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










Publication year range
1.
Leuk Lymphoma ; 61(7): 1653-1659, 2020 07.
Article in English | MEDLINE | ID: mdl-32154751

ABSTRACT

Buparlisib is an orally available pan-Class I PI3K inhibitor, that is more potent than idelalisib in vitro. Its distinct toxicities include hyperglycemia, hypertension, and mood disturbance. IND216 is a single arm phase II trial of buparlisib in Relapsed/refractory (R/R) chronic lymphocytic leukemia (CLL). Fourteen patients were enrolled, 13 were evaluable for response and toxicity. Six of 13 patients had a partial response (46%) with a median duration of response of 15.5 months, all 11 patients with tumor assessment experienced tumor shrinkage. The most common adverse events (≥15%) were hyperglycemia, fatigue, anxiety, and gastrointestinal toxicities; all were < grade 3 except for fatigue. Three patients stopped therapy for alterations in mood. Lower levels of raptor were significantly associated with greater tumor shrinkage, suggesting that raptor could be a biomarker for response. This requires further validation in a larger CLL patient cohort. The clinical activity of buparlisib is comparable to other phosphatidylinositol-3-kinase inhibitors, with a different toxicity profile.Novelty and impactBuparlisib, an oral, pan PI3 kinase inhibitor, is associated with a 46% partial response rate among patients with relapse chronic lymphocytic leukemia (CLL). This is a similar clinical activity to other phosphatidylinositol-3-kinase inhibitors tested. However, buparlisib has a distinct toxicity profile, characterized by hyperglycemia, hypertension, and mood alteration. In agreement with our previous preclinical study, our results suggest that basal raptor expression in CLL correlates with clinical response to buparlisib.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Aminopyridines , Biomarkers , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Morpholines , Phosphatidylinositol 3-Kinases , Phosphoinositide-3 Kinase Inhibitors , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases, 70-kDa
2.
Breast Cancer Res Treat ; 172(1): 23-32, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30039287

ABSTRACT

PURPOSE: PARP-3 is member of the PARP family of poly (ADP-ribose) polymerases involved in ADPribosylation. PARPs are involved in the basic mechanisms of DNA repair. PARP3, a critical player for efficient mitotic progression, is required for the stabilization of the mitotic spindle by regulation of the mitotic components, NuMA and Tankyrase 1. METHODS: The sensitization effect of vinorelbine on PARP3 inhibition-induced cytotoxicity was assessed by the SRB assay. The contribution of programed cell death and cell cycle arrest to the sensitization effect were determined by assessing changes in Annexin V, a marker of apoptosis. Alterations in cell cycle progression were assessed by cell cycle analysis. We used immunofluorescence to assess the effect of vinorelbine and/or PARP3 inhibitors on tubulin and microtubule depolarization. The PARP3 chemiluminescent assay kit was used for PARP3 activity. RESULTS: PARP3 inhibitors sensitize breast cancer cells to vinorelbine, a vinca alkaloid used in the treatment of metastatic breast cancer. Olaparib which was originally described as a PARP1 and 2 inhibitor has recently been shown to be a potent PARP3 inhibitor while ME0328 is a more selective PARP3 inhibitor. The combination of vinorelbine with nontoxic concentrations of ME0328 or olaparib reduces vinorelbine resistance by 10 and 17 fold, respectively, potentiating vinorelbine-induced arrest at the G2/M boundary. In addition, PARP3 inhibition potentiates vinorelbine interaction with tubulin. Furthermore, olaparib or ME0328 potentiates vinorelbine-induced PARP3 inhibition, mitotic arrest, and apoptosis. CONCLUSION: Our results indicated this approach with PARP3 inhibitors and vinorelbine is unique and promising for breast cancer patients with metastases. This combination could significantly increase the survival of breast cancer patients with metastases.


Subject(s)
Breast Neoplasms/drug therapy , Cell Cycle Proteins/genetics , Drug Resistance, Neoplasm/drug effects , Poly(ADP-ribose) Polymerases/genetics , Vinorelbine/pharmacology , Antigens, Nuclear/genetics , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/antagonists & inhibitors , DNA Repair/drug effects , Female , Humans , MCF-7 Cells , Nuclear Matrix-Associated Proteins/genetics , Phthalazines/pharmacology , Piperazines/pharmacology , Quinazolinones/pharmacology , Spindle Apparatus/genetics , Tankyrases/genetics
3.
Chem Sci ; 8(9): 6218-6229, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28989655

ABSTRACT

We report a spherical nucleic acid (SNA) system for the delivery of BKM120, an anticancer drug for treatment of chronic lymphocytic leukemia (CLL). While promising for cancer treatment, this drug crosses the blood-brain barrier causing significant side-effects in patients. The DNA nanoparticle encapsulates BKM120 in high efficiency, and is unparalleled in its monodispersity, ease of synthesis and stability in different biological media and in serum. These DNA nanostructures demonstrate efficient uptake in human cervical cancer (HeLa) cells, and increased internalization of cargo. In vitro studies show that BKM120-loaded nanoparticles promote apoptosis in primary patient CLL lymphocytes, and act as sensitizers of other antitumor drugs, without causing non-specific inflammation. Evaluation of this drug delivery system in vivo shows long circulation times up to 24 hours, full body distribution, accumulation at tumor sites and minimal leakage through the blood-brain barrier. Our results demonstrate the great potential of these delivery vehicles as a general platform for chemotherapeutic drug delivery.

4.
Front Pharmacol ; 6: 147, 2015.
Article in English | MEDLINE | ID: mdl-26257651

ABSTRACT

Enhanced DNA damage repair is one mechanism involved in colon cancer drug resistance. Thus, targeting molecular components of repair pathways with specific small molecule inhibitors may improve the efficacy of chemotherapy. ABT-888 and VE-821, inhibitors of poly-ADP-ribose-polymerase (PARP) and the serine/threonine-kinase Ataxia telangiectasia related (ATR), respectively, were used to treat colon cancer cell lines in combination with the topoisomerase-I inhibitor irinotecan (SN38). Our findings show that each of these DNA repair inhibitors utilized alone at nontoxic single agent concentrations resulted in sensitization to SN38 producing a 1.4-3 fold reduction in the 50% inhibitory concentration (IC50) of SN38 in three colon cancer cell lines. When combined together, nontoxic concentrations of ABT-888 and VE-821 produced a 4.5-27 fold reduction in the IC50 of SN38 with the HCT-116 colon cancer cells demonstrating the highest sensitization as compared to LoVo and HT-29 colon cancer cells. Furthermore, the combination of all three agents was associated with maximal G2 -M arrest and enhanced DNA-damage (γH2AX) in all three colon cancer cell lines. The mechanism of this enhanced sensitization was associated with: (a) maximal suppression of SN38 induced PARP activity in the presence of both inhibitors and (b) ABT-888 producing partial abrogation of the VE-821 enhancement of SN38 induced DNA-PK phosphorylation, resulting in more unrepaired DNA damage; these alterations were only present in the HCT-116 cells which have reduced levels of ATM. This novel combination of DNA repair inhibitors may be useful to enhance the activity of DNA damaging chemotherapies such as irinotecan and help produce sensitization to this drug in colon cancer.

5.
Front Pharmacol ; 4: 5, 2013.
Article in English | MEDLINE | ID: mdl-23386830

ABSTRACT

Many current chemotherapies function by damaging genomic DNA in rapidly dividing cells ultimately leading to cell death. This therapeutic approach differentially targets cancer cells that generally display rapid cell division compared to normal tissue cells. However, although these treatments are initially effective in arresting tumor growth and reducing tumor burden, resistance and disease progression eventually occur. A major mechanism underlying this resistance is increased levels of cellular DNA repair. Most cells have complex mechanisms in place to repair DNA damage that occurs due to environmental exposures or normal metabolic processes. These systems, initially overwhelmed when faced with chemotherapy induced DNA damage, become more efficient under constant selective pressure and as a result chemotherapies become less effective. Thus, inhibiting DNA repair pathways using target specific small molecule inhibitors may overcome cellular resistance to DNA damaging chemotherapies. Non-homologous end joining a major mechanism for the repair of double-strand breaks (DSB) in DNA is regulated in part by the serine/threonine kinase, DNA dependent protein kinase (DNA-PK). The DNA-PK holoenzyme acts as a scaffold protein tethering broken DNA ends and recruiting other repair molecules. It also has enzymatic activity that may be involved in DNA damage signaling. Because of its' central role in repair of DSBs, DNA-PK has been the focus of a number of small molecule studies. In these studies specific DNA-PK inhibitors have shown efficacy in synergizing chemotherapies in vitro. However, compounds currently known to specifically inhibit DNA-PK are limited by poor pharmacokinetics: these compounds have poor solubility and have high metabolic lability in vivo leading to short serum half-lives. Future improvement in DNA-PK inhibition will likely be achieved by designing new molecules based on the recently reported crystallographic structure of DNA-PK. Computer based drug design will not only assist in identifying novel functional moieties to replace the metabolically labile morpholino group but will also facilitate the design of molecules to target the DNA-PKcs/Ku80 interface or one of the autophosphorylation sites.

6.
Genes Chromosomes Cancer ; 52(5): 480-94, 2013 May.
Article in English | MEDLINE | ID: mdl-23341105

ABSTRACT

PALB2/FANCN is a BRCA1- and BRCA2-interacting Fanconi Anemia (FA) protein crucial for key BRCA2 genome caretaker functions. Heterozygous germline mutations in PALB2 predispose to breast cancer and biallelic mutations cause FA. FA proteins play a critical role in the telomere maintenance pathway, with telomeric shortening observed in FA cells. Less is known about telomere maintenance in the heterozygous state. Here, we investigate the roles of PALB2 heterozygous mutations in genomic instability, an important carcinogenesis precursor. Patient-derived lymphoblastoid (LCL) and fibroblast (FCL) cell lines with monoallelic truncating PALB2 mutations were investigated using a combination of molecular imaging techniques including centromeric FISH, telomeric Q-FISH and spectral karyotyping (SKY). Mitomycin C and Cisplatin sensitivity was assayed via cellular metabolism of WST-1. The PALB2 c.229delT FCL showed increases in telomere counts associated with increased mean intensity compared with two wild-type FCLs generated from first-degree relatives (P =1.04E-10 and P =9.68E-15) and it showed evidence of chromosomal rearrangements. Significant differences in centromere distribution were observed in one of three PALB2 heterozygous FCLs analyzed when compared with PALB2 wild-type, BRCA1 and BRCA2 heterozygous FCLs. No significant consistently increased sensitivity to Mitomycin C or Cisplatin was observed in LCLs. Our results are suggestive of an altered centromere distribution profile and a telomere instability phenotype. Together, these may indicate critical nuclear organization defects associated with the predisposition to transformation and early stage development of PALB2-related cancers.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Cell Nucleus/metabolism , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Case-Control Studies , Cell Survival/drug effects , Centromere/metabolism , Cisplatin/pharmacology , Fanconi Anemia Complementation Group N Protein , Female , Genetic Association Studies , Genetic Predisposition to Disease , Hereditary Breast and Ovarian Cancer Syndrome/pathology , Heterozygote , Humans , Karyotype , Male , Middle Aged , Mitomycin/pharmacology , Telomere/metabolism , Tumor Cells, Cultured
7.
Int J Cancer ; 133(1): 247-52, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23238639

ABSTRACT

BKM120, a pan class I PI3K inhibitor, was cytotoxic in the majority of primary B-chronic lymphocytic leukemia (CLL) lymphocytes, including samples from patients who have a high-risk for poor response to treatment (patient with del11 and del17) at clinically obtainable concentrations. The PI3Kδ inhibitor Cal-101 is cytotoxic in B-CLL lymphocytes in vitro and is active in the treatment of CLL in vivo. Interestingly, we demonstrated that BKM120 is 3.6 fold more toxic than Cal-101 in malignant B-CLL lymphocytes in vitro. BKM120 cytotoxicity correlated with the basal expression of proteins involved in the PI3K/Akt pathway. A protein signature of PI3K pathway proteins predicts the response to BKM120 treatment. In the primary B-CLL lymphocytes tested in vitro, BKM120 decreased the phosphorylation status of molecular biomarkers used as indicators of PI3K pathway inhibition in vivo. Also, BKM120 induced apoptosis in primary B-CLL cells culture in the presence and absence of stromal cell support. Our findings suggest that BKM120 should be tested clinically in CLL.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Adult , Aged , Apoptosis/drug effects , Blotting, Western , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Treatment Outcome , Tumor Cells, Cultured
8.
Cancer Prev Res (Phila) ; 5(4): 536-43, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22262811

ABSTRACT

Pharmacoepidemiologic studies provide evidence that use of metformin, a drug commonly prescribed for type II diabetes, is associated with a substantial reduction in cancer risk. Experimental models show that metformin inhibits the growth of certain neoplasms by cell autonomous mechanisms such as activation of AMP kinase with secondary inhibition of protein synthesis or by an indirect mechanism involving reduction in gluconeogenesis leading to a decline in insulin levels and reduced proliferation of insulin-responsive cancers. Here, we show that metformin attenuates paraquat-induced elevations in reactive oxygen species (ROS), and related DNA damage and mutations, but has no effect on similar changes induced by H(2)0(2), indicating a reduction in endogenous ROS production. Importantly, metformin also inhibited Ras-induced ROS production and DNA damage. Our results reveal previously unrecognized inhibitory effects of metformin on ROS production and somatic cell mutation, providing a novel mechanism for the reduction in cancer risk reported to be associated with exposure to this drug.


Subject(s)
DNA Damage , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Reactive Oxygen Species , Adenylate Kinase/metabolism , Animals , Cell Line , Diabetes Mellitus, Type 2/genetics , Enzyme-Linked Immunosorbent Assay/methods , Epidermal Growth Factor/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Humans , Hydrogen Peroxide/metabolism , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Mutagenesis , Mutation , NADP/metabolism
9.
Invest New Drugs ; 30(4): 1736-42, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21567185

ABSTRACT

The purpose of this study was to determine the degree to which the novel DNA-PKcs inhibitor, IC486241 (ICC), synergizes the cytotoxicity of DNA damaging agents in 3 genetically diverse breast cancer cell lines. The sulforhodamine B (SRB) assay was employed as a primary screening method to determine the in-vitro cytotoxicity and the degree of synergy of ICC in combination with the topoisomerase II inhibitor, doxorubicin, or the DNA cross linking agent, cisplatin. Molecular mechanisms underlying drug toxicity were probed using immunostaining and flow cytometry, as well as, the alkaline comet assay to detect DNA damage. In this study, improved cytotoxicity and significant synergy were observed with both anticancer agents in the presence of nontoxic concentrations of ICC. Moreover, ICC decreased doxorubicin-induced DNA-PKcs autophosphorylation on Ser2056 and increased doxorubicin-induced DNA fragmentation. In conclusion, the novel DNA-PKcs inhibitor, ICC, synergistically sensitized 3 breast cancer cell lines to doxorubicin and cisplatin. Enhanced efficacy of doxorubicin was achieved by inhibiting non-homologous end joining resulting in increased accumulation of DNA damage.


Subject(s)
Acridones/pharmacology , Breast Neoplasms/pathology , Cisplatin/pharmacology , DNA Damage , DNA, Neoplasm/metabolism , Doxorubicin/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Acridones/chemistry , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Cisplatin/therapeutic use , Comet Assay , Dimethyl Sulfoxide/pharmacology , Doxorubicin/therapeutic use , Drug Screening Assays, Antitumor , Drug Synergism , Female , Flow Cytometry , Humans , Inhibitory Concentration 50 , Protein Kinase Inhibitors/chemistry , Quinazolines/chemistry
10.
Invest New Drugs ; 30(3): 1248-56, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21221710

ABSTRACT

This study sought to measure the degree of synergy induced by specific small molecule inhibitors of DNA-PK [NU7026 and IC486241 (ICC)], a major component of the non-homologous end-joining (NHEJ) pathway, with SN38 or oxaliplatin. Synergy between the DNA damaging drugs and the DNA-PK inhibitors was assessed using the sulforhodamine-B assay (SRB). Effects of drug combinations on cell cycle and DNA-PK activity were determined using flow cytometry and western blot analysis. DNA damage was assessed via comet assay and quantification of γH2AX. The role of homologous recombination repair (HRR) was determined by nuclear Rad51 protein levels and a GFP reporter recombination assay. Significant reductions in the IC(50) values of SN38 were observed at 5 and 10 µM of DNA-PK inhibitors. Moreover, at 1-2 µM (attainable concentrations with ICC in mice) these DNA-PKcs inhibitors demonstrated synergistic reductions in the IC(50) of SN38. Flow cytometric data indicated that SN38 and SN38 in combination with DNA-PKcs inhibitors showed dramatic G2/M arrest at 24 h. Furthermore, reduced phosphorylation of DNA-PKcs and increased DNA damage were observed at this time point with SN38 in combination with DNA-PKcs inhibitors as compared to cells treated with SN38 alone. SN38 alone and in the presence of ICC increased nuclear Rad51 protein levels. Furthermore, inhibition of DNA-PKcs increased HRR suggesting that NHEJ is a negative regulator of HRR. These data indicate that small molecule inhibitors of DNA-PKcs dramatically enhance the efficacy of SN38 in colon cancer cell lines.


Subject(s)
Acridones/pharmacology , Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Chromones/pharmacology , DNA-Activated Protein Kinase/antagonists & inhibitors , Morpholines/pharmacology , Protein Kinase Inhibitors/pharmacology , Camptothecin/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Colonic Neoplasms/pathology , Comet Assay , Drug Synergism , HCT116 Cells , HT29 Cells , Humans , Irinotecan , Organoplatinum Compounds/pharmacology , Pyridines/pharmacology , Topoisomerase I Inhibitors/pharmacology
11.
Leuk Res ; 35(8): 1080-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21281966

ABSTRACT

Resistance to chlorambucil in chronic lymphocytic leukemia (CLL) has been associated with increased DNA repair. Specifically, inhibition of either c-abl, which modulates Rad51 directed homologous recombination or DNA-PK dependent nonhomologous end joining has been shown to sensitize primary CLL lymphocytes to chlorambucil. Here we report that inhibition of c-abl can result in a compensatory increase in DNA-PK and thus inhibition of both c-abl and DNA-PK optimally sensitizes CLL lymphocytes to chlorambucil. In this paper we report a drug-induced compensatory change between two DNA repair pathways with potential therapeutic implications in CLL therapy.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , B-Lymphocytes/pathology , Chlorambucil/pharmacology , DNA-Activated Protein Kinase/antagonists & inhibitors , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Piperazines/pharmacology , Pyrimidines/pharmacology , Recombination, Genetic , B-Lymphocytes/metabolism , Benzamides , Blotting, Western , Comet Assay , DNA-Activated Protein Kinase/metabolism , Drug Resistance, Neoplasm , Drug Synergism , Humans , Imatinib Mesylate , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Tumor Cells, Cultured
12.
Leuk Res ; 35(1): 99-102, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20573397

ABSTRACT

B-cell chronic lymphocytic leukemia (CLL) is the most common leukemia in adults and there is no cure for the disease. Although dasatinib is cytotoxic to primary CLL lymphocytes in vitro, the drug has been shown to be active in a small percent of CLL patients. Our previous results suggest that dasatinib targets del17 CLL lymphocytes which are the CLL patients with the worst prognosis. Here we present mechanistic evidence that dasatinib induces endoplasmic reticulum stress and autophagy in CLL lymphocytes. Furthermore we provide evidence suggesting that autophagy mediates resistance to the drugs, process that is modulated by p53.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , Tumor Suppressor Protein p53/physiology , Dasatinib , Drug Resistance, Neoplasm , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
13.
Cancer Chemother Pharmacol ; 68(3): 643-51, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21120481

ABSTRACT

PURPOSE: The tyrosine kinase inhibitor, imatinib, has the potential to indirectly inhibit DNA repair. This mechanism of action has been shown to mediate sensitization to chlorambucil in chronic lymphocytic leukemia (CLL). To evaluate this effect in vivo, we performed a phase I study of chlorambucil combined with imatinib in relapsed CLL patients. METHODS: The three dose levels studied included imatinib at 300, 400, or 600 mg/day. Imatinib was given on days 1-10, and chlorambucil (8 mg/m(2) daily) was given on days 3-7 of a 28-day cycle (up to 6 cycles). RESULTS: Eleven patients participated in this study. Low-grade gastrointestinal toxicities were observed in a dose-dependent manner. Forty-five percent of patients responded (two unconfirmed CRs and three PRs). Two responding patients were fludarabine refractory. The in vitro IC(50) of chlorambucil alone or in the presence of 5 µM imatinib in CLL lymphocytes correlated with the decrease in lymphocyte counts on day 15. Imatinib plasma concentrations achieved in patients were in the range of those effective in in vitro sensitization studies. CONCLUSION: The combination of chlorambucil and imatinib in patients with previously treated CLL was well tolerated and showed evidence of clinical efficacy. Based on our results, we recommend the 400 mg daily dose of imatinib on days 1-10 with 8 mg/m(3) chlorambucil on days 3-7 every 28 days as the phase II dose. This represents the first clinical trial examining the potential synergy between a tyrosine kinase inhibitor and a conventional alkylating agent for the treatment of CLL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Benzamides , Chlorambucil/administration & dosage , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Imatinib Mesylate , Lymphocyte Count , Lymphocytes/drug effects , Male , Middle Aged , Piperazines/administration & dosage , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrimidines/administration & dosage , Rad51 Recombinase/antagonists & inhibitors , Recombination, Genetic , Treatment Outcome , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use
14.
Endocr Relat Cancer ; 17(2): 351-60, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20228137

ABSTRACT

The molecular mechanisms responsible for the association of obesity with adverse colon cancer outcomes are poorly understood. We investigated the effects of a high-energy diet on growth of an in vivo colon cancer model. Seventeen days following the injection of 5x10(5) MC38 colon carcinoma cells, tumors from mice on the high-energy diet were approximately twice the volume of those of mice on the control diet. These findings were correlated with the observation that the high-energy diet led to elevated insulin levels, phosphorylated AKT, and increased expression of fatty acid synthase (FASN) by the tumor cells. Metformin, an antidiabetic drug, leads to the activation of AMPK and is currently under investigation for its antineoplastic activity. We observed that metformin blocked the effect of the high-energy diet on tumor growth, reduced insulin levels, and attenuated the effect of diet on phosphorylation of AKT and expression of FASN. Furthermore, the administration of metformin led to the activation of AMPK, the inhibitory phosphorylation of acetyl-CoA carboxylase, the upregulation of BNIP3 and increased apoptosis as estimated by poly (ADP-ribose) polymerase (PARP) cleavage. Prior work showed that activating mutations of PI3K are associated with increased AKT activation and adverse outcome in colon cancer; our results demonstrate that the aggressive tumor behavior associated with a high-energy diet has similar effects on this signaling pathway. Furthermore, metformin is demonstrated to reverse the effects of the high-energy diet, thus suggesting a potential role for this agent in the management of a metabolically defined subset of colon cancers.


Subject(s)
Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Energy Intake/drug effects , Fatty Acid Synthases/antagonists & inhibitors , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Apoptosis/drug effects , Carcinoma/enzymology , Colonic Neoplasms/enzymology , Fatty Acid Synthases/biosynthesis , Insulin/blood , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
16.
Br J Haematol ; 143(5): 698-706, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19062342

ABSTRACT

The dual c-abl/Src kinase inhibitor, dasatinib, utilized to treat chronic myeloid leukaemia (CML) when used at clinically attainable sublethal concentrations, synergistically sensitized primary chronic lymphocytic leukaemia (CLL) lymphocytes to chlorambucil and fludarabine. In contrast, dasatinib alone demonstrated toxicity to CLL lymphocytes at concentrations that are generally not clinically attainable. Dasatinib resistance and poorer dasatinib-mediated sensitization to chlorambucil and fludarabine was associated with higher expression of c-abl protein levels. In contrast, chlorambucil and fludarabine resistance correlated with basal p53 protein levels. Moreover, Western blot analysis after in vitro treatment of primary CLL lymphocytes with dasatinib, chlorambucil and/or fludarabine, showed that dasatinib: (i) inhibited c-abl function (e.g. downregulation of c-abl protein levels and decreased the phosphorylation of a c-abl downstream target, Dok2), (ii) decreased chlorambucil/fludarabine induced accumulation of p53 protein levels, (iii) altered the response to chlorambucil/fludarabine induced DNA-damage as evidenced by an increase in chlorambucil/fludarabine-induced H2AX phosphorylation, and (iv) accentuated the c-abl downregulation induced by chlorambucil/fludarabine. Our results suggest that dasatinib in combination with chlorambucil or fludarabine may improve the therapy of CLL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , Blotting, Western/methods , Cell Survival , Chlorambucil/administration & dosage , Cytotoxicity Tests, Immunologic , DNA Mutational Analysis , Dasatinib , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Linear Models , Proto-Oncogene Proteins c-abl/metabolism , Statistics, Nonparametric , Tumor Suppressor Protein p53/metabolism , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
17.
J Pharmacol Exp Ther ; 321(3): 848-55, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17351105

ABSTRACT

Chlorambucil (CLB) treatment is used in chronic lymphocytic leukemia (CLL) but resistance to CLB develops in association with accelerated repair of CLB-induced DNA damage. Phosphorylated histone H2AX (gammaH2AX) is located at DNA double-strand break (DSB) sites; furthermore, it recruits and retains damage-responsive proteins. This damage can be repaired by nonhomologous DNA end-joining (NHEJ) and/or homologous recombinational repair (HR) pathways. A key component of NHEJ is the DNA-dependent protein kinase (DNA-PK) complex. Increased DNA-PK activity is associated with resistance to CLB in CLL. We used the specific DNA-PK inhibitor 2-(morpholin-4-yl)-benzo[h]chomen-4-one (NU7026) to sensitize CLL cells to chlorambucil. Our results indicate that in a CLL cell line (I83) and in primary CLL-lymphocytes, chlorambucil plus NU7026 has synergistic cytotoxic activity at nontoxic doses of NU7026. CLB treatment results in G(2)/M phase arrest, and NU7026 increases this CLB-induced G(2)/M arrest. Moreover, a kinetic time course demonstrates that CLB-induced DNA-PK activity was inhibited by NU7026, providing direct evidence of the ability of NU7026 to inhibit DNA-PK function. DSBs, visualized as gammaH2AX, were enhanced 24 to 48 h after CLB and further increased by CLB plus NU7026, suggesting that the synergy of the combination is mediated by NU7026 inhibition of DNA-PK with subsequent inhibition of DSB repair.


Subject(s)
B-Lymphocytes/drug effects , Chlorambucil/pharmacology , Chromones/pharmacology , DNA Repair/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , Morpholines/pharmacology , Apoptosis/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA-Activated Protein Kinase/metabolism , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Inhibitors/pharmacology , Histones/metabolism , Humans , Inhibitory Concentration 50 , Phosphorylation/drug effects
18.
Cell Cycle ; 6(5): 606-11, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17351336

ABSTRACT

In vertebrate cells, Xrcc3 initiates the repair of exogenous induced-DNA breaks during S and G(2)/M phases of the cell cycle by homologous recombination. However, much less is known of the role of Xrcc3 in the response to spontaneous DNA breaks. Using a siRNA approach, we show that depletion of XRCC3 inhibits the proliferation of MCF7 breast cancer cells. This inhibition of replication coincides with the accumulation of DNA breaks, as shown by the comet assay. Cell cycle specific analysis of gammaH2AX expression shows that S and G2/M phase cells express the highest fraction of gammaH2AX positive cells. This is consistent with replication-dependent accumulation of DNA breaks and deficient homologous recombination. While the induction of gammaH2AX is followed by cell death in parental cells, a p53 knockdown derivative becomes more resistant to XRCC3 depletion-induced death without changes in the levels of gammaH2AX. These results show that XRCC3 is required for the proliferation of MCF7 cells, and that decrease in its expression leads to the accumulation of DNA breaks and the induction of p53-dependent cell death.


Subject(s)
DNA Breaks , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Tumor Suppressor Protein p53/physiology , Cell Death/genetics , DNA Repair/genetics , DNA-Binding Proteins/metabolism , Humans , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
19.
Cell Tissue Res ; 322(3): 365-78, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16082520

ABSTRACT

Nucleoside di-phosphate kinase enzyme (NDPK) isoforms, encoded by the nm23 family of genes, may be involved in various cellular differentiation and proliferation processes. We have therefore analyzed the expression of nm23-M1, -M2, -M3, and -M4 during embryonic mouse development. In situ hybridization data has revealed the differential expression of nm23 mRNA during organogenesis. Whereas nm23-M1 and -M3 are preferentially expressed in the nervous and sensory systems, nm23-M2 mRNA is found ubiquitously. Irrespective of the developmental state studied, nm23-M4 mRNA is only expressed at low levels in a few embryonic organs. In the cerebellum and cerebral cortex, nm23-M1, -M2, and -M3 are present in the neuronal differentiation layer, whereas nm23-M4 mRNA is distributed in the proliferating layer. Thus, nm23 mRNA is differentially expressed, and the diverse NDPK isoforms are sequentially involved in various developmental processes.


Subject(s)
Central Nervous System/embryology , Central Nervous System/metabolism , Monomeric GTP-Binding Proteins/biosynthesis , Monomeric GTP-Binding Proteins/genetics , Organogenesis/physiology , Animals , Base Sequence , Cells, Cultured , Gene Expression , In Situ Hybridization , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Monomeric GTP-Binding Proteins/metabolism , NM23 Nucleoside Diphosphate Kinases , Nucleoside Diphosphate Kinase D , Protein Isoforms , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
J Comp Neurol ; 444(4): 306-23, 2002 Mar 18.
Article in English | MEDLINE | ID: mdl-11891645

ABSTRACT

Nm23 has been identified as a gene family encoding different isoforms of nucleoside diphosphate kinase (NDPK). This protein is a key enzyme in nucleotide metabolism and has been shown to play important roles in various cellular functions. In the present study, we have investigated the expression of three isotypes in mouse dorsal root ganglia. In situ hybridization and reverse transcriptase-polymerase chain reaction analysis demonstrated high levels of nm23-M1, -M2, and -M3 mRNA expression in peripheral nervous tissue. Moreover, in situ hybridization also displayed a specific nuclear localization for nm23-M2 mRNA. Immunohistochemistry with light and electron microscopy on isoform-specific antibodies revealed a differential subcellular distribution of NDPK isoforms. Isoform A was mainly cytosolic, showing only partial association with organelles. In contrast, isoform B was also found in the nucleus, which is in agreement with its proposed role as a transcription factor. The results also indicate a preferential association of isoform C with endoplasmic reticulum and plasma membranes in neuronal cells. Furthermore, isoform C appeared to combine with other NDPK isoforms as demonstrated by double-labeling evidence by electron microscopy and might be responsible for binding NDPK oligomers to membranes. Thus, isoform C may be considered as a protein of importance for maintaining intracellular pools of GTP in the vicinity of membranes and, hence, for transmembrane signaling. The results indicate a high expression of NDPK isoforms, not only in the central but also in the peripheral nervous system. Their different subcellular compartmentalization suggests that they have isoform-specific roles in neuronal cell physiology.


Subject(s)
Ganglia, Spinal/physiology , Gene Expression , Monomeric GTP-Binding Proteins/genetics , Nucleoside-Diphosphate Kinase , Transcription Factors/genetics , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Ganglia, Spinal/ultrastructure , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Microscopy, Electron , Monomeric GTP-Binding Proteins/metabolism , NM23 Nucleoside Diphosphate Kinases , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...