Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Support Care Cancer ; 32(4): 249, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38530439

RESUMEN

OBJECTIVE: Megestrol acetate (MA) is used to manage anorexia and cachexia in patients with advanced cancer. This study investigated the prescription patterns of MA in patients with metastatic gastric cancer, as well as evaluated its impact on survival outcomes and the incidence of venous thromboembolism (VTE). METHODS: A Health Insurance Review and Assessment (HIRA) service database was used to investigate differences in baseline characteristics, survival, and the incidence of VTE according to MA prescription patterns (i.e., prescription vs. no prescription) in patients diagnosed with metastatic gastric cancer from July 2014 to December 2015. RESULTS: A total of 1938 patients were included in this study. In total, 65% of the patients were prescribed MA. Older age, treatment in tertiary hospitals, and palliative chemotherapy were statistically significant predictive factors for MA prescription. Continuous prescription of MA was observed in 37% of patients. There was no statistically significant difference in survival between the MA and non-MA prescription groups on multivariate analysis. Among the 1427 patients included in the analysis for VTE incidence, 4.3% and 2.9% were diagnosed with VTE during the follow-up period in the MA and non-MA prescription groups, respectively. However, there was no statistically significant difference in VTE diagnosis between the groups on multivariate analysis. CONCLUSION: MA is commonly prescribed for metastatic gastric cancer, especially in elderly patients and those undergoing palliative chemotherapy, without significantly affecting survival or VTE risk.


Asunto(s)
Neoplasias Gástricas , Tromboembolia Venosa , Humanos , Anciano , Acetato de Megestrol/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Tromboembolia Venosa/tratamiento farmacológico , Caquexia/etiología , Seguro de Salud , Factores de Transcripción/uso terapéutico , Proteínas de Ciclo Celular/uso terapéutico , Chaperonas de Histonas/uso terapéutico
2.
Cell Genom ; 4(2): 100487, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38278156

RESUMEN

Chemical genetic screens are a powerful tool for exploring how cancer cells' response to drugs is shaped by their mutations, yet they lack a molecular view of the contribution of individual genes to the response to exposure. Here, we present sci-Plex-Gene-by-Environment (sci-Plex-GxE), a platform for combined single-cell genetic and chemical screening at scale. We highlight the advantages of large-scale, unbiased screening by defining the contribution of each of 522 human kinases to the response of glioblastoma to different drugs designed to abrogate signaling from the receptor tyrosine kinase pathway. In total, we probed 14,121 gene-by-environment combinations across 1,052,205 single-cell transcriptomes. We identify an expression signature characteristic of compensatory adaptive signaling regulated in a MEK/MAPK-dependent manner. Further analyses aimed at preventing adaptation revealed promising combination therapies, including dual MEK and CDC7/CDK9 or nuclear factor κB (NF-κB) inhibitors, as potent means of preventing transcriptional adaptation of glioblastoma to targeted therapy.


Asunto(s)
Glioblastoma , Humanos , Glioblastoma/tratamiento farmacológico , Transducción de Señal , Proteínas Tirosina Quinasas Receptoras/uso terapéutico , Quinasas de Proteína Quinasa Activadas por Mitógenos/uso terapéutico , Genómica , Proteínas Serina-Treonina Quinasas , Proteínas de Ciclo Celular/uso terapéutico
3.
Naunyn Schmiedebergs Arch Pharmacol ; 397(2): 1015-1023, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37555854

RESUMEN

Evodiamine (EVO) was tested on acute gouty arthritis rats to investigate its anti-inflammatory effect. Seventy-two male Sprague-Dawley (SD) rats were randomly assigned into the control, model, high, medium, and low dose of EVO groups and colchicine group. The ankle swelling degrees were measured at 2 h, 6 h, and 24 h following sodium urate injection into ankle joint. Histopathological examination was performed 24 h after injection. Reactive oxygen species (ROS) content in the ankle joint was detected using chemical fluorescence. Serum interleukin-1ß (IL-1ß), interleukin-18 (IL-18), and tumor necrosis factor-α (TNF-α) content were determined by ELISA. Serum xanthine oxidase (XOD), superoxide dismutase (SOD), and malondialdehyde (MDA) were determined by spectrophotometry. The expressions of thioredoxin-interacting protein (TXNIP), NOD-like receptor thermal protein domain associated protein 3 (NLRP3), pro-caspase-1, caspase-1, and apoptosis-related spot like protein (ASC) in synovium were detected by Western blot. Evodiamine alleviated the ankle swelling of the affected foot in gouty arthritis rats and reduced inflammatory cell infiltration in joint synovial tissue. Evodiamine also decreased the content of serum inflammatory factors including IL-1ß, IL-18, and TNF-α, and increased serum SOD activity, while it decreased serum XOD, MDA activity, and ROS level. Moreover, evodiamine downregulated the protein expression levels of TXNIP, NLRP3, pro-caspase-1, cleaved caspae-1, and ASC. The mechanism of EVO in treating gouty arthritis is associated with the inhibition of NLRP3 inflammasome by regulating the ROS/TXNIP/NLRP3 signaling pathway.


Asunto(s)
Artritis Gotosa , Proteína con Dominio Pirina 3 de la Familia NLR , Quinazolinas , Ratas , Masculino , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Artritis Gotosa/inducido químicamente , Artritis Gotosa/tratamiento farmacológico , Artritis Gotosa/metabolismo , Interleucina-18/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Caspasa 1/metabolismo , Ratas Sprague-Dawley , Superóxido Dismutasa , Proteínas de Ciclo Celular/uso terapéutico
4.
Biotechnol J ; 18(11): e2300060, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37478121

RESUMEN

Certain cancers, such as triple-negative breast cancer (TNBC), pose a challenging prognosis due to the absence of identifiable hormone-related receptors and effective targeted therapies. Consequently, novel therapeutics are required for these cancers, offering minimal side effects and reduced drug resistance. Unexpectedly, siRNA-7, initially employed as a control, exhibited significant efficacy in inhibiting cell viability in MDA-MB-231 cells. Through a genome-wide search of seed sequences, the targets of siRNA-7 were identified as cancer-related genes, namely PRKCE, RBPJ, ZNF737, and CDC7 in MDA-MB-231 cells. The mRNA repression analysis confirmed the simultaneous suppression by siRNA-7. Combinatorial administration of single-targeting siRNAs demonstrated a comparable reduction in viability to that achieved by siRNA-7. Importantly, siRNA-7 selectively inhibited cell viability in MDA-MB-231 cells, while normal HDF-n cells remained unaffected. Furthermore, in a xenograft mouse model, siRNA-7 exhibited a remarkable 76% reduction in tumor volume without any loss in body weight. These findings position siRNA-7 as a promising candidate for a novel, safe, specific, and potent TNBC cancer therapeutic. Moreover, the strategy of multiple suppressing small interfering RNA holds potential for the treatment of various diseases associated with gene overexpression.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , ARN Interferente Pequeño/genética , Neoplasias de la Mama Triple Negativas/terapia , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Apoptosis , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/farmacología , Proteínas de Ciclo Celular/uso terapéutico
5.
Curr Opin Chem Biol ; 75: 102323, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37207401

RESUMEN

The bromodomain acts to recognize acetylated lysine in histones and transcription proteins and plays a fundamental role in chromatin-based cellular processes including gene transcription and chromatin remodeling. Many bromodomain proteins, particularly the bromodomain and extra terminal domain (BET) protein BRD4 have been implicated in cancers and inflammatory disorders and recognized as attractive drug targets. Although clinical studies of many BET bromodomain inhibitors have made substantial progress toward harnessing the therapeutic potential of targeting the bromodomain proteins, the development of this new class of epigenetic drugs is met with challenges, especially on-target dose-limiting toxicity. In this review, we highlight the current development of new-generation small molecule inhibitors for the BET and non-BET bromodomain proteins and discuss the research strategies used to target different bromodomain proteins for a wide array of human diseases including cancers and inflammatory disorders.


Asunto(s)
Neoplasias , Proteínas Nucleares , Humanos , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Histonas/metabolismo , Neoplasias/metabolismo , Cromatina , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/uso terapéutico
6.
Arch Pharm (Weinheim) ; 356(8): e2200602, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37211680

RESUMEN

Despite cancer research and therapy, breast cancer remains a complicated health crisis in women and represents a top biomedical research priority. Nowadays, breast cancer is an extremely heterogeneous disease and is known as the leading cause of death among women worldwide. The incidence and mortality rates of breast cancer have been increasing gradually for the past decades. Nowadays, common treatments for breast cancer are chemotherapy, endocrine therapy, immunotherapy, radiotherapy, and surgery. The most common targets in breast cancer treatment are human epidermal growth factor receptor 2 (HER2) and estrogen receptors. The literature suggests that several targets/pathways are also involved in the development of breast cancer, that is, poly(ADP-ribose) polymerase (PARP), bromodomain-containing protein 4 (BRD4), cyclin-dependent kinase 4/6 (CDK4/6), epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), polo-like kinase 1 (PLK1), phosphoinositide 3-kinases/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR), histone deacetylase (HDAC), nuclear factor kappa B (NF-κB), PD-L1, and aromatase inhibitors. Meanwhile, the study of breast cancer is a hot topic in the current scenario of basic/clinical research. This review article provides information on different targets associated with breast cancer and summarizes the progress of current research on synthesized inhibitors as anti-breast cancer agents from 2015 to 2021. The review aims to provide structure-activity relationship and docking studies for designing novel compounds for breast cancer therapy.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Femenino , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Nucleares/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Transcripción/uso terapéutico , Relación Estructura-Actividad , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Antineoplásicos/farmacología , Receptor ErbB-2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/uso terapéutico
7.
Mol Omics ; 19(8): 624-639, 2023 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-37232035

RESUMEN

Colorectal cancer (CRC), a common malignant tumour of the gastrointestinal tract, is a life-threatening cancer worldwide. Mutations in KRAS and BRAF, the major driver mutation subtypes in CRC, activate the RAS pathway, contribute to tumorigenesis in CRC and are being investigated as potential therapeutic targets. Despite recent advances in clinical trials targeting KRASG12C or RAS downstream signalling molecules for KRAS-mutant CRC, there is a lack of effective therapeutic interventions. Therefore, understanding the unique molecular characteristics of KRAS-mutant CRC is essential for identifying molecular targets and developing novel therapeutic interventions. We obtained in-depth proteomics and phosphoproteomics quantitative data for over 7900 proteins and 38 700 phosphorylation sites in cells from 35 CRC cell lines and performed informatic analyses, including proteomics-based coexpression analysis and correlation analysis between phosphoproteomics data and cancer dependency scores of the corresponding phosphoproteins. Our results revealed novel dysregulated protein-protein associations enriched specifically in KRAS-mutant cells. Our phosphoproteomics analysis revealed activation of EPHA2 kinase and downstream tight junction signalling in KRAS-mutant cells. Furthermore, the results implicate the phosphorylation site Y378 in the tight junction protein PARD3 as a cancer vulnerability in KRAS-mutant cells. Together, our large-scale phosphoproteomics and proteomics data across 35 steady-state CRC cell lines represent a valuable resource for understanding the molecular characteristics of oncogenic mutations. Our approach to predicting cancer dependency from phosphoproteomics data identified the EPHA2-PARD3 axis as a cancer vulnerability in KRAS-mutant CRC.


Asunto(s)
Neoplasias Colorrectales , Proteínas Proto-Oncogénicas p21(ras) , Humanos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/uso terapéutico , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/uso terapéutico , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/uso terapéutico , Transducción de Señal
8.
Inflammopharmacology ; 31(3): 1511-1527, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36976486

RESUMEN

BACKGROUND: The primary pathogenic factors of Alzheimer's disease (AD) have been identified as oxidative stress, inflammatory damage, and apoptosis. Chrysophanol (CHR) has a good neuroprotective effect on AD, however, the potential mechanism of CHR remains unclear. PURPOSE: In this study, we focused on the ROS/TXNIP/NLRP3 pathway to determine whether CHR regulates oxidative stress and neuroinflammation. METHODS: D-galactose and Aß25-35 combination were used to build an in vivo model of AD, and the Y-maze test was used to evaluate the learning and memory function of rats. Morphological changes of neurons in the rat hippocampus were observed using hematoxylin and eosin (HE) staining. AD cell model was established by Aß25-35 in PC12 cells. The DCFH-DA test identified reactive oxygen species (ROS). The apoptosis rate was determined using Hoechst33258 and flow cytometry. In addition, the levels of MDA, LDH, T-SOD, CAT, and GSH in serum, cell, and cell culture supernatant were detected by colorimetric method. The protein and mRNA expressions of the targets were detected by Western blot and RT-PCR. Finally, molecular docking was used to further verify the in vivo and in vitro experimental results. RESULTS: CHR could significantly improve learning and memory impairment, reduce hippocampal neuron damage, and reduce ROS production and apoptosis in AD rats. CHR could improve the survival rate, and reduce the oxidative stress and apoptosis in the AD cell model. Moreover, CHR significantly decreased the levels of MDA and LDH, and increased the activities of T-SOD, CAT, and GSH in the AD model. Mechanically, CHR significantly reduced the protein and mRNA expression of TXNIP, NLRP3, Caspase-1, IL-1ß, and IL-18, and increase TRX. CONCLUSIONS: CHR exerts neuroprotective effects on the Aß25-35-induced AD model mainly by reducing oxidative stress and neuroinflammation, and the mechanism may be related to ROS/TXNIP/NLRP3 signaling pathway.


Asunto(s)
Enfermedad de Alzheimer , Fármacos Neuroprotectores , Ratas , Animales , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedades Neuroinflamatorias , Simulación del Acoplamiento Molecular , Estrés Oxidativo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Superóxido Dismutasa/metabolismo , ARN Mensajero/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/farmacología , Proteínas de Ciclo Celular/uso terapéutico
9.
Nat Rev Drug Discov ; 22(1): 38-58, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36202931

RESUMEN

Replication stress is a major cause of genomic instability and a crucial vulnerability of cancer cells. This vulnerability can be therapeutically targeted by inhibiting kinases that coordinate the DNA damage response with cell cycle control, including ATR, CHK1, WEE1 and MYT1 checkpoint kinases. In addition, inhibiting the DNA damage response releases DNA fragments into the cytoplasm, eliciting an innate immune response. Therefore, several ATR, CHK1, WEE1 and MYT1 inhibitors are undergoing clinical evaluation as monotherapies or in combination with chemotherapy, poly[ADP-ribose]polymerase (PARP) inhibitors, or immune checkpoint inhibitors to capitalize on high replication stress, overcome therapeutic resistance and promote effective antitumour immunity. Here, we review current and emerging approaches for targeting replication stress in cancer, from preclinical and biomarker development to clinical trial evaluation.


Asunto(s)
Proteínas de Ciclo Celular , Neoplasias , Humanos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/uso terapéutico , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Daño del ADN , Replicación del ADN
10.
Curr Opin Urol ; 32(5): 472-480, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35869742

RESUMEN

PURPOSE OF REVIEW: Many clinical trials are currently underway to target the epigenome of castration-resistant prostate cancer. In this review, we summarize the major epigenetic alterations that occur during prostate cancer progression, describe their biological consequences, and highlight potential of therapies that target epigenetic regulators for use in patients. RECENT FINDINGS: Epigenetic alterations frequently occur in tumour suppressor genes, DNA repair genes, and genes that regulate cell proliferation and differentiation. Unlike genetic alterations, epigenetic changes are reversible, making them promising targets for cancer therapy. Epigenetic regulators can be divided into three broad groups: writers, readers, and erasers , each with specific drug targets that are being assessed in phase I and II clinical trials for prostate cancer. CBP/p300, and BRD4 are coregulators of the androgen receptor and inhibit androgen signalling, making bromodomain extra-terminal inhibitors and CBP/p300 inhibitors attractive targets in prostate cancer. Enhancer of zeste homolog 2, a histone methyltransferase, is also a potential target in castrate-resistant prostate cancer. An emerging direction is to combine epigenetic inhibitors with other compounds to enhance their efficacy. SUMMARY: Preclinical studies indicate that the epigenome is a potential target in prostate cancer, and clinical trials are testing multiple agents that target the epigenome in different ways. However, the process of translating these therapies into the clinic is ongoing and none have yet been approved for castrate-resistant prostate cancer.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/uso terapéutico , Proliferación Celular , Epigénesis Genética , Humanos , Masculino , Proteínas Nucleares/genética , Proteínas Nucleares/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Factores de Transcripción/genética , Factores de Transcripción/uso terapéutico
11.
Hematol Oncol ; 40(3): 417-429, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35544413

RESUMEN

B-cell progenitor fate determinant interferon regulatory factor 4 (IRF4) exerts key roles in the pathogenesis and progression of multiple myeloma (MM), a currently incurable plasma cell malignancy. Aberrant expression of IRF4 and the establishment of a positive auto-regulatory loop with oncogene MYC, drives a MM specific gene-expression program leading to the abnormal expansion of malignant immature plasma cells. Targeting the IRF4-MYC oncogenic loop has the potential to provide a selective and effective therapy for MM. Here we evaluate the use of bromodomain inhibitors to target the IRF4-MYC axis through combined inhibition of their known epigenetic regulators, BRD4 and CBP/EP300. Although all inhibitors induced cell death, we found no synergistic effect of targeting both of these regulators on the viability of MM cell-lines. Importantly, for all inhibitors over a time period up to 72 h, we detected reduced IRF4 mRNA, but a limited decrease in IRF4 protein expression or mRNA levels of downstream target genes. This indicates that inhibitor-induced loss of cell viability is not mediated through reduced IRF4 protein expression, as previously proposed. Further analysis revealed a long half-life of IRF4 protein in MM cells. In support of our experimental observations, gene network modeling of MM suggests that bromodomain inhibition is exerted primarily through MYC and not IRF4. These findings suggest that despite the autofeedback positive regulatory loop between IRF4 and MYC, bromodomain inhibitors are not effective at targeting IRF4 in MM and that novel therapeutic strategies should focus on the direct inhibition or degradation of IRF4.


Asunto(s)
Factores Reguladores del Interferón , Mieloma Múltiple , Proteínas Proto-Oncogénicas c-myc , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/farmacología , Proteínas de Ciclo Celular/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Clin Transl Med ; 12(5): e743, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35522902

RESUMEN

BACKGROUND: To date, 5-fluorouracil-based chemotherapy is very important for locally advanced or metastatic colorectal cancer (CRC). However, chemotherapy resistance results in tumor recurrence and metastasis, which is a major obstacle for treatment of CRC. METHODS: In the current research, we establish 5-fluorouracil resistant cell lines and explore the potential targets associated with 5-fluorouracil resistance in CRC. Moreover, we perform clinical specimen research, in vitro and in vivo experiments and molecular mechanism research, to reveal the biological effects and the mechanism of DCLK1 promoting 5-fluorouracil resistance, and to clarify the potential clinical value of DCLK1 as a target of 5-fluorouracil resistance in CRC. RESULTS: We discover that doublecortin-like kinase 1 (DCLK1), a cancer stem cell maker, is correlated with 5-fluorouracil resistance, and functionally promotes cancer stemness and 5-fluorouracil resistance in CRC. Mechanistically, we elucidate that DCLK1 interacts with cell cycle and apoptosis regulator 1 (CCAR1) through the C-terminal domain, and phosphorylates CCAR1 at the Ser343 site, which is essential for CCAR1 stabilisation. Moreover, we find that DCLK1 positively regulates ß-catenin signalling via CCAR1, which is responsible for maintaining cancer stemness. Subsequently, we prove that blocking ß-catenin inhibits DCLK1-mediated 5-fluorouracil resistance in CRC cells. Importantly, we demonstrate that DCLK1 inhibitor could block CCAR1/ß-catenin pathway-mediated cancer stemness and consequently suppresses 5-fluorouracil resistant CRC cells in vitro and in vivo. CONCLUSIONS: Collectively, our findings reveal that DCLK1 promotes 5-fluorouracil resistance in CRC by CCAR1/ß-catenin pathway-mediated cancer stemness, and suggest that targeting DCLK1 might be a promising method to eliminate cancer stem cells for overcoming 5-fluorouracil resistance in CRC.


Asunto(s)
Neoplasias Colorrectales , Quinasas Similares a Doblecortina , Fluorouracilo , beta Catenina , Proteínas Reguladoras de la Apoptosis/uso terapéutico , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/uso terapéutico , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Quinasas Similares a Doblecortina/genética , Fluorouracilo/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Recurrencia Local de Neoplasia , Proteínas Serina-Treonina Quinasas/genética , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
13.
Cancer Res Commun ; 2(11): 1426-1435, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36970056

RESUMEN

Purpose: We conducted a first-in-human, dose-escalation study, to evaluate the safety, tolerability, pharmacokinetics, pharmacodynamics, and activity of TAK-931, a cell division cycle 7 inhibitor, in Japanese patients with advanced solid tumors. Experimental Design: Patients ages ≥20 years received oral TAK-931: once daily for 14 days in 21-day cycles (schedule A; from 30 mg); once daily or twice daily for 7 days on, 7 days off in 28-day cycles (schedule B; from 60 mg); continuous once daily (schedule D; from 20 mg); or once daily for 2 days on, 5 days off (schedule E; from 100 mg) in 21-day cycles. Results: Of the 80 patients enrolled, all had prior systemic treatment and 86% had stage IV disease. In schedule A, 2 patients experienced dose-limiting toxicities (DLTs) of grade 4 neutropenia and the maximum tolerated dose (MTD) was 50 mg. In schedule B, 4 patients experienced DLTs of grade 3 febrile neutropenia (n = 3) or grade 4 neutropenia (n = 1); the MTD was 100 mg. Schedules D and E were discontinued before MTD determination. The most common adverse events were nausea (60%) and neutropenia (56%). Time to maximum plasma concentration of TAK-931 was approximately 1-4 hours postdose; systemic exposure was approximately dose proportional. Posttreatment pharmacodynamic effects correlating to drug exposure were observed. Overall, 5 patients achieved a partial response. Conclusions: TAK-931 was tolerable with a manageable safety profile. TAK-931 50 mg once daily days 1-14 in 21-day cycles was selected as a recommended phase II dose and achieved proof of mechanism. Trial registration ID: NCT02699749. Significance: This was the first-in-human study of the CDC7 inhibitor, TAK-931, in patients with solid tumors. TAK-931 was generally tolerable with a manageable safety profile. The recommend phase II dose was determined to be TAK-931 50 mg administered once daily on days 1-14 of each 21-day cycle. A phase II study is ongoing to confirm the safety, tolerability, and antitumor activity of TAK-931 in patients with metastatic solid tumors.


Asunto(s)
Neoplasias , Neutropenia , Humanos , Ciclo Celular , Proteínas de Ciclo Celular/uso terapéutico , Neoplasias/tratamiento farmacológico , Neutropenia/inducido químicamente , Proteínas Serina-Treonina Quinasas/uso terapéutico , Pirimidinas/efectos adversos
14.
Stat Med ; 40(29): 6523-6540, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34542190

RESUMEN

With the uptake of targeted therapies, instead of the "one-fits-all" approach, modern randomized controlled trials (RCTs) often aim to develop treatments that target a subgroup of patients. Motivated by analyzing the Age-Related Eye Disease Study (AREDS) data, a large RCT to study the efficacy of nutritional supplements in delaying the progression of an eye disease, age-related macular degeneration (AMD), we develop a simultaneous inference procedure to identify and infer subgroups with differential treatment efficacy in RCTs with time-to-event outcomes. Specifically, we formulate the multiple testing problem through contrasts and construct their simultaneous confidence intervals, which appropriately control both within- and across-marker multiplicity. Realistic simulations are conducted using real genotype data to evaluate the method performance under various scenarios. The method is then applied to AREDS to assess the efficacy of antioxidants and zinc combination in delaying AMD progression. Multiple gene regions including ESRRB-VASH1 on chromosome 14 have been identified with subgroups showing differential efficacy. We further validate our findings in an independent subsequent RCT, AREDS2, by discovering consistent differential treatment responses in the targeted and non-targeted subgroups identified from AREDS. This multiple-testing-based simultaneous inference approach provides a step forward to confidently identify and infer subgroups in modern drug development.


Asunto(s)
Degeneración Macular , Antioxidantes/uso terapéutico , Proteínas de Ciclo Celular/uso terapéutico , Progresión de la Enfermedad , Humanos , Degeneración Macular/tratamiento farmacológico , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
15.
Sci Rep ; 11(1): 1114, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441910

RESUMEN

Neovascularization of the erectile tissue emerges as a beneficial curative approach to treat erectile dysfunction (ED). Here we for the first time report the unexpected role of vasohibin-1 (VASH1), mainly known as an anti-angiogenic factor, in restoring erectile function in diabetic mice. A diabetic patient has lower cavernous VASH1 expression than in the potent man. VASH1 was mainly expressed in endothelial cells. There were significant decreases in cavernous endothelial cell and pericyte contents in VASH1 knockout mice compared with those in wild-type mice, which resulted in impairments in erectile function. Intracavernous injection of VASH1 protein successfully restored erectile function in the diabetic mice (~ 90% of control values). VASH1 protein reinstated endothelial cells, pericytes, and endothelial cell-cell junction proteins and induced phosphorylation of eNOS (Ser1177) in the diabetic mice. The induction of angiogenic factors, such as angiopoietin-1 and vascular endothelial growth factor, is responsible for cavernous angiogenesis and the restoration of erectile function mediated by VASH1. Altogether, these findings suggest that VASH1 is proangiogenic in diabetic penis and is a new potential target for diabetic ED.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/uso terapéutico , Diabetes Mellitus Experimental/complicaciones , Disfunción Eréctil/tratamiento farmacológico , Disfunción Eréctil/metabolismo , Erección Peniana , Pene/metabolismo , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/metabolismo , Animales , Proteínas de Ciclo Celular/administración & dosificación , Diabetes Mellitus Experimental/metabolismo , Regulación hacia Abajo , Células Endoteliales/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Pene/irrigación sanguínea , Pericitos/fisiología , Fosforilación , Proteínas de Uniones Estrechas/metabolismo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
J Nat Med ; 74(4): 777-787, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32666278

RESUMEN

Lung adenocarcinoma (LUAD) is the top prevalent histological kind of lung cancer worldwide. Recent evidences have demonstrated that Sauchinone plays an anticancer role in tumor cell invasion and migration. Therefore, we performed this investigation to explain the potential role of Sauchinone in LUAD as well as the potential mechanism involved. Cell counting kit 8 (CCK-8) and transwell experiments were implemented to measure the proliferative, invasive and migratory abilities of LUAD cells. qRT-PCR and Western blot were performed to detect the transfection efficiency of si-EIF4EBP1s. Additionally, Western blot was also implemented to evaluate the effect of Sauchinone on EIF4EBP1 expression level as well as cell cycle-related proteins. Our findings showed that Sauchinone remarkably suppressed the proliferative ability of LUAD cells in a dose-dependent and time-dependent manner. EIF4EBP1 was a candidate target gene of Sauchinone. EIF4EBP1 expression was increased in LUAD tissues, and its high expression induced a poorer prognosis of LUAD patients. EIF4EBP1 expression was positively associated with cell cycle in LUAD. Sauchinone treatment attenuated EIF4EBP1 expression and cell cycle-related protein levels. Knockdown of EIF4EBP1 repressed the proliferation, invasion and migration of LUAD cells; furthermore, Sauchinone stimulation enforced its inhibitory effect. Meanwhile, the treatment of Sauchinone intensified the arrest of cell cycle induced by EIF4EBP1 knockdown. To sum up, our discovery indicated that Sauchinone exerts an anticancer role through down-regulating EIF4EBP1 and mediating cell cycle in LUAD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/uso terapéutico , Adenocarcinoma del Pulmón/tratamiento farmacológico , Benzopiranos/uso terapéutico , Proteínas de Ciclo Celular/uso terapéutico , Dioxoles/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales/farmacología , Adenocarcinoma del Pulmón/patología , Benzopiranos/farmacología , Proteínas de Ciclo Celular/farmacología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Dioxoles/farmacología , Regulación hacia Abajo , Humanos , Neoplasias Pulmonares/patología , Invasividad Neoplásica , Estudios Prospectivos , Transfección
17.
Zhonghua Zhong Liu Za Zhi ; 41(5): 321-325, 2019 May 23.
Artículo en Chino | MEDLINE | ID: mdl-31137163

RESUMEN

Gastric cancer is the most common gastrointestinal cancer in China. The morbidity and mortality are extremely high and there are significant challenges in the treatment of gastric cancer. Recent studies have shown that the expressions of T lymphocyte subsets vary in the immune microenvironment of gastric cancer patients. T lymphocytes are not only the main effector cells of human cellular immunity, but also the important immunoregulatory cells. T lymphocytes not only reflect the state of the tumor microenvironment, but also closely relate with the prognosis of patients. T lymphocytes play a crucial guiding role in the clinical treatment. Currently, clinical trials related to immunological checkpoint inhibitors are still underway, among which PD-1/PD-L1 monoclonal antibody has been approved for the treatment of gastric cancer. The applications of tumor vaccines and adoptive cell therapies in gastric cancer are also being explored. How to screen patients suitable to immunotherapy, develop the best combination therapy and evaluate the effectiveness of immunotherapy need to be studied and solved.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunidad Celular/inmunología , Neoplasias Gástricas/inmunología , Subgrupos de Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Antígeno B7-H1/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Puntos de Control del Ciclo Celular/inmunología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/inmunología , Proteínas de Ciclo Celular/uso terapéutico , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunoterapia , Receptor de Muerte Celular Programada 1/uso terapéutico , Microambiente Tumoral/efectos de los fármacos
18.
J Gene Med ; 21(7): e3091, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30980444

RESUMEN

Gene therapy is a rapidly emerging remedial route for many serious incurable diseases, such as central nervous system (CNS) diseases. Currently, nucleic acid medicines, including DNAs encoding therapeutic or destructive proteins, small interfering RNAs or microRNAs, have been successfully delivered to the CNS with gene delivery vectors using various routes of administration and have subsequently exhibited remarkable therapeutic efficiency. Among these vectors, non-viral vectors are favorable for delivering genes into the CNS as a result of their many special characteristics, such as low toxicity and pre-existing immunogenicity, high gene loading efficiency and easy surface modification. In this review, we highlight the main types of therapeutic genes that have been applied in the therapy of CNS diseases and then outline non-viral gene delivery vectors.


Asunto(s)
Neoplasias Encefálicas/terapia , Enfermedades del Sistema Nervioso Central/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Neoplasias Encefálicas/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/uso terapéutico , Enfermedades del Sistema Nervioso Central/genética , Genes Transgénicos Suicidas , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/uso terapéutico , Humanos , MicroARNs/genética , MicroARNs/uso terapéutico , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/uso terapéutico
19.
Methods Mol Biol ; 1895: 111-122, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30539533

RESUMEN

Suicide gene therapy is based on the introduction of a foreign gene into tumor cells to sensitize cells to treatment, to convert a nontoxic compound into a lethal drug, or to produce a cytotoxic effect. We have constructed a suicide gene therapy vector that contains resveratrol-responsive CArG elements from the Egr-1 promoter and the GADD45α open reading frame. CArG elements are utilized as a "molecular switch" to drive the expression of GADD45α. When transfected into lung cancer cells, the vector is able to express GADD45α upon resveratrol treatment, and subsequently leads to cell cycle arrest at the G2/M transition. In this chapter, we describe a detailed protocol for vector construction, transfection, cell viability assay, and cell cycle analysis.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Genes Transgénicos Suicidas , Terapia Genética/métodos , Neoplasias/terapia , Proteínas Nucleares/genética , Células A549 , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/fisiopatología , Adenocarcinoma/terapia , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Proteínas de Ciclo Celular/uso terapéutico , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/fisiopatología , Neoplasias Pulmonares/terapia , Neoplasias/tratamiento farmacológico , Neoplasias/fisiopatología , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Proteínas Nucleares/uso terapéutico , Elementos de Respuesta , Resveratrol , Activación Transcripcional
20.
Cancer Sci ; 108(7): 1452-1457, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28498618

RESUMEN

Cell division associated 1 (CDCA1) was screened as an oncogene that is overexpressed on several cancers, including prostate cancer. A highly immunogenic HLA-A*2402-restricted epitope peptide corresponding to part of the CDCA1 protein was also identified. A phase I clinical trial was conducted for patients with castration resistant prostate cancer (CRPC) using a CDCA1 peptide vaccination. Twelve patients having HLA-A*2402 with CRPC after failure of docetaxel chemotherapy were enrolled. They received subcutaneous administration of the CDCA1 peptide as an emulsion with Montanide ISA51VG once a week in a dose-escalation manner (doses of 1.0 or 3.0 mg/body, six patients received each dose). The primary endpoint was safety, and the secondary endpoints were the immunological and clinical responses. Vaccination with CDCA1 peptide was well tolerated without any serious adverse events. Peptide-specific cytotoxic T lymphocyte (CTL) responses using ELISPOT assay and dextramer assay were observed in three patients receiving the 1.0 mg dose and five patients receiving the 3.0 mg dose. The median overall survival time was 11.0 months and specific CTL reacting to CDCA1 peptide were recognized in long-surviving patients. CDCA1-derived peptide vaccine treatment was tolerable and might effectively induce peptide-specific CTLs for CRPC patients. This novel peptide vaccine therapy for CRPC appears promising. (ClinicalTrials.gov number, NCT01225471).


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Proteínas de Ciclo Celular/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/terapia , Anciano , Anciano de 80 o más Años , Ensayo de Immunospot Ligado a Enzimas , Antígeno HLA-A24 , Humanos , Masculino , Persona de Mediana Edad , Linfocitos T Citotóxicos/inmunología , Vacunas de Subunidad/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...