Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 329
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Leukemia ; 21(3): 480-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17215857

ABSTRACT

beta-2 Microglobulin (beta2M), a subunit of human leukocyte antigen-class I (HLA-I), is well established as a marker of prognosis in various solid tumors and hematologic malignancies. The prognostic role of intact free-circulating HLA-I (sHLA-I) is less well understood. We compared the clinical relevance of plasma levels of sHLA-I and beta2M in patients with acute myeloid leukemia (AML; n=209) or advanced myelodysplastic syndrome (MDS; n=98). sHLA-1 and beta2M levels were significantly higher in AML and MDS patients than in control subjects, but did not differ significantly between the two disease groups. In AML patients, multivariate analysis showed both sHLA-1 and beta2-M to be highly predictive of complete remission (CR), survival and duration of complete response (CRD). In MDS, the predictive value of the two markers differed substantially from one another: beta2M was associated with survival, CR and CRD, whereas sHLA-I was not. These findings not only establish the role of sHLA-I as a tumor marker in AML but also support that MDS is clinically and biologically distinct from AML. sHLA-I has been reported to be an immunomodulator inhibiting the cytotoxic effects of T-lymphocytes, which may offset its predictive value for disease aggressiveness in patients with MDS.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/blood , HLA Antigens/blood , Leukemia, Myeloid/blood , Myelodysplastic Syndromes/blood , Neoplasm Proteins/blood , beta 2-Microglobulin/blood , Acute Disease , Adult , Aged , Aged, 80 and over , Cytarabine/administration & dosage , Disease Progression , Female , Humans , Idarubicin/administration & dosage , Kaplan-Meier Estimate , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/mortality , Male , Middle Aged , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/mortality , Predictive Value of Tests , Proportional Hazards Models , Remission Induction , Solubility , Topotecan/administration & dosage , Treatment Outcome , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
2.
J Clin Invest ; 100(7): 1716-24, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9312169

ABSTRACT

We hypothesized that 1,25-dihydroxyvitamin D3 (1,25D3) and its analogues may inhibit acute myelogenous leukemia (AML) proliferation by interrupting IL-1beta-mediated growth-stimulatory signals. The incubation of the IL-1beta- responsive AML cell line OCIM2 with 10 nM 1,25D3 reduced growth 80% in liquid culture, and a 100-1000-fold lower concentration of 20-epi analogues (MC1288 and MC1301) was sufficient to achieve similar growth inhibition. The growth inhibition was associated with a rapid but transient downregulation of IL-1beta and IL-1beta-converting enzyme (ICE) mRNAs in 1,25D3- and 20-epi analogue- treated cells, and the 20-epi analogue was more effective than 1,25D3 in repressing ICE expression. An examination of long-term changes in the levels of mature IL-1beta and its precursor revealed that 24-h incubation of OCIM2 with either 1,25D3 or its 20-epi analogues abolished the production of mature IL-1beta. The effect of 1,25D3 and its analogues on growth of fresh bone marrow cells from seven AML patients was tested by a clonogenic assay. Growth inhibition of 60% was reached in only one of seven 1,25D3-treated samples, but all seven samples were inhibited 60-90% by the 20-epi analogue MC1301. Growth inhibition by 1,25D3 and the analogue was reversible by addition of IL-1beta. These results suggest that 1,25D3 and its 20-epi analogues interrupt IL-1beta autocrine growth regulation by inhibiting IL-1beta production and processing but not the response to IL-1beta.


Subject(s)
Calcitriol/analogs & derivatives , Gene Expression Regulation, Neoplastic , Interleukin-1/biosynthesis , Leukemia, Myeloid, Acute/metabolism , Stem Cells/drug effects , Antineoplastic Agents , Autocrine Communication , Calcitriol/pharmacology , Caspase 1 , Cysteine Endopeptidases/biosynthesis , Dose-Response Relationship, Drug , Down-Regulation , Humans , Protein Processing, Post-Translational/drug effects , Structure-Activity Relationship , Transcription, Genetic/drug effects , Tumor Cells, Cultured
3.
J Clin Invest ; 108(6): 851-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11560954

ABSTRACT

The mitogen-activated protein kinase (MAPK) pathway regulates growth and survival of many cell types, and its constitutive activation has been implicated in the pathogenesis of a variety of malignancies. In this study we demonstrate that small-molecule MEK inhibitors (PD98059 and PD184352) profoundly impair cell growth and survival of acute myeloid leukemia (AML) cell lines and primary samples with constitutive MAPK activation. These agents abrogate the clonogenicity of leukemic cells but have minimal effects on normal hematopoietic progenitors. MEK blockade also results in sensitization to spontaneous and drug-induced apoptosis. At a molecular level, these effects correlate with modulation of the expression of cyclin-dependent kinase inhibitors (p27(Kip1) and p21(Waf1/CIP1)) and antiapoptotic proteins of the inhibitor of apoptosis proteins (IAP) and Bcl-2 families. Interruption of constitutive MEK/MAPK signaling therefore represents a promising therapeutic strategy in AML.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Tumor Suppressor Proteins , Apoptosis/drug effects , Benzamides/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/genetics , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/genetics , Cytarabine/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Leukemia, Myeloid, Acute/pathology , Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction , Tumor Cells, Cultured , X-Linked Inhibitor of Apoptosis Protein
4.
Leuk Res ; 31(6): 791-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17156841

ABSTRACT

FLT3 ligand (FL) has a significant role in the proliferation and differentiation of hematopoietic cells. Mutations in the FLT3 receptor gene have been reported in 30% of patients with AML. We investigated whether abnormal phosphorylation of FLT3 may be more common in AML. We evaluated FLT3 protein and its phosphorylation in the plasma from 85 patients with AML, 16 patients with myelodysplastic syndrome (MDS) and 5 patients with acute lymphoblastic leukemia (ALL). There were no significant differences in the level of plasma FLT3 protein level in the different diseases (p=0.57). AML patients had a significantly higher level of phospho-FLT3:FLT3 ratio (p=0.02). FLT3-ITD and FLT3 point mutations were present in 27 (32%) of the AML patients. Phosphorylated FLT3 was significantly higher in the plasma from patients with FLT3 mutation (p=0.002). Overall, there was no correlation between survival and the plasma level of FLT3 protein or its phosphorylated form. However, amongst the patients without FLT3 mutations, those with a higher level of phosphorylated FLT3 had a significantly shorter duration of remission (p=0.04). Other mechanisms may be responsible for abnormal phosphorylation of FLT3 and inhibitors of FLT3 should also be investigated in patients without mutations.


Subject(s)
Leukemia, Myeloid, Acute/blood , Protein Processing, Post-Translational , fms-Like Tyrosine Kinase 3/blood , Adolescent , Adult , Aged , Aged, 80 and over , Cell Differentiation/genetics , Cell Proliferation , Female , Hematopoietic Stem Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Male , Membrane Proteins/genetics , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Protein Processing, Post-Translational/genetics , Remission Induction , fms-Like Tyrosine Kinase 3/genetics
5.
Leukemia ; 31(3): 697-704, 2017 03.
Article in English | MEDLINE | ID: mdl-27654852

ABSTRACT

PR1, an HLA-A2-restricted peptide derived from both proteinase 3 and neutrophil elastase, is recognized on myeloid leukemia cells by cytotoxic T lymphocytes (CTLs) that preferentially kill leukemia and contribute to cytogenetic remission. To evaluate safety, immunogenicity and clinical activity of PR1 vaccination, a phase I/II trial was conducted. Sixty-six HLA-A2+ patients with acute myeloid leukemia (AML: 42), chronic myeloid leukemia (CML: 13) or myelodysplastic syndrome (MDS: 11) received three to six PR1 peptide vaccinations, administered subcutaneously every 3 weeks at dose levels of 0.25, 0.5 or 1.0 mg. Patients were randomized to the three dose levels after establishing the safety of the highest dose level. Primary end points were safety and immune response, assessed by doubling of PR1/HLA-A2 tetramer-specific CTL, and the secondary end point was clinical response. Immune responses were noted in 35 of 66 (53%) patients. Of the 53 evaluable patients with active disease, 12 (24%) had objective clinical responses (complete: 8; partial: 1 and hematological improvement: 3). PR1-specific immune response was seen in 9 of 25 clinical responders versus 3 of 28 clinical non-responders (P=0.03). In conclusion, PR1 peptide vaccine induces specific immunity that correlates with clinical responses, including molecular remission, in AML, CML and MDS patients.


Subject(s)
Cancer Vaccines/immunology , HLA-A2 Antigen/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Peptides/immunology , Biomarkers , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Epitopes, T-Lymphocyte/immunology , Female , HLA-A2 Antigen/chemistry , Humans , Immunologic Memory , Immunophenotyping , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Peptides/administration & dosage , Peptides/adverse effects , Survival Analysis , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Vaccination
6.
Leukemia ; 31(12): 2560-2567, 2017 12.
Article in English | MEDLINE | ID: mdl-28555084

ABSTRACT

DNA methyltransferase inhibitors sensitize leukemia cells to chemotherapeutics. We therefore conducted a phase 1/2 study of mitoxantrone, etoposide and cytarabine following 'priming' with 5-10 days of decitabine (dec/MEC) in 52 adults (median age 55 (range: 19-72) years) with relapsed/refractory acute myeloid leukemia (AML) or other high-grade myeloid neoplasms. During dose escalation in cohorts of 6-12 patients, all dose levels were well tolerated. As response rates appeared similar with 7 and 10 days of decitabine, a 7-day course was defined as the recommended phase 2 dose (RP2D). Among 46 patients treated at/above the RP2D, 10 (22%) achieved a complete remission (CR), 8 without measurable residual disease; five additional patients achieved CR with incomplete platelet recovery, for an overall response rate of 33%. Seven patients (15%) died within 28 days of treatment initiation. Infection/neutropenic fever, nausea and mucositis were the most common adverse events. While the CR rate compared favorably to a matched historic control population (observed/expected CR ratio=1.77), CR rate and survival were similar to two contemporary salvage regimens used at our institution (G-CLAC (granulocyte colony-stimulating factor (G-CSF); clofarabine; cytarabine) and G-CLAM (G-CSF; cladribine; cytarabine; mitoxantrone)). Thus, while meeting the prespecified efficacy goal, we found no evidence that dec/MEC is substantially better than other cytarabine-based regimens currently used for relapsed/refractory AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/analogs & derivatives , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Azacitidine/administration & dosage , Azacitidine/adverse effects , Azacitidine/therapeutic use , Biomarkers , Cytarabine , Decitabine , Drug Resistance, Neoplasm , Etoposide , Female , Humans , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Mitoxantrone , Neoplasm Grading , Recurrence , Treatment Outcome , Young Adult
7.
Leukemia ; 31(2): 318-324, 2017 02.
Article in English | MEDLINE | ID: mdl-27795561

ABSTRACT

Most clinical trials exclude patients with poor performance or comorbidities. To study whether patients with these characteristics can be treated within a clinical trial, we conducted a study for patients with acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS) with poor performance, organ dysfunction or comorbidities. Primary endpoint was 60-day survival. Study included stopping rules for survival and response. Treatment consisted on a combination of azacitidine and vorinostat. Thirty patients (16 with MDS, 14 with AML) were enrolled. Median follow-up was 7.4 months (0.3-29). Sixty-day survival was 83%. No stopping rules were met. Main adverse events (AEs) were grades 1 and 2 gastrointestinal toxicities. In view of these results, we expanded the study and treated 79 additional patients: 27 with azacitidine (AZA) and 52 with azacitidine and vorinostat (AZA+V). Median follow-up was 22.7 months (12.6-47.5). Sixty-day survival rate was 79% (AZA=67%, AZA+V=85%, P=0.07). Median overall survival was 7.6 months (4.5-10.7). Median event-free survival was 4.5 months (3.5-5.6). Main AEs included grades 1 and 2 gastrointestinal toxicities. Our results suggest this subset of patients can be safely treated within clinical trials and derive clinical benefit. Relaxation of standard exclusion criteria may increase the pool of patients likely to benefit from therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Bone Marrow/pathology , Chromosome Aberrations , Comorbidity , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Treatment Outcome
8.
Leukemia ; 31(11): 2347-2354, 2017 11.
Article in English | MEDLINE | ID: mdl-28322237

ABSTRACT

Therapy-related acute promyelocytic leukemia (t-APL) is relatively rare, with limited data on outcome after treatment with arsenic trioxide (ATO) compared to standard intensive chemotherapy (CTX). We evaluated 103 adult t-APL patients undergoing treatment with all-trans retinoic acid (ATRA) alone (n=7) or in combination with ATO (n=24), CTX (n=53), or both (n=19). Complete remissions were achieved after induction therapy in 57% with ATRA, 100% with ATO/ATRA, 78% with CTX/ATRA, and 95% with CTX/ATO/ATRA. Early death rates were 43% for ATRA, 0% for ATO/ATRA, 12% for CTX/ATRA and 5% for CTX/ATO/ATRA. Three patients relapsed, two developed therapy-related acute myeloid leukemia and 13 died in remission including seven patients with recurrence of the prior malignancy. Median follow-up for survival was 3.7 years. None of the patients treated with ATRA alone survived beyond one year. Event-free survival was significantly higher after ATO-based therapy (95%, 95% CI, 82-99%) as compared to CTX/ATRA (78%, 95% CI, 64-87%; P=0.042), if deaths due to recurrence of the prior malignancy were censored. The estimated 2-year overall survival in intensively treated patients was 88% (95% CI, 80-93%) without difference according to treatment (P=0.47). ATO when added to ATRA or CTX/ATRA is feasible and leads to better outcomes as compared to CTX/ATRA in t-APL.


Subject(s)
Arsenicals/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Neoplasms, Second Primary/drug therapy , Oxides/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenic Trioxide , Female , Humans , Leukemia, Promyelocytic, Acute/etiology , Leukemia, Promyelocytic, Acute/genetics , Male , Middle Aged , Neoplasms, Second Primary/etiology , Neoplasms, Second Primary/genetics , Remission Induction , Survival Analysis , Treatment Outcome , Young Adult
10.
Leukemia ; 19(11): 1912-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16167059

ABSTRACT

Little is known about the etiology of myelodysplastic syndromes (MDS). A hospital-based case-control study of 354 adult de novo MDS cases and 452 controls was conducted to investigate associations between lifestyle characteristics and MDS risk. The distribution by French-American-British (FAB) type was 67 (19%) refractory anemia (RA), 38 (11%) refractory anemia with ringed sideroblasts (RARS), 43 (12%) chronic myelomonocytic leukemia (CMML), 136 (38%) RA with excess blasts (RAEB), and 70 (20%) RAEB in transformation (RAEBT). Multivariate logistic regression analyses were performed among all MDS cases and among each FAB type and gender. For all MDS combined, family history of hematopoietic cancer (odds ratio (OR) = 1.92), smoking (OR = 1.65), and exposure to agricultural chemicals (OR = 4.55) or solvents (OR = 2.05) were associated with MDS risk. Among RA/RARS cases, smoking (OR = 2.23) and agricultural chemical exposure (OR = 5.68) were the only risk factors identified. For RAEB/RAEBT cases, family history of hematopoietic cancer (OR = 2.10), smoking (OR = 1.52), and exposure to agricultural chemicals (OR = 3.79) or solvents (OR = 2.71) were independent risk factors. Drinking wine reduced risk for all FAB types by almost 50% (OR = 0.54). We found a joint effect between smoking and chemical exposure with the highest risk among smokers exposed to solvents/agricultural chemicals (OR = 3.22). Results from this large study suggest that several factors play a role in MDS predisposition with possible joint effects. Risk profiles seem to differ by FAB type and gender.


Subject(s)
Environmental Exposure , Genetic Predisposition to Disease , Myelodysplastic Syndromes/etiology , Smoking/adverse effects , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Humans , Life Style , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Odds Ratio , Pesticides/poisoning , Risk Factors , Sex Factors , Solvents/poisoning
11.
Leukemia ; 19(9): 1543-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16001087

ABSTRACT

We investigated the constitutive activation of the MEK/ERK pathway in acute myelogenous leukemia (AML) via a flow cytometric technique to quantitate expression of phosphorylated ERK (p-ERK). A total of 42 AML samples (16 newly diagnosed, 26 relapsed/refractory) were analyzed. Normal bone marrow CD34+ cells (n = 10) had little or no expression of p-ERK, while G-CSF-mobilized CD34+ cells exhibited enhanced p-ERK levels. Markedly elevated p-ERK levels were found in 83.3% of the AML samples, with no differences observed between the newly diagnosed and relapsed/refractory samples. Treatment with a MEK inhibitor resulted in significantly decreased p-ERK levels in both the newly diagnosed and relapsed/refractory samples, which was associated with growth arrest, but not apoptosis induction. In summary, we defined conditions for the analysis of MAPK signaling in primary AML samples. Normal CD34+ cells expressed very low levels of p-ERK, and increased p-ERK levels were found in normal G-CSF-stimulated circulating CD34+ cells. Constitutively high p-ERK levels observed in the majority of AML samples suggest deregulation of this pathway that appears to be independent of disease status. The ability of ERK inhibition to promote growth arrest rather than apoptosis suggests that clinical trials of MEK/ERK inhibitors may be more effective when combined with chemotherapy.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Leukemia, Myeloid/metabolism , Acute Disease , Antigens, CD34/metabolism , Apoptosis/drug effects , Benzamides/pharmacology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flow Cytometry , Humans , Phosphorylation , Recurrence
12.
Leukemia ; 19(9): 1567-72, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16049515

ABSTRACT

Determining the percentage of peripheral blood (PB) and bone marrow (BM) blasts is important for diagnosing and classifying acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). Although most patients with acute leukemia or MDS have a higher percentage of BM blasts than PB blasts, the relative proportion is reversed in some patients. We explored the clinical relevance of this phenomenon in MDS (n = 446), AML (n = 1314), and acute lymphoblastic leukemia (ALL) (n = 385). Among patients with MDS or ALL, but not AML, having a higher blast percentage in PB than in BM was associated with significantly shorter survival. In multivariate analyses, these associations were independent of other relevant predictors, including cytogenetic status. Our findings suggest that MDS and ALL patients who have a higher percentage of PB blasts than BM blasts have more aggressive disease. These data also suggest that MDS classification schemes should take into account the percentage of blasts in PB differently from the percentage of blasts in BM.


Subject(s)
Blast Crisis/blood , Bone Marrow/blood supply , Leukemia, Lymphoid/blood , Leukemia, Myeloid/blood , Myelodysplastic Syndromes/blood , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Blast Crisis/pathology , Child , Female , Humans , Leukemia, Lymphoid/classification , Leukemia, Lymphoid/pathology , Leukemia, Myeloid/classification , Leukemia, Myeloid/pathology , Male , Middle Aged , Multivariate Analysis , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/pathology , Retrospective Studies , Survival Analysis
13.
Cancer Res ; 47(1): 251-7, 1987 Jan 01.
Article in English | MEDLINE | ID: mdl-3024817

ABSTRACT

Antineoplastic intercalating agents such as 4'-(9-acridinylamino)methanesulfon-m-anisidide (m-AMSA) stabilize a cleavable complex between topoisomerase II and DNA. The production of protein-associated DNA cleavage in whole cells exposed to m-AMSA is thought to represent the cellular correlate of this topoisomerase II-mediated reaction. Protein-associated DNA cleavage can be quantified in mammalian cells by using alkaline elution technology. In an attempt to understand the impact of phenotypic and biochemical cellular characteristics on protein-associated DNA cleavage, we quantified m-AMSA-induced DNA cleavage in quiescent or proliferative normal human fibroblasts (cell strain 1508) and human glioblastoma cells (line T98G) as well as in asynchronously proliferating HeLa cells. The magnitude of DNA cleavage in quiescent fibroblasts and quiescent glioblastoma cells was identical and low relative to that observed in the HeLa cells. The magnitude of DNA cleavage was enhanced in both cell types following proliferation. This enhancement was greater in the glioblastoma cells than in the fibroblasts. These results were not due to alterations in cellular m-AMSA uptake. Chromatin was more elongated (open) in the quiescent glioblastoma cells than in the quiescent fibroblasts (as visualized by using the premature chromosome condensation assay), suggesting chromatin accessibility to drug per se may not be a critical determinant of the magnitude of m-AMSA-induced DNA cleavage. The onset of the enhanced m-AMSA-induced DNA cleavability that accompanied proliferation closely followed the formation of regions of localized chromatin decondensation, a late G1 event, and coincided with the onset of enhanced thymidine uptake, a marker for the onset of S phase. m-AMSA-induced cytotoxicity was also enhanced in proliferating compared with quiescent cells. The major finding of this study is that the cellular target for m-AMSA, putatively topoisomerase II, is more susceptible to drug action in proliferating cells than in quiescent cells. Effects of chromatin conformation or cellular phenotype upon topoisomerase II-mediated events such as m-AMSA-induced DNA cleavage are less certain.


Subject(s)
Amsacrine/pharmacology , Chromatin/metabolism , DNA/metabolism , Intercalating Agents/pharmacology , Proteins/metabolism , Brain Neoplasms/metabolism , Cell Division , DNA Topoisomerases, Type II/physiology , Fibroblasts/metabolism , Humans , Molecular Conformation , Phenotype
14.
Cancer Res ; 51(3): 935-8, 1991 Feb 01.
Article in English | MEDLINE | ID: mdl-1988134

ABSTRACT

After Phase I studies of benzisoquinolinedione (amonafide) in solid tumors identified myelosuppression as the dose-limiting toxicity, we conducted a Phase I study in patients with relapsed or refractory acute leukemia to define the optimal dose. Amonafide was given i.v. over 2-4 h daily for 5 days. The starting dose was 600 mg/m2/day with subsequent escalation to 750, 900, 1100, 1400, and 1800 mg/m2/day. Thirty-eight courses were administered to 24 patients, of whom 12 participated in concomitant pharmacological studies. Nausea and vomiting, transient orange discoloration of the skin, and tinnitus occurred at all dose levels. The latter symptom, along with lightheadedness and flushing, was related to infusion duration; this was increased to 4 h with doses greater than or equal to 900 mg/m2. The dose-limiting toxicities were mucositis and painful skin erythema which occurred in all 4 patients treated with 1800 mg/m2. No remissions occurred. Clearing of peripheral blood blasts occurred in 67% of patients treated with 1100 mg/m2 and in all patients treated with greater than or equal to 1100 mg/m2/day. A decrease in marrow leukemic infiltrate (% blasts x % cellularity) to less than 10% occurred in 15 and 50% of patients treated at these levels, respectively. There were 10 deaths (42%), which were unrelated to dosage. The harmonic mean terminal plasma half-life was 4.6 h (range, 2.5-35.5 h). Three patients had long drug half-lives of 9.7, 16.4, and 35.5 h and each had initial bilirubin levels greater than 1.0 mg/dl. The average urinary excretion of amonafide over 5 days was 3.5% of the total dose. This establishes 1100-1400 mg/m2/day for 5 days as the maximally tolerated dose of amonafide for studies in acute leukemia.


Subject(s)
Antineoplastic Agents/therapeutic use , Imides , Isoquinolines/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Acute Disease , Adenine , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Drug Administration Schedule , Drug Evaluation , Female , Humans , Isoquinolines/administration & dosage , Isoquinolines/adverse effects , Isoquinolines/pharmacokinetics , Leukemia/metabolism , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Naphthalimides , Organophosphonates , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
15.
Cancer Res ; 47(11): 3005-11, 1987 Jun 01.
Article in English | MEDLINE | ID: mdl-3471322

ABSTRACT

Twenty-seven patients with refractory leukemia were treated with 1-beta-D-arabinofuranosylcytosine (ara-C), 0.3 to 3.0 g/m2 as i.v. infusions over 1, 2, 4, or 24 h. The pharmacokinetics of ara-C in plasma and its 5'-triphosphate (ara-CTP) in leukemic cells from peripheral blood were studied after a single infusion of 3 g/m2 over 2 h in 13 patients. Accumulation of ara-CTP in leukemic cells remained linear until 1 to 2 h after the infusion. At the time when the rate of ara-CTP accumulation deviated from linearity, the plasma concentration of ara-C was 5- to 20-fold lower [8.1 +/- 4.4 (SD) microM] than the steady-state level during the infusion. Plasma ara-C and cellular ara-CTP pharmacokinetics were studied after two serial infusions in 14 additional patients. Varying the duration of infusion of an ara-C dose between 1, 2, and 4 h (corresponding to infusion rates of 3000, 1500, and 750 mg/m2/h) did not substantially change the rate of ara-CTP accumulation by leukemic cells. The peak ara-CTP concentration and the area under the concentration times time curve (AUC) of ara-CTP in leukemic cells increased with prolongation of the infusion. Although steady-state concentration of ara-C and AUC of ara-C in plasma were proportionally reduced by 1.0 or 0.5 g/m2 infusion over 2 h, ara-CTP accumulation rate and AUC in leukemic cells did not change compared with administration of 3 g/m2 over 2 h. However, when the infusion rate was further reduced to 0.4 or 0.3 g/m2 over 2 h, resulting in steady-state plasma ara-C concentrations of less than 7 microM, the accumulation rate of ara-CTP was substantially reduced as was the ara-CTP intracellular AUC. The cellular elimination rate of ara-CTP remained constant under all infusion conditions. These findings support the conclusion that high-dose ara-C therapy, as currently administered, results in plasma ara-C concentrations that saturate the accumulation of ara-CTP by circulating leukemic cells. We recommend that intermediate dose rates, 200 to 250 mg/m2/h, be evaluated in future studies as an alternative to the substantially higher ara-C dose rates currently in use.


Subject(s)
Arabinofuranosylcytosine Triphosphate/metabolism , Arabinonucleotides/metabolism , Cytarabine/administration & dosage , Leukemia/metabolism , Blast Crisis , Cytarabine/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Infusions, Intravenous , Leukemia/drug therapy , Leukemia, Myeloid/metabolism , Leukemia, Myeloid, Acute/metabolism , Metabolic Clearance Rate
16.
Cancer Res ; 52(17): 4587-90, 1992 Sep 01.
Article in English | MEDLINE | ID: mdl-1511426

ABSTRACT

The retinoblastoma (RB) protein levels in blast-enriched mononuclear fractions from the peripheral blood of 33 newly diagnosed patients with acute myelogenous leukemia were studied. Ten patients who had previously been treated were also analyzed, nine of whom had achieved prior complete remission. Low RB protein expression was found in 13 of 43 (30%) of the acute myelogenous leukemia patients as determined by Western blotting and immunochemical analysis. Of particular interest among the 20 newly diagnosed patients treated with the same therapeutic regimen, the median survival was 39 days for those with low RB protein expression compared to 333 days for those with high levels of RB protein expression in their leukemic cells (P less than or equal to 0.02). This preliminary study suggests that decreases of RB protein expression in peripheral blood of myeloid leukemic cells occur frequently and may be associated with shortened survival of acute myelogenous leukemia patients.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Retinoblastoma Protein/metabolism , Blotting, Western , Humans , Prognosis , Survival Analysis
17.
Leukemia ; 30(7): 1456-64, 2016 07.
Article in English | MEDLINE | ID: mdl-27012865

ABSTRACT

Measurable ('minimal') residual disease (MRD) before or after hematopoietic cell transplantation (HCT) identifies adults with AML at risk of poor outcomes. Here, we studied whether peri-transplant MRD dynamics can refine risk assessment. We analyzed 279 adults receiving myeloablative allogeneic HCT in first or second remission who survived at least 35 days and underwent 10-color multiparametric flow cytometry (MFC) analyses of marrow aspirates before and 28±7 days after transplantation. MFC-detectable MRD before (n=63) or after (n=16) transplantation identified patients with high relapse risk and poor survival. Forty-nine patients cleared MRD with HCT conditioning, whereas two patients developed new evidence of disease. The 214 MRD(neg)/MRD(neg) patients had excellent outcomes, whereas both MRD(neg)/MRD(pos) patients died within 100 days following transplantation. For patients with pre-HCT MRD, outcomes were poor regardless of post-HCT MRD status, although survival beyond 3 years was only observed among the 58 patients with decreasing but not the seven patients with increasing peri-HCT MRD levels. In multivariable models, pre-HCT but not post-HCT MRD was independently associated with overall survival and risk of relapse. These data indicate that MRD(pos) patients before transplantation have a high relapse risk regardless of whether or not they clear MFC-detectable disease with conditioning and should be considered for pre-emptive therapeutic strategies.


Subject(s)
Flow Cytometry/methods , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid, Acute/pathology , Neoplasm, Residual/diagnosis , Adolescent , Adult , Aged , Bone Marrow Examination , Female , Hematopoietic Stem Cell Transplantation/mortality , Humans , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Multivariate Analysis , Neoplasm, Residual/mortality , Postoperative Period , Preoperative Period , Recurrence , Retrospective Studies , Survival Rate , Transplantation Conditioning , Treatment Outcome , Young Adult
18.
Leukemia ; 30(2): 268-73, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26365212

ABSTRACT

Decitabine may open the chromatin structure of leukemia cells making them accessible to the calicheamicin epitope of gemtuzumab ozogamicin (GO). A total of 110 patients (median age 70 years; range 27-89 years) were treated with decitabine and GO in a trial designed on model-based futility to accommodate subject heterogeneity: group 1: relapsed/refractory acute myeloid leukemia (AML) with complete remission duration (CRD) <1 year (N=28, 25%); group 2: relapsed/refractory AML with CRD ⩾1 year (N=5, 5%); group 3: untreated AML unfit for intensive chemotherapy or untreated myelodysplastic syndrome (MDS) or untreated myelofibrosis (MF; N=57, 52%); and group 4: AML evolving from MDS or relapsed/refractory MDS or MF (N=20, 18%). Treatment consisted of decitabine 20 mg/m(2) daily for 5 days and GO 3 mg/m(2) on day 5. Post-induction therapy included five cycles of decitabine+GO followed by decitabine alone. Complete remission (CR)/CR with incomplete count recovery was achieved in 39 (35%) patients; group 1= 5/28 (17%), group 2=3/5 (60%), group 3=24/57 (42%) and group 4=7/20 (35%). The 8-week mortality in groups 3 and 4 was 16% and 10%, respectively. Common drug-related adverse events included nausea, mucositis and hemorrhage. Decitabine and GO improved the response rate but not overall survival compared with historical outcomes in untreated AML ⩾60 years.


Subject(s)
Aminoglycosides/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/analogs & derivatives , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Adult , Aged , Aged, 80 and over , Aminoglycosides/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Azacitidine/administration & dosage , Azacitidine/adverse effects , Decitabine , Female , Gemtuzumab , Humans , Male , Middle Aged , Sialic Acid Binding Ig-like Lectin 3/analysis
19.
Oncogene ; 7(8): 1645-7, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1630824

ABSTRACT

Expression of the normal p53 gene promotes cell differentiation, maturation and apoptosis. The mutant p53 gene, which does not function normally, is frequently expressed at elevated levels in tumor cells [for review see Lane, D.P. & Benchimol, S. (1990). Genes Dev., 4, 1-8]. We have analysed the expression of and mutational change in the p53 gene in the peripheral blood cells of 49 primary acute myeloid leukemia (AML) patients. The p53 protein levels were elevated in 37 patients (75%) when measured by immunoprecipitation with antibodies PAb1801 and PAb421, which recognize both normal and mutant forms of the protein. The p53 protein from 32 of these 37 patients was immunoprecipitated by PAb240, which recognizes a conformation of p53 protein associated with point mutations. However, point mutations were detected by single-stranded conformation polymorphism (SSCP) assay and direct sequencing in only three patients at codons 178, 245, 273 and 290. Growth stimulation of normal lymphocytes also generated p53 which was immunoprecipitable by PAb240. Thus, alteration of p53 conformation, rather than acquisition of point mutations, could be the mechanism underlying the increased proliferation of myeloid cells in most AML patients.


Subject(s)
Genes, p53/genetics , Leukemia, Myeloid/metabolism , Tumor Suppressor Protein p53/metabolism , Acute Disease , DNA, Single-Stranded/genetics , Humans , Leukemia, Myeloid/blood , Leukemia, Myeloid/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/physiology , Lymphocytes/metabolism , Mutation/genetics , Polymorphism, Genetic/genetics , Precipitin Tests , Protein Conformation , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
20.
Oncogene ; 8(4): 983-8, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8455949

ABSTRACT

The t(8;21) translocation breakpoint, which is observed in acute myeloid leukemia (AML), has recently been cloned and a fusion transcript identified. We have now designed primer sets capable of amplifying the breakpoint junction of the fusion transcript by the reverse transcription-polymerase chain reaction (RT-PCR). Primer set 821U/821D1 amplified a 200-bp DNA fragment, and primer set 821U/821D2 amplified a 1.2-kb DNA fragment in all t(8;21)-positive AML tested. Sequence analysis of the amplified DNA fragments demonstrated that all fusion transcripts were fused at exactly the same site, indicating that this translocation breakpoint occurs within a single intron of the AML1 and ETO genes. Forty-five cycles of RT-PCR were used to detect residual t(8;21)-positive leukemia cells in three patients who had been in complete remission for 1, 3 and 5 years. Minimal residual disease was found in all three samples. Northern blot analysis demonstrated that two fusion transcripts of 7 and 10 kb were expressed in the t(8;21)-positive AML and that the ETO gene is not normally expressed in the hematopoietic system. Expression of a normal 5.5-kb ETO mRNA was found in the lung. From these results we concluded that expression of the ETO gene in t(8;21)-positive AML was activated as a result of the translocation.


Subject(s)
Leukemia, Myeloid, Acute/diagnosis , Base Sequence , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , DNA, Neoplasm/analysis , Gene Expression , Humans , Leukemia, Myeloid, Acute/genetics , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Polymerase Chain Reaction , RNA, Neoplasm/genetics , Restriction Mapping , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL