Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Cancer Immunol Res ; 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38631019

ABSTRACT

The intrinsic pharmacokinetic limitations of traditional peptide-based cancer vaccines hamper effective cross-presentation and codelivery of antigens and adjuvants, which are crucial for inducing robust antitumor CD8+ T-cell responses. Here, we report the development of a versatile strategy that simultaneously addresses the different pharmacokinetic challenges of soluble subunit vaccines composed of antigens and CpG to modulate vaccine efficacy via translating an engineered chimeric peptide, eTAT, as an intramolecular adjuvant. Linking antigens to eTAT enhanced cytosolic delivery of the antigens. This, in turn, led to improved activation and lymph node-trafficking of antigen-presenting cells and antigen cross-presentation, thus promoting antigen-specific T-cell immune responses. Simple mixing of eTAT-linked antigens and CpG significantly enhanced codelivery of antigens and CpG to the antigen-presenting cells, and this substantially augmented the adjuvant effect of CpG, maximized vaccine immunogenicity and elicited robust and durable CD8+ T-cell responses. Vaccination with this formulation altered the tumor microenvironment and exhibited potent antitumor effects, with generally further enhanced therapeutic efficacy when used in combination with anti-PD1. Altogether, the engineered chimeric peptide-based orchestrated codelivery of antigen and adjuvant may serve as a promising but simple strategy to improve the efficacy of peptide-based cancer vaccines.

2.
Head Neck ; 46(7): 1727-1736, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38429055

ABSTRACT

BACKGROUND: The long-term effect of intraoperative usage of carbon nanoparticles (CN) and parathyroid hormone (PTH) test strip using immune colloidal gold technique (ICGT) is unclear. This study aims to compare the effect of intraoperative usage of CN and ICGT test strips on PG function. METHODS: This randomized clinical study involved adult patients who underwent total thyroidectomy. They were randomly allocated into three groups (control, CN, and ICGT group). Clinical data were analyzed. RESULTS: Each group involved 98 patients. Serum calcium and PTH concentrations at 24 h postoperatively (PTH24h) were higher in CN group. The parathyroid function recovered quicker in CN group. Use of CN increased in situ PG preservation and PTH24h. Mediation analysis indicated that 23.05% of the total effect of CN on PTH24h was attributed to PGRIS. CONCLUSION: CN holds promise to improve in situ PG preservation and protect PG vasculature, thereby reducing the incidence of early hypoparathyroidism. The value of ICGT test strips for PG protection is dubious.


Subject(s)
Carbon , Gold Colloid , Hypoparathyroidism , Nanoparticles , Parathyroid Glands , Parathyroid Hormone , Thyroidectomy , Humans , Thyroidectomy/adverse effects , Male , Female , Middle Aged , Parathyroid Hormone/blood , Adult , Hypoparathyroidism/prevention & control , Hypoparathyroidism/etiology , Hypoparathyroidism/diagnosis , Aged
3.
Int J Surg ; 109(3): 429-437, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36912560

ABSTRACT

BACKGROUND: Routine prophylaxis for at-risk patients may reduce the occurrence of postoperative hypocalcemia but is not widely adopted due to a lack of evidence on the efficacy of available prophylactic strategies. In this study, we compared the relative efficacy of prophylactic strategies for postthyroidectomy hypocalcemia with a systematic review and network meta-analysis. METHODS: PubMed, Embase, and Cochrane Library were searched, covering the period from 1980 to May 2022, for randomized controlled trials (RCTs) comparing calcium, vitamin D 3 , activated vitamin D 3 , teriparatide, steroids, and magnesium with placebo or each other in patients receiving total or completion thyroidectomy. Involved RCTs reporting symptomatic or biochemical hypocalcemia. The primary outcome was symptomatic hypocalcemia, defined as circumoral tingling, and Chvostek and Trousseau signs. The secondary outcome was biochemical hypocalcemia. Risk of bias was assessed using the Cochrane risk of bias assessment tool for randomized trials. Pooled estimates were calculated using a random-effects inverse-variance weighting model. The network meta-analysis was performed under the frequentist framework. This meta-analysis was registered on the PROSPERO (International prospective register of systematic reviews) (CRD42022299982). RESULTS: Twenty-seven RCTs comprising 3382 patients are included. Prophylactic strategies of teriparatide, oral calcium plus vitamin D 3 , and oral calcium plus activated vitamin D 3 are superior to placebo in reducing symptomatic hypocalcemia. Teriparatide emerged as the most effective strategy for symptomatic hypocalcemia [relative risk (RR): 0.18; 95% CI: 0.03-0.98], followed by oral calcium plus activated vitamin D 3 (RR: 0.42; 95% CI: 0.25-0.73) and oral calcium plus vitamin D 3 (RR: 0.43; 95% CI: 0.26-0.71). Evidence on monotherapy with either oral calcium or vitamin D 3 in reducing symptomatic hypocalcemia is insufficient. Intravenous calcium and oral calcium are effective in reducing biochemical hypocalcemia. CONCLUSIONS: This network meta-analysis provides information on the relative efficacy of current prophylactic strategies for postthyroidectomy hypocalcemia. Teriparatide performed better than other interventions and would seem appropriate for deployment among high-risk populations.


Subject(s)
Hypocalcemia , Humans , Calcium , Cholecalciferol , Hypocalcemia/prevention & control , Network Meta-Analysis , Teriparatide
4.
Clin Cancer Res ; 27(24): 6772-6786, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34548317

ABSTRACT

PURPOSE: Human papillomavirus (HPV) plays a major role in oncogenesis and circular extrachromosomal DNA (ecDNA) is found in many cancers. However, the relationship between HPV and circular ecDNA in human cancer is not understood. EXPERIMENTAL DESIGN: Forty-four primary tumor tissue samples were obtained from a cohort of patients with HPV-positive oropharynx squamous cell carcinoma (OPSCC). Twenty-eight additional HPV oropharyngeal cancer (HPVOPC) tumors from The Cancer Genome Atlas (TCGA) project were analyzed as a separate validation cohort. Genomic, transcriptomic, proteomic, computational, and functional analyses of HPVOPC were applied to these datasets. RESULTS: Our analysis revealed circular, oncogenic DNA in nearly all HPVOPC, with circular human and human-viral hybrid ecDNA present in over a third of HPVOPC and viral circular DNA in remaining tumors. Hybrid ecDNA highly express fusion transcripts from HPV promoters and HPV oncogenes linked to downstream human transcripts that drive oncogenic transformation and immune evasion, and splice multiple, diverse human acceptors to a canonical SA880 viral donor site. HPVOPC have high E6*I expression with specific viral oncogene expression pattern related to viral or hybrid ecDNA composition. CONCLUSIONS: Nonchromosomal circular oncogenic DNA is a dominant feature of HPVOPC, revealing an unanticipated link between HPV and ecDNA that leverages the power of extrachromosomal inheritance to drive HPV and somatic oncogene expression.


Subject(s)
Alphapapillomavirus , Head and Neck Neoplasms , Oncogene Proteins, Viral , Oropharyngeal Neoplasms , Papillomavirus Infections , DNA, Circular , DNA, Viral/genetics , Head and Neck Neoplasms/genetics , Humans , Oncogene Proteins, Viral/genetics , Oncogenes/genetics , Oropharyngeal Neoplasms/genetics , Papillomaviridae/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Proteomics
5.
Nat Commun ; 12(1): 5131, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34446736

ABSTRACT

Protein delivery with cell-penetrating peptide is opening up the possibility of using targets inside cells for therapeutic or biological applications; however, cell-penetrating peptide-mediated protein delivery commonly suffers from ineffective endosomal escape and low tolerance in serum, thereby limiting in vivo efficacy. Here, we present an intracellular protein delivery system consisting of four modules in series: cell-penetrating peptide, pH-dependent membrane active peptide, endosome-specific protease sites and a leucine zipper. This system exhibits enhanced delivery efficiency and serum tolerance, depending on proteolytic cleavage-facilitated endosomal escape and leucine zipper-based dimerisation. Intravenous injection of protein phosphatase 1B fused with this system successfully suppresses the tumour necrosis factor-α-induced systemic inflammatory response and acetaminophen-induced acute liver failure in a mouse model. We believe that the strategy of using multifunctional chimaeric peptides is valuable for the development of cell-penetrating peptide-based protein delivery systems, and facilitate the development of biological macromolecular drugs for use against intracellular targets.


Subject(s)
Drug Delivery Systems/methods , Liver Failure, Acute/drug therapy , Peptides/chemistry , Protein Phosphatase 1/administration & dosage , Animals , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/metabolism , Endosomes/genetics , Endosomes/metabolism , Humans , Hydrogen-Ion Concentration , Liver Failure, Acute/genetics , Liver Failure, Acute/metabolism , Mice, Inbred BALB C , Peptides/genetics , Peptides/metabolism , Protein Phosphatase 1/chemistry , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Transport
6.
Int J Oncol ; 58(2): 226-237, 2021 02.
Article in English | MEDLINE | ID: mdl-33491747

ABSTRACT

Several comprehensive studies have demonstrated that the NOTCH pathway is altered in a bimodal manner in head and neck squamous cell carcinoma (HNSCC). In a previous study, it was found that the NOTCH4/HEY1 pathway was specifically upregulated in HNSCC and promoted epithelial­mesenchymal transition (EMT), and that HEY1 activation supported SOX2 expression. However, the interactions in this pathway have not yet been fully elucidated. The present study investigated the NOTCH4/HEY1/SOX2 axis in HNSCC using in vitro models and the Cancer Genome Atlas (TCGA) database. To explore the association, reporter and ChIP RT­qPCR assays using SOX2­overexpressing (SOX2­OE) cells were performed. The association between NOTCH4 and HEY1 was examined in the same manner using HEY1­overexpressing (HEY1­OE) cells. The results of the in vitro experiments indicated that HEY1 promoted EMT in the HNSCC cells. Furthermore, the overexpression of HEY1 also promoted sphere formation and increased murine xenograft tumorigenicity. Reporter assays and ChIP RT­qPCR experiments indicated that SOX2 regulated HEY1 expression via direct binding of the HEY1 promoter. HEY1 expression significantly correlated with SOX2 expression in primary lung SCC and other SCCs using the TCGA database. HEY1 also regulated NOTCH4 expression to create a positive reciprocal feedback loop. On the whole, the present study demonstrates that HEY1 expression in HNSCC is regulated via the promotion of SOX2 and promotes EMT. The NOTCH4/HEY1 pathway is specifically upregulated via a positive reciprocal feedback loop mediated by the HEY1­medaited regulation of NOTCH4 transcription, and SOX2 correlates with HEY1 expression in SCC from other primary sites.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Receptor, Notch4/genetics , SOXB1 Transcription Factors/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Datasets as Topic , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Humans , Mice , Receptor, Notch4/metabolism , Signal Transduction/genetics , Spheroids, Cellular , Squamous Cell Carcinoma of Head and Neck/pathology , Up-Regulation , Xenograft Model Antitumor Assays
7.
Int J Mol Sci ; 21(18)2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32899704

ABSTRACT

Precise gene editing is-or will soon be-in clinical use for several diseases, and more applications are under development. The programmable nuclease Cas9, directed by a single-guide RNA (sgRNA), can introduce double-strand breaks (DSBs) in target sites of genomic DNA, which constitutes the initial step of gene editing using this novel technology. In mammals, two pathways dominate the repair of the DSBs-nonhomologous end joining (NHEJ) and homology-directed repair (HDR)-and the outcome of gene editing mainly depends on the choice between these two repair pathways. Although HDR is attractive for its high fidelity, the choice of repair pathway is biased in a biological context. Mammalian cells preferentially employ NHEJ over HDR through several mechanisms: NHEJ is active throughout the cell cycle, whereas HDR is restricted to S/G2 phases; NHEJ is faster than HDR; and NHEJ suppresses the HDR process. This suggests that definitive control of outcome of the programmed DNA lesioning could be achieved through manipulating the choice of cellular repair pathway. In this review, we summarize the DSB repair pathways, the mechanisms involved in choice selection based on DNA resection, and make progress in the research investigating strategies that favor Cas9-mediated HDR based on the manipulation of repair pathway choice to increase the frequency of HDR in mammalian cells. The remaining problems in improving HDR efficiency are also discussed. This review should facilitate the development of CRISPR/Cas9 technology to achieve more precise gene editing.


Subject(s)
CRISPR-Cas Systems/genetics , DNA End-Joining Repair/genetics , Gene Editing/methods , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair/physiology , DNA Repair/genetics , DNA Repair/physiology , Endonucleases/metabolism , Gene Editing/trends , Humans , RNA, Guide, Kinetoplastida/genetics , Recombinational DNA Repair/genetics
8.
Oncogene ; 39(40): 6327-6339, 2020 10.
Article in English | MEDLINE | ID: mdl-32848210

ABSTRACT

The dominant paradigm for HPV carcinogenesis includes integration into the host genome followed by expression of E6 and E7 (E6/E7). We explored an alternative carcinogenic pathway characterized by episomal E2, E4, and E5 (E2/E4/E5) expression. Half of HPV positive cervical and pharyngeal cancers comprised a subtype with increase in expression of E2/E4/E5, as well as association with lack of integration into the host genome. Models of the E2/E4/E5 carcinogenesis show p53 dependent enhanced proliferation in vitro, as well as increased susceptibility to induction of cancer in vivo. Whole genomic expression analysis of the E2/E4/E5 pharyngeal cancer subtype is defined by activation of the fibroblast growth factor receptor (FGFR) pathway and this subtype is susceptible to combination FGFR and mTOR inhibition, with implications for targeted therapy.


Subject(s)
Carcinogenesis/genetics , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/genetics , Pharyngeal Neoplasms/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Uterine Cervical Neoplasms/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Proliferation/genetics , Datasets as Topic , Disease Models, Animal , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Viral/drug effects , Host-Pathogen Interactions/genetics , Human papillomavirus 16/genetics , Human papillomavirus 16/pathogenicity , Humans , Mice , Mice, Transgenic , Papillomavirus Infections/drug therapy , Papillomavirus Infections/mortality , Papillomavirus Infections/virology , Pharyngeal Neoplasms/drug therapy , Pharyngeal Neoplasms/mortality , Pharyngeal Neoplasms/virology , Primary Cell Culture , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/virology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/virology
9.
PLoS One ; 15(5): e0233380, 2020.
Article in English | MEDLINE | ID: mdl-32437477

ABSTRACT

Alternative mRNA splicing increases protein diversity, and alternative splicing events (ASEs) drive oncogenesis in multiple tumor types. However, the driving alterations that underlie the broad dysregulation of ASEs are incompletely defined. Using head and neck squamous cell carcinoma (HNSCC) as a model, we hypothesized that the genomic alteration of genes associated with the spliceosome may broadly induce ASEs across a broad range of target genes, driving an oncogenic phenotype. We identified 319 spliceosome genes and employed a discovery pipeline to identify 13 candidate spliceosome genes altered in HNSCC using The Cancer Genome Atlas (TCGA) HNSCC data. Phenotypic screens identified amplified and overexpressed CPSF1 as a target gene alteration that was validated in proliferation, colony formation, and apoptosis assays in cell line and xenograft systems as well as in primary HNSCC. We employed knockdown and overexpression assays followed by identification of ASEs regulated by CPSF1 overexpression to identify changes in ASEs, and the expression of these ASEs was validated using RNA from cell line models. Alterations in expression of spliceosome genes, including CPSF1, may contribute to HNSCC by mediating aberrant ASE expression.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Alternative Splicing , Biomarkers, Tumor , Cleavage And Polyadenylation Specificity Factor/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
10.
Otolaryngol Head Neck Surg ; 162(4): 512-519, 2020 04.
Article in English | MEDLINE | ID: mdl-32041482

ABSTRACT

OBJECTIVE: The incidence and survivorship of human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC) are increasing. Presence of HPV DNA and epigenetic alterations in salivary rinses are independently associated with clinical prognosis. We evaluated the utility of a combined panel in detecting disease recurrence during surveillance. We also assessed the assay's applicability in screening for HPV+ OPSCC. STUDY DESIGN: Retrospective cohort study. SETTING: Two tertiary academic hospitals. SUBJECTS AND METHODS: Forty-nine patients with posttreatment OPSCC were enrolled. Separately, 21 treatment-naive patients and 40 controls were included in the screening analysis. Salivary rinses were obtained from these cohorts and biomarker levels were quantified. Receiver operative characteristic (ROC) curves and multivariate logistic models were used to assess performance of biomarker combinations. RESULTS: Eight patients (16.3%) in the posttreatment cohort developed locoregional recurrence. Recurrence was associated with alcohol use (odds ratio [OR], 6.12; 95% confidence interval [CI], 0.26-3.79) and advanced nodal disease (OR, 2.21; 95% CI, 1.52-3.01). A panel of HPV DNA and methylated EDNRB improved detection of recurrent disease (area under the curve [AUC], 0.88) compared to single markers (AUC, 0.69-0.78). Positive biomarkers preceded clinical detection by 2.4 ± 1.6 months and was associated with nearly 40-fold risk of recurrence (OR, 36.4; 95% CI, 1.15-45.22). Within the screening analysis, single biomarkers demonstrated moderate sensitivity and specificity (AUC, 0.59-0.83) in the detection of primary disease. A panel combining HPV DNA markers with methylated EDNRB and methylated PAX5 improved AUC to 0.93. CONCLUSION: Detection of high-risk HPV DNA or aberrant hypermethylation in oral rinses is associated with presence and recurrence of OPSCC. Targeting both markers in saliva may have utility in long-term surveillance.


Subject(s)
DNA Methylation , DNA, Viral/analysis , DNA, Viral/genetics , Neoplasm Recurrence, Local/virology , Oropharyngeal Neoplasms/virology , Papillomaviridae/genetics , Promoter Regions, Genetic/genetics , Saliva/chemistry , Squamous Cell Carcinoma of Head and Neck/virology , Aged , Cohort Studies , Female , Humans , Male , Middle Aged , Retrospective Studies
11.
Clin Cancer Res ; 26(11): 2693-2703, 2020 06 01.
Article in English | MEDLINE | ID: mdl-31932491

ABSTRACT

PURPOSE: Human papillomavirus (HPV)-related head and neck squamous cell carcinoma (HNSCC) is associated with daily marijuana use and is also increasing in parallel with increased marijuana use in the United States. Our study is designed to define the interaction between cannabinoids and HPV-positive HNSCC. EXPERIMENTAL DESIGN: The expression of cannabinoid receptors CNR1 and CNR2 was analyzed using The Cancer Genome Atlas (TCGA) HNSCC data. We used agonists, antagonists, siRNAs, or shRNA-based models to explore the roles of CNR1 and CNR2 in HPV-positive HNSCC cell lines and animal models. Cannabinoid downstream pathways involved were determined by Western blotting and analyzed in a primary HPV HNSCC cohort with single-sample gene set enrichment analysis (ssGSEA) and the OncoGenome Positioning System (Onco-GPS). RESULTS: In TCGA cohort, the expression of CNR1 and CNR2 was elevated in HPV-positive HNSCC compared with HPV-negative HNSCC, and knockdown of CNR1/CNR2 expression inhibited proliferation in HPV-positive HNSCC cell lines. Specific CNR1 and CNR2 activation as well as nonselective cannabinoid receptor activation in cell lines and animal models promoted cell growth, migration, and inhibited apoptosis through p38 MAPK pathway activation. CNR1/CNR2 antagonists suppressed cell proliferation and migration and induced apoptosis. Using whole-genome expression analysis in a primary HPV HNSCC cohort, we identified specific p38 MAPK pathway activation signature in tumors from HPV HNSCC patients with objective measurement of concurrent cannabinoid exposure. CONCLUSIONS: Cannabinoids can promote progression of HPV-positive HNSCC through p38 MAPK pathway activation.


Subject(s)
Cannabinoids/pharmacology , Head and Neck Neoplasms/pathology , Papillomaviridae/isolation & purification , Papillomavirus Infections/complications , Receptors, Cannabinoid/chemistry , Squamous Cell Carcinoma of Head and Neck/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis , Cell Movement , Cell Proliferation , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/virology , Humans , Mice , Mice, Nude , Papillomavirus Infections/drug therapy , Papillomavirus Infections/virology , Prognosis , Receptors, Cannabinoid/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/virology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/genetics
12.
Head Neck ; 42(4): 688-697, 2020 04.
Article in English | MEDLINE | ID: mdl-31850594

ABSTRACT

BACKGROUND: We aimed to use genomic data for optimizing polymerase chain reaction (PCR) primer/probe sets for detection of human papillomavirus (HPV)-16 in body fluids of patients with HPV-related head and neck squamous cell carcinoma (HPV-HNSCC). METHODS: We used genomic HPV-HNSCC sequencing data from a single institutional and a TCGA cohort. Optimized primer/probe sets were designed and tested for analytical performance in CaSki HPV-16 genome and confirmed in salivary rinse samples from patients with HPV-HNSCC. RESULTS: The highest read density was observed between E5 and L2 regions. The E1 region contained a region that was universally present. Among candidate PCR primer/probe sets created, six reliably detected 30 HPV-16 copy number. In a CLIA certified laboratory setting, the combination of two novel primer/probe with E7 sets improved performance in salivary rinse samples with a sensitivity of 96% and specificity of 100%. CONCLUSIONS: PCR-based detection of HPV-16 DNA in HPV-HNSCC can be improved using rational genomic design.


Subject(s)
Head and Neck Neoplasms , Papillomavirus Infections , DNA, Viral/genetics , Genomics , Human papillomavirus 16/genetics , Humans , Papillomaviridae/genetics , Papillomavirus Infections/diagnosis , Squamous Cell Carcinoma of Head and Neck/genetics
13.
Cancer ; 126(4): 737-748, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31721164

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is one of the most frequently diagnosed cancers worldwide. LOXL2 demonstrates alternative splicing events in patients with human papillomavirus (HPV)-negative HNSCC. The current study explored the role of a dominant LOXL2 variant in HPV-negative HNSCC. METHODS: Expression of the LOXL2 variant was analyzed using The Cancer Genome Atlas cohorts and validated using quantitative reverse transcriptase-polymerase chain reaction in a separate primary tumor set. The authors defined the effect of LOXL2 splice variants in assays for cell proliferation using a cell viability assay and colony formation assay. Cell migration and invasion were examined using a cell scratch assay and transwell cell migration and invasion assay in LOXL2 splice variant gain and loss of expression cells. Western blot analysis and gene set enrichment analysis were used to explore the potential mechanism of the LOXL2 splice variant in HPV-negative HNSCC. RESULTS: Expression of a novel LOXL2 variant was found to be upregulated in The Cancer Genome Atlas HPV-negative HNSCC, and confirmed in the separate primary tumor validation set. Analyses of loss and gain of function demonstrated that this LOXL2 variant enhanced proliferation, migration, and invasion in HPV-negative HNSCC cells and activated the FAK/AKT pathway. A total of 837 upregulated and 820 downregulated genes and 526 upregulated and 124 downregulated pathways associated with LOXL2 variant expression were identified using gene set enrichment analysis, which helped in developing a better understanding of the networks activated by this LOXL2 variant in patients with HPV-negative HNSCC. CONCLUSIONS: The novel LOXL2 variant can promote the progression of HPV-negative HNSCC, in part through FAK/AKT pathway activation, which may provide a new potential therapeutic target among patients with HPV-negative HNSCC.


Subject(s)
Amino Acid Oxidoreductases/genetics , Carcinoma, Squamous Cell/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Alternative Splicing , Amino Acid Oxidoreductases/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Disease Progression , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Papillomaviridae/physiology , RNA Interference , Signal Transduction/genetics
14.
Cancer Sci ; 111(2): 356-368, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31778279

ABSTRACT

Metastasis is a critical determinant for the treatment strategy and prognosis in patients with squamous cell carcinoma of the head and neck (SCCHN). However, the mechanisms underlying SCCHN metastasis are poorly understood. Our study sought to determine the key microRNA and their functional mechanisms involved in SCCHN metastasis. For The Cancer Genome Atlas (TCGA) data analysis, quantitative PCR was used to quantify the level of miR-30e-5p in SCCHN and its clinical significance was further analyzed. A series of in vitro and in vivo experiments were applied to determine the effects of miR-30e-5p and its target AEG-1 on SCCHN metastasis. A mechanism investigation further revealed that AEG-1 was implicated in the angiogenesis and metastasis mediated by miR-30e-5p. Overall, our study confirms that miR-30e-5p is a valuable predictive biomarker and potential therapeutic target in SCCHN metastasis.


Subject(s)
Head and Neck Neoplasms/pathology , Lung Neoplasms/pathology , Membrane Proteins/genetics , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , RNA-Binding Proteins/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Humans , Lung Neoplasms/genetics , Male , Mice , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Prognosis , Squamous Cell Carcinoma of Head and Neck/genetics , Survival Analysis
15.
Nat Commun ; 10(1): 2188, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097695

ABSTRACT

Although promoter-associated CpG islands have been established as targets of DNA methylation changes in cancer, previous studies suggest that epigenetic dysregulation outside the promoter region may be more closely associated with transcriptional changes. Here we examine DNA methylation, chromatin marks, and transcriptional alterations to define the relationship between transcriptional modulation and spatial changes in chromatin structure. Using human papillomavirus-related oropharyngeal carcinoma as a model, we show aberrant enrichment of repressive H3K9me3 at the transcriptional start site (TSS) with methylation-associated, tumor-specific gene silencing. Further analysis identifies a hypermethylated subtype which shows a functional convergence on MYC targets and association with CREBBP/EP300 mutation. The tumor-specific shift to transcriptional repression associated with DNA methylation at TSSs was confirmed in multiple tumor types. Our data may show a common underlying epigenetic dysregulation in cancer associated with broad enrichment of repressive chromatin marks and aberrant DNA hypermethylation at TSSs in combination with MYC network activation.


Subject(s)
Chromatin/metabolism , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Transcription Initiation Site , CREB-Binding Protein/genetics , Cell Line, Tumor , Datasets as Topic , E1A-Associated p300 Protein/genetics , Gene Silencing , Histones/genetics , Histones/metabolism , Humans , Mutation , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/genetics
16.
J Cell Mol Med ; 23(7): 4711-4722, 2019 07.
Article in English | MEDLINE | ID: mdl-31111621

ABSTRACT

The canonical Wnt/ß-catenin signalling pathway and autophagy play critical roles in cancer progression. However, the role of Wnt-mediated autophagy in cancer radioresistance remains unclear. In this study, we found that irradiation activated the Wnt/ß-catenin and autophagic signalling pathways in squamous cell carcinoma of the head and neck (SCCHN). Wnt3a is a classical ligand that activated the Wnt/ß-catenin signalling pathway, induced autophagy and decreased the sensitivity of SCCHN to irradiation both in vitro and in vivo. Further mechanistic analysis revealed that Wnt3a promoted SCCHN radioresistance via protective autophagy. Finally, expression of the Wnt3a protein was elevated in both SCCHN tissues and patients' serum. Patients showing high expression of Wnt3a displayed a worse prognosis. Taken together, our study indicates that both the canonical Wnt and autophagic signalling pathways are valuable targets for sensitizing SCCHN to irradiation.


Subject(s)
Radiation Tolerance , Squamous Cell Carcinoma of Head and Neck/metabolism , Wnt3A Protein/metabolism , Animals , Autophagy/radiation effects , Beclin-1/metabolism , Cell Line, Tumor , Electrons , Female , Humans , Male , Mice, Nude , Middle Aged , Proportional Hazards Models , Radiation Tolerance/radiation effects , Survival Analysis , Wnt Signaling Pathway/radiation effects
17.
Nat Commun ; 10(1): 2415, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31142745

ABSTRACT

The original version of this Article contained an error in the author affiliations. Trey Ideker was incorrectly associated with 'Department of Medicine (Oncology), Stanford University School of Medicine, 875 Blake Wilbur Dr, Palo Alto, CA 94304, USA.' This has now been corrected in both the PDF and HTML versions of the Article.

18.
Cancer ; 125(14): 2423-2434, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30933315

ABSTRACT

BACKGROUND: Human papillomavirus (HPV)-associated oropharyngeal cancer is a disease clinically and biologically distinct from smoking-related head and neck squamous cell carcinoma (HNSCC). Despite its rapidly increasing incidence, the mutational landscape of HPV+ oropharyngeal squamous cell carcinoma (OPSCC) remains understudied. METHODS: This article presents the first mutational analysis of the 46 HPV+ OPSCC tumors within the newly expanded cohort of 530 HNSCC tumors from The Cancer Genome Atlas. A separate exome sequencing analysis was also performed for 46 HPV+ OPSCCs matched to their normal lymphocyte controls from the Johns Hopkins University cohort. RESULTS: There was a strikingly high 33% frequency of mutations within genes associated with chromatin regulation, including mutations in lysine methyltransferase 2C (KMT2C), lysine methyltransferase 2D (KMT2D), nuclear receptor binding SET domain protein 1 (NSD1), CREB binding protein (CREBBP), E1A-associated protein p300 (EP300), and CCCTC-binding factor (CTCF). In addition, the commonly altered genes phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) and fibroblast growth factor receptor 3 (FGFR3) showed distinct domain-specific hotspot mutations in comparison with their HPV- counterparts. PIK3CA showed a uniquely high rate of mutations within the helicase domain, and FGFR3 contained a predominance of hotspot S249C alterations that were not found in HPV- HNSCC. CONCLUSIONS: This analysis represents one of the largest studies to date of HPV+ OPSCC and lends novel insight into the genetic landscape of this biologically distinct disease, including a high rate of mutations in histone- and chromatin-modifying genes, which may offer novel therapeutic targets.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Human papillomavirus 16/immunology , Mutation , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/virology , Adult , Aged , Class I Phosphatidylinositol 3-Kinases/genetics , Cohort Studies , Female , Humans , Male , Middle Aged , Oropharyngeal Neoplasms/pathology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Receptor, Fibroblast Growth Factor, Type 3/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Exome Sequencing
19.
JCI Insight ; 4(1)2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30626753

ABSTRACT

Incidence of HPV+ oropharyngeal squamous cell carcinoma (OPSCC) has been increasing dramatically. Although long-term survival rates for these patients are high, they often suffer from permanent radiotherapy-related morbidity. This has prompted the development of de-escalation clinical protocols to reduce morbidity. However, a subset of patients do not respond even to standard therapy and have poor outcomes. It is unclear how to properly identify and treat the high- and low-risk HPV+ OPSCC patients. Since HPV positivity drives radiotherapy sensitivity, we hypothesized that variations in HPV biology may cause differences in treatment response and outcome. By analyzing gene expression data, we identified variations in HPV-related molecules among HPV+ OPSCC. A subset of tumors presented a molecular profile distinct from that of typical HPV+ tumors and exhibited poor treatment response, indicating molecular and clinical similarities with HPV- tumors. These molecular changes were also observed in vitro and correlated with radiation sensitivity. Finally, we developed a prognostic biomarker signature for identification of this subgroup of HPV+ OPSCC and validated it in independent cohorts of oropharyngeal and cervical carcinomas. These findings could translate to improved patient stratification for treatment deintensification and new therapeutic approaches for treatment-resistant HPV-related cancer.

20.
Int J Cancer ; 143(10): 2425-2436, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30070359

ABSTRACT

Human papillomavirus (HPV)-related oropharyngeal squamous cell carcinoma (OPSCC) exhibits a different composition of epigenetic alterations. In this study, we identified differentially methylated regions (DMRs) with potential utility in screening for HPV-positive OPSCC. Genome wide DNA methylation was measured using methyl-CpG binding domain protein-enriched genome sequencing (MBD-seq) in 50 HPV-positive OPSCC tissues and 25 normal tissues. Fifty-one DMRs were defined with maximal methylation specificity to cancer samples. The Cancer Genome Atlas (TCGA) methylation array data was used to evaluate the performance of the proposed candidates. Supervised hierarchical clustering of 51 DMRs found that HPV-positive OPSCC had significantly higher DNA methylation levels compared to normal samples, and non-HPV-related head and neck squamous cell carcinoma (HNSCC). The methylation levels of all top 20 DNA methylation biomarkers in HPV-positive OPSCC were significantly higher than those in normal samples. Further confirmation using quantitative methylation specific PCR (QMSP) in an independent set of 24 HPV-related OPSCCs and 22 controls showed that 16 of the 20 candidates had significant higher methylation levels in HPV-positive OPSCC samples compared with controls. One candidate, OR6S1, had a sensitivity of 100%, while 17 candidates (KCNA3, EMBP1, CCDC181, DPP4, ITGA4, BEND4, ELMO1, SFMBT2, C1QL3, MIR129-2, NID2, HOXB4, ZNF439, ZNF93, VSTM2B, ZNF137P and ZNF773) had specificities of 100%. The prediction accuracy of the 20 candidates rang from 56.2% to 99.8% by receiver operating characteristic analysis. We have defined 20 highly specific DMRs in HPV-related OPSCC, which can potentially be applied to molecular-based detection tests and improve disease management.


Subject(s)
DNA Methylation , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/virology , Biomarkers, Tumor/genetics , Case-Control Studies , Cohort Studies , Epigenesis, Genetic , Female , Humans , Male , Middle Aged , Oropharyngeal Neoplasms/pathology , Papillomaviridae/isolation & purification , Papillomavirus Infections/pathology , Squamous Cell Carcinoma of Head and Neck/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...