Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Cancer Res ; 80(2): 234-248, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31723002

ABSTRACT

The development of potent and selective therapeutic approaches to glioblastoma (GBM), one of the most aggressive primary brain tumors, requires identification of molecular pathways that critically regulate the survival and proliferation of GBM. Previous studies have reported that deregulated expression of N-myc downstream regulated gene 1 (NDRG1) affects tumor growth and clinical outcomes of patients with various types of cancer including glioma. Here, we show that high level expression of NDRG1 in tumors significantly correlated with better prognosis of patients with GBM. Loss of NDRG1 in GBM cells upregulated GSK3ß levels and promoted cell proliferation, which was reversed by selective inhibitors of GSK3ß. In contrast, NDRG1 overexpression suppressed growth of GBM cells by decreasing GSK3ß levels via proteasomal degradation and by suppressing AKT and S6 cell growth signaling, as well as cell-cycle signaling pathways. Conversely, GSK3ß phosphorylated serine and threonine sites in the C-terminal domain of NDRG1 and limited the protein stability of NDRG1. Furthermore, treatment with differentiation inducing factor-1, a small molecule derived from Dictyostelium discoideum, enhanced NDRG1 expression, decreased GSK3ß expression, and exerted marked NDRG1-dependent antitumor effects in vitro and in vivo. Taken together, this study revealed a novel molecular mechanism by which NDRG1 inhibits GBM proliferation and progression. Our study thus identifies the NDRG1/GSK3ß signaling pathway as a key growth regulatory program in GBM, and suggests enhancing NDRG1 expression in GBM as a potent strategy toward the development of anti-GBM therapeutics. SIGNIFICANCE: This study identifies NDRG1 as a potent and endogenous suppressor of glioblastoma cell growth, suggesting the clinical benefits of NDRG1-targeted therapeutics against glioblastoma.


Subject(s)
Brain Neoplasms/pathology , Cell Cycle Proteins/metabolism , Glioblastoma/pathology , Glycogen Synthase Kinase 3 beta/metabolism , Hexanones/administration & dosage , Intracellular Signaling Peptides and Proteins/metabolism , Adult , Aged , Aged, 80 and over , Brain/pathology , Brain/surgery , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Brain Neoplasms/surgery , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Glioblastoma/drug therapy , Glioblastoma/mortality , Glioblastoma/surgery , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Hexanones/therapeutic use , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Phosphorylation/drug effects , Primary Cell Culture , Prognosis , Protein Stability/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Thiadiazoles/pharmacology , Xenograft Model Antitumor Assays
2.
Drug Chem Toxicol ; 43(2): 113-126, 2020 Mar.
Article in English | MEDLINE | ID: mdl-29745774

ABSTRACT

The aim of this study was to evaluate the potentials of rutin on 2,5-hexanedione-induced toxicities. Two successive phases were involved using in silico and in vivo approaches. The in silico was adopted for potential oral toxicity and docking. The in vivo was carried-out in two stages for two weeks; the ameliorative (stage 1, first week), preventive, and curative studies (stage 2, extended to second week). In stage 1, rats were divided into four groups of seven each (distilled water, 3% (v/v) 2,5-hexanedione, 10 mg/kg rutin, and co-administration). In stage 2, the experimental groups were given either rutin or 2,5-hexanedione and treated in reverse order. Lipid peroxidation, protein carbonyl, and DNA fragmentation in tissues and bone marrow cells micronucleus were determined. The predicted Median lethal dose (LD50) of >5000 mg/kg and toxicity class of five (5) indicates the safety of rutin when orally administered. 2,5-Hexanedione comfortably docked in to the active sites of SOD (-22.857Kcal/mol; KI = 0.9621 µM), GPx (-11.2032Kcal/mol; KI = 0.9813 µM), and CAT (-16.446Kcal/mol; KI = 0.9726 µM) with strong hydrogen bond and hydrophobic interactions. However, only strong hydrophobic interaction was observed in the case of DNA (-3.3296Kcal/mol; KI = 0.9944). In vivo findings revealed deleterious effects of 2,5-hexanedione through induction of oxidative and chromosomal/DNA damage characterized by higher level of malondialdehyde, micronuclei formations, and DNA fragmentation. These have invariably, validates the findings from in silico experiments. Furthermore, rutin was able to ameliorate, protect, and reverse these effects, and was relatively non-toxic corroborating toxicity predictions. Rutin exhibited counteractive effects on 2,5-hexanedione-induced oxidative, chromosomal, and DNA damage.


Subject(s)
DNA Damage/drug effects , Hexanones/toxicity , Rutin/pharmacology , Animals , Chromosome Aberrations/drug effects , Computer Simulation , DNA Fragmentation/drug effects , Hexanones/administration & dosage , Lethal Dose 50 , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Molecular Docking Simulation , Oxidative Stress/drug effects , Rats , Rats, Wistar
3.
Am J Clin Oncol ; 41(3): 230-235, 2018 03.
Article in English | MEDLINE | ID: mdl-26709865

ABSTRACT

BACKGROUND: Imexon is a cyanoaziridine-derived iminopyrrolidone which has synergistic cytotoxicity with gemcitabine. A phase 1 study of the combination demonstrated good tolerance with encouraging clinical activity and thus we conducted this randomized phase II study. MATERIALS AND METHODS: Patients with measurable, metastatic, treatment-naive pancreatic adenocarcinoma were randomized 1:1 to receive gemcitabine at 1000 mg/m days 1, 8, and 15 with either imexon, 875 mg/m or placebo days 1, 8, and 15 every 28 days. The primary endpoint was overall survival. Secondary endpoints included progression-free survival and response rate. RESULTS: A total of 142 patients were randomized, 72 to the imexon containing arm and 70 to the placebo arm. Patients in the imexon arm received an average of 3.6 cycles (range, 1 to 23) compared with 4.4 (range, 1 to 21) in the placebo arm. There was no increased rate of ≥grade 3 toxicity in the imexon arm. Seven patients had objective responses in the imexon arm (13.7%), whereas 9 did in the placebo arm (17%). In the imexon arm, 23 patients had ≥50% reduction in CA 19-9 from baseline (33%), whereas 22 did in the placebo arm (31.4%). The median progression-free survival was 2.8 months in the imexon arm (95% confidence interval [CI], 2.0-4.1 m) and 3.8 months in the placebo arm (95% CI, 2.2-4.7 m), P=0.504. The median overall survival time in the imexon arm was 5.2 months (95% CI, 4.2-6.7 m) as compared with 6.8 m (95% CI, 4.9-8.5 m) in the placebo arm, P=0.6822. CONCLUSIONS: The combination of imexon and gemcitabine does not result in improved outcome as initial therapy of metastatic pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Hexanones/administration & dosage , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Deoxycytidine/administration & dosage , Double-Blind Method , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/mortality , Progression-Free Survival , Gemcitabine , Pancreatic Neoplasms
4.
Toxicol Pathol ; 44(5): 763-83, 2016 07.
Article in English | MEDLINE | ID: mdl-27025954

ABSTRACT

Occupational exposure to 2,3-butanedione (BD) vapors has been associated with severe respiratory disease leading to the use of potentially toxic substitutes. We compared the reactivity and respiratory toxicity of BD with that of two structurally related substitutes, 2,3-pentanedione (PD) and 2,3-hexanedione (HD). Chemical reactivity of the diketones with an arginine substrate decreased with increasing chain length (BD > PD > HD). Animals were evaluated the morning after a 2-week exposure to 0, 100, 150, or 200 ppm BD, PD, or HD (postexposure) or 2 weeks later (recovery). Bronchial fibrosis was observed in 5/5 BD and 5/5 PD rats at 200 ppm and in 4/6 BD and 6/6 PD rats at 150 ppm in the postexposure groups. Following recovery, bronchial fibrosis was observed in all surviving rats exposed to 200 ppm BD (5/5) or PD (3/3) and in 2/10 BD and 7/9 PD rats exposed to 150 ppm. Bronchial fibrosis was observed only in 2/12 HD-exposed rats in the 200 ppm postexposure group. Patchy interstitial fibrosis affected lungs of recovery groups exposed to 200 ppm PD (3/3) or BD (1/5) and to 150 ppm PD (4/9) or BD (7/10) and correlated with pulmonary function deficits. BD and PD were more reactive and produced more bronchial fibrosis than HD.


Subject(s)
Flavoring Agents/toxicity , Lung/drug effects , Lung/pathology , Animals , Diacetyl/administration & dosage , Diacetyl/toxicity , Dose-Response Relationship, Drug , Flavoring Agents/administration & dosage , Hexanones/administration & dosage , Hexanones/toxicity , Inhalation Exposure , Male , Pentanones/administration & dosage , Pentanones/toxicity , Rats
5.
J Pharmacol Sci ; 127(4): 446-55, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25913757

ABSTRACT

Differentiation-inducing factor-1 (DIF-1) produced by Dictyostelium discoideum strongly inhibits the proliferation of various types of cancer cells by suppression of the Wnt/ß-catenin signal transduction pathway. In the present study, we examined the effect of differentiation-inducing factor-3 (DIF-3), a monochlorinated metabolite of DIF-1 that is also produced by D. discoideum, on human colon cancer cell lines HCT-116 and DLD-1. DIF-3 strongly inhibited cell proliferation by arresting the cell cycle at the G0/G1 phase. DIF-3 reduced the expression levels of cyclin D1 and c-Myc by facilitating their degradation via activation of GSK-3ß in a time and dose-dependent manner. In addition, DIF-3 suppressed the expression of T-cell factor 7-like 2, a key transcription factor in the Wnt/ß-catenin signaling pathway, thereby reducing the mRNA levels of cyclin D1 and c-Myc. Subsequently, we examined the in vivo effects of DIF-3 in Mutyh(-/-) mice with oxidative stress-induced intestinal cancers. Repeated oral administration of DIF-3 markedly reduced the number and size of cancers at a level comparable to that of DIF-1. These data suggest that DIF-3 inhibits intestinal cancer cell proliferation in vitro and in vivo, probably by mechanisms similar to those identified in DIF-1 actions, and that DIF-3 may be a potential novel anti-cancer agent.


Subject(s)
Antineoplastic Agents , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Hexanones/pharmacology , Administration, Oral , Animals , Cell Cycle/drug effects , Cell Proliferation/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HCT116 Cells , Hexanones/administration & dosage , Humans , Mice, Transgenic , Oxidative Stress , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transcription Factor 7-Like 2 Protein/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/physiology
6.
Drug Chem Toxicol ; 38(4): 400-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25388508

ABSTRACT

2,5-Hexanedione (2,5-HD) is an aliphatic diketone identified as the main neurotoxic metabolite of the industrial chemicals n-hexane and methyl-n-butyl ketone. Considering the dearth of information on the female reproductive toxicity effects of 2,5-HD in the literature, we assessed the potential oxidative stress mechanisms of 2,5-HD in the ovary and uterus of Wistar rats. A total of 32 female rats were randomly allotted to four groups, in which rats were exposed to 2,5-HD at doses of 0% (control), 0.25%, 0.5% and 1.0% respectively in their drinking water for 21 days. The results showed that 2,5-HD significantly increased ovarian and uterine malondialdehyde (MDA) and hydrogen peroxide (H2O2) levels (p < 0.05). Additionally, while significant decreases in ovarian catalase, superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione-S-transferase (GST) activities occurred in all the 2,5-HD-treated groups, uterine catalase, GST, and GPx activities increased. Further, 2,5-HD increased follicle stimulating hormone, but decreased estrogen levels in all the 2,5-HD-treated groups, while prolactin increased in the 0.5, and 1.0% 2,5-HD-treated rats compared with the control (p < 0.05). Thus, these data imply that 2,5-HD exposure disrupts hormonal homeostasis and induces oxidative stress in the ovary and uterus of rats. These findings may therefore have toxicological implications in women occupationally exposed to n-hexane and methyl-n-butyl ketone.


Subject(s)
Hexanones/toxicity , Ovary/drug effects , Oxidative Stress/drug effects , Uterus/drug effects , Animals , Catalase/metabolism , Dose-Response Relationship, Drug , Endocrine Disruptors/administration & dosage , Endocrine Disruptors/toxicity , Estrogens/metabolism , Female , Follicle Stimulating Hormone/metabolism , Glutathione Peroxidase/metabolism , Hexanones/administration & dosage , Hydrogen Peroxide/metabolism , Malondialdehyde/metabolism , Ovary/pathology , Prolactin/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Uterus/pathology
7.
Environ Toxicol Pharmacol ; 38(3): 807-13, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25305742

ABSTRACT

The interference of N-acetylcysteine (NAC) on 2,5-hexanedione (2,5-HD) neurotoxicity was evaluated through behavioral assays and the analysis of urinary 2,5-HD, dimethylpyrrole norleucine (DMPN), and cysteine-pyrrole conjugate (DMPN NAC), by ESI-LC-MS/MS, in rats exposed to 2,5-HD and co-exposed to 2,5-HD and NAC. Wistar rats were treated with 4 doses of: 400mg 2,5-HD/kg bw (group I), 400mg 2,5-HD/kg bw+200mg NAC/kg bw (group II), 200mg NAC/kg bw (group III) and with saline (group IV). The results show a significant decrease (p<0.01) in urinary DMPN and free 2,5-HD, a significant increase (p<0.01) in DMPN NAC excretion, and a significant recovery (p<0.01) on motor activity in rats co-exposed to 2,5-HD+NAC, as compared with rats exposed to 2,5-HD alone. Taken together, our findings suggest that at the studied conditions NAC protects against 2,5-HD neurotoxicity and DMPN may be proposed as a new sensitive and specific biomarker of 2,5-HD neurotoxicity in animals treated with a toxic amount of 2,5-hexanedione.


Subject(s)
Acetylcysteine/administration & dosage , Hexanones/administration & dosage , Motor Activity/drug effects , Neuroprotective Agents/administration & dosage , Neurotoxins/administration & dosage , Pyrroles/urine , Acetylcysteine/pharmacology , Animals , Chromatography, Liquid , Hexanones/toxicity , Hexanones/urine , Male , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Neurotoxins/urine , Norleucine/urine , Rats , Rats, Wistar , Tandem Mass Spectrometry
8.
Blood ; 124(8): 1259-65, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25016003

ABSTRACT

Lymphoma cells are subject to higher levels of oxidative stress compared with their normal counterparts and may be vulnerable to manipulations of the cellular redox balance. We therefore designed a phase 2 study of imexon (Amplimexon/NSC-714597), a prooxidant molecule, in patients with relapsed/refractory B-cell non-Hodgkin lymphoma (NHL). Imexon was administered at 1000 mg/m(2) IV daily for 5 days in 21-day cycles. Gene expression analysis performed on pretreatment tumor specimens included 13 transcripts used to generate a redox signature score, previously demonstrated to correlate with lymphoma prognosis. Twenty-two patients were enrolled having follicular (n = 9), diffuse large B-cell (DLBCL) (n = 5), mantle cell (n = 3), transformed follicular (n = 2), small lymphocytic (n = 2), and Burkitt (n = 1) lymphoma. The most common grade 3/4 adverse events were anemia (14%) and neutropenia (9%). The overall response rate was 30%, including responses in follicular lymphoma (4 of 9) and DLBCL (2 of 5). Gene expression analyses revealed CD68 and the redox-related genes, GPX1 and SOD2, as well as a higher redox score to correlate with clinical responses. Therefore, pretreatment markers of oxidative stress may identify patients likely to respond to this therapeutic approach. This trial was registered at www.clinicaltrials.gov as #NCT01314014.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Hexanones/administration & dosage , Oxidants/administration & dosage , Oxidative Stress/drug effects , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/biosynthesis , Disease-Free Survival , Female , Glutathione Peroxidase/biosynthesis , Hexanones/adverse effects , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/pathology , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Oxidants/adverse effects , Recurrence , Superoxide Dismutase/biosynthesis , Survival Rate , Glutathione Peroxidase GPX1
9.
Article in English | MEDLINE | ID: mdl-23348767

ABSTRACT

BACKGROUND: Inhibin B is a heterodimer glycoprotein that downregulates follicle-stimulating hormone and is produced predominantly by Sertoli cells. The potential correlation between changes in plasma Inhibin B and Sertoli cell toxicity was evaluated in male rats administered testicular toxicants in eight studies. Inhibin B fluctuations over 24 hr were also measured. METHODS: Adult rats were administered one of eight testicular toxicants for 1 to 29 days. The toxicants were DL-ethionine, dibutyl phthalate, nitrofurazone, 2,5-hexanedione, 17-alpha ethinylestradiol, ethane dimethane sulfonate, hexachlorophene, and carbendazim. In a separate study plasma was collected throughout a 24-hr period via an automatic blood sampler. RESULTS: Histomorphologic testicular findings included seminiferous tubule degeneration, round and elongate spermatid degeneration/necrosis, seminiferous tubule vacuolation, aspermatogenesis, and interstitial cell degeneration. There was a varying response of plasma Inhibin B levels to seminiferous tubule toxicity, with three studies showing high correlation, three studies with a response only at a certain time or dose, and two studies with no Inhibin B changes. In a receiver operating characteristics exclusion model analysis, where treated samples without histopathology were excluded, Inhibin B showed a sensitivity of 70% at 90% specificity in studies targeting seminiferous tubule toxicity. CONCLUSION: Decreases in Inhibin B correlated with Sertoli cell toxicity in the majority of studies evaluated, demonstrating the value of Inhibin B as a potential biomarker of testicular toxicity. There was no correlation between decreases in Inhibin B and interstitial cell degeneration. In addition, a pattern of Inhibin B secretion could not be identified over 24 hr.


Subject(s)
Environmental Pollutants/administration & dosage , Environmental Pollutants/toxicity , Inhibins/blood , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/toxicity , Carbamates/administration & dosage , Carbamates/toxicity , Dibutyl Phthalate/administration & dosage , Dibutyl Phthalate/toxicity , Ethinyl Estradiol/administration & dosage , Ethinyl Estradiol/toxicity , Ethionine/administration & dosage , Ethionine/toxicity , Hexachlorophene/administration & dosage , Hexachlorophene/toxicity , Hexanones/administration & dosage , Hexanones/toxicity , Male , Mesylates/administration & dosage , Mesylates/toxicity , Nitrofurazone/administration & dosage , Nitrofurazone/toxicity , ROC Curve , Rats , Rats, Sprague-Dawley , Rats, Wistar , Testis/drug effects , Testis/pathology
10.
PLoS One ; 7(8): e44280, 2012.
Article in English | MEDLINE | ID: mdl-22952946

ABSTRACT

Current human reproductive risk assessment methods rely on semen and serum hormone analyses, which are not easily comparable to the histopathological endpoints and mating studies used in animal testing. Because of these limitations, there is a need to develop universal evaluations that reliably reflect male reproductive function. We hypothesized that toxicant-induced testicular injury can be detected in sperm using mRNA transcripts as indicators of insult. To test this, we exposed adult male Fischer 344 rats to low doses of model testicular toxicants and classically characterized the testicular injury while simultaneously evaluating sperm mRNA transcripts from the same animals. Overall, this study aimed to: 1) identify sperm transcripts altered after exposure to the model testicular toxicant, 2,5-hexanedione (HD) using microarrays; 2) expand on the HD-induced transcript changes in a comprehensive time course experiment using qRT-PCR arrays; and 3) test these injury indicators after exposure to another model testicular toxicant, carbendazim (CBZ). Microarray analysis of HD-treated adult Fischer 344 rats identified 128 altered sperm mRNA transcripts when compared to control using linear models of microarray analysis (q<0.05). All transcript alterations disappeared after 3 months of post-exposure recovery. In the time course experiment, time-dependent alterations were observed for 12 candidate transcripts selected from the microarray data based upon fold change and biological relevance, and 8 of these transcripts remained significantly altered after the 3-month recovery period (p<0.05). In the last experiment, 8 candidate transcripts changed after exposure to CBZ (p<0.05). The two testicular toxicants produced distinct molecular signatures with only 4 overlapping transcripts between them, each occurring in opposite directions. Overall, these results suggest that sperm mRNA transcripts are indicators of low dose toxicant-induced testicular injury in the rat.


Subject(s)
Benzimidazoles/administration & dosage , Benzimidazoles/toxicity , Carbamates/administration & dosage , Carbamates/toxicity , Hexanones/administration & dosage , Hexanones/toxicity , Spermatozoa/metabolism , Testis/metabolism , Testis/pathology , Animals , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Male , Oligonucleotide Array Sequence Analysis , Organ Size/drug effects , Organ Size/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Spermatozoa/drug effects , Testis/drug effects , Time Factors
11.
Cancer Chemother Pharmacol ; 69(4): 983-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22139443

ABSTRACT

PURPOSE: Differentiation-inducing factor-1 (DIF-1) is a morphogen originally identified in the amoebozoan Dictyostelium discoideum. In mammalian cells, it has been shown to activate GSK3ß, which in turn is expected to reduce levels of ß-catenin and cyclin D1, thus mediating DIF-1 antiproliferative properties. Since this could alter the expression and activity of E2F1 transcription factor and consequently those of the prognostic marker/chemotherapy target thymidylate synthase (TS), we evaluated (1) whether DIF-1 could effectively regulate these genes, (2) whether it could interfere with cell viability, and (3) whether DIF-1 activity could enhance the efficacy of the TS inhibitor 5-fluorouracil (5-FU). METHODS: We investigated the effects of DIF-1 in continuous human cell lines derived from two oral tumor histotypes (corresponding to an adenosquamous and a squamous carcinoma) and a gingival epithelium. We evaluated mRNA accumulation by means of quantitative real-time PCR and efficacy of drugs on cell viability by means of MTT assay. RESULTS: DIF-1 inhibited the accumulation of E2F1 mRNA and reduces TS mRNA levels in tumor cell lines, but did not alter mRNA levels in the gingival counterpart. As a result, it inhibited proliferation preferentially of tumor cell in time- and concentration-dependent manner. Moreover, it enhanced cytotoxic effects of 5-FU only in tumor cell, whereas reduced them in the gingival counterpart. CONCLUSIONS: These findings suggest a tumor-specific action of DIF-1 on oral carcinoma cells. Thus, interfering with E2F1 and TS transcription, DIF-1 potentiates TS enzymatic inhibitors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , E2F1 Transcription Factor/antagonists & inhibitors , Fluorouracil/pharmacology , Hexanones/pharmacology , Mouth Neoplasms/drug therapy , RNA, Messenger/antagonists & inhibitors , Thymidylate Synthase/antagonists & inhibitors , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , E2F1 Transcription Factor/biosynthesis , E2F1 Transcription Factor/genetics , Fluorouracil/administration & dosage , Gene Expression , Hexanones/administration & dosage , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Thymidylate Synthase/biosynthesis , Thymidylate Synthase/genetics
12.
Cancer Chemother Pharmacol ; 67(1): 183-92, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20339847

ABSTRACT

PURPOSE: Gemcitabine (GEM) is currently the standard first line treatment for pancreatic cancer; however, the overall survival of patients with this disease remains poor. Imexon is a pro-oxidant small molecule which produced a high response rate in combination with GEM in a phase I trial in pancreatic cancer. In this study, we investigate the combination of GEM with a novel redox-active agent, imexon, in vitro and in vivo. METHODS: Median effect analysis was used for in vitro combination cytotoxicity. The effect of imexon on GEM metabolism and uptake into cells and into DNA and effects on ribonucleotide reductase (RNR) were examined in vitro. The pharmacokinetics and antitumor efficacy of the imexon/GEM combination was evaluated in mouse models. RESULTS: In three human pancreatic cancer lines, there was additivity for the imexon/GEM combination. There was significantly greater efficacy for the drug combination in Panc-1 xenograft tumors. A pharmacokinetic study in mice showed a near doubling in the AUC of imexon when GEM was co-administered, with no effect of imexon on GEM's pharmacokinetic disposition. In vitro, imexon did not alter GEM's metabolism or uptake into DNA, but significantly inhibited RNR, and this effect was greater when combined with GEM. CONCLUSIONS: These results suggest that the interaction between imexon and GEM may be due to complimentary inhibition of RNR plus an enhanced exposure to imexon when the GEM is administered in vivo. This combination is currently being tested in a randomized phase II trial in pancreatic cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Pancreatic Neoplasms/drug therapy , Ribonucleotide Reductases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Synergism , Female , Hexanones/administration & dosage , Humans , Mice , Mice, Inbred BALB C , Mice, SCID , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays , Gemcitabine
13.
Cancer ; 116(15): 3683-91, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20564083

ABSTRACT

BACKGROUND: Imexon (Amplimexon) is an aziridine compound that increases reactive oxygen species, disrupts mitochondrial membranes, and induces apoptosis. Preclinical studies showed activity against melanoma cell lines and models in mice, and synergy with dacarbazine. The authors evaluated standard doses of dacarbazine combined with increasing doses of imexon to determine the maximal tolerated dose (MTD), toxicities, pharmacokinetics, and efficacy. METHODS: Sixty-eight chemotherapy-naive melanoma patients (1 inoperable stage III and 67 stage IV) were treated with dacarbazine (250 mg/m2) and imexon (570-1300 mg/m2), both daily for 5 days every 3 weeks. RESULTS: There were 18 patients in the phase 1, and 50 in the phase 2 component of the study. The MTD of imexon with dacarbazine was 1000 mg/m2. Dose-limiting toxicities were pulmonary edema and hepatorenal failure. At the MTD, therapy was well tolerated. The most common toxicities (any grade) were vomiting, diarrhea, anemia, thrombocytopenia, anorexia, fever, and constipation. Among 68 patients, there were 7 treatment-related serious adverse events. Partial response and stable disease rates were 5.9% and 25% for all subjects and 2% and 30% for the phase 2 patients, respectively. Median progression-free and overall survival of all patients were 2.0 and 11.7 months and 2 and 7.5 months for the phase 2 patients, respectively. Overall survival of the 31 patients with normal lactate dehydrogenase levels was >22.5 months. Pharmacokinetics of both drugs were similar to previous reports. CONCLUSIONS: Imexon plus dacarbazine was well tolerated. The survival data suggest further evaluation in a randomized phase 2 study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dacarbazine/administration & dosage , Hexanones/administration & dosage , Melanoma/drug therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Female , Humans , Male , Maximum Tolerated Dose , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology
14.
Cancer Chemother Pharmacol ; 66(2): 287-94, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19855966

ABSTRACT

PURPOSE: Imexon is an aziridine-derived iminopyrrolidone which has synergy with gemcitabine in pancreatic cancer cell lines. Gemcitabine is a standard therapy for pancreatic cancer. We performed a phase I trial of imexon and gemcitabine to evaluate safety, dose-limiting toxicity (DLT), and maximum tolerated dose (MTD) in patients with advanced pancreatic cancer. METHODS: Patients with untreated locally advanced or metastatic pancreatic adenocarcinoma received therapy in sequential cohorts on regimen A (n = 19; imexon 200 or 280 mg/m(2) intravenously (IV) over 30 min days 1-5, 15-19 and gemcitabine 800 or 1,000 mg/m(2) IV over 30 min on days 1,8,15 every 28 days) or regimen B (n = 86; imexon 280-1,300 mg/m(2) IV over 30-60 min days 1, 8, and 15 and gemcitabine 1,000 mg/m(2) IV over 30 min on days 1, 8, and 15 every 28 days). RESULTS: One hundred five patients received 340 treatment cycles (median 2, range 1-16). PATIENT CHARACTERISTICS: median age 63, 61% male, ECOG PS 0/1 50%/50%, 93% metastatic. DLT was abdominal cramping and pain, often with transient, acute diarrhea. Best response was confirmed partial response (PR) in 11.4%, 8.9% unconfirmed PR, and 48.1% with stable disease. There was a dose proportional increase in imexon AUC across the doses tested with terminal half life 69 min at the MTD and no alteration of gemcitabine pharmacokinetics. CONCLUSIONS: The recommended phase II dose of imexon is 875 mg/m(2) with gemcitabine 1,000 mg/m(2). DLT was acute abdominal pain and cramping. Encouraging antitumor responses support further evaluation of this combination in advanced pancreatic cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Cohort Studies , Cystine/blood , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Female , Half-Life , Hexanones/administration & dosage , Humans , Male , Middle Aged , Pancreatic Neoplasms/pathology , Sulfhydryl Compounds/metabolism , Tomography, X-Ray Computed , Gemcitabine
15.
Int J Pharm ; 340(1-2): 223-9, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17498897

ABSTRACT

The objective of these studies was to investigate the applicability of an online direct inject HPLC method for the preformulation screening of pharmaceutical agents in pressurized metered dose inhalers (MDIs). The technique was initially utilized for the solubility determination of solid solutes. This study explores the extension of the online direct inject method for the evaluation of drug stability in propellant systems as well as for the analysis of MDI vials crimped with metered valves. Through-life content analysis confirmed that a single vial may be repeatedly sampled, thus facilitating the stability evaluation of a single unit over time. The method was successfully used for evaluating the stability of a model drug, as a function of several different formulation configurations, with minimal sample numbers. Additionally, studies determined that after modifications were made to the injection coupler, the technique was also feasible for use with 50 and 100 microL metered valves, however further modifications are necessary for 25 microL valves.


Subject(s)
Aerosol Propellants/chemistry , Beclomethasone/chemistry , Chromatography, High Pressure Liquid , Hexanones/chemistry , Technology, Pharmaceutical/methods , Administration, Inhalation , Aerosol Propellants/administration & dosage , Aerosols , Beclomethasone/administration & dosage , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid/instrumentation , Drug Compounding , Drug Stability , Equipment Design , Ethanol/chemistry , Feasibility Studies , Hexanones/administration & dosage , Metered Dose Inhalers , Pressure , Reproducibility of Results , Solubility , Technology, Pharmaceutical/instrumentation , Temperature , Water/chemistry
16.
Cancer Chemother Pharmacol ; 59(6): 749-57, 2007 May.
Article in English | MEDLINE | ID: mdl-17333195

ABSTRACT

PURPOSE: This study evaluated the cytotoxic effects of imexon (NSC-714597) in tumor cells when combined with a broad panel of chemotherapeutic drugs. METHODS: The sulforhodamine B (SRB) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cytotoxicity assays were used to analyze the degree of growth inhibition for the combination studies in the A375 human malignant melanoma and RPMI 8226 human multiple myeloma cell lines, respectively. Cells were continuously exposed to both drugs at a constant molar ratio for 4-5 days. Combination effects were analyzed using the Median Effect method. Statistical significance was inferred if the 95% confidence interval for the combination interaction (C.I.) values for a particular two-drug combination did not include 1.0 (additivity). Synergy was inferred for C.I. values<1.0 and antagonism for CI values>1.0. RESULTS: Imexon was synergistic when combined with DNA-binding agents (cisplatin, dacarbazine, melphalan) and pyrimidine-based antimetabolites (cytarabine, fluorouracil, gemcitabine) in both cell lines. Antagonistic combinations with imexon included methotrexate and the topoisomerase I (TOPO I) and II (TOPO II) inhibitors irinotecan, doxorubicin, mitoxantrone and etoposide. Docetaxel was synergistic with imexon in both cell lines whereas paclitaxel and fludarabine showed a mixed result. Dexamethasone and the proteasome inhibitor bortezomib showed synergy in myeloma cells and additivity in the melanoma cells. The vinca alkaloid, vinorelbine, and the multi-targeted antifol, pemetrexed, were additive with imexon in both cell lines. DISCUSSION: The consistent synergy seen for imexon and alkylating agents may relate to the sulfhydryl-lowering effect of imexon, which would render cells more sensitive to electrophilic species from the alkylators. The marked synergy noted with pyrimidine-based antimetabolites was unexpected and may relate to the induction of cell cycle arrest in S-phase. The strong antagonism noted for imexon with topoisomerase I and II inhibitors may be due to the effect of imexon at increasing oxidant levels which are known to antagonize the cytotoxic effects of topoisomerase poisons. In contrast, the synergy seen with bortezomib in myeloma cells may be related to an increase in reactive oxygen species (ROS) from both drugs. These results suggest that combinations of imexon with alkylating agents and pyrimidine-based antimetabolites are rational to pursue in therapeutic studies in vivo.


Subject(s)
Hexanones/administration & dosage , Melanoma/drug therapy , Multiple Myeloma/drug therapy , Antineoplastic Combined Chemotherapy Protocols , Drug Screening Assays, Antitumor , Hexanones/therapeutic use , Humans , Tumor Cells, Cultured
17.
Front Biosci ; 11: 300-12, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16146732

ABSTRACT

Redox regulation has been shown to be an important component of malignant cell survival. Tipping the cellular redox balance through pharmacologic regulation in favor of increasing intracellular reactive oxygen species (ROS) and/or depleting protective reducing metabolites (such as glutathione and nicotinamide adenine dinucleotide phosphate) may lead to oxidative stress and resultant induction of apoptosis for the treatment of cancer. We review the biology and importance of ROS with regard to malignant and normal cells. Moreover, we discuss pre-clinical and clinical data regarding novel therapeutic agents that modulate the cellular redox system including buthionine sulfoximine, ascorbic acid, arsenic trioxide, imexon, and motexafin gadolinium as single-agents and in combination. Continued research is needed to better understand the mechanisms and specific apoptotic pathways involved in ROS-induced cell death, as well as, to determine the most rationale and effective combination of redox-active agents.


Subject(s)
Apoptosis , Neoplasms/pathology , Neoplasms/therapy , Oxidative Stress , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenic Trioxide , Arsenicals/administration & dosage , Ascorbic Acid/administration & dosage , Buthionine Sulfoximine/administration & dosage , Cell Death , Dose-Response Relationship, Drug , Glutathione/metabolism , Hexanones/administration & dosage , Humans , Metalloporphyrins/administration & dosage , Models, Biological , Models, Chemical , Oxidation-Reduction , Oxides/administration & dosage , Reactive Oxygen Species
18.
Biochem Biophys Res Commun ; 338(2): 903-9, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16243295

ABSTRACT

To determine the mechanism by which differentiation-inducing factor-1 (DIF-1), a morphogen of Dictyostelium discoideum, inhibits tumor cell proliferation, we examined the effect of DIF-1 on the gene expression of cyclin D1. DIF-1 strongly reduced the expression of cyclin D1 mRNA and correspondingly decreased the amount of beta-catenin in HeLa cells and squamous cell carcinoma cells. DIF-1 activated glycogen synthase kinase-3beta (GSK-3beta) and inhibition of GSK-3beta attenuated the DIF-1-induced beta-catenin degradation, indicating the involvement of GSK-3beta in this effect. Moreover, DIF-1 reduced the activities of T-cell factor (TCF)/lymphoid enhancer factor (LEF) reporter plasmid and a reporter gene driven by the human cyclin D1 promoter. Eliminating the TCF/LEF consensus site from the cyclin D1 promoter diminished the effect of DIF-1. These results suggest that DIF-1 inhibits Wnt/beta-catenin signaling, resulting in the suppression of cyclin D1 promoter activity.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Hexanones/administration & dosage , Cell Line, Tumor , HeLa Cells , Humans
19.
J Toxicol Sci ; 30(3): 195-206, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16141653

ABSTRACT

Predictive biomarkers of testicular toxicity are needed for an efficient development of drugs. The purpose of the present study was to obtain further insight into the toxicity mechanisms of various male reproductive toxicants and to detect genomic biomarkers for rapid screening of testicular toxicity. Four reproductive toxicants, 2,5-hexanedione (Sertoli cells toxicant), ethylene glycol monomethyl ether (EGME; spermatocytes toxicant), cyclophosphamide (spermatogonia toxicant) and sulfasalazine, were orally administered to male rats once. Six hours after the single dosing, gene expression in the testes was monitored by cDNA microarray and real-time RT-PCR and the testes were histopathologically examined. No histopathological abnormality was detected except for slight degeneration of spermatocytes in the EGME-treated testes. cDNA microarray analysis revealed differential gene expression profiles, and it was possible based on the profiles to characterize the action of the compounds in the testes. Interestingly, 3 spermatogenesis-related genes -- heat shock protein 70-2, insulin growth factor binding protein 3 and glutathione S transferase pi -- were affected by all the compounds. The above changes of gene expression were detectable within a short period after the dosing prior to the appearance of obvious pathological changes. These data suggest that cDNA microarray is a useful technique for evaluation of primary testicular toxicity. Furthermore, we propose the above 3 spermatogenesis-related genes as potential biomarkers of testicular toxicity.


Subject(s)
Epididymis/drug effects , Gene Expression/drug effects , Testis/drug effects , Administration, Oral , Animals , Cyclophosphamide/administration & dosage , Cyclophosphamide/toxicity , Epididymis/metabolism , Epididymis/pathology , Ethylene Glycols/administration & dosage , Ethylene Glycols/toxicity , Glutathione S-Transferase pi/genetics , HSP70 Heat-Shock Proteins/genetics , Hexanones/administration & dosage , Hexanones/toxicity , Insulin-Like Growth Factor Binding Protein 3/genetics , Male , Oligonucleotide Array Sequence Analysis/methods , Organ Size/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Spermatocytes/drug effects , Spermatocytes/metabolism , Spermatocytes/pathology , Sulfasalazine/administration & dosage , Sulfasalazine/toxicity , Testis/metabolism , Testis/pathology , Up-Regulation/drug effects , Up-Regulation/genetics
20.
Anticancer Drugs ; 16(7): 727-32, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16027520

ABSTRACT

Imexon is an investigational anti-cancer agent, pharmaceutically formulated as a lyophilized solid for i.v. infusion requiring reconstitution and subsequent dilution in infusion fluid before infusion. Imexon contains a highly reactive aziridine ring in its structure, which limits its stability in aqueous solutions. In the present study, several in vitro studies were conducted to determine the administration parameters for use in the forthcoming phase I clinical trial. The stability of Imexon in the reconstituted solution and infusion solutions was investigated, including its tendency to degrade to its main degradation product BM41.209, and to its hydroxy and chloride adducts. The compatibility of the infusion solution with glass, low-density polyethylene and freeflex polyolefin containers, and its potency to cause vascular irritation and hemolysis upon i.v. infusion were investigated. Imexon was found to be stable for 8 h in the reconstituted product and for 2 h after dilution to infusion solution concentrations in normal saline. The infusion solution was compatible with the freeflex polyolefin container and polyvinylchloride infusion lining, showing no sorption of Imexon during a 15-min infusion duration and no release of the plasticizer diethylhexyl phthalate. Furthermore, Imexon infusion solution showed no indication for vascular irritation or hemolysis upon i.v. infusion, as measured with a static in vitro model with incubation with whole blood. In conclusion, Imexon should be administered using a freeflex polyolefin infusion container within 2 h after preparation and a 15-min infusion duration. The results obtained with an in vitro model show that no vascular irritation or hemolysis is expected upon i.v. infusion.


Subject(s)
Antineoplastic Agents/chemistry , Hexanones/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid , Drug Incompatibility , Drug Packaging , Drug Stability , Freeze Drying , Hemolysis , Hexanones/administration & dosage , Hexanones/adverse effects , Humans , In Vitro Techniques , Infusion Pumps , Infusions, Intravenous
SELECTION OF CITATIONS
SEARCH DETAIL