Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Theranostics ; 11(13): 6427-6444, 2021.
Article in English | MEDLINE | ID: mdl-33995666

ABSTRACT

Background: Reportedly, nasopharyngeal carcinoma (NPC) patients with MHC I Class aberration are prone to poor survival outcomes, which indicates that the deficiency of tumor neoantigens might represent a mechanism of immune surveillance escape in NPC. Methods: To clearly delineate the landscape of neoantigens in NPC, we performed DNA and RNA sequencing on paired primary tumor, regional lymph node metastasis and distant metastasis samples from 26 patients. Neoantigens were predicted using pVACseq pipeline. Subtype prediction model was built using random forest algorithm. Results: Portraying the landscape of neoantigens in NPC for the first time, we found that the neoantigen load of NPC was above average compared to that of other cancers in The Cancer Genome Atlas program. While the quantity and quality of neoantigens were similar among primary tumor, regional lymph node metastasis and distant metastasis samples, neoantigen depletion was more severe in metastatic sites than in primary tumors. Upon tracking the clonality change of neoantigens, we found that neoantigen reduction occurred during metastasis. Building a subtype prediction model based on reported data, we observed that subtype I lacked T cells and suffered from severe neoantigen depletion, subtype II highly expressed immune checkpoint molecules and suffered from the least neoantigen depletion, and subtype III was heterogenous. Conclusions: These results indicate that neoantigens are conducive to the guidance of clinical treatment, and personalized therapeutic vaccines for NPC deserve deeper basic and clinical investigations to make them feasible in the future.


Subject(s)
Antigens, Neoplasm/immunology , Nasopharyngeal Carcinoma/secondary , Nasopharyngeal Neoplasms/immunology , Adult , Antigens, Neoplasm/genetics , DNA, Neoplasm/genetics , DNA, Viral/analysis , DNA, Viral/genetics , Female , HLA Antigens/genetics , HLA Antigens/immunology , Herpesvirus 4, Human/genetics , Humans , INDEL Mutation , Immune Checkpoint Inhibitors/therapeutic use , Kaplan-Meier Estimate , Lymphatic Metastasis/immunology , Male , Middle Aged , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/therapy , Nasopharyngeal Neoplasms/virology , Polymorphism, Single Nucleotide , Progression-Free Survival , RNA, Neoplasm/genetics , Receptors, Antigen, T-Cell/immunology , Tumor Escape , Tumor Microenvironment/immunology , Tumor Virus Infections/virology
3.
Cancer Res ; 79(23): 5930-5943, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31484669

ABSTRACT

The genetic events occurring in recurrent nasopharyngeal carcinoma (rNPC) are poorly understood. Here, we performed whole-genome and whole-exome sequencing in 55 patients with rNPC and 44 primarily diagnosed NPC (pNPC), with 7 patients having paired rNPC and pNPC samples. Previously published pNPC exome data were integrated for analysis. rNPC and pNPC tissues had similar mutational burdens, however, the number of clonal mutations was increased in rNPC samples. TP53 and three NF-κB pathway components (TRAF3, CYLD, and NFKBIA) were significantly mutated in both pNPC and rNPC. Notably, mutations in TRAF3, CYLD, and NFKBIA were all clonal in rNPC, however, 55.6% to 57.9% of them were clonal in pNPC. In general, the number of clonal mutations in NF-κB pathway-associated genes was significantly higher in rNPC than in pNPC. The NF-κB mutational clonality was selected and/or enriched during NPC recurrence. The amount of NF-κB translocated to the nucleus in samples with clonal NF-κB mutants was significantly higher than that in samples with subclonal NF-κB mutants. Moreover, the nuclear abundance of NF-κB protein was significantly greater in pNPC samples with locoregional relapse than in those without relapse. Furthermore, high nuclear NF-κB levels were an independent negative prognostic marker for locoregional relapse-free survival in pNPC. Finally, inhibition of NF-κB enhanced both radiosensitivity and chemosensitivity in vitro and in vivo. In conclusion, NF-κB pathway activation by clonal mutations plays an important role in promoting the recurrence of NPC. Moreover, nuclear accumulation of NF-κB is a prominent biomarker for predicting locoregional relapse-free survival. SIGNIFICANCE: This study uncovers genetic events that promote the progression and recurrence of nasopharyngeal carcinoma and has potential prognostic and therapeutic implications.See related commentary by Sehgal and Barbie, p. 5915.


Subject(s)
Carcinoma , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Humans , Mutation , NF-kappa B/genetics , Neoplasm Recurrence, Local
4.
Theranostics ; 9(4): 1115-1124, 2019.
Article in English | MEDLINE | ID: mdl-30867819

ABSTRACT

Rationale: Epstein-Barr virus (EBV) is associated with multiple malignancies with expression of viral oncogenic proteins and chronic inflammation as major mechanisms contributing to tumor development. A less well-studied mechanism is the integration of EBV into the human genome possibly at sites which may disrupt gene expression or genome stability. Methods: We sequenced tumor DNA to profile the EBV sequences by hybridization-based enrichment. Bioinformatic analysis was used to detect the breakpoints of EBV integrations in the genome of cancer cells. Results: We identified 197 breakpoints in nasopharyngeal carcinomas and other EBV-associated malignancies. EBV integrations were enriched at vulnerable regions of the human genome and were close to tumor suppressor and inflammation-related genes. We found that EBV integrations into the introns could decrease the expression of the inflammation-related genes, TNFAIP3, PARK2, and CDK15, in NPC tumors. In the EBV genome, the breakpoints were frequently at oriP or terminal repeats. These breakpoints were surrounded by microhomology sequences, consistent with a mechanism for integration involving viral genome replication and microhomology-mediated recombination. Conclusion: Our finding provides insight into the potential of EBV integration as an additional mechanism mediating tumorigenesis in EBV associated malignancies.


Subject(s)
DNA, Viral/analysis , Epstein-Barr Virus Infections/complications , Genome, Human , Herpesvirus 4, Human/genetics , Neoplasms/virology , Virus Integration , DNA, Viral/genetics , Genetic Loci , Humans , Sequence Analysis, DNA
5.
Nat Commun ; 9(1): 5009, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30479336

ABSTRACT

Epstein-Barr virus (EBV)-associated epithelial cancers, including nasopharyngeal carcinoma (NPC) and approximately 10% of gastric cancers, termed EBVaGC, represent 80% of all EBV-related malignancies. However, the exact role of EBV in epithelial cancers remains elusive. Here, we report that EBV functions in vasculogenic mimicry (VM). Epithelial cancer cells infected with EBV develop tumor vascular networks that correlate with tumor growth, which is different from endothelial-derived angiogenic vessels and is VEGF-independent. Mechanistically, activation of the PI3K/AKT/mTOR/HIF-1α signaling cascade, which is partly mediated by LMP2A, is responsible for EBV-induced VM formation. Both xenografts and clinical samples of NPC and EBVaGC exhibit VM histologically, which are correlated with AKT and HIF-1α activation. Furthermore, although anti-VEGF monotherapy shows limited effects, potent synergistic antitumor activities are achieved by combination therapy with VEGF and HIF-1α-targeted agents. Our findings suggest that EBV creates plasticity in epithelial cells to express endothelial phenotype and provides a novel EBV-targeted antitumor strategy.


Subject(s)
Epithelial Cells/pathology , Epithelial Cells/virology , Herpesvirus 4, Human/physiology , Nasopharyngeal Neoplasms/blood supply , Nasopharyngeal Neoplasms/virology , Neovascularization, Pathologic/pathology , Animals , Axitinib/pharmacology , Axitinib/therapeutic use , Cell Line, Tumor , Female , Gene Expression Profiling , Genome, Viral , Herpesvirus 4, Human/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred BALB C , Mice, Nude , Mustard Compounds/pharmacology , Mustard Compounds/therapeutic use , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/genetics , Neovascularization, Pathologic/genetics , Phenylpropionates/pharmacology , Phenylpropionates/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Viral Matrix Proteins/metabolism
6.
Cell Death Dis ; 9(7): 761, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29988031

ABSTRACT

The management of advanced nasopharyngeal carcinoma (NPC) remains a challenge. The ubiquitous nature of Epstein-Barr virus (EBV) infection in nonkeratinizing NPC has forced us to investigate novel drugs for NPC in the presence of EBV. In this study, we performed a small-scale screening of a library of compounds that target epigenetic regulators in paired EBV-positive and EBV-negative NPC cell lines. We found that bromodomain and extra-terminal (BET) inhibitor JQ1 preferentially inhibits the growth of EBV-positive NPC cells. JQ1 induces apoptosis, decreases cell proliferation and enhances the radiosensitivity in NPC cells, especially EBV-positive cells. Significantly, JQ1-induced cell death is c-Myc-dependent. Notably, RNA-seq analysis demonstrated that JQ1 represses TP63, TP53 and their targets. JQ1 also lessens the expression of PD-L1 in NPC. Moreover, the high potency of JQ1 in NPC cells was further confirmed in vivo in CNE2-EBV+ tumor-bearing mice. These findings indicate that JQ1 is a promising therapeutic candidate for advanced NPC.


Subject(s)
Azepines/therapeutic use , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/metabolism , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/metabolism , Proteins/antagonists & inhibitors , Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Triazoles/therapeutic use , Animals , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude
7.
PLoS Pathog ; 14(7): e1007208, 2018 07.
Article in English | MEDLINE | ID: mdl-30052682

ABSTRACT

Epstein-Barr virus (EBV) is a human cancer-related virus closely associated with lymphoid and epithelial malignancies, and EBV glycoprotein B (gB) plays an essential role in viral entry into both B cells and epithelial cells by promoting cell-cell fusion. EBV gB is exclusively modified with high-mannose-linked N-glycans and primarily localizes to the endoplasmic reticulum (ER) with low levels on the plasma membrane (PM). However, the mechanism through which gB is regulated within host cells is largely unknown. Here, we report the identification of F-box only protein 2 (FBXO2), an SCF ubiquitin ligase substrate adaptor that preferentially binds high-mannose glycans and attenuates EBV infectivity by targeting N-glycosylated gB for degradation. gB possesses seven N-glycosylation sites, and FBXO2 directly binds to these high-mannose moieties through its sugar-binding domain. The interaction promotes the degradation of glycosylated gB via the ubiquitin-proteasome pathway. Depletion of FBXO2 not only stabilizes gB but also promotes its transport from the ER to the PM, resulting in enhanced membrane fusion and viral entry. FBXO2 is expressed in epithelial cells but not B cells, and EBV infection up-regulates FBXO2 levels. In summary, our findings highlight the significance of high-mannose modification of gB and reveal a novel host defense mechanism involving glycoprotein homeostasis regulation.


Subject(s)
Cell Cycle Proteins/metabolism , Epstein-Barr Virus Infections/metabolism , F-Box Proteins/metabolism , Host-Parasite Interactions/physiology , Nerve Tissue Proteins/metabolism , Viral Envelope Proteins/metabolism , Animals , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/pathogenicity , Humans
8.
Cancer Res ; 78(13): 3469-3483, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29735548

ABSTRACT

Genetic susceptibility underlies the pathogenesis of cancer. We and others have previously identified a novel susceptibility gene TNFRSF19, which encodes an orphan member of the TNF receptor superfamily known to be associated with nasopharyngeal carcinoma (NPC) and lung cancer risk. Here, we show that TNFRSF19 is highly expressed in NPC and is required for cell proliferation and NPC development. However, unlike most of the TNF receptors, TNFRSF19 was not involved in NFκB activation or associated with TRAF proteins. We identified TGFß receptor type I (TßRI) as a specific binding partner for TNFRSF19. TNFRSF19 bound the kinase domain of TßRI in the cytoplasm, thereby blocking Smad2/3 association with TßRI and subsequent signal transduction. Ectopic expression of TNFRSF19 in normal epithelial cells conferred resistance to the cell-cycle block induced by TGFß, whereas knockout of TNFRSF19 in NPC cells unleashed a potent TGFß response characterized by upregulation of Smad2/3 phosphorylation and TGFß target gene transcription. Furthermore, elevated TNFRSF19 expression correlated with reduced TGFß activity and poor prognosis in patients with NPC. Our data reveal that gain of function of TNFRSF19 in NPC represents a mechanism by which tumor cells evade the growth-inhibitory action of TGFß.Significance:TNFRSF19, a susceptibility gene for nasopharyngeal carcinoma and other cancers, functions as a potent inhibitor of the TGFß signaling pathway.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/13/3469/F1.large.jpg Cancer Res; 78(13); 3469-83. ©2018 AACR.


Subject(s)
Carcinogenesis/pathology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Adolescent , Adult , Aged , Biopsy , Cell Line, Tumor , Cohort Studies , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , Nasopharynx/pathology , Phosphorylation , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation , Young Adult
9.
Cancer Lett ; 381(1): 14-22, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27424523

ABSTRACT

Despite advances in the development of radiation against nasopharyngeal carcinoma (NPC), the management of advanced NPC remains a challenge. Smac mimetics are designed to neutralize inhibitor of apoptosis (IAP) proteins, thus reactivating the apoptotic program in cancer cells. In this study, we investigated the effect of a novel bivalent Smac mimetic APG-1387 in NPC. In vitro, APG-1387 in combination with TNF-α potently decreased NPC cell viability by inducing apoptosis in majority of NPC cell lines. The in vitro antitumor effect was RIPK1-dependent, whereas it was independent on IAPs, USP11, or EBV. Of note, the inhibition of NF-κB or AKT pathway rendered resistant NPC cells responsive to the treatment of APG-1387/TNF-α. In vivo, APG-1387 displayed antitumor activity as a single agent at well-tolerated doses, even in an in vitro resistant cell line. In summary, our results demonstrate that APG-1387 exerts a potent antitumor effect on NPC. These findings support clinical evaluation of APG-1387 as a potential treatment for advanced NPC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Azepines/pharmacology , Carcinoma/drug therapy , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Molecular Mimicry , Nasopharyngeal Neoplasms/drug therapy , Sulfonamides/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Inhibitory Concentration 50 , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Burden/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...