Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 578
Filter
1.
Biomed Pharmacother ; 174: 116492, 2024 May.
Article in English | MEDLINE | ID: mdl-38537579

ABSTRACT

Targeting epigenetic mechanisms has emerged as a potential therapeutic approach for the treatment of kidney diseases. Specifically, inhibiting the bromodomain and extra-terminal (BET) domain proteins using the small molecule inhibitor JQ1 has shown promise in preclinical models of acute kidney injury (AKI) and chronic kidney disease (CKD). However, its clinical translation faces challenges due to issues with poor pharmacokinetics and side effects. Here, we developed engineered liposomes loaded with JQ1 with the aim of enhancing kidney drug delivery and reducing the required minimum effective dose by leveraging cargo protection. These liposomes efficiently encapsulated JQ1 in both the membrane and core, demonstrating superior therapeutic efficacy compared to freely delivered JQ1 in a mouse model of kidney ischemia-reperfusion injury. JQ1-loaded liposomes (JQ1-NPs) effectively targeted the kidneys and only one administration, one-hour after injury, was enough to decrease the immune cell (neutrophils and monocytes) infiltration to the kidney-an early and pivotal step to prevent damage progression. By inhibiting BRD4, JQ1-NPs suppress the transcription of pro-inflammatory genes, such as cytokines (il-6) and chemokines (ccl2, ccl5). This success not only improved early the kidney function, as evidenced by decreased serum levels of BUN and creatinine in JQ1-NPs-treated mice, along with reduced tissue expression of the damage marker, NGAL, but also halted the production of extracellular matrix proteins (Fsp-1, Fn-1, α-SMA and Col1a1) and the fibrosis development. In summary, this work presents a promising nanotherapeutic strategy for AKI treatment and its progression and provides new insights into renal drug delivery.


Subject(s)
Azepines , Bromodomain Containing Proteins , Disease Progression , Kidney , Liposomes , Mice, Inbred C57BL , Nuclear Proteins , Renal Insufficiency, Chronic , Reperfusion Injury , Triazoles , Animals , Azepines/pharmacology , Azepines/administration & dosage , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Triazoles/pharmacology , Triazoles/administration & dosage , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Mice , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Male , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Disease Models, Animal , Nanoparticles , Cell Cycle Proteins/antagonists & inhibitors
2.
Eur J Pharm Biopharm ; 171: 39-49, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34998911

ABSTRACT

Bromodomain and extraterminal domain protein inhibitors (BETi) for cancer treatment did not convince during their first clinical trials. Their epigenetic mechanism of action is still not well understood, even if MYC is generally considered as its main downstream target. In this context, we intended to assess two new nanoformulations of the BETi JQ1 for the treatment of colorectal cancer (CRC). JQ1 was encapsulated at 10 mg/mL in lipid nanocapsules (LNC) or polymeric micelles (PM), both compatible for an intravenous administration. Their effect was compared with free JQ1 on several CRC cell lines in vitro and with daily intraperitoneal cyclodextrin (CD)-loaded JQ1 on the CT26 CRC tumor model in vivo. We showed that LNC preferentially accumulated in tumor, liver, and lymph nodes. LNC-JQ1 and CD-JQ1 similarly delayed tumor growth and increased median survival from 15 to 23 or 20.5 days. JQ1 altered MYC in only two among four CRC cell lines. This MYC-independence found in CT26 was confirmed in vivo by PCR and immunohistochemistry. The main explanation of the JQ1 anticancer effect was an increase in apoptosis. The investigation of its impact on the tumor microenvironment did not show significant effects. Finally, JQ1 association with irinotecan did not synergize in vivo with JQ1 nanoformulations. In conclusion, we demonstrated that the JQ1 anticancer effect was not improved by nanoencapsulation even if their tumor delivery was probably higher. MYC inhibition was not associated to JQ1 efficacy in the case of the CT26 CRC murine model.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , Colorectal Neoplasms/drug therapy , Liposomes , Nanoparticles , Proteins/antagonists & inhibitors , Triazoles/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Azepines/administration & dosage , Azepines/therapeutic use , Cell Line, Tumor/drug effects , Colorectal Neoplasms/metabolism , Drug Delivery Systems , Female , Humans , Infusions, Intravenous , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-myc/metabolism , Triazoles/administration & dosage , Triazoles/therapeutic use
3.
Cancer Sci ; 113(1): 28-40, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34727389

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the commonest lethal malignancies worldwide, and often diagnosed at an advanced stage, without any curative therapy. Immune checkpoint blockers targeting the programmed death receptor 1 (PD-1) have shown impressive antitumor activity in patients with advanced-stage HCC, while the response rate is only 30%. Inducible PD-L1 overexpression may result in a lack of response to cancer immunotherapy, which is attributed to a mechanism of adaptive immune resistance. Our study investigated that the overexpression of PD-L1 promoted the invasion and migration of liver cancer cells in vitro, and the induced overexpression of PD-L1 in the tumor microenvironment could weaken the effects of anti-PD-1 immunotherapy in a BALB/c mouse model of liver cancer. CPI-203, a small-molecule bromodomain-containing protein 4 (BRD4) inhibitor, which can potently inhibit PD-L1 expression in vitro and in vivo, combined with PD-1 antibody improved the response to immunotherapy in a liver cancer model. Cell transfection and chromatin immunoprecipitation assay manifested that BRD4 plays a key role in PD-L1 expression; CPI-203 can inhibit PD-L1 expression by inhibiting the BRD4 occupation of the PD-L1 promoter region. This study indicates a potential clinical immunotherapy method to reduce the incidence of clinical resistance to immunotherapy in patients with HCC.


Subject(s)
Acetamides/administration & dosage , Azepines/administration & dosage , B7-H1 Antigen/genetics , Cell Cycle Proteins/metabolism , Immune Checkpoint Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , Transcription Factors/metabolism , Up-Regulation/drug effects , Acetamides/pharmacology , Animals , Azepines/pharmacology , Cell Line, Tumor , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Promoter Regions, Genetic/drug effects , Tumor Escape/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
4.
Behav Brain Res ; 418: 113644, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34757001

ABSTRACT

Epigenetic mechanisms play important roles in the neurobiology of substance use disorder. In particular, bromodomain and extra-terminal domain (BET) proteins, a class of histone acetylation readers, have been found to regulate cocaine conditioned behaviors, but their role in the behavioral response to other drugs of abuse remains unclear. To address this knowledge gap, we examined the effects of the BET inhibitor, JQ1, on nicotine, amphetamine, morphine, and oxycodone conditioned place preference (CPP). Similar to previous cocaine studies, systemic administration of JQ1 caused a dose-dependent reduction in the acquisition of amphetamine and nicotine CPP in male mice. However, in opioid studies, JQ1 did not alter morphine or oxycodone CPP. Investigating the effects of JQ1 on other types of learning and memory, we found that JQ1 did not alter the acquisition of contextual fear conditioning. Together, these results indicate that BET proteins play an important role in the acquisition of psychostimulant-induced CPP but not the acquisition of opioid-induced CPP nor contextual fear conditioning.


Subject(s)
Anesthetics, Local/pharmacology , Azepines/administration & dosage , Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Conditioning, Psychological/drug effects , Dose-Response Relationship, Drug , Triazoles/administration & dosage , Amphetamine/pharmacology , Animals , Cocaine/pharmacology , Epigenomics , Learning/drug effects , Male , Memory/drug effects , Mice , Morphine/pharmacology , Nicotine/pharmacology
5.
Curr Drug Metab ; 22(14): 1114-1123, 2021.
Article in English | MEDLINE | ID: mdl-34856893

ABSTRACT

BACKGROUND: MIDD0301 is an oral asthma drug candidate that binds GABAA receptors on airway smooth muscle and immune cells. OBJECTIVE: The objective of this study is to identify and quantify MIDD0301 metabolites in vitro and in vivo and determine the pharmacokinetics of oral, IP, and IV administered MIDD0301. METHODS: In vitro conversion of MIDD0301 was performed using liver and kidney microsomes/S9 fractions followed by quantification using liquid chromatography-tandem mass spectrometry (LC-MS/MS). A LC-MS/MS method was developed using synthesized standards to quantify MIDD0301 and its metabolites in urine and feces. Blood, lung, and brain were harvested from animals that received MIDD0301 by oral, IP, and IV administration, followed by LCMS/ MS quantification. Imaging mass spectrometry was used to demonstrate the presence of MIDD0301 in the lung after oral administration. RESULTS: MIDD0301 is stable in the presence of liver and kidney microsomes and S9 fractions for at least two hours. MIDD0301 undergoes conversion to the corresponding glucuronide and glucoside in the presence of conjugating cofactors. For IP and IV administration, unconjugated MIDD0301 together with significant amounts of MIDD0301 glucoside and MIDD0301 taurine were found in urine and feces. Less conjugation was observed following oral administration, with MIDD0301 glucuronide being the main metabolite. Pharmacokinetic quantification of MIDD0301 in blood, lung, and brain showed very low levels of MIDD0301 in the brain after oral, IV, or IP administration. The drug half-life in these tissues ranged between 4-6 hours for IP and oral and 1-2 hours for IV administration. Imaging mass spectrometry demonstrated that orally administered MIDD0301 distributes uniformly in the lung parenchyma. CONCLUSION: MIDD0301 undergoes no phase I and moderate phase II metabolism.


Subject(s)
Anti-Asthmatic Agents/pharmacokinetics , Azepines/pharmacokinetics , Imidazoles/pharmacokinetics , Kidney/metabolism , Microsomes, Liver/metabolism , Administration, Intravenous , Administration, Oral , Animals , Anti-Asthmatic Agents/administration & dosage , Azepines/administration & dosage , Chromatography, Liquid , Dogs , Female , Humans , Imidazoles/administration & dosage , Injections, Intraperitoneal , Lung/metabolism , Mice , Microsomes/metabolism , Rats , Tandem Mass Spectrometry , Tissue Distribution
6.
Article in English | MEDLINE | ID: mdl-34731742

ABSTRACT

Besifloxacin has been embraced for the treatment of ocular bacterial infections. While LC-MS/MS has been used in investigating BSF pharmacokinetics, those costly instruments are not universally available and have complicated requirements for operation and maintenance. Additionally, pharmacokinetics of besifloxacin in dose-intense regimens are still unknown. Herein, a new quantification method was developed employing the widely accessible HPLC with fluorescence detection and applied to an ocular pharmacokinetic study with an intense regimen. Biosamples were pre-treated using protein precipitation. Chromatographic separation was achieved on a C18 column using mobile phase of 0.1% trifluoroacetic acid and acetonitrile. To address the weak fluorescence issue of besifloxacin, effects of detection parameters, elution pattern, pH of mobile phase, and reconstitution solvents were investigated. The method was fully validated per US-FDA guidelines and demonstrated precision (<13%), accuracy (91-112%), lower limit of quantification (5 ng/mL), linearity over clinically relevant concentrations (R2 > 0.999), matrix-effects (93-105%), recoveries (95-106%), and excellent selectivity. The method showed agreement with agar disk diffusion assays for in vitro screening and comparable in vivo performance to LC-MS/MS (Deming Regression, y = 1.010x + 0.123, r = 0.997; Bland-Altman analysis, mean difference was -6.3%; n = 21). Pharmacokinetic parameters suggested superior surface-retentive properties of besifloxacin. Maximum concentrations were 1412 ± 1910 and 0.15 ± 0.12 µg/mL; area under the curve was 1,637 and 1.08 µg·h/g; and half-life was 4.9 and 4.1 h; and pharmacokinetic-to-pharmacodynamic ratios were ≥ 409 and ≤ 17.8 against ocular pathogens in tears and aqueous humor, respectively. This readily available method is sensitive for biosamples and practical for routine use, facilitating besifloxacin therapy development.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Azepines/chemistry , Azepines/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Fluoroquinolones/chemistry , Fluoroquinolones/pharmacokinetics , Keratitis/drug therapy , Tandem Mass Spectrometry/methods , Animals , Anti-Bacterial Agents/administration & dosage , Aqueous Humor/chemistry , Azepines/administration & dosage , Chromatography, High Pressure Liquid/instrumentation , Female , Fluorescence , Fluoroquinolones/administration & dosage , Humans , Limit of Detection , Male , Rabbits , Tears/chemistry
7.
Cell Death Dis ; 12(11): 1020, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34716294

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC), the second most common primary liver cancer, is a fatal malignancy with a poor prognosis and only very limited therapeutic options. Although molecular targeted therapy is emerged as a promising treatment strategy, resistance to molecular-targeted therapy occurs inevitably, which represents a major clinical challenge. In this study, we confirmed that mammalian target of rapamycin (mTOR) signaling is the most significantly affected pathways in ICC. As a novel phosphoinositide 3-kinase (PI3K)/mTOR dual inhibitor, BEZ235, exerts antitumour activity by effectively and specifically blocking the dysfunctional activation of the PI3K/serine/threonine kinase (AKT)/mTOR pathway. We generate the orthotopic ICC mouse model through hydrodynamic transfection of AKT and yes-associated protein (YAP) plasmids into the mouse liver. Our study confirmed that BEZ235 can suppress the proliferation, invasion and colony conformation abilities of ICC cells in vitro but cannot effectively inhibit ICC progression in vivo. Inhibition of PI3K/mTOR allowed upregulation of c-Myc and YAP through suppressed the phosphorylation of LATS1. It would be a novel mechanism that mediated resistance to PI3K/mTOR dual inhibitor. However, Bromo- and extraterminal domain (BET) inhibition by JQ1 downregulates c-Myc and YAP transcription, which could enhance the efficacy of PI3K/mTOR inhibitors. The efficacy results of combination therapy exhibited effective treatment on ICC in vitro and in vivo. Our data further confirmed that the combination of PI3K/mTOR dual inhibitor and BET inhibition induces M1 polarization and suppresses M2 polarization in macrophages by regulating the expression of HIF-1α. Our study provides a novel and efficient therapeutic strategy in treating primary ICC.


Subject(s)
Antineoplastic Agents/administration & dosage , Azepines/administration & dosage , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , Imidazoles/administration & dosage , MTOR Inhibitors/administration & dosage , Nerve Tissue Proteins/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Quinolines/administration & dosage , Receptors, Cell Surface/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triazoles/administration & dosage , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Disease Models, Animal , Drug Therapy, Combination/methods , Humans , Mice , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcriptome , Treatment Outcome
8.
Int J Mol Sci ; 22(16)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34445411

ABSTRACT

BACKGROUND: The present study investigated the role of proteins from the bromodomain and extra-terminal (BET) family in schizophrenia-like abnormalities in a neurodevelopmental model of schizophrenia induced by prenatal methylazoxymethanol (MAM) administration (MAM-E17). METHODS: An inhibitor of BET proteins, JQ1, was administered during adolescence on postnatal days (P) 23-P29, and behavioural responses (sensorimotor gating, recognition memory) and prefrontal cortical (mPFC) function (long-term potentiation (LTP), molecular and proteomic analyses) studies were performed in adult males and females. RESULTS: Deficits in sensorimotor gating and recognition memory were observed only in MAM-treated males. However, adolescent JQ1 treatment affected animals of both sexes in the control but not MAM-treated groups and reduced behavioural responses in both sexes. An electrophysiological study showed LTP impairments only in male MAM-treated animals, and JQ1 did not affect LTP in the mPFC. In contrast, MAM did not affect activity-dependent gene expression, but JQ1 altered gene expression in both sexes. A proteomic study revealed alterations in MAM-treated groups mainly in males, while JQ1 affected both sexes. CONCLUSIONS: MAM-induced schizophrenia-like abnormalities were observed only in males, while adolescent JQ1 treatment affected memory recognition and altered the molecular and proteomic landscape in the mPFC of both sexes. Thus, transient adolescent inhibition of the BET family might prompt permanent alterations in the mPFC.


Subject(s)
Azepines/administration & dosage , Methylazoxymethanol Acetate/analogs & derivatives , Prefrontal Cortex/growth & development , Schizophrenia/physiopathology , Triazoles/administration & dosage , Adolescent , Adolescent Development/drug effects , Animals , Azepines/pharmacology , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/drug effects , Humans , Long-Term Potentiation/drug effects , Male , Methylazoxymethanol Acetate/toxicity , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Proteomics , Rats , Recognition, Psychology/drug effects , Schizophrenia/chemically induced , Schizophrenia/metabolism , Sex Characteristics , Triazoles/pharmacology
9.
Molecules ; 26(11)2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34205930

ABSTRACT

BACKGROUND: Neurotic disturbances, anxiety, neurosis-like disorders, and stress situations are widespread. Benzodiazepine tranquillizers have been found to be among the most effective antianxiety drugs. The pharmacological action of benzodiazepines is due to their interaction with the supra-molecular membrane GABA-a-benzodiazepine receptor complex, linked to the Cl-ionophore. Benzodiazepines enhance GABA-ergic transmission and this has led to a study of the role of GABA in anxiety. The search for anxiolytics and anticonvulsive agents has involved glutamate-ergic, 5HT-ergic substances and neuropeptides. However, each of these well-known anxiolytics, anticonvulsants and cognition enhancers (nootropics) has repeatedly been reported to have many adverse side effects, therefore there is an urgent need to search for new drugs able to restore damaged cognitive functions without causing significant adverse reactions. OBJECTIVE: Considering the relevance of epilepsy diffusion in the world, we have addressed our attention to the discovery of new drugs in this field Thus our aim is the synthesis and study of new compounds with antiepileptic (anticonvulsant) and not only, activity. METHODS: For the synthesis of compounds classical organic methods were used and developed. For the evaluation of biological activity some anticonvulsant and psychotropic methods were used. RESULTS: As a result of multistep reactions 26 new, five-membered heterocyclic systems were obtained. PASS prediction of anticonvulsant activity was performed for the whole set of the designed molecules and probability to be active Pa values were ranging from 0.275 to 0.43. The studied compounds exhibit protection against pentylenetetrazole (PTZ) seizures, anti-thiosemicarbazides effect as well as some psychotropic effect. The biological assays evidenced that some of the studied compounds showed a high anticonvulsant activity by antagonism with pentylenetetrazole. The toxicity of compounds is low and they do not induce muscle relaxation in the studied doses. According to the study of psychotropic activity it was found that the selected compounds have an activating behavior and anxiolytic effects on the models of "open field" and "elevated plus maze" (EPM). The data obtained indicate the anxiolytic (anti-anxiety) activity of the derivatives of pyrimidines, especially pronounced in compounds 6n, 6b, and 7c. The studied compounds increase the latent time of first immobilization on the model of "forced swimming" (FST) and exhibit some antidepressant effect similarly to diazepam. Docking studies revealed that compound 6k bound tightly in the active site of GABAA receptor with a value of the scoring function that estimates free energy of binding (ΔG) at -7.95 kcal/mol, while compound 6n showed the best docking score and seems to be dual inhibitor of SERT transporter as well as 5-HT1A receptor. CONCLUSIONS: Тhe selected compounds have an anticonvulsant, activating behavior and anxiolytic effects, at the same time exhibit some antidepressant effect.


Subject(s)
Azepines/administration & dosage , Azepines/chemical synthesis , Pyrimidines/administration & dosage , Pyrimidines/chemical synthesis , Seizures/drug therapy , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/chemical synthesis , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/pharmacology , Anticonvulsants/administration & dosage , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Azepines/chemistry , Azepines/pharmacology , Disease Models, Animal , Male , Maze Learning/drug effects , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Pentylenetetrazole/adverse effects , Pyrimidines/chemistry , Pyrimidines/pharmacology , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Rats , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Seizures/chemically induced , Seizures/physiopathology
10.
Eur J Cancer ; 154: 102-110, 2021 09.
Article in English | MEDLINE | ID: mdl-34256279

ABSTRACT

AIM: Aurora kinase A (AURKA) is a pleiotropic serine/threonine kinase that orchestrates mitotic progression. Paclitaxel stabilises microtubules and disrupts mitotic spindle assembly. The combination of AURKA inhibitor (alisertib) plus paclitaxel may be synergistic in rapidly proliferative cancers. We evaluated the safety and maximum tolerated dose (MTD) of alisertib in combination with nab-paclitaxel and its preliminary efficacy in patients with refractory high-grade neuroendocrine tumours (NETs). METHOD: This is a two-part, Phase 1 study. In Part A (dose escalation), a standard 3 + 3 design was used to determine MTD. In Part B (dose expansion), patients with predominantly refractory high-grade NETs were enrolled. RESULTS: In total, 31 patients were enrolled and treated (16 in Part A and 15 in Part B). The MTD of alisertib was 40 mg BID on D1-3 per week and nab-paclitaxel 100mg/m2 weekly: 3 weeks, 1 week off. Dose-limiting toxicity was neutropenia, and other common side-effects included fatigue, mucositis, and diarrhoea. In Part A, a patient with small-cell lung cancer with partial response (PR) was treated for more than 2 years, whereas four other patients with pancreatic ductal adenocarcinoma (one patient), small cell lung cancer (SCLC) (two patients), or high-grade NET (one patient) achieved stable disease (SD). In Part B, 13 of 15 enrolled patients had high-grade NETs. Of these, one had PR, and four had SD for more than 10 months. CONCLUSIONS: The combination of alisertib and nab-paclitaxel has manageable side-effect profile and showed promising preliminary efficacy in high-grade NETs, warranting further testing. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT01677559.


Subject(s)
Albumins/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azepines/administration & dosage , Neuroendocrine Tumors/drug therapy , Paclitaxel/administration & dosage , Pyrimidines/administration & dosage , Adult , Aged , Albumins/adverse effects , Azepines/adverse effects , Female , Humans , Male , Middle Aged , Neuroendocrine Tumors/mortality , Paclitaxel/adverse effects , Pyrimidines/adverse effects
11.
J Psychopharmacol ; 35(12): 1488-1495, 2021 12.
Article in English | MEDLINE | ID: mdl-34330170

ABSTRACT

BACKGROUND: Cognitive decline after oral administration of sedatives, such as benzodiazepines, is a serious side effect. Suvorexant, an orexin receptor antagonist, has a favorable tolerability and a limited side-effect profile. AIM: The purpose of this study was to estimate the cognitive decline 1 day after oral medication with lormetazepam, a benzodiazepine, and suvorexant by comparing mismatch negativity (MMN) and P300 reflecting auditory discrimination function. METHODS: Sixty healthy subjects (42 males) were randomly assigned to three groups receiving suvorexant 20 mg, lormetazepam 2 mg, or placebo in this double-blind, randomized control study. Event-related potential recordings during an auditory oddball task and a digit symbol substitution test (DSST) were performed 1 day after oral administration. RESULTS: MMN, on the day after oral administration, was significantly attenuated in the lormetazepam group compared with the other two groups, but there was no difference between the suvorexant and placebo groups. No significant difference was found in P300 amplitudes and DSST scores among the three groups. CONCLUSION: These findings suggest that suvorexant, unlike benzodiazepine, is not associated with cognitive deficits, as revealed by MMN but not P300. This study shows a neurophysiological difference in the effects of suvorexant and benzodiazepine on cognitive function.


Subject(s)
Auditory Perception/drug effects , Azepines/pharmacology , Benzodiazepines/pharmacology , Cognitive Dysfunction/chemically induced , Discrimination, Psychological/drug effects , Evoked Potentials, Auditory/drug effects , Lorazepam/analogs & derivatives , Orexin Receptor Antagonists/pharmacology , Triazoles/pharmacology , Adult , Azepines/administration & dosage , Azepines/adverse effects , Benzodiazepines/administration & dosage , Benzodiazepines/adverse effects , Electroencephalography , Event-Related Potentials, P300/drug effects , Female , Humans , Lorazepam/administration & dosage , Lorazepam/adverse effects , Lorazepam/pharmacology , Male , Orexin Receptor Antagonists/administration & dosage , Orexin Receptor Antagonists/adverse effects , Triazoles/administration & dosage , Triazoles/adverse effects , Young Adult
12.
J Clin Psychopharmacol ; 41(4): 414-420, 2021.
Article in English | MEDLINE | ID: mdl-34181362

ABSTRACT

PURPOSE/BACKGROUND: This study was designed as an early assessment of the safety of the orexin receptor antagonist suvorexant, but also included exploratory assessments of balance and psychomotor performance that are the focus of this report. METHODS/PROCEDURES: This was a double-blind, randomized, 3-period, crossover, phase 1 study. Balance and psychomotor performance were evaluated during the night in 12 healthy elderly participants after bedtime administration of suvorexant 30 mg (a supratherapeutic dose), the GABAergic agonist zolpidem 5 mg (the recommended dose in the elderly), or placebo. Balance (body sway measured by platform stability) and psychomotor performance (measured by choice reaction time) were assessed predose and at 1.5, 4, and 8 hours postdose in each period. Memory (measured by word recall) was assessed predose and at 4 hours postdose. FINDINGS/RESULTS: At 1.5 hours after nighttime administration of each drug (the approximate time of their anticipated maximal plasma concentrations), both zolpidem and suvorexant increased body sway versus placebo, with a greater increase for zolpidem than suvorexant. Suvorexant increased choice reaction time compared with placebo or zolpidem at 1.5 hours. There were no treatment differences on body sway or choice reaction time at 4 or 8 hours, or on word recall at 4 hours. IMPLICATIONS/CONCLUSIONS: These exploratory data suggest that a 30-mg dose of suvorexant (supratherapeutic) and a 5-mg dose of zolpidem (recommended dose in the elderly) impaired balance at 1.5 hours in healthy elderly people, with potentially less impairment for suvorexant relative to zolpidem, but no treatment differences on body sway or psychomotor performance at 4 and 8 hours. Because of their exploratory nature, these findings and their clinical relevance, if any, require confirmation in a prospective study.


Subject(s)
Azepines , Memory/drug effects , Postural Balance/drug effects , Psychomotor Performance/drug effects , Triazoles , Zolpidem , Aged , Azepines/administration & dosage , Azepines/adverse effects , Cross-Over Studies , Double-Blind Method , Drug Chronotherapy , Drug Monitoring/methods , Female , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/adverse effects , Healthy Volunteers , Humans , Male , Neuropsychological Tests , Orexin Receptor Antagonists/administration & dosage , Orexin Receptor Antagonists/adverse effects , Reaction Time/drug effects , Sleep Aids, Pharmaceutical/administration & dosage , Sleep Aids, Pharmaceutical/adverse effects , Triazoles/administration & dosage , Triazoles/adverse effects , Zolpidem/administration & dosage , Zolpidem/adverse effects
13.
Mol Ther ; 29(10): 3011-3026, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34058385

ABSTRACT

Glioblastoma (GBM) is the deadliest brain malignancy without effective treatments. Here, we reported that epidermal growth factor receptor-targeted chimeric antigen receptor T cells (EGFR CAR-T) were effective in suppressing the growth of GBM cells in vitro and xenografts derived from GBM cell lines and patients in mice. However, mice soon acquired resistance to EGFR CAR-T cell treatment, limiting its potential use in the clinic. To find ways to improve the efficacy of EGFR CAR-T cells, we performed genomics and transcriptomics analysis for GBM cells incubated with EGFR CAR-T cells and found that a large cohort of genes, including immunosuppressive genes, as well as enhancers in vicinity are activated. BRD4, an epigenetic modulator functioning on both promoters and enhancers, was required for the activation of these immunosuppressive genes. Accordingly, inhibition of BRD4 by JQ1 blocked the activation of these immunosuppressive genes. Combination therapy with EGFR CAR-T cells and JQ1 suppressed the growth and metastasis of GBM cells and prolonged survival in mice. We demonstrated that transcriptional modulation by targeting epigenetic regulators could improve the efficacy of immunotherapy including CAR-T, providing a therapeutic avenue for treating GBM in the clinic.


Subject(s)
Azepines/administration & dosage , Brain Neoplasms/therapy , Cell Cycle Proteins/metabolism , ErbB Receptors/immunology , Glioblastoma/therapy , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/metabolism , Transcription Factors/metabolism , Triazoles/administration & dosage , Animals , Azepines/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Epigenesis, Genetic/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Neoplasm Metastasis , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Xenograft Model Antitumor Assays
15.
Pharmacol Biochem Behav ; 201: 173111, 2021 02.
Article in English | MEDLINE | ID: mdl-33444602

ABSTRACT

This study was designed to examine the effects of intra- nucleus accumbens (NAc) of BDNF receptor antagonist ANA-12 on the acquisition and expression and intra- medial-prefrontal cortex (mPFC) of ANA-12 on the extinction and reinstatement of morphine-induced conditioned place preference (CPP) and also BDNF levels and apoptotic neurons in the NAc and mPFC of rats. In this study, adult male Wistar rats (200-250 g) were used. Two separate cannulas were inserted bilaterally into the NAc and/or mPFC. ANA-12 (3 µg/0.5 µl/side) was injected into the NAc and/or mPFC to evaluate the rewarding effects of morphine using a CPP paradigm. Then, the levels of BDNF and apoptotic in the NAc and mPFC were assessed at the end of each treatment phase using ELISA and TUNEL methods, respectively. All of vehicle-treated rats following morphine CPP showed the increase of BDNF levels and apoptotic neurons in the NAc and mPFC. ANA-12 significantly attenuated the acquisition and expression of morphine-induced CPP, BDNF levels and apoptotic neurons in the NAc during the acquisition, but not the expression phase. Also, ANA-12 significantly facilitated the extinction, but no effect on reinstatement of morphine CPP, and decreased BDNF levels and apoptotic neurons in the mPFC during the extinction, but not the reinstatement. We conclude that blocking TrkB with ANA-12 showed therapeutic effects on morphine-associated reward memory and neuronal death in the NAc and mPFC induced by morphine CPP. Thus, the BDNF-TrkB signaling may be important in the acquisition, expression, extinction, but not the reinstatement of morphine CPP.


Subject(s)
Apoptosis/drug effects , Azepines/administration & dosage , Benzamides/administration & dosage , Brain-Derived Neurotrophic Factor/metabolism , Memory/drug effects , Microinjections/methods , Morphine/administration & dosage , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , Receptor, trkB/antagonists & inhibitors , Reward , Signal Transduction/drug effects , Animals , Conditioning, Classical/drug effects , Conditioning, Operant/drug effects , Extinction, Psychological/drug effects , Male , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Wistar
16.
Invest New Drugs ; 39(2): 362-376, 2021 04.
Article in English | MEDLINE | ID: mdl-32981006

ABSTRACT

Camptothecin (CPT) and its derivatives, irinotecan and topotecan are specific topoisomerase I (Top1) inhibitors and potent anticancer drugs. Mechanistically, they induce DNA double-strand breaks (DSBs). Although CPT is an effective chemotherapeutic agent used in the management of advanced colorectal cancer, there exist associated side effects. Herein, we aimed to establish novel drug combinations that can effectively aid in managing the CPT-related side effects. Besides, bromodomain and extra-terminal domain (BET) inhibitors have proved as promising drugs that target epigenetic mechanisms in various cancers, they alter DNA repair processes, hence are a potential candidate for CPT synthetic lethality. A novel BET inhibitor JQ1 synergized with CPT, exerted antiproliferative effects. Through cell cycle analyses and apoptosis assays, we revealed that a combination of CPT and JQ1 induces subG1-phase arrest and enhances cell apoptosis. This combination increased the intensity of γ-H2AX staining, a specific marker of DSBs. Moreover, colorectal cancer cells highly expressing Top1 showed greater sensitivity to JQ1, which was lowered through the lentiviral shRNA-mediated knockdown of Top1. JQ1, combined with CPT, impeded the recruitment of the Mre11-mediated MRN complex. Finally, JQ1 enhanced the in vivo sensitivity of tumors to CPT without inducing toxicity. These results demonstrate that a combination of BET inhibitor with Top1 inhibitor is safe and exerts positive chemotherapeutic effects in colorectal cancer.


Subject(s)
Azepines/pharmacology , Camptothecin/pharmacology , DNA Repair/drug effects , MRE11 Homologue Protein/drug effects , Topoisomerase I Inhibitors/pharmacology , Triazoles/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Apoptosis , Azepines/administration & dosage , Camptothecin/administration & dosage , Cell Line, Tumor , Cell Survival , Colorectal Neoplasms/pathology , Humans , Triazoles/administration & dosage
17.
Epigenetics ; 16(1): 54-63, 2021 01.
Article in English | MEDLINE | ID: mdl-32603264

ABSTRACT

(1) Background: BET bromodomain proteins regulate transcription by binding acetylated histones and attracting key factors for, e.g., transcriptional elongation. BET inhibitors have been developed to block pathogenic processes such as cancer and inflammation. Despite having potent biological activities, BET inhibitors have still not made a breakthrough in clinical use for treating cancer. Multiple resistance mechanisms have been proposed but thus far no attempts to block this in glioma has been made. (2) Methods: Here, we have conducted a pharmacological synergy screen in glioma cells to search for possible combination treatments augmenting the apoptotic response to BET inhibitors. We first used HMBA, a compound that was developed as a differentiation therapy four decades ago but more recently was shown to primarily inhibit BET bromodomain proteins. Data was also generated using other BET inhibitors. (3) Results: In the synergy screen, we discovered that several MEK inhibitors can enhance apoptosis in response to HMBA in rat and human glioma cells in vitro as well as in vivo xenografts. The combination is not unique to HMBA but also other BET inhibitors such as JQ1 and I-BET-762 can synergize with MEK inhibitors. (4) Conclusions: Our findings validate a combination therapy previously demonstrated to exhibit anti-cancer activities in multiple other tumour types but which appears to have been lost in translation to the clinic.


Subject(s)
Acetamides/therapeutic use , Antineoplastic Agents/therapeutic use , Azepines/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Triazoles/therapeutic use , Acetamides/administration & dosage , Acetamides/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Azepines/administration & dosage , Azepines/pharmacology , Cell Line, Tumor , Drug Synergism , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Rats , Transcription Factors/antagonists & inhibitors , Triazoles/administration & dosage , Triazoles/pharmacology
18.
Br J Cancer ; 124(4): 744-753, 2021 02.
Article in English | MEDLINE | ID: mdl-33311588

ABSTRACT

BACKGROUND: Bromodomain and extra-terminal (BET) proteins are epigenetic readers that can drive carcinogenesis and therapy resistance. RO6870810 is a novel, small-molecule BET inhibitor. METHODS: We conducted a Phase 1 study of RO6870810 administered subcutaneously for 21 or 14 days of 28- or 21-day cycles, respectively, in patients with the nuclear protein of the testis carcinoma (NC), other solid tumours, or diffuse large B-cell lymphoma (DLBCL) with MYC deregulation. RESULTS: Fatigue (42%), decreased appetite (35%) and injection-site erythema (35%) were the most common treatment-related adverse events. Pharmacokinetic parameters demonstrated linearity over the dose range tested and support once-daily dosing. Pharmacodynamic assessments demonstrated sustained decreases in CD11b levels in peripheral blood mononuclear cells. Objective response rates were 25% (2/8), 2% (1/47) and 11% (2/19) for patients with NC, other solid tumours and DLBCL, respectively. Responding tumours had evidence of deregulated MYC expression. CONCLUSIONS: This trial establishes the safety, favourable pharmacokinetics, evidence of target engagement and preliminary single-agent activity of RO6870810. Responses in patients with NC, other solid tumours and DLBCL provide proof-of-principle for BET inhibition in MYC-driven cancers. The results support further exploration of RO6870810 as monotherapy and in combinations. CLINICAL TRIALS REGISTRATION: NCT01987362.


Subject(s)
Azepines/administration & dosage , Azepines/adverse effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Nuclear Proteins/metabolism , Proteins/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Azepines/blood , Azepines/pharmacokinetics , Dose-Response Relationship, Drug , Female , Humans , Lymphoma, Large B-Cell, Diffuse/blood , Male , Middle Aged , Neoplasms/blood , Neoplasms/metabolism , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/adverse effects , Small Molecule Libraries/pharmacokinetics
19.
Int J Radiat Oncol Biol Phys ; 109(5): 1440-1453, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33186615

ABSTRACT

PURPOSE: To investigate whether the vascular collapse in tumors by conventional dose rate (CONV) irradiation (IR) would also occur by the ultra-high dose rate FLASH IR. METHODS AND MATERIALS: Lewis lung carcinoma (LLC) cells were subcutaneously implanted in mice. This was followed by CONV or FLASH IR at 15 Gy. Tumors were harvested at 6 or 48 hours after IR and stained for CD31, phosphorylated myosin light chain (p-MLC), γH2AX (a surrogate marker for DNA double strand break), intracellular reactive oxygen species (ROS), or immune cells such as myeloid and CD8α T cells. Cell lines were irradiated with CONV IR for Western blot analyses. ML-7 was intraperitoneally administered daily to LLC-bearing mice for 7 days before 15 Gy CONV IR. Tumors were similarly harvested and analyzed. RESULTS: By immunostaining, we observed that CONV IR at 6 hours resulted in constricted vessel morphology, increased expression of p-MLC, and much higher numbers of γH2AX-positive cells in tumors, which were not observed with FLASH IR. Mechanistically, MLC activation by ROS is unlikely, because FLASH IR produced significantly more ROS than CONV IR in tumors. In vitro studies demonstrated that ML-7, an inhibitor of MLC kinase, abrogated IR-induced γH2AX formation and disappearance kinetics. Lastly, we observed that CONV IR when combined with ML-7 produced some effects similar to FLASH IR, including reduction in the vasculature collapse, fewer γH2AX-positive cells, and increased immune cell influx to the tumors. CONCLUSIONS: FLASH IR produced novel changes in the tumor microenvironment that were not observed with CONV IR. We believe that MLC activation in tumors may be responsible for some of the microenvironmental changes differentially regulated between CONV and FLASH IR.


Subject(s)
Carcinoma, Lewis Lung/radiotherapy , Myosin Light Chains/radiation effects , Tumor Microenvironment/radiation effects , Animals , Azepines/administration & dosage , Blood Vessels/pathology , Blood Vessels/radiation effects , CD8-Positive T-Lymphocytes/cytology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/metabolism , Histones/metabolism , Histones/radiation effects , Male , Mice , Mice, Inbred C57BL , Myosin Light Chains/antagonists & inhibitors , Myosin Light Chains/metabolism , Naphthalenes/administration & dosage , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/radiation effects , Radiotherapy/methods , Radiotherapy Dosage , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/radiation effects
20.
Behav Brain Res ; 399: 113015, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33212086

ABSTRACT

Post-traumatic stress disorder (PTSD) is a psychological disorder affecting many around the world. Growing evidence suggests that orexin-A is involved in the pathophysiology of depression and panic anxiety disorder. However, the role of orexin-A in PTSD remains unclear. Therefore, pharmacological manipulation of orexin-A can be a potential approach for the treatment of PTSD. Male Wistar rats were subjected to stress re-stress (SRS) by restraining them for 2 h followed by foot shock (FS) and halothane exposure on day-2 (D-2). Then the rats were weekly exposed to FS as re-stress cue . Suvorexant, an orexin antagonist (10, 20 and 30 mg/kg p.o.) and paroxetine (10 mg/kg p.o.) were administered from D-8 to D-32. Plasma and cerebrospinal fluid (CSF) were collected for corticosterone and orexin-A measurement. The analysis of serotonin and corticotropin-releasing factor receptor-1 (CRF-R1) were performed in the amygdalar tissue. SRS-induced PTSD-like symptoms like fear response, anxiety-like behaviour and hypocorticosteronism were attenuated by suvorexant and paroxetine. Interestingly, SRS exposed rats showed activation of orexin-A and serotonergic systems, which were also attenuated by suvorexant. Additionally, suvorexant ameliorated the extrahypothalamic induced upregulation of CRH-R1 in SRS-exposed rats. Therefore, orexin-A may be considered as a neurochemical-marker for PTSD and suvorexant alleviated PTSD-like symptoms through modulating orexinergic, serotonergic and neuroendocrine systems.


Subject(s)
Amygdala , Azepines/pharmacology , Corticosterone , Orexin Receptor Antagonists/pharmacology , Orexins , Receptors, Corticotropin-Releasing Hormone , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin , Stress Disorders, Post-Traumatic/drug therapy , Triazoles/pharmacology , Amygdala/drug effects , Amygdala/metabolism , Animals , Azepines/administration & dosage , Corticosterone/blood , Corticosterone/cerebrospinal fluid , Disease Models, Animal , Male , Orexin Receptor Antagonists/administration & dosage , Orexins/blood , Orexins/cerebrospinal fluid , Orexins/drug effects , Paroxetine/pharmacology , Rats , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/administration & dosage , Stress Disorders, Post-Traumatic/etiology , Triazoles/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...