Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters

Country/Region as subject
Publication year range
1.
CA Cancer J Clin ; 73(1): 72-112, 2023 01.
Article in English | MEDLINE | ID: mdl-35916666

ABSTRACT

Sinonasal malignancies make up <5% of all head and neck neoplasms, with an incidence of 0.5-1.0 per 100,000. The outcome of these rare malignancies has been poor, whereas significant progress has been made in the management of other cancers. The objective of the current review was to describe the incidence, causes, presentation, diagnosis, treatment, and recent developments of malignancies of the sinonasal tract. The diagnoses covered in this review included sinonasal undifferentiated carcinoma, sinonasal adenocarcinoma, sinonasal squamous cell carcinoma, and esthesioneuroblastoma, which are exclusive to the sinonasal tract. In addition, the authors covered malignances that are likely to be encountered in the sinonasal tract-primary mucosal melanoma, NUT (nuclear protein of the testis) carcinoma, and extranodal natural killer cell/T-cell lymphoma. For the purpose of keeping this review as concise and focused as possible, sarcomas and malignancies that can be classified as salivary gland neoplasms were excluded.


Subject(s)
Carcinoma , Maxillary Sinus Neoplasms , Melanoma , Nose Neoplasms , Paranasal Sinuses , Humans , Carcinoma/diagnosis , Maxillary Sinus Neoplasms/diagnosis , Maxillary Sinus Neoplasms/pathology , Nasal Cavity/pathology , Nose Neoplasms/diagnosis , Nose Neoplasms/epidemiology , Nose Neoplasms/therapy , Paranasal Sinuses/pathology
2.
Cell ; 149(5): 1098-111, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22632973

ABSTRACT

Akt kinase plays a central role in cell growth, metabolism, and tumorigenesis. The TRAF6 E3 ligase orchestrates IGF-1-mediated Akt ubiquitination and activation. Here, we show that Akt ubiquitination is also induced by activation of ErbB receptors; unexpectedly, and in contrast to IGF-1 induced activation, the Skp2 SCF complex, not TRAF6, is a critical E3 ligase for ErbB-receptor-mediated Akt ubiquitination and membrane recruitment in response to EGF. Skp2 deficiency impairs Akt activation, Glut1 expression, glucose uptake and glycolysis, and breast cancer progression in various tumor models. Moreover, Skp2 overexpression correlates with Akt activation and breast cancer metastasis and serves as a marker for poor prognosis in Her2-positive patients. Finally, Skp2 silencing sensitizes Her2-overexpressing tumors to Herceptin treatment. Our study suggests that distinct E3 ligases are utilized by diverse growth factors for Akt activation and that targeting glycolysis sensitizes Her2-positive tumors to Herceptin treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/drug therapy , Cell Transformation, Neoplastic , F-Box Proteins/metabolism , Glycolysis , S-Phase Kinase-Associated Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Humans , Mice , Receptor, ErbB-2/metabolism , S-Phase Kinase-Associated Proteins/genetics , Trastuzumab , Ubiquitination
3.
Ann Hum Genet ; 87(6): 274-284, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37712180

ABSTRACT

The complex mechanism of colorectal cancer development is closely associated with epigenetic modifications and is caused by overexpression and/or inactivation of oncogenes. Histone modifying enzymes catalyze histone modifications to alter gene expression, which plays a crucial role in the development and progression of colorectal cancer. Currently, there is more frequent study on histone acetylation, methylation, and phosphorylation, and their mechanisms in colorectal cancer development are clearer. This article elaborates on the role of histone modification in epigenetics in colorectal cancer development and discusses recent advances in using it as biomarkers and therapeutic targets for the treatment of colorectal cancer. The review aims to demonstrate the significant role of histone modification as a new therapeutic target in colorectal cancer and provides insights into the novel diagnostic and therapeutic options it offers.

4.
BMC Gastroenterol ; 23(1): 292, 2023 Aug 31.
Article in English | MEDLINE | ID: mdl-37653392

ABSTRACT

BACKGROUND: Several proteins in the tripartite-motif (TRIM) family are associated with the development of colorectal cancer (CRC), but research on the role of TRIM69 was lacking. The present study examined the correlation between TRIM69 expression and colon adenocarcinoma (COAD). METHODS: mRNA sequencing data for COAD patients was extracted from The Cancer Genome Atlas to analyze correlations between TRIM69 expression and patients' clinical features as well as survival. Potential associations with immune cells and chemosensitivity also were predicted using various algorithms in the TIMER, Limma, clusterProfiler, GeneMANIA, and Gene Set Cancer Analysis platforms. Subsequently, polymerase chain reaction analysis and immunohistochemical staining were used to detect TRIM69 expression in COAD tissue samples from real-world patients. RESULTS: TRIM69 expression was lower in COAD tissues than in normal tissues and correlated with the pathologic stage and metastasis (M category). Additionally, TRIM69 was found to be involved in several immune-related pathways, notably the NOD-like signaling pathway. These results suggest that high TRIM69 expression has the potential to enhance tumor sensitivity to 5-fluorouracil and programmed cell death protein 1 (PD-1) blockers. CONCLUSIONS: From our findings that TRIM69 expression was significantly reduced in COAD compared with non-cancer tissues and associated with pathologic stage and metastasis, we conclude that increasing TRIM69 expression and/or activity may help to improve therapeutic outcomes. Accordingly, TRIM69 represents a potentially valuable marker of metastasis and target for adjuvant therapy in COAD.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Humans , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Fluorouracil/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Programmed Cell Death 1 Receptor , Algorithms , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics
6.
Oncology (Williston Park) ; 35(7): 410-420, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34264569

ABSTRACT

Bladder cancer is the sixth most common cancer in the United States, with an estimated 81,400 new cases in 2020. Although bladder cancer has 4 stages, for systemic treatment we recognize 3 clinical stages: non-muscle-invasive bladder cancer (NMIBC), muscle-invasive bladder cancer (MIBC), and locally advanced/metastatic urothelial carcinoma (mUC). Approximately 70% to 80% of patients present with NMIBC at diagnosis and have an excellent 5-year overall survival (OS) of 69.2% to 95.8%.1 About 10% to 15% of patients present with MIBC at the time of diagnosis and have about a 50% chance of progressing to metastatic disease.2 mUC accounts for 10% to 15% of all bladder cancers at diagnosis, with a 5-year OS for patients of fewer than 10% with platinum-based chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Immunoconjugates/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Immunoconjugates/administration & dosage , Immunoconjugates/adverse effects , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Randomized Controlled Trials as Topic , Urinary Bladder Neoplasms/pathology
7.
Jpn J Clin Oncol ; 48(9): 815-821, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30053015

ABSTRACT

OBJECTIVE: To analyze the failure pattern and clinical efficacy of elective nodal irradiation in patients with esophageal squamous cell carcinoma who received neoadjuvant chemoradiotherapy. METHODS: We retrospectively analyzed 173 esophageal squamous cell carcinoma patients who received neoadjuvant chemoradiotherapy including elective nodal irradiation from 2002 to 2015. Failure pattern, survival and recurrence sites were analyzed. For patients with regional recurrences, the recurrence sites were analyzed in relation to an imaginary field of involved field irradiation. RESULTS: After a median follow-up of 55.5 months, 58 patients (33.5%) developed recurrences. Among 22 patients with regional recurrences, infield failure occurred in 19 patients (86.4%) and outfield failure occurred in 3 patients (13.6%), of whom only 1 patient had an outfield failure without synchronous distant metastasis. Compared with the involved field irradiation field, 6 patients' failure sites (27.3%) were located in the involved field irradiation field and 13 patients' failure sites (59.1%) were out of the involved field irradiation field but within the elective nodal irradiation field. CONCLUSIONS: Since only a minority of patients had outfield regional recurrences, neoadjuvant chemoradiotherapy with elective nodal irradiation yields satisfactory infield control. More than half of the regional recurrences occurred within the elective nodal irradiation field but out of the involved field irradiation field. Prospective evaluation of whether elective nodal irradiation could lead to an improved survival outcome is necessary.


Subject(s)
Carcinoma, Squamous Cell/therapy , Chemoradiotherapy , Esophageal Neoplasms/therapy , Neoadjuvant Therapy , Adult , Aged , Disease-Free Survival , Esophageal Squamous Cell Carcinoma , Female , Follow-Up Studies , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local/therapy , Prognosis , Retrospective Studies , Treatment Failure
8.
Biochem Biophys Res Commun ; 445(3): 566-71, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24561244

ABSTRACT

The homing ability of hematopoietic stem cells (HSCs) was a critical step for transplantation and subsequent hematopoiesis. Although the HSC transplantation was widely used for many diseases, the mechanism by which HSC homing was regulated remained poorly understood. F-box protein S-phase kinase associated protein2 (Skp2), a component of the Skp2-SCF E3 ligase complex, was regarded as a cell cycle regulator by controlling the level of p21 and p27 through ubiquitination. We recently reported an important role of Skp2 in maintaining HSC pool size, quiescent stage and self-renewal ability. In this current study, we showed that Skp2 was a novel and critical regulator for maintaining the homing of HSCs as well as their residence in the endosteal niche. Microarray analysis together with biochemical validations revealed that Skp2 deficiency profoundly reduced the expression of ß-catenin and its target genes. Knockdown of ß-catenin mimicked the decline of HSC homing upon Skp2 deficiency, suggesting that Skp2 may regulate ß-catenin and its target gene expression to orchestrate HSC homing. Our study not only identified Skp2 as a new regulator for maintaining ß-catenin expression and HSC homing, but also suggested that Skp2 may serve as a predictive marker for monitoring the transplantation efficiency.


Subject(s)
Down-Regulation , Hematopoietic Stem Cells/cytology , S-Phase Kinase-Associated Proteins/metabolism , beta Catenin/genetics , Animals , Cell Cycle , Cell Movement , Cells, Cultured , Gene Deletion , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , S-Phase Kinase-Associated Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , beta Catenin/metabolism
9.
BMC Cancer ; 13: 302, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23800247

ABSTRACT

BACKGROUND: There is a strong need for early assessment of tumor response to chemotherapy in order to avoid the adverse effects of unnecessary chemotherapy and to allow early transition to second-line therapy. The purpose of this study was to determine the feasibility of ultrasonic spectral analysis for the in vivo characterization of changes in tumor microstructure in the evaluation of tumor response to chemotherapy using diagnostic ultrasound. METHODS: Experiments were approved by the regional animal care committee. Twenty-four MCF-7 breast cancer bearing nude mice were treated with adriamycin or sterile saline administered by intraperitoneal injection. Ultrasonic radio-frequency (RF) data was collected using a clinically available ultrasound scanner (6-MHz linear transducer). Linear regression parameters (spectral slope and midband-fit) regarding the calibrated power spectra from the RF signals were tested to monitor tumor response to treatment. The section equivalent to the ultrasound imaging plane was stained with hematoxylin and eosin to allow for assessment of the density of tumor cell nuclei. RESULTS: Treatment with adriamycin significantly reduced tumor growth in comparison with the control group (p = 0.003). Significant changes were observed in the ultrasonic parameters of the treated relative to the untreated tumors (p < 0.05). The spectral slope increased by 48.5%, from -10.66 ± 2.96 to -5.49 ± 2.69; the midband-fit increased by 12.8%, from -57.10 ± 7.68 to -49.81 ± 5.40. Treated tumors were associated with a significant decrease in the density of tumor cell nuclei as compared with control tumors (p < 0.001). CONCLUSIONS: Ultrasonic spectral analysis can detect changes in tumor microstructure after chemotherapy, and this will be helpful in the early evaluation tumor response to chemotherapy.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Spectrum Analysis/methods , Animals , Antibiotics, Antineoplastic/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Female , Fourier Analysis , Linear Models , Mice , Mice, Nude , Random Allocation , Signal Processing, Computer-Assisted , Single-Blind Method , Ultrasonography
10.
Digit Health ; 9: 20552076231203902, 2023.
Article in English | MEDLINE | ID: mdl-37766908

ABSTRACT

Background: Although surgical methods are the most effective treatments for colon adenocarcinoma (COAD), the cure rates remain low, and recurrence rates remain high. Furthermore, platelet adhesion-related genes are gaining attention as potential regulators of tumorigenesis. Therefore, identifying the mechanisms responsible for the regulation of these genes in patients with COAD has become important. The present study aims to investigate the underlying mechanisms of platelet adhesion-related genes in COAD patients. Methods: The present study was an experimental study. Initially, the effects of platelet number and related genomic alteration on survival were explored using real-world data and the cBioPortal database, respectively. Then, the differentially expressed platelet adhesion-related genes of COAD were analyzed using the TCGA database, and patients were further classified by employing the non-negative matrix factorization (NMF) analysis method. Afterward, some of the clinical and expression characteristics were analyzed between clusters. Finally, least absolute shrinkage and selection operator regression analysis was used to establish the prognostic nomogram. All data analyses were performed using the R package. Results: High platelet counts are associated with worse survival in real-world patients, and alternations to platelet adhesion-related genes have resulted in poorer prognoses, based on online data. Based on platelet adhesion-related genes, patients with COAD were classified into two clusters by NMF-based clustering analysis. Cluster2 had a better overall survival, when compared to Cluster1. The gene copy number and enrichment analysis results revealed that two pathways were differentially enriched. In addition, the differentially expressed genes between these two clusters were enriched for POU6F1 in the transcription factor signaling pathway, and for MATN3 in the ceRNA network. Finally, a prognostic nomogram, which included the ALOX12 and ACTG1 genes, was established based on the platelet adhesion-related genes, with a concordance (C) index of 0.879 (0.848-0.910). Conclusion: The mRNA expression-based NMF was used to reveal the potential role of platelet adhesion-related genes in COAD. The series of experiments revealed the feasibility of targeting platelet adhesion-associated gene therapy.

11.
J Virol ; 85(21): 11291-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21880770

ABSTRACT

Epstein-Barr virus (EBV)-encoded molecules have been detected in the tumor tissues of several cancers, including nasopharyngeal carcinoma (NPC), suggesting that EBV plays an important role in tumorigenesis. However, the nature of EBV with respect to genome width in vivo and whether EBV undergoes clonal expansion in the tumor tissues are still poorly understood. In this study, next-generation sequencing (NGS) was used to sequence DNA extracted directly from the tumor tissue of a patient with NPC. Apart from the human sequences, a clinically isolated EBV genome 164.7 kb in size was successfully assembled and named GD2 (GenBank accession number HQ020558). Sequence and phylogenetic analyses showed that GD2 was closely related to GD1, a previously assembled variant derived from a patient with NPC. GD2 contains the most prevalent EBV variants reported in Cantonese patients with NPC, suggesting that it might be the prevalent strain in this population. Furthermore, GD2 could be grouped into a single subtype according to common classification criteria and contains only 6 heterozygous point mutations, suggesting the monoclonal expansion of GD2 in NPC. This study represents the first genome-wide analysis of a clinical isolate of EBV directly extracted from NPC tissue. Our study reveals that NGS allows the characterization of genome-wide variations of EBV in clinical tumors and provides evidence of monoclonal expansion of EBV in vivo. The pipeline could also be applied to the study of other pathogen-related malignancies. With additional NGS studies of NPC, it might be possible to uncover the potential causative EBV variant involved in NPC.


Subject(s)
DNA, Viral/genetics , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Nasopharyngeal Neoplasms/virology , Carcinoma , China , Cluster Analysis , DNA, Viral/chemistry , Herpesvirus 4, Human/classification , High-Throughput Nucleotide Sequencing , Humans , Molecular Sequence Data , Nasopharyngeal Carcinoma , Phylogeny , Sequence Analysis, DNA , Sequence Homology
12.
J Transl Med ; 10: 249, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23232108

ABSTRACT

BACKGROUND: Epidermal growth factor receptor (EGFR) is suggested to predict the radiosensitivity and/or prognosis of human esophageal squamous cell carcinoma (ESCC). The objective of this study was to investigate the efficacy of Nimotuzumab (an anti-EGFR monoclonal antibody) on ESCC radiotherapy (RT) and underlying mechanisms. METHODS: Nimotuzumab was administrated to 2 ESCC cell lines KYSE30 and TE-1 treated with RT. Cell growth, colony formation and apoptosis were used to measure anti-proliferation effects. The method of RNA interference was used to investigate the role of insulin-like growth factor binding protein-3 (IGFBP-3) in ESCC cells radiosensitivity treated with Nimotuzumab. In vivo effect of Nimotuzumab on ESCC radiotherapy was done using a mouse xenograft model. RESULTS: Nimotuzumab enhanced radiation response of KYSE30 cells (with high EGFR expression) in vitro, as evidenced by increased radiation-inhibited cell growth and colony formation and radiation-mediated apoptosis. Mechanism study revealed that Nimotuzumab inhibited phosphorylated EGFR (p-EGFR) induced by EGF in KYSE30 cells. In addition, knockdown of IGFBP-3 by short hairpin RNA significantly reduced KYSE30 cells radiosensitivity (P<0.05), and even after the administration of Nimotuzumab, the RT response of IGFBP-3 silenced KYSE30 cells was not enhanced (P>0.05). In KYSE30 cell xenografts, Nimotuzumab combined with radiation led to significant tumor growth delay, compared with that of radiation alone (P=0.029), and also with IGFBP-3 up-regulation in tumor tissue. CONCLUSIONS: Nimotuzumab could enhance the RT effect of ESCC cells with a functional active EGFR pathway. In particular, the increased ESCC radiosensitivity by Nimotuzumab might be dependent on the up-regulation of IGFBP-3 through EGFR-dependent pathway.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Carcinoma, Squamous Cell/metabolism , ErbB Receptors/metabolism , Esophageal Neoplasms/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Radiation Tolerance , Up-Regulation , Apoptosis , Blotting, Western , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Esophageal Neoplasms/pathology , Humans , Immunohistochemistry
13.
Clin Genitourin Cancer ; 20(5): 415-422, 2022 10.
Article in English | MEDLINE | ID: mdl-35688679

ABSTRACT

BACKGROUND: Overexpression of the epidermal growth factor receptor (EGFR) and its ligands occur frequently in renal cell carcinoma (RCC). Combined vascular endothelial growth factor receptor (VEGF-R) and EGFR inhibition may overcome resistance to VEGF-R inhibitor monotherapy. We performed a dose-escalation phase II study of sunitinib plus erlotinib in advanced renal cell carcinoma. PATIENTS AND METHODS: Patients with metastatic clear cell or papillary RCC were eligible. Prior therapy was allowed except sunitinib or erlotinib. Three dose levels of erlotinib (50, 100, 150 mg daily) were evaluated in combination with sunitinib 50 mg. Thirty-seven patients were treated at maximum tolerated dose to determine efficacy. The primary endpoint was 8-month progression-free survival (PFS) rate. The trial was powered to assess for a difference between a median PFS of less than 50% with a targeted 70% PFS for the combination. RESULTS: The 8-month PFS rate was 40% (95% CI: 23-56). Median PFS was 5.8 months (95% CI: 4.1-9.7) and median overall survival (OS) was 26.3 months (95% CI: 16.1-34.0). The objective response rate was 22% and an additional 59% of patients had stable disease for at least 6 weeks. The most common adverse events for all grades were diarrhea, rash, fatigue, and dysgeusia. Dose reduction in 1 or both of the drugs was undertaken in 17 (46%) patients, while 5 (14%) discontinued study therapy due to toxicity. CONCLUSION: While sunitinib and erlotinib are combinable,the 8-month PFS rate did not suggest efficacy improvement over sunitinib monotherapy (NCT00425386).


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Renal Cell/pathology , Disease-Free Survival , ErbB Receptors , Erlotinib Hydrochloride/adverse effects , Humans , Kidney Neoplasms/pathology , Receptors, Vascular Endothelial Growth Factor , Sunitinib/therapeutic use , Vascular Endothelial Growth Factor A
14.
Drugs Aging ; 38(2): 111-123, 2021 02.
Article in English | MEDLINE | ID: mdl-33559101

ABSTRACT

Prostate cancer is common, particularly in older patients, as the risk of getting prostate cancer increases with age. Cancer therapy brings unique challenges in older patients, as this population is vulnerable to many side effects and drug interactions, and they have varying degrees of frailty, which may limit the use of these therapies. The US FDA has recently approved several novel next-generation hormonal therapies for patients with various stages of prostate cancer, giving patients more treatment options. These therapies (e.g., apalutamide, enzalutamide, darolutamide, and abiraterone) have unique side effects that the practitioner must consider when evaluating therapeutic treatments in any patient, and these side effects also affect older patients differently. Here, we review the mechanism of action and metabolism of the next-generation hormonal therapies; report efficacy and safety data from trials of these agents in non-metastatic castration-resistant prostate cancer, metastatic hormone-sensitive prostate cancer, and metastatic castration-resistant prostate cancer; and discuss the intricacies of treating older men with prostate cancer. Key takeaways include the fact that enzalutamide and apalutamide may increase the risk of falls and fractures in older patients. Abiraterone requires the concurrent use of low-dose glucocorticoids, which can lead to side effects in older patients. Lastly, drug-drug interactions should be considered in older patients using multiple medications.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Aged , Androgen Antagonists , Humans , Male , Phenylthiohydantoin
15.
Radiother Oncol ; 149: 228-235, 2020 08.
Article in English | MEDLINE | ID: mdl-32474127

ABSTRACT

PURPOSE: To compare survival outcomes and radiation pneumonitis (RP) between intensity-modulated radiotherapy (IMRT) and three-dimensional conformal radiotherapy (3DCRT) in patients with esophageal cancer (EC) who underwent definitive chemoradiation therapy (CRT). METHODS: Clinical characteristics and dose-volume histogram parameters were collected for 388 EC patients who received definitive CRT with either IMRT (n = 297) or 3DCRT (n = 91) from 2010 through 2017. Dosimetric parameters, survival end-points, and symptomatic RP (grade ≥2) were compared between groups. Propensity score matching (PSM) was performed to balance potential confounding factors. Univariate and multivariate logistic regression analyses were applied to identify predictors of RP. RESULTS: Compared with 3DCRT, IMRT was significantly associated with better overall survival (OS; P = 0.001), progression-free survival (PFS; P = 0.008), and distant metastasis-free survival (P = 0.011), but not with locoregional failure-free survival (P = 0.721). Moreover, IMRT demonstrated a remarkably lower risk of RP than 3DCRT (5.4% vs 23.1%, P < 0.001). PSM analysis further confirmed the clinical benefit of IMRT. In the matched cohort, radiation modality was independently correlated with OS and PFS. On multivariate analysis, smoking history (odds ratio [OR]: 4.225, P = 0.002), primary tumor length (OR: 2.764, P = 0.049), radiation modality (OR: 10.760, P < 0.001), planning target volume (OR: 1.004, P < 0.001), and lung V20 (OR: 1.286, P = 0.002) were found to be significant predictors of RP. CONCLUSIONS: Compared with 3DCRT, IMRT was associated with more favorable survival and a reduced risk of RP after definitive CRT, supporting the routine use of IMRT for EC.


Subject(s)
Esophageal Neoplasms , Radiation Pneumonitis , Radiotherapy, Conformal , Radiotherapy, Intensity-Modulated , Esophageal Neoplasms/therapy , Humans , Propensity Score , Radiation Pneumonitis/etiology , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted , Radiotherapy, Conformal/adverse effects , Radiotherapy, Intensity-Modulated/adverse effects , Retrospective Studies , Treatment Outcome
16.
J Med Chem ; 59(1): 253-263, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26652247

ABSTRACT

Methylation of histone lysine residues plays important roles in gene expression regulation as well as cancer initiation. Lysine specific demethylase 1 (LSD1) is responsible for maintaining balanced methylation levels at histone H3 lysine 4 (H3K4). LSD1 is a drug target for certain cancers, due to important functions of methylated H3K4 or LSD1 overexpression. We report the design, synthesis, and structure-activity relationships of 3-(piperidin-4-ylmethoxy)pyridine containing compounds as potent LSD1 inhibitors with Ki values as low as 29 nM. These compounds exhibited high selectivity (>160×) against related monoamine oxidase A and B. Enzyme kinetics and docking studies suggested they are competitive inhibitors against a dimethylated H3K4 substrate and provided a possible binding mode. The potent LSD1 inhibitors can increase cellular H3K4 methylation and strongly inhibit proliferation of several leukemia and solid tumor cells with EC50 values as low as 280 nM, while they had negligible effects on normal cells.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Piperidines/chemical synthesis , Piperidines/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Drug Screening Assays, Antitumor , Histone Demethylases/genetics , Histones/metabolism , Humans , Kinetics , Methylation , Models, Molecular , Molecular Docking Simulation , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/pharmacology , Structure-Activity Relationship , Substrate Specificity
17.
J Hematol Oncol ; 9: 24, 2016 Mar 12.
Article in English | MEDLINE | ID: mdl-26970896

ABSTRACT

BACKGROUND: Mixed lineage leukemia (MLL) gene translocations are found in ~75% infant and 10% adult acute leukemia, showing a poor prognosis. Lysine-specific demethylase 1 (LSD1) has recently been implicated to be a drug target for this subtype of leukemia. More studies using potent LSD1 inhibitors against MLL-rearranged leukemia are needed. METHODS: LSD1 inhibitors were examined for their biochemical and biological activities against LSD1 and MLL-rearranged leukemia as well as other cancer cells. RESULTS: Potent LSD1 inhibitors with biochemical IC50 values of 9.8-77 nM were found to strongly inhibit proliferation of MLL-rearranged leukemia cells with EC50 of 10-320 nM, while these compounds are generally non-cytotoxic to several other tumor cells. LSD1 inhibition increased histone H3 lysine 4 (H3K4) methylation, downregulated expression of several leukemia-relevant genes, induced apoptosis and differentiation, and inhibited self-renewal of stem-like leukemia cells. Moreover, LSD1 inhibitors worked synergistically with inhibition of DOT1L, a histone H3 lysine 79 (H3K79) methyltransferase, against MLL-rearranged leukemia. The most potent LSD1 inhibitor showed significant in vivo activity in a systemic mouse model of MLL-rearranged leukemia without overt toxicities. Mechanistically, LSD1 inhibitors caused significant upregulation of several pathways that promote hematopoietic differentiation and apoptosis. CONCLUSIONS: LSD1 is a drug target for MLL-rearranged leukemia, and LSD1 inhibitors are potential therapeutics for the malignancy.


Subject(s)
Enzyme Inhibitors/pharmacology , Gene Rearrangement , Histone Demethylases/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Leukemia/drug therapy , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Child , Drug Synergism , Enzyme Inhibitors/chemistry , Gene Expression Regulation, Leukemic/drug effects , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/metabolism , Humans , Leukemia/genetics , Leukemia/metabolism , Lysine/metabolism , MCF-7 Cells , Methylation/drug effects , Methyltransferases/antagonists & inhibitors , Methyltransferases/genetics , Methyltransferases/metabolism , Mice, Inbred NOD , Mice, SCID , Molecular Structure , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , U937 Cells , Xenograft Model Antitumor Assays
18.
Nat Commun ; 6: 6641, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25818643

ABSTRACT

Understanding the mechanism by which cell growth, migration, polyploidy, and tumorigenesis are regulated may provide important therapeutic strategies for cancer therapy. Here we identify the Skp2-macroH2A1 (mH2A1)-cyclin-dependent kinase 8 (CDK8) axis as a critical pathway for these processes, and deregulation of this pathway is associated with human breast cancer progression and patient survival outcome. We showed that mH2A1 is a new substrate of Skp2 SCF complex whose degradation by Skp2 promotes CDK8 gene and protein expression. Strikingly, breast tumour suppression on Skp2 deficiency can be rescued by mH2A1 knockdown or CDK8 restoration using mouse tumour models. We further show that CDK8 regulates p27 protein expression by facilitating Skp2-mediated p27 ubiquitination and degradation. Our study establishes a critical role of Skp2-mH2A1-CDK8 axis in breast cancer development and targeting this pathway offers a promising strategy for breast cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Carcinogenesis/genetics , Carcinoma/metabolism , Cyclin-Dependent Kinase 8/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Histones/metabolism , S-Phase Kinase-Associated Proteins/genetics , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fibroblasts , Humans , Mice , Mice, Knockout , S-Phase Kinase-Associated Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitination
20.
Sci Signal ; 6(257): ra3, 2013 Jan 08.
Article in English | MEDLINE | ID: mdl-23300340

ABSTRACT

K63-linked ubiquitination of Akt is a posttranslational modification that plays a critical role in growth factor-mediated membrane recruitment and activation of Akt. Although E3 ligases involved in growth factor-induced ubiquitination of Akt have been defined, the deubiquitinating enzyme (DUB) that triggers deubiquitination of Akt and the function of Akt deubiquitination remain largely unclear. We showed that CYLD was a DUB for Akt and suppressed growth factor-mediated ubiquitination and activation of Akt. CYLD directly removed ubiquitin moieties from Akt under serum-starved conditions. CYLD dissociated from Akt upon growth factor stimulation, thereby allowing E3 ligases to induce ubiquitination and activation of Akt. CYLD deficiency also promoted cancer cell proliferation, survival, glucose uptake, and, when injected into mice, growth of prostate tumors. Our findings reveal the crucial role of cycles of ubiquitination and deubiquitination of Akt in determining its plasma membrane localization and activation--and further identify CYLD as a molecular switch for these processes.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Ubiquitination/physiology , Animals , Cell Membrane/metabolism , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Deubiquitinating Enzyme CYLD , HEK293 Cells , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Knockout , Phosphorylation , Statistics, Nonparametric , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL