Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Cancer ; 126(20): 4521-4531, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32749686

ABSTRACT

BACKGROUND: The objective of the current study was to investigate the clinical activity of, safety of, and predictive biomarkers for afatinib, an irreversible pan-ErbB kinase inhibitor, in patients with recurrent and/or metastatic esophageal squamous cell carcinoma (R/M-ESCC). METHODS: Patients with R/M-ESCC that was refractory to platinum-based chemotherapy were enrolled in the current multicenter, single-arm, phase 2 study and received afatinib at a dose of 40 mg/day. The primary endpoint was the objective response rate. Secondary endpoints included progression-free survival, overall survival, the disease control rate, and the safety profile. To identify predictive biomarkers, single-nucleotide variations, short insertions/deletions, and somatic copy number alterations were assessed using whole-exome sequencing and their associations with clinical outcomes were analyzed. RESULTS: Among 49 enrolled patients, the objective response rate and disease control rate were 14.3% and 73.3%, respectively. With a median follow-up of 6.6 months, the median progression-free survival and overall survival were 3.4 months and 6.3 months, respectively. Treatment-related adverse events were noted to have occurred in 33 patients (67.3%), with the majority being of grade 1 to 2 (adverse events were graded and recorded based on the National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.03]). Whole-exome sequencing demonstrated that the ESCC genomes of patients who demonstrated a response to afatinib were enriched with genomic alterations of TP53 and epidermal growth factor receptor (EGFR). As a predictive marker, a score derived from TP53 disruptive mutations and EGFR amplifications and/or missense mutations demonstrated a significant association with the response to afatinib. The score based on the mutational status of EGFR and TP53 achieved a performance of an area under the curve of 0.86 in predicting the sensitivity of afatinib. CONCLUSIONS: The results of the current study demonstrated that afatinib can confer modest clinical benefits with manageable toxicity in patients with platinum-resistant R/M-ESCC. Identification of TP53 alterations and EGFR amplifications may serve as predictive markers with which to identify patients with R/M-ESCC who may benefit from afatinib. LAY SUMMARY: Esophageal squamous cell carcinoma (ESCC) is a type of cancer with a dismal prognosis and very limited treatment options. The clinical efficacy of afatinib was evaluated in patients with recurrent and/or metastatic ESCC, with adverse events demonstrating the modest efficacy with manageable toxicity of this irreversible, pan-ErbB kinase inhibitor. Whole-exome sequencing analysis of 41 cases of ESCC further revealed that the patients harboring epidermal growth factor receptor (EGFR) amplifications and disruptive TP53 mutations are more likely to benefit from treatment with afatinib. The results of the current study have highlighted the clinical value of EGFR and TP53 as predictive biomarkers of platinum-resistant recurrent and/or metastatic ESCC for afatinib sensitivity.


Subject(s)
Afatinib/therapeutic use , Esophageal Squamous Cell Carcinoma/drug therapy , Adult , Afatinib/pharmacology , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local
2.
Br J Cancer ; 123(12): 1720-1729, 2020 12.
Article in English | MEDLINE | ID: mdl-32963347

ABSTRACT

BACKGROUND: Recurrent and/or metastatic squamous cell carcinoma of head and neck (R/M SCCHN) is a common cancer with high recurrence and mortality. Current treatments have low response rates (RRs). METHODS: Fifty-three patients with R/M SCCHN received continuous oral buparlisib. In parallel, patient-derived xenografts (PDXs) were established in mice to evaluate resistance mechanisms and efficacy of buparlisib/cetuximab combination. Baseline and on-treatment tumour genomes and transcriptomes were sequenced. Based on the integrated clinical and PDX data, 11 patients with progression under buparlisib monotherapy were treated with a combination of buparlisib and cetuximab. RESULTS: For buparlisib monotherapy, disease control rate (DCR) was 49%, RR was 3% and median progression-free survival (PFS) and overall survival (OS) were 63 and 143 days, respectively. For combination therapy, DCR was 91%, RR was 18% and median PFS and OS were 111 and 206 days, respectively. Four PDX models were originated from patients enrolled in the current clinical trial. While buparlisib alone did not inhibit tumour growth, combination therapy achieved tumour inhibition in three of seven PDXs. Genes associated with apoptosis and cell-cycle arrest were expressed at higher levels with combination treatment than with buparlisib or cetuximab alone. CONCLUSIONS: The buparlisib/cetuximab combination has significant promise as a treatment strategy for R/M SCCHN. CLINICAL TRIAL REGISTRATION: NCT01527877.


Subject(s)
Aminopyridines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cetuximab/therapeutic use , Head and Neck Neoplasms/drug therapy , Morpholines/therapeutic use , Adult , Aged , Aged, 80 and over , Aminopyridines/adverse effects , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Cell Survival/drug effects , Cetuximab/adverse effects , DNA Copy Number Variations , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/mortality , Humans , Male , Mice , Mice, Nude , Mice, SCID , Middle Aged , Morpholines/adverse effects , Mutation , Neoplasm Transplantation , Progression-Free Survival , Reproducibility of Results , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/mortality , Treatment Outcome , Up-Regulation/genetics , Whole Genome Sequencing/methods
3.
Cancer Med ; 9(21): 8243-8257, 2020 11.
Article in English | MEDLINE | ID: mdl-33017516

ABSTRACT

Acquisition of recurrent/metastatic potential by a tumor cell defines a critical step in malignant progression. However, understanding of metastatic progression at the molecular level is scarce for cervical carcinomas (CES). In this study, we performed genomic, transcriptomic, and viral profiling of five pairs of primary (CES-P) and matched recurrent/metastatic tumors (CES-R/M) with high risk human papillomavirus. Whole exome sequencing revealed mutation features of CES-R/M including elevated mutation burdens and prevalent copy number alterations compared to their matched CES-P. A relative deficit of APOBEC-related mutation signatures accompanying the transcriptional downregulation of APOBEC3A was observed for CES-R/M. Mutations in genes encoding epigenetic regulators were commonly observed as CES-R/M-specific alterations. Immunoprofiling and gene set analysis revealed CES-Ps were enriched with transcripts representing activated anticancer immunity such as interferon-gamma pathway, while CES-R/M exhibited upregulation of genes involved in epithelial-mesenchymal transition and angiogenesis. Viral capture sequencing revealed that integration sites remained enriched in viral E1 protein domain during malignant progression. Moreover, we found transcriptional upregulation of POSTN and downregulation of APOBEC3A were associated with unfavorable clinical outcomes in CES. Comprehensive genomic and transcriptomic profiling of a rare cohort including CES-R/M identified metastases-specific features to advance the molecular understanding into CES metastatic progression with potential clinical implications.


Subject(s)
Alphapapillomavirus/genetics , Carcinoma/genetics , Neoplasm Recurrence, Local/genetics , Papillomavirus Infections/complications , Uterine Cervical Neoplasms/genetics , Virus Integration/genetics , Adult , Carcinoma/immunology , Carcinoma/secondary , Carcinoma/virology , Cell Adhesion Molecules/genetics , Cytidine Deaminase/genetics , DNA Copy Number Variations , DNA, Neoplasm/genetics , Disease-Free Survival , Down-Regulation , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Female , Genome , Genomics , Humans , INDEL Mutation , Middle Aged , Neoplasm Metastasis/genetics , Neoplasm Recurrence, Local/virology , Neovascularization, Pathologic/genetics , Proteins/genetics , Risk Factors , Sequence Analysis, RNA , Survival Rate , Transcriptome , Up-Regulation , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Viral Proteins/genetics , Exome Sequencing
4.
IEEE/ACM Trans Comput Biol Bioinform ; 16(5): 1656-1662, 2019.
Article in English | MEDLINE | ID: mdl-29993813

ABSTRACT

Mutation hotspots are either solitary amino acid residues or stretches of amino acids that show elevated mutation frequency in cancer-related genes, but their prevalence and biological relevance are not completely understood. Here, we developed a Smith-Waterman algorithm-based mutation hotspot discovery method, MutClustSW, to identify mutation hotspots of either single or clustered amino acid residues. We identified 181 missense mutation hotspots from COSMIC and TCGA mutation databases. In addition to 77 single amino acid residue hotspots (42.5 percent) including well-known mutation hotspots such as IDH1 (p.R132) and BRAF (p.V600), we identified 104 mutation hotspots (57.5 percent) as clusters or stretches of multiple amino acids, and the hotspots on MUC2, EPPK1, KMT2C, and TP53 were larger than 50 amino acids. Twelve of 27 nonsense mutation hotspots (44.4 percent) were observed in four cancer-related genes, TP53, ARID1A, CDKN2A, and PTEN, suggesting that truncating mutations on some tumor suppressor genes are not randomly distributed as previously assumed. We also show that hotspot mutations have higher mutation allele frequency than non-hotspots, and the hotspot information can be used to prioritize the cancer drivers. Together, the proposed algorithm and the mutation hotspot information can serve as valuable resources in the selection of functional driver mutations and associated genes.


Subject(s)
Computational Biology/methods , Mutation/genetics , Neoplasms/genetics , Algorithms , Amino Acid Sequence/genetics , Cluster Analysis , Databases, Genetic , Humans
5.
Cancer Immunol Res ; 6(1): 87-97, 2018 01.
Article in English | MEDLINE | ID: mdl-29141981

ABSTRACT

Surgical archives of tumor specimens are often impure. The presence of RNA transcripts from nontumor cells, such as immune and stromal cells, can impede analyses of cancer expression profiles. To systematically analyze the impact of tumor purity, the gene expression profiles and tumor purities were obtained for 7,794 tumor specimens across 21 tumor types (available in The Cancer Genome Atlas consortium). First, we observed that genes with roles in immunity and oxidative phosphorylation were significantly inversely correlated and correlated with the tumor purity, respectively. The expression of genes implicated in immunotherapy and specific immune cell genes, along with the abundance of immune cell infiltrates, was substantially inversely correlated with tumor purity. This relationship may explain the correlation between immune gene expression and mutation burden, highlighting the need to account for tumor purity in the evaluation of expression markers obtained from bulk tumor transcriptome data. Second, examination of cluster membership of gene pairs, with or without controlling for tumor purity, revealed that tumor purity may have a substantial impact on gene clustering across tumor types. Third, feature genes for molecular taxonomy were analyzed for correlation with tumor purity, and for some tumor types, feature genes representing the mesenchymal and classical subtypes were inversely correlated and correlated with tumor purity, respectively. Our findings indicate that tumor purity is an important confounder in evaluating the correlation between gene expression and clinicopathologic features such as mutation burden, as well as gene clustering and molecular taxonomy. Cancer Immunol Res; 6(1); 87-97. ©2017 AACR.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Immunity/genetics , Neoplasms/genetics , Neoplasms/immunology , Alleles , Cluster Analysis , Computational Biology/methods , Databases, Genetic , Gene Expression Profiling , Genetic Variation , Humans , Kaplan-Meier Estimate , Mutation , Neoplasms/mortality , Neoplasms/pathology , Organ Specificity/genetics , Prognosis , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL