Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bioorg Chem ; 139: 106711, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37473479

RESUMEN

Polo-like kinase 1 (PLK1) is an attractive therapeutic target for the treatment of tumors, as it is an essential cell-cycle regulator frequently overexpressed in tumor tissues. PLK1 can promote tumor invasion and metastasis, and is often associated with poor prognosis in cancer patients. However, no PLK1 inhibitor has been granted marketing approval until now. Therefore, more potentially promising PLK1 inhibitors need to be investigated. In this study, a series of novel inhibitors targeting PLK1 was designed and optimized derived from a new scaffold. After synthesis and characterization, we obtained the structure-activity relationship and led to the discovery of the most promising compound 30e for PLK1. The antiproliferative activity against HCT116 cells (IC50 = 5 nM versus 45 nM for onvansertib) and the cellular permeability and efflux ratio were significantly improved (PappA→B = 2.03 versus 0.345 and efflux ratio = 1.65 versus 94.7 for 30e and onvansertib, respectively). Further in vivo studies indicated that 30e had favorable antitumor activity with 116.2% tumor growth inhibition (TGI) in comparison with TGI of 43.0% for onvansertib. Furthermore, 30e improved volume of tumor tissue distribution in mice as compared to onvansertib. This initial study on 30e holds promise for further development of an antitumor agent.


Asunto(s)
Neoplasias , Inhibidores de Proteínas Quinasas , Animales , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Proteínas de Ciclo Celular , Proteínas Serina-Treonina Quinasas , Línea Celular Tumoral , Proliferación Celular , Quinasa Tipo Polo 1
2.
Food Chem Toxicol ; 176: 113800, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37100235

RESUMEN

LPM6690061 is a novel compound with 5-HT2A receptor antagonist and inverse agonist activities. To support the clinical trial and marketing application of LPM6690061, a series of pharmacology and toxicology studies have been conducted. In vitro and in vivo pharmacology studies showed that LPM6690061 had high inverse agonism and antagonism activities against human 5-HT2A receptors, and demonstrated significant antipsychotic-like effects in two rat models: the DOI-induced head-twitch model and the MK-801-induced hyperactivity model, which was more effective than the control drug pimavanserin. LPM6690061 did not have detectable side effects on the neurobehavioral activities and respiratory function in rats, or on the ECG or blood pressure in dogs at the doses of 2 and 6 mg/kg. The half maximal inhibitory concentration (IC50) of LPM6690061 for inhibiting hERG current was 1.02 µM. Three in vivo toxicology studies were conducted. In the single dose toxicity study in rats and dogs, the maximum tolerated dose of LPM6690061 was 100 mg/kg. In the 4-week repeat dose toxicity study in rats, the main detectable toxic reactions of LPM6690061 included moderate artery wall hypertrophy, minimal to mild mixed cell inflammation and increased macrophages in the lung, which generally recovered after a 4-week drug withdrawal period. In the 4-week repeat dose toxicity study in dogs, no detectable toxicity was observed. The doses of no-observed-adverse-effect-level (NOAEL) in rats and dogs were 10 mg/kg and 20 mg/kg, respectively. In conclusion, both in vitro and in vivo pharmacological and toxicological studies showed that LPM6690061 was a safe and efficacious 5-HT2A receptor antagonist/inverse agonist which supports the clinical development as a novel antipsychotic drug.


Asunto(s)
Antipsicóticos , Serotonina , Ratas , Humanos , Animales , Perros , Agonismo Inverso de Drogas , Receptor de Serotonina 5-HT2A , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico
3.
Cell Discov ; 9(1): 3, 2023 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-36609558

RESUMEN

SARS-CoV-2 Omicron subvariants have demonstrated extensive evasion from monoclonal antibodies (mAbs) developed for clinical use, which raises an urgent need to develop new broad-spectrum mAbs. Here, we report the isolation and analysis of two anti-RBD neutralizing antibodies BA7208 and BA7125 from mice engineered to produce human antibodies. While BA7125 showed broadly neutralizing activity against all variants except the Omicron sublineages, BA7208 was potently neutralizing against all tested SARS-CoV-2 variants (including Omicron BA.1-BA.5) except Mu. By combining BA7208 and BA7125 through the knobs-into-holes technology, we generated a biparatopic antibody BA7208/7125 that was able to neutralize all tested circulating SARS-CoV-2 variants. Cryo-electron microscopy structure of these broad-spectrum antibodies in complex with trimeric Delta and Omicron spike indicated that the contact residues are highly conserved and had minimal interactions with mutational residues in RBD of current variants. In addition, we showed that administration of BA7208/7125 via the intraperitoneal, intranasal, or aerosol inhalation route showed potent therapeutic efficacy against Omicron BA.1 and BA.2 in hACE2-transgenic and wild-type mice and, separately, effective prophylaxis. BA7208/7125 thus has the potential to be an effective candidate as an intervention against COVID-19.

4.
Biol Trace Elem Res ; 201(2): 1006-1018, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35507137

RESUMEN

The traceability of different cultivation modes is critical for ensuring the commercial viability of high-value Dendrobium officinale. In this study, by means of polarizing microscopy, SEM-EDX, ICP-MS and ICP-AES, the possibility of combining microscopic characteristics, multielemental analysis and multivariate statistical authenticity analysis was realized to determine the origins of the fresh stem and dried stem powder of D. officinale derived from three different cultivation modes from six provinces of China. The microscopic structure, chemical elements on the surface of the main microstructures and concentrations of Ca, K, Ba, Cs, As and Cu varied among specimens derived from different cultivation modes. The fresh stems of D. officinale derived from different cultivation modes can be effectively and quickly identified by various microscopic characteristics and different contents of Ca on the surface of the parenchyma, phloem and xylem. Meanwhile, linear discriminant analysis showed that 98.1% of the dried stem powder samples were correctly classified, and the accuracy of cross-validation was 95.3%. This study facilitated an effective integrated method for determining the traceability of the fresh stem and dried stem powder of D. officinale derived from three different cultivation modes. This approach offers a potential method for identifying the origins of medicinal plants derived from different cultivation modes.


Asunto(s)
Dendrobium , Plantas Medicinales , Dendrobium/química , Polvos , Análisis Discriminante , China
5.
Future Med Chem ; 14(13): 991-1003, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35638444

RESUMEN

Aim: To separate and evaluate 9-cyclopropylmethoxy-dihydrotetrabenazine (13a) and its stereoisomers for their high affinity for vesicular monoamine transporter-2 (VMAT2). Method: Stereoisomers of 13a were separated and configurations were ascertained by chiral chromatography and crystal diffraction combined with 1H-1H NOESY assay. Possible binding modes of eight stereoisomers and VMAT2 were explored by molecular docking assays. The VMAT2 affinity of the stereoisomers, inhibition in vivo and pharmacokinetics in rats were evaluated. Results: Three stereoisomers were obtained: P1, P2 and P3, and all had similar VMAT2 binding modes. P2 [(2R, 3R, 11bR)-13a] showed the highest potential VMAT2 binding activity (Ki = 0.75 nM), decreased locomotor activity in rats and had an oral absolute bioavailability of 92.0%. Conclusion: P2 has good efficacy and pharmacokinetic properties and warrants further development to treat tardive dyskinesia.


Asunto(s)
Moduladores del Transporte de Membrana/farmacología , Tetrabenazina , Proteínas de Transporte Vesicular de Monoaminas , Animales , Simulación del Acoplamiento Molecular , Ratas , Estereoisomerismo , Tetrabenazina/análogos & derivados , Tetrabenazina/química , Tetrabenazina/farmacología , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
6.
Eur J Med Chem ; 234: 114246, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35279606

RESUMEN

Pimavanserin is a selective 5-HT2A receptor antagonist and inverse agonist approved by the FDA in 2016, which is used to treat patients with Parkinson's disease psychosis (PDP). But pimavanserin has potential risk with increasing mortality in elderly patients and also increasing the risk of QT interval prolongation in patients. Therefore, searching for new drugs with high efficacy and low toxicity is urgently needed. Based on the docking study of pimavanserin, a series of novel pimavanserin derivatives (7-1∼7-37) were designed and synthesized. The biological activities were evaluated by cell assays and compound 7-16 exhibited 50-fold higher 5-HT2A receptor antagonist activity (IC50 = 0.54 vs 27.3 nM) and 23-fold higher inverse agonist activity (IC50 = 2.1 vs 50 nM) than pimavanserin. Moreover, 7-16 showed increased potency window between the 5-HT2A and hERG activities than pimavanserin. Furthermore, compound 7-16 demonstrated excellent in vitro and in vivo pharmacokinetics, 4-fold more improvement in functional activity in vivo, and good safety profile. Therefore, compound 7-16 represents a potentially promising candidate as a novel anti-PDP agent that warrants further investigation.


Asunto(s)
Antipsicóticos , Enfermedad de Parkinson , Trastornos Psicóticos , Anciano , Antipsicóticos/uso terapéutico , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Trastornos Psicóticos/tratamiento farmacológico , Receptor de Serotonina 5-HT2A , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Agonistas del Receptor de Serotonina 5-HT2/uso terapéutico , Urea
7.
Front Pharmacol ; 12: 770377, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34950030

RESUMEN

Valbenazine and deutetrabenazine are the only two therapeutic drugs approved for tardive dyskinesia based on blocking the action of vesicular monoamine transporter 2 (VMAT2). But there exist demethylated inactive metabolism at the nine position for both them resulting in low availability, and CYP2D6 plays a major role in this metabolism resulting in the genetic polymorphism issue. 9-trifluoroethoxy-dihydrotetrabenazine (13e) was identified as a promising lead compound for treating tardive dyskinesia. In this study, we separated 13e via chiral chromatography and acquired R,R,R-13e [(+)-13e] and S,S,S-13e [(-)-13e], and we investigated their VMAT2-inhibitory activity and examined the related pharmacodynamics and pharmacokinetics properties using in vitro and in vivo models (+)-13e displayed high affinity for VMAT2 (Ki = 1.48 nM) and strongly inhibited [3H]DA uptake (IC50 = 6.11 nM) in striatal synaptosomes. Conversely, its enantiomer was inactive. In vivo, (+)-13e decreased locomotion in rats in a dose-dependent manner. The treatment had faster, stronger, and longer-lasting effects than valbenazine at an equivalent dose. Mono-oxidation was the main metabolic pathway in the liver microsomes and in dog plasma after oral administration, and glucuronide conjugation of mono-oxidized and/or demethylated products and direct glucuronide conjugation were also major metabolic pathways in dog plasma. O-detrifluoroethylation of (+)-13e did not occur. Furthermore, CYP3A4 was identified as the primary isoenzyme responsible for mono-oxidation and demethylation metabolism, and CYP2C8 was a secondary isoenzyme (+)-13e displayed high permeability across the Caco-2 cell monolayer, and it was not a P-glycoprotein substrate as demonstrated by its high oral absolute bioavailability (75.9%) in dogs. Thus, our study findings highlighted the potential efficacy and safety of (+)-13e in the treatment of tardive dyskinesia. These results should promote its clinical development.

8.
Eur J Med Chem ; 224: 113718, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34329999

RESUMEN

Vesicular monoamine transporter 2 (VMAT2) is essential for synaptic transmission of all biogenic amines in the brain including serotonin, norepinephrine, histamine, and dopamine (DA). Given its crucial role in the neurophysiology and pharmacology of the central nervous system, VMAT2 is recognized as an important therapeutic target for various neurological disorders such as tardive dyskinesia (TD). Here, a novel series of dihydrotetrabenazine derivative analogs were designed and synthesized to evaluate their effects on [3H]dihydrotetrabenazine (DTBZ) binding and [3H]DA uptake at VMAT2. Of these analogs, compound 13e showed a high binding affinity for VMAT2 (IC50 = 5.13 ± 0.16 nM) with excellent inhibition of [3H]DA uptake (IC50 = 6.04 ± 0.03 nM) in striatal synaptosomes. In human liver microsomes, 13e was more stable (T1/2 = 161.2 min) than other reported VMAT2 inhibitors such as DTBZ (T1/2 = 119.5 min). In addition, 13e effectively inhibited the spontaneous locomotor activity (percent inhibition at 3 µmol/kg = 64.7%) in Sprague-Dawley rats. Taken together, our results indicate that 13e might be a promising lead compound for the development of novel treatments of TD.


Asunto(s)
Tetrabenazina/análogos & derivados , Proteínas de Transporte Vesicular de Monoaminas/antagonistas & inhibidores , Animales , Humanos , Tetrabenazina/metabolismo , Tetrabenazina/farmacología , Tetrabenazina/uso terapéutico
9.
Commun Biol ; 4(1): 500, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893388

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the ongoing COVID-19 pandemic, which has resulted in more than two million deaths at 2021 February . There is currently no approved therapeutics for treating COVID-19. The SARS-CoV-2 Spike protein is considered a key therapeutic target by many researchers. Here we describe the identification of several monoclonal antibodies that target SARS-CoV-2 Spike protein. One human antibody, CA521FALA, demonstrated neutralization potential by immunizing human antibody transgenic mice. CA521FALA showed potent SARS-CoV-2-specific neutralization activity against SARS-CoV-2 pseudovirus and authentic SARS-CoV-2 infection in vitro. CA521FALA also demonstrated having a long half-life of 9.5 days in mice and 9.3 days in rhesus monkeys. CA521FALA inhibited SARS-CoV-2 infection in SARS-CoV-2 susceptible mice at a therapeutic setting with virus titer of the lung reduced by 4.5 logs. Structural analysis by cryo-EM revealed that CA521FALA recognizes an epitope overlapping with angiotensin converting enzyme 2 (ACE2)-binding sites in SARS-CoV-2 RBD in the Spike protein. CA521FALA blocks the interaction by binding all three RBDs of one SARS-CoV-2 spike trimer simultaneously. These results demonstrate the importance for antibody-based therapeutic interventions against COVID-19 and identifies CA521FALA a promising antibody that reacts with SARS-CoV-2 Spike protein to strongly neutralize its activity.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , COVID-19/prevención & control , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Enzima Convertidora de Angiotensina 2/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , COVID-19/epidemiología , COVID-19/virología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Pandemias , Unión Proteica/efectos de los fármacos , Receptores Virales/inmunología , Receptores Virales/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo
10.
Int J Immunopathol Pharmacol ; 34: 2058738420950584, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32962460

RESUMEN

INTRODUCTION: Indoleamine 2,3-dioxygenase (IDO) was a potential tumor immunotherapy target. IDO inhibitors showed inconsistent results in clinical trials, but no preclinical comparative study was reported. The purpose of this study was to evaluate the differences of representative IDO inhibitors (PCC0208009, INCB024360, NLG919) from the pharmacological perspective. METHODS: In vitro experiments included: inhibition effects on IDO activity in cell and enzyme-based assay, effects on IDO expression in HeLa cells, and enhancement of proliferation and activation of peripheral blood mononuclear cell (PBMC). In vivo experiments included: pharmacokinetics and tumor distribution in CT26-bearing mice, effects on Kyn/Trp and anti-tumor effect and immunological mechanism in CT26 and B16F10 tumor-bearing mice. RESULTS: Compared with INCB024360 and NLG919, PCC0208009 effectively inhibited IDO activity at lower dose 2 nM and longer duration more than 72 h, had higher enhancements on PBMC proliferation and activation, and could inhibit the IDO expression in Hela cells. The pharmacokinetics characteristics of three IDO inhibitors were similar in CT26-bearing mice. In CT26 and B16F10 tumor-bearing mice, PCC0208009 and INCB024360 had similar effects in Kyn/Trp reduction, and more potent than NLG919; three IDO inhibitors had similar effects in tumor suppression, changes of the percentages of CD3+CD8+ and CD3+CD4+ T cells, and activation of tumor infiltrating lymphocytes, while PCC0208009 had a better tendency than INCB024360 and NLG919. CONCLUSION: PCC0208009, INCB024360, and NLG919 were all effective IDO inhibitors, but the comprehensive pharmacological activity of PCC0208009 was better than INCB024360 and NLG919, which was basically consistent with the results or progresses of clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Isoindoles/farmacología , Neoplasias/tratamiento farmacológico , Oximas/farmacología , Sulfonamidas/farmacología , Tetrazoles/farmacología , Animales , Antineoplásicos/farmacocinética , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacocinética , Células HeLa , Humanos , Imidazoles/farmacocinética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Isoindoles/farmacocinética , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/enzimología , Leucocitos Mononucleares/inmunología , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/enzimología , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/enzimología , Neoplasias/inmunología , Neoplasias/patología , Oximas/farmacocinética , Sulfonamidas/farmacocinética , Tetrazoles/farmacocinética , Distribución Tisular , Carga Tumoral/efectos de los fármacos
11.
Toxicol Appl Pharmacol ; 398: 115019, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32335126

RESUMEN

The non-receptor tyrosine phosphatase SHP2, encoded by PTPN11, plays an indispensable role in tumors driven by oncogenic KRAS mutations, which frequently occur in colorectal cancer. Here, PCC0208023, a potent SHP2 allosteric inhibitor, was synthesized to evaluate its inhibitory effects against the SHP2 enzyme, and the KRAS mutant colorectal cancer in vitro and in vivo, and its impart on the RAS/MAPK pathway. Consistent with an allosteric mode of inhibition, PCC0208023 can non-competitively inhibit the activity of full-length SHP2 enzyme, but lacks activity against the free catalytic domain of SHP2. Furthermore, PCC0208023 inhibited the proliferation of KRAS mutation-driven human colorectal cancer cells by inhibiting the RAS/MAPK signaling pathway in vitro. Importantly, PCC0208023 displayed good anti-tumor efficacy against KRAS-driven LS180 and HCT116 xenograft models in nude mice with the decreased Ki67 and p-ERK level, and increased cleaved caspase-3 expression in tumors. Interestingly, PCC0208023 maintained high levels in LS180 tumors within 24 h after administration and was mainly distributed in both intestines and lungs. Molecular docking studies revealed a higher affinity of PCC0208023 with key residues in the SHP2 allosteric pocket than RMC-4550. PCC0208023 deserves further optimization to identify additional low-toxic and potent SHP2 allosteric inhibitors with novel scaffolds for the treatment of patients with KRAS mutation-positive colorectal cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Mutación/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Femenino , Células HCT116 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación/genética , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
12.
PLoS One ; 15(3): e0228339, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32214351

RESUMEN

The increased PD-L1 expression induces poorer prognosis in melanoma. The small molecule inhibitors of PD-1/PD-L1 pathways have been an encouraging drug development strategy because of good affinity and oral bioavailability without immunogenicity and immunotoxicities of PD-1/PD-L1 antibodies. In this study, we studied the effects of PCC0208025 (BMS202), a small molecule inhibitor of PD-L1, on PD-1/PD-L1 binding and the cytokines secretion in human CD3+ cells in vitro. We also investigated the antitumor and immunomodulatory activity of PCC0208025 and the pharmacokinetics properties in B16-F10 melanoma-bearing mice. The results showed that PCC0208025 inhibited the PD-1/PD-L1 proteins binding, and rescued PD-L1-mediated inhibition of IFN-γ production in human CD3+ T cells in vitro. Furthermore, in B16-F10 melanoma-bearing mice, PCC0208025 presented the antitumor effects, enhanced IFN-γ levels in plasma, increased the frequency of CD3+CD8+ T and CD8+IFN-γ+ T and the ratios of CD8+/Treg, and deceased the CD4+CD25+CD127low/- (Treg) number in tumor. Pharmacokinetics study found that PCC0208025 was absorbed and distributed into the tumors with much higher concentrations than those of the blockade against PD-1/PD-L1 binding. Our work suggests that PCC0208025 exhibited anti-tumor effects through inhibiting Treg expansion and increasing cytotoxic activity of tumor-infiltrating CD8+ T cells by the blockade of PD-1/PD-L1 binding, which may provide the pharmacological basis to develop small molecule inhibitors of PD-1/PD-L1 binding for PCC0208025 as a lead compound.


Asunto(s)
Acetamidas/farmacología , Antineoplásicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Melanoma Experimental/tratamiento farmacológico , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Acetamidas/farmacocinética , Acetamidas/uso terapéutico , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Humanos , Factores Inmunológicos/farmacocinética , Factores Inmunológicos/farmacología , Factores Inmunológicos/uso terapéutico , Interferón gamma/metabolismo , Masculino , Melanoma Experimental/patología , Ratones , Piridinas/farmacocinética , Piridinas/uso terapéutico , Bibliotecas de Moléculas Pequeñas/farmacocinética , Bibliotecas de Moléculas Pequeñas/uso terapéutico
13.
Biochem Pharmacol ; 177: 113926, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32217098

RESUMEN

BACKGROUND AND PURPOSE: Indoleamine 2, 3-dioxygenase 1 (IDO1) has been linked to neuropathic pain and IDO1 inhibitors have been shown to reduce pain in animals. Some studies have indicated that IDO1 expression increased after neuropathic pain in hippocampus and spinal cord, whether these changes existing in anterior cingulate cortex (ACC) and amygdala remains obscure and how IDO1 inhibition leads to analgesia is largely unknown. Here, we evaluated the antinociceptive effect of PCC0208009, an indirect IDO1 inhibitor, on neuropathic pain and examined the related neurobiological mechanisms. EXPERIMENTAL APPROACH: The effects of PCC0208009 on pain, cognition and anxiogenic behaviors were evaluated in a rat model of neuropathic pain. Motor disorder, sedation and somnolence were also assessed. Biochemical techniques were used to measure IDO1-mediated signaling changes in ACC and amygdala. KEY RESULTS: In rats receiving spinal nerve ligation (SNL), IDO1 expression level was increased in ACC and amygdala. PCC0208009 attenuated pain-related behaviors in the formalin test and SNL model and increased cognition and anxiogenic behaviors in SNL rats at doses that did not affect locomotor activity and sleeping. PCC0208009 inhibited IDO1 expression in ACC and amygdala by inhibiting the IL-6-JAK2/STAT3-IDO1-GCN2-IL-6 pathway. In addition, PCC0208009 reversed synaptic plasticity at the functional and structural levels by suppressing NMDA2B receptor and CDK5/MAP2 or CDK5/Tau pathway in ACC and amygdala. CONCLUSION AND IMPLICATIONS: These results support the role of IDO1-mediated molecular mechanisms in neuropathic pain and suggest that the IDO1 inhibitor PCC0208009 demonstrates selective pain suppression and could be a useful pharmacological therapy for neuropathic pain.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Analgésicos/uso terapéutico , Giro del Cíngulo/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Neuralgia/tratamiento farmacológico , Plasticidad Neuronal/efectos de los fármacos , Tetrazoles/uso terapéutico , Analgésicos/farmacocinética , Animales , Conducta Animal/efectos de los fármacos , Comorbilidad , Modelos Animales de Enfermedad , Formaldehído/farmacología , Hiperalgesia/tratamiento farmacológico , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Neuralgia/metabolismo , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Tetrazoles/farmacocinética
14.
Int J Immunopathol Pharmacol ; 33: 2058738419843366, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30968715

RESUMEN

Poor prognosis is associated with melanoma due to immunosuppression profiles, suggesting that immune alterations have an important role in the occurrence, growth, and metastasis of melanoma. Here, we found that PCC0208018, a small-molecule compound, enhanced T cell proliferation and activation to release interferon gamma (IFN-γ) and interleukin-2 (IL-2) without blocking the programmed cell death 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) binding and did not directly affect tumor cell viability in vitro. Furthermore, PCC0208018 increased the phosphorylation of protein kinase B (PKB/AKT) as well as extracellular regulated protein kinases (ERK) in human peripheral blood mononuclear cells (PBMCs) in vitro. The secretion of cytokines induced by PCC0208018 was significantly suppressed by the PI3K inhibitor GDC-0941. In B16-F10 melanoma-harboring mice, PCC0208018 significantly inhibited tumor growth as well as increasing CD3+, CD3+CD4+, and CD3+CD8+ T cell abundance in tumors without affecting PD-L1 expression. This study showed that PCC0208018 potentially increased PBMCs proliferation and function by activating the phosphatidylinositol 3 kinase (PI3K)/AKT and mitogen-activated protein kinase (MEK)/ERK pathways to exert antitumor effects.


Asunto(s)
Antineoplásicos/farmacología , Melanoma Experimental/metabolismo , Neoplasias Cutáneas/metabolismo , Linfocitos T/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Células CHO , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Masculino , Melanoma Experimental/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Neoplasias Cutáneas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Xenobiotica ; 49(11): 1338-1343, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30507339

RESUMEN

1. S-EPA is a sulfur-substitution analog of epacadostat (EPA), an effective small molecule indoleamine 2,3-dioxgenase1 (IDO) inhibitor. By in vitro and in vivo experiments, pharmacokinetic differences of two closely related analogs, S-EPA and EPA was investigated in this study. 2. Liver microsomes clearance experiments showed S-EPA had comparable metabolic stability with EPA in rat and human liver microsomes. The whole blood distribution experiments showed the distribution ratio of S-EPA in blood cells to plasma in mice, rats, dogs and monkey was 1.2, 4.8, 2.2 and 40.6, respectively. While the distribution ratio of EPA ranged from 0.94 to 1.30 in mice, rats, dogs and was 3.1 in monkeys. 3. The pharmacokinetic study in rats showed the exposure (AUClast) of S-EPA in plasma and blood cells was 1.7-fold and 3.9-fold higher than that of EPA, respectively. Moreover, the exposure ratio of S-EPA in blood cells to plasma was 3.7, while the ratio of EPA was 1.6. 4. In CT26 tumor bearing mice, the IDO inhibition of S-EPA and EPA on plasma or tumor kynurenine was generally consistent. And the inhibition ratio could reach at more than 50% at 3 h after single dose, at least lasting up to 8 h.


Asunto(s)
Inhibidores Enzimáticos/farmacocinética , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Oximas/farmacocinética , Sulfuros/farmacocinética , Sulfonamidas/farmacocinética , Animales , Línea Celular Tumoral , Perros , Estabilidad de Medicamentos , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/química , Humanos , Quinurenina/sangre , Quinurenina/metabolismo , Macaca fascicularis , Ratones Endogámicos BALB C , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Trasplante de Neoplasias , Oximas/sangre , Oximas/química , Ratas Sprague-Dawley , Sulfuros/sangre , Sulfuros/química , Sulfonamidas/sangre , Sulfonamidas/química
16.
Bioanalysis ; 10(16): 1335-1344, 2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-30130131

RESUMEN

AIM: Tryptophan (Trp) and kynurnine (Kyn) are a pair of biomarkers for indoleamine 2,3-dioxygenase which closely related to the tumor immune escape. To evaluate the effect of drugs on the indoleamine 2,3-dioxygenase activity, the specific and accurate LC-MS/MS methods were developed and validated for simultaneous determination Kyn and Trp in mouse plasma and tumor tissues using surrogate analytes Kyn-d4 and Trp-d5 calibrators. RESULTS: Plasma and tumor homogenates samples were pretreated with the solid phase extraction which assured the method having high recovery (>90% in plasma and >80% in tumor) and no matrix effect. The methods were validated for specificity, linearity, accuracy and precision, recovery, matrix effect and stability using surrogate analytes Kyn-d4 and Trp-d5 in authentic matrices. CONCLUSION: The validated methods have been successfully applied to the pharmacodynamic study of INCB024360 in CT26 tumor bearing mice after single dose and multiple dosing.


Asunto(s)
Análisis Químico de la Sangre/métodos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Quinurenina/sangre , Neoplasias/sangre , Triptófano/sangre , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Cromatografía Liquida , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Quinurenina/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Espectrometría de Masas en Tándem , Triptófano/metabolismo
17.
Int J Immunopathol Pharmacol ; 32: 2058738418787991, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29993291

RESUMEN

Indoleamine 2,3-dioxygenase (IDO), which is highly expressed in human glioblastoma and involved in tumor immune escape and resistance to chemotherapy, is clinically correlated with tumor progression and poor clinical outcomes, and is a promising therapeutic target for glioblastoma. IDO inhibitors are marginally efficacious as single-agents; therefore, combination with other therapies holds promise for cancer therapy. The aim of this study was to investigate the anti-tumor effects and mechanisms of the IDO inhibitor PCC0208009 in combination with temozolomide. The effects of PCC0208009 on IDO activity inhibition, and mRNA and protein expression in HeLa cells were observed. In the mouse glioma GL261 heterotopic model, the effects of PCC0208009 on l-kynurenine/tryptophan (Kyn/Trp), tumor growth, flow cytometry for T cells within tumors, and immunohistochemistry for IDO and Ki67 were examined. In the rat glioma C6 orthotopic model, animal survival, flow cytometry for T cells within tumors, and immunohistochemistry for proliferating cell nuclear antigen (PCNA) and IDO were examined. The results show that PCC0208009 is a highly effective IDO inhibitor, not only directly inhibiting IDO activity but also participating in the gene regulation of IDO expression at the transcription and translation levels. PCC0208009 significantly enhanced the anti-tumor effects of temozolomide in GL261 and C6 models, by increasing the percentages of CD3+, CD4+, and CD8+ T cells within tumors and suppressing tumor proliferation. These findings indicate that PCC0208009 can potentiate the anti-tumor efficacy of temozolomide and suggest that combination of IDO inhibitor-based immunotherapy with chemotherapy is a potential strategy for brain tumor treatment.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/enzimología , Glioma/enzimología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Temozolomida/farmacología , Tetrazoles/farmacología , Animales , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimioterapia Combinada , Glioma/tratamiento farmacológico , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Masculino , Ratones Endogámicos C57BL , Ratas Sprague-Dawley , Temozolomida/uso terapéutico , Tetrazoles/uso terapéutico , Transcripción Genética/efectos de los fármacos
18.
Int J Immunopathol Pharmacol ; 30(3): 215-226, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28604143

RESUMEN

Indoleamine 2,3-dioxygenase (IDO) is involved in tumor immune escape and resistance to chemotherapy, and is clinically correlated with tumor progression. IDO inhibitors show marginal efficacy as single agents; therefore, combinations of these inhibitors with other therapies hold promise for cancer therapy. The aim of this study was to investigate the synergistic antitumor effects of IDO inhibitor NLG919 in combination with different regimens of paclitaxel in a murine B16-F10 melanoma model. NLG919 increased the cytotoxic activity of paclitaxel toward B16-F10 cells in the presence of pretreatment with interferon (IFN)-γ in vitro. In B16-F10 tumor-bearing mice, NLG919 was uniformly distributed throughout tumors and decreased kynurenine levels and kynurenine/tryptophan ratios in tumors and plasma for 6-12 h. NLG919 suppressed tumor growth in a dose-dependent manner and exhibited maximum efficacy at 100 mg/kg. In combination with different regimens of paclitaxel, NLG919 displayed synergistic antitumor effects, and NLG919 did not increase the side effects of paclitaxel. Within the tumors, the percentage of CD3+, CD8+, and CD4+ T cells and secretion of IFN-γ and interleukin-2 were synergistically increased, whereas the percentage of CD4+CD25+ regulatory T cells was decreased. NLG919 can potentiate the antitumor efficacy of paclitaxel without increasing its side effects, suggesting that the combination of IDO inhibitor-based immunotherapy with chemotherapy could be a potential strategy for cancer treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Imidazoles/uso terapéutico , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Isoindoles/uso terapéutico , Melanoma Experimental/tratamiento farmacológico , Paclitaxel/uso terapéutico , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Quimioterapia Combinada , Imidazoles/farmacocinética , Imidazoles/farmacología , Interferón gamma/inmunología , Interleucina-2/inmunología , Isoindoles/farmacocinética , Isoindoles/farmacología , Quinurenina/sangre , Quinurenina/metabolismo , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Paclitaxel/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Triptófano/sangre , Triptófano/metabolismo , Carga Tumoral/efectos de los fármacos
19.
Tumour Biol ; 36(3): 2143-53, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25398692

RESUMEN

The purposes of this study are to investigate the antitumor activities of NSK-01105, a novel sorafenib derivative, in in vitro and in vivo models, and explore the potential mechanisms. The effects of NSK-01105 on proliferation and apoptosis of prostate cancer cells were established by cytotoxicity assays, apoptosis analysis, flow cytometry analysis, and Western blot analysis. Two xenograft tumor models were used to verify the therapeutic effect of NSK-01105 in vivo. NSK-01105 exhibited broad-spectrum antitumor activity, particularly in prostate cancer cells. Characterization of apoptosis morphology was observed, and the percentage of apoptosis-positive cells significantly increased after NSK-01105 treatment for 24 h. Furthermore, a significant increase of the "sub-G1" population in LNCaP and PC-3 cells after NSK-01105 treatment was determined by cell cycle analysis. Tumor growth was significantly suppressed by once daily oral 30 mg/kg dose of NSK-01105 with the inhibition rates of 63.82% in LNCaP models and 64.29% in PC-3 models, respectively. The activation of Raf-1 kinase and epidermal growth factor receptor was downregulated by NSK-01105 at 10 µmol/L. Consequently, the dual inhibitions of Raf/MEK/ERK and PI3K/Akt/mTOR signal pathways were observed by Western blot analysis. Collectively, our results suggest a role of NSK-01105 in treatment for human prostate tumors by inhibiting cell proliferation and inducing apoptosis. NSK-01105 appears to be a promising orally active anticancer drug and deserves further investigation.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HCT116 , Células HT29 , Células Hep G2 , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Células MCF-7 , Masculino , Niacinamida/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Sorafenib , Serina-Treonina Quinasas TOR/metabolismo
20.
PLoS One ; 9(12): e114688, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25506928

RESUMEN

Lx2-32c is a novel taxane that has been demonstrated to have robust antitumor activity against different types of tumors including several paclitaxel-resistant neoplasms. Since the delivery vehicles for taxane, which include cremophor EL, are all associated with severe toxic effects, liposome-based Lx2-32c has been developed. In the present study, the pharmacokinetics, biodistribution, antitumor efficacy and safety characteristics of liposome-based Lx2-32c were explored and compared with those of cremophor-based Lx2-32c. The results showed that liposome-based Lx2-32c displayed similar antitumor effects to cremophor-based Lx2-32c, but with significantly lower bone marrow toxicity and cardiotoxicity, especially with regard to the low ratio of hypersensitivity reaction. In comparing these two delivery modalities, targeting was superior using the Lx2-32c liposome formulation; it achieved significantly higher uptake in tumor than in bone marrow and heart. Our data thus suggested that the Lx2-32c liposome was a novel alternative formulation with comparable antitumor efficacy and a superior safety profiles to cremophor-based Lx2-32c, which might be related to the improved pharmacokinetic and biodistribution characteristics. In conclusion, the Lx2-32c liposome could be a promising alternative formulation for further development.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Paclitaxel/análogos & derivados , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Liposomas , Masculino , Ratones Endogámicos C57BL , Paclitaxel/administración & dosificación , Paclitaxel/farmacocinética , Paclitaxel/uso terapéutico , Paclitaxel/toxicidad , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...