Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 440
Filter
1.
Leukemia ; 32(4): 920-930, 2018 04.
Article in English | MEDLINE | ID: mdl-29099493

ABSTRACT

Acalabrutinib, a highly selective Bruton's tyrosine kinase inhibitor, is associated with high overall response rates and durable remission in previously treated chronic lymphocytic leukemia (CLL); however, complete remissions were limited. To elucidate on-target and pharmacodynamic effects of acalabrutinib, we evaluated several laboratory endpoints, including proteomic changes, chemokine modulation and impact on cell migration. Pharmacological profiling of samples from acalabrutinib-treated CLL patients was used to identify strategies for achieving deeper responses, and to identify additive/synergistic combination regimens. Peripheral blood samples from 21 patients with relapsed/refractory CLL in acalabrutinib phase I (100-400 mg/day) and II (100 mg BID) clinical trials were collected prior to and on days 8 and 28 after treatment initiation and evaluated for plasma chemokines, reverse phase protein array, immunoblotting and pseudoemperipolesis. The on-target pharmacodynamic profile of acalabrutinib in CLL lymphocytes was comparable to ibrutinib in measures of acalabrutinib-mediated changes in CCL3/CCL4 chemokine production, migration assays and changes in B-cell receptor signaling pathway proteins and other downstream survival proteins. Among several CLL-targeted agents, venetoclax, when combined with acalabrutinib, showed optimal complementary activity in vitro, ex vivo and in vivo in TCL-1 adoptive transfer mouse model system of CLL. These findings support selective targeting and combinatorial potential of acalabrutinib.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Adenine/analogs & derivatives , Adoptive Transfer/methods , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Benzamides/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cell Movement/drug effects , Chemokine CCL3/metabolism , Chemokine CCL4/metabolism , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Combined Modality Therapy/methods , Drug Resistance, Neoplasm/drug effects , Humans , Mice , Piperidines , Protein Kinase Inhibitors/administration & dosage , Protein-Tyrosine Kinases/metabolism , Proteomics , Pyrazines/administration & dosage , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Signal Transduction/drug effects , Sulfonamides/administration & dosage
2.
Blood Cancer J ; 6(9): e465, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27588518

ABSTRACT

Approximately 10-20% of chronic lymphocytic leukemia (CLL) patients exhibit del(11q22-23) before treatment, this cohort increases to over 40% upon progression following chemoimmunotherapy. The coding sequence of the DNA damage response gene, ataxia-telangiectasia-mutated (ATM), is contained in this deletion. The residual ATM allele is frequently mutated, suggesting a relationship between gene function and clinical response. To investigate this possibility, we sought to develop and validate an assay for the function of ATM protein in these patients. SMC1 (structural maintenance of chromosomes 1) and KAP1 (KRAB-associated protein 1) were found to be unique substrates of ATM kinase by immunoblot detection following ionizing radiation. Using a pool of eight fluorescence in situ hybridization-negative CLL samples as a standard, the phosphorylation of SMC1 and KAP1 from 46 del (11q22-23) samples was analyzed using normal mixture model-based clustering. This identified 13 samples (28%) that were deficient in ATM function. Targeted sequencing of the ATM gene of these samples, with reference to genomic DNA, revealed 12 somatic mutations and 15 germline mutations in these samples. No strong correlation was observed between ATM mutation and function. Therefore, mutation status may not be taken as an indicator of ATM function. Rather, a direct assay of the kinase activity should be used in the development of therapies.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Chromosome Deletion , Chromosomes, Human, Pair 11 , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mutation , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , DNA Methylation , Gene Deletion , Germ-Line Mutation , Humans , Phosphorylation , Promoter Regions, Genetic , Tripartite Motif-Containing Protein 28/metabolism
4.
Oncogene ; 35(43): 5663-5673, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27065330

ABSTRACT

Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in the western countries and is currently incurable due, in part, to difficulty in eliminating the leukemia cells protected by stromal microenvironment. Based on previous observations that CLL cells exhibit mitochondrial dysfunction and altered lipid metabolism and that carnitine palmitoyltransferases (CPT) have a major role in transporting fatty acid into mitochondria to support cancer cell metabolism, we tested several clinically relevant inhibitors of lipid metabolism for their ability to eliminate primary CLL cells. We discovered that perhexiline, an antiangina agent that inhibits CPT, was highly effective in killing CLL cells in stromal microenvironment at clinically achievable concentrations. These effective concentrations caused low toxicity to normal lymphocytes and normal stromal cells. Mechanistic study revealed that CLL cells expressed high levels of CPT1 and CPT2. Suppression of fatty acid transport into mitochondria by inhibiting CPT using perhexiline resulted in a depletion of cardiolipin, a key component of mitochondrial membranes, and compromised mitochondrial integrity, leading to rapid depolarization and massive CLL cell death. The therapeutic activity of perhexiline was further demonstrated in vivo using a CLL transgenic mouse model. Perhexiline significantly prolonged the overall animal survival by only four drug injections. Our study suggests that targeting CPT using an antiangina drug is able to effectively eliminate leukemia cells in vivo, and is a novel therapeutic strategy for potential clinical treatment of CLL.


Subject(s)
Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Perhexiline/pharmacology , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents/pharmacology , Cardiolipins/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Gene Expression , Glucose/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Models, Biological , Oxygen Consumption , Xenograft Model Antitumor Assays
5.
Ann Oncol ; 27(6): 1100-1106, 2016 06.
Article in English | MEDLINE | ID: mdl-26912560

ABSTRACT

BACKGROUND: Information on the impact of other cancers (OCs) in long-term survivors (LTSs) of chronic lymphocytic leukemia (CLL) is limited. PATIENTS AND METHODS: Patients with CLL who survived >10 years were defined as LTSs of CLL. We calculated standardized incidence ratios (SIRs) to compare the incidence of OC in LTS of CLL versus the general population. A multivariable model was used to identify independent predictors of OC. Overall survival was analyzed as a function of the presence of OC. RESULTS: Among 797 LTSs of CLL, the cumulative frequency of OC was 36%, similar between 570 patients (72%) who required treatment for CLL (TRT) and 227 (28%) who remained untreated (UT). The most common OC in both groups was non-melanoma skin cancer, followed by prostate cancer, breast cancer, melanoma, lung cancer, and leukemia in TRT patients, and by prostate cancer, breast cancer, melanoma, lung cancer, and gastrointestinal tumors in the UT group. The SIR for all OC was 1.2 (P = 0.034). It was higher in males (SIR 1.31; P = 0.013) and patients <60 years (SIR 1.27; P = 0.027). A higher SIR was shown for secondary leukemia, melanoma, and head-and-neck cancers, whereas a lower SIR was found for gastrointestinal and bladder cancers. Independent predictors of OC development were advanced age, male gender, and lower platelets. The survival of patients with OC was 16.2 months and that of patients without OC 22.9 years. CONCLUSIONS: LTSs of CLL have an increased incidence of OC compared with the general population. CLL therapy is not a risk factor for OC in LTSs of CLL. The presence of an OC in these patients may be associated with shorter survival.


Subject(s)
Cancer Survivors , Leukemia, Lymphocytic, Chronic, B-Cell/epidemiology , Neoplasms, Second Primary/epidemiology , Prognosis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Female , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/pathology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/epidemiology , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasms, Second Primary/drug therapy , Neoplasms, Second Primary/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/pathology , Risk Factors , SEER Program
6.
Leukemia ; 30(2): 325-30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26290497

ABSTRACT

Acute leukemia (AL) and myelodysplastic syndrome (MDS) are uncommon in chronic lymphocytic leukemia (CLL). We retrospectively identified 95 patients with CLL, also diagnosed with AL (n=38) or MDS (n=57), either concurrently (n=5) or subsequent (n=90) to CLL diagnosis and report their outcomes. Median number of CLL treatments prior to AL and MDS was 2 (0-9) and 1 (0-8), respectively; the most common regimen was purine analog combined with alkylating agent±CD20 monoclonal antibody. Twelve cases had no prior CLL treatment. Among 38 cases with AL, 33 had acute myelogenous leukemia (AML), 3 had acute lymphoid leukemia (ALL; 1 Philadelphia chromosome positive), 1 had biphenotypic and 1 had extramedullary (bladder) AML. Unfavorable AML karyotype was noted in 26, and intermediate risk in 7 patients. There was no association between survival from AL and number of prior CLL regimens or karyotype. Expression of CD7 on blasts was associated with shorter survival. Among MDS cases, all International Prognostic Scoring System (IPSS) were represented; karyotype was unfavorable in 36, intermediate in 6 and favorable in 12 patients; 10 experienced transformation to AML. Shorter survival from MDS correlated with higher risk IPSS, poor-risk karyotype and increased number of prior CLL treatments. Overall, outcomes for patients with CLL subsequently diagnosed with AL or MDS were very poor; AL/MDS occurred without prior CLL treatment. Effective therapies for these patients are desperately needed.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Myeloid, Acute/mortality , Myelodysplastic Syndromes/mortality , Aged , Female , Humans , Karyotype , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Male , Retrospective Studies
7.
Leukemia ; 29(9): 1811-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25917267

ABSTRACT

The functional relevance of the B-cell receptor (BCR) and the evolution of protein kinases as therapeutic targets have recently shifted the paradigm for treatment of B-cell malignancies. Inhibition of p110δ with idelalisib has shown clinical activity in chronic lymphocytic leukemia (CLL). The dynamic interplay of isoforms p110δ and p110γ in leukocytes support the hypothesis that dual blockade may provide a therapeutic benefit. IPI-145, an oral inhibitor of p110δ and p110γ isoforms, sensitizes BCR-stimulated and/or stromal co-cultured primary CLL cells to apoptosis (median 20%, n=57; P<0.0001) including samples with poor prognostic markers, unmutated IgVH (n=28) and prior treatment (n=15; P<0.0001). IPI-145 potently inhibits the CD40L/IL-2/IL-10 induced proliferation of CLL cells with an IC50 in sub-nanomolar range. A corresponding dose-responsive inhibition of pAKT(Ser473) is observed with an IC50 of 0.36 nM. IPI-145 diminishes the BCR-induced chemokines CCL3 and CCL4 secretion to 17% and 37%, respectively. Pre-treatment with 1 µM IPI-145 inhibits the chemotaxis toward CXCL12; reduces pseudoemperipolesis to median 50%, inferring its ability to interfere with homing capabilities of CLL cells. BCR-activated signaling proteins AKT(Ser473), BAD(Ser112), ERK(Thr202/Tyr204) and S6(Ser235/236) are mitigated by IPI-145. Importantly, for clinical development in hematological malignancies, IPI-145 is selective to CLL B cells, sparing normal B- and T-lymphocytes.


Subject(s)
Apoptosis/drug effects , Isoquinolines/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Purines/pharmacology , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Adenine/analogs & derivatives , Aged , Aged, 80 and over , Bone Marrow Cells/metabolism , Case-Control Studies , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Chemokine CCL3/metabolism , Chemokine CCL4/metabolism , Chemotaxis/drug effects , Chemotaxis/immunology , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Phosphorylation , Piperidines , Prognosis , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinazolinones/pharmacology , Receptors, Antigen, B-Cell/metabolism , Stromal Cells/metabolism
8.
Leukemia ; 29(4): 783-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25138588

ABSTRACT

The BTK (Bruton's tyrosine kinase) inhibitor ibrutinib is associated with an increased risk of bleeding. A previous study reported defects in collagen- and adenosine diphosphate (ADP)-dependent platelet responses when ibrutinib was added ex vivo to patient samples. Whereas the collagen defect is expected given the central role of BTK in glycoprotein VI signaling, the ADP defect lacks a mechanistic explanation. In order to determine the real-life consequences of BTK platelet blockade, we performed light transmission aggregometry in 23 patients receiving ibrutinib treatment. All patients had reductions in collagen-mediated platelet aggregation, with a significant association between the degree of inhibition and the occurrence of clinical bleeding or bruising (P=0.044). This collagen defect was reversible on drug cessation. In contrast to the previous ex vivo report, we found no in vivo ADP defects in subjects receiving standard doses of ibrutinib. These results establish platelet light transmission aggregometry as a method for gauging, at least qualitatively, the severity of platelet impairment in patients receiving ibrutinib treatment.


Subject(s)
Antineoplastic Agents/adverse effects , Blood Platelets/drug effects , Collagen/pharmacology , Hemorrhage/diagnosis , Platelet Aggregation/drug effects , Pyrazoles/adverse effects , Pyrimidines/adverse effects , Adenine/analogs & derivatives , Adenosine Diphosphate/pharmacology , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Blood Platelets/pathology , Cells, Cultured , Female , Hemorrhage/chemically induced , Hemorrhage/pathology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/pathology , Male , Middle Aged , Piperidines , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Severity of Illness Index
9.
Leukemia ; 28(1): 118-28, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23608884

ABSTRACT

Chronic lymphocytic leukemia (CLL) patients with deletion of chromosome 17p, where the p53 gene is located, often develop more aggressive disease with poor clinical outcomes. To investigate the underlying mechanisms for the highly malignant phenotype of 17p- CLL and to facilitate in vivo evaluation of potential drugs against CLL with p53 deletion, we have generated a mouse model with TCL1-Tg:p53(-/-) genotype. These mice develop B-cell leukemia at an early age with an early appearance of CD5+ / IgM+ B cells in the peritoneal cavity and spleen, and exhibit an aggressive path of disease development and drug resistance phenotype similar to human CLL with 17p deletion. The TCL1-Tg:p53(-/-) leukemia cells exhibit higher survival capacity and are more drug resistant than the leukemia cells from TCL1-Tg:p53wt mice. Analysis of microRNA expression reveals that p53 deletion resulted in a decrease of miR-15a and miR-16-1, leading to an elevated expression of Mcl-1. Primary leukemia cells from CLL patients with 17p deletion also show a decrease in miR-15a/miR-16-1 and an increase in Mcl-1. Our study suggests that the p53/miR15a/16-1/Mcl-1 axis may be an important pathway that regulates Mcl-1 expression and contributes to drug resistance and aggressive phenotype in CLL cells with loss of p53.


Subject(s)
Disease Models, Animal , Genes, p53 , MicroRNAs/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Line, Tumor , Humans , Mice , Mice, Transgenic
11.
Leukemia ; 26(8): 1812-20, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22460735

ABSTRACT

The tissue microenvironment in chronic lymphocytic leukemia (CLL) has an increasingly recognized role in disease progression, but the molecular mechanisms of cross talk between CLL cells and their microenvironment remain incompletely defined. Bone marrow stromal cells (BMSC) protect CLL cells from apoptosis in a contact-dependent fashion, and have been used for the identification of key pathways such as the CXCR4-CXCL12 axis. To further dissect the molecular impact of BMSC on survival and the molecular activation signature of CLL cells, we co-cultured CLL cells with different BMSC. Gene expression profiling of CLL cells revealed that the lymphoid proto-oncogene TCL1 was among the top genes upregulated in CLL cells by BMSC. TCL1 mRNA and protein upregulation by BMSC was paralleled by decreases of TCL1-interacting FOS/JUN, and confirmed by qRT-PCR, immunoblotting, immunoprecipitations, and flow cytometry. Stroma mediated increases in TCL1 were also associated with decreased levels of TCL1-regulatory micro-RNAs (miR-29b, miR-181b, miR-34b). These findings demonstrate that the microenvironment has a proactive role in the regulation of the known signaling enhancer and pro-survival molecule TCL1 in CLL. This provides a further rationale for therapeutically targeting the cross talk between CLL and BMSC.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , MicroRNAs/genetics , Proto-Oncogene Proteins/genetics , Stromal Cells/metabolism , Transcription Factor AP-1/metabolism , Bone Marrow Cells/metabolism , Cell Communication , Cell Survival/drug effects , Cluster Analysis , Coculture Techniques , Gene Expression Profiling , Humans , Interleukin-4/pharmacology , MicroRNAs/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Transcription, Genetic
12.
Leukemia ; 26(7): 1576-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22362000

ABSTRACT

Syk is a protein tyrosine kinase that couples B-cell receptor (BCR) activation with downstream signaling pathways, affecting cell survival and proliferation. Moreover, Syk is involved in BCR-independent functions, such as B-cell migration and adhesion. In chronic lymphocytic leukemia (CLL), Syk becomes activated by external signals from the tissue microenvironment, and was targeted in a first clinical trial with R788 (fostamatinib), a relatively nonspecific Syk inhibitor. Here, we characterize the activity of two novel, highly selective Syk inhibitors, PRT318 and P505-15, in assays that model CLL interactions with the microenvironment. PRT318 and P505-15 effectively antagonize CLL cell survival after BCR triggering and in nurse-like cell-co-cultures. Moreover, they inhibit BCR-dependent secretion of the chemokines CCL3 and CCL4 by CLL cells, and leukemia cell migration toward the tissue homing chemokines CXCL12, CXCL13, and beneath stromal cells. PRT318 and P505-15 furthermore inhibit Syk and extracellular signal-regulated kinase phosphorylation after BCR triggering. These findings demonstrate that the selective Syk inhibitors PRT318 and P505-15 are highly effective for inhibition of CLL survival and tissue homing circuits, and support the therapeutic development of these agents in patients with CLL, other B-cell malignancies and autoimmune disorders.


Subject(s)
Cell Movement/drug effects , Cyclohexylamines/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Chemotaxis , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Phosphorylation/drug effects , Protein-Tyrosine Kinases/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Syk Kinase , Tumor Cells, Cultured
14.
Leukemia ; 22(6): 1191-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18385754

ABSTRACT

Mutation of Bcr-Abl is an important mechanism by which chronic myelogenous leukemia (CML) cells become resistant to Gleevec. The T315I mutation is clinically significant since CML cells harboring this mutation are insensitive to Gleevec and other Bcr-Abl-targeted drugs. Identification of new agents capable of effectively killing CML cells with T315I mutation would have important therapeutic implications in Gleevec-resistant CML. Here, we showed that beta-phenylethyl isothiocyanate (PEITC), a natural compound found in vegetables, is effective in killing CML cells expressing T315I BCR-ABL. Treatment of leukemia cell lines harboring wild-type or mutant Bcr-Abl with 10 microM PEITC resulted in an elevated ROS stress and a redox-mediated degradation of the BCR-ABL protein, leading to massive death of the leukemia cells. Antioxidant NAC attenuated the PEITC-induced oxidative stress in CML cells and prevented the degradation of BCR-ABL, caspase-3 activation and cell death. We further showed that the ROS-induced degradation of BCR-ABL was mediated partially by caspase-3 and the proteasome pathway. The ability of PEITC to effectively kill T315I-positive CML cells was further confirmed using primary leukemia cells isolated from CML patients. Our results suggest that PEITC is a promising compound capable of killing Gleevec-resistant CML cells through a ROS-mediated mechanism and warrants further investigations.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/genetics , Isothiocyanates/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Aldehyde Dehydrogenase/antagonists & inhibitors , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Benzamides , Caspase 3/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Fusion Proteins, bcr-abl/metabolism , Glutathione/metabolism , Humans , Imatinib Mesylate , Immunoblotting , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mutation/genetics , Oxidation-Reduction , Precursor Cells, B-Lymphoid/drug effects , Precursor Cells, B-Lymphoid/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Reactive Oxygen Species/metabolism
15.
Br J Clin Pharmacol ; 64(3): 278-91, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17506867

ABSTRACT

AIMS: To characterize alemtuzumab pharmacokinetics and its exposure-response relationship with white blood cell (WBC) count in patients with B-cell chronic lymphocytic leukaemia (CLL). METHODS: Nonlinear mixed effects models were used to characterize plasma concentration-time data and WBC count-time data from 67 patients. Logistic regression was used to relate summary measures of drug exposure to tumour response. RESULTS: Alemtuzumab pharmacokinetics were best characterized by a two-compartment model with nonlinear elimination where V(max) (microg h(-1)) was [1020 x (WBC count/10 x 10(9) l(-1))(0.194)], K(m) was 338 microg l(-1), V(1) was 11.3 l, Q was 1.05 l h(-1) and V(2) was 41.5 l. Intersubject variability (ISV) in V(max), K(m), V(1) and V(2) was 32%, 145%, 84% and 179%, respectively. The reduction in WBC over time was modelled by a stimulatory loss indirect response model with values of 18.2 for E(max), 306 microg l(-1) for EC(50), 1.56 x 10(9) cells l(-1) h(-1) for K(in) and 0.029 per h for K(out). The probability of achieving a complete or partial response was >/=50% when the maximal trough concentration exceeded 13.2 microg ml(-1) or when AUC(0-tau) exceeded 484 microg h(-1) ml(-1). CONCLUSIONS: Alemtuzumab displayed time- and concentration-dependent pharmacokinetics with large interpatient variability, both in pharmacokinetics and pharmacodynamics, which was probably reflective of differences in tumour burden among patients. A direct relationship between maximal trough concentrations and clinical outcomes was observed, with increasing alemtuzumab exposure resulting in a greater probability of positive tumour response.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Immunoglobulin G/blood , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Adult , Aged , Alemtuzumab , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukocyte Count , Logistic Models , Male , Middle Aged , Models, Biological , Nonlinear Dynamics
16.
Bone Marrow Transplant ; 39(3): 157-64, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17245424

ABSTRACT

Pneumocandins have concentration-dependent antifungal activity and higher dose of caspofungin (HD-CAP) in combination with other licensed antifungal therapy (OLAT) may improve response. Thirty-four patients who received HD-CAP were compared with 63 patients who received standard dose (SD)-CAP. There were no differences between the groups in either patient or disease characteristics. Significantly more patients in the HD-CAP arm had extrapulmonary infections (29 vs 8% in SD group; P=0.0053), and non-Aspergillus species infection (21 vs 6%; P=0.05) and had received prior antifungal therapy (71 vs 33%; P=0.0004). No serious adverse reactions were noted in patients receiving HD- or SD-CAP therapy. Twelve weeks after treatment commenced 44% had a complete or partial response compared with 29% in SD-CAP group (P=0.1). Logistic regression analysis showed a significant probability of a favorable outcome at 12 weeks in patients who received HD-CAP (OR 3.066, 95% CI, 1.092-8.61; P=0.033). This may in part reflect higher number of patients in HD group had received granulocyte-macrophage colony-stimulating factor (41 vs 14% in SD group; P=0.04) and/or interferon gamma (26 vs 5% in SD group; P=0.003) immune enhancement. Further studies are needed to evaluate efficacy of HD-CAP in severely immunosuppressed cancer patients with invasive fungal infections.


Subject(s)
Antifungal Agents/therapeutic use , Hematologic Neoplasms/complications , Hematologic Neoplasms/drug therapy , Hematopoietic Stem Cell Transplantation/methods , Peptides, Cyclic/administration & dosage , Adult , Aged , Antifungal Agents/toxicity , Caspofungin , Dose-Response Relationship, Drug , Drug Therapy, Combination , Echinocandins , Female , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infections/chemically induced , Interferon-gamma/therapeutic use , Lipopeptides , Male , Middle Aged , Peptides, Cyclic/toxicity , Remission Induction , Retrospective Studies , Treatment Outcome
18.
Leukemia ; 20(4): 610-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16482209

ABSTRACT

17-Allylamino-17-demethoxygeldanamycin (17-AAG) is a new anticancer agent currently in clinical trials. The ability of 17-AAG to abrogate the function of heat-shock protein Hsp90 and modulate cellular sensitivity to anticancer agents has prompted recent research to use this compound in drug combination therapy. Here we report that 17-AAG has striking opposite effects on the activity of arsenic trioxide (ATO) and ara-C. Combination of 17-AAG with ATO exhibited a synergistic effect in leukemia cells, whereas coincubation of 17-AAG and ara-C showed antagonistic activity. Mechanistic studies revealed that ATO exerted cytotoxic action by reactive oxygen species generation, and activated Akt survival pathway. 17-AAG abrogated Akt activation and enhanced the activity of ATO. In contrast, treatment of leukemia cells with 17-AAG caused a G1 arrest, a decrease in DNA synthesis and reduced ara-C incorporation into DNA, leading to antagonism. The ability of 17-AAG to enhance the antileukemia activity of ATO was further demonstrated in primary leukemia cells isolated from patients with acute myeloid leukemia and chronic lymphocytic leukemia, including cells from refractory patients. Our data suggest that combination of 17-AAG and ATO may be an effective therapeutic regimen. Caution should be exercised in using 17-AAG together with ara-C, as their combination effects are schedule dependent.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Benzoquinones/pharmacology , Cytarabine/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Myeloid/metabolism , Oxides/pharmacology , Acute Disease , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals/antagonists & inhibitors , Cytarabine/antagonists & inhibitors , DNA/biosynthesis , DNA/drug effects , DNA/metabolism , Drug Administration Schedule , Drug Antagonism , Drug Screening Assays, Antitumor , Drug Synergism , Drug Therapy, Combination , G1 Phase/drug effects , HSP90 Heat-Shock Proteins/physiology , HeLa Cells , Humans , Jurkat Cells , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Myeloid/drug therapy , Oxides/antagonists & inhibitors , Sensitivity and Specificity , Structure-Activity Relationship , Tumor Cells, Cultured
19.
Leukemia ; 19(12): 2153-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16193082

ABSTRACT

The mammalian target of rapamycin (mTOR) pathway plays important roles in regulating nutrient metabolism and promoting the growth and survival of cancer cells, which exhibit increased glycolysis for ATP generation. In this study, we tested the hypothesis that inhibition of the mTOR pathway and glycolysis would synergistically impact the energy metabolism in cancer cells and may serve as an effective therapeutic strategy to kill malignant cells. Using human lymphoma cells and leukemia cells, we demonstrated that the combination of rapamycin, an mTOR inhibitor, with a glycolytic inhibitor produced synergistic cytotoxic effect, as evidenced by apoptosis and cell growth inhibition assays. Mechanistic studies showed that inhibition of the mTOR pathway by rapamycin alone sufficiently suppressed the phosphorylation of the downstream molecules p70S6K and 4E-BP-1, but only caused a moderate cytostatic effect. Combination of mTOR inhibition and blockage of glycolysis synergistically suppressed glucose uptake and severely depleted cellular ATP pools, leading to significant enhancement of cell killing. In contrast, combination of rapamycin and ara-C did not increase cytotoxicity in vitro. Our findings suggest that targeting mTOR pathway in combination with inhibition of glycolysis may be an effective therapeutic strategy for hematological malignancies. This mechanism-based drug combination warrants further investigation in preclinical and clinical settings.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Leukemia/metabolism , Lymphoma/metabolism , Protein Kinases/drug effects , Sirolimus/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cytarabine/pharmacology , Drug Synergism , Energy Metabolism/drug effects , Glycolysis/drug effects , Humans , Leukemia/drug therapy , Leukemia/pathology , Lymphoma/drug therapy , Lymphoma/pathology , TOR Serine-Threonine Kinases , Tumor Cells, Cultured
20.
Leukemia ; 19(10): 1788-93, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16094420

ABSTRACT

Patients with chronic lymphocytic leukemia (CLL) have defects in both cellular and humoral immunity. Since CD152 (CTLA-4) plays a critical role in downregulating T-cell responses, we studied the expression of surface and cytoplasmic CD152 (sCD152 and cCD152, respectively) in freshly isolated T cells from treatment-naïve patients with CLL. CD4+ and CD8+ T cells from these patients demonstrated significantly increased sCD152 and cCD152 compared to normal donors. Furthermore, these patients had an increased proportion of the regulatory CD4(+)/CD25(+)/CD152+ subset that correlated with advanced Rai stage, unfavorable cytogenetics and low serum IgG and IgA levels. The expression of sCD152 by T cells also correlated with ZAP-70 expression by CLL B cells. The proportion of CD4(+)/CD25+ cells was also correlated with unmutated immunoglobulin heavy chain variable gene status. Blockade of CD152 with monoclonal antibody (mAb) in proliferation assays was associated with potent T-cell proliferation in response to autologous and allogeneic CD40-activated CLL B cells. In summary, T cells from patients with CLL may be primed for anergy by expressing increased amounts of CD152; anti-CD152 mAb may represent a therapeutic opportunity to enhance an immune response against autologous leukemia cells.


Subject(s)
Antigens, Differentiation/metabolism , Gene Expression Regulation, Neoplastic , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , T-Lymphocytes/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/immunology , Antigens, CD , Antigens, Neoplasm/immunology , CTLA-4 Antigen , Case-Control Studies , Cell Proliferation , Coculture Techniques , Female , Flow Cytometry , Humans , Kinetics , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Lymphocyte Activation , Male , Middle Aged , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...