Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pharmacol Res ; 203: 107160, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38547937

RESUMEN

Immunostimulatory antibody conjugates (ISACs) as a promising new generation of targeted therapeutic antibody-drug conjugates (ADCs), that not only activate innate immunity but also stimulate adaptive immunity, providing a dual therapeutic effect to eliminate tumor cells. However, several ISACs are still in the early stages of clinical development or have already failed. Therefore, it is crucial to design ISACs more effectively to overcome their limitations, including high toxicity, strong immunogenicity, long development time, and poor pharmacokinetics. This review aims to summarize the composition and function of ISACs, incorporating current design considerations and ongoing clinical trials. Additionally, the review delves into the current issues with ISACs and potential solutions, such as adjusting the drug-antibody ratio (DAR) to improve the bioavailability of ISACs. By leveraging the affinity and bioavailability-enhancing properties of bispecific antibodies, the utility between antibodies and immunostimulatory agents can be balanced. Commonly used immunostimulatory agents may induce systemic immune reactions, and BTK (Bruton's tyrosine kinase) inhibitors can regulate immunogenicity. Finally, the concept of grafting ADC's therapeutic principles is simple, but the combination of payload, linker, and targeted functional molecules is not a simple permutation and combination problem. The development of conjugate drugs faces more complex pharmacological and toxicological issues. Standing on the shoulders of ADC, the development and application scenarios of ISAC are endowed with broader space.


Asunto(s)
Inmunoconjugados , Humanos , Inmunoconjugados/uso terapéutico , Inmunoconjugados/farmacología , Animales , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología
2.
J Cell Mol Med ; 25(12): 5534-5546, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33960636

RESUMEN

Glioblastoma (GBM) is the most common malignant intracranial tumour with intrinsic infiltrative characteristics, which could lead to most patients eventually relapse. The prognosis of recurrent GBM patients remains unsatisfactory. Cancer cell infiltration and their interaction with the tumour microenvironment (TME) could promote tumour recurrence and treatment resistance. In our study, we aimed to identify potential tumour target correlated with rGBM microenvironment based on the gene expression profiles and clinical information of rGBM patients from The Cancer Genome Atlas (TCGA) database. LRRC15 gene with prognostic value was screened by univariate and multivariate analysis, and the correlation between macrophages and LRRC15 was identified as well. Furthermore, the prognosis correlation and immune characteristics of LRRC15 were validated using the Chinese Glioma Genome Atlas (CGGA) database and our clinical tissues by immunochemistry assay. Additionally, we utilized the transwell assay and carboxy fluorescein succinimidyl ester (CFSE) tracking to further confirm the effects of LRRC15 on attracting microglia/macrophages and tumour cell proliferation in the TME. Gene profiles-based rGBM microenvironment identified that LRRC15 could act in collusion with microglia/macrophages in the rGBM microenvironment to promote the poor prognosis, especially in mesenchymal subtype, indicating the strategies of targeting LRRC15 to improve macrophages-based immunosuppressive effects could be promising for rGBM treatments.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Recurrencia Local de Neoplasia/patología , Microambiente Tumoral/inmunología , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Perfilación de la Expresión Génica , Glioblastoma/genética , Glioblastoma/inmunología , Humanos , Proteínas de la Membrana/genética , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Pronóstico , Tasa de Supervivencia
3.
J Cell Physiol ; 234(7): 11888-11899, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30523640

RESUMEN

MiR-137 has been identified as potential hepatocellular carcinoma (HCC) prognostic biomarkers. Highly relevant HCC prognostic biomarkers may be derived from combinations of miR-137 with its target genes involved in the regulation of liver microenvironment. This study aimed at the discovery of such a combination with improved HCC prognosis performance than miR-137 or its target gene alone in a significantly higher number of HCC patients than previous studies. Analysis of the differentially expressed micro RNAs (miRNAs) between cancer and noncancer tissues reconfirmed miR-137 to be among the most relevant prognostic miRNAs and the data of 375 HCC patients and 50 normal cases were from the Cancer Genome Atlas (TCGA) data sets. Target genes were identified by the established search methods and Kaplan-Meier survival analysis of HCC patients was used to evaluate the overall survival (OS) and recurrence-free survival (RFS). Cox proportional hazards regression indicated that the miR-137 and its target gene AFM combination is an independent prognostic factor for the OS and RFS in HCC. In vitro experiments validated that miR-137 could bind to 3'-untranslated region of the AFM and promote the invasion and metastasis of HCC cell lines. The expressions of miR-137 and its liver microenvironment regulatory target gene AFM in combination significantly correlated with HCC progression in a higher number of patients than in previous studies, which suggested their potential as prognostic biomarkers for HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , Biomarcadores de Tumor/genética , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Microambiente Tumoral/genética
4.
J Transl Med ; 17(1): 259, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31395064

RESUMEN

BACKGROUND: Ovarian cancer is the leading cause of death in gynecological cancer. Cancer stem cells (CSCs) contribute to the occurrence, progression and resistance. Small nucleolar RNAs (SnoRNAs), a class of small molecule non-coding RNA, involve in the cancer cell stemness and tumorigenesis. METHODS: In this study, we screened out SNORNAs related to ovarian patient's prognosis by analyzing the data of 379 cases of ovarian cancer patients in the TCGA database, and analyzed the difference of SNORNAs expression between OVCAR-3 (OV) sphere-forming (OS) cells and OV cells. After overexpression or knockdown SNORD89, the expression of Nanog, CD44, and CD133 was measured by qRT-PCR or flow cytometry analysis in OV, CAOV-3 (CA) and OS cells, respectively. CCK-8 assays, plate clone formation assay and soft agar colony formation assay were carried out to evaluate the changes of cell proliferation and self-renewal ability. Scratch migration assay and trans-well invasion analysis were used for assessing the changes of migration and invasion ability. RESULTS: High expression of SNORD89 indicates the poor prognosis of ovarian cancer patients and was associated with patients' age, therapy outcome. SNORD89 highly expressed in ovarian cancer stem cells. The overexpression of SNORD89 resulted in the increased stemness markers, S phase cell cycle, cell proliferation, invasion and migration ability in OV and CA cells. Conversely, these phenomena were reversed after SNORD89 silencing in OS cells. Further, we found that SNORD89 could upregulate c-Myc and Notch1 expression in mRNA and protein levels. SNORD89 deteriorates the prognosis of ovarian cancer patients by regulating Notch1-c-Myc pathway to promote cell stemness and acts as an oncogene in ovarian tumorigenesis. Consequently, SNORD89 can be a novel prognostic biomarker and therapeutic target for ovarian cancer.


Asunto(s)
Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , ARN Nucleolar Pequeño/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Autorrenovación de las Células/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , Fenotipo , Pronóstico , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Nucleolar Pequeño/genética
5.
Tumour Biol ; 37(4): 5001-11, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26537584

RESUMEN

Wnt/ß-catenin signaling pathway plays a major role in the cancer metastasis. Several microRNAs (miRNAs) are contributed to the inhibition of breast cancer metastasis. Here, we attempted to find novel targets and mechanisms of microRNA-100 (miR-100) in regulating the migration and invasion of breast cancer cells. In this study, we found that miR-100 expression was downregulated in human breast cancer tissues and cell lines. The overexpression of miR-100 inhibited the migration and invasion of MDA-MB-231 breast cancer cells. Inversely, the downregulation of miR-100 increased the migration and invasion of MCF-7 breast cancer cells. Furthermore, FZD-8, a receptor of Wnt/ß-catenin signaling pathway, was demonstrated a direct target of miR-100. The overexpression of miR-100 decreased the expression levels not only FZD-8 but also the key components of Wnt/ß-catenin pathway, including ß-catenin, metalloproteniase-7 (MMP-7), T-cell factor-4 (TCF-4), and lymphoid enhancing factor-1 (LEF-1), and increased the protein expression levels of GSK-3ß and p-GSK-3ß in MDA-MB-231 cells, and the transfection of miR-100 inhibitor in MCF-7 cells showed the opposite effects. In addition, the expression of miR-100 was negatively correlated with the FZD-8 expression in human breast cancer tissues. Overall, these findings suggest that miR-100 suppresses the migration and invasion of breast cancer cells by targeting FZD-8 and inhibiting Wnt/ß-catenin signaling pathway and manipulation of miR-100 may provide a promoting therapeutic strategy for cancer breast treatment.


Asunto(s)
Neoplasias de la Mama/genética , MicroARNs/genética , Invasividad Neoplásica/genética , Receptores de Superficie Celular/biosíntesis , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/biosíntesis , Neoplasias de la Mama/patología , Movimiento Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/biosíntesis , Humanos , Células MCF-7 , Metaloproteinasa 7 de la Matriz/biosíntesis , MicroARNs/biosíntesis , Invasividad Neoplásica/patología , Receptores de Superficie Celular/genética , Factor de Transcripción 4 , Factores de Transcripción/biosíntesis , Vía de Señalización Wnt , beta Catenina/genética
6.
Jpn J Clin Oncol ; 46(10): 893-902, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27432453

RESUMEN

OBJECTIVE: Previous studies have revealed the association of multidrug resistance with histone deacetylases inhibitors treatment in cancer cells. But little data were available for the correlation of histone deacetylases and drug-resistant-related proteins in breast cancer tissue. This study aimed to exploring the association of histone deacetylases expression with clinicopathological features, drug-resistant-related proteins, prognosis and therapeutic responses in breast cancer patients. METHODS: We performed immunohistochemistry to study the expression of HDAC1 and HDAC2 in 226 breast cancer and 34 breast fibroadenoma patients, and the expression of breast cancer resistance protein, P-glycoprotein, lung resistance protein and multidrug resistance protein in 226 breast cancer. RESULTS: In breast cancer, HDAC2 expression was significantly increased than in fibroadenoma (P = 0.015), and correlated with lymph node metastasis (P = 0.002), advanced clinical stages (P = 0.016) and high histological grade (P = 0.001). Significant positive correlations were found between HDAC2 and Ki67, HDAC1 and multidrug resistance protein, HDAC2 and breast cancer resistance protein, HDAC2 and multidrug resistance protein. HDAC2 positive expression was associated with shorter overall survival (P = 0.035) of breast cancer patients. In addition, HDAC2-positive expression was significantly associated with shorter overall survival in multidrug resistance protein-positive patients (P = 0.034), but not in multidrug resistance protein-negative patients (P = 0.530). HDAC2-positive expression was associated with shorter survival in patients who received chemotherapy containing anthracyclines (overall survival, P = 0.041; disease-free survival, P = 0.084), but not in patients who received chemotherapy without anthracyclines (overall survival, P = 0.679; disease-free survival, P = 0.708). CONCLUSIONS: HDAC2 overexpression correlated with the metastasis, progression and the increased Ki67, multidrug resistance protein expression in breast cancer, and HDAC2 could be a prognostic factor of breast cancer patients, especially the patients who received anthracyclines therapy.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antraciclinas/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Histona Desacetilasa 2/metabolismo , Adulto , Anciano , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Femenino , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Metástasis Linfática , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Oportunidad Relativa , Pronóstico , Modelos de Riesgos Proporcionales
7.
Mol Carcinog ; 54(9): 707-19, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24464625

RESUMEN

DNA methyltransferases (DNMTs), including DNMT1, 3a, and 3b, play an important role in the progression of many malignant tumors. However, it remains unclear whether expression of DNMTs is associated with the development of breast cancer. This study aimed to explore the clinical significance of DNMT proteins in sporadic breast cancer. We investigated the expression of DNMT1, 3a, and 3b in 256 breast cancer and 36 breast fibroadenoma, using immunohistochemistry. The expression of DNMT1 and 3a was significantly higher in breast cancer than in fibroadenoma. In breast cancer, the expression of DNMT1 was significantly correlated with lymph node metastasis (P = 0.020), and the expression of DNMT3a and 3b was significantly correlated with advanced clinical stages (P = 0.046 and 0.012, respectively). Overexpression of DNMT1/3a was correlated with promoter hypermethylation and reduced expression of ERα and BRCA1. The expression levels of DNMT1 or DNMT3a were associated with a significantly shorter DFS or OS in a subgroup of breast cancer patients (patients with the age ≤50 years old, ERα-negative status, or HER2-postive status). The expression of DNMT1 or a combined expression of DNMT1 and 3a was associated with poor prognosis in patients who received chemotherapy and endocrine therapy, but not in patients who received chemotherapy alone. These findings suggest that DNMT1 and 3a may be involved in the progression and prognosis of sporadic breast cancer.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/diagnóstico , Mama/patología , ADN (Citosina-5-)-Metiltransferasas/análisis , Receptor alfa de Estrógeno/genética , Fibroadenoma/diagnóstico , Adulto , Anciano , Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , ADN Metiltransferasa 3A , Regulación hacia Abajo , Femenino , Fibroadenoma/genética , Fibroadenoma/patología , Regulación Neoplásica de la Expresión Génica , Genes BRCA1 , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Pronóstico , Regiones Promotoras Genéticas , Análisis de Supervivencia , Regulación hacia Arriba
8.
Clin Sci (Lond) ; 129(9): 809-22, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26201092

RESUMEN

BCSCs (breast cancer stem cells) have been shown to be resistant to chemotherapy. However, the mechanisms underlying BCSC-mediated chemoresistance remain poorly understood. The Hh (Hedgehog) pathway is important in the stemness maintenance of CSCs. Nonetheless, it is unknown whether the Hh pathway is involved in BCSC-mediated chemoresistance. In the present study, we cultured breast cancer MCF-7 cells in suspension in serum-free medium to obtain BCSC-enriched MCF-7 MS (MCF-7 mammosphere) cells. We showed that MCF-7 MS cells are sensitive to salinomycin, but not paclitaxel, distinct from parent MCF-7 cells. The expression of the critical components of Hh pathway, i.e., PTCH (Patched), SMO (Smoothened), Gli1 and Gli2, was significantly up-regulated in MCF-7 MS cells; salinomycin, but not paclitaxel, treatment caused a remarkable decrease in expression of those genes in MCF-7 MS cells, but not in MCF-7 cells. Salinomycin, but not paclitaxel, increased apoptosis, decreased the migration capacity of MCF-7 MS cells, accompanied by a decreased expression of c-Myc, Bcl-2 and Snail, the target genes of the Hh pathway. The salinomycin-induced cytotoxic effect could be blocked by Shh (Sonic Hedgehog)-mediated Hh signalling activation. Inhibition of the Hh pathway by cyclopamine could sensitize MCF-7 MS cells to paclitaxel. In addition, salinomycin, but not paclitaxel, significantly reduced the tumour growth, accompanied by decreased expression of PTCH, SMO, Gli1 and Gli2 in xenograft tumours. Furthermore, the expression of SMO and Gli1 was positively correlated with the expression of CD44+ / CD24-, and the expression of SMO and Gli1 in CD44+ / CD24- tissues was associated with a significantly shorter OS (overall survival) and DFS (disease-free survival) in breast cancer patients receiving chemotherapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Hedgehog/metabolismo , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Antígeno CD24/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Receptores de Hialuranos/metabolismo , Estimación de Kaplan-Meier , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Piranos/farmacología , Piranos/uso terapéutico , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc
9.
Tumour Biol ; 35(7): 7047-56, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24752577

RESUMEN

Bone morphogenetic proteins and activin membrane-bound inhibitor (BAMBI) and drosophila mothers against decapentaplegic protein 7 (Smad7) are known to negatively regulate the transforming growth factor-ß (TGF-ß) signaling and play an important role in the progression of many malignant tumors. However, it remains unclear whether expression of BAMBI alone or in combination with Smad7 is associated with the progression of gastric cancer. In the present study, we investigated the expression of BAMBI and Smad7 in 276 cancer tissues and 263 tumor-adjacent tissues from gastric cancer patients, using tissue-microarray-based immunohistochemistry. The expression of BAMBI and Smad7 was significantly higher in cancer tissues than in tumor-adjacent tissues. The expression of BAMBI was significantly correlated with increased depth of invasion (P = 0.010), lymphatic invasion (P < 0.001), lymph node metastasis (P = 0.001), TNM stage (P = 0.008), and decreased differentiation (P = 0.046). The expression of BAMBI was associated with a significantly shorter overall survival (OS) (P = 0.006) and disease-free survival (DFS) (P = 0.011). The combined expression of BAMBI and Smad7 was associated with more invasion and metastasis as well as less survival time in gastric cancer patients. The univariate analysis showed that the expression of BAMBI alone or in combination with Smad7 was significantly associated with the OS and DFS. These findings suggest that BAMBI and Smad7 may cooperatively inhibit the TGF-ß signaling, and thus promote the progression of gastric cancer.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Proteínas de la Membrana/biosíntesis , Proteína smad7/biosíntesis , Neoplasias Gástricas/genética , Anciano , Biomarcadores de Tumor/genética , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Invasividad Neoplásica/genética , Pronóstico , Transducción de Señal/genética , Proteína smad7/genética , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
10.
World J Gastrointest Oncol ; 16(2): 259-272, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38425391

RESUMEN

Approximately 20% of colorectal cancer (CRC) patients present with metastasis at diagnosis. Among Stage I-III CRC patients who undergo surgical resection, 18% typically suffer from distal metastasis within the first three years following initial treatment. The median survival duration after the diagnosis of metastatic CRC (mCRC) is only 9 mo. mCRC is traditionally considered to be an advanced stage malignancy or is thought to be caused by incomplete resection of tumor tissue, allowing cancer cells to spread from primary to distant organs; however, increasing evidence suggests that the mCRC process can begin early in tumor development. CRC patients present with high heterogeneity and diverse cancer phenotypes that are classified on the basis of molecular and morphological alterations. Different genomic and nongenomic events can induce subclone diversity, which leads to cancer and metastasis. Throughout the course of mCRC, metastatic cascades are associated with invasive cancer cell migration through the circulatory system, extravasation, distal seeding, dormancy, and reactivation, with each step requiring specific molecular functions. However, cancer cells presenting neoantigens can be recognized and eliminated by the immune system. In this review, we explain the biological factors that drive CRC metastasis, namely, genomic instability, epigenetic instability, the metastatic cascade, the cancer-immunity cycle, and external lifestyle factors. Despite remarkable progress in CRC research, the role of molecular classification in therapeutic intervention remains unclear. This review shows the driving factors of mCRC which may help in identifying potential candidate biomarkers that can improve the diagnosis and early detection of mCRC cases.

11.
Int J Biol Macromol ; 259(Pt 2): 129320, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38218276

RESUMEN

Polysaccharides possess excellent moisturizing effects due to their abundance of hydrophilic groups and film-forming properties. Additionally, they can produce a refreshing aroma during the pyrolysis process. However, there is scarce research on their application in the tobacco field. Herein, we investigated the effects of low molecular weight fenugreek polysaccharide (FP) obtained through ethanol fractionation and DEAE-52 cellulose column chromatography on moisture retention and aroma enhancement in tobacco. The moisture retention test revealed that the addition of FP increased the moisture retention index (MRI) of tobacco by 11.72 %-16.69 %, indicating that the hydrophilic nature of polysaccharides facilitated the migration of free water in tobacco to bound water, resulting in reduced water activity. Moreover, the contact angle between polysaccharide and tobacco was <90°, enabling better infiltration into tobacco and slowing down tobacco shrinkage caused by water loss. Among all the components, EFP-20 and EFP-40 demonstrated superior performance. Furthermore, FP exhibited excellent thermal stability below 200 °C and can decomposed to produce aromatic substances at high temperatures. It also demonstrated the ability to adsorb ethyl heptanoate and thermally decompose to produce a substantial amount of heptanoic acid. Consequently, the incorporation of FP in tobacco demonstrated favorable effects on both moisturization and aroma enhancement.


Asunto(s)
Productos de Tabaco , Trigonella , Odorantes/análisis , Peso Molecular , Polisacáridos/farmacología , Polisacáridos/química , Agua/química
12.
Tumour Biol ; 34(6): 3945-58, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23860775

RESUMEN

Breast cancer is a complex disease; the molecular mechanisms involved in sporadic breast carcinogenesis remain to be elucidated. The present study aimed to explore the deficiency of breast cancer susceptibility gene 1 (BRCA1), including protein loss expression, promoter hypermethylation and gene copy deletion, its correlationship with other tumor markers expression (TP53, MYC, etc.), and clinical significance in sporadic breast cancer. BRCA1 protein expression was negative in 226 of 374 (60.4%) cases of this study. Cases negative for BRCA1 protein were more often with pathological tumor-node-metastasis stage III, positive for lymph node metastasis and MYC overexpression than BRCA1-positive tumors. BRCA1 hypermethylation was detected in 16.4% (31 of 189) breast cancers, which correlated with BRCA1 negative, ER negative, MYC overexpression, and triple-negative phenotype. In addition, the percentage of cells with BRCA1 gene copy deletion was significantly increased in BRCA1-methylated tumors. Kaplan-Meier survival analysis showed that patients with BRCA1-negative expression showed a worse overall survival (OS) than those with BRCA1-positive expression, and patients with BRCA1-methylated tumors had a significantly worse disease-free survival than did patients with unmethylated tumors. Furthermore, BRCA1 hypermethylation showed an inverse association with OS in LN-positive or p53-negative subgroup patients. Importantly, uni- and multivariate Cox regression analyses revealed that BRCA1 was an independent prognostic indicator of OS in sporadic breast cancer. Thus, we found MYC overexpression and poor prognosis in sporadic breast cancer with BRCA1 deficiency. The targeting of BRCA1 deficiency in combination with MYC-pathways inhibitors may provide a promising strategy for sporadic breast cancer care, the triple-negative subtype in particular.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-myc/genética , Adulto , Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Variaciones en el Número de Copia de ADN , Metilación de ADN , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica/estadística & datos numéricos , Hibridación Fluorescente in Situ/estadística & datos numéricos , Estimación de Kaplan-Meier , Metástasis Linfática , Persona de Mediana Edad , Análisis Multivariante , Mutación , Estadificación de Neoplasias , Pronóstico , Regiones Promotoras Genéticas/genética , Modelos de Riesgos Proporcionales , Proteínas Proto-Oncogénicas c-myc/metabolismo
13.
Acta Pharm Sin B ; 13(2): 498-516, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36873165

RESUMEN

Peptide-drug conjugates (PDCs) are the next generation of targeted therapeutics drug after antibody-drug conjugates (ADCs), with the core benefits of enhanced cellular permeability and improved drug selectivity. Two drugs are now approved for market by US Food and Drug Administration (FDA), and in the last two years, the pharmaceutical companies have been developing PDCs as targeted therapeutic candidates for cancer, coronavirus disease 2019 (COVID-19), metabolic diseases, and so on. The therapeutic benefits of PDCs are significant, but poor stability, low bioactivity, long research and development time, and slow clinical development process as therapeutic agents of PDC, how can we design PDCs more effectively and what is the future direction of PDCs? This review summarises the components and functions of PDCs for therapeutic, from drug target screening and PDC design improvement strategies to clinical applications to improve the permeability, targeting, and stability of the various components of PDCs. This holds great promise for the future of PDCs, such as bicyclic peptide‒toxin coupling or supramolecular nanostructures for peptide-conjugated drugs. The mode of drug delivery is determined according to the PDC design and current clinical trials are summarised. The way is shown for future PDC development.

14.
Front Oncol ; 13: 1107631, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36895477

RESUMEN

Advanced non-small cell lung cancer (NSCLC) is a severe disease and still has high mortality rate after conventional treatment (e.g., surgical resection, chemotherapy, radiotherapy and targeted therapy). In NSCLC patients, cancer cells can induce immunosuppression, growth and metastasis by modulating cell adhesion molecules of both cancer cells and immune cells. Therefore, immunotherapy is increasingly concerned due to its promising anti-tumor effect and broader indication, which targets cell adhesion molecules to reverse the process. Among these therapies, immune checkpoint inhibitors (mainly anti-PD-(L)1 and anti-CTLA-4) are most successful and have been adapted as first or second line therapy in advanced NSCLC. However, drug resistance and immune-related adverse reactions restrict its further application. Further understanding of mechanism, adequate biomarkers and novel therapies are necessary to improve therapeutic effect and alleviate adverse effect.

15.
Mol Carcinog ; 51 Suppl 1: E83-93, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22213216

RESUMEN

Xeroderma pigmentosum complementation group C and G (XPC, XPG) play important roles in DNA damage repairing machinery. Genetic variations in the XPC and XPG may be associated with increased risk for colorectal carcinoma (CRC). In this study, we evaluated the relation between the XPC Lys939Gln, XPG Asp1104His polymorphisms, and CRC susceptibility in a population-based case-control study, which included 1,028 CRC cases and 1,085 controls. Compared with the corresponding wild genotypes, we found that individuals with at least one copy of the XPC Lys939Gln (AC or CC genotype) and XPG Asp1104His (GC or CC genotype) had an increased risk for CRC. In addition, the variant genotypes of the XPC Lys939Gln AC/CC (P = 0.027) or XPG Asp1104His GC/CC (P = 0.003) reduced the elevation of preoperative carcinoembryonic antigen (CEA) level. Moreover a significantly longer progression-free survival (PFS) after Oxaliplatin-based adjuvant chemotherapy was observed in patients with XPG Asp1104His wide-type GG genotype (n = 432, Log-rank test: P = 0.033). Cox proportional hazards analyses demonstrated that variant genotypes of XPG Asp1104His [hazard ratio (HR) = 1.692, 95% confidence interval (95%CI): 1.202-2.383, P = 0.003] as well as pathology grade (HR = 2.545, 95%CI: 2.139-3.030, P < 0.001), and lymph node metastases (HR = 1.851, 95%CI: 1.306-2.625, P < 0.001) were predictive of shorter PFS for the CRC patients with Oxaliplatin-based adjuvant chemotherapy. In conclusion, the current data suggested that XPC Lys939Gln and XPG Asp1104His polymorphisms might contribute to the identification of patients with increased risk for CRC.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Proteínas de Unión al ADN/genética , Compuestos Organoplatinos/uso terapéutico , Polimorfismo Genético , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/sangre , Antígeno Carcinoembrionario/sangre , Quimioterapia Adyuvante , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Reparación del ADN/genética , Supervivencia sin Enfermedad , Endonucleasas/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Oxaliplatino , Polimorfismo de Nucleótido Simple , Modelos de Riesgos Proporcionales , Factores de Transcripción/genética , Resultado del Tratamiento , Xerodermia Pigmentosa/genética , Adulto Joven
17.
Int J Oncol ; 60(5)2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35348188

RESUMEN

Following the publication of the above paper, a concerned reader drew to the Editor's attention that one of the fluorescence microscopic images featured in Fig. 4A had previously appeared in a different form (a portion of data in a different orientation) in another article published by the same authors [Yu J, Zhao L, Li Y, Li N, He M, Bai H, Yu Z, Zheng Z, Mi X, Wang E and We M: Silencing of Fanconi anemia complementation group F exhibits potent chemosensitization of mitomycin C activity in breast cancer cells. J Breast Cancer 16: 291­299, 2013]. Furthermore, the data panel shown for the 'MDA­MB­231/untreated' experiment in Fig. 4A in the above paper appeared to be duplicated as the 'MDA­MB­231/MMC + control shRNA' experiment, albeit stained differently. After having received a request from the authors to publish a corrigendum in view of the errors identified in Fig. 4 of the above paper, the Editor of International Journal of Oncology has conducted an independent investigation of the matter and determined that this article should be retracted from the Journal on account of a lack of confidence in the presented data. Upon receiving this decision, the authors were not in agreement that the paper should be retracted. The Editor regrets any inconvenience that has been caused to the readership of the Journal. [the original article was published in International Journal of Oncology 45: 129­138, 2014; DOI: 10.3892/ijo.2014.2400].

18.
Oncol Rep ; 47(6)2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35417034

RESUMEN

Subsequently to the publication of the above article, an interested reader drew to the authors' attention that the data panel for the MDA­MB­231/migration/NC experiment in Fig. 2B on p. 1428 was strikingly similar to the data shown for the MDA­MB­231/invasion/Blank experiment in Fig. 2C, such that these data appeared to have been derived from the same original source. The authors have referred back to their original data, and realize that the data panel was selected incorrectly for Fig. 2B. The corrected version of Fig. 2, showing the correct data for the MDA­MB­231/migration/NC experiment in Fig. 2B, is shown on the next page. The authors regret the error that was made during the preparation of this figure, and can confirm that the error in the assembly of this figure did not adversely affect the conclusions reported in the study. The authors are grateful to the Editor of Oncology Reports for allowing them the opportunity to publish a Corrigendum, and all the authors agree to this Corrigendum. Furthermore, they apologize to the readership for any inconvenience caused. [the original article was published in Oncology Reports 35: 1425­1432, 2016; DOI: 10.3892/or.2015.4502].

19.
Int Immunopharmacol ; 109: 108783, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35561479

RESUMEN

The rapid development of bioengineering technology has introduced Fc-fusion proteins, representing a novel kind of recombinant protein, as promising biopharmaceutical products in tumor therapy. Numerous related anti-tumor Fc-fusion proteins have been investigated and are in different stages of development. Fc-fusion proteins are constructed by fusing the Fc-region of the antibody with functional proteins or peptides. They retain the bioactivity of the latter and partial properties of the former. This structural and functional advantage makes Fc-fusion proteins an effective tool in tumor immunotherapy, especially for the recruitment and activation of natural killer (NK) cells, which play a critical role in tumor immunotherapy. Even though tumor cells have developed mechanisms to circumvent the cytotoxic effect of NK cells or induce defective NK cells, Fc-fusion proteins have been proven to effectively activate NK cells to kill tumor cells in different ways, such as antibody-dependent cell-mediated cytotoxicity (ADCC), activate NK cells in different ways in order to promote killing of tumor cells. In this review, we focus on NK cell-based immunity for cancers and current research progress of the Fc-fusion proteins for anti-tumor therapy by activating NK cells.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas , Células Asesinas Naturales , Citotoxicidad Celular Dependiente de Anticuerpos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoterapia , Proteínas Recombinantes de Fusión/genética
20.
Oncol Rep ; 47(6)2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35543152

RESUMEN

After the publication of the article, an interested reader drew to the authors' attention that there appeared to be a pair of overlapping data panels in Fig. 4C on p. 1726 [specifically, the 'Untransfected' and 'Control shRNA' data panels for the ADM (24 h) experiments]. The authors have consulted their original data, and have realized that this figure was inadvertently assembled incorrectly. Furthermore, they have noticed that Fig. 1 on p. 1724 also contained errors that arose during its assembly; essentially, several of the data panels in Fig. 1C, showing the detection of FANCD2 focus formation via immunofluorescence experiments, were selected inappropriately. The corrected versions of Figs. 1 and 4, containing the corrected data panels for Figs. 1C and 4C respectively, are shown on the next page. Note that these errors did not affect the results or the conclusions reported in this work. The authors all agree to this Corrigendum, and are grateful to the Editor of Oncology Reports for allowing them to have the opportunity to correct these mistakes. Lastly, the authors apologize to the readership for any inconvenience these errors may have caused. [Oncology Reports 29: 1721­1729, 2013; DOI: 10.3892/or.2013.2295].

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA