Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
J Immunol Res ; 2022: 7538649, 2022.
Article in English | MEDLINE | ID: mdl-35211631

ABSTRACT

BACKGROUND: The leading cause of mortality in patients with Marfan syndrome (MFS) is thoracic aortic aneurysm and dissection. Notch signaling is essential for vessel morphogenesis and function. However, the role of Notch signaling in aortic pathology and aortic smooth muscle cell (SMC) differentiation in Marfan syndrome (MFS) is not completely understood. METHODS: RNA-sequencing on ascending aortic tissue from a mouse model of MFS, Fbn1mgR/mgR , and wild-type controls was performed. Notch 3 expression and activation in aortic tissue were confirmed with real-time RT-PCR, immunohistochemistry, and Western blot. Fbn1mgR/mgR and wild-type mice were treated with a γ-secretase inhibitor, DAPT, to block Notch activation. Aortic aneurysms and rupture were evaluated with connective tissue staining, ultrasound, and life table analysis. RESULTS: The murine RNA-sequencing data were validated with mouse and human MFS aortic tissue, demonstrating elevated Notch3 activation in MFS. Data further revealed that upregulation and activation of Notch3 were concomitant with increased expression of SMC contractile markers. Inhibiting Notch3 activation with DAPT attenuated aortic enlargement and improved survival of Fbn1mgR/mgR mice. DAPT treatment reduced elastin fiber fragmentation in the aorta and reversed the differentiation of SMCs. CONCLUSIONS: Our data demonstrated that matrix abnormalities in the aorta of MFS are associated with increased Notch3 activation. Enhanced Notch3 activation in MFS contributed to aortic aneurysm formation in MFS. This might be mediated by inducing a contractile phenotypic change of SMC. Our results suggest that inhibiting Notch3 activation may provide a strategy to prevent and treat aortic aneurysms in MFS.


Subject(s)
Aorta/pathology , Aortic Aneurysm/metabolism , Marfan Syndrome/metabolism , Myocytes, Smooth Muscle/physiology , Receptor, Notch3/metabolism , Animals , Aortic Aneurysm/genetics , Diamines/administration & dosage , Diamines/pharmacology , Disease Models, Animal , Elastin/metabolism , Fibrillin-1/genetics , Fibrillin-1/metabolism , Humans , Marfan Syndrome/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Targeted Therapy , Receptor, Notch3/antagonists & inhibitors , Thiazoles/administration & dosage , Thiazoles/pharmacology
2.
Sci Rep ; 12(1): 1688, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35105922

ABSTRACT

Organophosphorus (OP) compounds that inhibit acetylcholinesterase are a common cause of poisoning worldwide, resulting in several hundred thousand deaths each year. The pathways activated during OP compound poisoning via overstimulation of muscarinic acetylcholine receptors (mAChRs) play a decisive role in toxidrome. The antidotal therapy includes atropine, which is a nonspecific blocker of all mAChR subtypes. Atropine is efficient for mitigating depression in respiratory control centers but does not benefit patients with OP-induced skeletal muscle weakness. By using an ex vivo model of OP-induced muscle weakness, we studied the effects of the M1/M4 mAChR antagonist pirenzepine and the M2/M4 mAChR antagonist methoctramine on the force of mouse diaphragm muscle contraction. It was shown that weakness caused by the application of paraoxon can be significantly prevented by methoctramine (1 µM). However, neither pirenzepine (0.1 µM) nor atropine (1 µM) was able to prevent muscle weakness. Moreover, the application of pirenzepine significantly reduced the positive effect of methoctramine. Thus, balanced modulation of neuromuscular synaptic transmission via M1 and M2 mAChRs contributes to paraoxon-induced muscle weakness. It was shown that methoctramine (10 µmol/kg, i.p.) and atropine (50 µmol/kg, i.p.) were equieffective toward increasing the survival of mice poisoned with a 2xLD50 dose of paraoxon.


Subject(s)
Antidotes/administration & dosage , Atropine/administration & dosage , Cholinesterase Inhibitors/adverse effects , Diamines/administration & dosage , Muscarinic Antagonists/administration & dosage , Muscle Weakness/chemically induced , Muscle Weakness/prevention & control , Paraoxon/adverse effects , Parasympatholytics/administration & dosage , Protective Agents/administration & dosage , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M2/metabolism , Synaptic Transmission/drug effects , Animals , Cholinesterase Inhibitors/administration & dosage , Cholinesterases/metabolism , Diaphragm/drug effects , Disease Models, Animal , Mice , Muscle Contraction/drug effects , Muscle Weakness/metabolism , Paraoxon/administration & dosage , Pirenzepine/administration & dosage , Receptor, Muscarinic M1/antagonists & inhibitors , Receptor, Muscarinic M2/antagonists & inhibitors , Signal Transduction/drug effects , Treatment Outcome
3.
Biomolecules ; 11(7)2021 07 19.
Article in English | MEDLINE | ID: mdl-34356678

ABSTRACT

Allograft kidney transplantation, which triggers host cellular- and antibody-mediated rejection of the kidney, is a major contributor to kidney damage during transplant. Here, we asked whether PrC-210 would suppress damage seen in allograft kidney transplant. Brown Norway (BN) rat kidneys were perfused in situ (UW Solution) with or without added 30 mM PrC-210, and then immediately transplanted into Lewis (LEW) rats. 20 h later, the transplanted BN kidneys and LEW rat plasma were analyzed. Kidney histology, and kidney/serum levels of several inflammation-associated cytokines, were measured to assess mismatch-related kidney pathology, and PrC-210 protective efficacy. Twenty hours after the allograft transplants: (i) significant histologic kidney tubule damage and mononuclear inflammatory cell infiltration were seen in allograft kidneys; (ii) kidney function metrics (creatinine and BUN) were significantly elevated; (iii) significant changes in key cytokines, i.e., TIMP-1, TNF-alpha and MIP-3A/CCL20, and kidney activated caspase levels were seen. In PrC-210-treated kidneys and recipient rats, (i) kidney histologic damage (Banff Scores) and mononuclear infiltration were reduced to untreated background levels; (ii) creatinine and BUN were significantly reduced; and (iii) activated caspase and cytokine changes were significantly reduced, some to background. In conclusion, the results suggest that PrC-210 could provide broadly applicable organ protection for many allograft transplantation conditions; it could protect transplanted kidneys during and after all stages of the transplantation process-from organ donation, through transportation, re-implantation and the post-operative inflammation-to minimize acute and chronic rejection.


Subject(s)
Diamines/pharmacology , Inflammation/drug therapy , Kidney Transplantation/adverse effects , Kidney/drug effects , Sulfhydryl Compounds/pharmacology , Adenosine , Allografts , Allopurinol , Animals , Caspases/metabolism , Creatinine/blood , Cytokines/metabolism , Diamines/administration & dosage , Free Radical Scavengers/pharmacology , Glutathione , Inflammation/pathology , Insulin , Kidney/pathology , Kidney Transplantation/methods , Male , Mitochondria/drug effects , Organ Preservation Solutions , Raffinose , Rats, Inbred BN , Rats, Inbred Lew , Sulfhydryl Compounds/administration & dosage
4.
Nat Commun ; 11(1): 2397, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32409697

ABSTRACT

Obesity is a health problem affecting more than 40% of US adults and 13% of the global population. Anti-obesity treatments including diet, exercise, surgery and pharmacotherapies have so far failed to reverse obesity incidence. Herein, we target obesity with a pharmacotherapeutic approach that decreases caloric efficiency by mitochondrial uncoupling. We show that a recently identified mitochondrial uncoupler BAM15 is orally bioavailable, increases nutrient oxidation, and decreases body fat mass without altering food intake, lean body mass, body temperature, or biochemical and haematological markers of toxicity. BAM15 decreases hepatic fat, decreases inflammatory lipids, and has strong antioxidant effects. Hyperinsulinemic-euglycemic clamp studies show that BAM15 improves insulin sensitivity in multiple tissue types. Collectively, these data demonstrate that pharmacologic mitochondrial uncoupling with BAM15 has powerful anti-obesity and insulin sensitizing effects without compromising lean mass or affecting food intake.


Subject(s)
Diamines/administration & dosage , Insulin Resistance , Mitochondria/drug effects , Obesity/drug therapy , Oxadiazoles/administration & dosage , Pyrazines/administration & dosage , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Administration, Oral , Animals , Blood Glucose/analysis , Body Temperature/drug effects , Body Weight/drug effects , Diamines/adverse effects , Diet, Western/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Glucose Clamp Technique , Humans , Liver/drug effects , Liver/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Obesity/blood , Obesity/etiology , Obesity/metabolism , Oxadiazoles/adverse effects , Oxidative Stress/drug effects , Pyrazines/adverse effects
5.
Cancer Chemother Pharmacol ; 85(4): 673-683, 2020 04.
Article in English | MEDLINE | ID: mdl-32062691

ABSTRACT

PURPOSE: This study aimed to determine the safety, tolerability, and recommended phase II doses of trametinib plus uprosertib (GSK2141795) in patients with solid tumors likely to be sensitive to MEK and/or AKT inhibition. METHODS: This was a phase I, open-label, dose-escalation, and dose-expansion study in patients with triple-negative breast cancer or BRAF-wild type advanced melanoma. The primary outcome of the expansion study was investigator-assessed response. Among 126 enrolled patients, 63 received continuous oral daily dosing of trametinib and uprosertib, 29 received various alternative dosing schedules, and 34 were enrolled into expansion cohorts. Doses tested in the expansion cohort were trametinib 1.5 mg once daily (QD) + uprosertib 50 mg QD. RESULTS: Adverse events (AEs) were consistent with those reported in monotherapy studies but occurred at lower doses and with greater severity. Diarrhea was the most common dose-limiting toxicity; diarrhea and rash were particularly difficult to tolerate. Overall, 59% and 6% of patients reported AEs with a maximum severity of grade 3 and 4, respectively. Poor tolerability prevented adequate delivery of uprosertib with trametinib at a concentration predicted to have clinical activity. The study was terminated early based on futility in the continuous-dosing expansion cohorts and a lack of pharmacological or therapeutic advantage with intermittent dosing. The objective response rate was < 5% (1 complete response, 5 partial responses). CONCLUSIONS: Continuous and intermittent dosing of trametinib in combination with uprosertib was not tolerated, and minimal clinical activity was observed in all schedules tested.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Melanoma/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Cohort Studies , Diamines/administration & dosage , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Melanoma/pathology , Middle Aged , Prognosis , Pyrazoles/administration & dosage , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Tissue Distribution , Triple Negative Breast Neoplasms/pathology , Young Adult
6.
Gynecol Oncol ; 155(3): 420-428, 2019 12.
Article in English | MEDLINE | ID: mdl-31623857

ABSTRACT

OBJECTIVE: We sought to determine safety and efficacy of the AKT inhibitor, GSK2141795, combined with the MEK inhibitor, trametinib, in endometrial cancer. METHODS: Patients with measurable recurrent endometrial cancer were eligible. One to two prior cytotoxic regimens were allowed; prior use of a MEK or PI3K pathway inhibitor was excluded. Initial trial design consisted of a KRAS mutation stratified randomized phase II with a safety lead-in evaluating the combination. For the safety lead in, the previously recommended phase 2 dose (RP2D; trametinib 1.5 mg, GSK2141795 50 mg) was chosen for Dose Level 1 (DL1). RESULTS: Of 26 enrolled patients, 14 were treated on DL1 and 12 were treated on DL-1 (trametinib 1.5 mg, GSK2141795 25 mg). Most common histologies were endometrioid (58%) and serous (27%). Four of 25 (16%) patients were KRAS mutant. Dose limiting toxicities (DLTs) were assessed during cycle 1. DL1 had 8 DLTs (hypertension (n = 2), mucositis (2), rash (2), dehydration, stroke/acute kidney injury). DL1 was deemed non-tolerable so DL-1 was explored. DL-1 had no DLTs. Sixty-five percent of patients had ≥ grade 3 toxicity. There were no responses in DL1 (0%, 90%CI 0-15%) and 1 response in DL-1 (8.3%, 90%CI 0.4-33.9%). Proportion PFS at 6 months for DL1 is 14%, and 25% for DL-1. CONCLUSION: The combination of trametinib and GSK2141795 had high levels of toxicity in endometrial cancer at the previously RP2D but was tolerable at a reduced dose. Due to insufficient preliminary efficacy at a tolerable dose, the Phase II study was not initiated.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Endometrial Neoplasms/drug therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Diamines/administration & dosage , Diamines/adverse effects , Endometrial Neoplasms/enzymology , Female , Humans , Middle Aged , Progression-Free Survival , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyridones/administration & dosage , Pyridones/adverse effects , Pyrimidinones/administration & dosage , Pyrimidinones/adverse effects
7.
Int J Neurosci ; 129(12): 1203-1212, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31393204

ABSTRACT

Purpose of the study: Kaempferol (KM) is a flavonoid found in plant-derived foods and medicinal plants. Recently, it is well established that KM plays a protective role to develop Alzheimer's disease. The current study aimed at evaluating the effect of intracerebroventricular micro-injection of KM on memory retention of passive avoidance learning (MRPAM) and identifying the potentially related cholinergic mechanisms (ChMs) in rats. Materials and methods: In the current study, male Wistar rats randomly divided into control, vehicle and KM (10, 20 and 40 µg/rat) groups. Moreover, MRPAM was evaluated by shuttle box test. The role of ChM was studied using non-selective and selective acetylcholine antagonists (scopolamine [SCN], 4-DAMP and methoctramine [MN], respectively) as well as pirenzepine (PZ) in combination with KM. Results: The employment of KM (40 µg/rat) improved the SCN-induced memory impairment in MRPAM. Co-treatment with KM (40 µg/rat) plus 4-DAMP significantly increased the step-through latency (STL, P < 0.05; 167 ± 28 s) and decreased the total dark chamber (TDC, P < 0.05; 121 ± 31 s) compared with those of the 4-DAMP group (STL: 75 ± 13 s; TDC: 178 ± 46 s). Co-treatment with KM (40 µg/rat) plus PZ attenuated STL, and also increased TDC (P < 0.01; 220 ± 28 s) compared with those of the PZ group. Co-treatment with KM (10 and 20 µg/rat) and MN increased STL (P < 0.05), and deceased TDC compared with those of the MN group (P < 0.01). Conclusions: Totally, the results of the present study showed that cholinergic system may be involved in improving effect of KM on SCN-induced memory impairment.


Subject(s)
Acetylcholine/physiology , Avoidance Learning/drug effects , Cholinergic Antagonists/administration & dosage , Kaempferols/administration & dosage , Memory/drug effects , Muscarinic Antagonists/administration & dosage , Animals , Avoidance Learning/physiology , Diamines/administration & dosage , Injections, Intraventricular , Male , Memory/physiology , Microinjections , Piperidines/administration & dosage , Pirenzepine/administration & dosage , Rats, Wistar , Scopolamine/administration & dosage
8.
Pharmacol Res Perspect ; 7(4): e00500, 2019 08.
Article in English | MEDLINE | ID: mdl-31338199

ABSTRACT

Managing myocardial infarction (MI) to reduce cardiac cell death relies primarily on timely reperfusion of the affected coronary site, but reperfusion itself induces cell death through a toxic, ROS-mediated process. In this study, we determined whether the PrC-210 aminothiol ROS-scavenger could prevent ROS-induced damage in post-MI hearts. In a series of both in vitro and in vivo experiments, we show that: (a) in vitro, PrC-210 was the most potent and effective ROS-scavenger when functionally compared to eight of the most commonly studied antioxidants in the MI literature, (b) in vitro PrC-210 ROS-scavenging efficacy was both immediate (seconds) and long-lasting (hours), which would make it effective in both (1) real-time (seconds), as post-MI or cardiac surgery hearts are reperfused with PrC-210-containing blood, and (2) long-term (hours), as hearts are bathed with systemic PrC-210 after MI or surgery, (c) systemic PrC-210 caused a significant 36% reduction of mouse cardiac muscle death following a 45-minute cardiac IR insult; in a striking coincidence, the PrC-210 36% reduction in cardiac muscle death equals the 36% of the MI-induced cardiac cell death estimated 6 years ago by Ovize and colleagues to result from "reperfusion injury," (d) hearts in PrC-210-treated mice performed better than controls after heart attacks when functionally analyzed using echocardiography, and (e) the PrC-210 ROS-scavenging mechanism of action was corroborated by its ability to prevent >85% of the direct, H2O2-induced killing of neonate cardiomyocytes in cell culture. PrC-210 does not cause the nausea, emesis, nor hypotension that preclude clinical use of the WR-1065/amifostine aminothiol. PrC-210 is a highly effective ROS-scavenger that significantly reduces IR injury-associated cardiac cell death.


Subject(s)
Diamines/administration & dosage , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/administration & dosage , Animals , Cell Death/drug effects , Cells, Cultured , Diamines/pharmacology , Disease Models, Animal , Hydrogen Peroxide/adverse effects , Male , Mice , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Sulfhydryl Compounds/pharmacology
9.
Clin Lymphoma Myeloma Leuk ; 19(7): 431-440.e13, 2019 07.
Article in English | MEDLINE | ID: mdl-31056348

ABSTRACT

BACKGROUND: With proven single-agent activity and favorable toxicity profile of MEK-1/2 inhibition in advanced leukemia, investigation into combination strategies to overcome proposed resistance pathways is warranted. Resistance to MEK inhibition is secondary to upstream hyperactivation of RAS/RAF or activation of the PI3K/PTEN/AKT/mTOR pathway. This phase II multi-institution Cancer Therapy Evaluation Program-sponsored study was conducted to determine efficacy and safety of the combination of the ATP-competitive pan-AKT inhibitor GSK2141795, targeting the PI3K/AKT pathway, and the MEK inhibitor trametinib in RAS-mutated relapsed/refractory acute myeloid leukemia (AML). PATIENTS AND METHODS: The primary objective was to determine the proportion of patients achieving a complete remission. Secondary objectives included assessment of toxicity profile and biologic effects of this combination. Twenty-three patients with RAS-mutated AML received the combination. Two dose levels were explored (dose level 1: 2 mg trametinib, 25 mg GSK2141795 and dose level 2: 1.5 mg trametinib, 50 mg GSK2141795). RESULTS: Dose level 1 was identified as the recommended phase II dose. No complete remissions were identified in either cohort. Minor responses were recognized in 5 (22%) patients. The most common drug-related toxicities included rash and diarrhea, with dose-limiting toxicities of mucositis and colitis. Longitudinal correlative assessment of the modulation of MEK and AKT pathways using reverse phase protein array and phospho-flow analysis revealed significant and near significant down-modulation of pERK and pS6, respectively. Combined MEK and AKT inhibition had no clinical activity in patients with RAS-mutated AML. CONCLUSION: Further investigation is required to explore the discrepancy between the activity of this combination on leukemia cells and the lack of clinical efficacy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Genes, ras , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Mutation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Diamines/administration & dosage , Female , Humans , Leukemia, Myeloid, Acute/diagnosis , Male , Middle Aged , Pyrazoles/administration & dosage , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Treatment Outcome , Young Adult
10.
Gynecol Oncol ; 154(1): 95-101, 2019 07.
Article in English | MEDLINE | ID: mdl-31118140

ABSTRACT

BACKGROUND: Improved treatment for advanced cervical cancer is needed; currently, treatment options include combined chemotherapy and bevacizumab or pembrolizumab monotherapy for PD-L1 positive disease. PIK3CA and KRAS mutations have been reported in cervical cancers; this study therefore tested dual inhibition of PI3K and RAS signaling by combining the MEK inhibitor trametinib and the AKT inhibitor GSK2141795 in recurrent cervical cancer. METHODS: This was an investigator-initiated phase II study combining trametinib and GSK2141795 in patients with recurrent cervical cancer. Primary endpoint was best tumor response; secondary endpoints included progression free survival, overall survival, and safety assessment. Translational objectives included characterization of molecular alterations in PI3K and RAS signaling pathway genes. RESULTS: Planned accrual was 35 patients; 14 patients were enrolled and received at least one dose of study drug before the study was terminated due to discontinuation of GSK2141795 development. There were no confirmed responses; 1 patient had an unconfirmed PR, 8 had stable disease, 3 had progression as best response, and 2 were unevaluable. Toxicities were mostly grade 1 and 2, although 57% of patients experienced grade 3/4 adverse events and 50% patients required a dose reduction. CONCLUSIONS: The combination of trametinib and GSK2141795 was feasible but required dose holds and modifications for adverse events; however, anti-cancer activity was minimal, even in patients with PI3K or RAS pathway alterations. Although the study was terminated early after GSK2141795 development was halted, the findings in these 14 patients do not support further development of this combination in cervical cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Uterine Cervical Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Diamines/administration & dosage , Diamines/adverse effects , Female , Humans , Kaplan-Meier Estimate , MAP Kinase Kinase Kinases/antagonists & inhibitors , Middle Aged , Neoplasm Recurrence, Local/enzymology , Phosphoinositide-3 Kinase Inhibitors , Progression-Free Survival , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyridones/administration & dosage , Pyridones/adverse effects , Pyrimidinones/administration & dosage , Pyrimidinones/adverse effects , Signal Transduction/drug effects , Uterine Cervical Neoplasms/enzymology
11.
Drug Deliv ; 26(1): 470-480, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30957572

ABSTRACT

Although a variety of drug delivery strategies have been designed for enhancing the treatment of Triple negative breast cancer (TNBC), combating with TNBCs is still dramatically challenged by the selection of appropriate therapeutic targets and insufficient tumor accumulation or inner penetration of chemotherapeutics. To address these issues, the classical EGFR-inhibitor, erlotinib (EB), was selected as the model drug here and PLA-based nano-platform (NP-EB) was prepared for tumor site drug delivery. Given the significant role of Notch-EGFR interplay in raising severe resistance to EGFR inhibition of EB, gamma secretase inhibitor (GSI)-DAPT was further entrapped into the core of nanoparticles to inhibit the activation of Notch signaling (NP-EB/DART). For achieving the goal of tumor targeting drug delivery, we developed a new peptide CF and decorating it on the surface of EB/DART-dual loaded nanoparticles (CF-NP-EB/DART). Such CF peptide was designed by conjugating two separated peptide CREKA, tumor-homing peptide, and F3, cell penetrating peptide, to together via a pH-sensitive hydrazone bond. By this way, the tumor unspecific property of F3 was sealed and significantly reduced the site effects. However, after the nanoparticles reach the tumor site, the pH-sensitive linkage can be broken down by the unique acidic environment of tumor, and subsequently discovered the F3 peptide to penetrate into tumor cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell-Penetrating Peptides/chemistry , Diamines/administration & dosage , Drug Carriers/chemistry , Erlotinib Hydrochloride/administration & dosage , Nanoparticles/chemistry , Oligopeptides/chemistry , Thiazoles/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Diamines/therapeutic use , Drug Liberation , Erlotinib Hydrochloride/therapeutic use , Female , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, Notch/antagonists & inhibitors , Thiazoles/therapeutic use , Xenograft Model Antitumor Assays
12.
Invest New Drugs ; 36(6): 1016-1025, 2018 12.
Article in English | MEDLINE | ID: mdl-29611022

ABSTRACT

Background We sought to determine the recommended phase II dose (RP2D) and schedule of GSK2141795, an oral pan-AKT kinase inhibitor. Patients and Methods Patients with solid tumors were enrolled in the dose-escalation phase. Pharmacokinetic (PK) analysis after a single dose (Cycle 0) informed dose escalation using accelerated dose titration. Once one grade 2 toxicity or dose-limiting toxicity was observed in Cycle 1, the accelerated dose titration was terminated and a 3 + 3 dose escalation was started. Continuous daily dosing was evaluated along with two intermittent regimens (7 days on/7 days off and 3 times per week). In the expansion phase at RP2D, patients with endometrial or prostate cancer, as well as those with select tumor types with a PIK3CA mutation, AKT mutation or PTEN loss, were enrolled. Patients were evaluated for adverse events (AEs), PK parameters, blood glucose and insulin levels, and tumor response. Results The RP2D of GSK2141795 for once-daily dosing is 75 mg. The most common (>10%) treatment-related AEs included diarrhea, fatigue, vomiting, and decreased appetite. Most AEs were low grade. The frequency of hyperglycemia increased with dose; however, at the RP2D, grade 3 hyperglycemia was only reported in 4% of patients and no grade 4 events were observed. PK characteristics were favorable, with a prolonged half-life and low peak-to-trough ratio. There were two partial responses at the RP2D in patients with either a PIK3CA mutation or PTEN loss. Conclusion GSK2141795 was safe and well-tolerated, with clinical activity seen as monotherapy at the RP2D of 75 mg daily. NCT00920257.


Subject(s)
Diamines/pharmacokinetics , Diamines/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Administration, Oral , Adult , Aged , Aged, 80 and over , Cohort Studies , Diamines/administration & dosage , Diamines/adverse effects , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Mutation/genetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/administration & dosage , Pyrazoles/adverse effects
13.
Biochem Biophys Res Commun ; 495(3): 2289-2295, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29284118

ABSTRACT

Nuclear factor-κB (NF-κB) proteins are transcription factors that play key roles in regulating most immune responses and cell death. Constitutively active NF-κB has been shown to exhibit chemoresistance by inducing anti-apoptosis in tumor cells. Multiple myeloma is known as a constitutive NF-κB activating disease, and the proteasome inhibitor bortezomib is used to treat multiple myeloma and mantle cell lymphoma. We demonstrate here that DANFIN (N,N'-bis-(2,4-dimethyl-phenyl)-ethane-1,2-diamine) functions as an inhibitor of the p65 family proteins and induces chemosensitization to bortezomib in multiple myeloma. DANFIN was found to be an inhibitor of interactions between p65 and IκBα without the inhibition of the DNA binding activity of the p65 protein. In addition, DANFIN affected the IκBα binding region in Rel Homology Domain (RHD) and suppressed the nuclear translocalization of the p65 protein in cells. Furthermore, in multiple myeloma cells, DANFIN suppressed the expression level of NF-κB target genes and induced apoptosis. The combination therapy of DANFIN with bortezomib dramatically enhanced the apoptosis of multiple myeloma cells and indicated a remarkable anti-tumor effect in a multiple-myeloma xenograft mouse model.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Bortezomib/administration & dosage , Diamines/administration & dosage , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , NF-kappa B/metabolism , Animals , Cell Line, Tumor , Drug Synergism , Female , Mice , Mice, Inbred BALB C , Mice, Nude , Multiple Myeloma/pathology , Transcription Factors/metabolism , Treatment Outcome
14.
Swiss Dent J ; 127(10): 857-864, 2017.
Article in English | MEDLINE | ID: mdl-29199414

ABSTRACT

This study analyzed the anti-erosive effect of a self-assembling peptide fibre gel. One hundred and twelve bovine enamel samples were ground flat and subjected to a three times de- and re-mineralization cycle: erosion (5 min, HCl, pH 2.6) alternated with storage in artificial saliva under agitation. Then, samples were covered with different anti-erosive compounds (2 min): Duraphat toothpaste (DT), Elmex Erosion Protection Toothpaste (EET) or Elmex Gelée (EG) ­ all mixed with saliva (1:3) ­, Elmex Erosion Protection Mouthwash (EEM), Curodont Protect (CP; self-assembling peptide gel) or MI Paste Plus (MIP). Untreated, water stored samples served as control. In experiment 1, half of the samples of each group were continuously superfused with HCl (pH 2.6, 60 µl/min, 8 min). In experiment 2, the second half of samples were subjected to eight cycles, each consisting of application of the respective anti-erosive compound followed by an erosion (60 s, HCl, pH 2.6), followed by remineralization in artificial saliva (45 min). Enamel loss was profilometrically determined. In experiment 1, EEM and EET performed significantly better compared to all other compounds. Substance loss of all other compounds did not differ significantly from control. In experiment 2, significantly better performance was achieved by EEM and EET. EG showed significantly lower protection than the control. All other applied compounds yielded no significant difference compared to control. Under the chosen conditions, the self-assembling peptide-containing compound showed no anti-erosive effect.


Subject(s)
Cariostatic Agents/administration & dosage , Diamines/administration & dosage , Fluorides, Topical/administration & dosage , Fluorides/administration & dosage , Peptides/administration & dosage , Sodium Fluoride/administration & dosage , Tooth Erosion/therapy , Tooth Remineralization , Animals , Cattle , In Vitro Techniques , Mouthwashes/administration & dosage , Toothpastes/administration & dosage
15.
Anim Reprod Sci ; 181: 69-78, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28400072

ABSTRACT

The Notch signaling pathway regulates cell proliferation, differentiation and apoptosis involved in development of the organs and tissues such as nervous system, cartilage, lungs, kidneys and prostate as well as the ovarian follicles. This study aimed to investigate the mRNA expression and localization of NOTCH2, as the key factor in Notch signaling pathway. This was determined by PCR, real-time PCR and immunohistochemistry. Additionally, the effects of inhibiting Notch signaling pathway with different concentrations (5µM, 10µM and 20µM) of N-[N-(3, 5-Difuorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT), an inhibitor of Notch signaling pathway, on ovine granulosa cells was determined in vitro by detecting estradiol production using enzyme linked immunosorbent assay and expressions of the genes related to the cell cycle and apoptosis using real-time polymerase chain reaction (PCR). NOTCH2, the key member of Notch signaling pathway, was found in ovine follicles, and the expression of NOTCH2 mRNA was highest in the theca cells of the follicles in medium sizes (3-5mm in diameter) and granulosa cells of the follicles in large sizes (>5mm in diameter). Immunohistochemical results demonstrated that NOTCH2 protein was expressed in granulosa cells of preantral follicles, in both granulosa cells and theca cells of antral follicles. Compared with DAPT-treated groups, the control group had a higher number of granulosa cells (P<0.05) and a higher estradiol production (P<0.05). Compared with the control group, the mRNA abundances of HES1, MYC, BAX, BCL2 and CYP19A1 in DAPT-treated groups was lower (P<0.05), respectively; whereas, the expression of CCND2, CDKN1A and TP53 mRNA showed no remarkable difference compared with control group. Collectively, Notch signaling pathway could be involved in the ovine follicular development by regulating the growth and estradiol production of granulosa cells.


Subject(s)
Gene Expression Regulation/physiology , Granulosa Cells/physiology , Receptor, Notch2/metabolism , Sheep/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Diamines/administration & dosage , Diamines/pharmacology , Dose-Response Relationship, Drug , Female , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction/veterinary , Receptor, Notch2/genetics , Theca Cells/physiology , Thiazoles/administration & dosage , Thiazoles/pharmacology
16.
Dalton Trans ; 45(33): 13052-60, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27430044

ABSTRACT

Platinum-based antineoplastic drugs are among the first-line chemotherapeutic agents against a variety of solid tumors, but toxic side-effects and drug resistance issues limit their clinical optimization. Novel strategies and platforms to conquer cisplatin resistance are highly desired. Herein, we assembled a multimodal nanoplatform utilizing 808 nm-excited and biocompatible core-shell-shell upconversion nanoparticles (UCNPs) [NaGdF4:Yb/Nd@NaGdF4:Yb/Er@NaGdF4] that were covalently loaded with not only photosensitizers (PSs), but also Pt(iv) prodrugs, which were rose bengal (RB) and c,c,t-[Pt(NH3)2Cl2(OCOCH2CH2NH2)2], respectively. The UCNPs had the capability to convert near infrared (NIR) light to visible light, which was further utilized by RB to generate singlet oxygen. At the same time, the nanoplatform delivered the Pt(iv) prodrug into cancer cells. Thus, this upconversion nanoplatform was able to carry out combined and simultaneous photodynamic therapy (PDT) and Pt chemotherapy. The nanoplatform was well characterized and the energy transfer efficiency was confirmed. Compared with free cisplatin or UCNPs loaded with RB only, our nanoplatform showed significantly improved cytotoxicity upon 808 nm irradiation in both cisplatin-sensitive and -resistant human ovarian cancer cells. A mechanistic study showed that the nanoparticles efficiently delivered the Pt(iv) prodrug into cancer cells, resulting in Pt-DNA damage, and that the nanoplatform generated cellular singlet oxygen to kill cancer cells. We, therefore, provide a comprehensive strategy to use UCNPs for combined Pt chemotherapy and PDT against cisplatin resistance, and our nanoplatform can also be used as a theranostic tool due to its NIR bioimaging capacity.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm/drug effects , Nanoparticles/administration & dosage , Photochemotherapy , Platinum/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Diamines/administration & dosage , Diamines/chemistry , Diamines/pharmacology , Humans , Light , Nanoparticles/chemistry , Platinum/chemistry , Platinum/pharmacology , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/pharmacology , Reactive Oxygen Species/metabolism , Rose Bengal/administration & dosage , Rose Bengal/chemistry , Rose Bengal/pharmacology
17.
Int J Oncol ; 49(1): 422-30, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27176495

ABSTRACT

We investigated the effect of valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, and the mechanism of VPA-induced growth inhibition on three cervical cancer cell lines with different molecular and genetic background. We found that VPA induced proliferation suppression, cell apoptosis and cell cycle arrest in all tested cell lines, with an increase of Notch1 active form ICN1 as a tumor suppressor and its target gene HES1. Noteworthy, blocking of Notch signaling with DAPT resulted in growth inhibition in ICN1-overexpressing CaSki and HT-3 cells. Thus, endogenous Notch signaling may be necessary for survival of ICN1-overexpressing cervical cancer cell lines. Furthermore, G1 phase arrest was induced in HeLa and CaSki cells by VPA while G2 phase arrest was induced in HT-3 cells, suggesting different mechanism in this cycle arrest. We also found VPA suppressed oncogene E6 in a Notch-independent manner, and induced significant apoptosis in E6-overexpressing HPV positive CaSki cells. Cell morphological change was also observed in HeLa and HT-3 cell lines after VPA treatment with an upregulation of EMT transcription factor Snail1. Notch signaling inhibitor DAPT partly reversed VPA-induced Snail1 upregulation in HeLa cells. This discovery supports that VPA may induce EMT at least partly via Notch activation.


Subject(s)
Oncogene Proteins, Viral/biosynthesis , Receptor, Notch1/biosynthesis , Repressor Proteins/biosynthesis , Uterine Cervical Neoplasms/drug therapy , Valproic Acid/administration & dosage , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Diamines/administration & dosage , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Viral/drug effects , HeLa Cells/drug effects , Human papillomavirus 16/genetics , Human papillomavirus 16/pathogenicity , Humans , Oncogene Proteins, Viral/genetics , Receptor, Notch1/genetics , Repressor Proteins/genetics , Snail Family Transcription Factors/biosynthesis , Thiazoles/administration & dosage , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology
18.
Oncogene ; 35(40): 5304-5316, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27041563

ABSTRACT

Metastasis is a critical factor contributing to poor prognosis in cancer, but the underlying mechanisms of metastasis are still poorly understood. We established a highly metastatic cell subline (HOC313-LM) derived from an oral squamous cell carcinoma cell line (HOC313) for uncovering the mechanisms of metastasis, and identified deoxyhypusine synthase (DHPS) as a metastasis-associated gene within the specific amplification at 19p13.2-p13.13 in HOC313-LM. DHPS-mediated hypusine-modification of eukaryotic translation factor 5A facilitated the translation of RhoA, resulting in the activation of the RhoA signaling pathway and leading to not only increased cell motility, invasion and metastasis of cancer cells in vitro, but also increased tumor growth in vivo. Moreover, the use of N1-Guanyl-1,7-diaminoheptane, a DHPS inhibitor, resulted in a significant decrease in tumor formation in vivo. In patients with esophageal squamous cell carcinoma (ESCC), overexpression of DHPS in ESCC tumors was significantly associated with worse recurrence-free survival, and correlated with distant metastasis. The elucidation of these molecular mechanisms within the hypusine cascade suggests opportunities for novel therapeutic targets in SCC.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Esophageal Neoplasms/drug therapy , Lysine/analogs & derivatives , Oxidoreductases Acting on CH-NH Group Donors/genetics , rhoA GTP-Binding Protein/genetics , Adult , Aged , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Diamines/administration & dosage , Disease Progression , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lysine/biosynthesis , Lysine/genetics , Male , Mice , Middle Aged , Neoplasm Metastasis , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Signal Transduction/drug effects
19.
J Nucl Med ; 56(12): 1828-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26429956

ABSTRACT

UNLABELLED: AKT (a serine/threonine-specific protein kinase) regulates many cellular processes contributing to cytotoxic drug resistance. This study's primary objective examined the relationship between GSK2141795, an oral, pan-AKT inhibitor, and (18)F-FDG PET markers of glucose metabolism in tumor tissue to determine whether (18)F-FDG PET could be used to guide personalized dosing of GSK2141795. Biomarker analysis of biopsies was also undertaken. METHODS: Twelve patients were enrolled in 3 cohorts; all underwent dynamic (18)F-FDG PET scans and serial pharmacokinetic sampling at baseline, week 2, and week 4 with tumor biopsies before treatment and at week 4. Response was evaluated by RECIST v1.1 and Gynecologic Cancer Intergroup criteria. Biopsy samples were analyzed for mutations and protein expression. RESULTS: GSK2141795 did not significantly influence blood glucose levels. No dose-response relationship was observed between GSK2141795 pharmacokinetics and (18)F-FDG PET pharmacodynamic measures; however, an exposure-response relationship was seen between maximum drug concentrations and maximal decrease in (18)F-FDG uptake in the best-responding tumor. This relationship also held for pharmacokinetic parameters of exposure and 1,5-anhydroglucitol (a systemic measure of glucose metabolism). Phospho-AKT upregulation at week 4 in biopsies confirmed AKT inhibition by GSK2141795. Single-agent activity was observed with a clinical benefit rate of 27% (3/11) and 30% (3/10) CA125 response in the study's platinum-resistant ovarian patients. AKT pathway activation by PIK3CA/PIK3R1 mutation did not correlate with clinical activity, whereas RAS/RAF pathway mutations did segregate with resistance to AKT inhibition. CONCLUSION: GSK2141795 demonstrated an exposure-response relationship with decreased (18)F-FDG uptake and is active and tolerable. This study's design integrating (18)F-FDG PET, pharmacokinetics, and biomarker analyses demonstrates the potential for clinical development for personalized treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Diamines/administration & dosage , Diamines/therapeutic use , Fluorodeoxyglucose F18/pharmacokinetics , Genital Neoplasms, Female/diagnostic imaging , Genital Neoplasms, Female/drug therapy , Oncogene Protein v-akt/antagonists & inhibitors , Positron-Emission Tomography/methods , Pyrazoles/administration & dosage , Pyrazoles/therapeutic use , Radiopharmaceuticals/pharmacokinetics , Antineoplastic Agents/adverse effects , Biomarkers , Biopsy , Blood Glucose/metabolism , Deoxyglucose , Diamines/adverse effects , Drug Interactions , Drug Resistance, Neoplasm/genetics , Female , Humans , Oncogene Protein v-akt/genetics , Pyrazoles/adverse effects , Treatment Outcome
20.
Implant Dent ; 24(6): 675-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26134690

ABSTRACT

OBJECTIVES: Acidic pH and high fluoride (F(-)) concentration impair the corrosion resistance of titanium (Ti). Caries-preventive products contain high amounts of F(-) and are applied at low pH. The purpose of this study was to evaluate whether fluoride applied in different forms has different short-, mid-, and long-term effects on the growth of the bacteria Streptococcus mutans. MATERIALS AND METHODS: Ti discs with polished surface were treated with a rinse containing 0.025% olaflur, a gel containing 1.25% olaflur, or a 1% aqueous solution of NaF (pH 4.5), and they were incubated with S mutans for 21 days. Control discs did not get prophylactic treatment. Protein assay analysis was performed at regular intervals to estimate the amount of S mutans. Scanning electron microscopic (SEM) images were also taken. RESULTS: Bacterial protein quantity became significantly different only by the 21st day. Fluoride in rinse and gel proved to be superior to NaF in aqueous solution or no treatment (P < 0.01 and P < 0.05, respectively). However, the discs treated with fluoride in gel showed signs of corrosion in SEM images. CONCLUSION: The results suggest that the use of fluoride-containing mouthwashes might be the best and safest oral hygienic choice for patients with oral implants. Furthermore, olaflur seems to be superior to NaF for long-term use at low pH.


Subject(s)
Antibiotic Prophylaxis/methods , Fluorides/therapeutic use , Streptococcus mutans/drug effects , Titanium , Diamines/administration & dosage , Diamines/therapeutic use , Fluorides/administration & dosage , Microscopy, Electron, Scanning , Sodium Fluoride/administration & dosage , Sodium Fluoride/therapeutic use , Streptococcus mutans/growth & development , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...