Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 321
Filtrar
1.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 220-225, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38814212

RESUMEN

This study explored the impact of penehyclidine hydrochloride on cognitive function in rats with brain injury. Sprague-Dawley rats (n=36) were randomly assigned to sham-operation, model, and penehyclidine hydrochloride groups. Rats in the sham-operation group underwent craniotomy, while the model and penehyclidine hydrochloride groups received brain injury models and interventions with normal saline and penehyclidine hydrochloride, respectively. Specimens were obtained two weeks post-intervention. Neurological deficits were evaluated using Zea-Longa scores, and memory was assessed with the Morris water maze test. ELISA determined brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) content. mRNA expressions of BDNF and NGF were assessed via qPCR, and phosphorylated CREB (p-CREB) protein expression was measured by Western blotting. Compared to the sham-operation group, both model and penehyclidine hydrochloride groups showed increased Zea-Longa scores. Escape latencies were longer and platform crossings were fewer in model and penehyclidine hydrochloride groups compared to the sham-operation group, but penehyclidine hydrochloride demonstrated a shorter latency and more platform crossings than the model group. BDNF and NGF content decreased in model and penehyclidine hydrochloride groups compared to the sham-operation group, with an increase in the penehyclidine hydrochloride group compared to the model group. mRNA expression levels declined in model and penehyclidine hydrochloride groups but were higher in the latter. p-CREB protein expression was lower in model and penehyclidine hydrochloride groups compared to the sham-operation group but higher in the penehyclidine hydrochloride group than the model group. Penehyclidine hydrochloride exhibited neuroprotective effects by upregulating the cAMP/CREB signaling pathway, improving cognitive function in rats with brain injury.


Asunto(s)
Lesiones Encefálicas , Factor Neurotrófico Derivado del Encéfalo , Cognición , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , AMP Cíclico , Quinuclidinas , Ratas Sprague-Dawley , Transducción de Señal , Animales , Transducción de Señal/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Quinuclidinas/farmacología , Quinuclidinas/uso terapéutico , Cognición/efectos de los fármacos , Masculino , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , AMP Cíclico/metabolismo , Ratas , Factor de Crecimiento Nervioso/metabolismo , Factor de Crecimiento Nervioso/genética , Fosforilación/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Modelos Animales de Enfermedad
2.
BMC Pulm Med ; 24(1): 207, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38671448

RESUMEN

OBJECTIVE: The aim of this research was to examine how penehyclidine hydrochloride (PHC) impacts the occurrence of pyroptosis in lung tissue cells within a rat model of lung ischemia-reperfusion injury. METHODS: Twenty-four Sprague Dawley (SD) rats, weighing 250 g to 270 g, were randomly distributed into three distinct groups as outlined below: a sham operation group (S group), a control group (C group), and a test group (PHC group). Rats in the PHC group received a preliminary intravenous injection of PHC at a dose of 3 mg/kg. At the conclusion of the experiment, lung tissue and blood samples were collected and properly stored for subsequent analysis. The levels of malondialdehyde, superoxide dismutase, and myeloperoxidase in the lung tissue, as well as IL-18 and IL-1ß in the blood serum, were assessed using an Elisa kit. Pyroptosis-related proteins, including Caspase1 p20, GSDMD-N, and NLRP3, were detected through the western blot method. Additionally, the dry-to-wet ratio (D/W) of the lung tissue and the findings from the blood gas analysis were also documented. RESULTS: In contrast to the control group, the PHC group showed enhancements in oxygenation metrics, reductions in oxidative stress and inflammatory reactions, and a decrease in lung injury. Additionally, the PHC group exhibited lowered levels of pyroptosis-associated proteins, including the N-terminal segment of gasdermin D (GSDMD-N), caspase-1p20, and nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3). CONCLUSION: Pre-administration of PHC has the potential to mitigate lung ischemia-reperfusion injuries by suppressing the pyroptosis of lung tissue cells, diminishing inflammatory reactions, and enhancing lung function. The primary mechanism behind anti-pyroptotic effect of PHC appears to involve the inhibition of oxidative stress.


Asunto(s)
Gasderminas , Pulmón , Piroptosis , Quinuclidinas , Ratas Sprague-Dawley , Daño por Reperfusión , Animales , Piroptosis/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/prevención & control , Ratas , Quinuclidinas/farmacología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Masculino , Malondialdehído/metabolismo , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Interleucina-18/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Superóxido Dismutasa/metabolismo , Peroxidasa/metabolismo , Estrés Oxidativo/efectos de los fármacos , Caspasa 1/metabolismo , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo
3.
J Pharmacol Exp Ther ; 384(3): 439-444, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36635086

RESUMEN

Cancer treatments are frequently associated with nausea and vomiting despite greatly improved preventive medication. Administration of antinausea agents as eye drops might provide easy and rapid access to the systemic circulation for prevention of nausea and vomiting and for the treatment of breakthrough nausea, but the ocular administration route has rarely been evaluated. Palonosetron is a second-generation 5-hydroxytryptamine 3 receptor antagonist approved for prevention and treatment of chemotherapy-induced nausea and vomiting. We compared ocular administration of palonosetron to non-active vehicle eye drops and to intravenous palonosetron in the prevention of cisplatin-induced nausea and vomiting in beagle dogs. Palonosetron ocular drops at the dose of 30 µg/kg reduced cumulative nausea over time as measured with the area under the visual analog scale curve by 98% compared with the vehicle and reduced nausea-associated dog behavior by 95%. Vomiting was completely prevented with repeated palonosetron ocular dosing. Hydroxypropyl-ß-cyclodextrin (HP-ß-CD) palonosetron formulation was well tolerated locally at the palonosetron concentration of 3 mg/ml. Absorption of palonosetron from eye drops was fast. Ten minutes after ocular administration, palonosetron plasma concentrations were similar compared with intravenous administration, and remained similar for six hours. We conclude that palonosetron is rapidly absorbed into the systemic circulation from eye drops. Ocularly administered palonosetron was well tolerated in the HP-ß-CD formulation and was highly effective in the prevention of cisplatin-induced nausea and vomiting. Evaluation of the safety and efficacy of ocular administration of palonosetron is warranted in the prevention and treatment of chemotherapy-induced nausea and vomiting in clinical trials. SIGNIFICANCE STATEMENT: Palonosetron, an effective and well-tolerated antiemetic drug was rapidly absorbed into the systemic blood circulation when administered as eye drops. The achieved palonosetron blood concentrations prevented cisplatin-induced nausea and vomiting in beagle dogs. Palonosetron eye drops might provide an easy and quick method for administering palonosetron when parenteral administration is desired and intravenous administration is not feasible.


Asunto(s)
Antineoplásicos , Cisplatino , Animales , Perros , Palonosetrón/efectos adversos , 2-Hidroxipropil-beta-Ciclodextrina , Administración Oftálmica , Isoquinolinas/farmacología , Quinuclidinas/farmacología , Vómitos/inducido químicamente , Náusea/inducido químicamente , Antineoplásicos/uso terapéutico , Dexametasona
4.
Leukemia ; 36(9): 2269-2280, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35835991

RESUMEN

TP53 mutations correlate with inferior survival in many cancers. APR-246 is a compound to shift mutant p53 and exhibits anti-cancer effects. Among its effects, APR-246 facilitates the binding of restored p53 mutants to target genes and their transcription. A set of 2464 DLBCL cases from multiple cohorts including our center, was integrated to identify the type and localization of TP53 mutations and clinical impacts. APR-246 was applied in TP53-mutated DLBCL cells and xenograft mouse models to explore the anti-tumor effect. TP53 mutations frequency was 16% and TP53 mutations correlated with poor overall survival (OS) and progression-free survival (PFS) in all cases, especially in germinal center B-cell-like (GCB) and unclassified (UNC) subtypes. Notably, TP53 single mutations in the DNA binding domain (DBD) led to poor OS and PFS. Specifically, mutations in exon 7 correlated with poorer OS, while mutations in exons 5 and 6 associated with inferior PFS. APR-246 induces p53-dependent ferritinophagy of DLBCL cells with TP53 missense mutation on exon 7 and ferroptosis of DLBCL cells harboring wild-type TP53 and other TP53 mutations. TP53 mutations on exons 5, 6 and 7 are predictors of progression and survival. Targeting mutant p53 by APR-246 is a promising therapeutic approach for DLBCL patients.


Asunto(s)
Ferroptosis , Linfoma de Células B Grandes Difuso , Quinuclidinas , Animales , Ferroptosis/efectos de los fármacos , Humanos , Hierro/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , Ratones , Mutación , Pronóstico , Quinuclidinas/farmacología , Proteína p53 Supresora de Tumor
5.
Oncol Rep ; 47(4)2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35234267

RESUMEN

Pharmacological reactivation of tumor­suppressor protein p53 has acted as a promising strategy for more than 50% of human cancers that carry a non­functional mutant p53 (mutp53). p53 plays a critical role in preserving genomic integrity and DNA fidelity through numerous biological processes, including cell cycle arrest, DNA repair, senescence and apoptosis. By contrast, non­functional mutp53 compromises the aforementioned genome stabilizing mechanisms through gain of function, thereby increasing genomic instability in human cancers. Restoring the functional activity of p53 using both genetic and pharmacological approaches has gained prominence in targeting p53­mutated tumors. Thus, the present study aimed to investigate the reactivation of p53 in DNA repair mechanisms and the maintenance of genomic stability using PRIMA­1MET/APR­246 small molecules, in both MDA­MB­231 and MCF­7 breast cancer cell lines, which carry mutp53 and wild­type p53, respectively. Results of the present study revealed that reactivation of p53 through APR­246 led to an increase in the functional activity of DNA repair. Prolonged treatment of MDA­MB­231 cells with APR­246 in the presence of cisplatin led to a reduction in mutational accumulation, compared with cells treated with cisplatin alone. These findings demonstrated that APR­246 may act as a promising small molecule to control the genomic instability in p53­mutated tumors.


Asunto(s)
Neoplasias , Proteína p53 Supresora de Tumor , Inestabilidad Genómica , Humanos , Mutación , Neoplasias/patología , Quinuclidinas/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
J Pharmacol Exp Ther ; 380(2): 94-103, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34782407

RESUMEN

Studies performed in healthy smokers have documented a diminished responsiveness to tussive challenges, and several lines of experimental evidence implicate nicotine as an antitussive component in both cigarette smoke and the vapors generated by electronic cigarettes (eCigs). We set out to identify the nicotinic receptor subtype involved in the antitussive actions of nicotine and to further evaluate the potential of nicotinic receptor-selective agonists as cough-suppressing therapeutics. We confirmed an antitussive effect of nicotine in guinea pigs. We additionally observed that the alpha-4 beta-2 (α 4 ß 2)-selective agonist Tc-6683 was without effect on evoked cough responses in guinea pigs, while the α 7-selective agonist PHA 543613 dose-dependently inhibited evoked coughing. We subsequently describe the preclinical evidence in support of ATA-101, a potent and highly selective (α 7) selective nicotinic receptor agonist, as a potential candidate for antitussive therapy in humans. ATA-101, formerly known as Tc-5619, was orally bioavailable and moderately central nervous system (CNS) penetrant and dose-dependently inhibited coughing in guinea pigs evoked by citric acid and bradykinin. Comparing the effects of airway targeted administration versus systemic dosing and the effects of repeated dosing at various times prior to tussive challenge, our data suggest that the antitussive actions of ATA-101 require continued engagement of α 7 nicotinic receptors, likely in the CNS. Collectively, the data provide the preclinical rationale for α 7 nicotinic receptor engagement as a novel therapeutic strategy for cough suppression. The data also suggest that α 7 nicotinic acetylcholine receptor (nAChR) activation by nicotine may be permissive to nicotine delivery in a way that may promote addiction. SIGNIFICANCE STATEMENT: This study documents the antitussive actions of nicotine and identifies the α7 nicotinic receptor subtype as the target for nicotine during cough suppression described in humans. We additionally present evidence suggesting that ATA-101 and other α7 nicotinic receptor-selective agonists may be promising candidates for the treatment of chronic refractory cough.


Asunto(s)
Antitusígenos/uso terapéutico , Benzofuranos/uso terapéutico , Tos/tratamiento farmacológico , Agonistas Nicotínicos/uso terapéutico , Quinuclidinas/uso terapéutico , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Antitusígenos/farmacología , Benzofuranos/farmacología , Tos/metabolismo , Cobayas , Masculino , Nicotina/metabolismo , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Quinuclidinas/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/agonistas
7.
Nat Commun ; 12(1): 7057, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34862374

RESUMEN

In response to genotoxic stress, the tumor suppressor p53 acts as a transcription factor by regulating the expression of genes critical for cancer prevention. Mutations in the gene encoding p53 are associated with cancer development. PRIMA-1 and eprenetapopt (APR-246/PRIMA-1MET) are small molecules that are converted into the biologically active compound, methylene quinuclidinone (MQ), shown to reactivate mutant p53 by binding covalently to cysteine residues. Here, we investigate the structural basis of mutant p53 reactivation by MQ based on a series of high-resolution crystal structures of cancer-related and wild-type p53 core domains bound to MQ in their free state and in complexes with their DNA response elements. Our data demonstrate that MQ binds to several cysteine residues located at the surface of the core domain. The structures reveal a large diversity in MQ interaction modes that stabilize p53 and its complexes with DNA, leading to a common global effect that is pertinent to the restoration of non-functional p53 proteins.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Aza/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Neoplasias/tratamiento farmacológico , Quinuclidinas/farmacología , Proteína p53 Supresora de Tumor/agonistas , Antineoplásicos/uso terapéutico , Compuestos Aza/química , Compuestos Aza/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Cristalografía por Rayos X , Humanos , Mutación con Pérdida de Función/efectos de los fármacos , Neoplasias/genética , Dominios Proteicos/efectos de los fármacos , Quinuclidinas/química , Quinuclidinas/uso terapéutico , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/aislamiento & purificación , Proteína p53 Supresora de Tumor/ultraestructura
8.
Adv Sci (Weinh) ; 8(21): e2101936, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34605226

RESUMEN

The neurokinin-1 receptor (NK-1R) antagonists are approved as treatment for chemotherapy-associated nausea and vomiting in cancer patients. The emerging role of the substance P-NK-1R system in oncogenesis raises the possibility of repurposing well-tolerated NK-1R antagonists for cancer treatment. This study reports that human colorectal cancer (CRC) patients with high NK-1R expression have poor survival, and NK-1R antagonists SR140333 and aprepitant induce apoptotic cell death in CRC cells and inhibit CRC xenograft growth. This cytotoxicity induced by treatment with NK-1R antagonists is mediated by induction of endoplasmic reticulum (ER) stress. ER stress triggers calcium release, resulting in the suppression of prosurvival extracellular signal-regulated kinase (ERK)-c-Myc signaling. Along with ER calcium release, one ER stress pathway mediated by protein kinase RNA-like ER kinase (PERK) is specifically activated, leading to increased expression of proapoptotic C/EBP-homologous protein (CHOP). Moreover, NK-1R antagonists enhance the efficacy of chemotherapy by increasing the sensitivity and overcoming resistance to 5-fluorouracil in CRC cells through the induction of sustained ER stress and the consequent suppression of ERK-c-Myc signaling both in vitro and in vivo. Collectively, the findings provide novel mechanistic insights into the efficacy of NK-1R antagonists either as a single agent or in combination with chemotherapy for cancer treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Antagonistas del Receptor de Neuroquinina-1/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Aprepitant/farmacología , Aprepitant/uso terapéutico , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones , Ratones Desnudos , Antagonistas del Receptor de Neuroquinina-1/uso terapéutico , Piperidinas/farmacología , Piperidinas/uso terapéutico , Quinuclidinas/farmacología , Quinuclidinas/uso terapéutico , Tasa de Supervivencia , Trasplante Heterólogo
9.
Br J Cancer ; 125(11): 1523-1532, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34599296

RESUMEN

BACKGROUND: Mutations in p53, identified in 90% of oesophageal squamous cell carcinoma (ESCC), are associated with unfavourable prognosis and chemo-resistance. APR-246 induces apoptosis by restoring transcriptional ability of mutant p53, and may be a promising therapeutic agent to overcome chemo-resistance in ESCC. METHODS: In ESCC cell lines differing in p53 status, we performed in vitro cell viability and apoptosis assays, evaluated reactive oxygen species (ROS) generation, and assessed signal changes by western blot after APR-246 administration with/without chemo-agent. Antitumour effects and signal changes were evaluated in in vivo experiments using xenograft and patient-derived xenograft (PDX) mouse models. RESULTS: APR-246 administration induced significant apoptosis by upregulating p73 and Noxa via ROS induction in ESCC cell lines harbouring p53 missense mutations. Moreover, APR-246 plus chemotherapy exerted combined antitumour effects in ESCC with p53 missense mutations. This effect was also mediated through enhanced ROS activity, leading to massive apoptosis via upregulation of p73 and Noxa. These findings were confirmed by xenograft and PDX models with p53 mutant ESCC. CONCLUSION: APR-246 strongly induced apoptosis by inducing ROS activity and p73-Noxa signalling, specifically in ESCC with p53 missense mutation. This antitumour effect was further enhanced by combination with 5-FU, which we first confirmed in ESCC preclinical model.


Asunto(s)
Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Quinuclidinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proteína Tumoral p73/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Humanos , Ratones , Mutación Missense , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Med Rep ; 24(5)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34468006

RESUMEN

The aim of the present study was to investigate the effect of penehyclidine hydrochloride (PHC) pretreatment on mice with lipopolysaccharide (LPS)­induced acute lung injury (ALI) and its possible underlying mechanisms. Mice were randomly separated into six groups: i) Sham group; ii) LPS group; iii) LPS + PHC group; iv) tumor necrosis factor a­induced protein 8­like protein 2 (TIPE2) group; v) LPS + TIPE2 group; and vi) LPS + TIPE2 + PHC group. The ALI model was induced using LPS through intratracheal injection. The mice received adenovirus gene to induce the overexpression of TIPE2. After mice were sacrificed, lung injury indices were assessed, and arterial blood, bronchoalveolar lavage fluid and lung tissues were collected for subsequent assays. Expression levels of related proteins were detected by using western blotting. It was found that compared with the sham group, the mice treated with LPS showed increased lung injury and dysfunctions of gas exchange. However, these trends were significantly ameliorated in the LPS + PHC group. Evaluation of protein expression in lung tissues showed that the increased expression of nuclear NF­κB p65 and p­c­Jun N­terminal kinase (JNK) induced by LPS were suppressed in the LPS + PHC group and the expression of TIPE2 was increased. The mice that received adenovirus gene to induce TIPE2 overexpression could also showed protective effects compared with the mice in the LPS group. However, the expression of TIPE2 decreased rather than increased in LPS group. In the mice pretreated with PHC, the expression of TIPE2 increased in mice with LPS­induced ALI. To conclude, PHC pretreatment could inhibit the occurrence of inflammation and apoptosis in LPS­induced ALI. This process may be related to the activation of TIPE2 and the inhibition of NF­κB and JNK signaling pathway in the lungs of mice.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipopolisacáridos/efectos adversos , Quinuclidinas/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Inflamación , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pulmón/patología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo
11.
Drugs ; 81(11): 1331-1342, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34292534

RESUMEN

Netupitant/palonosetron (NEPA; Akynzeo®), available in oral and intravenous (IV) formulations, is a fixed-dose combination of the neurokinin 1 (NK1) receptor antagonist netupitant (or the prodrug, fosnetupitant, in the IV formulation) and the second-generation serotonin 3 (5-HT3) receptor antagonist palonosetron. Administered as a single dose, (fos)netupitant/palonosetron (in combination with dexamethasone) is indicated for the prevention of acute and delayed chemotherapy-induced nausea and vomiting (CINV) in adults. In clinical trials, (fos)netupitant/palonosetron plus dexamethasone was associated with high complete response rates (no emesis and no rescue medication) in the acute, delayed and overall phases in patients receiving highly or moderately emetogenic chemotherapy, with efficacy maintained over multiple cycles. Further, oral netupitant/palonosetron was found to be superior to palonosetron and non-inferior to aprepitant plus granisetron in preventing CINV in individual trials. Both the oral and IV formulations of the drug combination are well tolerated. The fixed-dose combination is concordant with guideline recommendations and provides a simple and convenient option for prophylaxis against acute and delayed CINV in patients receiving highly or moderately emetogenic chemotherapy.


Chemotherapy-induced nausea and vomiting (CINV) is a common problem during cancer treatment. Netupitant/palonosetron (NEPA; Akynzeo®) is a fixed-dose combination of two drugs (netupitant, a neurokinin 1 receptor antagonist; and palonosetron, a serotonin 3 receptor antagonist) which target two different signalling pathways involved in the induction of vomiting. Approved for use in the prevention of acute and delayed CINV in adults, netupitant/palonosetron is given orally or via intravenous infusion as a single dose prior to chemotherapy. In clinical trials, high proportions of patients who received netupitant/palonosetron (used in combination with the corticosteroid dexamethasone) prior to chemotherapy reported no vomiting, no requirement for rescue medication, and no significant nausea in the 5 days post chemotherapy. Both the oral and intravenous formulations of the drug combination are well tolerated. In conclusion, netupitant/palonosetron is a simple, convenient and effective drug combination for the prevention of acute and delayed CINV in patients receiving chemotherapy that has a moderate to high potential to cause nausea and vomiting.


Asunto(s)
Antieméticos/uso terapéutico , Isoquinolinas/uso terapéutico , Náusea/tratamiento farmacológico , Piridinas/uso terapéutico , Quinuclidinas/uso terapéutico , Vómitos/tratamiento farmacológico , Administración Intravenosa , Administración Oral , Antieméticos/administración & dosificación , Antieméticos/efectos adversos , Antieméticos/farmacología , Antineoplásicos/efectos adversos , Ensayos Clínicos como Asunto , Dexametasona/uso terapéutico , Combinación de Medicamentos , Interacciones Farmacológicas , Humanos , Isoquinolinas/administración & dosificación , Isoquinolinas/efectos adversos , Isoquinolinas/farmacología , Náusea/inducido químicamente , Piridinas/administración & dosificación , Piridinas/efectos adversos , Piridinas/farmacología , Quinuclidinas/administración & dosificación , Quinuclidinas/efectos adversos , Quinuclidinas/farmacología , Vómitos/inducido químicamente
12.
Cell Death Dis ; 12(7): 709, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34267184

RESUMEN

Asparaginase depletes extracellular asparagine in the blood and is an important treatment for acute lymphoblastic leukemia (ALL) due to asparagine auxotrophy of ALL blasts. Unfortunately, resistance occurs and has been linked to expression of the enzyme asparagine synthetase (ASNS), which generates asparagine from intracellular sources. Although TP53 is the most frequently mutated gene in cancer overall, TP53 mutations are rare in ALL. However, TP53 mutation is associated with poor therapy response and occurs at higher frequency in relapsed ALL. The mutant p53-reactivating compound APR-246 (Eprenetapopt/PRIMA-1Met) is currently being tested in phase II and III clinical trials in several hematological malignancies with mutant TP53. Here we present CEllular Thermal Shift Assay (CETSA) data indicating that ASNS is a direct or indirect target of APR-246 via the active product methylene quinuclidinone (MQ). Furthermore, combination treatment with asparaginase and APR-246 resulted in synergistic growth suppression in ALL cell lines. Our results thus suggest a potential novel treatment strategy for ALL.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Asparaginasa/farmacología , Proliferación Celular/efectos de los fármacos , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Quinuclidinas/farmacología , Proteína p53 Supresora de Tumor/agonistas , Ligasas de Carbono-Nitrógeno con Glutamina como Donante de Amida-N/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
Int J Mol Sci ; 22(11)2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071360

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer lacking targeted therapy. Here, we evaluated the anti-cancer activity of APR-246, a P53 activator, and CX-5461, a RNA polymerase I inhibitor, in the treatment of TNBC cells. We tested the efficacy of individual and combination therapy of CX-5461 and APR-246 in vitro, using a panel of breast cancer cell lines. Using publicly available breast cancer datasets, we found that components of RNA Pol I are predominately upregulated in basal-like breast cancer, compared to other subtypes, and this upregulation is associated with poor overall and relapse-free survival. Notably, we found that the treatment of breast cancer cells lines with CX-5461 significantly hampered cell proliferation and synergistically enhanced the efficacy of APR-246. The combination treatment significantly induced apoptosis that is associated with cleaved PARP and Caspase 3 along with Annexin V positivity. Likewise, we also found that combination treatment significantly induced DNA damage and replication stress in these cells. Our data provide a novel combination strategy by utilizing APR-246 in combination CX-5461 in killing TNBC cells that can be further developed into more effective therapy in TNBC therapeutic armamentarium.


Asunto(s)
Benzotiazoles/farmacología , Daño del ADN , Replicación del ADN/efectos de los fármacos , Naftiridinas/farmacología , Quinuclidinas/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Replicación del ADN/genética , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , ARN Polimerasa I/antagonistas & inhibidores , ARN Polimerasa I/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
15.
Biochem Pharmacol ; 190: 114600, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33992630

RESUMEN

Cynandione A, an acetophenone isolated from Cynanchum Wilfordii Radix, attenuates inflammation. The present study aimed to study the mechanisms underlying cynandione A-induced antiinflammation. Treatment with cynandione A and the specific α7 nicotinic acetylcholine receptor (α7 nAChR) agonist PHA-543613 remarkably reduced overexpression of proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1ß in lipopolysaccharide (LPS)-treated RAW264.7 cells and primary peritoneal macrophages, and endotoxemic mice. Both cynandione A and PHA-543613 also stimulated IL-10 expression in naïve and LPS-treated macrophages and endotoxemic mice. Cynandione A- and PHA-543613-inhibited proinflammatory cytokine expression was completely blocked by the α7 nAChR antagonist methyllycaconitine and the IL-10 antibody. The stimulatory effect of cynandione A and PHA-543613 on IL-10 expression were suppressed by methyllycaconitine and knockdown of α7 nAChRs using siRNA/α7 nAChR. Cynandione A significantly stimulated STAT3 phosphorylation, which was attenuated by methyllycaconitine and the IL-10 neutralizing antibody. The STAT3 activation inhibitor NSC74859 also blocked cynandione A-inhibited proinflammatory cytokine expression. Taken together, our results, for the first time, demonstrate that cynandione A and PHA-543613 inhibit inflammation through macrophageal α7 nAChR activation and subsequent IL-10 expression.


Asunto(s)
Compuestos de Bifenilo/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Mediadores de Inflamación/antagonistas & inhibidores , Interleucina-10/agonistas , Macrófagos/efectos de los fármacos , Quinuclidinas/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Animales , Compuestos de Bifenilo/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Células Cultivadas , Cynanchum , Relación Dosis-Respuesta a Droga , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-10/biosíntesis , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Quinuclidinas/uso terapéutico , Células RAW 264.7 , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
16.
Biomed Pharmacother ; 139: 111571, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33857915

RESUMEN

This study aimed to investigate the effects of various concentrations of cevimelines (CVMs) and compare them with commercial drugs in a murine model of dry eye. The experimental mouse model used male and female NOD.B10.H2b mice over 12 weeks of age. Desiccation stress was performed at 30-40% ambient humidity, and subcutaneous injection of 0.5 mg/0.2 mL scopolamine hydrobromide was performed four times a day for 10 days. The efficacy of various concentrations of CVMs (seven experimental groups) was first evaluated, and then 2% CVM was compared with commercial drugs, such as cyclosporine A (CsA), diquafosol (DQS), and rebamipide (REB) (seven experimental groups). The clinical changes, including tear production, corneal irregularity, and fluorescein staining, were measured after the instillation of various concentrations of CVMs and commercial drugs for 0, 3, 5, 7, and 10 days. Histological changes, such as corneal detachment, conjunctival goblet cell and mucin density staining, were assessed by staining the cornea or conjunctiva with hematoxylin-eosin, periodic acid-Schiff, and alcian blue. The expression of inflammatory markers and mucin factors was detected by immunohistochemistry and immunofluorescence in the lacrimal gland, cornea, and conjunctiva. Tear production was significantly increased in the 2% CVM group and was similar to that in the DQS and REB groups (P < 0.05). The corneal smoothness and fluorescein staining score were significantly improved in the 2% CVM group and were similar to those in the REB group (P < 0.05). Corneal epithelial cells were significantly decreased in the 2% CVM group, with similar observations made in the DQS and REB groups (P < 0.05). The conjunctival goblet cells and mucin density recovered in the 2% CVM group were similar to those in the CsA and REB groups (P < 0.05). The 2% CVM group showed suppressed expression of inflammatory factors in the lacrimal gland and was comparable to that seen in the CsA and REB groups. The expression of mucin factors was upregulated in the cornea and conjunctiva of the 2% CVM group and was similar to that of the CsA and REB groups. In conclusion, administration of CVM resulted in recovery or clinical and histological improvement of the murine dry eye model, and all the observed parameters were comparable to those with commercial drugs.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Córnea/metabolismo , Síndromes de Ojo Seco/tratamiento farmacológico , Mucinas/biosíntesis , Quinuclidinas/uso terapéutico , Tiofenos/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/farmacología , Conjuntiva/patología , Córnea/efectos de los fármacos , Córnea/patología , Síndromes de Ojo Seco/metabolismo , Síndromes de Ojo Seco/patología , Células Epiteliales/efectos de los fármacos , Femenino , Aparato Lagrimal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos NOD , Soluciones Oftálmicas , Quinuclidinas/farmacología , Lágrimas/efectos de los fármacos , Tiofenos/farmacología , Regulación hacia Arriba/efectos de los fármacos
17.
Oncol Rep ; 45(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33846805

RESUMEN

p53­reactivation and induction of massive apoptosis­1, APR­017 methylated (PRIMA­1met; APR246) targets mutant p53 to restore its wild­type structure and function. It was previously demonstrated that PRIMA­1met effectively inhibited the growth of colorectal cancer (CRC) cells in a p53­independent manner, and distinctly induced apoptosis by upregulating Noxa in p53­mutant cell lines. The present study including experiments of western blotting, acridine orange staining and transmission electron microscopy revealed that PRIMA­1met induced autophagy in CRC cells independently of p53 status. Importantly, PRIMA­1met not only promoted autophagic vesicle (AV) formation and AV­lysosome fusion, but also increased lysosomal degradation. Furthermore, Cell Counting Kit­8 assay, colony formation assay and small interfering RNA transfection were performed to investigate the underling mechanisms. The study indicated that activation of the mTOR/AMPK­ULK1­Vps34 autophagic signaling cascade was key for PRIMA­1met­induced autophagy. Additionally, autophagy served a crucial role in the inhibitory effect of PRIMA­1met in cells harboring wild­type p53, which was closely associated with the increased expression of Noxa. Taken together, the results determined the effect of PRIMA­1met on autophagy, and further revealed mechanistic insights into different CRC cell lines. It was concluded that PRIMA­1met­based therapy may be an effective strategy for CRC treatment.


Asunto(s)
Autofagia/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Quinuclidinas/farmacología , Proteína p53 Supresora de Tumor/agonistas , Proteínas Quinasas Activadas por AMP/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mutación , Quinuclidinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
18.
Int J Oncol ; 58(3): 312-330, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33650649

RESUMEN

Glioblastoma is the most malignant brain tumor and presents high resistance to chemotherapy and radiotherapy. Surgery, radiotherapy and chemotherapy with temozolomide are the only treatments against this tumor. New targeted therapies, including epigenetic modulators such as 3­deazaneplanocin A (DZ­Nep; an EZH2 inhibitor) and panobinostat (a histone deacetylase inhibitor) are being tested in vitro, together with temozolomide. The present study combined APR­246 with DZ­Nep, panobinostat and teomozolomide in order to explore the possibility of restoring p53 function in mutated cases of glioblastoma. Following the Chou­Talalay method it was demonstrated that APR­246 acts in an additive manner together with the other compounds, reducing clonogenicity and inducing apoptosis in glioblastoma cells independently of p53 status.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Quinuclidinas/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosina/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/patología , Humanos , Mutación , Panobinostat/farmacología , Panobinostat/uso terapéutico , Quinuclidinas/uso terapéutico , Temozolomida/farmacología , Temozolomida/uso terapéutico , Proteína p53 Supresora de Tumor/genética
19.
Sci Rep ; 11(1): 4480, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33627789

RESUMEN

The prevalence and dire implications of mutations in the tumour suppressor, p53, highlight its appeal as a chemotherapeutic target. We recently showed that impairing cellular antioxidant systems via inhibition of SLC7A11, a component of the system xc- cystine-glutamate antiporter, enhances sensitivity to mutant-p53 targeted therapy, APR-246. We investigated whether this synergy extends to other genes, such as those encoding enzymes of the pentose phosphate pathway (PPP). TKT, one of the major enzymes of the PPP, is allegedly regulated by NRF2, which is in turn impaired by accumulated mutant-p53 protein. Therefore, we investigated the relationship between mutant-p53, TKT and sensitivity to APR-246. We found that mutant-p53 does not alter expression of TKT, nor is TKT modulated directly by NRF2, suggesting a more complex mechanism at play. Furthermore, we found that in p53null cells, knockdown of TKT increased sensitivity to APR-246, whilst TKT overexpression conferred resistance to the drug. However, neither permutation elicited any effect on cells overexpressing mutant-p53 protein, despite mediating oxidative stress levels in a similar fashion to that in p53-null cells. In sum, this study has unveiled TKT expression as a determinant for sensitivity to APR-246 in p53-null cells.


Asunto(s)
Estrés Oxidativo/efectos de los fármacos , Quinuclidinas/farmacología , Transcetolasa/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Antioxidantes/metabolismo , Línea Celular Tumoral , Células HCT116 , Células HEK293 , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción/efectos de los fármacos
20.
BMC Anesthesiol ; 21(1): 49, 2021 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-33581727

RESUMEN

BACKGROUND: Postoperative nausea and vomiting (PONV) is one of the most frequent complications following strabismus surgery. Penehyclidine, an anticholinergic agent, is widely used as premedication. This study investigated the effect of preoperative penehyclidine on PONV in patients undergoing strabismus surgery. METHODS: In this prospective, randomized, double-blind study, patients scheduled for strabismus surgery under general anesthesia were randomly assigned to either penehyclidine (n = 114) or normal saline (n = 104) group. Penehyclidine was administrated immediately after anesthesia induction, and normal saline was substituted as control. PONV was investigated from 0 to 48 h after surgery. Intraoperative oculocardiac reflex (OCR) was also recorded. RESULTS: Compared with normal saline, penehyclidine significantly reduced PONV incidence (30.7% vs. 54.8%, P < 0.01) and mitigated PONV severity as indicated by severity scoring (P < 0.01). Compared with normal saline, penehyclidine also significantly reduced OCR incidence (57.9% vs. 77.9%, P < 0.01) and mitigated OCR severity, as indicated by the requirement for atropine rescue (77.3% vs. 90.1%, P < 0.05) and the maximum decrease of heart rate during OCR (23.1 ± 9.4 bpm vs. 27.3 ± 12.4 bpm, P < 0.05). The recovery course did not differ between groups. CONCLUSIONS: Penehyclidine administrated after anesthesia induction significantly reduced the incidence of PONV and alleviated intraoperative OCR in patients undergoing strabismus surgery. TRIAL REGISTRATION: ClinicalTrials.gov ( NCT04054479 ). Retrospectively registered August 13, 2019.


Asunto(s)
Complicaciones Intraoperatorias/prevención & control , Náusea y Vómito Posoperatorios/prevención & control , Quinuclidinas/farmacología , Reflejo Oculocardíaco/efectos de los fármacos , Estrabismo/cirugía , Adolescente , Adulto , Anciano , Niño , Preescolar , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA