5.
Ecancermedicalscience ; 2: 121, 2008.
Article in English | MEDLINE | ID: mdl-22275991

ABSTRACT

Azacitidine (AZA), as demonstrated in the phase III trial (AZA-001), is the first MDS treatment to significantly prolong overall survival (OS) in higher risk MDS pts ((2007) Blood 110 817). Approximately, one-third of the patients (pts) enrolled in AZA-001 were FAB RAEB-T (≥20-30% blasts) and now meet the WHO criteria for acute myeloid leukaemia (AML) ((1999) Blood 17 3835). Considering the poor prognosis (median survival <1 year) and the poor response to chemotherapy in these pts, this sub-group analysis evaluated the effects of AZA versus conventional care regimens (CCR) on OS and on response rates in pts with WHO AML.

6.
Ecancermedicalscience ; 2: 118, 2008.
Article in English | MEDLINE | ID: mdl-22275990

ABSTRACT

The international, phase III, multi-centre AZA-001 trial demonstrated azacitidine (AZA) is the first treatment to significantly extend overall survival (OS) in higher risk myelodysplastic syndromes (MDS) patients (Fenaux (2007) Blood110 817). The current treatment paradigm, which is based on a relationship between complete remission (CR) and survival, is increasingly being questioned (Cheson (2006) Blood108 419). Results of AZA-001 show CR is sufficient but not necessary to prolong OS (List (2008) Clin Oncol26 7006). Indeed, the AZA CR rate in AZA-001 was modest (17%), while partial remission (PR, 12%) and haematological improvement (HI, 49%) were also predictive of prolonged survival. This analysis was conducted to assess the median number of AZA treatment cycles associated with achievement of first response, as measured by IWG 2000-defined CR, PR or HI (major + minor). The number of treatment cycles from first response to best response was also measured.

7.
J Assoc Physicians India ; 54: 441-4, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16909690

ABSTRACT

OBJECTIVES: To study the inheritance pattern of diabetes mellitus in Western Indian population by analysing the pedigree of diabetes patients. METHODS: 3,921 individuals from 300 families were interviewed for family history in this study, out of which 770 were diabetic individuals. Statistical analysis of the data was carried out using T-test and Chi-square test. RESULTS: 37% cases of Type 1 DM and 58% cases of Type 2 DM showed family history of the disease. Of the cases showing family history for diabetes, 92% in case of Type 1 DM and 59% in case of Type 2 DM showed family history of Type 2 DM with a decrease in age of onset in the successive generations. Both the parents, when diabetic conferred equal risk of inheriting diabetes in offspring. The sex ratio of offspring suffering from diabetes was not influenced when only one of the parents was diabetic. However it was observed that the male offspring were highly susceptible when both parents were diabetic (Chi-square value=4.55 with 1 d.f.). The age of onset of diabetes did not show significant correlation with whether one or both the parents were diabetic. However, it was noteworthy that in case of familial history of diabetes there was a decrease in the age of onset in successive generations. CONCLUSION: This study suggests that family history of diabetes results in predisposition to early onset of the disease in successive generations and a cluster of genes involved in Type 2 DM may show a parental effect for predisposition to Type 1 DM in the offspring in this set of Indian population.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Pedigree , Adolescent , Adult , Age Factors , Aged , Child , Child, Preschool , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Epidemiologic Studies , Female , Humans , India/epidemiology , Infant , Male , Middle Aged , Risk Assessment , Risk Factors
8.
Appl Biochem Biotechnol ; 102-103(1-6): 21-39, 2002.
Article in English | MEDLINE | ID: mdl-12396108

ABSTRACT

Thirty-seven strains of Acinetobacter isolated and characterized from rhizosphere of wheat were screened for indole-3-acetic acid (IAA) production. Only eight Acinetobacter strains showed IAA production. The genus Acinetobacter was confirmed by chromosomal DNA transformation assay. Biotyping of eight strains was carried out and they were found to be genospecies of A. junii, A. baumannii, A. genospecies 3, and A. haemolyticus. Five of eight strains produced IAA at the early stationary phase: A. haemolyticus (A19), A. baumannii (A18, A16, A13), and Acinetobactergenospecies 3 (A15). A. junii A6 showed maximum IAA production at log phase and A. genospecies 3 and A. baumannii (A28, A30) at late stationary phase. IAA was extracted by ethyl acetate and purified by preparative thin-layer chromatography. Purified IAA was confirmed by 1H-nuclear magnetic resonance and infrared spectrum analysis. Pot experiments showed a significant increase in plant growth inoculated with eight Acinetobacter genospecies as compared to control plants. IAA production was found to be encoded by plasmid pUPI126. All eight strains of Acinetobacter contain a plasmid pUPI126 with a molecular weight of 40 kb. Plasmid pUPI126 was transformed into Escherichia coli HB101 at a frequency of 5 x 10(-5), and E. coli HB101 (pUPI126) transformants also showed IAA activity. PUPI126 also encoded resistance to selenium, tellurium, and lead. This is the first report of plasmid-encoded IAA production in the genus Acinetobacter.


Subject(s)
Acinetobacter/genetics , Acinetobacter/metabolism , Indoleacetic Acids/metabolism , Plant Roots/microbiology , Plasmids/genetics , Triticum/microbiology , Acinetobacter/isolation & purification , Analysis of Variance , Chromatography, Thin Layer , Hydrogen-Ion Concentration , Indoleacetic Acids/isolation & purification , Indoleacetic Acids/pharmacology , Magnetic Resonance Spectroscopy , Plant Growth Regulators/pharmacology , Plant Roots/drug effects , Plant Roots/growth & development , Plant Shoots/drug effects , Plant Shoots/growth & development , Spectrophotometry, Infrared , Time Factors , Transformation, Genetic , Triticum/drug effects , Triticum/growth & development
9.
Leuk Res ; 26(5): 461-71, 2002 May.
Article in English | MEDLINE | ID: mdl-11916520

ABSTRACT

We report single institution outcome of brief, intensive ara-C-based chemotherapy using bone marrow transplantation as primary intensification for untreated adult patients with acute lymphoblastic leukemia (ALL). Overall disease-free and overall survival were inferior to those reported with prolonged chemotherapy modeled on pediatric protocols. Survival and disease-free survival were superior for patients receiving allogeneic BMT compared with chemopurged autologous transplant or maintenance chemotherapy (patients ineligible for or declining BMT). In multivariate analysis, non-L2-FAB, higher ara-C dose, absence of CNS disease, non-Ph1+ karyotype, allogeneic BMT, T cell phenotype, and younger age were associated with improved disease-free survival. Autologous BMT was not superior to chemotherapy, and appears unlikely to provide adequate curative treatment for most adult ALL patients if not followed by maintenance.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Bone Marrow Transplantation , Cytarabine/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adult , Aged , Follow-Up Studies , Humans , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Transplantation, Autologous
10.
Clin Cancer Res ; 7(8): 2330-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489809

ABSTRACT

Sodium phenylbutyrate (PB) is an aromatic fatty acid with cytostatic and differentiating activity against malignant myeloid cells (ID(50), 1-2 mM). Higher doses induce apoptosis. Patients with myelodysplasia (n = 11) and acute myeloid leukemia (n = 16) were treated with PB as a 7-day continuous infusion repeated every 28 days in a Phase I dose escalation study. The maximum tolerated dose was 375 mg/kg/day; higher doses led to dose-limiting reversible neurocortical toxicity. At the maximum tolerated dose, PB was extremely well tolerated, with no significant toxicities; median steady-state plasma concentration at this dose was 0.29 +/- 0.16 mM. Although no patients achieved complete or partial remission, four patients achieved hematological improvement (neutrophils in three, platelet transfusion-independence in one). Other patients developed transient increases in neutrophils or platelets and decrements in circulating blasts. Monitoring of the percentage of clonal cells using centromere fluorescence in situ hybridization over the course of PB administration showed that hematopoiesis remained clonal. Hematological response was often associated with increases in both colony-forming units-granulocyte-macrophage and leukemic colony-forming units. PB administration was also associated with increases in fetal erythrocytes. These data document the safety of continuous infusion PB and provide preliminary evidence of clinical activity in patients with myeloid malignancies.


Subject(s)
Leukemia, Myeloid/drug therapy , Myelodysplastic Syndromes/drug therapy , Phenylbutyrates/therapeutic use , Acute Disease , Aged , Aged, 80 and over , Alopecia/chemically induced , Antigens, CD34/analysis , Apoptosis/drug effects , Cell Cycle/drug effects , Clone Cells , Diarrhea/chemically induced , Dose-Response Relationship, Drug , Fetal Hemoglobin/drug effects , Fetal Hemoglobin/metabolism , Flow Cytometry , Hemorrhage/chemically induced , Humans , Middle Aged , Mouth Mucosa/drug effects , Mouth Mucosa/pathology , Myelodysplastic Syndromes/immunology , Nausea/chemically induced , Phenylbutyrates/adverse effects , Phenylbutyrates/pharmacokinetics , Stomatitis/chemically induced , Treatment Outcome , Vomiting/chemically induced
11.
Expert Opin Investig Drugs ; 9(12): 2923-34, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11093362

ABSTRACT

Compounds that inhibit histone deacetylase may enable the re-expression of silenced regulatory genes in neoplastic cells, reversing the malignant phenotype. Although several molecules that inhibit histone deacetylase are undergoing preclinical development, butyric acid derivatives have undergone clinical investigation for several years, initially for non-malignant indications and more recently for the treatment of cancer. Of the butyric acid derivatives, sodium phenylbutyrate has undergone the most extensive systematic investigation. Administration of phenylbutyrate by iv. and oral routes is well-tolerated clinically at concentrations which effect acetylation of histones in vitro. Higher doses lead to reversible CNS depression. The studies presented to date have been Phase I studies and do not enable assessment of efficacy. However, current development of phenylbutyrate is proceeding in combination with other agents based on rational biologically-based in vitro studies. The parallel development of combination therapy including phenylbutyrate and early clinical development of other, more potent histone deacetylase inhibitors will hopefully lead to feasible, clinically tolerable strategies for altering the malignant phenotype of cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Histones/physiology , Neoplasms/drug therapy , Phenylbutyrates/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Histones/metabolism , Humans , Neoplasms/enzymology , Phenylbutyrates/therapeutic use
12.
Blood ; 96(12): 3922-31, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11090079

ABSTRACT

Recent studies have suggested that variations in levels of caspases, a family of intracellular cysteine proteases, can profoundly affect the ability of cells to undergo apoptosis. In this study, immunoblotting was used to examine levels of apoptotic protease activating factor-1 (Apaf-1) and procaspases-2, -3, -7, -8, and -9 in bone marrow samples (at least 80% leukemia) harvested before chemotherapy from adults with newly diagnosed acute myelogenous leukemia (AML, 42 patients) and acute lymphocytic leukemia (ALL, 18 patients). Levels of each of these polypeptides varied over a more than 10-fold range between specimens. In AML samples, expression of procaspase-2 correlated with levels of Apaf-1 (R(s) = 0.52, P <.02), procaspase-3 (R(s) = 0.56, P <.006) and procaspase-8 (R(s) = 0.64, P <.002). In ALL samples, expression of procaspases-7 and -9 was highly correlated (R(s) = 0.90, P <.003). Levels of these polypeptides did not correlate with prognostic factors or response to induction chemotherapy. In further studies, 16 paired samples (13 AML, 3 ALL), the first harvested before induction therapy and the second harvested at the time of leukemia regrowth, were also examined. There were no systematic alterations in levels of Apaf-1 or procaspases at relapse compared with diagnosis. These results indicate that levels of initiator caspases vary widely among different leukemia specimens but cast doubt on the hypothesis that this variation is a major determinant of drug sensitivity for acute leukemia in the clinical setting. (Blood. 2000;96:3922-3931)


Subject(s)
Caspases/metabolism , Enzyme Precursors/metabolism , Leukemia/diagnosis , Acute Disease , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptotic Protease-Activating Factor 1 , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Bone Marrow Cells/chemistry , Bone Marrow Cells/enzymology , Caspases/drug effects , Caspases/immunology , Cohort Studies , Enzyme Precursors/drug effects , Enzyme Precursors/immunology , HL-60 Cells , Humans , Immunoblotting , Leukemia/metabolism , Leukemia/therapy , Leukemia, Myeloid/diagnosis , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/therapy , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prognosis , Proteins/drug effects , Proteins/immunology , Proteins/metabolism
13.
Leukemia ; 13(8): 1243-53, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10450753

ABSTRACT

The aromatic fatty acid phenylbutyrate (PB) induces cytostasis, differentiation, and apoptosis in primary myeloid leukemic cells at clinically achievable concentrations. In the present study, we have investigated the structural and cellular basis for PB-induced cytostasis, using the ML-1 human myeloid leukemia cell line as a model system. PB induced a dose-dependent increase in cells in G1 with a corresponding decrease in cells in S-phase of the cell cycle. At comparable doses, PB induced expression of CD11b, indicating myeloid differentiation. At higher doses, the drug induced apoptosis. The antitumor activity was independent of the aromatic ring, as butyric acid (BA) was of equal or greater potency at producing these biological changes. In contrast, shortening of the fatty acid carbon chain length, as demonstrated with phenylacetate (PA), significantly diminished drug potency. Consistent with their effects on cell cycle, PB and BA, but not PA, induced the cyclin-dependent kinase inhibitor, p21(WAF1/CIP1), and led to the appearance of hypophosphorylated Rb, suggesting a role for p21(WAF1/CIP1) in PB-induced cytostasis. Therefore, it appears that the fatty acid moiety of PB, rather than its aromatic ring, is critical for its activity in myeloid leukemic cells. These data provide a potential mechanistic basis for the increased potency of PB over PA previously demonstrated in primary leukemic samples, and support the further clinical development of PB in the treatment of hematologic malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , G1 Phase/drug effects , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/pathology , Phenylbutyrates/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Cell Division/drug effects , Dose-Response Relationship, Drug , Humans , Phenylbutyrates/chemistry , Phenylbutyrates/therapeutic use , Structure-Activity Relationship , Tumor Cells, Cultured
14.
Leukemia ; 13(8): 1258-65, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10450755

ABSTRACT

Despite preliminary evidence of clinical activity of the putative differentiating agent sodium phenylbutyrate (PB) in the treatment of myeloid neoplasms, it has proven difficult to maintain therapeutic levels of PB above 0.5 mM, well below the ED50 of 1-2 mM. We have studied the impact of combining PB with all-trans retinoic acid (ATRA) on the ML-1 myeloid leukemia cell line. ATRA augmented PB-induced differentiation, cell-cycle arrest, and apoptosis. ATRA augmented PB induction of the myelomonocytic marker CD11b at all doses of ATRA tested (0.0025-1 microM). Although ATRA did not significantly affect the ED50 of PB, the combination of ATRA (1 microM) and PB (0.5 mM) augmented PB-induced CD11b expression eight-fold. Compared to PB alone, this combination of ATRA and PB induced greater cell cycle arrest (S-phase 14% vs 38%; G0/G1-phase cells 72% vs 52%) and greater apoptosis (24% vs 16% by TUNEL assay). Treatment with ATRA (0.5 microM) in combination with PB (0.5 mM) led to significantly greater inhibition of colony formation (4.8% vs 48% inhibition). ATRA combined synergistically with PB to augment CD11b expression and inhibit colony formation. This combination also showed significant interaction in terms of S-phase inhibition. However, this interaction varied as a function of ATRA concentration: antagonistic at low concentrations of ATRA, synergistic at higher concentrations of ATRA. These data suggest that retinoids may significantly augment the cytostatic and differentiating activity of PB, leading to increased potency of the latter drug at clinically achievable doses.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/pathology , Phenylbutyrates/pharmacology , Tretinoin/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Differentiation/drug effects , Cell Division/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Humans , Phenylbutyrates/therapeutic use , Tretinoin/therapeutic use , Tumor Cells, Cultured
15.
In Vivo ; 13(1): 1-6, 1999.
Article in English | MEDLINE | ID: mdl-10218124

ABSTRACT

BACKGROUND: Hematopoietic growth factors (HGF) can suppress chemotherapy-induced programmed cell death (apoptosis) in hematopoietic cells. Although HGF can modulate the expression of apoptosis-regulatory genes, including bcl-2, bax, and p21WAF1/CIP1 in cell lines, few data address whether HGF regulate the expression of these proteins in primary acute myeloid leukemia (AML). MATERIALS AND METHODS: We evaluated the expression of bcl-2, bax, and p21WAF1/CIP1 in primary samples from patients with AML in the presence and absence of HGF. The potential association of HGF-induced changes in the levels of these proteins with inhibition of chemotherapy-induced apoptosis was further investigated. RESULTS: While a combination of steel factor (SF) and PIXY321 inhibited etoposide-induced apoptosis in 8/11 primary AML samples studied, Bcl-2 and bax protein levels were unaffected by exposure to HGF and/or etoposide. In contrast, HGF enhanced basal and etoposide-induced p21WAF1/CIP1 protein levels in 9/11 and 7/11 of the cases, respectively. In several cases, inhibition of apoptosis by HGF was seen without up-regulation of p21WAF1/CIP1 levels, suggesting that modulation of p21WAF1/CIP1 is not required for HGF-mediated inhibition of apoptosis. CONCLUSIONS: These data indicate that HGF-mediated inhibition of chemotherapy-induced apoptosis in primary AML samples is not mediated through changes in Bcl-2, bax, and p21WAF1/CIP1 protein levels.


Subject(s)
Apoptosis , Cyclins/metabolism , Hematopoietic Cell Growth Factors/metabolism , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myelomonocytic, Acute/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Phytogenic/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Etoposide/pharmacology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Humans , Interleukin-3/pharmacology , Leukemia, Erythroblastic, Acute/blood , Leukemia, Myeloid, Acute/blood , Leukemia, Myelomonocytic, Acute/blood , Male , Middle Aged , Recombinant Fusion Proteins/pharmacology , Stem Cell Factor/pharmacology , Tumor Cells, Cultured , bcl-2-Associated X Protein
16.
Semin Oncol ; 26(1): 21-34, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10073559

ABSTRACT

Allogeneic (alloBMT) and autologous bone marrow transplantation (ABMT) have become standard approaches for the management of adults with acute myeloid leukemia (AML). The indications for transplantation remain controversial as parallel improvements in intensive chemotherapy have resulted in excellent outcomes for many patients. AlloBMT is the therapy of choice for patients who fail to respond to induction chemotherapy. For those patients in first remission (CRI), a policy of intensive postremission chemotherapy with transplantation upon relapse appears to be optimal. There are no data to support transplantation in CRI, allogeneic or autologous, for those patients with leukemia characterized by favorable cytogenetic abnormalities [ie, core-binding factor type or t(15;17)], as these patients do well with nonmyeloablative strategies. Patients with relapsed disease appear to be best served with allogeneic transplantation from a human leukocyte antigen (HLA)-matched sibling or one-antigen-mismatched family member, whereas for those patients lacking a related donor, unrelated donor alloBMT or ABMT provides similar long-term overall survival. Randomized studies for the optimal management of relapsed disease are lacking but are needed. The objective of this review is to discuss the data supporting the use of alloBMT or ABMT at various points during the course of de novo adult AML.


Subject(s)
Antineoplastic Agents/adverse effects , Bone Marrow Transplantation , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Adult , Bone Marrow Transplantation/methods , Clinical Trials as Topic , Humans , Leukemia, Myeloid, Acute/drug therapy , Neoplasm Staging , Prospective Studies , Transplantation, Autologous , Transplantation, Homologous , Treatment Outcome
17.
Clin Cancer Res ; 4(11): 2677-89, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9829730

ABSTRACT

To further evaluate the activity of topotecan (TPT) in acute leukemia, TPT was administered (2.1 mg/m2/day for 5 days by continuous i.v. infusion) to adult patients with previously untreated acute lymphoblastic leukemia (ALL) with high-risk features (13 patients) or relapsed ALL (1 patient). Patients achieving a partial response or significant hematological improvement received a second course. All patients subsequently received standard treatment for ALL. Because complete response was achieved in only 1 of 14 patients, the study was terminated prematurely. An additional patient achieved minimal response, and a third patient normalized her hemogram despite ongoing leukemia in the marrow. Overall, six patients had significant hematological improvement (normalization of platelet and/or absolute neutrophil count). Two patients expired due to infections during induction chemotherapy. The primary nonhematological toxicities were mucositis and diarrhea. Exposure to TPT did not appear to influence the response to subsequent standard chemotherapy. The mean steady-state TPT plasma concentration, 16.1+/-1 nM, overlapped the range of LD90 values of primary human leukemia specimens. Cellular topo I content varied over a 3-fold range, encompassing levels found previously in relapsed patients. No relationship was found between topo I expression and markers of cellular proliferation or response to therapy. In contrast, low expression of the apoptosis inhibitor Bcl-2 was associated with response to TPT therapy. TPT has significant, albeit modest, single-agent activity against high-risk adult ALL. This study demonstrates the feasibility of evaluating promising new therapeutic agents in untreated patients with acute leukemia at high risk for failure with conventional therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Topotecan/therapeutic use , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Bone Marrow/drug effects , Bone Marrow/pathology , DNA Topoisomerases, Type I/metabolism , Female , Humans , Leukocyte Count/drug effects , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Risk Factors , Topoisomerase I Inhibitors , Topotecan/adverse effects , Topotecan/pharmacokinetics , Treatment Outcome
18.
Br J Haematol ; 102(4): 1042-9, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9734656

ABSTRACT

Despite extensive investigation into mechanisms of drug resistance in acute myeloid leukaemia (AML), the aetiology of therapeutic resistance is unclear. We found that five leukaemia cell lines (K562, HL-60, CEM. CEM induced to overexpress bcl-2, and REH) displayed parallel sensitivity to four antileukaemia drugs with different mechanisms of action, with K562 generally being the least sensitive and REH being the most sensitive. The amount of spontaneous apoptosis in the cell lines after serum-free culture paralleled their drug sensitivity: K562 cells displayed the least apoptosis at 24h (2.50 +/- 0.24%) and REH the most (24.47 +/- 8.22%). The extent of spontaneous apoptosis of leukaemic blasts from 39 patients with newly diagnosed de novo AML also correlated with the success of the intensive, infusional cytarabine-based induction therapy. There was a median of 19.5% (range 3.6-64%) apoptotic AML cells after 24 h of serum-free culture in patients who entered a complete remission compared with 4.2% (1.8-7.0%) apoptotic AML cells in patients who did not achieve a complete remission (P = 0.0007). Thus, inhibited apoptosis was associated with both in vitro and in vivo pan-resistance to antileukaemic chemotherapy. The cause of inhibited apoptosis in AML is probably a function of interactions among multiple signals that influence apoptosis. Assessment of spontaneous apoptosis may serve as an important prognostic factor for AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/physiology , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/pathology , Acute Disease , Adult , Aged , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Culture Media, Serum-Free , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/physiology , Humans , Leukemia/pathology , Middle Aged , Treatment Outcome , Tumor Cells, Cultured
19.
Leuk Lymphoma ; 29(3-4): 217-37, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9684921

ABSTRACT

A number of recent studies have investigated the expression of topoisomerase II in clinical leukemia specimens. Here we outline the rationale for these studies, identify potential pitfalls, summarize recent results, and discuss unanswered questions in this area.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia/drug therapy , Topoisomerase II Inhibitors , Acute Disease , Adult , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Child , DNA Damage , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/physiology , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Humans , RNA, Messenger/analysis
20.
Leuk Lymphoma ; 29(3-4): 339-50, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9684931

ABSTRACT

The biological heterogeneity of AML makes growth factor augmentation of cell cycle-dependent chemotherapy unlikely to be successful for all patients. Patients whose leukemic cells empirically demonstrate cytokine-induced chemosensitization in vitro might benefit from the concurrent administration of growth factors during consolidation chemotherapy. We have explored the growth factor-dependence and response of primary bone marrow samples from patients with AML at diagnosis, remission, and relapse to determine whether minimal residual leukemia remains growth factor-responsive. Most cases of AML studied at all phases of treatment were growth factor-responsive. Growth factor response of occult remission clonogenic leukemic precursors (CFU-L) was usually concordant with their response at diagnosis. Occult CFU-L were markedly resistant to cytosine arabinoside (median LD99% 20 microM); preincubation with IL-3 or GM-CSF did not significantly improve their ara-C sensitivity. While occult remission CFU-L appear to remain growth factor-responsive, it appears unlikely that growth factor augmentation of consolidation chemotherapy will overcome the important problem of drug resistance of residual leukemia.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Interleukin-3/therapeutic use , Leukemia, Myeloid/drug therapy , Acute Disease , Adult , Aged , Amsacrine/administration & dosage , Cytarabine/administration & dosage , Daunorubicin/administration & dosage , Etoposide/administration & dosage , Humans , Ki-67 Antigen/analysis , Ki-67 Antigen/drug effects , Leukemia, Myeloid/pathology , Middle Aged , Neoplasm, Residual , Remission Induction , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...