Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
Cell Death Differ ; 22(12): 2133-42, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26045051

ABSTRACT

MLN4924 (pevonedistat), an inhibitor of the Nedd8 activating enzyme (NAE), has exhibited promising clinical activity in acute myelogenous leukemia (AML). Here we demonstrate that MLN4924 induces apoptosis in AML cell lines and clinical samples via a mechanism distinct from those observed in other malignancies. Inactivation of E3 cullin ring ligases (CRLs) through NAE inhibition causes accumulation of the CRL substrate c-Myc, which transactivates the PMAIP1 gene encoding Noxa, leading to increased Noxa protein, Bax and Bak activation, and subsequent apoptotic changes. Importantly, c-Myc knockdown diminishes Noxa induction; and Noxa siRNA diminishes MLN4924-induced killing. Because Noxa also neutralizes Mcl-1, an anti-apoptotic Bcl-2 paralog often upregulated in resistant AML, further experiments have examined the effect of combining MLN4924 with BH3 mimetics that target other anti-apoptotic proteins. In combination with ABT-199 or ABT-263 (navitoclax), MLN4924 exerts a synergistic cytotoxic effect. Collectively, these results provide new insight into MLN4924-induced engagement of the apoptotic machinery that could help guide further exploration of MLN4924 for AML.


Subject(s)
Apoptosis/drug effects , Cyclopentanes/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyrimidines/pharmacology , Up-Regulation/drug effects , Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Drug Synergism , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Sulfonamides/pharmacology
2.
Leukemia ; 25(12): 1808-14, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21760592

ABSTRACT

This study of vosaroxin evaluated dose-limiting toxicity (DLT), maximum-tolerated dose (MTD), pharmacokinetics (PK), clinical activity and pharmacodynamics in relapsed/refractory leukemia. Dosing was weekly (days 1, 8 and 15) or twice weekly (days 1, 4, 8 and 11). Seventy-three treated patients had a median age of 65 years, 85% had acute myeloid leukemia and 78% had refractory disease. Weekly schedule: 42 patients received 18-90 mg/m(2); MTD was 72 mg/m(2). Twice-weekly schedule: 31 patients received 9-50 mg/m(2); MTD was 40 mg/m(2). DLT was stomatitis; primary non-hematologic toxicity was reversible gastrointestinal symptoms and febrile neutropenia. Thirty-day all-cause mortality was 11%. Five patients had complete or incomplete remissions; median duration was 3.1 months. A morphologic leukemia-free state (bone marrow blast reduction to <5%) occurred in 11 additional patients. Antileukemic activity was associated with total dose or weekly time above 1 µmol/l plasma vosaroxin concentration (P<0.05). Vosaroxin exposure was dose proportional over 9-90 mg/m(2). The average terminal half-life was ~25 h and clearance was non-renal. No induction or inhibition of vosaroxin metabolism was evident. Vosaroxin-induced DNA damage was detected as increased intracellular γH2AX. Vosaroxin had an acceptable safety profile, linear PK and encouraging clinical activity in relapsed/refractory leukemia.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Naphthyridines/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Salvage Therapy , Thiazoles/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Staging , Treatment Outcome , Young Adult
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.
Leukemia ; 21(9): 1915-20, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17611566

ABSTRACT

Despite extensive study in many malignancies, maintenance therapy has clinically benefited only two diseases: acute lymphocytic leukemia (ALL) and acute promyelocytic leukemia (APL). ALL maintenance therapy utilizes low-dose 6-mercaptopurine (6MP) and methotrexate (MTX), while maintenance in APL primarily consists of all-trans-retinoic acid (ATRA). 6MP and MTX as used in ALL are also now usually added to maintenance ATRA for APL, based on data suggesting an improved disease-free survival. Although the mechanism of action of MTX and 6MP as maintenance is unknown, low-dose cytotoxic agents are potent inducers of differentiation in vitro. Thus, we studied whether maintenance therapy in ALL, like ATRA in APL, may be inducing terminal differentiation of ALL progenitors. The APL cell line NB4, the ALL cell lines REH and RS4;11, and patients' ALL blasts were incubated with ATRA, 6MP, and MTX in vitro. All three drugs inhibited the clonogenic growth of the APL and ALL cell lines without inducing immediate apoptosis, but associated with induction of phenotypic differentiation. The three drugs similarly upregulated lymphoid antigen expression, while decreasing CD34 expression, on patients' ALL blasts. These data suggest that induction of leukemia progenitor differentiation plays an important role in the mechanism of action of maintenance therapy in ALL.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Mercaptopurine/pharmacology , Methotrexate/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tretinoin/pharmacology , Adolescent , Adult , Cell Differentiation/drug effects , Cell Line, Tumor , Clone Cells , Cytotoxins/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , In Vitro Techniques , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Remission Induction
5.
Mutat Res ; 629(1): 24-31, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17276720

ABSTRACT

Although there has been a rapid rise in the application of fluorescent in situ hybridization (FISH) analysis of bone marrow tissue for the staging and prognosis determination of hematopoietic malignacies such as the chronic and acute leukemias, it's application as a surveillance tool for leukemogen exposed high risk occupational cohorts is understandably limited by the invasiveness of sample collection. While some small occupational studies have been performed using FISH in peripheral blood with promising results, some of the basic assumptions made in utilizing the FISH technique have not been fully explored. These include selection of the correct hematopoietic cell to assay (myeloid or lymphoid); selection of appropriate chromosomal markers and the sensitivity of peripheral blood FISH in detecting unbalanced genomic abnormalities. In this study, we performed a pilot 'validation' exercise utilizing the FISH technique and standard metaphase cytogenetics, comparing results in tandem pairs of peripheral blood with bone marrow cells, where clonal abnormalities arise. Samples were taken from patients with known chromosomal lesions associated with active leukemia. We carefully chose markers most frequently associated with leukemogen-inducing DNA damage and probes that have been utilized successfully in clinical practice. Ten de novo or therapy-related acute myeloid leukemia (t-AML) patients underwent bone marrow cell karyotyping and fluorescent in situ hybridization (FISH) analysis. Parallel peripheral blood samples were concommitently drawn and evaluated with FISH using the same probes. In six of eight paired samples treated with a 3-day phytohemagglutinin (PHA) stimulation, typically used to assay lymphocytes and their progenitors, we detected abnormal clones. In one of the two remaining cases, we identified an abnormal clone in both bone marrow and PHA-stimulated peripheral blood, although at a level in the peripheral blood sample that would typically be reported as "non-diagnostic" for clinical purposes. These results suggest that use of FISH in PHA stimulated peripheral blood samples with probes commonly employed in t-AML evaluations (chromosomes 5q, 7q, 8, 11q) to detect cytogenetic abnormalities in peripheral blood represents a potentially promising though as yet, under-utilized approach for the occupational surveillance of workers exposed to leukemogens, especially if it could be linked to automated high-throughput assays for increased sensitivity.


Subject(s)
Bone Marrow Cells/pathology , In Situ Hybridization, Fluorescence/methods , Leukemia, Myeloid, Acute/pathology , Leukocytes, Mononuclear/metabolism , Occupational Exposure/adverse effects , Population Surveillance , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 5 , Chromosomes, Human, Pair 7 , Chromosomes, Human, Pair 8 , Cohort Studies , Cytogenetic Analysis/methods , Humans , Karyotyping , Pilot Projects , Reproducibility of Results
6.
Leukemia ; 20(6): 971-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16598306

ABSTRACT

Signal transducer and activator of transcription (STAT) proteins are phosphorylated and activated by Janus kinases (JAKs). Recently, several groups identified a recurrent somatic point mutation constitutively activating the hematopoietic growth factor receptor-associated JAK2 tyrosine kinase in diverse chronic myeloid disorders - most commonly classic myeloproliferative disorders (MPD), especially polycythemia vera. We hypothesized that the JAK2 V617F mutation might also be present in samples from patients with acute myeloid leukemia (AML), especially erythroleukemia (AML-M6) or megakaryoblastic leukemia (AML-M7), where it might mimic erythropoietin or thrombopoietin signaling. First, we documented STAT3 activation by immunoblotting in AML-M6 and other AML subtypes. Immunoperoxidase staining confirmed phosphorylated STAT3 in malignant myeloblasts (21% of cases, including all AML-M3 samples tested). We then analyzed genomic DNA from 162 AML, 30 B-cell lymphoma, and 10 chronic lymphocytic leukemia (CLL) samples for JAK2 mutations, and assayed a subset for SOCS1 and FLT3 mutations. Janus kinase2 V617F was present in 13/162 AML samples (8%): 10/13 transformed MPD, and three apparent de novo AML (one of 12 AML-M6, one of 24 AML-M7, and one AML-M2 - all mixed clonality). FLT3 mutations were present in 5/32 (16%), while SOCS1 mutations were totally absent. Lymphoproliferative disorder samples were both JAK2 and SOCS1 wild type. Thus, while JAK2 V617F is uncommon in de novo AML and probably does not occur in lymphoid malignancy, unexplained STAT3 activation is common in AML. Janus kinase2 extrinsic regulators and other proteins in the JAK-STAT pathway should be interrogated to explain frequent STAT activation in AML.


Subject(s)
Leukemia, Myeloid/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , STAT3 Transcription Factor/metabolism , Acute Disease , Blotting, Western , Humans , Janus Kinase 2 , Leukemia, Myeloid/metabolism , Phosphorylation , Point Mutation , Signal Transduction/genetics , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , fms-Like Tyrosine Kinase 3/genetics
7.
J Biol Regul Homeost Agents ; 19(1-2): 23-32, 2005.
Article in English | MEDLINE | ID: mdl-16178271

ABSTRACT

Acute myeloid leukemia (AML) and myelodysplasia (MDS) continue to present formidable treatment challenges as there is not yet a "standard approach" that reliably and safely cures the majority of adults with these disorders. Understanding the basic cellular and molecular biology of leukemia is vital to the development of better treatment approaches. All malignant cells, including leukemias, carry survival advantages compared to their normal counterparts. These survival advantages are particularly evident when the cells face both normal environmental stressors as well as extreme stressors of potentially lethal DNA damage. The impact of such stressors is ultimately determined by the signaling pathways and resultant cellular activation, including alterations in cellular growth, differentiation, survival and death. It is our improving understanding of these signaling pathways, both normal and malignant, that provide the targets for novel therapies. In this review article, we will focus on several signaling pathways, as well as the regulation of the cell cycle, that not only represent potentially accessible sites for intervention in myeloid malignancies, but those that are actively being studied.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/therapy , Neural Tube Defects/therapy , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bevacizumab , Carbazoles/therapeutic use , Cell Cycle , Cell Death , Cell Differentiation , Cell Survival , DNA Damage , Drug Resistance , Flavonoids/therapeutic use , Furans , Humans , Indoles/therapeutic use , Piperidines/therapeutic use , Quinolones/therapeutic use , RNA Processing, Post-Transcriptional , Signal Transduction , Staurosporine/analogs & derivatives , Staurosporine/therapeutic use , Stem Cells/metabolism , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
8.
Ann Hematol ; 83 Suppl 1: S87-8, 2004.
Article in English | MEDLINE | ID: mdl-15124688

ABSTRACT

Farnesyltransferase inhibitors (FTIs) are small-molecule inhibitors that selectively inhibit farnesylation of a number of intracellular substrate proteins such as Ras. Preclinical work has revealed their ability to effectively inhibit tumor growth in vitro and in vivo in animal models across a wide range of malignant phenotypes. Myeloid malignancies are appropriate disease targets, in that they express relevant biologic targets, such as Ras, Mitogen-Activated Protein Kinase (MAPK), AKT, and others that may depend upon farnesyl protein transferase (FTase) activity to promote proliferation and survival. Phase I trials in acute leukemias and myelodysplasia have demonstrated biologic and clinical activities as determined by target enzyme inhibition, low toxicity, and both complete and partial responses. As a result, phase II trials have been initiated in a variety of hematologic malignancies and disease settings, in order to further validate clinical activity and to identify downstream signal transduction targets that may be modified by these agents. It is anticipated that these studies will serve to define the optimal roles of FTIs in patients with hematologic malignancies and provide insight into effective methods by which to combine FTIs with other agents.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Leukemia, Myeloid/drug therapy , Clinical Trials as Topic , Farnesyltranstransferase , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Myeloproliferative Disorders/drug therapy , Signal Transduction/drug effects
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.
Cancer Treat Res ; 108: 231-55, 2001.
Article in English | MEDLINE | ID: mdl-11702601

Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid/drug therapy , Neoplasms, Second Primary/drug therapy , Proto-Oncogenes , Transcription Factors , Acute Disease , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/therapeutic use , Chromosome Deletion , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/ultrastructure , Chromosomes, Human, Pair 7/genetics , Chromosomes, Human, Pair 7/ultrastructure , DNA Methylation , DNA-Binding Proteins/genetics , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Forecasting , Genes, Tumor Suppressor , Hematopoietic Stem Cells/drug effects , Histone Deacetylase Inhibitors , Histone-Lysine N-Methyltransferase , Humans , Leukemia, Experimental/etiology , Leukemia, Experimental/pathology , Leukemia, Myeloid/chemically induced , Leukemia, Myeloid/genetics , Leukemia, Radiation-Induced/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Myeloid-Lymphoid Leukemia Protein , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/radiotherapy , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/genetics , Neoplastic Stem Cells/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Oncogene Proteins, Fusion/genetics , Stromal Cells/pathology , Topoisomerase II Inhibitors , Translocation, Genetic , Xenograft Model Antitumor Assays
11.
Ann Intern Med ; 135(7): 477-83, 2001 Oct 02.
Article in English | MEDLINE | ID: mdl-11578150

ABSTRACT

BACKGROUND: Severe aplastic anemia is a life-threatening bone marrow failure disorder. High-dose cyclophosphamide therapy followed by allogeneic bone marrow transplantation cures the disease. However, it requires a suitable donor and carries the risk for graft-versus-host disease. A small pilot study demonstrated that high-dose cyclophosphamide therapy without bone marrow transplantation leads to durable, treatment-free complete remission. OBJECTIVE: To confirm the safety and efficacy of high-dose cyclophosphamide therapy alone in patients with severe aplastic anemia. DESIGN: Uncontrolled clinical trial. SETTING: Three tertiary care hospitals. PATIENTS: 19 patients with untreated severe aplastic anemia. INTERVENTION: Cyclophosphamide, 50 mg/kg of body weight per day for 4 consecutive days. MEASUREMENTS: Probability of response and overall survival were measured. Complete remission was defined as normal blood count for age and sex. Partial remission was defined as independence from transfusion and an absolute neutrophil count greater than 0.5 x 10(9) cells/L without growth factor support. Nonresponders were patients who remained transfusion dependent or died. Relapse was defined as no longer meeting criteria for partial or complete remission. RESULTS: The median time to an absolute neutrophil count of 0.5 x 10(9) cells/L was 49 days. The probability of survival was 84% (95% CI, 59% to 95%) at 24 months. The probability of achieving treatment-free remission was 73% (CI, 51% to 91%) at 24 months, and the probability of achieving complete remission was 65% (CI, 39% to 89%) at 50 months. No responding patients have had relapse or have developed secondary clonal disorders. CONCLUSIONS: High-dose cyclophosphamide therapy without bone marrow transplantation produces durable treatment-free remission in severe aplastic anemia. This approach deserves further study in patients with severe aplastic anemia who are not suitable candidates for allogeneic bone marrow transplantation.


Subject(s)
Anemia, Aplastic/drug therapy , Cyclophosphamide/administration & dosage , Immunosuppressive Agents/administration & dosage , Adolescent , Adult , Aged , Anemia, Aplastic/blood , Anemia, Aplastic/mortality , Anti-Bacterial Agents/therapeutic use , Antiemetics/therapeutic use , Cyclophosphamide/adverse effects , Drug Administration Schedule , Drug Therapy, Combination , Female , Hematopoiesis , Humans , Immunosuppressive Agents/adverse effects , Leukocyte Count , Male , Middle Aged , Neutrophils , Ondansetron/therapeutic use , Pilot Projects , Platelet Transfusion , Prospective Studies , Remission Induction , Survival Analysis
12.
Curr Opin Oncol ; 13(6): 470-6, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11673687

ABSTRACT

Farnesyltransferase inhibitors represent a new class of agents that target signal transduction pathways responsible for the proliferation and survival of diverse malignant cell types. Although these agents were developed to prevent a processing step necessary for membrane attachment and maturation of Ras proteins, recent studies suggest that farnesyltransferase inhibitors block the farnesylation of additional cellular polypeptides, thereby exerting antitumor effects independent of the presence of activating ras gene mutations. Clinical trials of two farnesyltransferase inhibitors--the tricyclic SCH66336 and the methylquinolone R115777--as single agents have demonstrated disease stabilization or objective responses in 10 to 15% of patients with refractory malignancies. Combinations of farnesyltransferase inhibitors with cytotoxic chemotherapies are yielding complete and partial responses in patients with advanced solid tumors. A phase I trial of R115777 in refractory and relapsed acute leukemias induced responses in 8 (32%) of 25 patients with acute myelogenous leukemia (including two complete remissions) and in two of three with chronic myelogenous leukemia in blast crisis. In patients with solid tumors, accessible normal tissues such as peripheral blood lymphocytes or, perhaps more germane to epithelial malignancies, buccal mucosa have provided surrogate tissues that allow confirmation that farnesyltransferase is inhibited in vivo at clinically achievable drug doses. In conjunction with the R115777 acute leukemia trial, serial measurements provided evidence of farnesyltransferase enzyme inhibition, interference with farnesyltransferase function ( ie, protein processing), and blockade of signal transduction pathways in leukemic bone marrow cells. Preclinical studies of farnesyltransferase inhibitor resistance and clinical trials of farnesyltransferase inhibitors in combination with other agents currently are in progress.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Quinolones/pharmacology , Alkyl and Aryl Transferases/metabolism , Clinical Trials as Topic , Farnesyltranstransferase , Humans , Leukemia/drug therapy , Leukemia/physiopathology , Neoplasms/drug therapy , Signal Transduction
13.
Semin Hematol ; 38(3 Suppl 7): 16-23, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11523024

ABSTRACT

Farnesyl protein transferase inhibitors (FTIs) represent a new class of anticancer agents specifically targeting aberrant biologic processes involved with cellular transformation and malignancy. Originally developed to inhibit tumors by preventing activation of oncogenic ras genes via suppression of their posttranslational farnesylation, their anticancer activity appears to stem from their ability to inhibit farnesylation of various proteins that mediate signal transduction, growth, apoptosis, and angiogenesis. The safety, biologic activity, clinical response, and pharmacokinetics of R115777, a potent, orally active FTI, were recently investigated in a phase I dose-ranging study in patients with acute leukemias. Patients with acute myelogenous leukemia (AML), acute lymphocytic leukemia (ALL), or chronic myelogenous leukemia (CML) in blast crisis received R115777 100 mg, 300 mg, 600 mg, 900 mg, or 1,200 mg twice daily for 21 days. Cycles were repeated every 28 to 31 days for up to four cycles. An overall response rate of 29% (10/34 evaluable patients) was observed across all R115777 doses. R115777 was well tolerated; common adverse events included fatigue, increased creatinine, nausea, and neutropenia. Dose-limiting toxicity occurred at 1,200 mg twice daily. Farnesylation of lamin A and HDJ-2, examined as biologic end points, was inhibited by R115777 doses > or = 600 mg twice daily. Pharmacokinetic evaluation suggests that R115777 is concentrated in bone marrow at steady state. The biologic and antitumor activity and favorable tolerability of R115777 support further clinical evaluation alone and in combination therapy in hematologic malignancies.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Hematologic Neoplasms/drug therapy , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase , Humans , Quinolones/pharmacokinetics , Quinolones/pharmacology , Quinolones/therapeutic use , Signal Transduction/drug effects
15.
Blood ; 97(11): 3361-9, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11369625

ABSTRACT

R115777 is a nonpeptidomimetic enzyme-specific inhibitor of farnesyl protein transferase (FT) that was developed as a potential inhibitor of Ras protein signaling, with antitumor activity in preclinical models. This study was a phase 1 trial of orally administered R115777 in 35 adults with poor-risk acute leukemias. Cohorts of patients received R115777 at doses ranging from 100 mg twice daily (bid) to 1200 mg bid for up to 21 days. Dose-limiting toxicity occurred at 1200 mg bid, with central neurotoxicity evidenced by ataxia, confusion, and dysarthria. Non-dose-limiting toxicities included reversible nausea, renal insufficiency, polydipsia, paresthesias, and myelosuppression. R115777 inhibited FT activity at 300 mg bid and farnesylation of FT substrates lamin A and HDJ-2 at 600 mg bid. Extracellular signal-regulated kinase (ERK), an effector enzyme of Ras-mediated signaling, was detected in its phosphorylated (activated) form in 8 (36.4%) of 22 pretreatment marrows and became undetectable in 4 of those 8 after one cycle of treatment. Pharmacokinetics revealed a linear relationship between dose and maximum plasma concentration or area under the curve over 12 hours at all dose levels. Weekly marrow samples demonstrated that R115777 accumulated in bone marrow in a dose-dependent fashion, with large increases in marrow drug levels beginning at 600 mg bid and with sustained levels throughout drug administration. Clinical responses occurred in 10 (29%) of the 34 evaluable patients, including 2 complete remissions. Genomic analyses failed to detect N-ras gene mutations in any of the 35 leukemias. The results of this first clinical trial of a signal transduction inhibitor in patients with acute leukemias suggest that inhibitors of FT may have important clinical antileukemic activity. (Blood. 2001;97:3361-3369)


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Quinolones/therapeutic use , Adult , Aged , Bone Marrow/metabolism , Cohort Studies , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Farnesyltranstransferase , Female , Genes, ras , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Prenylation , Quinolones/adverse effects , Quinolones/pharmacokinetics , Recurrence , Remission Induction , Treatment Outcome
16.
J Natl Cancer Inst Monogr ; (28): 15-23, 2001.
Article in English | MEDLINE | ID: mdl-11158202

ABSTRACT

BACKGROUND: The growing number of human immunodeficiency virus type 1 (HIV-1) infections worldwide and the increasing use of immunosuppressive modalities for organ transplantation have contributed to an epidemic of Kaposi's sarcoma (KS), which has been etiologically linked to human herpesvirus 8 (HHV8) or KS-associated virus. Since the onset of the acquired immunodeficiency syndrome epidemic, inflammation has been recognized as an essential component of KS pathology. HHV8 bears a gene (K1) encoding a transmembrane protein with an immunoreceptor tyrosine-based activation motif. This motif is present in receptors that mediate inflammation. PURPOSE: To dissect the cellular effects of K1 function and the eventual role of K1 in KS, we developed a cell model for studying K1 expression. METHODS: K1 was cloned from BC-3 lymphoma cells. To monitor transcriptional activation, K1 was coexpressed with plasmids containing luciferase under control of various promoters. K1 expression was monitored by indirect immunofluorescence and by combined immunoprecipitation/immunoblot analysis. Inflammatory cytokines were measured by enzyme-linked immunosorbent assay. RESULTS: Cellular transfection of the K1 gene induced reporter expression under control of nuclear factor-kappa B (NF-kappaB), which controls the transcription of numerous proteins involved in inflammation. Treatment of cells with aspirin, an agent that targets this intracellular pathway and blocks cell inflammatory responses, blocked K1-induced NF-kappaB-dependent promoter activity. When a second KS cofactor, i.e., the HIV-1-transactivating gene tat, was coexpressed with K1, we observed an additive effect on NF-kappaB-dependent transcription. K1 transfection stimulated the secretion of cytokines interleukin (IL) 6, granulocyte-macrophage colony-stimulating factor, and IL-12. Cells treated with the conditioned media of K1 transfectants exhibited similar characteristics of K1 transfectants, indicating that a paracrine loop was being activated. CONCLUSION: Thus, K1 may activate cells in which it is expressed, as well as other cells in a paracrine manner. K1 cooperates in signaling with HIV-1 Tat, suggesting that both of the proteins from these viruses converge to reach an enhanced level of inflammation that may underlie progressive KS.


Subject(s)
Cytokines/metabolism , Dermatitis/etiology , Gene Expression Regulation, Viral , Herpesvirus 8, Human/physiology , NF-kappa B/physiology , Sarcoma, Kaposi/complications , Skin Neoplasms/complications , Viral Proteins/physiology , Amino Acid Motifs , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , COS Cells , Chlorocebus aethiops , Culture Media, Conditioned/pharmacology , Cytokines/biosynthesis , Gene Products, tat/physiology , Genes, tat , HIV-1/genetics , Herpesvirus 8, Human/genetics , Humans , Hyperplasia , Lymphoma, Non-Hodgkin/pathology , Lymphoma, Non-Hodgkin/virology , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/physiopathology , Paracrine Communication , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/physiology , Sarcoma, Kaposi/blood supply , Sarcoma, Kaposi/virology , Skin/pathology , Skin/virology , Skin Neoplasms/blood supply , Skin Neoplasms/virology , Transcriptional Activation , Transfection , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/virology , Viral Proteins/chemistry , Viral Proteins/genetics , tat Gene Products, Human Immunodeficiency Virus
17.
Leukemia ; 14(11): 1867-75, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11069021

ABSTRACT

Aberrant expression of homeobox genes has been described in primary leukemia blasts. We recently cloned a new cDNA, BP1, which is a member of the homeobox gene family. BP1 expression was investigated in bone marrow samples from acute myeloid leukemia (AML), acute T cell lymphocytic leukemia (ALL) and pre-B cell ALL. Expression levels of two apparent isoforms of BP1, DLX7 and DLX4, were measured in the same samples. They are weakly if at all detectable in normal bone marrow, PHA-stimulated T cells or B cells. BP1 RNA was highly expressed in 63% of AML cases, including 81% of the pediatric and 47% of the adult cases, and in 32% of T-ALL cases, but was not found in any of the pre-B ALL cases. Coexpression of BP1, DLX7 and DLX4 occurred in a significant number of leukemias. Our data, including co-expression of BP1 with c-myb and GATA-1, markers of early progenitors, suggest that BP1 expression occurs in primitive cells in AML. Analysis of CD34+ and CD34- normal bone marrow cells revealed BP1 is expressed in CD34- cells and virtually extinguished in CD34+ cells. Ectopic expression of BP1 in the leukemia cell line K562 increased clonogenicity, consistent with a role for BP1 in leukemogenesis. The presence of BP1 RNA in leukemic blasts may therefore be a molecular marker for primitive cells and/or may indicate that BP1 is an important upstream factor in an oncogenic pathway.


Subject(s)
Genes, Homeobox , Homeodomain Proteins/genetics , Leukemia/genetics , Neoplasm Proteins/genetics , Oncogene Proteins , Protein Isoforms/genetics , Transcription Factors , Acute Disease , Age Factors , Alternative Splicing , Biomarkers, Tumor/genetics , Bone Marrow Examination , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , DNA, Complementary/genetics , DNA, Neoplasm/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/isolation & purification , Humans , K562 Cells/cytology , Leukemia/metabolism , Molecular Sequence Data , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/isolation & purification , Protein Isoforms/biosynthesis , Protein Isoforms/isolation & purification , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Recombinant Fusion Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Stem Cell Assay
18.
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
19.
Blood ; 96(1): 365-8, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10891476

ABSTRACT

Breast cancer resistance protein (BCRP) is a novel member of the adenosine triphosphate-binding cassette superfamily of transport proteins. Transfection and enforced expression of BCRP in drug-sensitive cells confer resistance to mitoxantrone, doxorubicin, daunorubicin, and topotecan. We studied blast cells from 21 acute leukemia patients (20 acute myeloid leukemia, 1 acute lymphocytic leukemia) for the expression of BCRP mRNA using a quantitative reverse-transcription polymerase chain reaction assay. BCRP mRNA expression varied more than 1000-fold among the samples tested, with low or barely detectable expression in half of the samples. Seven samples (33%) had relatively high expression of BCRP mRNA. High expression of BCRP did not correlate strongly with high expression of P-glycoprotein, suggesting that BCRP may cause resistance to certain antileukemic drugs in P-glycoprotein-negative cases. High expression of BCRP mRNA is sufficiently frequent in AML to warrant more extensive investigations to determine the relation of disease subtype and treatment outcome to BCRP expression and function.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Blast Crisis/pathology , Leukemia, Myeloid/pathology , Neoplasm Proteins , Transcription, Genetic , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/analysis , Acute Disease , Adult , Aged , Blast Crisis/metabolism , Bone Marrow/metabolism , Bone Marrow/pathology , Breast Neoplasms , Cells, Cultured , Female , HL-60 Cells , Humans , Leukemia, Myeloid/genetics , Male , Middle Aged , Tumor Cells, Cultured
20.
Oncology (Williston Park) ; 14(5): 659-66, 671-2; discussion 672-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10853459

ABSTRACT

Several recent studies have addressed the management of infectious problems in patients with acute leukemia. Although those studies have served to emphasize the fundamental management principles formulated and proven almost 30 years ago, they have also contributed important new insights. This article describes recent developments in the management of infectious illnesses in patients who are neutropenic due to leukemia or its treatment. The discussion will focus on the increasing armamentarium of antimicrobial drugs and adjunctive agents. These expanding therapeutic options must be viewed in the context of newly emerging resistant organisms and special problems, such as the increased use of indwelling venous catheters.


Subject(s)
Anti-Infective Agents/therapeutic use , Antineoplastic Agents/adverse effects , Leukemia/drug therapy , Neutropenia/complications , Opportunistic Infections/drug therapy , Opportunistic Infections/prevention & control , Acute Disease , Antifungal Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Infections/prevention & control , Humans , Mycoses/drug therapy , Mycoses/prevention & control , Neutropenia/chemically induced , Opportunistic Infections/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...