Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
World J Clin Oncol ; 15(3): 391-410, 2024 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-38576597

RESUMO

BACKGROUND: Ferroptosis has recently been associated with multiple degenerative diseases. Ferroptosis induction in cancer cells is a feasible method for treating neoplastic diseases. However, the association of iron proliferation-related genes with prognosis in HER2+ breast cancer (BC) patients is unclear. AIM: To identify and evaluate fresh ferroptosis-related biomarkers for HER2+ BC. METHODS: First, we obtained the mRNA expression profiles and clinical information of HER2+ BC patients from the TCGA and METABRIC public databases. A four-gene prediction model comprising PROM2, SLC7A11, FANCD2, and FH was subsequently developed in the TCGA cohort and confirmed in the METABRIC cohort. Patients were stratified into high-risk and low-risk groups based on their median risk score, an independent predictor of overall survival (OS). Based on these findings, immune infiltration, mutations, and medication sensitivity were analyzed in various risk groupings. Additionally, we assessed patient prognosis by combining the tumor mutation burden (TMB) with risk score. Finally, we evaluated the expression of critical genes by analyzing single-cell RNA sequencing (scRNA-seq) data from malignant vs normal epithelial cells. RESULTS: We found that the higher the risk score was, the worse the prognosis was (P < 0.05). We also found that the immune cell infiltration, mutation, and drug sensitivity were different between the different risk groups. The high-risk subgroup was associated with lower immune scores and high TMB. Moreover, we found that the combination of the TMB and risk score could stratify patients into three groups with distinct prognoses. HRisk-HTMB patients had the worst prognosis, whereas LRisk-LTMB patients had the best prognosis (P < 0.0001). Analysis of the scRNA-seq data showed that PROM2, SLC7A11, and FANCD2 were significantly differentially expressed, whereas FH was not, suggesting that these genes are expressed mainly in cancer epithelial cells (P < 0.01). CONCLUSION: Our model helps guide the prognosis of HER2+ breast cancer patients, and its combination with the TMB can aid in more accurate assessment of patient prognosis and provide new ideas for further diagnosis and treatment.

2.
Eur J Neurosci ; 59(10): 2836-2849, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38488530

RESUMO

Glioblastoma multiforme (GBM) represents the deadliest form of brain tumour, characterized by its low survival rate and grim prognosis. Cytokines released from glioma-associated microglia/macrophages are involved in establishing the tumour microenvironment, thereby crucially promoting GBM progression. MS4A6A polymorphism was confirmed to be associated with neurodegenerative and polymorphism disease pathobiology, but whether it participates in the regulation of GBM and the underlying mechanisms is still not elucidated. Here, we found that MS4A6A was significantly upregulated in GBM patient samples. The results from the single-cell RNA-sequencing (scRNA-seq) database and immunostaining demonstrated the specific expression of MS4A6A in microglial cells. In vitro, microglial overexpression of MS4A6A stimulated the proliferation and migration of glioblastoma cells. Moreover, high MS4A6A mRNA expression was related to poor prognosis in GBM patients. Our study highlights the potential of MS4A6A as a promising biomarker for GBM, which may provide novel strategies for its prevention, diagnosis and treatment.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Microglia , Humanos , Glioblastoma/genética , Glioblastoma/metabolismo , Microglia/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Movimento Celular , Prognóstico , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/genética
4.
Ultraschall Med ; 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38052240

RESUMO

PURPOSE: To investigate the feasibility of deep learning radiomics (DLR) based on multimodal ultrasound to differentiate the primary cancer sites of metastatic cervical lymphadenopathy (CLA). MATERIALS AND METHODS: This study analyzed 280 biopsy-confirmed metastatic CLAs from 280 cancer patients, including 54 from head and neck squamous cell carcinoma (HNSCC), 58 from thyroid cancer (TC), 92 from lung cancer (LC), and 76 from gastrointestinal cancer (GIC). Before biopsy, patients underwent conventional ultrasound (CUS), ultrasound elastography (UE), and contrast-enhanced ultrasound (CEUS). Based on CUS, DLR models using CUS, CUS+UE, CUS+CEUS, and CUS+UE+CEUS data were developed and compared. The best model was integrated with key clinical indicators selected by univariate analysis to achieve the best classification performance. RESULTS: All DLR models achieved similar performance with respect to classifying four primary tumor sites of metastatic CLA (AUC:0.708~0.755). After integrating key clinical indicators (age, sex, and neck level), the US+UE+CEUS+clinical model yielded the best performance with an overall AUC of 0.822 in the validation cohort, but there was no significance compared with the basal CUS+clinical model (P>0.05), both of which identified metastasis from HNSCC, TC, LC, and GIC with 0.869 and 0.911, 0.838 and 0.916, 0.750 and 0.610, and 0.829 and 0.769, respectively. CONCLUSION: The ultrasound-based DLR model can be used to classify the primary cancer sites of metastatic CLA, and the CUS combined with clinical indicators is adequate to provide a high discriminatory performance. The addition of the combination of UE and CEUS data is expected to further improve performance.

5.
Curr Drug Deliv ; 2023 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-37491853

RESUMO

BACKGROUND: Tumor-associated macrophages (TAMs) are crucial for non-small cell lung cancer (NSCLC) development. OBJECTIVE: In this study, polylactic acid-co-glycolic acid (PLGA)-polyethylenimine (PEI) nanobubbles (NBs) carrying STAT6 siRNA were prepared and combined with ultrasound-mediated nanobubbles destruction (UMND) to silence the STAT6 gene, ultimately repolarizing TAMs from the M2 to the M1 phenotype, treating NSCLC in vitro. METHODS: PLGA-PEI NBs-siRNA were prepared and characterised, and their respective ultrasound imaging, biological stabilities and cytotoxicities were detected. Transfection efficiency was evaluated by fluorescence microscopy and flow cytometry. Repolarization of THP-1-derived M2-like macrophages was determined by qPCR and flow cytometry. NSCLC cells (A549) were co-cultured with transfected M2-like macrophages or their associated conditioned medium (CM). Western blotting was used to detect STAT6 gene silencing in M2-like macrophages and markers of epithelial and mesenchymal in A549 cells. The proliferation of A549 cells was detected using CCK-8 and cell colony formation assays. Transwell assays were used to detect the migration and invasion of A549 cells. RESULTS: PLGA-PEI NBs-siRNA had an average size of 223.13±0.92nm and a zeta potential of about -5.59±0.97 mV. PLGA-PEI NBs showed excellent ultrasonic imaging capability in addition to biological stability to protect siRNA from degradation. UMND enhanced PLGA-PEI NBs-STAT6 siRNA transfection in M2-like macrophages, which made M2-like macrophages repolarize to M1-like macrophages and prevented proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) in A549 cells. CONCLUSION: UMND enhanced PLGA-PEI NBs-STAT6 siRNA to repolarize TAMs from the M2 to the M1 phenotype, thus treating NSCLC. These findings provide a promising therapeutic approach for enhancing NSCLC immunotherapy.

6.
Bioconjug Chem ; 34(6): 1157-1165, 2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37235785

RESUMO

Triple-negative breast cancer (TNBC) remains the most challenging breast cancer subtype due to its lack of targeted therapies and poor prognosis. In order to treat patients with these tumors, efforts have been made to explore feasible targets. Epidermal growth factor receptor (EGFR)-targeted therapy is currently in clinical trials and regarded to be a promising treatment strategy. In this study, an EGFR-targeting nanoliposome (LTL@Rh2@Lipo-GE11) using ginsenoside Rh2 as a wall material was developed, in which GE11 was used as the EGFR-binding peptide to deliver more ginsenoside Rh2 and luteolin into TNBC. In comparison to non-targeted liposomes (Rh2@Lipo and LTL@Rh2@Lipo), the nanoliposomes LTL@Rh2@Lipo-GE11 demonstrated a high specificity to MDA-MB-231 cells that expressed a high level of EGFR both in vitro and in vivo, contributing to the strong inhibitory effects on the growth and migration of TNBC. These results suggest that LTL@Rh2@Lipo-GE11 is a prospective candidate for targeted therapy of TNBC, with a remarkable capability to inhibit tumor development and metastasis.


Assuntos
Ginsenosídeos , Neoplasias de Mama Triplo Negativas , Humanos , Lipossomos/uso terapêutico , Neoplasias de Mama Triplo Negativas/metabolismo , Receptores ErbB/metabolismo , Ginsenosídeos/farmacologia , Ginsenosídeos/uso terapêutico , Linhagem Celular Tumoral
7.
Biomater Sci ; 11(5): 1677-1691, 2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36625328

RESUMO

Epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) is a first-line targeted drug for the treatment of advanced non-small cell lung cancer (NSCLC) in clinical practice, but EGFR-TKI-acquired resistance limits its therapeutic effect. To address this challenge, a novel multifunctional gold-based targeted nanoparticle-based drug delivery system is fabricated. The gold-based nanoparticle is loaded with the EGFR-TKI (gefitinib) and IR780, and the surface-modified gold nanoshell layer has a photothermal effect for thermally triggered drug release. Finally, the unique binding of cyclic arginine-glycine-aspartic acid (cRGD) to the αvß3 receptor ensured that the nanoparticle (cRGD-GIPG) targeted transport into drug-resistant NSCLC cells was functional. Due to the sonodynamic properties of IR780, ultrasound (US) irradiation promoted reactive oxygen species (ROS) generation, while low-temperature photothermal therapy (PTT) not only promoted the release of drug, but also further enhanced the cytotoxic effects of ROS. In turn, it blocked the activation of TGF-ß/PDLIM5/SMAD resistance pathway and induced apoptosis of drug-resistant cells through mitochondrial apoptosis, enabling the treatment of EGFR-TKI-resistant NSCLC. The low-temperature PTT combined with sonodynamic therapy (SDT) by cRGD-GIPG thus shows potent anticancer activity against EGFR-TKI-resistant NSCLC cells in vitro and in vivo. The present work provides a valuable strategy for highly targeted and EGFR-TKI-resistant reversal therapy in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Nanopartículas , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Ouro/farmacologia , Terapia Fototérmica , Espécies Reativas de Oxigênio , Temperatura , Receptores ErbB/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Resistencia a Medicamentos Antineoplásicos , Linhagem Celular Tumoral
8.
Eur J Pharm Sci ; 182: 106372, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36621614

RESUMO

Epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKIs) was one of the main drugs in the treatment of non-small cell lung cancer (NSCLC). Previous studies had demonstrated that PDZ and LIM Domain 5 (PDLIM5) played an important role in EGFR TKIs resistance. However, there was no feasible method to eliminate EGFR TKIs resistance by suppressing this gene. Here, we formulated a novel mesoporous silica-loaded PDLIM5 siRNA (Small interfering RNA) nanoplatforms. The results have shown that PDLIM5 siRNA could be effectively bound to the nanoplatforms and had good biocompatibility. Further exploration suggested that the nano-platform combined with ultrasonic irradiation could be very effective for siRNA delivery and ultrasound imaging. Moreover, Epithelial-mesenchymal transformation (EMT) changes occurred in PC-9 Gefitinib resistance (PC-9/GR) cells during the development of drug resistance. When PDLIM5 siRNA entered PC-9/GR cells, the sensitivity of drug-resistant cells to gefitinib could be restored through the transforming growth factor-ß (TGF-ß)/EMT pathway. Therefore, PDLIM5 may be an important reason for the resistance of NSCLC cells to gefitinib, and this nanoplatform may become a novel treatment for EGFR TKIs resistance in NSCLC patients.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Gefitinibe/farmacologia , Gefitinibe/uso terapêutico , RNA Interferente Pequeno/genética , Neoplasias Pulmonares/metabolismo , Transição Epitelial-Mesenquimal , Receptores ErbB , Quinazolinas , Resistencia a Medicamentos Antineoplásicos , Ultrassonografia , Linhagem Celular Tumoral , Inibidores de Proteínas Quinases/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/uso terapêutico , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Proteínas com Domínio LIM/farmacologia
9.
Biomaterials ; 291: 121868, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36332286

RESUMO

Triple-negative breast cancer (TNBC) is a form of breast cancer that is more aggressive and harder to treat than others, with a higher probability of relapse. Its nefarious capabilities for migrating and invading other parts of the body together with the current lack of clinically established effective therapies account for a low survival rate. In this work, we demonstrate the in-tandem use of two complementary therapeutic routes to effectively combat TNBC. A versatile magnetic-photothermal converter (MPC) consisting of zinc-doped ferrite nanoparticles and polyethene glycol, is shown to display excellent therapeutic efficiency, being capable to fight TNBC via two distinct routes: magneto-mechanical force (MMF) and near-infrared-II (NIR-II) hypothermal ablation. The combined use of these two complementary and synergistic therapies, which are less aggressive to the human body compared to conventional chemotherapeutic approaches, results in the splendid suppression of TNBC migration and invasion. Remotely controlling the MPCs by an external magnetic field, results in cellular MMF effects that cause direct mechanical destruction to the cancer cell membrane, leading to its necrosis. Furthermore, the MMF disrupts intracellular lysosomes, thereby triggering the release of large amounts of protein hydrolases, which induce intracellular oxidative stress, and accelerate the induction of apoptosis. Complementing the therapeutic approach based on MMF, the excellent photothermal performance of the MPC in the NIR-II region (1064 nm) is exploited to enable effective hypothermal ablation of the tumours, which can be achieved in deep tissue layers. The proposed multifunctional nanocomposites, together with the demonstrated "double-punch" therapeutic approach, hold significant potential to pave the way for future cutting-edge weapons against the dreadful TNBC.


Assuntos
Nanopartículas , Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/patologia , Fototerapia/métodos , Linhagem Celular Tumoral , Recidiva Local de Neoplasia
10.
Cancer Cell Int ; 22(1): 293, 2022 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-36154921

RESUMO

BACKGROUND: Ultrasound-targeted microbubble destruction (UTMD) technology is a new drug and gene delivery strategy. This study investigates novel ultrasound (US) sensitive siRNA-loaded nanobubbles (siRNA-NBs) to explore the relationship between PDLIM5 mediated autophagy and drug resistance development using epidermal growth factor tyrosine kinase inhibitors (EGFR-TKIs) in the treatment of non-small cell lung cancer (NSCLC). METHODS: US sensitive siRNA-NBs were designed to inhibit the expression of PDLIM5 in gefitinib-resistant human NSCLC PC9GR cells in vitro. The expression of autophagy-related proteins (P62 and LC3-II/I) and autophagosomes in PC9GR cells after PDLIM5 gene silencing were explored. RESULTS: US-sensitive PDLIM5-targeted siRNA-NBs were effectively delivered into PC9GR cells, inhibiting PDLIM5 expression, increasing LC3-II/I and p62 expressions and increasing autophagosomes in PC9GR cells in vitro. CONCLUSIONS: Using UTMD, US-sensitive siRNA-NBs have the potential as an ideal delivery vector to mediate highly effective RNA interference for NSCLC cells. Furthermore, PDLIM5 plays a role in the autophagy-mediated resistance in gefitinib-resistant PC9GR cells.

11.
Comput Math Methods Med ; 2022: 1809186, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35572834

RESUMO

The study was aimed at exploring the diagnostic value of artificial intelligence reconstruction algorithm combined with CT image parameters on hepatic ascites, expected to provide a reference for the etiological evaluation of clinical abdominal effusion. Specifically, the adaptive iterative hard threshold (AIHT) algorithm for CT image reconstruction was proposed. Then, 100 patients with peritoneal effusion were selected as the research subjects. After 8 cases were excluded, the remaining was divided into 50 cases of the S1 group (hepatic ascites) and 42 cases of the D0 group (cancerous peritoneal effusion). Gemstone energy spectrum CT scanning was performed on all patients, and CT image parameters of the two groups were compared. It was found that CT value of mixed energy, CT value of 60-100 KeV single energy, concentration value of water (calcium), concentration value of water (iodine), and slope of energy spectrum curve in the S1 group were significantly lower than those in the D0 group (P < 0.05). The effective atomic number in the S1 group was significantly higher than that in the D0 group (P < 0.05). Of the 50 patients in the S1 group, 3 (6%) had an ascending and 47 (94%) had a descending spectral curve. Of the 42 patients in the D0 group, 37 (88.1%) had an ascending and 5 (11.9%) had a descending spectral curve. The sensitivity and specificity of water (iodine) were 0.927 and 0.836, respectively. The sensitivity and specificity of water (calcium) were 0.863 and 0.887, respectively. For different scan ranges ([0,90]; [0,120]), root mean square error (RMSE) of AIHT reconstructed image was significantly smaller than that of traditional algorithm, while peak signal-to-noise ratio (PSNR) was opposite. The differences were statistically significant (P < 0.05). In conclusion, AIHT-based CT images can better display the distribution of hepatic ascites, and the parameters of CT value, effective atomic number, water (iodine), water (calcium), and spectral curve can all provide help for the identification of hepatic ascites. Especially, water (iodine) and water (calcium) demonstrated high diagnostic performance of hepatic ascites.


Assuntos
Inteligência Artificial , Iodo , Algoritmos , Ascite/diagnóstico por imagem , Cálcio , Humanos , Interpretação de Imagem Radiográfica Assistida por Computador/métodos , Tomografia Computadorizada por Raios X/métodos , Água
12.
Jpn J Radiol ; 40(6): 595-606, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35064441

RESUMO

OBJECTIVES: To establish a preoperative nomogram based on the multimodal ultrasonographic features and biopsy results of primary lesion to predict the risk of sentinel lymph node metastasis (SLNM) in patients with T1-2N0 breast cancer. METHODS: This study included 114 patients with T1-2N0 breast cancer who underwent ultrasound-guided core needle biopsy and multimodal ultrasound (Gray scale, Elastography, and Contrast-enhanced ultrasound) preoperatively. The pathological results of SLN were obtained from sentinel lymph node biopsy. Factors associated with sentinel lymph node metastasis were studied. RESULTS: The regression analysis identified the maximum diameter of tumor (p = 0.003), Doppler resistive index (p = 0.030), HER-2 status (p = 0.016) and the extended range of enhancement lesion (p = 0.010), which were used to establish a nomogram. The prediction model indicated that the value of area under the receiver-operating characteristic curve was 0.798. The calibration curve revealed that the nomogram possesses an excellent consistency between the predicted value and the actual value of SLNM (Hosmer-Lemeshow test: p = 0.436). CONCLUSIONS: The preoperative nomogram can effectively guide clinicians in predicting SLNM of breast cancer, and assist management of breast cancer patients through intuitive risk values to develop personalized treatment strategies.


Assuntos
Neoplasias da Mama , Linfadenopatia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/patologia , Neoplasias da Mama/cirurgia , Feminino , Humanos , Linfonodos/patologia , Metástase Linfática/diagnóstico por imagem , Nomogramas , Curva ROC , Fatores de Risco
13.
Int J Pharm ; 613: 121412, 2022 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-34942327

RESUMO

Ultrasound is one of the safest and most advanced medical imaging technologies that is widely used in clinical practice. Ultrasound microbubbles, traditionally used for contrast-enhanced imaging, are increasingly applied in Ultrasound-targeted Microbubble Destruction (UTMD) technology which enhances tissue and cell membrane permeability through cavitation and sonoporation, to result in a promising therapeutic gene/drug delivery strategy. Here, we review recent developments in the application of UTMD-mediated gene and drug delivery in the diagnosis and treatment of tumors, including the concept, mechanism of action, clinical application status, and advantages of UTMD. Furthermore, the future perspectives that should be paid more attention to in this field are prospected.


Assuntos
Neoplasias , Preparações Farmacêuticas , Técnicas de Transferência de Genes , Terapia Genética , Humanos , Microbolhas , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico
14.
In Vitro Cell Dev Biol Anim ; 57(10): 987-997, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34888748

RESUMO

Gastric cancer is a kind of malignant tumor in the world. Emerging studies have proved the regulatory role of nucleoporin 37 in the development of several malignant tumors. However, the potential effect of NUP37 in gastric cancer is still unclear. In this study, we searched for the Cancer Genome Atlas analysis to explore the potential correlation between NUP37 and gastric cancer. Then, we analyzed NUP37 expression in gastric cancer tissues and cell lines. After constructing a NUP37-silenced model in NCI-N87 cells and a NUP37-overexpressed model in MKN45 cells, we evaluated the role of NUP37 in cell proliferation, migration, and invasion as well as its underlying mechanism. TCGA analysis showed that NUP37 expression was highly expressed in stomach adenocarcinoma, which showed a lower survival rate than normal samples. Moreover, NUP37 was found to be highly expressed in gastric cancer tissues and cell lines. Functionally, NUP37 deficiency promoted gastric cancer cell apoptosis and inhibited cell proliferation, migration, and invasion, whereas NUP37 overexpression exhibited the opposite results. Mechanically, upregulation of NUP37 activated the PI3K/AKT/mTOR signaling pathway. Furthermore, the rescue assay exhibited that the mTOR inhibitor rapamycin significantly reversed the promoting effect of NUP37 in cell proliferation, migration, and invasion. In conclusion, our study identified that NUP37 promoted malignant behavior of gastric cancer cells including invasion, proliferation, and migration through activating the PI3K and its downregulated signaling pathway, indicating that NUP37 might become a novel prognostic target for further gastric cancer therapy.


Assuntos
Complexo de Proteínas Formadoras de Poros Nucleares , Neoplasias Gástricas , Humanos , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Regulação Neoplásica da Expressão Gênica , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Serina-Treonina Quinases TOR/metabolismo
15.
Balkan Med J ; 38(6): 331-340, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34860160

RESUMO

BACKGROUND: Previous studies have published the promoting effect of serum and glucocorticoid-regulated kinase 1 (SGK1) in various malignant tumors. However, whether SGK1 promotes gastric cancer remains a mystery. AIMS: To clarify the function of SGK1 in gastric cancer and its potential regulatory mechanism. STUDY DESIGN: Cell culture study. METHODS: The SGK1-silenced model was generated in two gastric cancer cell lines and further evaluated their malignant behavior and susceptibility to cisplatin. The interaction between miR-15a-5p and SGK1 was evaluated by the luciferase reporter assay. The knockdown efficiency of SGK1 was confirmed by RT- qPCR and Western blot assays. Cell proliferation rate was assessed with CCK-8 assay, and flow cytometry was used to determine cell cycle progression and apoptosis. RESULTS: Western blot data displayed an elevated level of SGK1 in gastric cancer cell lines. Functionally, SGK1 deficiency suppressed gastric cancer cell proliferation (P < .01) by acting on cell-cycle progression. Moreover, SGK1 deficiency suppressed cell invasion and migration of gastric cancer cells (P < .01). Further, the silencing of SGK1 obviously suppressed cell proliferation and induced apoptosis of the cells after cisplatin treatment (P < .01), indicating that SGK1 deficiency facilitated the chemosensitivity of these 2 gastric cancer cell lines to cisplatin. Mechanically, downregulation of SGK1 repressed the cytoplasm- to-nucleus translocation of NF-κB p65. Interestingly, we found that miR-15a-5p binds to the 3'UTR of SGK1, which was confirmed using luciferase activity assay (P < .05). Moreover, the data suggested that SGK1 reversed the suppression effect of miR-15a-5p on gastric cancer cell migration (P < .01). CONCLUSION: Loss of SGK1 suppresses the malignant behavior of gastric cancer cells and increases cisplatin sensitivity by restraining the NF-κB signaling pathway. Moreover, SGK1 may exert an inhibitory effect in gastric cancer by being targeted by miR-15a-5p. Therefore, SGK1 may be a prospective target for future gastric cancer therapy.


Assuntos
Cisplatino/farmacologia , Proteínas Imediatamente Precoces/sangue , NF-kappa B/genética , Proteínas Serina-Treonina Quinases/sangue , Neoplasias Gástricas/tratamento farmacológico , Western Blotting , Cisplatino/uso terapêutico , Glucocorticoides , Humanos , MicroRNAs/genética , Estudos Prospectivos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Neoplasias Gástricas/sangue , Neoplasias Gástricas/genética
16.
BMC Cancer ; 21(1): 657, 2021 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-34078310

RESUMO

BACKGROUND: Long non-coding RNAs exert vital roles in several types of cancer. The objective of this study was to explore the role of LINC_00355 in gastric cancer (GC) progression and its potential mechanism. METHODS: The expression levels of LINC_00355 in GC tissues and cells were detected by quantitative real-time PCR, followed by assessing the effects of LINC_00355 knockdown or overexpression on cell properties. Dual-luciferase reporter assay was utilized to identify the relationship between LINC_00355 and microRNA (miR)-15a-5p and miR-15a-5p and PHD finger protein 19 (PHF19), followed by the rescue experiments. RESULTS: The results showed that LINC_00355 was highly expressed in GC tissues and cells compared with the corresponding control. LINC_00355 knockdown decreased the viability, migration, and invasion and increased the accumulation of GC cells in G1 phase and apoptosis. Meanwhile, LINC_00355 downregulation markedly increased cleaved caspase 3 and cleaved poly (ADP-ribose) polymerase protein levels, whereas decreased cyclin D1, cyclin E, matrix metalloproteinase (MMP) 9, MMP2, and N-cadherin protein levels in GC cells. However, LINC_00355 overexpression had the opposite effects. It was verified that LINC_00355 upregulated the expression of PHF19 through sponging miR-15a-5p. Furthermore, PHF19 overexpression reversed the effect of LINC_00355 knockdown on GC cell properties, including cell viability, migration, invasion, and apoptosis. CONCLUSIONS: Collectively, these results suggest that LINC_00355 promotes GC progression by up-regulating PHF19 through sponging miR-15a-5p. Our findings may provide an important clinical basis for reversing the malignant phenotype of GC.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Neoplasias Gástricas/genética , Fatores de Transcrição/genética , Apoptose/genética , Biópsia , Linhagem Celular Tumoral , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Mucosa Gástrica/patologia , Técnicas de Silenciamento de Genes , Humanos , RNA Longo não Codificante/genética , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patologia , Ativação Transcricional , Regulação para Cima
17.
RSC Adv ; 11(21): 12915-12928, 2021 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-35423829

RESUMO

The non-specific distribution, non-selectivity towards cancerous cells, and adverse off-target side effects of anticancer drugs and other therapeutic molecules lead to their inferior clinical efficacy. Accordingly, ultrasound-based targeted delivery of therapeutic molecules loaded in smart nanocarriers is currently gaining wider acceptance for the treatment and management of cancer. Nanobubbles (NBs) are nanosize carriers, which are currently used as effective drug/gene delivery systems because they can deliver drugs/genes selectively to target sites. Thus, combining the applications of ultrasound with NBs has recently demonstrated increased localization of anticancer molecules in tumor tissues with triggered release behavior. Consequently, an effective therapeutic concentration of drugs/genes is achieved in target tumor tissues with ultimately increased therapeutic efficacy and minimal side-effects on other non-cancerous tissues. This review illustrates present developments in the field of ultrasound-nanobubble combined strategies for targeted cancer treatment. The first part of this review discusses the composition and the formulation parameters of NBs. Next, we illustrate the interactions and biological effects of combining NBs and ultrasound. Subsequently, we explain the potential of NBs combined with US for targeted cancer therapeutics. Finally, the present and future directions for the improvement of current methods are proposed.

18.
Curr Pharm Des ; 26(26): 3171-3186, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32067605

RESUMO

BACKGROUND: Metastatic colorectal cancer (mCRC) is one of the most common and deadly cancers worldwide. For most patients diagnosed with mCRC and managed with 5-fluorouracil (5-FU)/leucovorin plus oxaliplatin (FOLFOX), the median survival time is still less than 2 years. Small molecule selective vascular endothelial growth factor receptor (VEGFR) inhibitors have been demonstrated to have strong anti-tumour activity in various cancer models. OBJECTIVE: To demonstrate the efficacy and safety of selective VEGFR inhibitors in the management of mCRC. METHODS: A comprehensive search in PubMed, EMBASE, Web of Science, Ovid MEDLINE, Google Scholar, Springer and Cochrane Central databases was performed for randomized controlled trials (RCTs) focusing on the effect of selective VEGFR inhibitors on mCRC. The primary outcome measures were progression-free survival (PFS) rates, overall survival (OS) rates, complete response (CR), partial response (PR), stable disease (SD), progressive disease (PD), objective response rates (ORRs), disease control rates (DCRs) and adverse effect (AE) rates. The dates of the included studies ranged from the inception of the database to January 15, 2020. RESULTS: Twenty-two RCTs were included. A total of 9362 patients met the inclusion criteria. Compared with placebo, selective VEGFR inhibitors significantly increased the PFS rate, SD, PR and DCR, reduced PD, caused more treatment-emergent adverse events (TEAEs), hypertension, hand-foot skin reaction, diarrhoea, fatigue, and thrombocytopaenia and increased aspartate aminotransferase(AST) concentration. There was no significant difference between selective VEGFR inhibitors and placebo regarding OS rate, CR, ORR, proteinuria, hyperbilirubinaemia or alkaline phosphatase(ALP) concentration. Additionally, compared with FOLFOX4+placebo, FOLFOX4+ selective VEGFR inhibitors, clearly reduced PD, and caused more 3-4 AEs, serious AEs, hypertension, hand-foot syndrome, diarrhoea, nausea, vomiting, decreased appetite, dehydration, fatigue, dizziness, neutropaenia and thrombocytopaenia. For PFS rate, OS rate, CR, PR, SD, ORR, abdominal pain, peripheral sensory neuropathy, asthaenia, anaemia and hypokalaemia rates, there was no significant difference between FOLFOX4+ selective VEGFR inhibitors and FOLFOX4+placebo. However, compared with FOLFOX4+bevacizumab, FOLFOX4+selective VEGFR inhibitors, led to increased hypertension, neutropaenia, fatigue, thrombocytopaenia and asthaenia. There is no clear difference between FOLFOX4+selective VEGFR inhibitors and FOLFOX4+ bevacizumab with regard to PFS rate, OS rate, CR, PR, SD, PD, ORR, diarrhoea, nausea, vomiting, peripheral neuropathy and abdominal pain rates. Selective VEGFR inhibitors+cetuximab increased PFS and PR and reduced PD compared to cetuximab, but there was no statistical difference between the two groups for OS and SD. CONCLUSION: Compared with placebo or cetuximab, selective VEGFR inhibitors alone or combined with cetuximab seemed to be more efficacious for mCRC respectively; however, the effects were not better than FOLFOX4 alone or when combined with bevacizumab for mCRC. Additionally, selective VEGFR inhibitors were not as safe as placebo or FOLFOX4 alone or in combination with bevacizumab in mCRC.


Assuntos
Neoplasias Colorretais , Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica , Bevacizumab/uso terapêutico , Cetuximab/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Fluoruracila , Humanos , Receptores de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Resultado do Tratamento
19.
Medicine (Baltimore) ; 97(41): e12784, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30313101

RESUMO

Retrospective studies have suggested that capecitabine combined with temozolomide (CAPTEM) is effective for treating patients with advanced neuroendocrine neoplasms (NENs); however, the efficacy and safety of this regimen needs to be verified by high-quality evidence or results of randomized controlled trials.We carried out a meta-analysis to evaluate the safety and effectiveness of a CAPTEM protocol for patients with advanced NENs. Systematic electronic literature searches were conducted using PubMed, EMBASE, and the Cochrane Library, and among meeting abstracts of the American Society of Clinical Oncology, European Society for Medical Oncology, European Neuroendocrine Tumor Society, and North American Neuroendocrine Tumor Society, up to June 30, 2017. We selected studies describing CAPTEM regimens for treating advanced NENs and reported on tumor response and/or toxicities according to clear World Health Organization (WHO) grading of patients. Three reviewers independently and repeatedly identified studies, extracted data, and assessed the quality of the literature. A single-proportion meta-analysis was applied to included articles.Fifteen studies with a total of 384 individuals were included. Medium overall survival in most studies was more than 12 months, whereas medium progression-free survival was similar or slightly higher than that in studies using other treatment regimes. Disease control rate of CAPTEM administration for patients with NENs was 72.89% (95% confidence interval, 64.04-81.73%; I = 82.4%; P < .01). WHO grade 3 to 4 toxicities, such as thrombocytopenia (3.36%), neutropenia (0.69%), lymphopenia (0.65%), anemia (0.59%), mucositis (0.57%), fatigue (0.54%), diarrhea (0.49%), nausea (0.39%), and transaminase elevation (0.13%) were reported in the trials included.CAPTEM is effective and relatively safe for treating patients with advanced NENs.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Capecitabina/uso terapêutico , Dacarbazina/análogos & derivados , Tumores Neuroendócrinos/tratamento farmacológico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Capecitabina/administração & dosagem , Capecitabina/efeitos adversos , Dacarbazina/administração & dosagem , Dacarbazina/efeitos adversos , Dacarbazina/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Estudos Observacionais como Assunto , Análise de Sobrevida , Temozolomida
20.
Gastrointest Endosc ; 86(5): 831-838, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28286094

RESUMO

BACKGROUND AND AIMS: Esophageal endoscopic submucosal dissection (ESD) is technically challenging because of the thinner wall and narrow lumen. The tunnel technique was proposed previously. This current retrospective study aimed to evaluate the efficacy of the tunnel technique in ESD of superficial esophageal squamous cell carcinoma (ESCC). METHODS: Patients who underwent ESD for superficial ESCC between October 2013 and September 2015 were included in the study. Propensity score matching was used to compensate for the differences in age, sex, resected specimen size, and pathology. Treatment outcomes were compared with conventional statistic methods between the tunnel ESD group and conventional ESD group after matching. To further explore the potential variables relevant to procedure time, univariate and multivariate logistic regression analyses were applied. RESULTS: A total of 115 lesions were included in the analysis. Propensity score matching analysis created 38 matched pairs. There was no difference on en bloc resection rate, complete resection rate, and curative rate between the 2 groups. The ESD procedure time was 38.0 (range 29.5-46.0) minutes in the tunnel ESD group and 48.0 (35.4-83.3) minutes in the conventional ESD group (P = .006). There was no difference in adverse events including postprocedural bleeding, perforation, and chest pain, but a lower rate of muscular injury (28.9% vs 52.6%; P = .036) and a less-frequent use of coagulation forceps (36.8% vs 65.8%; P = .012) were shown in the tunnel ESD group. In multivariate regression analysis for procedure time, the tunnel ESD technique (odds ratio [OR] 3.42; 95% confidence interval [CI], 1.32-8.85; P = .011) and specimen size <40 mm (OR 8.74; 95% CI, 1.30-58.5; P = .026) were associated with a shorter procedure time. CONCLUSIONS: The endoscopic submucosal tunnel dissection improved the efficacy and safety of the ESD procedure by shortening the procedure time and reducing injury to the muscular layer.


Assuntos
Carcinoma de Células Escamosas/cirurgia , Ressecção Endoscópica de Mucosa/métodos , Neoplasias Esofágicas/cirurgia , Duração da Cirurgia , Complicações Pós-Operatórias/epidemiologia , Idoso , Carcinoma de Células Escamosas/patologia , Dor no Peito/epidemiologia , Neoplasias Esofágicas/patologia , Perfuração Esofágica/epidemiologia , Carcinoma de Células Escamosas do Esôfago , Esofagoscopia/métodos , Feminino , Hemorragia Gastrointestinal/epidemiologia , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Razão de Chances , Hemorragia Pós-Operatória/epidemiologia , Pontuação de Propensão , Estudos Retrospectivos , Resultado do Tratamento , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA