Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
J Vet Pharmacol Ther ; 45(4): 325-351, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35460083

ABSTRACT

Robenacoxib is a veterinary-approved non-steroidal anti-inflammatory drug (NSAID) of the coxib group. It possesses anti-hyperalgesic, anti-inflammatory and anti-pyretic properties. Robenacoxib inhibits the cyclooxygenase (COX)-2 isoform of COX selectively (in vitro IC50 ratios COX-1:COX-2, 129:1 in dogs, 32:1 in cats). At registered dosages (2 mg/kg subcutaneously in dogs and cats, 1-4 mg/kg orally in dogs and 1-2.4 mg/kg orally in cats), robenacoxib produces significant inhibition of COX-2 whilst sparing COX-1. The pharmacokinetic (PK) profile of robenacoxib is characterized by a high degree of binding to plasma proteins (>98%) and moderate volume of distribution (at steady state, 240 ml/kg in dogs and 190 ml/kg in cats). In consequence, the terminal half-life in blood (<2 h) is short, despite moderate body clearance (0.81 L/kg/h) in dogs and low clearance (0.44 L/kg/h) in cats. Excretion is principally in the bile (65% in dogs and 72% in cats). Robenacoxib concentrates in inflamed tissues, and clinical efficacy is achieved with once-daily dosing, despite the short blood terminal half-life. In dogs, no relevant breed differences in robenacoxib PK have been detected. Robenacoxib has a wide safety margin; in healthy laboratory animals daily oral doses 20-fold (dog, 1 month), eight-fold (cat, 6 weeks) and five-fold (dog, 6 months) higher than recommended clinical doses were well tolerated. Clinical efficacy and safety have been demonstrated in orthopaedic and soft tissue surgery, and in musculoskeletal disorders in dogs and cats.


Subject(s)
Cat Diseases , Dog Diseases , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Cat Diseases/chemically induced , Cat Diseases/drug therapy , Cats , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/adverse effects , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Dog Diseases/drug therapy , Dogs , Phenylacetates/pharmacology , Phenylacetates/therapeutic use
2.
Sci Rep ; 11(1): 7721, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833276

ABSTRACT

The main objective of this pilot clinical trial was to evaluate outcome measures for the assessment of the nonsteroidal anti-inflammatory drug (NSAID) robenacoxib in cats with degenerative joint disease-associated pain (DJD-pain). Otherwise healthy cats (n = 109) with DJD-pain entered a parallel group, randomized, blinded clinical trial. Cats received placebo (P) or robenacoxib (R) for two consecutive 3-week periods. Treatment groups were PP, RR, and RP. Actimetry and owner-assessment data were collected. Data were analyzed using mixed-effects and generalized mixed-effects linear models. Activity data showed high within-cat and between-cat variability, and 82.4% of the values were zero. Compared to placebo, mean total activity was higher (5.7%) in robenacoxib-treated cats (p = 0.24); for the 80th percentile of activity, more robenacoxib-treated cats had a > 10% increase in activity after 3 (p = 0.046) and 6 weeks (p = 0.026). Robenacoxib treatment significantly decreased owner-assessed disability, (p = 0.01; 49% reduction in disability; effect size ~ 0.3), and improved temperament (p = 0.0039) and happiness (p = 0.021) after 6 weeks. More robenacoxib-treated cats were successes at 6 weeks (p = 0.018; NNT: 3.8). Adverse effect frequencies were similar across groups. Results identified suitable endpoints for confirmatory studies, while also indicating efficacy of robenacoxib in cats with DJD-pain.


Subject(s)
Cat Diseases/drug therapy , Diphenylamine/analogs & derivatives , Joint Diseases/veterinary , Osteoarthritis/veterinary , Pain Management/veterinary , Pain/veterinary , Phenylacetates/therapeutic use , Animals , Cats , Diphenylamine/therapeutic use , Female , Joint Diseases/complications , Male , Osteoarthritis/complications , Pain/complications , Pilot Projects , Placebos
3.
Commun Biol ; 4(1): 9, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33398072

ABSTRACT

The mitogen-activated protein kinase (MAPK) pathways are crucial regulators of the cellular processes that fuel the malignant transformation of normal cells. The molecular aberrations which lead to cancer involve mutations in, and transcription variations of, various MAPK pathway genes. Here, we examine the genome sequences of 40,848 patient-derived tumours representing 101 distinct human cancers to identify cancer-associated mutations in MAPK signalling pathway genes. We show that patients with tumours that have mutations within genes of the ERK-1/2 pathway, the p38 pathways, or multiple MAPK pathway modules, tend to have worse disease outcomes than patients with tumours that have no mutations within the MAPK pathways genes. Furthermore, by integrating information extracted from various large-scale molecular datasets, we expose the relationship between the fitness of cancer cells after CRISPR mediated gene knockout of MAPK pathway genes, and their dose-responses to MAPK pathway inhibitors. Besides providing new insights into MAPK pathways, we unearth vulnerabilities in specific pathway genes that are reflected in the re sponses of cancer cells to MAPK targeting drugs: a revelation with great potential for guiding the development of innovative therapies.


Subject(s)
MAP Kinase Signaling System/genetics , Mutation , Neoplasms/metabolism , A549 Cells , Benzamides/pharmacology , Benzamides/therapeutic use , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cell Survival , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drug Evaluation, Preclinical , Humans , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Neoplasms/genetics , Transcription, Genetic/drug effects
4.
J Clin Oncol ; 39(7): 797-806, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33507822

ABSTRACT

PURPOSE: Patients with neurofibromatosis type 1 (NF1) frequently develop plexiform neurofibromas (PNs), which can cause significant morbidity. We performed a phase II trial of the MAPK/ERK kinase inhibitor, mirdametinib (PD-0325901), in patients with NF1 and inoperable PNs. The primary objective was response rate based on volumetric magnetic resonance imaging analysis. METHODS: Inclusion criteria included age ≥ 16 years and a PN that was either progressive or causing significant morbidity. First-dose pharmacokinetics were performed. Patients completed patient-reported outcome measures. Patients received mirdametinib by mouth twice a day at 2 mg/m2/dose (maximum dose = 4 mg twice a day) in a 3-week on/1-week off sequence. Each course was 4 weeks in duration. Evaluations were performed after four courses for the first year and then after every six courses. Patients could receive a maximum of 24 total courses. RESULTS: Nineteen patients were enrolled, and all 19 received mirdametinib. The median age was 24 years (range, 16-39 years); the median baseline tumor volume was 363.8 mL (range, 3.9-5,161 mL). Eight of the 19 patients (42%) achieved a partial response of the target PN by course 12, and 10 (53%) had stable disease. One patient (5%) developed progressive disease at course 8. Significant and durable decreases were observed in pain ratings. CONCLUSION: To our knowledge, this analysis represents the first characterization of the activity and pharmacokinetics of mirdametinib in patients with NF1 and PNs and is the first published response study for MAPK/ERK kinase inhibitors in adults with NF1 and PNs. Mirdametinib given at 2 mg/m2/dose (maximum dose, 4 mg) twice daily in a 3-week on/1-week off sequence resulted in a 42% partial response rate with preliminary evidence of reduction in pain.


Subject(s)
Benzamides/therapeutic use , Diphenylamine/analogs & derivatives , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neurofibroma, Plexiform/drug therapy , Neurofibromatosis 1/drug therapy , Protein Kinase Inhibitors/therapeutic use , Adolescent , Adult , Benzamides/adverse effects , Benzamides/pharmacokinetics , Diphenylamine/adverse effects , Diphenylamine/pharmacokinetics , Diphenylamine/therapeutic use , Female , Humans , Magnetic Resonance Imaging , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurofibroma, Plexiform/diagnostic imaging , Neurofibroma, Plexiform/enzymology , Neurofibromatosis 1/diagnostic imaging , Neurofibromatosis 1/enzymology , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Time Factors , Treatment Outcome , United States , Young Adult
5.
Cancer Discov ; 10(2): 214-231, 2020 02.
Article in English | MEDLINE | ID: mdl-31771968

ABSTRACT

Spleen tyrosine kinase (SYK) is a nonmutated therapeutic target in acute myeloid leukemia (AML). Attempts to exploit SYK therapeutically in AML have shown promising results in combination with chemotherapy, likely reflecting induced mechanisms of resistance to single-agent treatment in vivo. We conducted a genome-scale open reading frame (ORF) resistance screen and identified activation of the RAS-MAPK-ERK pathway as one major mechanism of resistance to SYK inhibitors. This finding was validated in AML cell lines with innate and acquired resistance to SYK inhibitors. Furthermore, patients with AML with select mutations activating these pathways displayed early resistance to SYK inhibition. To circumvent SYK inhibitor therapy resistance in AML, we demonstrate that a MEK and SYK inhibitor combination is synergistic in vitro and in vivo. Our data provide justification for use of ORF screening to identify resistance mechanisms to kinase inhibitor therapy in AML lacking distinct mutations and to direct novel combination-based strategies to abrogate these. SIGNIFICANCE: The integration of functional genomic screening with the study of mechanisms of intrinsic and acquired resistance in model systems and human patients identified resistance to SYK inhibitors through MAPK signaling in AML. The dual targeting of SYK and the MAPK pathway offers a combinatorial strategy to overcome this resistance.This article is highlighted in the In This Issue feature, p. 161.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Syk Kinase/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Cell Line, Tumor , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Gene Expression Regulation, Leukemic/drug effects , Humans , Indazoles/pharmacology , Indazoles/therapeutic use , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutagenesis, Site-Directed , Mutation , Open Reading Frames/genetics , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Pyrazines/pharmacology , Pyrazines/therapeutic use , Syk Kinase/metabolism , Xenograft Model Antitumor Assays
6.
Clin Sci (Lond) ; 133(16): 1797-1811, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31387935

ABSTRACT

Background: Aneurysmal subarachnoid haemorrhage (SAH) is a variant of haemorrhagic stroke with a striking 50% mortality rate. In addition to the initial insult, secondary delayed brain injury may occur days after the initial ischemic insult and is associated with vasospasms leading to delayed cerebral ischemia. We have previously shown that the MEK1/2 inhibitor U0126 improves neurological assessment after SAH in rats. Aim: The purpose of the present study was to analyse the impact of a broad selection of high potency MEK1/2 inhibitors in an organ culture model and use the IC50 values obtained from the organ culture to select highly potent inhibitors for pre-clinical in vivo studies. Results: Nine highly potent mitogen activated protein kinase kinase (MEK1/2) inhibitors were screened and the two most potent inhibitors from the organ culture screening, trametinib and PD0325901, were tested in an in vivo experimental rat SAH model with intrathecal injections. Subsequently, the successful inhibitor trametinib was administered intraperitoneally in a second in vivo study. In both regimens, trametinib treatment caused significant reductions in the endothelin-1 induced contractility after SAH, which is believed to be associated with endothelin B receptor up-regulation. Trametinib treated rats showed improved neurological scores, evaluated by the ability to traverse a rotating pole, after induced SAH. Conclusion: The PD0325901 treatment did not improve the neurological score after SAH, nor showed any beneficial therapeutic effect on the contractility, contrasting with the reduction in neurological deficits seen after trametinib treatment. These data show that trametinib might be a potential candidate for treatment of SAH.


Subject(s)
Cerebral Arteries/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Subarachnoid Hemorrhage/drug therapy , Animals , Basilar Artery/drug effects , Basilar Artery/metabolism , Benzamides/pharmacology , Benzamides/therapeutic use , Cerebral Arteries/metabolism , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Male , Muscle Contraction/drug effects , Organ Culture Techniques , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Rats , Rats, Sprague-Dawley
7.
Vet Surg ; 48(4): 578-583, 2019 May.
Article in English | MEDLINE | ID: mdl-30637777

ABSTRACT

OBJECTIVE: To compare meloxicam and robenacoxib for short-term postoperative pain management after combined laparoscopic ovariectomy and laparoscopic-assisted gastropexy. STUDY DESIGN: Double-blind, prospective, randomised clinical trial. ANIMALS: Twenty-six client-owned female dogs. METHODS: Dogs undergoing combined laparoscopic ovariectomy and laparoscopic-assisted gastropexy were randomly divided into 2 groups. Before induction of anesthesia, 13 dogs received meloxicam (0.2 mg/kg subcutaneously), and 13 dogs received robenacoxib (2 mg/kg subcutaneously). Pain was scored with the Glasgow Composite Pain Scale (short form) before surgery and at 1, 6, 12, 18, and 24 hours after extubation. Rescue analgesia (tramadol, 3 mg/kg) was provided to dogs with a Glasgow pain score (GPS) ≥5. Glasgow pain scores were analyzed by ANOVA with treatment, age, and surgical time as fixed factors. RESULTS: Glasgow pain scores were higher at 24 hours postsurgery in dogs treated with robenacoxib (2.18 ± 0.29) compared with those treated with meloxicam (0.68 ± 0.41, P = .04). Two dogs treated with meloxicam and 7 dogs treated with robenacoxib required rescue analgesia. Regardless of the treatment, the overall GPS was lower at 18 and 24 hours postsurgery when the surgical time was >40 minutes compared with surgical times ≤40 minutes, but surgical site inflammation was likely a confounding factor in this finding. Glasgow pain score was not affected by patient age. CONCLUSION: Meloxicam was more effective than robenacoxib at controlling pain in the population of dogs reported here. CLINICAL SIGNIFICANCE: Preoperative administration of meloxicam effectively controls pain for 24 hours after combined laparoscopic ovariectomy and laparoscopic-assisted gastropexy, but rescue analgesia may be required.


Subject(s)
Diphenylamine/analogs & derivatives , Gastropexy/veterinary , Meloxicam/therapeutic use , Ovariectomy/veterinary , Pain, Postoperative/veterinary , Phenylacetates/therapeutic use , Analgesia/veterinary , Anesthesia , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Diphenylamine/therapeutic use , Dogs , Double-Blind Method , Female , Gastropexy/adverse effects , Laparoscopy , Ovariectomy/adverse effects , Pain Management/veterinary , Pain Measurement/veterinary , Pain, Postoperative/prevention & control , Prospective Studies , Random Allocation
8.
Cancer Biol Ther ; 20(1): 21-30, 2019.
Article in English | MEDLINE | ID: mdl-30261145

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) progression and chemotherapy insensitivity have been associated with aberrant PI3K/mTOR/MEK signalling. However, cell death responses activated by inhibitors of these pathways can differ - contextually varying with tumour genetic background. Here, we demonstrate that combining the dual PI3K/mTOR inhibitor PF5212384 (PF384) and MEK inhibitor PD325901 (PD901) more effectively induces apoptosis compared with either agent alone, independent of KRAS mutational status in PDAC cell lines. Additionally, a non-caspase dependent decrease in cell viability upon PF384 treatment was observed, and may be attributed to autophagy and G0/G1 cell cycle arrest. Using reverse phase protein arrays, we identify key molecular events associated with the conversion of cytostatic responses (elicited by single inhibitor treatments) into a complete cell death response when PF384 and PD901 are combined. This response was also independent of KRAS mutation, occurring in both BxPC3 (KRAS wildtype) and MIA-PaCa-2 (KRASG12C mutated) cells. In both cell lines, Bim expression increased in response to PF384/PD901 treatment (by 60% and 48%, respectively), while siRNA-mediated silencing of Bim attenuated the apoptosis induced by combination treatment. In parallel, Mcl-1 levels decreased by 36% in BxPC3, and 30% in MIA-PaCa-2 cells. This is consistent with a functional role for Mcl-1, and siRNA-mediated silencing enhanced apoptosis in PF384/PD901-treated MIA-PaCa-2 cells, whilst Mcl-1 overexpression decreased apoptosis induction by 24%. Moreover, a novel role was identified for PDCD4 loss in driving the apoptotic response to PF384/PD901 in BxPC3 and MIA-PaCa-2 cell lines. Overall, our data indicates PF384/PD901 co-treatment activates the same apoptotic mechanism in wild-type or KRAS mutant PDAC cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , MAP Kinase Signaling System/drug effects , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Benzamides/pharmacology , Benzamides/therapeutic use , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Survival/drug effects , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drug Screening Assays, Antitumor , Humans , Morpholines/pharmacology , Morpholines/therapeutic use , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Small Interfering/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Triazines/pharmacology , Triazines/therapeutic use
9.
Int J Chron Obstruct Pulmon Dis ; 14: 2611-2624, 2019.
Article in English | MEDLINE | ID: mdl-32063702

ABSTRACT

Background: Unlike p38 mitogen-activated protein Kinases (MAPK) that has been extensively studied in the context of lung-associated pathologies in COPD, the role of the dual-specificity mitogen-activated protein kinase kinase (MEK1/2) or its downstream signaling molecule extracellular signal-regulated kinases 1/2 (ERK1/2) in COPD is poorly understood. Objectives: The aim of this study was to address whether MEK1/2 pathway activation is linked to COPD and that targeting this pathway can improve lung inflammation through decreased immune-mediated inflammatory responses without compromising bacterial clearance. Methods: Association of MEK1/2 pathway activation to COPD was investigated by immunohistochemistry using lung tissue biopsies from COPD and healthy individuals and through analysis of sputum gene expression data from COPD patients. The anti-inflammatory effect of MEK1/2 inhibition was assessed on cytokine release from lipopolysaccharide-stimulated alveolar macrophages. The effect of MEK1/2 inhibition on bacterial clearance was assessed using Staphylococcus aureus killing assays with RAW 264.7 macrophage cell line and human neutrophils. Results: We report here MEK1/2 pathway activation demonstrated by increased pERK1/2 staining in bronchial epithelium and by the presence of MEK gene activation signature in sputum samples from COPD patients. Inhibition of MEK1/2 resulted in a superior anti-inflammatory effect in human alveolar macrophages in comparison to a p38 inhibitor. Furthermore, MEK1/2 inhibition led to an increase in bacterial killing in human neutrophils and RAW 264.7 cells that was not observed with the p38 inhibitor. Conclusion: Our data demonstrate the activation of MEK1/2 pathway in COPD and highlight a dual function of MEK1/2 inhibition in improving host defense responses whilst also controlling inflammation.


Subject(s)
Benzamides/pharmacology , Benzamides/therapeutic use , Diphenylamine/analogs & derivatives , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Pulmonary Disease, Chronic Obstructive/drug therapy , Adult , Aged , Cells, Cultured , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Female , Humans , Inflammation/drug therapy , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/immunology , Signal Transduction/drug effects , Young Adult
11.
J Vis Exp ; (136)2018 06 01.
Article in English | MEDLINE | ID: mdl-29912180

ABSTRACT

Embryonic stem (ES) cells have the potential to differentiate into any of the three germ layers (endoderm, mesoderm, or ectoderm), and can generate many lineages for regenerative medicine. ES cell culture in vitro has long been the subject of widespread concerns. Classically, mouse ES cells are maintained in serum and leukemia inhibitory factor (LIF)-containing medium. However, under serum/LIF conditions, cells show heterogeneity in morphology and the expression profile of pluripotency-related genes, and are mostly in a metastable state. Moreover, cultured ES cells exhibit global hypermethylation, but naïve ES cells of the inner cell mass (ICM) and primordial germ cells (PGCs) are in a state of global hypomethylation. The hypomethylated state of ICM and PGCs is closely associated with their pluripotency. To improve mouse ES cell culture methods, we have recently developed a new method based on the selectively combined utilization of two small-molecule compounds to maintain the DNA hypomethylated and pluripotent state. Here, we present that the co-treatment of vitamin C (Vc) and PD0325901 can erase about 90% of 5-methylcytosine (5mC) at 5 days in mouse ES cells. The generated 5mC content is comparable to that in PGCs. The mechanistic investigation shows that PD0325901 up-regulates Prdm14 expression to suppress Dnmt3b (de novo DNA methyltransferase) and Dnmt3l (the cofactor of Dnmt3b), by reducing de novo 5mC synthesis. Vc facilitates the conversion of 5mC to 5-hydroxymethylcytosine (5hmC) catalyzed mainly by Tet1 and Tet2, indicating the involvement of both passive and active DNA demethylations. Moreover, under Vc/PD0325901 conditions, mouse ES cells show homogeneous morphology and pluripotent state. Collectively, we propose a novel and chemical-synergy culture method for achieving DNA hypomethylation and maintenance of pluripotency in mouse ES cells. The small-molecule chemical-dependent method overcomes the major shortcomings of serum culture, and holds promise to generate homogeneous ES cells for further clinical applications and researches.


Subject(s)
Antioxidants/therapeutic use , Ascorbic Acid/therapeutic use , Benzamides/therapeutic use , Diphenylamine/analogs & derivatives , Genomics/methods , Mouse Embryonic Stem Cells/metabolism , Transcription Factors/genetics , Animals , Antioxidants/pharmacology , Ascorbic Acid/metabolism , Ascorbic Acid/pharmacology , Benzamides/pharmacology , Cell Differentiation , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Mice , Proto-Oncogene Proteins
12.
J Vet Med Sci ; 80(6): 989-996, 2018 Jun 29.
Article in English | MEDLINE | ID: mdl-29695672

ABSTRACT

The objective of this study was to evaluate the efficacy of a non-selective COX inhibitor (tolfenamic acid) and a selective COX-2 inhibitor (robenacoxib) for post-operative pain control in cats. Thirty cats undergoing ovariohysterectomy were randomly divided into three groups: the control (placebo) group, the tolfenamic acid (4 mg/kg/day) group, and the robenacoxib (1 mg/kg/day) group. Non-steroidal anti-inflammatory drugs (NSAIDs) were administered orally 2 hr before anesthesia induction and 24 and 48 hr post-operation. Buccal mucosal bleeding times (BMBTs) were assessed prior to anesthesia induction. Colorado pain scores and composite pain scores were evaluated in a blinded fashion before induction and 2, 8, 24, 30 and 48 hr post-operation. The Colorado pain scores of cats receiving robenacoxib were significantly lower than those of cats in the control group at 30 (P=0.0126) and 48 (P=0.0439) hr post-operation. The composite pain scores of cats from the robenacoxib group were lower than those of cats in the control group at 30 (P=0.0299) and 48 (P=0.0103) hr post-operation. The Colorado pain scores of cats receiving tolfenamic acid were significantly lower than those of cats in the control group at 30 hr (P=0.0186) post-operation. The composite pain scores in cats in the tolfenamic acid group were lower than the scores of cats in the control group at 24 (P=0.0403) and 48 (P=0.0413) hr post-operation. BMBTs remained within normal limits in all groups. Both tolfenamic acid and robenacoxib are useful for post-operative pain control in cats.


Subject(s)
Analgesia/veterinary , Analgesics/therapeutic use , Cats , Diphenylamine/analogs & derivatives , Pain, Postoperative/veterinary , Phenylacetates/therapeutic use , ortho-Aminobenzoates/therapeutic use , Analgesia/methods , Animals , Anti-Inflammatory Agents, Non-Steroidal , Diphenylamine/therapeutic use , Female , Hysterectomy/veterinary , Ovariectomy/veterinary , Pain Measurement/veterinary , Pain, Postoperative/drug therapy
13.
Biochem Pharmacol ; 151: 26-37, 2018 05.
Article in English | MEDLINE | ID: mdl-29499167

ABSTRACT

BACKGROUND: Blood reperfusion of the ischemic tissue after stroke promotes increases in the inflammatory response as well as accumulation of unfolded/misfolded proteins in the cell, leading to endoplasmic reticulum (ER) stress. Both Inflammation and ER stress are critical processes in the delayed death of the cells damaged after ischemia. The aim of this study is to check the putative synergic neuroprotective effect by combining anti-inflammatory and anti-ER stress agents after ischemia. METHODS: The study was performed on a two-vessel occlusion global cerebral ischemia model. Animals were treated with salubrinal one hour after ischemia and with robenacoxib at 8 h and 32 h after ischemia. Parameters related to the integrity of the blood-brain barrier (BBB), such as matrix metalloproteinase 9 and different cell adhesion molecules (CAMs), were analyzed by qPCR at 24 h and 48 h after ischemia. Microglia and cell components of the neurovascular unit, including neurons, endothelial cells and astrocytes, were analyzed by immunofluorescence after 48 h and seven days of reperfusion. RESULTS: Pharmacologic control of ER stress by salubrinal treatment after ischemia, revealed a neuroprotective effect over neurons that reduces the transcription of molecules involved in the impairment of the BBB. Robenacoxib treatment stepped neuronal demise forward, revealing a detrimental effect of this anti-inflammatory agent. Combined treatment with robenacoxib and salubrinal after ischemia prevented neuronal loss and changes in components of the neurovascular unit and microglia observed when animals were treated only with robenacoxib. CONCLUSION: Combined treatment with anti-ER stress and anti-inflammatory agents is able to provide enhanced neuroprotective effects reducing glial activation, which opens new avenues in therapies against stroke.


Subject(s)
Brain Ischemia/drug therapy , Cinnamates/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Diphenylamine/analogs & derivatives , Endoplasmic Reticulum Stress/drug effects , Neuroprotective Agents/therapeutic use , Phenylacetates/therapeutic use , Thiourea/analogs & derivatives , Animals , Blood-Brain Barrier/metabolism , Brain Ischemia/diagnostic imaging , Brain Ischemia/immunology , Cinnamates/administration & dosage , Cyclooxygenase 2 Inhibitors/administration & dosage , Diphenylamine/administration & dosage , Diphenylamine/therapeutic use , Drug Administration Schedule , Drug Therapy, Combination , Inflammation , Male , Neuroprotective Agents/administration & dosage , Phenylacetates/administration & dosage , Rats, Sprague-Dawley , Thiourea/administration & dosage , Thiourea/therapeutic use
14.
Sci Rep ; 7(1): 17038, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29213099

ABSTRACT

Recent advances in molecular subtyping of Pancreatic Ductal Adenocarcinoma (PDAC) support individualization of therapeutic strategies in this most aggressive disease. With the emergence of various novel therapeutic strategies and neoadjuvant approaches in this quickly deteriorating disease, robust approaches for fast evaluation of therapy response are urgently needed. To this aim, we designed a preclinical imaging-guided therapy trial where genetically engineered mice harboring endogenous aggressive PDAC were treated with the MEK targeting drug refametinib, which induces rapid and profound tumor regression in this model system. Multi-parametric non-invasive imaging was used for therapy response monitoring. A significant increase in the Diffusion-Weighted Magnetic Resonance Imaging derived Apparent Diffusion Coefficient (ADC) was noted already 24 hours after treatment onset. Histopathological analyses showed increased apoptosis and matrix remodeling at this time point. Our findings suggest the ADC parameter as an early predictor of therapy response in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/drug therapy , Diffusion Magnetic Resonance Imaging , Diphenylamine/analogs & derivatives , Diphenylamine/therapeutic use , Disease Models, Animal , Humans , Image Processing, Computer-Assisted , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Mice , Neoadjuvant Therapy , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Response Evaluation Criteria in Solid Tumors , Sulfonamides/therapeutic use , Pancreatic Neoplasms
15.
Target Oncol ; 12(6): 775-785, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29067643

ABSTRACT

BACKGROUND: This phase I, four-arm, open-label study (NCT01347866) evaluated the PI3K/mTOR inhibitors PF-04691502 (arms A, B) and gedatolisib (PF-05212384; arms C, D) in combination with the MEK inhibitor PD-0325901 (arm A, D) or irinotecan (arm B, C) in patients with advanced solid tumors. OBJECTIVES: Primary endpoint was dose-limiting toxicity with each combination. Secondary endpoints included safety, pharmacokinetics and preliminary antitumor activity. PATIENTS AND METHODS: Dose escalation followed a 3 + 3 design in arm C and a zone-based design in arm D. RESULTS: The PF-04691502 combination arms were closed prematurely due to low tolerability, and the maximum tolerated doses (MTDs) were not determined for either arm. The MTD for the combination of gedatolisib with irinotecan 180 mg/m2 was estimated to be 110 mg weekly and for the combination with PD-0325901 was not reached at the highest dose evaluated (gedatolisib 154 mg weekly). Plasma concentrations of gedatolisib were generally similar across dose groups in arm C (with irinotecan) and arm D (with PD-0325901). Frequent dose delays or dose reductions were required for both combinations, potentially preventing sustained therapeutic drug concentrations. Gedatolisib plus irinotecan produced a response rate of ~5% and clinical benefit in 16% of patients with advanced colorectal cancer (progression-free survival, 2.8 months). Preliminary evidence of clinical activity was observed with gedatolisib plus PD-0325901 in patients with ovarian cancer (three partial responses, n = 5) or endometrial cancer (one partial response, n = 1) and KRAS mutations. CONCLUSIONS: Further evaluations of gedatolisib are warranted in patients with advanced solid malignancies.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/therapeutic use , Camptothecin/analogs & derivatives , Diphenylamine/analogs & derivatives , Morpholines/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triazines/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides/pharmacology , Camptothecin/pharmacology , Camptothecin/therapeutic use , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Female , Humans , Irinotecan , Male , Morpholines/pharmacology , Neoplasms/pathology , Triazines/pharmacology
16.
BMC Vet Res ; 13(1): 197, 2017 Jun 26.
Article in English | MEDLINE | ID: mdl-28651573

ABSTRACT

BACKGROUND: Non-steroidal anti-inflammatory drugs (NSAIDs) have been proven to be effective in controlling peri-operative pain in dogs. Robenacoxib is an NSAID with high selectivity for the cyclooxygenase (COX)-2 isoform. The objective of this study was to assess the efficacy and safety of an oral tablet formulation of robenacoxib in client-owned dogs undergoing soft tissue surgery. The study was a prospective, multi-center, randomized, masked, placebo-controlled, parallel-group clinical trial. A total of 239 dogs were included and randomly allocated in a 1:1 ratio to receive either robenacoxib or placebo. Each dog received an oral tablet administration of either robenacoxib, at a target dose of 2 mg/kg, or placebo once prior to surgery and for two additional days post-operatively. All dogs also received a pre-anesthetic dose of 0.2 mg/kg butorphanol (intravenous or intramuscular). Pain assessments were performed using the short form of the Glasgow Composite Measure Pain Scale. Robenacoxib was compared to the placebo group on a success/failure basis. Treatment failure was defined as the need for rescue therapy to control post-operative pain. RESULTS: Significantly (P = 0.019) more dogs administered robenacoxib were considered treatment successes (89 of 116, 76.72%) compared to dogs given placebo (74 of 115, 64.35%). The percentage of treatment failure was therefore 23.28% in the robenacoxib and 35.65% in the placebo group. The least squares mean total pain scores were significantly different between groups and in favor of robenacoxib at 3 and 5 hours (P < 0.05) and 8 hours post-extubation (P < 0.01). Pain at the surgery sites (response to touch) was also significantly improved at 3, 5 and 8 hours post-extubation in dogs receiving robenacoxib versus placebo (P < 0.01). In addition, a significant overall improvement in posture/activity was revealed with robenacoxib having lower scores versus placebo (P < 0.01). No significant differences between the robenacoxib and placebo groups in the frequency of reported adverse events were observed. CONCLUSIONS: Robenacoxib by oral (tablet) administration was effective and well tolerated in the control of peri-operative pain and inflammation associated with soft tissue surgery in dogs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Diphenylamine/analogs & derivatives , Dogs/surgery , Pain, Postoperative/veterinary , Phenylacetates/therapeutic use , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Diphenylamine/administration & dosage , Diphenylamine/adverse effects , Diphenylamine/therapeutic use , Double-Blind Method , Female , Inflammation/drug therapy , Inflammation/veterinary , Male , Pain Measurement/veterinary , Pain, Postoperative/drug therapy , Phenylacetates/administration & dosage , Phenylacetates/adverse effects , Prospective Studies , Tablets
17.
J Vet Intern Med ; 31(3): 832-841, 2017 May.
Article in English | MEDLINE | ID: mdl-28514527

ABSTRACT

BACKGROUND: Nonsteroidal anti-inflammatory drugs (NSAIDs) are used routinely to control pain and inflammation after surgery in dogs. Robenacoxib is a cyclooxygenase-2 selective NSAID. HYPOTHESIS/OBJECTIVE: Assess the clinical efficacy and safety of an injectable formulation of robenacoxib in dogs undergoing surgery. ANIMALS: Three hundred and seventeen client-owned dogs (N = 159 robenacoxib or N = 158 placebo). METHODS: In this prospective, multicenter, randomized, masked, placebo-controlled, parallel-group study, dogs received a SC injection of either robenacoxib, at a target dose of 2.0 mg/kg, or placebo once prior to surgery and for 2 additional days postoperatively. Pain assessments were performed using the short form of the Glasgow Composite Measure Pain Scale (CMPS-SF). The primary efficacy variable was treatment success/failure, with failure defined as the need for rescue therapy to control pain or withdrawal of the dog from the study due to an adverse event. RESULTS: Significantly (P = .006) more dogs administered robenacoxib were considered treatment successes (108 of 151, 73.7%) compared to dogs given placebo (85 of 152, 58.1%). Total pain scores (P < .01), pain at the surgery sites (response to touch, P < .01), and posture/activity (P < .05) were significantly improved at 3, 5, and 8 hours postextubation in dogs receiving robenacoxib versus placebo. CONCLUSIONS AND CLINICAL IMPORTANCE: Robenacoxib administered by SC injection prior to surgery and for 2 additional days postoperatively was effective and well tolerated in the control of postoperative pain and inflammation associated with soft tissue surgery in dogs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Diphenylamine/analogs & derivatives , Dogs/surgery , Pain Management/veterinary , Phenylacetates/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Diphenylamine/administration & dosage , Diphenylamine/adverse effects , Diphenylamine/therapeutic use , Female , Injections, Subcutaneous/veterinary , Male , Pain Management/methods , Pain Measurement/veterinary , Pain, Postoperative/prevention & control , Pain, Postoperative/veterinary , Phenylacetates/administration & dosage , Phenylacetates/adverse effects
18.
Physiol Rep ; 5(5)2017 Mar.
Article in English | MEDLINE | ID: mdl-28270590

ABSTRACT

Aortic valve disease (AVD) is a common condition with a progressive natural history, and presently, there are no pharmacologic treatment strategies. Elastic fiber fragmentation (EFF) is a hallmark of AVD, and increasing evidence implicates developmental elastic fiber assembly defects. Emilin1 is a glycoprotein necessary for elastic fiber assembly that is present in both developing and mature human and mouse aortic valves. The Emilin1-deficient mouse (Emilin1-/- ) is a model of latent AVD, characterized by activated TGFß/MEK/p-Erk signaling and upregulated elastase activity. Emilin1-/- aortic valves demonstrate early EFF and aberrant angiogenesis followed by late neovascularization and fibrosis. The objective of this study was to test the effectiveness of three different targeted therapies. Aged (12-14 months) Emilin1-/- mice were treated with refametinib (RDEA-119, MEK1/2 inhibitor), doxycycline (elastase inhibitor), or G6-31 (anti-VEGF-A mouse antibody) for 4 weeks. Refametinib- and doxycycline-treated Emilin1-/- mice markedly reduced MEK/p-Erk activation in valve tissue. Furthermore, both refametinib and doxycycline attenuated elastolytic cathepsin K, L, MMP-2, and MMP-9 activation, and abrogated macrophage and neutrophil infiltration in Emilin1-/- aortic valves. RNAseq analysis was performed in aortic valve tissue from adult (4 months) and aged (14 months) Emilin1-/- and age-matched wild-type control mice, and demonstrated upregulation of genes associated with MAPK/MEK/p-Erk signaling and elastases at the adult stage and inflammatory pathways at the aged stage controlling for age. These results suggest that Erk1/2 signaling is an important modulator of early elastase activation, and pharmacological inhibition using refametinib may be a promising treatment to halt AVD progression.


Subject(s)
Antibodies/therapeutic use , Aortic Valve/drug effects , Diphenylamine/analogs & derivatives , Doxycycline/therapeutic use , Heart Defects, Congenital/drug therapy , Heart Valve Diseases/drug therapy , MAP Kinase Signaling System/drug effects , Membrane Glycoproteins/genetics , Sulfonamides/therapeutic use , Vascular Endothelial Growth Factor A/immunology , Animals , Antibodies/pharmacology , Aortic Valve/metabolism , Aortic Valve/pathology , Bicuspid Aortic Valve Disease , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Disease Models, Animal , Disease Progression , Doxycycline/pharmacology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heart Valve Diseases/metabolism , Heart Valve Diseases/pathology , Mice , Mice, Knockout , Pancreatic Elastase/metabolism , Sulfonamides/pharmacology , Up-Regulation/drug effects
19.
Vet J ; 221: 38-47, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28283079

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs) have been suggested as effective adjunctive anti-tumour agents in human and veterinary medicine. However, the molecular mechanisms associated with their anti-tumour effects and correlations with the expression of cyclooxygenase (COX) and related molecules in tumours remain controversial. The objective of this study was to compare the expression profiles of COX and related molecules with NSAID sensitivity and to explore the molecular mechanisms of anti-tumour effects. The expression profiles of COXs, prostaglandins (PGs), PGD2 synthases, and PGE2 synthases were obtained, and their correlations with in vitro sensitivity to the NSAIDs piroxicam, carprofen, and robenacoxib were examined, using 26 canine cancer cell lines. Subsequently, microarray analysis was performed using one melanoma cell line to gain insight into mechanisms by which NSAIDs could exert cytotoxic effects. No strong correlation was observed between the cellular expression of COX and related molecules and sensitivity to NSAID treatment. Additionally, NSAIDs inhibited cell growth only at considerably higher concentrations than those required for functional COX inhibition. Microarray data demonstrated that five genes (SLC16A6, PER2, SLC9A8, HTR2B, and BRAF) were significantly upregulated and that four genes (LOC488305, H2AFJ, LOC476445, and ANKRD43) were significantly downregulated by NSAID exposure to the melanoma cell line. These results suggest that the direct in vitro anti-tumour effects of NSAIDs might be mediated by COX/PG-independent pathways. Novel candidate genes that could potentially be involved in the anti-tumour effects of NSAIDs were identified. Further validation and elucidation of their associated mechanisms will contribute to patient selection in clinical settings and the development of effective combination therapies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/therapeutic use , Dog Diseases/drug therapy , Neoplasms/veterinary , Animals , Carbazoles/therapeutic use , Cell Line, Tumor , Cyclooxygenase Inhibitors/therapeutic use , Dinoprostone/metabolism , Diphenylamine/analogs & derivatives , Diphenylamine/therapeutic use , Dogs , Dose-Response Relationship, Drug , Gene Expression Profiling/veterinary , Neoplasms/drug therapy , Phenylacetates/therapeutic use , Piroxicam/therapeutic use , Prostaglandin D2/metabolism
20.
Cell ; 168(5): 817-829.e15, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28215705

ABSTRACT

Investigating therapeutic "outliers" that show exceptional responses to anti-cancer treatment can uncover biomarkers of drug sensitivity. We performed preclinical trials investigating primary murine acute myeloid leukemias (AMLs) generated by retroviral insertional mutagenesis in KrasG12D "knockin" mice with the MEK inhibitor PD0325901 (PD901). One outlier AML responded and exhibited intrinsic drug resistance at relapse. Loss of wild-type (WT) Kras enhanced the fitness of the dominant clone and rendered it sensitive to MEK inhibition. Similarly, human colorectal cancer cell lines with increased KRAS mutant allele frequency were more sensitive to MAP kinase inhibition, and CRISPR-Cas9-mediated replacement of WT KRAS with a mutant allele sensitized heterozygous mutant HCT116 cells to treatment. In a prospectively characterized cohort of patients with advanced cancer, 642 of 1,168 (55%) with KRAS mutations exhibited allelic imbalance. These studies demonstrate that serial genetic changes at the Kras/KRAS locus are frequent in cancer and modulate competitive fitness and MEK dependency.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Colorectal Neoplasms/genetics , Diphenylamine/analogs & derivatives , MAP Kinase Signaling System , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Line, Tumor , Clonal Evolution , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drug Resistance, Neoplasm , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System/drug effects , Mice , Mutation , Retroviridae
SELECTION OF CITATIONS
SEARCH DETAIL
...