Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.142
Filter
1.
Respir Res ; 25(1): 233, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840238

ABSTRACT

BACKGROUND: There is inconclusive evidence to suggest that the expression of programmed cell death ligand 1 (PD-L1) is a putative predictor of response to EGFR-TKI therapy in advanced EGFR-mutant non-small cell lung cancer (NSCLC). We evaluated the heterogeneity in PD-L1 expression in the primary lung site and metastatic lymph nodes to analyze the association between PD-L1 expression and response for patients treated with EGFR-TKI. METHODS: This study reviewed 184 advanced NSCLC patients with EGFR mutations who received first-generation EGFR-TKI as first-line treatment from 2020 to 2021 at Shanghai Chest Hospital. The patients were divided into the primary lung site group (n = 100) and the metastatic lymph nodes group (n = 84) according to the biopsy site. The patients in each group were divided into TPS < 1%, TPS 1-49%, and TPS ≥ 50% groups according to PD-L1 expression. RESULTS: The median PFS was 7 (95% CI: 5.7-8.3) months, and the median OS was 26 (95% CI: 23.5-28.5) months for all patients. No correlation existed between PFS or OS and PD-L1 expression. The median PFS in the primary lung site group was 11 months (95% CI: 9.6-12.4) in the TPS < 1% group, 8 months (95% CI: 6.6-9.4) in TPS 1-49% group, and 4 months (95% CI: 3.2-4.8) in TPS ≥ 50% group, with statistically significant differences (p = 0.000). The median OS of the TPS < 1% group and TPS ≥ 50% group showed a statistically significant difference (p = 0.008) in the primary lung site group. In contrast, PD-L1 expression in the lymph nodes of EGFR-mutant patients was unrelated to PFS or OS after EGFR-TKI therapy. CONCLUSION: PD-L1 expression from the primary lung site might predict clinical benefit from EGFR-TKI, whereas PD-L1 from metastatic lymph nodes did not. TRIAL REGISTRATION: This retrospective study was approved by the Ethics Committee of Shanghai Chest Hospital (ID: IS23060) and performed following the Helsinki Declaration of 1964 (revised 2008).


Subject(s)
B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung , ErbB Receptors , Lung Neoplasms , Lymphatic Metastasis , Protein Kinase Inhibitors , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Female , Male , Middle Aged , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , Aged , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies , Lymph Nodes/pathology , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Adult , Aged, 80 and over , Treatment Outcome , Predictive Value of Tests , Mutation , Biomarkers, Tumor/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/analysis
2.
Oncol Rep ; 48(2)2022 08.
Article in English | MEDLINE | ID: mdl-35775375

ABSTRACT

The clinical introduction of molecular imaging for the management of oropharyngeal squamous cell carcinoma (OPSCC) relies on the identification of relevant cancer­specific biomarkers. The application of three membrane­bound receptors, namely urokinase­type plasminogen activator receptor (uPAR), tissue factor (TF) and EGFR have been previously explored for targeted imaging and therapeutic strategies in a broad range of solid cancers. The present study aimed to investigate the expression patterns of uPAR, EGFR and TF by immunohistochemistry (IHC) to evaluate their potential for targeted imaging and prognostic value in OPSCC. In a retrospective cohort of 93 patients with primary OPSCC, who were balanced into the 45 human papillomavirus (HPV)­positive and 48 HPV­negative groups, the IHC­determined expression profiles of uPAR, TF and EGFR in large biopsy or tumor resection specimens were analyzed. Using the follow­up data, overall survival (OS) and recurrence­free survival were measured. Specifically, associations between survival outcome, biomarker expression and clinicopathological factors were examined using Cox proportional hazards model and log­rank test following Kaplan­Meier statistics. After comparing the expression pattern of biomarkers within the tumor compartment with that in the adjacent normal tissues, uPAR and TF exhibited a highly tumor­specific expression pattern, whereas EGFR showed a homogeneous expression within the tumor compartment as well as a consistent expression in the normal mucosal epithelium and salivary gland tissues. The positive expression rate of uPAR, TF and EGFR in the tumors was 98.9, 76.3 and 98.9%, respectively. No statistically significant association between biomarker expression and survival outcome could be detected. Higher uPAR expression levels had a trend towards reduced OS according to results from univariate analysis (P=0.07; hazard ratio=2.01; 95% CI=0.92­4.37). Taken together, these results suggest that uPAR, TF and EGFR may be suitable targets for molecular imaging and therapy in OPSCC. In particular, uPAR may be an attractive target owing to their high positive expression rates in tumors and a highly tumor­specific expression pattern.


Subject(s)
Oropharyngeal Neoplasms , Papillomavirus Infections , Squamous Cell Carcinoma of Head and Neck , Biomarkers, Tumor/biosynthesis , ErbB Receptors/biosynthesis , Humans , Molecular Imaging , Molecular Targeted Therapy , Oropharyngeal Neoplasms/diagnostic imaging , Oropharyngeal Neoplasms/drug therapy , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/virology , Papillomaviridae , Papillomavirus Infections/diagnostic imaging , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Prognosis , Receptors, Urokinase Plasminogen Activator/biosynthesis , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/diagnostic imaging , Squamous Cell Carcinoma of Head and Neck/drug therapy , Thromboplastin/biosynthesis
3.
Sci Rep ; 12(1): 10401, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35729260

ABSTRACT

Sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) might be involved in the activation of important pathways related to tumor immune escape, along with programmed death-ligand 1 (PD-L1). Here, we aimed to investigate the correlation between the expression of Siglec-15 and PD-L1 in nasopharyngeal carcinoma (NPC) patients. We determined the expression of PD-L1 via immunohistochemical staining and that of Siglec-15 via immunofluorescence staining in 182 NPC tissue samples. A significant correlation was identified between the PD-L1 and Siglec-15 expression (P = 0.000). Moreover, Kaplan-Meier survival curves showed that PD-L1 expression was associated with improved overall survival (OS) (P = 0.025) and Siglec-15 expression was associated with improved distant failure-free survival (D-FFS) (P = 0.048). Moreover, multivariate Cox analysis showed that PD-L1 and Siglec-15 were independent predictors of OS (P = 0.020) and D-FFS (P = 0.047), respectively. The results of the log-rank test and Cox regression analyses showed that patients exhibiting no PD-L1/Siglec-15 expression had significant advantages regarding OS, compared to other groups (P = 0.037). PD-L1 and Siglec-15 may represent novel biomarkers for predicting the prognosis of NPC patients. Siglec-15 may be considered as a potential target for the development of therapeutics for NPC treatment in the future.


Subject(s)
B7-H1 Antigen , Immunoglobulins , Membrane Proteins , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Humans , Immunoglobulins/biosynthesis , Membrane Proteins/biosynthesis , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Prognosis
4.
Cell Oncol (Dordr) ; 45(3): 463-477, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35543859

ABSTRACT

PURPOSE: Reliable biomarkers to predict the outcome and treatment response of estrogen receptor (ER)-negative breast cancer (BC) are urgently needed. Since immune-related signaling plays an important role in the tumorigenesis of ER-negative BC, we asked whether Notch genes, alone or in combination with other immune genes, can be used to predict the clinical outcome and immune checkpoint blockade (ICB) for this type of cancer. METHODS: We analyzed transcriptome data of 6918 BC samples from five independent cohorts, 81 xenograft triple-negative BC tumors that respond differently to ICB treatment and 754 samples of different cancer types from patients treated with ICB agents. RESULTS: We found that among four Notch genes, the expression levels of NOTCH1 and NOTCH4 were positively associated with recurrence of ER-negative BC, and that combined expression of these two genes (named Notch14) further enhanced this association, which was comparable with that of the Notch pathway signature. Analysis of 1182 immune-related genes revealed that the expression levels of most HLA genes, particularly HLA-DMA and -DRA, were reversely associated with recurrence in ER-negative BC with low, but not high Notch14 expression. A combined expression signature of NOTCH1, NOTCH4, HLA-DMA and HLA-DRA was more prognostic for ER-negative and triple-negative BCs than previously reported immune-related signatures. Furthermore, we found that the expression levels of these four genes were also synergistically associated with T cell exclusion score, infiltration of specific T cells and ICB efficacy in ER-negative BC, thereby providing a potential molecular mechanism for the synergistic effect of these genes on BC. CONCLUSIONS: Our data indicate that a gene signature composed of NOTCH1, NOTCH4, HLA-DMA and HLA-DRA may serve as a potential promising biomarker for predicting ICB therapy efficacy and recurrence in ER-negative/triple-negative BCs.


Subject(s)
HLA-DR alpha-Chains , Receptor, Notch1 , Receptor, Notch4 , T-Lymphocytes , Triple Negative Breast Neoplasms , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Female , HLA-DR alpha-Chains/biosynthesis , HLA-DR alpha-Chains/genetics , HLA-DR alpha-Chains/immunology , Humans , Receptor, Notch1/biosynthesis , Receptor, Notch1/genetics , Receptor, Notch1/immunology , Receptor, Notch4/biosynthesis , Receptor, Notch4/genetics , Receptor, Notch4/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
5.
J Clin Lab Anal ; 36(6): e24411, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35446997

ABSTRACT

PURPOSE: To find a useful disease marker for early diagnosis of gastric cancer, we tried to explore the expression of serum miR-181, miR-652, and carbohydrate antigen 72-4 (CA72-4). PATIENTS AND METHODS: According to clinical pathologic stages, 112 patients with gastric cancer were divided into early gastric cancer group (n = 60) and advanced gastric cancer group (n = 52), stage I-II (n = 65), and stage III-IV (n = 47). Another 50 cases of gastric benign lesions and 40 healthy controls were also selected. Real-time quantitative PCR together with chemiluminescence were applied to detect expression levels. ROC curve was applied to judge their diagnostic efficiency. Pearson's correlation analysis was put into use to investigate the relevance of three indicators. RESULTS: Compared with benign lesions group and control group, significantly higher expression levels were found in patients of gastric cancer (all p < 0.001). Similarly, compared with early gastric cancer group, significantly higher expression levels were found in advanced gastric cancer group (all p < 0.001). The same result was also found in stage III-IV (all p < 0.001). The best cutoff values were 0.93, 2.38, and 16.94 U/ml, respectively. The area under the curve (0.917, 95%CI: 0.856-0.975) of the three combined diagnosis of early gastric cancer was the largest, and its sensitivity and specificity were 92.5% and 86.8%. And miR-181 and miR-652 were positively correlated with CA72-4 (r = 0.772, p < 0.001, r = 0.853, p < 0.001). CONCLUSION: Serum miR-181, miR-652, and CA72-4 are closely linked to the occurrence and development of gastric cancer. Combination of three indicators has diagnostic value for early gastric cancer.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate , Early Detection of Cancer , MicroRNAs , Stomach Neoplasms , Antigens, Tumor-Associated, Carbohydrate/biosynthesis , Antigens, Tumor-Associated, Carbohydrate/blood , Antigens, Tumor-Associated, Carbohydrate/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Early Detection of Cancer/methods , Humans , MicroRNAs/biosynthesis , MicroRNAs/blood , MicroRNAs/genetics , Prognosis , ROC Curve , Stomach Neoplasms/blood , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
6.
BMC Gastroenterol ; 22(1): 207, 2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35473611

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is the third most common of cancer-related deaths. Nucleolar protein 14 (NOP14) is known to play different roles in diverse types of cancers. However, little is known about its roles in CRC. Here, we assessed the prognostic value and functions of NOP14 in CRC using the data from The Cancer Genome Atlas (TCGA) and validated them based on the data from Gene Expression Omnibus (GEO). METHODS: NOP14 mRNA and protein data in CRC was obtained from the TCGA, GEO, human protein atlas (HPA), and UALCAN databases. Survival and Cox regression analysis was performed to assess the prognostic value of NOP14 in CRC patients. Next, to evaluate the potential functions of NOP14, a protein-protein interaction (PPI) network was constructed and gene set enrichment analysis (GSEA) of differential expression genes (DEGs) associated with dysregulated NOP14 was performed. Finally, to investigate the immune response associated with NOP14 expression in CRC, we analyzed the correlations between immune cells infiltration and NOP14 expression level. Additionally, the correlations between immune molecule expression levels with NOP14 expression level were analyzed. RESULTS: High NOP14 mRNA expression was observed in CRC tissues based on the data from TCGA and GEO datasets. Similarly, high NOP14 protein levels were found in CRC tissues according to the immunohistochemical images from HPA. Interestingly, high NOP14 expression level was associated with an improved prognosis in CRC patients. Univariate and multivariate Cox regression analysis indicated that high NOP14 expression level was an independent protective factor for CRC patients. With the support of PPI network analysis, we found several risk genes interacted with NOP14. GSEA revealed that high NOP14 expression inhibited several signal pathways involved in tumor formation and development. Additionally, high NOP14 expression was positively associated with most kinds of immune cell infiltrations and the expression levels of some molecules related to immune activation. CONCLUSION: Altogether, these results indicated that high NOP14 expression leads to improved prognosis in CRC patients by inhibiting the signaling pathways involved in tumor growth and promoting the immune responses.


Subject(s)
Colorectal Neoplasms , Nuclear Proteins , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Humans , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Prognosis , RNA, Messenger/genetics , RNA, Messenger/immunology
7.
Exp Mol Pathol ; 126: 104761, 2022 06.
Article in English | MEDLINE | ID: mdl-35390309

ABSTRACT

The accurate identification of different components in testicular germ cell tumors (GCT) is essential for tailoring treatment and informing the clinical prognosis. PRAME (preferentially expressed antigen in melanoma), a member in the family of cancer testis antigens, plays critical roles in regulating pluripotency and suppressing somatic/germ cell differentiation in seminomas (SEM). To investigate the potential diagnostic value of PRAME in testicular GCT, here we comparatively examined the expression patterns of PRAME and SOX17 by immunohistochemistry in both pure and mixed GCT. Tissue microarrays constructed from 66 pure or mixed GCT were examined, including 25 seminomas (13 pure and 12 mixed), 35 embryonal carcinomas (EC; 7 pure and 28 mixed), 23 teratomas (TER; 10 pure and 13 mixed), 15 yolk sac tumors (YST; 1 pure and 14 mixed), and 5 choriocarcinomas (CC; 1 pure and 4 mixed), with 11 germ cell neoplasia in situ (GCNIS) and 6 normal testicular tissue as controls. The expression levels of PRAME or SOX17 were evaluated by a scoring system counting for intensity and extent of staining. PRAME nuclear expression was present in 92% (23/25) of SEM, including all 13 pure SEM, and 10 out of 12 seminomatous component of mixed GCT. In contrast, all EC and TER were completely negative for PRAME, and focal expression was demonstrated in 33.3% of YST and 20% of CC. As for SOX17, 96% of SEM and 73% of YST stained positively, whereas EC and CC were negative. Focal nuclear positivity was identified in the epithelial cell component of 17.4% (4/23) of TER. We found the sensitivity of PRAME to detect SEM to be comparable to SOX17, although SOX17 staining is more diffuse and stronger in the majority of cases. The specificity of PRAME for SEM appeared to be superior to that of SOX17 (92% versus 81%). In conclusion, PRAME is preferentially expressed in SEM or within the seminomatous component of mixed GCT with only focal variable expression in YST and CC, but shows no expression in EC and TER. These findings suggest that PRAME can be explored as a diagnostic marker for SEM.


Subject(s)
Antigens, Neoplasm , Neoplasms, Germ Cell and Embryonal , SOXF Transcription Factors , Seminoma , Testicular Neoplasms , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Humans , Male , Neoplasms, Germ Cell and Embryonal/diagnosis , Neoplasms, Germ Cell and Embryonal/genetics , SOXF Transcription Factors/genetics , Seminoma/diagnosis , Seminoma/genetics , Seminoma/metabolism , Testicular Neoplasms/diagnosis , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism
8.
Future Oncol ; 18(14): 1745-1756, 2022 May.
Article in English | MEDLINE | ID: mdl-35227076

ABSTRACT

Purpose: The authors aimed to evaluate the prognostic and predictive value of androgen receptor (AR) expression in patients with luminal/human EGFR2 negative (HER2-) T1N0 breast cancer. Methods: The cohort in this retrospective study comprised 471 patients with luminal/HER2- T1N0 breast cancer who had undergone surgery between 2013 and 2017 in the authors' center. Results: AR+ tumors were associated with favorable characteristics. AR+ patients had better 5-year recurrence-free survival rates and the risk of recurrence was greater for AR- than for AR+ patients. AR- status predicted the failure of adjuvant endocrine therapy with aromatase inhibitors and of adjuvant chemotherapy with docetaxel plus cyclophosphamide. Conclusion: AR+ expression is significantly related to a better prognosis. AR expression may be an additional biomarker for both endocrine and chemotherapy responsiveness.


Subject(s)
Breast Neoplasms , Receptor, ErbB-2 , Receptors, Androgen , Androgens , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Disease-Free Survival , Docetaxel/therapeutic use , Female , Humans , Neoplasm Staging , Prognosis , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Retrospective Studies
9.
Int J Biol Markers ; 37(2): 140-148, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35296171

ABSTRACT

BACKGROUND: This study aimed to investigate the expression pattern and prognostic significance of HOXB13 in rectal cancer. METHODS: HOXB13 expression in rectal cancer and normal adjacent tissues was detected by reverse transcriptase-polymerase chain reaction and immunohistochemistry, and its clinicopathological characteristics and prognosis were statistically tested. Furthermore, we evaluated the association between tumor immune infiltrating cells and HOXB13 using the tumor immune estimation resource (TIMER) database. The potential biological mechanism associated with HOXB13 overexpression was investigated by gene set enrichment analysis (GSEA). RESULTS: The expression of HOXB13 messenger RNA and protein in human rectal cancer tissues were significantly higher than those in the normal adjacent tissues (P < 0.05). HOXB13 expression was significantly correlated with depth of invasion, lymphatic invasion, lymph node metastasis, distant metastasis, and pathological tumor node metastasis stage (P < 0.05). Kaplan-Meier survival curves confirmed that HOXB13 overexpression was correlated negatively with overall survival and disease-free survival in rectal cancer (P < 0.05). Also, multivariate Cox regression analysis demonstrated that HOXB13 expression, age, and lymphatic invasion were independent prognostic factors in rectal cancer (P < 0.05). Plus, the results from the TIMER database indicated that HOXB13 expression has a significant association with several immune cell infiltrates. Finally, the GSEA results indicated that HOXB13 participated in the various immune-associated processes, including natural killer cell-mediated cytotoxicity and the T-cell receptor signaling pathway. CONCLUSION: Our study showed an essential role of HOXB13 in rectal cancer immunity and prognosis. Significantly, the overexpression of HOXB13 leads to the worse prognosis for patients with rectal cancer, which will contribute to understanding molecular mechanisms associated with tumor pathogenesis and prognosis in this disease.


Subject(s)
Homeodomain Proteins , Rectal Neoplasms , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Prognosis , Rectal Neoplasms/genetics , Rectal Neoplasms/metabolism
10.
Dis Markers ; 2022: 8567642, 2022.
Article in English | MEDLINE | ID: mdl-35265227

ABSTRACT

Background: Recent studies have shown that nicotinamide adenosine dinucleotide phosphate oxidase 4 (NOX4) is related to cancer development, proliferation, invasion, epithelial-to-mesenchymal transition, and metastasis. The prognostic value of NOX4 expression although has been reported in various cancers, it remains unclear as several studies have reported conflicting results. Therefore, the purpose of this study was to systematically investigate the prognostic value of NOX4 expression in cancer patients. Method: Appropriate studies were collected by searching the PubMed, EMBASE, and Cochrane library databases, and the prognostic value of NOX4 expression in cancer patients was assessed through a meta-analysis. Results: Nine eligible studies involving 2675 cancer patients were included in this meta-analysis. We found that NOX4 expression is related to prognosis in cancer patients. In particular, high expression of NOX4 was significantly associated with overall survival in patients with gastrointestinal cancer (hazard ratio [HR]: 1.83, 95% confidence interval [CI]: 1.39-2.42, p < 0.001). Conclusion: NOX4 expression is significantly correlated with overall survival in patients with gastrointestinal cancer, indicating that it could be a potential prognostic marker.


Subject(s)
Biomarkers, Tumor/biosynthesis , NADPH Oxidase 4/biosynthesis , Neoplasms/metabolism , Humans , Neoplasms/mortality , Prognosis , Survival Rate
11.
Kaohsiung J Med Sci ; 38(2): 157-164, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35142429

ABSTRACT

Cancer stem cell is considered as an important cause to exacerbate the prognosis. NANOG and POU5F1 are markers for cancer stem cells. The associations between NANOG and POU5F1 expressions with the sorafenib anticancer effects in primary cultured hepatocellular carcinoma (HCC) cells were investigated. Eight primary cultured HCC parent cell lines and 13 subgroups established by flow cytometric sorting using NANOG and POU5F1 as targets were investigated with clinically achievable sorafenib plasma concentrations (5 and 10 µg/mL). Sorafenib showed obvious downregulation of RAF/MEK/ERK signaling pathways and dose-dependent anti-proliferative effects only on s003 parent cell line, which showed the lowest expression of NANOG among all tested cell lines except one downregulated NANOG with upregulated POU5F1 s020 subgroup. Sorafenib also inhibited proliferation in this s020 subgroup but promoted proliferation in its parent cell line. For the only one downregulated NANOG alone s015 subgroup, sorafenib which had no influence on its parent cell line inhibited proliferation in this subgroup. Only the above three cell lines could demonstrate sorafenib antiproliferative effects. On the contrary, sorafenib promoted proliferation in three (s003, s015, s071) out of four upregulated NANOG alone subgroups. On the other hand, Sorafenib showed diverse influence on proliferation among four upregulated POU5F1 alone subgroups. In conclusion, NANOG rather than POU5F1 expression is a critical marker for the anticancer effects of sorafenib on HCC. The sorafenib anticancer effects on HCC cells with high NANOG expression were limited. Sorafenib should be combined with other drug able to target cancer cells with high NANOG expression.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/biosynthesis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Nanog Homeobox Protein/biosynthesis , Octamer Transcription Factor-3/biosynthesis , Sorafenib/therapeutic use , Humans , Treatment Outcome , Tumor Cells, Cultured
12.
Biomolecules ; 12(2)2022 02 11.
Article in English | MEDLINE | ID: mdl-35204793

ABSTRACT

Immune checkpoint inhibitors (ICIs) such as PD1/PD-L1 blockers are an established treatment for many solid cancers. There are currently no approved ICIs for sarcomas, but satisfactory results have been seen in some patients with disseminated disease in certain histological types. Most studies on PD-L1 in sarcoma have used small specimens and there are no clear cutoff values for scoring. We investigated PD-L1 immunoreactivity in high-grade chondrosarcomas (CS), abdominal liposarcoma (LS) and undifferentiated pleomorphic sarcomas (UPS). In total, 230 tumors were stained with SP142 and SP263 assays and evaluated by two clinical pathologists. Immunoreactivity in tumor and immune cells was correlated with clinical outcome. Overall, ≥1% PD-L1 immunoreactivity in tumor cells was found in 11 CS, 26 LS and 59 UPS (SP142 assay) and in 10 CS, 26 LS and 77 UPS (SP263 assay). Most tumors exhibited ≤10% PD-L1 immunoreactivity, but a subset across all three subtypes had >50%. Kaplan-Meier survival analysis showed no significant difference in metastasis-free or overall survival in relation to PD-L1 immunoreactivity in tumor or immune cells for any subtype. As there is a lack of clinical data regarding PD-L1/PD-1 status and therapy response, it is not currently possible to establish clear cutoff values. Patients with high (>50%) PD-L1 immunoreactivity in tumor cells (TC) with the SP263 assay would be a logical group to investigate for potentially beneficial PD1/PD-L1-targeted treatment.


Subject(s)
B7-H1 Antigen , Bone Neoplasms , Chondrosarcoma , Liposarcoma , Sarcoma , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/immunology , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Chondrosarcoma/immunology , Chondrosarcoma/pathology , Humans , Liposarcoma/immunology , Liposarcoma/pathology , Sarcoma/immunology , Sarcoma/pathology , Staining and Labeling
13.
Sci Rep ; 12(1): 2767, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177765

ABSTRACT

In a quest for prognostic biomarkers in early-stage colorectal cancer, we investigated NNMT (nicotinamide N-methyltransferase) in large cohorts of patients. Immunohistochemical examination of 679 patients illustrates that NNMT protein is predominantly expressed in the cancer stroma at varying levels, and about 20% of cancer tissues overexpress NNMT when compared to levels observed in normal colorectal mucosa. Clinical correlation analyses of 572 patients with early-stage cancers reveal that NNMT protein overexpression is significantly associated with shorter overall and disease-free survival, but no such correlation is found in late-stage colorectal cancer. Analyses of TCGA and CPTAC colorectal cancer cohorts show that NNMT mRNA expression is positively correlated with protein levels, is significantly higher in CIMP-high or MSI subtypes than in CIMP-low or MSS subtypes, and is positively correlated with its paralog INMT but not with its interaction partners such as PNMT, ADK, APP, ATF6, BMF, BRD4, CDC37, or CRYZ. In early-stage cancers, NNMT expression is higher in BRAF-mutated than in BRAF wild type tumors but is not affected by KRAS or PIK3CA mutation status. As a cancer stromal protein with important roles in metabolism and cancer epigenetics, NNMT is emerging as a promising biomarker for risk stratification of early-stage cancers.


Subject(s)
Biomarkers, Tumor/biosynthesis , Colorectal Neoplasms , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Nicotinamide N-Methyltransferase/biosynthesis , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/mortality , Disease-Free Survival , Female , Humans , Male , Retrospective Studies , Survival Rate
14.
Life Sci ; 293: 120327, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35065165

ABSTRACT

AIMS: Transcriptional enhanced associate domain (TEAD) transcription factor family, a very important family in the hippo signaling pathway, has been found to play oncogenic functions in the occurrence of various malignant tumors. However, the expression of TEADs in pan-cancer and the important role of TEAD4 in clear cell renal cell carcinoma (ccRCC) have not been analyzed. Herein, we aim to evaluate the expression of TEADs in pan-cancer, and focus on analyzing the role of TEAD4 in the progression of ccRCC. MAIN METHODS: Data from the Cancer Genome Atlas (TCGA) was used to analyze the expression of TEADs in pan-cancer and its clinical correlation. TEAD4 expression in ccRCC tissues, biological functions in vitro and in vivo were analyzed by immunohistochemistry (IHC), western blotting, RNAi and Xenograft assay. Mircode, BioGRID and g: Profiler website were used to build a ceRNA network and downstream pathway prediction. KEY FINDINGS: TEAD1, TEAD2, TEAD3 and TEAD4 were highly expressed in 3, 6, 5, and 12 types of cancer tissues, respectively, indicating that TEAD4 is most closely related to tumor progression. Among the cancers with high TEAD4 expression, the expression of TEAD4 has the greatest correlation with the poor prognosis of ccRCC. We also found the malignant phenotypes of ccRCC cells in vitro and vivo have been significantly suppressed by silencing TEAD4. SIGNIFICANCE: TEADs, especially TEAD4, were overexpressed in many human tumors. This study is the first to show that TEAD4 acts as an oncogene in ccRCC and may be an important factor in progress of ccRCC.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinogens/metabolism , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , TEA Domain Transcription Factors/biosynthesis , Animals , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Gene Regulatory Networks/physiology , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , TEA Domain Transcription Factors/genetics , Xenograft Model Antitumor Assays/methods
15.
JCI Insight ; 7(4)2022 02 22.
Article in English | MEDLINE | ID: mdl-35015732

ABSTRACT

Sustained proliferative signaling and resisting cell death are hallmarks of cancer. Zinc finger protein 277 (ZNF277; murine Zfp277), a transcription factor regulating cellular senescence, is overexpressed in colon cancer, but its actions in intestinal homeostasis and neoplasia are unclear. Using human and murine intestine, human colon cancer cells, and ApcMin/+ mice with dysregulated ß-catenin signaling and exuberant intestinal neoplasia, we explored the actions of ZNF277/Zfp277 and defined the underlying mechanisms. In normal human and murine intestine, ZNF277/Zfp277 was expressed uniquely in early stem cell progenitors, undifferentiated transit-amplifying cells (TACs). Zfp277 was overexpressed in the ApcMin/+ mouse colon, implicating ZNF277/Zfp277 as a transcriptional target of ß-catenin signaling. We confirmed this by showing ß-catenin knockdown reduced ZNF277 expression and, using chromatin IP, identified 2 ß-catenin binding sites in the ZNF277 promoter. Zfp277 deficiency attenuated intestinal epithelial cell proliferation and tumor formation, and it strikingly prolonged ApcMin/+ mouse survival. RNA-Seq and PCR analyses revealed that Zfp277 modulates expression of genes in key cancer pathways, including ß-catenin signaling, the HOXD family that regulates development, and p21WAF1, a cell cycle inhibitor and tumor suppressor. In both human colon cancer cells and the murine colon, ZNF277/Zfp277 deficiency induced p21WAF1 expression and promoted senescence. Our findings identify ZNF277/Zfp277 as both a TAC marker and colon cancer oncogene that regulates cellular proliferation and senescence, in part by repressing p21WAF1 expression.


Subject(s)
Colon/metabolism , Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Intestinal Mucosa/metabolism , Neoplasms, Experimental , Zinc Fingers/genetics , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Proliferation , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA-Binding Proteins/biosynthesis , Humans , Intestinal Mucosa/pathology , Mice , Promoter Regions, Genetic , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Transcription Factors , Wnt Signaling Pathway/genetics
16.
Biomed Res Int ; 2022: 2067540, 2022.
Article in English | MEDLINE | ID: mdl-35036428

ABSTRACT

BACKGROUND: This study is aimed at constructing a risk signature to predict survival outcomes of ORCA patients. METHODS: We identified differentially expressed autophagy-related genes (DEARGs) based on the RNA sequencing data in the TCGA database; then, four independent survival-related ARGs were identified to construct an autophagy-associated signature for survival prediction of ORCA patients. The validity and robustness of the prognostic model were validated by clinicopathological data and survival data. Subsequently, four independent prognostic DEARGs that composed the model were evaluated individually. RESULTS: The expressions of 232 autophagy-related genes (ARGs) in 127 ORCA and 13 control tissues were compared, and 36 DEARGs were filtered out. We performed functional enrichment analysis and constructed protein-protein interaction network for 36 DEARGs. Univariate and multivariate Cox regression analyses were adopted for searching prognostic ARGs, and an autophagy-associated signature for ORCA patients was constructed. Eventually, 4 desirable independent survival-related ARGs (WDR45, MAPK9, VEGFA, and ATIC) were confirmed and comprised the prognostic model. We made use of multiple ways to verify the accuracy of the novel autophagy-related signature for survival evaluation, such as receiver-operator characteristic curve, Kaplan-Meier plotter, and clinicopathological correlational analyses. Four independent prognostic DEARGs that formed the model were also associated with the prognosis of ORCA patients. CONCLUSIONS: The autophagy-related risk model can evaluate OS for ORCA patients independently since it is accurate and stable. Four prognostic ARGs that composed the model can be studied deeply for target treatment.


Subject(s)
Autophagy , Biomarkers, Tumor , Models, Biological , Mouth Neoplasms , Neoplasm Proteins , Nomograms , Transcriptome , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Databases, Nucleic Acid , Disease-Free Survival , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/mortality , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Survival Rate
17.
Eur J Cancer ; 163: 35-43, 2022 03.
Article in English | MEDLINE | ID: mdl-35032815

ABSTRACT

BACKGROUND: Low human epidermal growth factor receptor 2 (HER2) expression is emerging as an actionable biomarker for the treatment of breast cancer (BC) with novel anti-HER2 drugs. However, the evolution of this biomarker during the course of disease is still poorly characterised, and controversial data exist on its prognostic implications. METHODS: We reviewed data of patients with HER2-negative BC according to the latest ASCO/CAP guidelines referred between January 2014 and December 2020. We grouped patients based on the immunohistochemistry (IHC) expression of HER2, HER2-zero (IHC 0) and HER2-low subgroup (IHC 1+ or 2+/ISH-negative) and evaluated the evolution of HER2 expression between the primary tumour and the first biopsy collected in the advanced setting. Disease-free survival, overall survival and progression-free survival were compared between patients with HER2-zero and HER2-low expression on the primary tumour. RESULTS: 232 patients were included in the analysis. Among the overall population, there was a relevant discordance in HER2 expression between the primary tumour and the matched biopsy (K = 0.33, 95%CI 0.21-0.44): 44% of the HER2-zero primary tumour showed an increased HER2 score on biopsy, and 22% of the HER2-low primary tumours turned into HER2-IHC 0. The findings in the sub-populations of hormone-receptors positive (K = 0.32, 95%CI 0.19-0.45) and triple-negative tumours (K = 0.18, 95%CI -0.09-0.46) were consistent with the primary analysis. No difference in survival outcomes was observed between HER2-low and HER2-zero primary tumours. CONCLUSIONS: HER2-low expression is dynamic in BC and may be enriched in the advanced-stage setting. No prognostic significance was demonstrated for HER2-low expression.


Subject(s)
Breast Neoplasms , Receptor, ErbB-2 , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Female , Humans , Immunohistochemistry , Prognosis , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/metabolism
18.
Signal Transduct Target Ther ; 7(1): 9, 2022 01 14.
Article in English | MEDLINE | ID: mdl-35027529

ABSTRACT

Lung adenocarcinoma (LUAD) and squamous carcinoma (LUSC) are two major subtypes of non-small cell lung cancer with distinct pathologic features and treatment paradigms. The heterogeneity can be attributed to genetic, transcriptional, and epigenetic parameters. Here, we established a multi-omics atlas, integrating 52 single-cell RNA sequencing and 2342 public bulk RNA sequencing. We investigated their differences in genetic amplification, cellular compositions, and expression modules. We revealed that LUAD and LUSC contained amplifications occurring selectively in subclusters of AT2 and basal cells, and had distinct cellular composition modules associated with poor survival of lung cancer. Malignant and stage-specific gene analyses further uncovered critical transcription factors and genes in tumor progression. Moreover, we identified subclusters with proliferating and differentiating properties in AT2 and basal cells. Overexpression assays of ten genes, including sub-cluster markers AQP5 and KPNA2, further indicated their functional roles, providing potential targets for early diagnosis and treatment in lung cancer.


Subject(s)
Adenocarcinoma of Lung , Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism
19.
Life Sci ; 293: 120264, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35031262

ABSTRACT

AIMS: This study evaluated the association of mucinous metaplasia (MM) with tumor cell proliferation, androgen receptor (AR) expression and invasiveness in Pten conditional knockout mice and the prognostic value of MM markers for patients with PCa. MAIN METHODS: Prostatic lobes samples from genetic engineered mouse model Ptenf/f and Pb-Cre4/Ptenf/f were submitted for histopathological analysis and tissue expression of AR, the proliferation marker Ki67, alpha-smooth muscle actin, and laminin. RNAseq data of prostatic lobes samples were analyzed searching for MM gene expression patterns. We also investigated gene and protein expression related to MM in human PCa public databases. KEY FINDINGS: All knockout animals analyzed showed at least one area of stroma-invading MM, which was absent in the control animals. The tumoral regions of MM showed a proliferative index 5 times higher than other tumoral areas and low expression of the AR (less than 20% of the cells were AR-positive). Disrupted basement membrane areas were observed in MM. The mouse and human PCa transcriptomes exhibited increased expression of the MM markers such as MUC1, MUC19, MUC4, MUC5AC, MUC5B, and TFF3. Gene expression profile was associated with castration-resistant prostate cancer (CRPC) and with a lower probability of freedom from biochemical recurrence. SIGNIFICANCE: The expression of goblet cell genes, such as MUC1, MUC5AC, MUC5B, and TFF3 have significant prognostic value for PCa patients and represent another class of potential therapeutic targets.


Subject(s)
Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/deficiency , Mucins/biosynthesis , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms, Castration-Resistant/metabolism , Animals , Biomarkers, Tumor/genetics , Databases, Genetic , Male , Metaplasia/genetics , Metaplasia/metabolism , Metaplasia/pathology , Mice , Mice, Knockout , Mice, Transgenic , Mucins/genetics , PTEN Phosphohydrolase/genetics , Prognosis , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology
20.
Life Sci ; 293: 120328, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35051418

ABSTRACT

Paclitaxel (PTX) resistance contributes to mortality in epithelial ovarian cancer (EOC). Aerobic glycolysis is elevated in the tumor environment and may influence resistance to PTX in EOC. KH domain-containing, RNA-binding signal transduction-associated protein 3 (KHDRBS3) is an RNA binding protein that is up-regulated in EOC, but its underlying mechanism in EOC is unclear. Here, we investigate the role of KHDRBS3 in glycolysis and increased resistance to PTX. Expression of KHDRBS3 and Claudin (CLDN6) were measured in EOC tissue and cells by quantitative real-time PCR, western blotting and immunohistochemistry. The biological functions of KHDRBS3, MIR17HG and CLDN6 were examined using MTT, colony formation, apoptosis and seahorse assays in vitro. For in vivo experiments, a xenograft model was used to investigate the effects of KHDRBS3 and MIR17HG in EOC. Here, we investigate the role of KHDRBS3 in glycolysis and increased resistance to PTX. The expression of KHDRBS3 was up-regulated in PTX-resistant cells. KHDRBS3 knockdown restrained the IC50 of PTX, cell proliferation, colony formation and glycolysis in SKOV3-R and A2780-R cells in vitro and enhanced PTX sensitivity in a xenograft mouse model in vivo. KHDRBS3 interacts with lncRNA MIR17HG, which is down-regulated in EOC tissue and cells. The effect of KHDRBS3 overexpression on PTX resistance and glycolysis was rescued by MIR17HG overexpression. Additionally, MIR17HG interacts with the 3'UTR of CLDN6 and negatively regulates CLDN6 expression. MIR17HG overexpression suppressed the IC50 of PTX and glycolysis by targeting CLDN6. Our results reveal a KHDRBS3-MIR17HG-CLDN6 regulatory axis that contributes to enhanced glycolysis in EOC and represents a potential target for therapy.


Subject(s)
Claudins/biosynthesis , Drug Resistance, Neoplasm/drug effects , Glycolysis/drug effects , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , RNA, Long Noncoding/biosynthesis , RNA-Binding Proteins/biosynthesis , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/physiology , Female , Glycolysis/physiology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/physiology , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL