Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Br J Cancer ; 115(4): 442-53, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27441498

ABSTRACT

BACKGROUND: Albumin-bound paclitaxel (nab-paclitaxel, nab-PTX) plus gemcitabine (GEM) combination has demonstrated efficient antitumour activity and statistically significant overall survival of patients with metastatic pancreatic ductal adenocarcinoma (PDAC) compared with GEM monotherapy. This regimen is currently approved as a standard of care treatment option for patients with metastatic PDAC. It is unclear whether cremophor-based PTX combined with GEM provide a similar level of therapeutic efficacy in PDAC. METHODS: We comprehensively explored the antitumour efficacy, effect on metastatic dissemination, tumour stroma and survival advantage following GEM, PTX and nab-PTX as monotherapy or in combination with GEM in a locally advanced, and a highly metastatic orthotopic model of human PDAC. RESULTS: Nab-PTX treatment resulted in significantly higher paclitaxel tumour plasma ratio (1.98-fold), robust stromal depletion, antitumour efficacy (3.79-fold) and survival benefit compared with PTX treatment. PTX plus GEM treatment showed no survival gain over GEM monotherapy. However, nab-PTX in combination with GEM decreased primary tumour burden, metastatic dissemination and significantly increased median survival of animals compared with either agents alone. These therapeutic effects were accompanied by depletion of dense fibrotic tumour stroma and decreased proliferation of carcinoma cells. Notably, nab-PTX monotherapy was equivalent to nab-PTX plus GEM in providing survival advantage to mice in a highly aggressive metastatic PDAC model, indicating that nab-PTX could potentially stop the progression of late-stage pancreatic cancer. CONCLUSIONS: Our data confirmed that therapeutic efficacy of PTX and nab-PTX vary widely, and the contention that these agents elicit similar antitumour response was not supported. The addition of PTX to GEM showed no survival advantage, concluding that a clinical combination of PTX and GEM may unlikely to provide significant survival advantage over GEM monotherapy and may not be a viable alternative to the current standard-of-care nab-PTX plus GEM regimen for the treatment of PDAC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Kidney Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Splenic Neoplasms/drug therapy , Albumins/administration & dosage , Animals , Carcinoma, Pancreatic Ductal/secondary , Cell Proliferation , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Humans , Kidney Neoplasms/secondary , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Neoplasm Metastasis , Neovascularization, Pathologic , Paclitaxel/administration & dosage , Pancreatic Neoplasms/pathology , Polyethylene Glycols/administration & dosage , Splenic Neoplasms/secondary , Xenograft Model Antitumor Assays , Gemcitabine
2.
Clin Cancer Res ; 21(21): 4811-8, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26169969

ABSTRACT

PURPOSE: nab-Paclitaxel plus gemcitabine was superior to gemcitabine alone for patients with metastatic pancreatic cancer (MPC) in the phase III MPACT trial. This study evaluated the association of secreted protein acidic and rich in cysteine (SPARC) levels with efficacy as an exploratory endpoint. EXPERIMENTAL DESIGN: Patients with previously untreated MPC (N = 861) received nab-paclitaxel plus gemcitabine or gemcitabine alone. Baseline SPARC level was measured in the tumor stroma and epithelia (archival biopsies) and plasma. Experiments were performed in pancreatic cancer mouse models in which SPARC was intact or deleted. RESULTS: SPARC was measured in the tumor stroma of 256 patients (30%), the tumor epithelia of 301 patients (35%), and plasma of 343 patients (40%). Stroma-evaluable samples were from metastases (71%), from the pancreas (11%), or of unidentifiable origin (insufficient tissue to determine; 17%). For all patients, stromal SPARC level [high (n = 71) vs. low (n = 185)] was not associated with overall survival (OS; HR, 1.019; P = 0.903); multivariate analysis confirmed this lack of association. There was no association between stromal SPARC level and OS in either treatment arm. Neither tumor epithelial SPARC nor plasma SPARC was associated with OS. Results from a SPARC knockout mouse model treated with nab-paclitaxel plus gemcitabine revealed no correlation between SPARC expression and tumor progression or treatment efficacy. CONCLUSIONS: SPARC levels were not associated with efficacy in patients with MPC. This exploratory analysis does not support making treatment decisions regarding nab-paclitaxel plus gemcitabine or gemcitabine alone in MPC based on SPARC expression.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Gene Expression , Osteonectin/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Albumins/administration & dosage , Animals , Biomarkers , Deoxycytidine/administration & dosage , Deoxycytidine/therapeutic use , Disease Models, Animal , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Mice, Knockout , Neoplasm Metastasis , Osteonectin/blood , Osteonectin/metabolism , Paclitaxel/administration & dosage , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Stromal Cells/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays , Gemcitabine
3.
Bioorg Med Chem Lett ; 25(12): 2514-8, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25971769

ABSTRACT

A series of bicyclic pyrazole carboxamides was synthesized and tested for inhibitory activity against the class III deacetylase sirtuin enzymes. Moderate to low micromolar inhibitory activities were obtained against SIRT1 and SIRT2. These bicyclic pyrazole compounds represent a new class of sirtuin inhibitors with a preference for SIRT1 over SIRT2.


Subject(s)
Bridged Bicyclo Compounds/chemistry , Pyrazoles/chemistry , Sirtuin 1/antagonists & inhibitors , Sirtuin 2/antagonists & inhibitors , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Drug Evaluation, Preclinical , Molecular Dynamics Simulation , Protein Binding , Pyrazoles/chemical synthesis , Pyrazoles/metabolism , Sirtuin 1/metabolism , Sirtuin 2/metabolism , Structure-Activity Relationship
4.
Br J Haematol ; 165(6): 870-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24635723

ABSTRACT

Sotatercept (ACE-011), a recombinant human fusion protein containing the extracellular domain of the human Activin receptor IIA, binds to and inhibits activin and other members of the transforming growth factor -ß (TGF-ß) superfamily. Administration of sotatercept led to a rapid and sustained increase in red blood cell (RBC) count and haemoglobin (Hb) in healthy volunteers (phase I clinical trials), but the mechanism is not fully understood. Mice treated with RAP-011 (murine ortholog of ACE-011) respond with a rapid (within 24 h) increase in haematocrit, Hb, and RBC count. These effects are accompanied by an equally rapid stimulation of late-stage erythroid precursors in the bone marrow (BM). RAP-011 also induces a significant increase in erythroid burst-forming units and erythropoietin, which could contribute to additional, sustained effects on RBC production. Further in vitro co-culture studies demonstrate that BM accessory cells are required for RAP-011 effects. To better understand which TGF-ß family ligand(s) mediate RAP-011 effects, we evaluated the impact of several of these ligands on erythroid differentiation. Our data suggest that RAP-011 may act to rescue growth differentiation factor 11/Activin A-induced inhibition of late-stage erythropoiesis. These data define the mechanism of action of a novel agent that regulates RBC differentiation and provide the rationale to develop sotatercept for the treatment of anaemia and ineffective erythropoiesis.


Subject(s)
Activin Receptors, Type II/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythropoiesis/drug effects , Erythropoiesis/physiology , Hemoglobins/biosynthesis , Recombinant Fusion Proteins/pharmacology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cellular Microenvironment/physiology , Colony-Forming Units Assay , Erythrocyte Indices/drug effects , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoietin/biosynthesis , Female , Humans , Ligands , Mice , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism
5.
Br J Haematol ; 160(4): 487-502, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23252516

ABSTRACT

Durable responses with lenalidomide monotherapy have been reported in patients with non-Hodgkin lymphoma. In relapsed/refractory diffuse large B-cell lymphoma (DLBCL), higher responses were observed in the activated B-cell-like (ABC) subtype than in the germinal centre B-cell-like subtype. Herein, the molecular mechanisms involved in the differential efficacy of lenalidomide in DLBCL subtypes were investigated. Using DLBCL cell lines, lenalidomide treatment was found to preferentially suppress proliferation of ABC-DLBCL cells in vitro and delay tumour growth in a human tumour xenograft model, with minimal effect on non-ABC-DLBCL cells. This tumouricidal effect was associated with downregulation of interferon regulatory factor 4 (IRF4), a hallmark of ABC-DLBCL cells. IRF4 inhibition by lenalidomide induced downregulation of B-cell receptor (BCR)-dependent NF-κB. Whereas IRF4-specific small, interfering RNA mimicked the effects of lenalidomide reducing NF-κB activation, IRF4 overexpression enhanced NF-κB activation and conferred resistance to lenalidomide. These findings indicate the crucial role of IRF4 inhibition in lenalidomide efficacy in ABC cells. Furthermore, lenalidomide-induced IRF4 downregulation required the expression of cereblon, a molecular target of lenalidomide. Taken together, these findings suggest that lenalidomide has direct antitumour activity against DLBCL cells, preferentially ABC-DLBCL cells, by blocking IRF4 expression and the BCR-NF-κB signalling pathway in a cereblon-dependent manner.


Subject(s)
Antineoplastic Agents/pharmacology , Interferon Regulatory Factors/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Peptide Hydrolases/metabolism , Thalidomide/analogs & derivatives , Adaptor Proteins, Signal Transducing/drug effects , Animals , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/drug effects , CARD Signaling Adaptor Proteins/genetics , Caspases/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Drug Resistance, Neoplasm/genetics , Female , Guanylate Cyclase/drug effects , Guanylate Cyclase/genetics , Humans , Interleukin-1 Receptor-Associated Kinases/drug effects , Lenalidomide , Lymphoma, Large B-Cell, Diffuse/metabolism , Mice , Mice, SCID , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Mutation , Myeloid Differentiation Factor 88/drug effects , NF-kappa B/drug effects , Neoplasm Proteins/drug effects , Neoplasm Proteins/metabolism , Neoplasm Transplantation/methods , Thalidomide/pharmacology , Transplantation, Heterologous , Ubiquitin-Protein Ligases
6.
J Exp Ther Oncol ; 10(3): 219-33, 2013.
Article in English | MEDLINE | ID: mdl-24416998

ABSTRACT

Inflammatory breast cancer (IBC) is the most metastatic variant of locally advanced breast cancer. IBC has distinctive characteristics including invasion of tumor emboli into the skin and rapid disease progression. Given our previous studies suggesting that HDAC inhibitors have promise in targeting IBC, the present study revealed that the class I HDAC inhibitor Romidepsin (FK-288, Istodax; Celgene Corporation, Summit, NJ) potently induced destruction of IBC tumor emboli and lymphatic vascular architecture. associated with inhibition of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor 1alpha, (HIF1alpha) proteins in the Mary-X pre-clinical model of IBC. Romidepsin treatment induced clinically relevant biomarkers in including induction of acetylated Histone 3 (Ac-H3) proteins, apoptosis, and increased p21WAF1/CIP1. Romidepsin, alone and synergistically when combined with Paclitaxel, effectively eliminated both primary tumors and metastatic lesions at multiple sites formed by the SUM149 IBC cell line. This is the first report of the ability of an HDAC inhibitor to eradicate IBC tumor emboli, to destroy the integrity of lymphatic vessel architecture and to target metastasis. Furthermore, Romidepsin, in combination with a taxane, warrants evaluation as a therapeutic strategy that may effectively target the skin involvement and rapid metastasis that are hallmarks of IBC.


Subject(s)
Depsipeptides/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Inflammatory Breast Neoplasms/drug therapy , Neoplastic Cells, Circulating/drug effects , Paclitaxel/administration & dosage , Animals , Apoptosis/drug effects , Cell Line, Tumor , Depsipeptides/administration & dosage , Drug Synergism , Female , Humans , Inflammatory Breast Neoplasms/pathology , Mice , Neoplasm Metastasis/prevention & control , Spheroids, Cellular , Xenograft Model Antitumor Assays
7.
Br J Haematol ; 159(2): 154-63, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22881386

ABSTRACT

We present data from a phase II study investigating a novel treatment strategy for relapsed/refractory mantle cell lymphoma (MCL). Twenty-six patients received lenalidomide 25 mg/d (days 1-21 of a 28-d cycle) for up to 6 cycles followed by low-dose maintenance lenalidomide (15 mg) in responding patients. Eight patients achieved complete or partial response to give an overall response rate of 31% with median response duration of 22·2 months [95% confidence interval (CI) 0·0-53·6] and median progression-free survival (PFS) of 3·9 months (95% CI 0·0-11·1). An additional six patients (23%) achieved stable disease. Eleven patients received maintenance with median PFS of 14·6 months (95% CI 7·3-21·9). Correlative studies showed that peripheral T and Natural Killer (NK) cells increased in responding patients by 40-60% over the first 6 cycles with an initial dip in NK cells suggestive of tumour infiltration. Peripheral regulatory T cells were increased in MCL patients (P = 0·001) and expanded further following lenalidomide. Sequential plasma analysis showed increased IL12 p40 and IL7 alongside decreased MMP9, IL10, and adiponectin. Finally, a significant correlation (P = 0·02) between gender and response suggested that female MCL patients were more sensitive to lenalidomide than males. In summary, we confirm the activity, safety and immunomodulatory properties of lenalidomide in MCL and highlight its potential as a low-dose maintenance agent.


Subject(s)
Antineoplastic Agents/administration & dosage , Lymphoma, Mantle-Cell , Sex Characteristics , Thalidomide/analogs & derivatives , Adiponectin/blood , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Interleukin-10/blood , Interleukin-12 Subunit p40/blood , Lenalidomide , Leukocyte Count , Lymphoma, Mantle-Cell/blood , Lymphoma, Mantle-Cell/mortality , Lymphoma, Mantle-Cell/prevention & control , Male , Middle Aged , Recurrence , Sex Factors , Survival Rate , Thalidomide/administration & dosage , United Kingdom/epidemiology
8.
Cancer Chemother Pharmacol ; 69(4): 965-76, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22120960

ABSTRACT

PURPOSE: Multi-drug resistance and cumulative cardiotoxicity are major limitations for the clinical use of anthracyclines. Here, we evaluated and compared the cross-resistance of amrubicin, a third-generation synthetic anthracycline and potent topoisomerase (topo)-II inhibitor with little or no observed cardiotoxicity to other anthracyclines and the topo-II inhibitor etoposide in drug-resistant tumor models in order to elucidate its potential mechanisms of action. METHODS: Amrubicin activity was assessed in multi-drug-resistant cell lines and human tumor explants using cytotoxicity assays, confocal microscopy, fluorescence time-lapse imaging, flow cytometry, immunoblotting, and gene expression profiling techniques. RESULTS: We demonstrate that both doxorubicin-resistant tumor cell lines and several drug-resistant human ovarian and breast tumor explants retain sensitivity to amrubicin. In addition, we observed similar levels of amrubicin uptake and accumulation in doxorubicin-sensitive versus doxorubicin-resistant cell lines. Although amrubicin is a weak P-glycoprotein substrate, transport and retention of amrubicin were not solely modulated by P-glycoprotein in the resistant cell lines overexpressing drug efflux pumps. The cellular retention of amrubicin is likely to be a result of rapid influx due to its high intrinsic permeability and lipophilic properties, and this may explain why amrubicin overcomes pleiotropic drug resistance. Consistent with drug accumulation studies, amrubicin induced DNA damage, G(2)-M cell cycle arrest, and apoptosis in both doxorubicin-sensitive and doxorubicin-resistant lines. Using gene expression profiling studies, several classes of genes were significantly and uniquely regulated following amrubicin, but not doxorubicin or etoposide, treatment. CONCLUSIONS: Amrubicin appears to have a distinct mode of action that overcomes typical anthracycline resistance mechanisms. Therefore, amrubicin may be useful in the treatment of anthracycline-refractory or anthracycline-resistant tumors.


Subject(s)
Anthracyclines/pharmacology , Anthracyclines/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Caspase 3/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , DNA Damage , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Epirubicin/pharmacology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Topoisomerase II Inhibitors/pharmacokinetics , Topoisomerase II Inhibitors/pharmacology , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
9.
Cancer Sci ; 102(6): 1201-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21375679

ABSTRACT

Histone deacetylase inhibitors are a group of recently developed compounds that modulate cell growth and survival. We evaluated the effects of the histone deacetylase inhibitor MGCD0103 on growth of pancreatic carcinoma models following single agent treatment and in combination with gemcitabine. MGCD0103 inhibited tumor cell growth and acted synergistically with gemcitabine to enhance its cytotoxic effects. Gene expression analysis identified the cell cycle pathway as one of the most highly modulated gene groups. Our data suggest that MGCD0103 + gemcitabine might be an effective treatment for gemcitabine-refractory pancreatic cancer.


Subject(s)
Benzamides/pharmacology , Deoxycytidine/analogs & derivatives , Histone Deacetylase Inhibitors/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pyrimidines/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Drug Synergism , Gene Expression , Humans , Immunoblotting , Pancreas/enzymology , Gemcitabine
10.
Expert Rev Anticancer Ther ; 10(10): 1663-72, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20942636

ABSTRACT

Lenalidomide has demonstrated clinical activity in myelodysplastic syndromes, particularly in patients with a deletion in the long arm of chromosome 5 (del[5q] abnormality). It has a direct effect on the del(5q) clone, which may contribute to its ability to induce cytogenetic responses. Lenalidomide also stimulates erythropoiesis, leading to erythroid responses in certain patients. Other effects include immunomodulation, anti-inflammatory effects and angiogenesis inhibition. New findings indicate that lenalidomide modulates key genes located within the del(5q) region, including tumor suppressor genes, and regulators of the actin cytoskeleton and cell membrane. Its effects on cytoskeleton regulation may explain its direct antitumor and immunomodulatory effects. This article provides an overview of the known effects of lenalidomide and new insights into its activity in del(5q) myelodysplastic syndromes.


Subject(s)
Antineoplastic Agents/pharmacology , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Thalidomide/analogs & derivatives , Antineoplastic Agents/therapeutic use , Erythropoiesis/drug effects , Erythropoiesis/genetics , Genes/drug effects , Humans , Immunologic Factors/genetics , Immunologic Factors/pharmacology , Lenalidomide , Myelodysplastic Syndromes/pathology , Thalidomide/pharmacology , Thalidomide/therapeutic use , Treatment Outcome
11.
Br J Haematol ; 151(4): 387-96, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20880107

ABSTRACT

Inhibition of histone deacetylase 6 (HDAC6)-dependent aggresome function by pan HDAC inhibitors was recently reported to be a key mechanism underlying the synergistic activity between proteasome inhibitors and HDAC inhibitors in a variety of tumour types. Because these combinations induce significant thrombocytopenia in vivo, we examined whether less toxic, isotype-selective HDAC inhibitors may still synergize with proteasome inhibitors, and if so, by what mechanisms. Here, we showed that the class I HDAC inhibitor, MGCD0103, has a potent antiproliferative activity in Hodgkin lymphoma (HL) cell lines. Furthermore, MGCD0103 induced tumour necrosis factor α (TNF-α) expression and secretion, which was associated with nuclear factor (NF)-κB activation. Selective inhibition of TNF-α expression by short interfering mRNA, or inhibition of MGCD0103-induced NF-kB activation by proteasome inhibitors enhanced MGCD0103-induced cell death. Thus, our results demonstrate that MGCD0103 may synergize with proteasome inhibitors by HDAC6-independent mechanisms, providing mechanistic rationale for exploring this potentially less toxic combination for the treatment of lymphoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hodgkin Disease/pathology , Pyrimidines/pharmacology , Boronic Acids/pharmacology , Bortezomib , Cytokines/biosynthesis , Cytokines/genetics , Drug Screening Assays, Antitumor/methods , Drug Synergism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase 6 , Histone Deacetylases/physiology , Humans , NF-kappa B/metabolism , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/drug effects
12.
PLoS One ; 5(2): e9001, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20126405

ABSTRACT

BACKGROUND: The cytidine nucleoside analogs azacitidine (AZA) and decitabine (DAC) are used for the treatment of patients with myelodysplastic syndromes and acute myeloid leukemia (AML). Few non-clinical studies have directly compared the mechanisms of action of these agents in a head-to-head fashion, and the agents are often viewed as mechanistically similar DNA hypomethylating agents. To better understand the similarities and differences in mechanisms of these drugs, we compared their in vitro effects on several end points in human AML cell lines. METHODOLOGY/PRINCIPAL FINDINGS: Both drugs effected DNA methyltransferase 1 depletion, DNA hypomethylation, and DNA damage induction, with DAC showing equivalent activity at concentrations 2- to 10-fold lower than AZA. At concentrations above 1 microM, AZA had a greater effect than DAC on reducing cell viability. Both drugs increased the sub-G1 fraction and apoptosis markers, with AZA decreasing all cell cycle phases and DAC causing an increase in G2-M. Total protein synthesis was reduced only by AZA, and drug-modulated gene expression profiles were largely non-overlapping. CONCLUSIONS/SIGNIFICANCE: These data demonstrate shared mechanisms of action of AZA and DAC on DNA-mediated markers of activity, but distinctly different effects in their actions on cell viability, protein synthesis, cell cycle, and gene expression. The differential effects of AZA may be mediated by RNA incorporation, as the distribution of AZA in nucleic acid of KG-1a cells was 65:35, RNA:DNA.


Subject(s)
Apoptosis/drug effects , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Cycle/drug effects , Protein Biosynthesis/drug effects , Acute Disease , Antimetabolites, Antineoplastic/pharmacology , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage , DNA Methylation/drug effects , Decitabine , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression/drug effects , HL-60 Cells , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Oligonucleotide Array Sequence Analysis/methods , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism
13.
Lung Cancer (Auckl) ; 1: 119-140, 2010.
Article in English | MEDLINE | ID: mdl-28210112

ABSTRACT

Azacitidine (AZA) and decitabine (DAC) are cytidine azanucleoside analogs with clinical activity in myelodysplastic syndromes (MDS) and potential activity in solid tumors. To better understand the mechanism of action of these drugs, we examined the effects of AZA and DAC in a panel of non-small cell lung cancer (NSCLC) cell lines. Of 5 NSCLC lines tested in a cell viability assay, all were sensitive to AZA (EC50 of 1.8-10.5 µM), while only H1299 cells were equally sensitive to DAC (EC50 of 5.1 µM). In the relatively DAC-insensitive cell line A549, both AZA and DAC caused DNA methyltransferase I depletion and DNA hypomethylation; however, only AZA significantly induced markers of DNA damage and apoptosis, suggesting that mechanisms in addition to, or other than, DNA hypomethylation are important for AZA-induced cell death. Cell cycle analysis indicated that AZA induced an accumulation of cells in sub-G1 phase, whereas DAC mainly caused an increase of cells in G2/M. Gene expression analysis of AZA- and DAC-treated cells revealed strikingly different profiles, with many genes distinctly regulated by each drug. In summary, while both AZA and DAC caused DNA hypomethylation, distinct effects were demonstrated on regulation of gene expression, cell cycle, DNA damage, and apoptosis.

14.
J Med Chem ; 52(2): 278-92, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-19113866

ABSTRACT

The inhibition of key receptor tyrosine kinases (RTKs) that are implicated in tumor vasculature formation and maintenance, as well as tumor progression and metastasis, has been a major focus in oncology research over the last several years. Many potent small molecule inhibitors of vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR) kinases have been evaluated. More recently, compounds that act through the complex inhibition of multiple kinase targets have been reported and may exhibit improved clinical efficacy. We report herein a series of potent, orally efficacious 4-amino-3-benzimidazol-2-ylhydroquinolin-2-one analogues as inhibitors of VEGF, PDGF, and fibroblast growth factor (FGF) receptor tyrosine kinases. Compounds in this class, such as 5 (TKI258), are reversible ATP-competitive inhibitors of VEGFR-2, FGFR-1, and PDGFRbeta with IC(50) values <0.1 microM. On the basis of its favorable in vitro and in vivo properties, compound 5 was selected for clinical evaluation and is currently in phase I clinical trials.


Subject(s)
Drug Design , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Quinolones/chemistry , Quinolones/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Mice, Inbred NOD , Mice, SCID , Models, Molecular , Protein Kinase Inhibitors/pharmacokinetics , Quinolones/pharmacokinetics , Structure-Activity Relationship
15.
Clin Cancer Res ; 14(7): 2075-81, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381947

ABSTRACT

PURPOSE: To determine the maximum tolerated dose (MTD) dose-limiting toxicity, and pharmacokinetic and pharmacodynamic profile of TKI258 (formerly CHIR-258). EXPERIMENTAL DESIGN: A phase I dose escalating trial in patients with advanced solid tumors was performed. Treatment was initially as single daily doses on an intermittent 7-day on/7-day off schedule. Following a protocol amendment, a second schedule comprised, during cycle 1, 7-day on/7-day off treatment followed by 14 days of continuous daily dosing; subsequent cycles comprised 28 days of daily dosing. Pharmacokinetics and evaluation of phosphorylated extracellular signal-regulated kinase (ERK) in peripheral blood mononuclear cells were done during the first 28 days of each schedule. RESULTS: Thirty-five patients were treated in four intermittent (25-100 mg/d) and three continuous (100-175 mg/d) dosing cohorts. Observed drug-related toxicities were nausea and vomiting, fatigue, headache, anorexia, and diarrhea. Dose-limiting toxicities were grade 3 hypertension in one patient at 100 mg continuous dosing, grade 3 anorexia in a second patient at 175 mg, and grade 3 alkaline phosphatase elevation in a third patient at 175 mg. One patient had a partial response (melanoma) and two patients had stable disease >6 months. TKI258 pharmacokinetics were linear over the dose range of 25 to 175 mg. Five of 14 evaluable patients had modulation of phosphorylated ERK levels. CONCLUSIONS: The MTD was defined as 125 mg/d. Evidence of antitumor activity in melanoma and gastrointestinal stromal tumors warrants further investigation, and other phase I studies are ongoing. Further pharmacodynamic evaluation is required in these studies to evaluate the biological effects of TKI258.


Subject(s)
Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Neoplasms/drug therapy , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Quinolones/adverse effects , Quinolones/pharmacokinetics , Administration, Oral , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Benzimidazoles/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/drug effects , Quinolones/administration & dosage , Treatment Outcome
16.
J Immunother ; 30(1): 64-74, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17198084

ABSTRACT

Recombinant interleukin-2 (rIL-2) is a pleiotropic cytokine that activates select immune effector cell responses associated with antitumor activity, including antibody-dependent cellular cytotoxicity (ADCC). Rituximab is an anti-CD20 monoclonal antibody that activates ADCC in non-Hodgkin lymphoma (NHL). The ability of rIL-2 to augment rituximab-dependent tumor responses was investigated. The efficacy of rIL-2 in combination with rituximab was evaluated in 2 NHL tumor xenograft models: the CD20hi, rituximab-sensitive, low-grade Daudi model and the CD20lo, aggressive, rituximab-resistant Namalwa model. Combination of rIL-2 plus rituximab was synergistic in a rituximab-sensitive Daudi tumor model, as evidenced by significant tumor regressions and increased time to tumor progression, compared with rIL-2 and rituximab single agents. In contrast, rituximab-resistant Namalwa tumors were responsive to single-agent rIL-2 and showed an increased response when combined with rituximab. Using in vitro killing assays, rIL-2 was shown to enhance activity of rituximab by activating ADCC and lymphokine-activated killer activity. Additionally, the activity of rIL-2 plus rituximab F(ab')2 was similar to that of rIL-2 alone, indicating a critical role for immunoglobulin G1 Fc-FcgammaR-effector responses in mediating ADCC. Antiproliferative and apoptotic tumor responses, along with an influx of immune effector cells, were observed by immunohistochemistry. Collectively, the data suggest that rIL-2 mediates potent tumoricidal activity against NHL tumors, in part, through activation and trafficking of monocytes and natural killer cells to tumors. These data support the mechanistic and therapeutic rationale for combination of rIL-2 with rituximab in NHL clinical trials and for single-agent rIL-2 in rituximab-resistant NHL patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Interleukin-2/pharmacology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD20/immunology , Drug Synergism , Female , Humans , Immunoglobulin Fc Fragments/immunology , Interleukin-2/administration & dosage , Interleukin-2/immunology , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Monocytes/immunology , Rituximab , Xenograft Model Antitumor Assays
17.
Clin Cancer Res ; 12(16): 4908-15, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16914579

ABSTRACT

PURPOSE: The ectopically expressed and deregulated fibroblast growth factor receptor 3 (FGFR3) results from a t(4;14) chromosomal translocation that occurs in approximately 15% of multiple myeloma (MM) patients and confers a particularly poor prognosis. This study assesses the antimyeloma activity of CHIR-258, a small-molecule inhibitor of multiple receptor tyrosine kinases that is currently in phase I trials, in a newly developed FGFR3-driven preclinical MM animal model. EXPERIMENTAL DESIGN: We developed an orthotopic MM model in mice using a luciferase-expressing human KMS-11-luc line that expresses mutant FGFR3 (Y373C). The antimyeloma activity of CHIR-258 was evaluated at doses that inhibited FGFR3 signaling in vivo in this FGFR3-driven animal model. RESULTS: Noninvasive bioluminescence imaging detected MM lesions in nearly all mice injected with KMS-11-luc cells, which were mainly localized in the spine, skull, and pelvis, resulting in frequent development of paralysis. Daily oral administration of CHIR-258 at doses that inhibited FGFR3 signaling in KMS-11-luc tumors in vivo resulted in a significant inhibition of KMS-11-luc tumor growth, which translated into a significant improvement in animal survival. CONCLUSIONS: Our data provide a relevant preclinical basis for clinical trials of CHIR-258 in FGFR3-positive MM patients.


Subject(s)
Benzimidazoles/pharmacology , Multiple Myeloma/drug therapy , Quinolones/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/biosynthesis , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, SCID , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Multiple Myeloma/enzymology , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/blood , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Xenograft Model Antitumor Assays
18.
Blood ; 108(10): 3465-71, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16849642

ABSTRACT

Overexpression of fibroblast growth factor receptor 3 (FGFR3) is a hallmark of t(4;14) multiple myeloma (MM). To dissect the mechanism of FGFR3 oncogenesis in MM, we used 3 FGFR selective kinase inhibitors-CHIR258, PD173074, and SU5402-and FGFR3-specific siRNA to modulate FGFR3 activity. Conversely, the ligand FGF was used to stimulate FGFR3 function in human MM cells. The transcriptional response to FGFR3 modification was recorded, and gene expression changes common to all 5 modifiers were documented. Ten genes were commonly regulated. Macrophage inflammatory protein-1 alpha (MIP-1alpha) was the single most differentially altered gene. MIP-1 alpha promoter function, gene expression, and protein secretion were each down-regulated following inhibition of FGFR3 signaling. Down-regulation of MIP-1 alpha was not, however, observed following FGFR3 inhibition in MM cells with RAS mutations implicating RAS-MAPK in MIP-1 alpha regulation. As confirmation, inhibition of ERK1 also down-regulated MIP-1 alpha in FGFR3 inhibitor-resistant cells harboring RAS mutations. MIP-1 alpha is implicated in the survival and proliferation of MM cells and the pathogenesis of MM bone disease. Our observation is the first to directly link an initiating IgH translocation not only to MM-cell growth and survival but also to the disease-associated bone disease.


Subject(s)
MAP Kinase Signaling System/physiology , Macrophage Inflammatory Proteins/metabolism , Multiple Myeloma/etiology , Receptor, Fibroblast Growth Factor, Type 3/genetics , ras Proteins/metabolism , Bone Diseases/etiology , Chemokine CCL3 , Chemokine CCL4 , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Macrophage Inflammatory Proteins/genetics , Multiple Myeloma/complications , Multiple Myeloma/genetics , Mutation , Neoplasm Proteins , RNA, Small Interfering/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/physiology , Tumor Cells, Cultured , ras Proteins/genetics
20.
Clin Cancer Res ; 11(14): 5281-91, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16033847

ABSTRACT

PURPOSE: Fms-like tyrosine kinase 3 (FLT3) encodes a receptor tyrosine kinase (RTK) for which activating mutations have been identified in a proportion of acute myelogenous leukemia (AML) patients and associated with poor clinical prognosis. Given the relevance of FLT3 mutations in AML, we investigated the activity of CHIR-258, an orally active, multitargeted small molecule, with potent activity against FLT3 kinase and class III, IV, and V RTKs involved in endothelial and tumor cell proliferation in AML models. EXPERIMENTAL DESIGN: CHIR-258 was tested on two human leukemic cell lines in vitro and in vivo with differing FLT3 mutational status [MV4;11 cells express FLT3 internal tandem duplications (ITD) versus RS4;11 cells with wild-type (WT) FLT3]. RESULTS: Antiproliferative activity of CHIR-258 against MV4;11 was approximately 24-fold greater compared with RS4;11, indicating more potent inhibition against cells with constitutively activated FLT3 ITD. Dose-dependent down modulation of receptor phosphorylation and downstream signaling [signal transducer and activator of transcription 5 (STAT5) and extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase] in MV4;11 cells with CHIR-258 confirmed the molecular mechanism of action. Target modulation of phospho-FLT3, phospho-STAT5, and phospho-ERK in MV4;11 tumors was achieved at biologically active doses of CHIR-258. Tumor regressions and eradication of AML cells from the bone marrow were shown in s.c. and bone marrow engraftment leukemic xenograft models. Tumor responses were characterized by decreased cellular proliferation and positive immunohistochemical staining for active caspase-3 and cleaved poly(ADP-ribose) polymerase, suggesting cell death was mediated in part via apoptosis. CONCLUSIONS: Our data indicate that CHIR-258 may be an effective therapy in FLT3-associated AML and warrants clinical trials.


Subject(s)
Benzimidazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins/genetics , Quinolones/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Animals , Cell Proliferation , DNA Mutational Analysis , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/veterinary , Mice , Mice, SCID , Neoplasm Transplantation , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Tandem Repeat Sequences , Transplantation, Heterologous , Tumor Cells, Cultured , fms-Like Tyrosine Kinase 3
SELECTION OF CITATIONS
SEARCH DETAIL
...