Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
1.
Br J Surg ; 111(1)2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38215239

RESUMEN

BACKGROUND: The aim of this multicentre cohort study was to compare the long-term oncological outcomes of robotic gastrectomy (RG) and laparoscopic gastrectomy (LG) for patients with gastric cancer. METHODS: Patients with gastric cancer who underwent radical gastrectomy by robotic or laparoscopic approaches from 1 March 2010 to 31 December 2018 at 10 high-volume centres in China were selected from institutional databases. Patients receiving RG were matched 1 : 1 by propensity score with patients undergoing LG. The primary outcome was 3-year disease-free survival. Secondary outcomes were overall survival and disease recurrence. RESULTS: Some 2055 patients who underwent RG and 4309 patients who had LG were included. The propensity score-matched cohort comprised 2026 RGs and 2026 LGs. Median follow-up was 41 (i.q.r. 39-58) months for the RG group and 39 (38-56) months for the LG group. The 3-year disease-free survival rates were 80.8% in the RG group and 79.5% in the LG group (log rank P = 0.240; HR 0.92, 95% c.i. 0.80 to 1.06; P = 0.242). Three-year OS rates were 83.9 and 81.8% respectively (log rank P = 0.068; HR 0.87, 0.75 to 1.01; P = 0.068) and the cumulative incidence of recurrence over 3 years was 19.3% versus 20.8% (HR 0.95, 0.88 to 1.03; P = 0.219), with no difference between groups. CONCLUSION: RG and LG in patients with gastric cancer are associated with comparable disease-free and overall survival.


Asunto(s)
Laparoscopía , Levamisol/análogos & derivados , Procedimientos Quirúrgicos Robotizados , Neoplasias Gástricas , Humanos , Resultado del Tratamiento , Estudios de Cohortes , Neoplasias Gástricas/cirugía , Gastrectomía , Puntaje de Propensión , Estudios Retrospectivos , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/cirugía
3.
Acta Haematol ; 146(4): 307-315, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37023729

RESUMEN

INTRODUCTION: Despite the promising clinical trial data regarding programmed death 1 (PD-1) inhibitors in relapsed/refractory classical Hodgkin lymphoma (R/R cHL), there remains a paucity of studies describing the outcomes of patients in a real-world setting, especially for Asian cohort. METHODS: We present a multicenter retrospective analysis of patients with R/R cHL who had failed ≥2 prior lines of therapies and received sintilimab or tislelizumab developed in China (sintilimab or tislelizumab monotherapy) at 3 medical centers from January 2019 to September 2021. Efficacy was evaluated with progression-free survival (PFS), overall survival, duration of response (DOR), best overall response (BOR) including objective response rate (ORR), complete response rate (CRR). Safety data were also recorded. RESULTS: 74 patients were reviewed. The median age was 38 years (range, 14-85 years). The ORR, CRR, and disease control rate were 78.3%, 52.7%, and 91.9%, respectively. The median duration of follow-up was 22 (4-36) months. Four patients (5.4%) died of disease progression. The median PFS and DOR was 22.1 and 23.5 months. BOR as a new emergent endpoint was found to be the only independent prognostic factor for PFS in our study (HR = 6.234, p = 0.005), suggesting this endpoint carries stronger prognostic value over traditional endpoints in the immunotherapy era. 66 (89.2%) patients reported adverse event (AE) with any grade, with the majority of AEs being grade 1 or 2. CONCLUSION: We presented a unique real-life experience and conducted a relatively long follow-up of PD-1 antibodies developed in China for R/R HL patients which confirmed their promising effectiveness and manageable side effects given in real world in an Asian cohort. Even for those who would usually be excluded in most of clinical trials such as elderly or minor patients, anti-PD-1 monotherapy also showed a significant improvement of outcomes. Furthermore, the depth of response seemed to be a more powerful predictive tool in new era, which might serve as a basis for future immune risk-adapted strategies.


Asunto(s)
Enfermedad de Hodgkin , Adulto , Anciano , Humanos , Enfermedad de Hodgkin/tratamiento farmacológico , Enfermedad de Hodgkin/patología , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Inducción de Remisión , Estudios Retrospectivos , Adolescente , Adulto Joven , Persona de Mediana Edad , Anciano de 80 o más Años
4.
Adv Sci (Weinh) ; 10(15): e2300169, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36999833

RESUMEN

Direct photoelectrochemical 2-electron water oxidation to renewable H2 O2 production on an anode increases the value of solar water splitting. BiVO4 has a theoretical thermodynamic activity trend toward highly selective water oxidation H2 O2 formation, but the challenges of competing 4-electron O2 evolution and H2 O2 decomposition reaction need to overcome. The influence of surface microenvironment has never been considered as a possible activity loss factor in the BiVO4 -based system. Herein, it is theoretically and experimentally demonstrated that the situ confined O2 , where coating BiVO4 with hydrophobic polymers, can regulate the thermodynamic activity aiming for water oxidation H2 O2 . Also, the hydrophobicity is responsible for the H2 O2 production and decomposition process kinetically. Therefore, after the addition of hydrophobic polytetrafluoroethylene on BiVO4 surface, it achieves an average Faradaic efficiency (FE) of 81.6% in a wide applied bias region (0.6-2.1 V vs RHE) with the best FE of 85%, which is 4-time higher than BiVO4 photoanode. The accumulated H2 O2 concentration can reach 150 µm at 1.23 V versus RHE under AM 1.5 illumination in 2 h. This concept of modifying the catalyst surface microenvironment via stable polymers provides a new approach to tune the multiple-electrons competitive reactions in aqueous solution.

5.
Ann Surg ; 277(1): e87-e95, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34225299

RESUMEN

OBJECTIVE: A large-scale multicenter retrospective cohort study was conducted to compare the short- and long-term outcomes of robotic gastrectomy (RG) and laparoscopic gastrectomy (LG) for gastric cancer. SUMMARY OF BACKGROUND DATA: RG is being increasingly used worldwide, but data from large-scale multicenter studies on the short- and long-term oncologic outcomes of RG versus LG are limited. The potential benefits of RG compared with LG for gastric cancer remain controversial. METHODS: Data from eligible patients who underwent RG or LG for gastric cancer of 11 experienced surgeons from 7 centers in China between March 2010 and October 2019 were collected. The RG group was matched 1:1 with the LG group by using propensity score matching. The primary outcome was postoperative complications. RESULTS: After propensity score matching, a well-balanced cohort of 3552 patients was included for further analysis. The occurrence of overall complications (12.6% vs 15.2%, P = 0.023) was lower in the RG group than in the LG group. RG was associated with less blood loss (126.8 vs 142.5 mL, P < 0.001) and more retrieved lymph nodes in total (32.5 vs 30.7, P < 0.001) and in suprapancreatic areas (13.3 vs 11.6, P < 0.001).The long-term oncological outcomes were comparable between the two groups. CONCLUSIONS: The results of this multicenter study demonstrate that RG is a safe and effective treatment for gastric cancer when performed by experienced surgeons, although longer operation time and higher costs are still concerns about RG. This study provides evidence suggesting that RG may represent an alternative surgical treatment to LG.


Asunto(s)
Laparoscopía , Procedimientos Quirúrgicos Robotizados , Neoplasias Gástricas , Humanos , Procedimientos Quirúrgicos Robotizados/métodos , Estudios Retrospectivos , Neoplasias Gástricas/cirugía , Resultado del Tratamiento , Gastrectomía/métodos , Complicaciones Posoperatorias/cirugía , China
6.
Int J Biol Sci ; 18(16): 6229-6230, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36439882

RESUMEN

[This corrects the article DOI: 10.7150/ijbs.65255.].

7.
Acta Pharmacol Sin ; 43(12): 3015-3020, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36424452

RESUMEN

Anticancer drug discovery has yielded unprecedented progress in recent decades, resulting in the approval of innovative treatment options for patients and the successful implementation of personalized medicine in clinical practice. This remarkable progress has also reshaped the research scope of pharmacological research. This article, as a tribute to cancer research at Shanghai Institute of Materia Medica in celebration of the institute's 90th birthday, provides an overview of the conceptual revolution occurring in anticancer therapy, and summarizes our recent progress in the development of molecularly targeted therapeutics and exploration of new strategies in personalized medicine. With this review, we hope to provide a glimpse into how antitumor pharmacological researchers have embraced the new era of personalized medicine research and to propose a future path for anticancer drug discovery and pharmacological research.


Asunto(s)
Antineoplásicos , Medicina de Precisión , Humanos , China , Descubrimiento de Drogas , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
8.
BMC Genomics ; 23(1): 467, 2022 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-35751016

RESUMEN

BACKGROUND: T cell acute lymphoblastic leukemia (T-ALL) defines a group of hematological malignancies with heterogeneous aggressiveness and highly variable outcome, making therapeutic decisions a challenging task. We tried to discover new predictive model for T-ALL before treatment by using a specific pipeline designed to discover aberrantly active gene. RESULTS: The expression of 18 genes was significantly associated with shorter survival, including ACTRT2, GOT1L1, SPATA45, TOPAZ1 and ZPBP (5-GEC), which were used as a basis to design a prognostic classifier for T-ALL patients. The molecular characterization of the 5-GEC positive T-ALL unveiled specific characteristics inherent to the most aggressive T leukemic cells, including a drastic shut-down of genes located on the mitochondrial genome and an upregulation of histone genes, the latter characterizing high risk forms in adult patients. These cases fail to respond to the induction treatment, since 5-GEC either predicted positive minimal residual disease (MRD) or a short-term relapse in MRD negative patients. CONCLUSION: Overall, our investigations led to the discovery of a homogenous group of leukemic cells with profound alterations of their biology. It also resulted in an accurate predictive tool that could significantly improve the management of T-ALL patients.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Adulto , Expresión Génica Ectópica , Humanos , Neoplasia Residual/diagnóstico , Neoplasia Residual/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Pronóstico , Linfocitos T/patología , Resultado del Tratamiento
9.
Int J Biol Sci ; 18(1): 30-42, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34975316

RESUMEN

Osteosarcoma (OS) is a malignant bone tumor among adolescents and young adults. IRF7 belongs to the transcription factor family of interferon regulatory factors (IRFs) and has previously been described to function as a tumor suppressor in multiple cancer types. However, the biological functions and cellular mechanism of IRF7 in OS remain elusive. In this study, by quantitative real-time PCR (qRT-PCR) and western blotting, we found that IRF7 was downregulated in OS, and the higher expression of IRF7 was correlated with a better survival prognosis. Moreover, loss-of-function and gain-of-function studies have proved the critical functions of IRF7 in suppressing aerobic glycolysis of osteosarcoma cells as evidenced by glucose uptake, lactate production, extracellular acidification rate, and oxygen consumption rate. Mechanistically, IRF7 inhibited the expression of key glycolytic gene PKM2 via direct transcriptional regulation. Moreover, the in vitro and in vivo tumor-suppressive roles of IRF7 were uncovered in OS and these effects were largely glycolysis-dependent. Therefore, our study unveils a previous unprecedented role of IRF7 in glucose metabolism reprogram and suggests that IRF7 might serve as a potential therapeutic target for patients with OS.


Asunto(s)
Neoplasias Óseas/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Osteosarcoma/metabolismo , Factores de Transcripción/metabolismo , Efecto Warburg en Oncología , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Acta Pharmacol Sin ; 43(1): 209-219, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33782541

RESUMEN

PI3Kδ is expressed predominately in leukocytes and overexpressed in B-cell-related malignances. PI3Kδ has been validated as a promising target for cancer therapy, and specific PI3Kδ inhibitors were approved for clinical practice. However, the substantial toxicity and relatively low efficacy as a monotherapy in diffuse large B-cell lymphoma (DLBCL) limit their clinical use. In this study, we described a novel PI3Kδ inhibitor SAF-248, which exhibited high selectivity for PI3Kδ (IC50 = 30.6 nM) over other PI3K isoforms at both molecular and cellular levels, while sparing most of the other human protein kinases in the kinome profiling. SAF-248 exhibited superior antiproliferative activity against 27 human lymphoma and leukemia cell lines compared with the approved PI3Kδ inhibitor idelalisib. In particular, SAF-248 potently inhibited the proliferation of a panel of seven DLBCL cell lines (with GI50 values < 1 µM in 5 DLBCL cell lines). We demonstrated that SAF-248 concentration-dependently blocked PI3K signaling followed by inducing G1 phase arrest and apoptosis in DLBCL KARPAS-422, Pfeiffer and TMD8 cells. Its activity against the DLBCL cells was negatively correlated to the protein level of PI3Kα. Oral administration of SAF-248 dose-dependently inhibited the growth of xenografts derived from Pfeiffer and TMD8 cells. Activation of mTORC1, MYC and JAK/STAT signaling was observed upon prolonged treatment and co-targeting these pathways would potentiate the activity of SAF-248. Taken together, SAF-248 is a promising selective PI3Kδ inhibitor for the treatment of DLBCL and rational drug combination would further improve its efficacy.


Asunto(s)
Antineoplásicos/farmacología , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Animales , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Inhibidores de las Quinasa Fosfoinosítidos-3/química , Relación Estructura-Actividad
11.
J Colloid Interface Sci ; 606(Pt 2): 1469-1476, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34500151

RESUMEN

A Z-scheme photosystems combining Schottky junction and loading of applicable bandgap semiconductor is beneficial for enhancing the charge carriers' separation/transfer as well as maintain their excellent redox ability. Here, CdxZn1-xS@Au was in-situ deposited on the (010) facets of BiVO4 taking Au as a bridge for constructing a sandwich structure CdxZn1-xS@Au/BiVO4 Z-scheme photocatalyst. The electrons in BiVO4 (010) migrate unidirectionally to Au nanoparticles across the Schottky junction and effectively suppress opposite electrons flow, then be captured by the excited holes in CdxZn1-xS. Furthermore, Zn-doping also contributes to an appropriate redox ability and charge carriers separation. Benefiting from the dual-facilitated effects, the ternary CdxZn1-xS@Au/BiVO4 exhibited superior photocatalytic activity for CO2 reduction under visible light irradiation using H2O as a reducing agent, as compared with CdS and CdS@Au/BiVO4. Furthermore, the intermediate product HCOO* fixed on the surface of CdxZn1-xS@Au/BiVO4 is identified by in-situ FT-IR, playing a key role in the conversion of CO2 to CO and then improve photocatalytic selectivity.

12.
Cell Death Dis ; 12(8): 717, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34285190

RESUMEN

Renal cell carcinoma (RCC) cells have increased lipogenesis and cholesterol synthesis. Sterol regulatory element-binding protein-1 (SREBP1) is cleaved by site 1 protease (S1P) to release the transcriptionally active amino-terminal domain. PF-429242 is a potent and competitive S1P inhibitor. We here tested its activity in RCC cells. In established and primary human RCC cells, PF-429242 potently inhibited cell proliferation, migration, and invasion. The S1P inhibitor provoked apoptosis activation in RCC cells. Furthermore, shRNA-mediated S1P silencing or CRISPR/Cas9-induced S1P knockout led to RCC cell growth inhibition and apoptosis activation. Conversely, ectopic overexpression of SREBP1 or S1P augmented RCC cell proliferation and migration. Daily i.v. injection of a single dose of PF-429242 robustly inhibited RCC xenograft growth in severe combined immunodeficiency mice. Additionally, intratumoral injection of S1P shRNA lentivirus inhibited RCC xenograft growth in mice. SREBP1, S1P, and its target gene low density lipoprotein receptor (LDLR) were significantly elevated in human RCC tissues. These results suggest that targeting S1P by PF-429242 inhibited RCC cell growth in vitro and in vivo.


Asunto(s)
Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Proproteína Convertasas/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Silenciador del Gen/efectos de los fármacos , Humanos , Túbulos Renales/patología , Masculino , Persona de Mediana Edad , Proproteína Convertasas/metabolismo , Pirrolidinas , Serina Endopeptidasas/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Acta Pharmacol Sin ; 42(10): 1653-1664, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33441995

RESUMEN

Rheumatoid arthritis (RA) is characterized by joint leukocyte infiltration, synovial inflammation and bone damage result from osteoclastogenesis. Bruton's tyrosine kinase (BTK) is a key regulator of B cell receptor (BCR) and Fc gamma receptor (FcγR) signaling involved in the pathobiology of RA and other autoimmune disorders. SOMCL-17-016 is a potent and selective tricyclic BTK inhibitor, structurally distinct from other known BTK inhibitors. In present study we investigated the therapeutic efficacy of SOMCL-17-016 in a mouse collagen-induced arthritis (CIA) model and underlying mechanisms. CIA mice were administered SOMCL-17-016 (6.25, 12.5, 25 mg·kg-1·d-1, ig), or ibrutinib (25 mg·kg-1·d-1, ig) or acalabrutinib (25 mg·kg-1·d-1, ig) for 15 days. We showed that oral administration of SOMCL-17-016 dose-dependently ameliorated arthritis severity and bone damage in CIA mice; it displayed a higher in vivo efficacy than ibrutinib and acalabrutinib at the corresponding dosage. We found that SOMCL-17-016 administration dose-dependently inhibited anti-IgM-induced proliferation and activation of B cells from CIA mice, and significantly decreased anti-IgM/anti-CD40-stimulated RANKL expression in memory B cells from RA patients. In RANKL/M-CSF-stimulated RAW264.7 cells, SOMCL-17-016 prevented osteoclast differentiation and abolished RANK-BTK-PLCγ2-NFATc1 signaling. In summary, this study demonstrates that SOMCL-17-016 presents distinguished therapeutic effects in the CIA model. SOMCL-17-016 exerts a dual inhibition of B cell function and osteoclastogenesis, suggesting that it to be a promising drug candidate for RA treatment.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Células B de Memoria/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Agammaglobulinemia Tirosina Quinasa/metabolismo , Animales , Autoanticuerpos/metabolismo , Inflamación/tratamiento farmacológico , Activación de Linfocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos DBA , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Pirimidinas/uso terapéutico , Alcaloides de Pirrolicidina/uso terapéutico , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal/efectos de los fármacos
14.
Acta Pharmacol Sin ; 42(6): 998-1004, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32918045

RESUMEN

The ROS1 fusion kinase is an attractive antitumor target. Though with significant clinical efficacy, the well-known first-generation ROS1 inhibitor (ROS1i) crizotinib inevitably developed acquired resistance due to secondary point mutations in the ROS1 kinase. Novel ROS1is effective against mutations conferring secondary crizotinib resistance, especially G2032R, are urgently needed. In the present study, we evaluated the antitumor efficacy of SAF-189s, the new-generation ROS1/ALK inhibitor, against ROS1 fusion wild-type and crizotinib-resistant mutants. We showed that SAF-189s potently inhibited ROS1 kinase and its known acquired clinically resistant mutants, including the highly resistant G2032R mutant. SAF-189s displayed subnanomolar to nanomolar IC50 values against ROS1 wild-type and mutant kinase activity and a selectivity vs. other 288 protein kinases tested. SAF-189s blocked cellular ROS1 signaling, and in turn potently inhibited the cell proliferation in HCC78 cells and BaF3 cells expressing ROS1 fusion wild-type and resistance mutants. In nude mice bearing BaF3/CD74-ROS1 or BaF3/CD74-ROS1G2032R xenografts, oral administration of SAF-189s dose dependently suppressed the growth of both ROS1 wild-type- and G2032R mutant-driven tumors. In a patient-derived xenograft model of SDC4-ROS1 fusion NSCLC, oral administration of SAF-189s (20 mg/kg every day) induced tumor regression and exhibited notable prolonged and durable efficacy. In addition, SAF-189s was more potent than crizotinib and comparable to lorlatinib, the most advanced ROS1i known against the ROS1G2032R. Collectively, these results suggest the promising potential of SAF-189s for the treatment of patients with the ROS1 fusion G2032R mutation who relapse on crizotinib. It is now recruiting both crizotinib-relapsed and naive ROS1-positive NSCLC patients in a multicenter phase II trial (ClinicalTrials.gov Identifier: NCT04237805).


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Crizotinib/uso terapéutico , Femenino , Humanos , Ratones Desnudos , Mutación , Neoplasias/enzimología , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Nat Commun ; 11(1): 3789, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32709895

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

16.
World J Gastrointest Oncol ; 12(4): 424-434, 2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32368320

RESUMEN

BACKGROUND: Reports in the field of robotic surgery for rectal cancer are increasing year by year. However, most of these studies enroll patients at a relatively early stage and have small sample sizes. In fact, studies only on patients with locally advanced rectal cancer (LARC) and with relatively large sample sizes are lacking. AIM: To investigate whether the short-term outcomes differed between robotic-assisted proctectomy (RAP) and laparoscopic-assisted proctectomy (LAP) for LARC. METHODS: The clinicopathological data of patients with LARC who underwent robotic- or laparoscopic-assisted radical surgery between January 2015 and October 2019 were collected retrospectively. To reduce patient selection bias, we used the clinical baseline characteristics of the two groups of patients as covariates for propensity-score matching (PSM) analysis. Short-term outcomes were compared between the two groups. RESULTS: The clinical features were well matched in the PSM cohort. Compared with the LAP group, the RAP group had less intraoperative blood loss, lower volume of pelvic cavity drainage, less time to remove the pelvic drainage tube and urinary catheter, longer distal resection margin and lower rates of conversion (P < 0.05). However, the time to recover bowel function, the harvested lymph nodes, the postoperative length of hospital stay, and the rate of unplanned readmission within 30 days postoperatively showed no difference between the two groups (P > 0.05). The rates of total complications and all individual complications were similar between the RAP and LAP groups (P > 0.05). CONCLUSION: This retrospective study indicated that RAP is a safe and feasible method for LARC with better short-term outcomes than LAP, but we have to admit that the clinically significant of part of indicators are relatively small in the practical situation.

17.
Acta Pharmacol Sin ; 41(5): 731-732, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32081977

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

18.
Acta Pharmacol Sin ; 41(2): 270-277, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31316177

RESUMEN

KRAS is one of the most important proto-oncogenes. Its mutations occur in almost all tumor types, and KRAS mutant cancer is still lack of effective therapy. Prenyl-binding protein phosphodiesterase-δ (PDEδ) is required for the plasma membrane association and subsequent activation of KRAS oncogenic signaling. Recently, targeting PDEδ has provided new promise for KRAS mutant tumors. However, the therapeutic potential of PDEδ inhibition remains obscure. In this study, we explored how PDEδ inhibition was responded in KRAS mutant cancer cells, and identified KRAS mutant subset responsive to PDEδ inhibition. We first performed siRNA screen of KRAS growth dependency of a small panel of human cancer lines, and identified a subset of KRAS mutant cancer cells that were highly dependent on KRAS signaling. Among these cells, only a fraction of KRAS-dependent cells responded to PDEδ depletion, though KRAS plasma membrane association was effectively impaired. We revealed that the persistent RAF/MEK/ERK signaling seemed responsible for the lack of response to PDEδ depletion. A kinase array further identified that the feedback activation of EPH receptor A2 (EPHA2) accounted for the compensatory activation of RAF/MEK/ERK signaling in these cells. Simultaneous inhibition of EPHA2 and PDEδ led to the growth inhibition of KRAS mutant cancer cells. Together, this study gains a better understanding of PDEδ-targeted therapeutic strategy and suggests the combined inhibition of EPHA2 and PDEδ as a potential therapy for KRAS mutant cancer.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/antagonistas & inhibidores , Neoplasias/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptor EphA2/metabolismo , Línea Celular Tumoral , Humanos , Mutación , Neoplasias/genética , ARN Interferente Pequeño/genética , Transducción de Señal/genética
19.
Cancer Lett ; 459: 145-155, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31173854

RESUMEN

Radiotherapy is one of the standard therapies for esophageal squamous cell carcinoma (ESCC), but the efficacy is far from desirable. Large scale genome sequencing reveals PI3Kα is frequently hyper-activated in ESCC. We found that ESCC cells harboring alterations in PI3K pathway were more resistant to radiation and combination of a clinical PI3Kα-selective inhibitor CYH33 and radiation synergistically inhibited cell proliferation in 14 ESCC cell lines. Radiation induced phosphorylation of FOXO1 and Akt, which sensitized ESCC cells to PI3Kα inhibitors. Both S1PR3 and DNA-PK contributed to radiation-induced Akt phosphorylation, which were revealed to be collectively dependent on PI3Kα. By contrast, constitutively active Akt abrogated the synergism between PI3Kα inhibitors and radiation. PI3Kα inhibition enhanced radiation-induced DNA damage, G2/M arrest and apoptosis. Combination of CYH33 and radiation significantly inhibited the growth of xenografts derived from ESCC patients, which was accompanied with abrogation of radiation-induced phosphorylation of Akt and filtration of M2-like macrophages. Taken together, combination of CYH33 and radiation possesses synergism in ESCC, which provides promising rationale to test this combinatorial regimen in ESCC patients.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/radioterapia , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/radioterapia , Morfolinas/farmacología , Piperazinas/farmacología , Pirroles/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Técnicas de Cocultivo , Daño del ADN , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Proteína Forkhead Box O1/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de la radiación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Células THP-1 , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Acta Pharmacol Sin ; 40(5): 677-688, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30224636

RESUMEN

Pancreatic adenocarcinoma is a highly malignant cancer that often involves a deregulation of c-Myc. It has been shown that c-Myc plays a pivotal role in the regulation of a variety of physiological processes and is involved in early neoplastic development, resulting in poor progression. Hence, suppression of c-Myc overexpression is a potential strategy for pancreatic cancer therapy. CUDC-907 is a novel dual-acting inhibitor of phosphoinositide 3-kinase (PI3K) and histone deacetylase (HDAC). It has shown potential efficiency in patients with lymphoma, multiple myeloma, or thyroid cancer, as well as in solid tumors with c-Myc alterations, but the evidence is lacking for how CUDC-907 regulates c-Myc. In this study, we investigated the effect of CUDC-907 on human pancreatic cancer cells in vitro and in vivo. Our results showed that CUDC-907 potently inhibited the proliferation of 9 pancreatic cancer cell lines in vitro with IC50 values ranging from 6.7 to 54.5 nM. Furthermore, we revealed the antitumor mechanism of CUDC-907 in Aspc-1, PANC-1, and Capan-1 pancreatic cancer cells: it suppressed the HDAC6 subunit, thus downregulating c-Myc protein levels, which was a mode of action distinct from the existing mechanisms. Consistently, the extraordinary antitumor activity of CUDC-907 accompanied by downregulation of c-Myc and Ki67 expression in tumor tissue was observed in a human pancreatic cancer Aspc-1 xenograft nude mouse model in vivo. Our results suggest that CUDC-907 can be a valuable therapeutic option for treating pancreatic adenocarcinoma.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Histona Desacetilasa 6/antagonistas & inhibidores , Morfolinas/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pirimidinas/uso terapéutico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Antígeno Ki-67/metabolismo , Masculino , Ratones Endogámicos BALB C , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...