1
|
Oketch DJA, Giulietti M, Piva F. A Comparison of Tools That Identify Tumor Cells by Inferring Copy Number Variations from Single-Cell Experiments in Pancreatic Ductal Adenocarcinoma. Biomedicines 2024; 12:1759. [PMID: 39200223 PMCID: PMC11351975 DOI: 10.3390/biomedicines12081759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) technique has enabled detailed analysis of gene expression at the single cell level, enhancing the understanding of subtle mechanisms that underly pathologies and drug resistance. To derive such biological meaning from sequencing data in oncology, some critical processing must be performed, including identification of the tumor cells by markers and algorithms that infer copy number variations (CNVs). We compared the performance of sciCNV, InferCNV, CopyKAT and SCEVAN tools that identify tumor cells by inferring CNVs from scRNA-seq data. Sequencing data from Pancreatic Ductal Adenocarcinoma (PDAC) patients, adjacent and healthy tissues were analyzed, and the predicted tumor cells were compared to those identified by well-assessed PDAC markers. Results from InferCNV, CopyKAT and SCEVAN overlapped by less than 30% with InferCNV showing the highest sensitivity (0.72) and SCEVAN the highest specificity (0.75). We show that the predictions are highly dependent on the sample and the software used, and that they return so many false positives hence are of little use in verifying or filtering predictions made via tumor biomarkers. We highlight how critical this processing can be, warn against the blind use of these software and point out the great need for more reliable algorithms.
Collapse
Affiliation(s)
| | - Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| |
Collapse
|
2
|
Si Y, Wang X, Su X, Weng Z, Hu Q, Li Q, Fan C, Zhang DY, Wang Y, Luo S, Song P. Extended Enrichment for Ultrasensitive Detection of Low-Frequency Mutations by Long Blocker Displacement Amplification. Angew Chem Int Ed Engl 2024; 63:e202400551. [PMID: 38416545 DOI: 10.1002/anie.202400551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 02/29/2024]
Abstract
Detecting low-frequency DNA mutations hotspots cluster is critical for cancer diagnosis but remains challenging. Quantitative PCR (qPCR) is constrained by sensitivity, and allele-specific PCR is restricted by throughput. Here we develop a long blocker displacement amplification (LBDA) coupled with qPCR for ultrasensitive and multiplexed variants detection. By designing long blocker oligos to perfectly match wildtype sequences while mispairing with mutants, long blockers enable 14-44 nt enrichment regions which is 2-fold longer than normal BDA in the experiments. For wild template with a specific nucleotide, LBDA can detect different mutation types down to 0.5 % variant allele frequency (VAF) in one reaction, with median enrichment fold of 1,000 on 21 mutant DNA templates compared to the wild type. We applied LBDA-qPCR to detect KRAS and NRAS mutation hotspots, utilizing a single plex assay capable of covering 81 mutations and tested in synthetic templates and colorectal cancer tissue samples. Moreover, the mutation types were verified through Sanger sequencing, demonstrating concordance with results obtained from next generation sequencing. Overall, LBDA-qPCR provides a simple yet ultrasensitive approach for multiplexed detection of low VAF mutations hotspots, presenting a powerful tool for cancer diagnosis and monitoring.
Collapse
Affiliation(s)
- Yunpei Si
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiawen Wang
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinglei Su
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- Institute of Molecular Medicine (IMM) Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhi Weng
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qiongzheng Hu
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250014, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | | | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Shihua Luo
- Department of Traumatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Ping Song
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
3
|
Ma M, Guo B, Lu H, Hong L. SCN4B inhibits the progression of lung adenocarcinoma and is associated with better prognosis. THE CLINICAL RESPIRATORY JOURNAL 2023; 17:1233-1245. [PMID: 37826914 PMCID: PMC10730470 DOI: 10.1111/crj.13709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023]
Abstract
INTRODUCTION Lung adenocarcinoma (LUAD) is the major type of non-small cell lung cancer with low a survival rate caused by metastasis. SCN4B encoding voltage-gated sodium channel β subunit is regarded as a metastasis-suppressor gene. We aim to explore how SCN4B influences the progression and prognosis of LUAD. METHODS The gene expression profiles of 585 LUAD samples in TCGA and GSE31210, GSE116959, and GSE72094 datasets from the GEO database were downloaded for analysis. Differentially expressed genes were obtained through the "limma" package. The "clusterProfiler" package was used to conduct GSEA. Survival analysis was conducted via "survival" and "survminer" packages. Transcription factors regulating SCN4B expression were screened by correlation analysis and further predicted by FIMO. Infiltration of immune cells was analyzed by CIBERSORT. ESTIMATE algorithm was used to evaluate the immune-related scores. RESULTS SCN4B expressed higher in normal samples than in LUAD samples and higher in female samples than male samples. One hundred and twenty-six pathways were significantly enriched between high and low SCN4B expression groups. Six transcription factors' expressions were positively related to SCN4B expression, and ChIP-seq data from "Cistrome" verified that TAL1 and ERG might bind to the upstream sequence of SCN4B. SCN4B expression was significantly correlated with activated memory CD4 T cells, resting mast cells, and monocytes. TMB status, three scores based on ESTIMATE algorithm, and expression of three immune checkpoints showed significant differences between SCN4B high- and low-expression groups. SCN4B could be considered as an independent prognostic signature of LUAD patients that higher expression represents a better prognosis. CONCLUSION SCN4B expresses higher in normal samples, and SCN4B is able to be an independent prognostic signature for LUAD patients. TAL1 and ERG may regulate the expression of SCN4B by binding its upstream sequences. Our research is valuable in improving the effectiveness of treatment in LUAD.
Collapse
Affiliation(s)
- Minting Ma
- Department of OncologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| | - Bin Guo
- Department of Thoracic SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| | - Hongwei Lu
- Department of OphthalmologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| | - Lei Hong
- Department of OncologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| |
Collapse
|
4
|
Konda P, Garinet S, Van Allen EM, Viswanathan SR. Genome-guided discovery of cancer therapeutic targets. Cell Rep 2023; 42:112978. [PMID: 37572322 DOI: 10.1016/j.celrep.2023.112978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/22/2023] [Accepted: 07/28/2023] [Indexed: 08/14/2023] Open
Abstract
The success of precision oncology-which aims to match the right therapies to the right patients based on molecular status-is predicated on a robust pipeline of molecular targets against which therapies can be developed. Recent advances in genomics and functional genetics have enabled the unbiased discovery of novel molecular targets at scale. We summarize the promise and challenges in integrating genomic and functional genetic landscapes of cancer to establish the next generation of cancer targets.
Collapse
Affiliation(s)
- Prathyusha Konda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Simon Garinet
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Srinivas R Viswanathan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Ornos ED, Cando LF, Catral CD, Quebral EP, Tantengco OA, Arevalo MVP, Dee EC. Molecular basis of sex differences in cancer: Perspective from Asia. iScience 2023; 26:107101. [PMID: 37404373 PMCID: PMC10316661 DOI: 10.1016/j.isci.2023.107101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2023] Open
Abstract
Cancer is a leading cause of mortality and morbidity globally. Sex differences in cancer are evident in death rates and treatment responses in several cancers. Asian patients have unique cancer epidemiology influenced by their genetic ancestry and sociocultural factors in the region. In this review, we show molecular associations that potentially mediate sex disparities observed in cancer in Asian populations. Differences in sex characteristics are evident at the cytogenetic, genetic, and epigenetic levels mediating processes that include cell cycle, oncogenesis, and metastasis. Larger clinical and in vitro studies that explore mechanisms can confirm the associations of these molecular markers. In-depth studies of these markers can reveal their importance as diagnostics, prognostics, and therapeutic efficacy markers. Sex differences should be considered in designing novel cancer therapeutics in this era of precision medicine.
Collapse
Affiliation(s)
- Eric David Ornos
- Department of Medical Microbiology, College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
- College of Medicine, University of the Philippines Manila, Manila, 1000, Philippines
| | - Leslie Faye Cando
- College of Medicine, University of the Philippines Manila, Manila, 1000, Philippines
| | | | - Elgin Paul Quebral
- College of Medicine, University of the Philippines Manila, Manila, 1000, Philippines
- Virology Laboratory, Department of Medical Microbiology, College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Ourlad Alzeus Tantengco
- College of Medicine, University of the Philippines Manila, Manila, 1000, Philippines
- Department of Physiology, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
- Department of Biology, College of Science, De La Salle University, Manila 0922, Philippines
| | | | - Edward Christopher Dee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10028, USA
| |
Collapse
|
6
|
Fernández-Serra A, López-Reig R, Márquez R, Gallego A, de Sande LM, Yubero A, Pérez-Segura C, Ramchandani-Vaswani A, Barretina-Ginesta MP, Mendizábal E, Esteban C, Gálvez F, Sánchez-Heras AB, Guerra-Alía EM, Gaba L, Quindós M, Palacio I, Alarcón J, Oaknin A, Aliaga J, Ramírez-Calvo M, García-Casado Z, Romero I, López-Guerrero JA. The Scarface Score: Deciphering Response to DNA Damage Agents in High-Grade Serous Ovarian Cancer-A GEICO Study. Cancers (Basel) 2023; 15:cancers15113030. [PMID: 37296992 DOI: 10.3390/cancers15113030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Genomic Instability (GI) is a transversal phenomenon shared by several tumor types that provide both prognostic and predictive information. In the context of high-grade serous ovarian cancer (HGSOC), response to DNA-damaging agents such as platinum-based and poly(ADP-ribose) polymerase inhibitors (PARPi) has been closely linked to deficiencies in the DNA repair machinery by homologous recombination repair (HRR) and GI. In this study, we have developed the Scarface score, an integrative algorithm based on genomic and transcriptomic data obtained from the NGS analysis of a prospective GEICO cohort of 190 formalin-fixed paraffin-embedded (FFPE) tumor samples from patients diagnosed with HGSOC with a median follow up of 31.03 months (5.87-159.27 months). In the first step, three single-source models, including the SNP-based model (accuracy = 0.8077), analyzing 8 SNPs distributed along the genome; the GI-based model (accuracy = 0.9038) interrogating 28 parameters of GI; and the HTG-based model (accuracy = 0.8077), evaluating the expression of 7 genes related with tumor biology; were proved to predict response. Then, an ensemble model called the Scarface score was found to predict response to DNA-damaging agents with an accuracy of 0.9615 and a kappa index of 0.9128 (p < 0.0001). The Scarface Score approaches the routine establishment of GI in the clinical setting, enabling its incorporation as a predictive and prognostic tool in the management of HGSOC.
Collapse
Affiliation(s)
- Antonio Fernández-Serra
- Molecular Biology Lab, Molecular Biology Department, Instituto Valenciano de Oncologia, 46009 Valencia, Spain
- Joint IVO-CIPF Cancer Research Unit, 46012 Valencia, Spain
| | - Raquel López-Reig
- Molecular Biology Lab, Molecular Biology Department, Instituto Valenciano de Oncologia, 46009 Valencia, Spain
- Joint IVO-CIPF Cancer Research Unit, 46012 Valencia, Spain
| | - Raúl Márquez
- Medical Oncology Department, MD Anderson Cancer Center, 28033 Madrid, Spain
| | - Alejandro Gallego
- Medical Oncology Department, Hospital Universitario La Paz, 28046 Madrid, Spain
| | | | - Alfonso Yubero
- Medical Oncology Department, Hospital Clínico Universitario Lozano Blesa, 50009 Zaragoza, Spain
| | - Cristina Pérez-Segura
- Medical Oncology Department, Hospital de Sant Pau i Santa Tecla, 08025 Barcelona, Spain
| | | | | | - Elsa Mendizábal
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Carmen Esteban
- Medical Oncology Department, Hospital Virgen de la Salud, 45005 Toledo, Spain
| | - Fernando Gálvez
- Medical Oncology Department, Complejo Hospitalario de Jaén, 23007 Jaén, Spain
| | | | - Eva María Guerra-Alía
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Lydia Gaba
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - María Quindós
- Medical Oncology Department, Complejo Hospitalario Universitario A Coruña, 15006 A Coruña, Spain
| | - Isabel Palacio
- Medical Oncology Department, Hospital Central Asturias, 33011 Oviedo, Spain
| | - Jesús Alarcón
- Medical Oncology Department, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Ana Oaknin
- Medical Oncology Department, Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Jessica Aliaga
- Pathology Department, Instituto Valenciano de Oncologia, 46009 Valencia, Spain
| | - Marta Ramírez-Calvo
- Molecular Biology Lab, Molecular Biology Department, Instituto Valenciano de Oncologia, 46009 Valencia, Spain
| | - Zaida García-Casado
- Molecular Biology Lab, Molecular Biology Department, Instituto Valenciano de Oncologia, 46009 Valencia, Spain
| | - Ignacio Romero
- Medical Oncology Department, Instituto Valenciano de Oncología, 46010 Valencia, Spain
| | - José Antonio López-Guerrero
- Molecular Biology Lab, Molecular Biology Department, Instituto Valenciano de Oncologia, 46009 Valencia, Spain
- Joint IVO-CIPF Cancer Research Unit, 46012 Valencia, Spain
- Department of Pathology, Catholic University of Valencia, 46001 Valencia, Spain
| |
Collapse
|
7
|
Kouidhi S, Zidi O, Belkhiria Z, Rais H, Ayadi A, Ben Ayed F, Mosbah A, Cherif A, El Gaaied ABA. Gut microbiota, an emergent target to shape the efficiency of cancer therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:240-265. [PMID: 37205307 PMCID: PMC10185446 DOI: 10.37349/etat.2023.00132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/04/2023] [Indexed: 05/21/2023] Open
Abstract
It is now well-acknowledged that microbiota has a profound influence on both human health and illness. The gut microbiota has recently come to light as a crucial element that influences cancer through a variety of mechanisms. The connections between the microbiome and cancer therapy are further highlighted by a number of preclinical and clinical evidence, suggesting that these complicated interactions may vary by cancer type, treatment, or even by tumor stage. The paradoxical relationship between gut microbiota and cancer therapies is that in some cancers, the gut microbiota may be necessary to maintain therapeutic efficacy, whereas, in other cancers, gut microbiota depletion significantly increases efficacy. Actually, mounting research has shown that the gut microbiota plays a crucial role in regulating the host immune response and boosting the efficacy of anticancer medications like chemotherapy and immunotherapy. Therefore, gut microbiota modulation, which aims to restore gut microbial balance, is a viable technique for cancer prevention and therapy given the expanding understanding of how the gut microbiome regulates treatment response and contributes to carcinogenesis. This review will provide an outline of the gut microbiota's role in health and disease, along with a summary of the most recent research on how it may influence the effectiveness of various anticancer medicines and affect the growth of cancer. This study will next cover the newly developed microbiota-targeting strategies including prebiotics, probiotics, and fecal microbiota transplantation (FMT) to enhance anticancer therapy effectiveness, given its significance.
Collapse
Affiliation(s)
- Soumaya Kouidhi
- Laboratory BVBGR-LR11ES31, Biotechnopole Sidi Thabet, University Manouba, ISBST, Ariana 2020, Tunisia
- Association Tunisienne de Lutte contre le Cancer (ATCC), Tunis, Tunisia
| | - Oumaima Zidi
- Laboratory BVBGR-LR11ES31, Biotechnopole Sidi Thabet, University Manouba, ISBST, Ariana 2020, Tunisia
- Department of Biologu, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis 1068, Tunisia
| | | | - Henda Rais
- Association Tunisienne de Lutte contre le Cancer (ATCC), Tunis, Tunisia
- Service d’Oncologie Médicale, Hôpital Salah-Azaïz, Tunis 1006, Tunisia
| | - Aida Ayadi
- Department of Pathology, Abderrahman Mami Hospital, University of Tunis El Manar, Ariana 2080, Tunisia
| | - Farhat Ben Ayed
- Association Tunisienne de Lutte contre le Cancer (ATCC), Tunis, Tunisia
| | - Amor Mosbah
- Laboratory BVBGR-LR11ES31, Biotechnopole Sidi Thabet, University Manouba, ISBST, Ariana 2020, Tunisia
| | - Ameur Cherif
- Laboratory BVBGR-LR11ES31, Biotechnopole Sidi Thabet, University Manouba, ISBST, Ariana 2020, Tunisia
| | - Amel Ben Ammar El Gaaied
- Laboratory of Genetics, Immunology and Human Pathology, Department of Biology, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis 1068, Tunisia
| |
Collapse
|
8
|
Landau J, Tsaban L, Yaacov A, Ben Cohen G, Rosenberg S. Shared Cancer Dataset Analysis Identifies and Predicts the Quantitative Effects of Pan-Cancer Somatic Driver Variants. Cancer Res 2023; 83:74-88. [PMID: 36264175 DOI: 10.1158/0008-5472.can-22-1038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/02/2022] [Accepted: 10/18/2022] [Indexed: 02/03/2023]
Abstract
Driver mutations endow tumors with selective advantages and produce an array of pathogenic effects. Determining the function of somatic variants is important for understanding cancer biology and identifying optimal therapies. Here, we compiled a shared dataset from several cancer genomic databases. Two measures were applied to 535 cancer genes based on observed and expected frequencies of driver variants as derived from cancer-specific rates of somatic mutagenesis. The first measure comprised a binary classifier based on a binomial test; the second was tumor variant amplitude (TVA), a continuous measure representing the selective advantage of individual variants. TVA outperformed all other computational tools in terms of its correlation with experimentally derived functional scores of cancer mutations. TVA also highly correlated with drug response, overall survival, and other clinical implications in relevant cancer genes. This study demonstrates how a selective advantage measure based on a large cancer dataset significantly impacts our understanding of the spectral effect of driver variants in cancer. The impact of this information will increase as cancer treatment becomes more precise and personalized to tumor-specific mutations. SIGNIFICANCE A new selective advantage estimation assists in oncogenic driver identification and relative effect measurements, enabling better prognostication, therapy selection, and prioritization.
Collapse
Affiliation(s)
- Jakob Landau
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Linoy Tsaban
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Adar Yaacov
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gil Ben Cohen
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shai Rosenberg
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
9
|
Qin H, Sheng W, Zhang G, Yang Q, Yao S, Yue Y, Zhang P, Zhu Y, Wang Q, Chen Y, Zeng H, Weng J, Yu F, Yang J. Comprehensive analysis of cuproptosis-related prognostic gene signature and tumor immune microenvironment in HCC. Front Genet 2023; 14:1094793. [PMID: 36891150 PMCID: PMC9986498 DOI: 10.3389/fgene.2023.1094793] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Background: Copper is an indispensable mineral element involved in many physiological metabolic processes. Cuproptosis is associated with a variety of cancer such as hepatocellular carcinoma (HCC). The objective of this study was to examine the relationships between the expression of cuproptosis-related genes (CRGs) and tumor characteristics, including prognosis and microenvironment of HCC. Methods: The differentially expressed genes (DEGs) between high and low CRGs expression groups in HCC samples were identified, and further were analyzed for functional enrichment analysis. Then, CRGs signature of HCC was constructed and analyzed utilizing LASSO and univariate and multivariate Cox regression analysis. Prognostic values of CRGs signature were evaluated by Kaplan-Meier analysis, independent prognostic analysis and nomograph. The expression of prognostic CRGs was verified by Real-time quantitative PCR (RT-qPCR) in HCC cell lines. In addition, the relationships between prognostic CRGs expression and the immune infiltration, tumor microenvironment, antitumor drugs response and m6A modifications were further explored using a series of algorithms in HCC. Finally, ceRNA regulatory network based on prognostic CRGs was constructed. Results: The DEGs between high and low CRG expression groups in HCC were mainly enriched in focal adhesion and extracellular matrix organization. Besides, we constructed a prognostic model that consists of CDKN2A, DLAT, DLST, GLS, and PDHA1 CRGs for predicting the survival likelihood of HCC patients. And the elevated expression of these five prognostic CRGs was substantially in HCC cell lines and associated with poor prognosis. Moreover, immune score and m6A gene expression were higher in the high CRG expression group of HCC patients. Furthermore, prognostic CRGs have higher mutation rates in HCC, and are significantly correlated with immune cell infiltration, tumor mutational burden, microsatellite instability, and anti-tumor drug sensitivity. Then, eight lncRNA-miRNA-mRNA regulatory axes that affected the progression of HCC were predicted. Conclusion: This study demonstrated that the CRGs signature could effectively evaluate prognosis, tumor immune microenvironment, immunotherapy response and predict lncRNA-miRNA-mRNA regulatory axes in HCC. These findings extend our knowledge of cuproptosis in HCC and may inform novel therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Haotian Qin
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Weibei Sheng
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | | | - Qi Yang
- Department of Medical Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Sen Yao
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yaohang Yue
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peng Zhang
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yuanchao Zhu
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qichang Wang
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yixiao Chen
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hui Zeng
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jian Weng
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Yu
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
10
|
Gao GF, Oh C, Saksena G, Deng D, Westlake LC, Hill BA, Reich M, Schumacher SE, Berger AC, Carter SL, Cherniack AD, Meyerson M, Tabak B, Beroukhim R, Getz G. Tangent normalization for somatic copy-number inference in cancer genome analysis. Bioinformatics 2022; 38:4677-4686. [PMID: 36040167 PMCID: PMC9563697 DOI: 10.1093/bioinformatics/btac586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/28/2022] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Somatic copy-number alterations (SCNAs) play an important role in cancer development. Systematic noise in sequencing and array data present a significant challenge to the inference of SCNAs for cancer genome analyses. As part of The Cancer Genome Atlas, the Broad Institute Genome Characterization Center developed the Tangent normalization method to generate copy-number profiles using data from single-nucleotide polymorphism (SNP) arrays and whole-exome sequencing (WES) technologies for over 10 000 pairs of tumors and matched normal samples. Here, we describe the Tangent method, which uses a unique linear combination of normal samples as a reference for each tumor sample, to subtract systematic errors that vary across samples. We also describe a modification of Tangent, called Pseudo-Tangent, which enables denoising through comparisons between tumor profiles when few normal samples are available. RESULTS Tangent normalization substantially increases signal-to-noise ratios (SNRs) compared to conventional normalization methods in both SNP array and WES analyses. Tangent and Pseudo-Tangent normalizations improve the SNR by reducing noise with minimal effect on signal and exceed the contribution of other steps in the analysis such as choice of segmentation algorithm. Tangent and Pseudo-Tangent are broadly applicable and enable more accurate inference of SCNAs from DNA sequencing and array data. AVAILABILITY AND IMPLEMENTATION Tangent is available at https://github.com/broadinstitute/tangent and as a Docker image (https://hub.docker.com/r/broadinstitute/tangent). Tangent is also the normalization method for the copy-number pipeline in Genome Analysis Toolkit 4 (GATK4). SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Galen F Gao
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Coyin Oh
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gordon Saksena
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Davy Deng
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | | | - Barbara A Hill
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Reich
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Division of Medical Genetics, University of California, San Diego, La, Jolla, CA, USA
| | - Steven E Schumacher
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ashton C Berger
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott L Carter
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Matthew Meyerson
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Barbara Tabak
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rameen Beroukhim
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Gad Getz
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
11
|
Identification of Copy Number Alterations from Next-Generation Sequencing Data. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1361:55-74. [DOI: 10.1007/978-3-030-91836-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
12
|
Raguraman P, Balachandran AA, Chen S, Diermeier SD, Veedu RN. Antisense Oligonucleotide-Mediated Splice Switching: Potential Therapeutic Approach for Cancer Mitigation. Cancers (Basel) 2021; 13:5555. [PMID: 34771719 PMCID: PMC8583451 DOI: 10.3390/cancers13215555] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
Splicing is an essential process wherein precursor messenger RNA (pre-mRNA) is reshaped into mature mRNA. In alternative splicing, exons of any pre-mRNA get rearranged to form mRNA variants and subsequently protein isoforms, which are distinct both by structure and function. On the other hand, aberrant splicing is the cause of many disorders, including cancer. In the past few decades, developments in the understanding of the underlying biological basis for cancer progression and therapeutic resistance have identified many oncogenes as well as carcinogenic splice variants of essential genes. These transcripts are involved in various cellular processes, such as apoptosis, cell signaling and proliferation. Strategies to inhibit these carcinogenic isoforms at the mRNA level are promising. Antisense oligonucleotides (AOs) have been developed to inhibit the production of alternatively spliced carcinogenic isoforms through splice modulation or mRNA degradation. AOs can also be used to induce splice switching, where the expression of an oncogenic protein can be inhibited by the induction of a premature stop codon. In general, AOs are modified chemically to increase their stability and binding affinity. One of the major concerns with AOs is efficient delivery. Strategies for the delivery of AOs are constantly being evolved to facilitate the entry of AOs into cells. In this review, the different chemical modifications employed and delivery strategies applied are discussed. In addition to that various AOs in clinical trials and their efficacy are discussed herein with a focus on six distinct studies that use AO-mediated exon skipping as a therapeutic strategy to combat cancer.
Collapse
Affiliation(s)
- Prithi Raguraman
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia; (P.R.); (A.A.B.); (S.C.)
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Akilandeswari Ashwini Balachandran
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia; (P.R.); (A.A.B.); (S.C.)
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Suxiang Chen
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia; (P.R.); (A.A.B.); (S.C.)
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Sarah D. Diermeier
- Department of Biochemistry, University of Otago, Dunedin 9016, New Zealand;
| | - Rakesh N. Veedu
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia; (P.R.); (A.A.B.); (S.C.)
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| |
Collapse
|
13
|
Wei Q, Ramsey SA. Predicting chemotherapy response using a variational autoencoder approach. BMC Bioinformatics 2021; 22:453. [PMID: 34551729 PMCID: PMC8456615 DOI: 10.1186/s12859-021-04339-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/17/2021] [Indexed: 01/14/2023] Open
Abstract
Background Multiple studies have shown the utility of transcriptome-wide RNA-seq profiles as features for machine learning-based prediction of response to chemotherapy in cancer. While tumor transcriptome profiles are publicly available for thousands of tumors for many cancer types, a relatively modest number of tumor profiles are clinically annotated for response to chemotherapy. The paucity of labeled examples and the high dimension of the feature data limit performance for predicting therapeutic response using fully-supervised classification methods. Recently, multiple studies have established the utility of a deep neural network approach, the variational autoencoder (VAE), for generating meaningful latent features from original data. Here, we report the first study of a semi-supervised approach using VAE-encoded tumor transcriptome features and regularized gradient boosted decision trees (XGBoost) to predict chemotherapy drug response for five cancer types: colon, pancreatic, bladder, breast, and sarcoma. Results We found: (1) VAE-encoding of the tumor transcriptome preserves the cancer type identity of the tumor, suggesting preservation of biologically relevant information; and (2) as a feature-set for supervised classification to predict response-to-chemotherapy, the unsupervised VAE encoding of the tumor’s gene expression profile leads to better area under the receiver operating characteristic curve and area under the precision-recall curve classification performance than the original gene expression profile or the PCA principal components or the ICA components of the gene expression profile, in four out of five cancer types that we tested. Conclusions Given high-dimensional “omics” data, the VAE is a powerful tool for obtaining a nonlinear low-dimensional embedding; it yields features that retain biological patterns that distinguish between different types of cancer and that enable more accurate tumor transcriptome-based prediction of response to chemotherapy than would be possible using the original data or their principal components. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04339-6.
Collapse
Affiliation(s)
- Qi Wei
- School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, OR, USA.
| | - Stephen A Ramsey
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
14
|
Li Y, Sun L, Guo X, Mo N, Zhang J, Li C. Frontiers in Bladder Cancer Genomic Research. Front Oncol 2021; 11:670729. [PMID: 34094968 PMCID: PMC8173177 DOI: 10.3389/fonc.2021.670729] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/28/2021] [Indexed: 02/05/2023] Open
Abstract
Most of the etiology studies of bladder cancer focus on genetic changes, mainly including mutation and activation of oncogenes, mutation and inactivation of tumor suppressor genes, and rearrangement or heterozygous deletion of chromosomes. Moreover, bladder cancer is highly heterogeneous mainly due to abnormal changes in the genome and proteome of tumor cells. Surgery is the main treatment for bladder cancer, but because the recurrence rate is high after surgery and most of the muscle-invasive bladder cancer acquires distant metastasis. Therefore, there is a need to combine with chemotherapy to consolidate the treatment effect. However, there are differences in chemosensitivity among patients. In this article, we review the up-to-date genomic researches on bladder cancer occurrence, development, metastasis, and chemosensitivity in patients, in order to provide some theoretical support for the diagnosis and treatment strategy for bladder cancer.
Collapse
Affiliation(s)
- Yi Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Lihui Sun
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Na Mo
- Department of Pathology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Jinku Zhang
- Department of Pathology, First Central Hospital of Baoding, Baoding, China.,Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, First Central Hospital of Baoding, Baoding, China
| | - Chong Li
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, First Central Hospital of Baoding, Baoding, China.,Department of Immunology, Beijing Jianlan Institute of Medicine, Beijing, China.,Department of Immunology, Beijing Zhongke Jianlan Biotechnology Co., Ltd., Beijing, China
| |
Collapse
|
15
|
Cosmetic, Biomedical and Pharmaceutical Applications of Fish Gelatin/Hydrolysates. Mar Drugs 2021; 19:md19030145. [PMID: 33800149 PMCID: PMC8000627 DOI: 10.3390/md19030145] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
There are several reviews that separately cover different aspects of fish gelatin including its preparation, characteristics, modifications, and applications. Its packaging application in food industry is extensively covered but other applications are not covered or covered alongside with those of collagen. This review is comprehensive, specific to fish gelatin/hydrolysate and cites recent research. It covers cosmetic applications, intrinsic activities, and biomedical applications in wound dressing and wound healing, gene therapy, tissue engineering, implants, and bone substitutes. It also covers its pharmaceutical applications including manufacturing of capsules, coating of microparticles/oils, coating of tablets, stabilization of emulsions and drug delivery (microspheres, nanospheres, scaffolds, microneedles, and hydrogels). The main outcomes are that fish gelatin is immunologically safe, protects from the possibility of transmission of bovine spongiform encephalopathy and foot and mouth diseases, has an economic and environmental benefits, and may be suitable for those that practice religious-based food restrictions, i.e., people of Muslim, Jewish and Hindu faiths. It has unique rheological properties, making it more suitable for certain applications than mammalian gelatins. It can be easily modified to enhance its mechanical properties. However, extensive research is still needed to characterize gelatin hydrolysates, elucidate the Structure Activity Relationship (SAR), and formulate them into dosage forms. Additionally, expansion into cosmetic applications and drug delivery is needed.
Collapse
|
16
|
Han XJ, Ma XL, Yang L, Wei YQ, Peng Y, Wei XW. Progress in Neoantigen Targeted Cancer Immunotherapies. Front Cell Dev Biol 2020; 8:728. [PMID: 32850843 PMCID: PMC7406675 DOI: 10.3389/fcell.2020.00728] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/14/2020] [Indexed: 02/05/2023] Open
Abstract
Immunotherapies that harness the immune system to kill cancer cells have showed significant therapeutic efficacy in many human malignancies. A growing number of studies have highlighted the relevance of neoantigens in recognizing cancer cells by intrinsic T cells. Cancer neoantigens are a direct consequence of somatic mutations presenting on the surface of individual cancer cells. Neoantigens are fully cancer-specific and exempt from central tolerance. In addition, neoantigens are important targets for checkpoint blockade therapy. Recently, technological innovations have made neoantigen discovery possible in a variety of malignancies, thus providing an impetus to develop novel immunotherapies that selectively enhance T cell reactivity for the destruction of cancer cells while leaving normal tissues unharmed. In this review, we aim to introduce the methods of the identification of neoantigens, the mutational patterns of human cancers, related clinical trials, neoantigen burden and sensitivity to immune checkpoint blockade. Moreover, we focus on relevant challenges of targeting neoantigens for cancer treatment.
Collapse
|
17
|
Lv LC, Huang QY, Ding W, Xiao XH, Zhang HY, Xiong LX. Fish gelatin: The novel potential applications. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103581] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
18
|
Kumar P, Mistri TK. Transcription factors in SOX family: Potent regulators for cancer initiation and development in the human body. Semin Cancer Biol 2019; 67:105-113. [PMID: 31288067 DOI: 10.1016/j.semcancer.2019.06.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022]
Abstract
Transcription factors (TFs) have a key role in controlling the gene regulatory network that sustains explicit cell states in humans. However, an uncontrolled regulation of these genes potentially results in a wide range of diseases, including cancer. Genes of the SOX family are indeed crucial as deregulation of SOX family TFs can potentially lead to changes in cell fate as well as irregular cell growth. SOX TFs are a conserved group of transcriptional regulators that mediate DNA binding through a highly conserved high-mobility group (HMG) domain. Accumulating evidence demonstrates that cell fate and differentiation in major developmental processes are controlled by SOX TFs. Besides; numerous reports indicate that both up- and down-regulation of SOX TFs may induce cancer progression. In this review, we discuss the involvement of key TFs of SOX family in human cancers.
Collapse
Affiliation(s)
- Prasann Kumar
- The Division of Research and Development, Lovely Professional University, Jalandhar, Punjab, 144411, India; The Department of Agronomy, Lovely Professional University, Jalandhar, Punjab, 144411, India
| | - Tapan Kumar Mistri
- The Division of Research and Development, Lovely Professional University, Jalandhar, Punjab, 144411, India; The Department of Chemistry, Lovely Professional University, Jalandhar, Punjab, 144411, India.
| |
Collapse
|
19
|
Chen J, Jian X, Deng S, Ma Z, Shou X, Shen Y, Zhang Q, Song Z, Li Z, Peng H, Peng C, Chen M, Luo C, Zhao D, Ye Z, Shen M, Zhang Y, Zhou J, Fahira A, Wang Y, Li S, Zhang Z, Ye H, Li Y, Shen J, Chen H, Tang F, Yao Z, Shi Z, Chen C, Xie L, Wang Y, Fu C, Mao Y, Zhou L, Gao D, Yan H, Zhao Y, Huang C, Shi Y. Identification of recurrent USP48 and BRAF mutations in Cushing's disease. Nat Commun 2018; 9:3171. [PMID: 30093687 PMCID: PMC6085354 DOI: 10.1038/s41467-018-05275-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 06/13/2018] [Indexed: 12/15/2022] Open
Abstract
Cushing's disease results from corticotroph adenomas of the pituitary that hypersecrete adrenocorticotropin (ACTH), leading to excess glucocorticoid and hypercortisolism. Mutations of the deubiquitinase gene USP8 occur in 35-62% of corticotroph adenomas. However, the major driver mutations in USP8 wild-type tumors remain elusive. Here, we report recurrent mutations in the deubiquitinase gene USP48 (predominantly encoding p.M415I or p.M415V; 21/91 subjects) and BRAF (encoding p.V600E; 15/91 subjects) in corticotroph adenomas with wild-type USP8. Similar to USP8 mutants, both USP48 and BRAF mutants enhance the promoter activity and transcription of the gene encoding proopiomelanocortin (POMC), which is the precursor of ACTH, providing a potential mechanism for ACTH overproduction in corticotroph adenomas. Moreover, primary corticotroph tumor cells harboring BRAF V600E are sensitive to the BRAF inhibitor vemurafenib. Our study thus contributes to the understanding of the molecular mechanism of the pathogenesis of corticotroph adenoma and informs therapeutic targets for this disease.
Collapse
Affiliation(s)
- Jianhua Chen
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xuemin Jian
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Siyu Deng
- Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zengyi Ma
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Xuefei Shou
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Yue Shen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Qilin Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Zhijian Song
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Zhiqiang Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Hong Peng
- Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Cheng Peng
- Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Min Chen
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cheng Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Dan Zhao
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhao Ye
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Ming Shen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Yichao Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Juan Zhou
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Aamir Fahira
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yongfei Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Shiqi Li
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Zhaoyun Zhang
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Hongying Ye
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Yiming Li
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Jiawei Shen
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Hong Chen
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Feng Tang
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Zhenwei Yao
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Department of Radiology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Chunjui Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Lu Xie
- Shanghai Center for Bioinformation Technology (SCBIT), Shanghai Academy of Science and Technology, Shanghai, 201203, China
| | - Ye Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Chaowei Fu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
- State Key Laboratory of Medical Neurobiology, Institute of Neurosurgery, Shanghai Medical College, Fudan University, 200040, Shanghai, China
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China
| | - Daming Gao
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hai Yan
- Department of Pathology, Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
- Shanghai Pituitary Tumor Center, Shanghai, 200040, China.
- Institute of Neurosurgery, Fudan University, Shanghai, 200040, China.
- State Key Laboratory of Medical Neurobiology, Institute of Neurosurgery, Shanghai Medical College, Fudan University, 200040, Shanghai, China.
| | - Chuanxin Huang
- Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yongyong Shi
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China.
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
- Department of Psychiatry, First Teaching Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, Shandong, 266003, China.
| |
Collapse
|
20
|
Ding QS, Zhang L, Wang BC, Zeng Z, Zou XQ, Cao PB, Zhou GM, Tang M, Wu L, Wu LL, Yu HG, Guo Y, Zhou FX. Aberrant high expression level of MORC2 is a common character in multiple cancers. Hum Pathol 2018; 76:58-67. [PMID: 29555576 DOI: 10.1016/j.humpath.2018.03.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 02/28/2018] [Accepted: 03/09/2018] [Indexed: 12/26/2022]
Abstract
Microrchidia 2 (MORC2) plays important roles in DNA damage repair and lipogenesis, but the clinical and functional role of MORC2 in cancer remains largely unexplored. In this study, we showed that MORC2 was widely expressed in human tissues while significantly up-regulated in most cancer types using immunohistochemical staining and analysis of messenger RNA expression profile of more than 2000 human tissue samples from 15 different organs (lung, prostate, liver, breast, brain, stomach, colon/rectum, pancreas, ovary, endometrium, skin, nasopharynx, kidney, esophagus, and bladder). We also found that the MORC2 expression level in high-grade cancer tissues was much more elevated and associated with unfavorable pathological characteristics, poor overall survival, and disease-free survival in several kinds of cancers such as non-small cell lung cancer and breast cancer. Gene set enrichment analysis was used to predict the genes modulated by MORC2, and the results showed that dysregulation of MORC2 in tumor may take part in the cell cycle regulation and genomic instability. We observed that MORC2 knockdown would arrest the cell cycle progress, and the genome of tumors with high MORC2 expression contained more point mutations and gene copy number variation, which validates our gene set enrichment analysis results. The results also showed that MORC2 knockdown would significantly inhibit the proliferation, colony forming, migration, and invasion in multiple cancer cell lines. Taken together, these results highlight the importance of MORC2 in tumorigenesis and cancer progression, and it may act as a potential diagnostic marker and therapeutic target for these diseases.
Collapse
Affiliation(s)
- Qian-Shan Ding
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Clinical Cancer Study Center, Zhongnan Hospital, Wuhan University, Wuhan 430071, China; Department of Gastroenterology, Institute for Gastroenterology and Hepatology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Li Zhang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Bi-Cheng Wang
- Department of Pathology, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xian-Qiong Zou
- College of Biotechnology, Guilin Medical University, Guilin 541100, China
| | - Peng-Bo Cao
- The State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Guang-Ming Zhou
- The State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Meng Tang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Clinical Cancer Study Center, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Lu Wu
- Department of Gastroenterology, Institute for Gastroenterology and Hepatology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lian-Lian Wu
- Department of Gastroenterology, Institute for Gastroenterology and Hepatology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hong-Gang Yu
- Department of Gastroenterology, Institute for Gastroenterology and Hepatology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yong Guo
- College of Biotechnology, Guilin Medical University, Guilin 541100, China.
| | - Fu-Xiang Zhou
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Clinical Cancer Study Center, Zhongnan Hospital, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
21
|
Park S, Lee M, Cho KJ, Kim SB, Roh JL, Choi SH, Nam SY, Kim SY, Song JS. Association Between Fibroblast Growth Factor Receptor 1 Gene Amplification and Human Papillomavirus Prevalence in Tonsillar Squamous Cell Carcinoma With Clinicopathologic Analysis. J Histochem Cytochem 2018; 66:511-522. [PMID: 29553868 DOI: 10.1369/0022155418761652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Amplification of fibroblast growth factor receptor 1 ( FGFR1) has been reported in many squamous cell carcinomas, and human papillomavirus (HPV)-related oropharyngeal squamous cell carcinoma has been characterized as a distinct subset with favorable prognosis. Here, we investigated the FGFR1 amplification and HPV status in tonsillar squamous cell carcinoma (TSCC) and analyzed the clinical characteristics. HPV in situ hybridization (HPV ISH) and FGFR1 fluorescence in situ hybridization (FISH) were performed using tissue microarray from 89 cases of TSCC. Fourteen of 89 (15.7%) TSCC cases had FGFR1 amplification, and HPV was detected in 59 of 89 (66.3%) cases. FGFR1 amplification status was not associated with HPV positivity ( p=0.765). Outcomes were not significantly different between FGFR1 amplified and non-amplified patients. Although FGFR1 amplified patients ( n=4) in the HPV ISH-negative group ( n=30) had a tendency for poorer overall survival, no statistical significance was identified ( p=0.150, log-rank). FGFR1 protein overexpression showed better disease-free survival ( p=0.031, log-rank) in HPV-negative TSCC. This study suggests FGFR1 amplification may be important in the pathogenesis of TSCC regardless of HPV status.
Collapse
Affiliation(s)
- Soonchan Park
- Department of Radiology, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Miji Lee
- Department of Pathology, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Kyung-Ja Cho
- Departments of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Sung Bae Kim
- Medical Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jong-Lyel Roh
- Head and Neck Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Seung-Ho Choi
- Head and Neck Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Soon Yuhl Nam
- Head and Neck Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Sang Yoon Kim
- Head and Neck Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Joon Seon Song
- Departments of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
22
|
Kim JS, Kim MW, Jeong HY, Kang SJ, Park SI, Lee YK, Kim HS, Kim KS, Park YS. Sendai viroplexes for epidermal growth factor receptor-directed delivery of interleukin-12 and salmosin genes to cancer cells. J Gene Med 2018; 18:112-23. [PMID: 27191929 DOI: 10.1002/jgm.2884] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/13/2016] [Accepted: 05/13/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The effective delivery of therapeutic genes to target cells has been a fundamental goal in cancer gene therapy because of its advantages with respect to both safety and transfection efficiency. In the present, study we describe a tumor-directed gene delivery system that demonstrates remarkable efficacy in gene delivery and minimizes the off-target effects of gene transfection. METHODS The system consists of a well-verified cationic O,O'-dimyristyl-N-lysyl glutamate (DMKE), Sendai virus fusion (F) protein and hemagglutinin-neuraminidase (HN) protein, referred to as cationic Sendai F/HN virosomes. To achieve tumor-specific recognition, anti-epidermal growth factor (EGF) receptor antibody was coupled to the surface of the virosomes containing interleukin-12 (IL-12) and/or salmosin genes that have potent anti-angiogenetic functions. RESULTS Among the virosomal formulations, the anti-EGF receptor (EGFR) viroplexes, prepared via complexation of plasmid DNA (pDNA) with cationic DMKE lipid, exhibited more efficient gene transfection to tumor cells over-expressing EGF receptors compared to the neutrally-charged anti-EGFR virosomes encapsulating pDNA. In addition, the anti-EGFR viroplexes with IL-12 and salmosin genes exhibited the most effective therapeutic efficacy in a mouse tumor model. Especially when combined with doxorubicin, transfection of the two genes via the anti-EGFR viroplexes exhibited an enhanced inhibitory effect on tumor growth and metastasis in lungs. CONCLUSIONS The results of the present study suggest that anti-EGFR viroplexes can be utilized as an effective strategy for tumor-directed gene delivery. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jung Seok Kim
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Min Woo Kim
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Hwa Yeon Jeong
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Seong Jae Kang
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Sang Il Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Yeon Kyung Lee
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Hong Sung Kim
- Department of Biomedical Laboratory Science, Korea Nazarene University, Cheonan, Republic of Korea
| | - Keun Sik Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon, Republic of Korea
| | - Yong Serk Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| |
Collapse
|
23
|
Giordano TJ. Genomic Hallmarks of Thyroid Neoplasia. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 13:141-162. [PMID: 29083981 DOI: 10.1146/annurev-pathol-121808-102139] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The genomic landscape of thyroid cancers that are derived from follicular cells has been substantially elucidated through the coordinated application of high-throughput genomic technologies. Here, I review the common genetic alterations across the spectrum of thyroid neoplasia and present the resulting model of thyroid cancer initiation and progression. This model illustrates the striking correlation between tumor differentiation and overall somatic mutational burden, which also likely explains the highly variable clinical behavior and outcome of patients with thyroid cancers. These advances are yielding critical insights into thyroid cancer pathogenesis, which are being leveraged for the development of new diagnostic tools, prognostic and predictive biomarkers, and novel therapeutic approaches.
Collapse
Affiliation(s)
- Thomas J Giordano
- Departments of Pathology and Internal Medicine, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA;
| |
Collapse
|
24
|
Kuriyama K, Enomoto Y, Suzuki R, Watanuki J, Hosoi H, Yamashita Y, Murata S, Mushino T, Tamura S, Hanaoka N, Dyer M, Siebert R, Kiyonari H, Nakakuma H, Kitamura T, Sonoki T. Enforced expression of MIR142, a target of chromosome translocation in human B-cell tumors, results in B-cell depletion. Int J Hematol 2017; 107:345-354. [PMID: 29071477 DOI: 10.1007/s12185-017-2360-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 10/16/2017] [Accepted: 10/18/2017] [Indexed: 12/22/2022]
Abstract
MicroRNA142 (MIR142) is a target of chromosome translocations and mutations in human B-cell lymphomas. We analyzed an aggressive B-cell lymphoma carrying t(8;17)(q24;q22) and t(6;14)(p21;q32), and sought to explore the role(s) of MIR142 in lymphomagenesis. t(8;17)(q24;q22) involved MYC on 8q24 and pri-MIR142 on 17q22. MYC was activated by a promoter substitution by t(8;17)(q24;q22). t(8;17)(q24;q22) was an additional event after t(6;14) (p21;q32), which caused the over-expression of CCND3. Southern blot analyses revealed that the MIR142 locus was deleted from the affected allele, whereas Northern analyses showed over-expression of MIR142 in tumor cells. Although previous studies reported an over-expression of mutations in MIR142 in B-cell lymphomas, limited information is available on the functions of MIR142 in lymphomagenesis. Therefore, we generated bone marrow transplantation (BMT) and transgenic (Eμ/mir142) mice, which showed enforced expression in hematopoietic progenitor cells and B cells, respectively. BMT mice showed decreased numbers of all lineage-positive cells, particularly B cells, in peripheral blood. Eμ/mir142 mice showed decreased numbers of IgM-positive splenocytes, and exhibited altered B-cell phenotypic changes induced by lipopolysaccharide. Our results suggest that over-expression of MIR142 alters B-cell differentiation, implying multi-step lymphomagenesis together with MYC activation and CCND3 over-expression.
Collapse
Affiliation(s)
- Kodai Kuriyama
- Hematology/Oncology, Wakayama Medical University, 811-1 Kimi-idera, Wakayama, 641-8510, Japan
| | - Yutaka Enomoto
- Division of Cellular Therapy and Division of Stem Cell Signaling, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Ritsuro Suzuki
- Hematology and Oncology, Shimane Medical University, Shimane, Japan
| | - Jyuri Watanuki
- Hematology/Oncology, Wakayama Medical University, 811-1 Kimi-idera, Wakayama, 641-8510, Japan
| | - Hiroki Hosoi
- Hematology/Oncology, Wakayama Medical University, 811-1 Kimi-idera, Wakayama, 641-8510, Japan
| | - Yusuke Yamashita
- Hematology/Oncology, Wakayama Medical University, 811-1 Kimi-idera, Wakayama, 641-8510, Japan
| | - Shogo Murata
- Hematology/Oncology, Wakayama Medical University, 811-1 Kimi-idera, Wakayama, 641-8510, Japan
| | - Toshiki Mushino
- Hematology/Oncology, Wakayama Medical University, 811-1 Kimi-idera, Wakayama, 641-8510, Japan
| | - Shinobu Tamura
- Hematology/Oncology, Wakayama Medical University, 811-1 Kimi-idera, Wakayama, 641-8510, Japan
| | - Nobuyoshi Hanaoka
- Hematology/Oncology, Wakayama Medical University, 811-1 Kimi-idera, Wakayama, 641-8510, Japan
| | - Martin Dyer
- Department of Cancer Studies and Molecular Medicine, Leicester Medical School, University of Leicester, Leicester, UK
| | - Reiner Siebert
- Institute of Human Genetics, Christian Albrechts University Kiel, Kiel, Germany.,Institute of Human Genetics, University of Ulm and University of Ulm Medical Center, Ulm, Germany
| | - Hiroshi Kiyonari
- Animal Resource Development Unit, RIKEN Center for Life Science Technologies, Kobe, Japan.,Genetic Engineering Team, RIKEN Center for Life Science Technologies, Kobe, Japan
| | - Hideki Nakakuma
- Hematology/Oncology, Wakayama Medical University, 811-1 Kimi-idera, Wakayama, 641-8510, Japan
| | - Toshio Kitamura
- Division of Cellular Therapy and Division of Stem Cell Signaling, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takashi Sonoki
- Hematology/Oncology, Wakayama Medical University, 811-1 Kimi-idera, Wakayama, 641-8510, Japan.
| |
Collapse
|
25
|
Zare F, Dow M, Monteleone N, Hosny A, Nabavi S. An evaluation of copy number variation detection tools for cancer using whole exome sequencing data. BMC Bioinformatics 2017; 18:286. [PMID: 28569140 PMCID: PMC5452530 DOI: 10.1186/s12859-017-1705-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 05/22/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Recently copy number variation (CNV) has gained considerable interest as a type of genomic/genetic variation that plays an important role in disease susceptibility. Advances in sequencing technology have created an opportunity for detecting CNVs more accurately. Recently whole exome sequencing (WES) has become primary strategy for sequencing patient samples and study their genomics aberrations. However, compared to whole genome sequencing, WES introduces more biases and noise that make CNV detection very challenging. Additionally, tumors' complexity makes the detection of cancer specific CNVs even more difficult. Although many CNV detection tools have been developed since introducing NGS data, there are few tools for somatic CNV detection for WES data in cancer. RESULTS In this study, we evaluated the performance of the most recent and commonly used CNV detection tools for WES data in cancer to address their limitations and provide guidelines for developing new ones. We focused on the tools that have been designed or have the ability to detect cancer somatic aberrations. We compared the performance of the tools in terms of sensitivity and false discovery rate (FDR) using real data and simulated data. Comparative analysis of the results of the tools showed that there is a low consensus among the tools in calling CNVs. Using real data, tools show moderate sensitivity (~50% - ~80%), fair specificity (~70% - ~94%) and poor FDRs (~27% - ~60%). Also, using simulated data we observed that increasing the coverage more than 10× in exonic regions does not improve the detection power of the tools significantly. CONCLUSIONS The limited performance of the current CNV detection tools for WES data in cancer indicates the need for developing more efficient and precise CNV detection methods. Due to the complexity of tumors and high level of noise and biases in WES data, employing advanced novel segmentation, normalization and de-noising techniques that are designed specifically for cancer data is necessary. Also, CNV detection development suffers from the lack of a gold standard for performance evaluation. Finally, developing tools with user-friendly user interfaces and visualization features can enhance CNV studies for a broader range of users.
Collapse
Affiliation(s)
- Fatima Zare
- Computer Science and Engineering Department, University of Connecticut, Storrs, CT, USA
| | - Michelle Dow
- Biomedical Informatics Department, University of California San Diego, San Diego, CA, USA
| | - Nicholas Monteleone
- Computer Science and Engineering Department, University of Connecticut, Storrs, CT, USA
| | - Abdelrahman Hosny
- Computer Science and Engineering Department, University of Connecticut, Storrs, CT, USA
| | - Sheida Nabavi
- Computer Science and Engineering Department and Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
26
|
Ye M, He Y, Lin H, Yang S, Zhou Y, Zhou L, Zhong J, Lu G, Zheng J, Xue ZX, Cai ZZ. High expression of atonal homolog 8 predicts a poor clinical outcome in patients with colorectal cancer and contributes to tumor progression. Oncol Rep 2017; 37:2955-2963. [PMID: 28393252 DOI: 10.3892/or.2017.5554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/02/2016] [Indexed: 12/16/2022] Open
Abstract
Hitherto, it has been identified that numerous basic-helix-loop-helix (bHLH) transcription factors play vital roles in tumor initiation and progression. Atonal homolog 8 (ATOH8) is a member of the bHLH family of transcription factors, which participates in embryogenesis and the development of various tissues. Several studies have demonstrated that ATOH8 is involved in the progression of malignancies; however, the effects of ATOH8 in colorectal cancer (CRC) remain unknown. The aim of the present study was to explore the expression and function of ATOH8 in CRC. The present study included 106 paired CRCs and peritumoral samples. The expression of ATOH8 was evaluated by immunohistochemistry, and the results were compared with the clinical outcomes of the patients. Furthermore, cell proliferation, cell cycle distribution, wound healing and cytotoxicity assays were performed in colon cancer cell line SW620. Immunohistochemical analyses revealed that the expression of ATOH8 in CRC tissues was significantly increased compared with the peritumoral tissues, and that the high expression of ATOH8 was associated with a high serum carcinoembryonic antigen (CEA) level and a worse overall survival. In vitro assays revealed that ATOH8 knockdown in colon cancer cells inhibited cell proliferation, induced cell cycle arrest at the S phase, and increased the percentage of apoptotic cells and sensitivity to 5-fluorouracil (5-FU). The present study suggests that ATOH8 promotes the progression of CRC and may potentially serve as a novel prognostic predictor and potential therapeutic target in CRC.
Collapse
Affiliation(s)
- Mengsi Ye
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| | - Yun He
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| | - Hao Lin
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| | - Shouxing Yang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuhui Zhou
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| | - Lingling Zhou
- Department of Pathology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| | - Jinwei Zhong
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| | - Guangrong Lu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| | - Jihang Zheng
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhan-Xiong Xue
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhen-Zhai Cai
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Lucheng, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
27
|
Zeng Y, Zhou Z, Fan M, Gong T, Zhang Z, Sun X. PEGylated Cationic Vectors Containing a Protease-Sensitive Peptide as a miRNA Delivery System for Treating Breast Cancer. Mol Pharm 2016; 14:81-92. [PMID: 28043137 DOI: 10.1021/acs.molpharmaceut.6b00726] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Several targeted drug delivery systems have recently been developed to increase the bioavailability of a drug at its site of action, allowing simultaneous reduction of the total necessary drug dose as well as side effects. Here, we designed a cationic gene vector containing matrix metalloproteinase-2 (MMP2)-cleavable substrate peptides that specifically target tumor sites where MMP2 levels are high. The targeted delivery system is fabricated by linking enzyme-cleavable polyethylene glycol (PEG) derivatives to cationic β-cyclodextrin-polyethylenimine conjugates, which reduce the toxicity of polyethylenimine and condense the therapeutic cargo. In the present study, tumor suppressor microRNA miR-34a, which suppresses onset and progression of many types of cancers, was investigated for its therapeutic potential for treating breast cancer. The PEG coating markedly reduces nonspecific interaction between cationic particles and serum proteins, permitting accumulation at the target site; subsequent peptide cleavage by MMP2 facilitates miR-34a delivery into tumor cells. The nanopreparation shows excellent stability, and its internalization, tumor targeting, and antitumor efficacy in vitro and in vivo are better than those of a nanopreparation containing MMP2-uncleavable peptide.
Collapse
Affiliation(s)
- Ye Zeng
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| | - Zixuan Zhou
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| | - Minmin Fan
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
28
|
Kaveh F, Baumbusch LO, Nebdal D, Børresen-Dale AL, Lingjærde OC, Edvardsen H, Kristensen VN, Solvang HK. A systematic comparison of copy number alterations in four types of female cancer. BMC Cancer 2016; 16:913. [PMID: 27876019 PMCID: PMC5120489 DOI: 10.1186/s12885-016-2899-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/30/2016] [Indexed: 01/06/2023] Open
Abstract
Background Detection and localization of genomic alterations and breakpoints are crucial in cancer research. The purpose of this study was to investigate, in a methodological and biological perspective, different female, hormone-dependent cancers to identify common and diverse DNA aberrations, genes, and pathways. Methods In this work, we analyzed tissue samples from patients with breast (n = 112), ovarian (n = 74), endometrial (n = 84), or cervical (n = 76) cancer. To identify genomic aberrations, the Circular Binary Segmentation (CBS) and Piecewise Constant Fitting (PCF) algorithms were used and segmentation thresholds optimized. The Genomic Identification of Significant Targets in Cancer (GISTIC) algorithm was applied to the segmented data to identify significantly altered regions and the associated genes were analyzed by Ingenuity Pathway Analysis (IPA) to detect over-represented pathways and functions within the identified gene sets. Results and Discussion Analyses of high-resolution copy number alterations in four different female cancer types are presented. For appropriately adjusted segmentation parameters the two segmentation algorithms CBS and PCF performed similarly. We identified one region at 8q24.3 with focal aberrations that was altered at significant frequency across all four cancer types. Considering both, broad regions and focal peaks, three additional regions with gains at significant frequency were revealed at 1p21.1, 8p22, and 13q21.33, respectively. Several of these events involve known cancer-related genes, like PPP2R2A, PSCA, PTP4A3, and PTK2. In the female reproductive system (ovarian, endometrial, and cervix [OEC]), we discovered three common events: copy number gains at 5p15.33 and 15q11.2, further a copy number loss at 8p21.2. Interestingly, as many as 75% of the aberrations (75% amplifications and 86% deletions) identified by GISTIC were specific for just one cancer type and represented distinct molecular pathways. Conclusions Our results disclose that some prominent copy number changes are shared in the four examined female, hormone-dependent cancer whereas others are definitive to specific cancer types. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2899-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fatemeh Kaveh
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Medical Genetics Department, Oslo University Hospital Ullevål, Oslo, Norway.,Department of Pediatric Research, Division of Pediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars O Baumbusch
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Department of Pediatric Research, Division of Pediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Daniel Nebdal
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Ole Christian Lingjærde
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Department of Computer Science, University of Oslo, Oslo, Norway
| | - Hege Edvardsen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Vessela N Kristensen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway. .,Department of Clinical Molecular Biology (EpiGen), Medical Division, Akershus University Hospital, Lørenskog, Norway.
| | - Hiroko K Solvang
- Marine Mammals Research Group, Institute of Marine Research, Bergen, Norway
| |
Collapse
|
29
|
Falasca M, Hamilton JR, Selvadurai M, Sundaram K, Adamska A, Thompson PE. Class II Phosphoinositide 3-Kinases as Novel Drug Targets. J Med Chem 2016; 60:47-65. [DOI: 10.1021/acs.jmedchem.6b00963] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Marco Falasca
- Metabolic
Signalling Group, School of Biomedical Sciences, CHIRI Biosciences, Curtin University, Perth, Western Australia 6845, Australia
| | - Justin R. Hamilton
- Australian
Centre for Blood Diseases and Department of Clinical Haematology, Monash University, 99 Commercial Road, Melbourne, Victoria 3004, Australia
| | - Maria Selvadurai
- Australian
Centre for Blood Diseases and Department of Clinical Haematology, Monash University, 99 Commercial Road, Melbourne, Victoria 3004, Australia
| | - Krithika Sundaram
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Aleksandra Adamska
- Metabolic
Signalling Group, School of Biomedical Sciences, CHIRI Biosciences, Curtin University, Perth, Western Australia 6845, Australia
| | - Philip E. Thompson
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
30
|
Narang P, Dhapola P, Chowdhury S. BreCAN-DB: a repository cum browser of personalized DNA breakpoint profiles of cancer genomes. Nucleic Acids Res 2016; 44:D952-8. [PMID: 26586806 PMCID: PMC4702892 DOI: 10.1093/nar/gkv1264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 10/29/2015] [Accepted: 11/03/2015] [Indexed: 11/20/2022] Open
Abstract
BreCAN-DB (http://brecandb.igib.res.in) is a repository cum browser of whole genome somatic DNA breakpoint profiles of cancer genomes, mapped at single nucleotide resolution using deep sequencing data. These breakpoints are associated with deletions, insertions, inversions, tandem duplications, translocations and a combination of these structural genomic alterations. The current release of BreCAN-DB features breakpoint profiles from 99 cancer-normal pairs, comprising five cancer types. We identified DNA breakpoints across genomes using high-coverage next-generation sequencing data obtained from TCGA and dbGaP. Further, in these cancer genomes, we methodically identified breakpoint hotspots which were significantly enriched with somatic structural alterations. To visualize the breakpoint profiles, a next-generation genome browser was integrated with BreCAN-DB. Moreover, we also included previously reported breakpoint profiles from 138 cancer-normal pairs, spanning 10 cancer types into the browser. Additionally, BreCAN-DB allows one to identify breakpoint hotspots in user uploaded data set. We have also included a functionality to query overlap of any breakpoint profile with regions of user's interest. Users can download breakpoint profiles from the database or may submit their data to be integrated in BreCAN-DB. We believe that BreCAN-DB will be useful resource for genomics scientific community and is a step towards personalized cancer genomics.
Collapse
Affiliation(s)
- Pankaj Narang
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Parashar Dhapola
- GNR Center for Genome Informatics, Institute of Genomics and Integrative Biology, CSIR, Delhi, India
| | - Shantanu Chowdhury
- GNR Center for Genome Informatics, Institute of Genomics and Integrative Biology, CSIR, Delhi, India Proteomics and Structural Biology Unit, Institute of Genomics and Integrative Biology, CSIR, Delhi, India
| |
Collapse
|
31
|
Studies of Tumor Suppressor Genes via Chromosome Engineering. Cancers (Basel) 2015; 8:cancers8010004. [PMID: 26729168 PMCID: PMC4728451 DOI: 10.3390/cancers8010004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/19/2015] [Accepted: 12/21/2015] [Indexed: 12/01/2022] Open
Abstract
The development and progression of malignant tumors likely result from consecutive accumulation of genetic alterations, including dysfunctional tumor suppressor genes. However, the signaling mechanisms that underlie the development of tumors have not yet been completely elucidated. Discovery of novel tumor-related genes plays a crucial role in our understanding of the development and progression of malignant tumors. Chromosome engineering technology based on microcell-mediated chromosome transfer (MMCT) is an effective approach for identification of tumor suppressor genes. The studies have revealed at least five tumor suppression effects. The discovery of novel tumor suppressor genes provide greater understanding of the complex signaling pathways that underlie the development and progression of malignant tumors. These advances are being exploited to develop targeted drugs and new biological therapies for cancer.
Collapse
|
32
|
Lund HL, Hughesman CB, McNeil K, Clemens S, Hocken K, Pettersson R, Karsan A, Foster LJ, Haynes C. Initial diagnosis of chronic myelogenous leukemia based on quantification of M-BCR status using droplet digital PCR. Anal Bioanal Chem 2015; 408:1079-94. [DOI: 10.1007/s00216-015-9204-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 11/11/2015] [Accepted: 11/18/2015] [Indexed: 01/25/2023]
|
33
|
Seeger-Nukpezah T, Geynisman DM, Nikonova AS, Benzing T, Golemis EA. The hallmarks of cancer: relevance to the pathogenesis of polycystic kidney disease. Nat Rev Nephrol 2015; 11:515-34. [PMID: 25870008 PMCID: PMC5902186 DOI: 10.1038/nrneph.2015.46] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a progressive inherited disorder in which renal tissue is gradually replaced with fluid-filled cysts, giving rise to chronic kidney disease (CKD) and progressive loss of renal function. ADPKD is also associated with liver ductal cysts, hypertension, chronic pain and extra-renal problems such as cerebral aneurysms. Intriguingly, improved understanding of the signalling and pathological derangements characteristic of ADPKD has revealed marked similarities to those of solid tumours, even though the gross presentation of tumours and the greater morbidity and mortality associated with tumour invasion and metastasis would initially suggest entirely different disease processes. The commonalities between ADPKD and cancer are provocative, particularly in the context of recent preclinical and clinical studies of ADPKD that have shown promise with drugs that were originally developed for cancer. The potential therapeutic benefit of such repurposing has led us to review in detail the pathological features of ADPKD through the lens of the defined, classic hallmarks of cancer. In addition, we have evaluated features typical of ADPKD, and determined whether evidence supports the presence of such features in cancer cells. This analysis, which places pathological processes in the context of defined signalling pathways and approved signalling inhibitors, highlights potential avenues for further research and therapeutic exploitation in both diseases.
Collapse
Affiliation(s)
- Tamina Seeger-Nukpezah
- Department I of Internal Medicine and Centre for Integrated Oncology, University of Cologne, Kerpenerstrasse 62, D-50937 Cologne, Germany
| | - Daniel M Geynisman
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Anna S Nikonova
- Department of Developmental Therapeutics, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Thomas Benzing
- Department II of Internal Medicine and Centre for Molecular Medicine Cologne, University of Cologne, Kerpenerstrasse 62, D-50937 Cologne, Germany
| | - Erica A Golemis
- Department of Developmental Therapeutics, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| |
Collapse
|
34
|
Abstract
Cancer is widely recognized as a genetic disease in which somatic mutations are sequentially accumulated to drive tumor progression. Although genomic landscape studies are informative for individual cancer types, a comprehensive comparative study of tumorigenic mutations across cancer types based on integrative data sources is still a pressing need. We systematically analyzed ~10(6) non-synonymous mutations extracted from COSMIC, involving ~8000 genome-wide screened samples across 23 major human cancers at both the amino acid and gene levels. Our analysis identified cancer-specific heterogeneity that traditional nucleotide variation analysis alone usually overlooked. Particularly, the amino acid arginine (R) turns out to be the most favorable target of amino acid alteration in most cancer types studied (P < 10(-9), binomial test), reflecting its important role in cellular physiology. The tumor suppressor gene TP53 is mutated exclusively with the HYDIN, KRAS, and PTEN genes in large intestine, lung, and endometrial cancers respectively, indicating that TP53 takes part in different signaling pathways in different cancers. While some of our analyses corroborated previous observations, others indicated relevant candidates with high priority for further experimental validation. Our findings have many ramifications in understanding the etiology of cancer and the underlying molecular mechanisms in particular cancers.
Collapse
|
35
|
Gregory KB, Momin AA, Coombes KR, Baladandayuthapani V. Latent Feature Decompositions for Integrative Analysis of Multi-Platform Genomic Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2014; 11:984-994. [PMID: 26146492 PMCID: PMC4486317 DOI: 10.1109/tcbb.2014.2325035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Increased availability of multi-platform genomics data on matched samples has sparked research efforts to discover how diverse molecular features interact both within and between platforms. In addition, simultaneous measurements of genetic and epigenetic characteristics illuminate the roles their complex relationships play in disease progression and outcomes. However, integrative methods for diverse genomics data are faced with the challenges of ultra-high dimensionality and the existence of complex interactions both within and between platforms. We propose a novel modeling framework for integrative analysis based on decompositions of the large number of platform-specific features into a smaller number of latent features. Subsequently we build a predictive model for clinical outcomes accounting for both within- and between-platform interactions based on Bayesian model averaging procedures. Principal components, partial least squares and non-negative matrix factorization as well as sparse counterparts of each are used to define the latent features, and the performance of these decompositions is compared both on real and simulated data. The latent feature interactions are shown to preserve interactions between the original features and not only aid prediction but also allow explicit selection of outcome-related features. The methods are motivated by and applied to a glioblastoma multiforme data set from The Cancer Genome Atlas to predict patient survival times integrating gene expression, microRNA, copy number and methylation data. For the glioblastoma data, we find a high concordance between our selected prognostic genes and genes with known associations with glioblastoma. In addition, our model discovers several relevant cross-platform interactions such as copy number variation associated gene dosing and epigenetic regulation through promoter methylation. On simulated data, we show that our proposed method successfully incorporates interactions within and between genomic platforms to aid accurate prediction and variable selection. Our methods perform best when principal components are used to define the latent features.
Collapse
Affiliation(s)
- Karl B. Gregory
- PhD candidate in Department of Statistics, Texas A&M University, College Station, TX, 77843-3143, USA
| | - Amin A. Momin
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, 77230-1402, USA
| | - Kevin R. Coombes
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, USA
| | | |
Collapse
|
36
|
Mesri M. Advances in Proteomic Technologies and Its Contribution to the Field of Cancer. Adv Med 2014; 2014:238045. [PMID: 26556407 PMCID: PMC4590950 DOI: 10.1155/2014/238045] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 06/30/2014] [Indexed: 12/12/2022] Open
Abstract
Systematic studies of the cancer genome have generated a wealth of knowledge in recent years. These studies have uncovered a number of new cancer genes not previously known to be causal targets in cancer. Genetic markers can be used to determine predisposition to tumor development, but molecularly targeted treatment strategies are not widely available for most cancers. Precision care plans still must be developed by understanding and implementing basic science research into clinical treatment. Proteomics is continuing to make major strides in the discovery of fundamental biological processes as well as more recent transition into an assay platform capable of measuring hundreds of proteins in any biological system. As such, proteomics can translate basic science discoveries into the clinical practice of precision medicine. The proteomic field has progressed at a fast rate over the past five years in technology, breadth and depth of applications in all areas of the bioscience. Some of the previously experimental technical approaches are considered the gold standard today, and the community is now trying to come to terms with the volume and complexity of the data generated. Here I describe contribution of proteomics in general and biological mass spectrometry in particular to cancer research, as well as related major technical and conceptual developments in the field.
Collapse
Affiliation(s)
- Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
37
|
Schröck A, Bode M, Göke FJM, Bareiss PM, Schairer R, Wang H, Weichert W, Franzen A, Kirsten R, van Bremen T, Queisser A, Kristiansen G, Heasley L, Bootz F, Lengerke C, Perner S. Expression and role of the embryonic protein SOX2 in head and neck squamous cell carcinoma. Carcinogenesis 2014; 35:1636-42. [PMID: 24743512 DOI: 10.1093/carcin/bgu094] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Recently, SOX2 has been identified as a potential lineage-specific oncogene in lung squamous cell carcinomas. Since head and neck squamous cell carcinomas (HNSCC) are morphologically and clinically highly related to lung squamous cell carcinomas, we hypothesized that SOX2 also plays an oncogenic role in this tumor entity. We assembled a cohort of 496 patients with HNSCC, including 253 metastases and 135 recurrences. SOX2 amplification (FISH) and SOX2 protein expression (immunohistochemistry) were correlated with molecular and clinicopathological parameters. In order to investigate the functional role of SOX2 in human HNSCC, SOX2 knockdown and overexpression in SCC-25 cells were generated by lentiviral constructs and subjected to cell cycle analysis, proliferation and apoptosis assays. Furthermore, SOX2 expression was correlated with the expression of proliferation and apoptosis-related proteins in primary HNSCC samples. SOX2 amplification was detected in 21% of primary HNSCC and mostly observed in a concordant manner between primary tumors and corresponding metastatic tissues. Overall, SOX2 amplification resulted in protein overexpression and was mutually exclusive with human papillomavirus infection. SOX2 protein overexpression was associated with clinicopathological parameters of worse outcome. Functionally, SOX2 induced the expression of the antiapoptotic protein BCL-2 and enhanced resistance to apoptosis-inducing agents including cisplatin, indicating SOX2 as a mediator of therapy resistance in human HNSCC. Targeting SOX2 and related molecular downstream pathways such as BCL-2 may enhance therapy efficacy in SOX2-expressing HNSCC.
Collapse
Affiliation(s)
- Andreas Schröck
- Department of Otorhinolaryngology/Head and Neck Surgery, University Hospital of Bonn, 53127 Bonn, Germany, Department of Prostate Cancer Research, Institute of Pathology, University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Maike Bode
- Department of Prostate Cancer Research, Institute of Pathology, University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Friederike Johanna Maria Göke
- Department of Prostate Cancer Research, Institute of Pathology, University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | | | - Rebekka Schairer
- Medical Center II, University Hospital of Tuebingen, 72076 Tuebingen, Germany
| | - Hui Wang
- Medical Center II, University Hospital of Tuebingen, 72076 Tuebingen, Germany
| | - Wilko Weichert
- Institute of Pathology, University Hospital of Heidelberg, 69120 Heidelberg, Germany and
| | - Alina Franzen
- Department of Prostate Cancer Research, Institute of Pathology, University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Robert Kirsten
- Department of Prostate Cancer Research, Institute of Pathology, University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Tobias van Bremen
- Department of Otorhinolaryngology/Head and Neck Surgery, University Hospital of Bonn, 53127 Bonn, Germany
| | - Angela Queisser
- Department of Prostate Cancer Research, Institute of Pathology, University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Glen Kristiansen
- Department of Prostate Cancer Research, Institute of Pathology, University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Lynn Heasley
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Friedrich Bootz
- Department of Otorhinolaryngology/Head and Neck Surgery, University Hospital of Bonn, 53127 Bonn, Germany
| | - Claudia Lengerke
- Medical Center II, University Hospital of Tuebingen, 72076 Tuebingen, Germany
| | - Sven Perner
- Department of Prostate Cancer Research, Institute of Pathology, University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany,
| |
Collapse
|
38
|
Imielinski M, Hammerman PS, Thomas R, Meyerson M. Somatic Genome Alterations in Human Lung Cancers. Lung Cancer 2014. [DOI: 10.1002/9781118468791.ch4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
39
|
Yeste-Velasco M, Mao X, Grose R, Kudahetti SC, Lin D, Marzec J, Vasiljević N, Chaplin T, Xue L, Xu M, Foster JM, Karnam SS, James SY, Chioni AM, Gould D, Lorincz AT, Oliver RTD, Chelala C, Thomas GM, Shipley JM, Mather SJ, Berney DM, Young BD, Lu YJ. Identification of ZDHHC14 as a novel human tumour suppressor gene. J Pathol 2014; 232:566-77. [PMID: 24407904 DOI: 10.1002/path.4327] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 12/20/2013] [Accepted: 01/03/2014] [Indexed: 01/19/2023]
Abstract
Genomic changes affecting tumour suppressor genes are fundamental to cancer. We applied SNP array analysis to a panel of testicular germ cell tumours to search for novel tumour suppressor genes and identified a frequent small deletion on 6q25.3 affecting just one gene, ZDHHC14. The expression of ZDHHC14, a putative protein palmitoyltransferase with unknown cellular function, was decreased at both RNA and protein levels in testicular germ cell tumours. ZDHHC14 expression was also significantly decreased in a panel of prostate cancer samples and cell lines. In addition to our findings of genetic and protein expression changes in clinical samples, inducible overexpression of ZDHHC14 led to reduced cell viability and increased apoptosis through the classic caspase-dependent apoptotic pathway and heterozygous knockout of ZDHHC14 increased [CORRECTED] cell colony formation ability. Finally, we confirmed our in vitro findings of the tumour suppressor role of ZDHHC14 in a mouse xenograft model, showing that overexpression of ZDHHC14 inhibits tumourigenesis. Thus, we have identified a novel tumour suppressor gene that is commonly down-regulated in testicular germ cell tumours and prostate cancer, as well as given insight into the cellular functional role of ZDHHC14, a potential protein palmitoyltransferase that may play a key protective role in cancer.
Collapse
Affiliation(s)
- Marc Yeste-Velasco
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Schröck A, Göke F, Wagner P, Bode M, Franzen A, Huss S, Agaimy A, Ihrler S, Kirsten R, Kristiansen G, Bootz F, Lengerke C, Perner S. Fibroblast growth factor receptor-1 as a potential therapeutic target in sinonasal cancer. Head Neck 2014; 36:1253-7. [PMID: 23913758 DOI: 10.1002/hed.23443] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 04/18/2013] [Accepted: 07/25/2013] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Despite multimodal treatment, sinonasal malignancies have an unfavorable prognosis. The purpose of this study was to elucidate if these tumors harbor amplifications of the fibroblast growth factor receptor 1 (FGFR1) gene, which has recently been identified as a potential therapeutic target in squamous cell lung cancer. METHODS One hundred twelve primary tumors (including squamous cell carcinoma [SCC], carcinoma associated with an inverted papilloma, sinonasal undifferentiated carcinoma [SNUC], adenocarcinoma, adenoid cystic carcinoma [ACC], esthesioneuroblastoma, and 9 corresponding lymph node metastases) were assessed by fluorescence in situ hybridization (FISH) for FGFR1 copy number status. Human papillomavirus (HPV) status was assessed by p16 immunohistochemical as a surrogate marker. RESULTS FGFR1 amplification was found in subsets of sinonasal SCCs (20%), carcinomas associated with an inverted papilloma (33%), and SNUCs (5%). In all cases, metastatic tumor samples shared the same FGFR1 amplification status as the corresponding primary tumor tissue. None of the FGFR1-amplified tumors expressed p16. CONCLUSION FGFR1 amplification represents a potential molecular target in a subset of patients with sinonasal cancer. © 2014 Wiley Periodicals, Inc. Head Neck 36: 1253-1257, 2014.
Collapse
Affiliation(s)
- Andreas Schröck
- Department of Otorhinolaryngology / Head and Neck Surgery, University of Bonn, Germany; Department of Prostate Cancer Research, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Castellano E, Sheridan C, Thin M, Nye E, Spencer-Dene B, Diefenbacher M, Moore C, Kumar M, Murillo M, Grönroos E, Lassailly F, Stamp G, Downward J. Requirement for interaction of PI3-kinase p110α with RAS in lung tumor maintenance. Cancer Cell 2013; 24:617-30. [PMID: 24229709 PMCID: PMC3826036 DOI: 10.1016/j.ccr.2013.09.012] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 08/27/2013] [Accepted: 09/24/2013] [Indexed: 12/18/2022]
Abstract
RAS proteins directly activate PI3-kinases. Mice bearing a germline mutation in the RAS binding domain of the p110α subunit of PI3-kinse are resistant to the development of RAS-driven tumors. However, it is unknown whether interaction of RAS with PI3-kinase is required in established tumors. The need for RAS interaction with p110α in the maintenance of mutant Kras-driven lung tumors was explored using an inducible mouse model. In established tumors, removal of the ability of p110α to interact with RAS causes long-term tumor stasis and partial regression. This is a tumor cell-autonomous effect, which is improved significantly by combination with MEK inhibition. Total removal of p110α expression or activity has comparable effects, albeit with greater toxicities.
Collapse
Affiliation(s)
- Esther Castellano
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Clare Sheridan
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - May Zaw Thin
- In Vivo Imaging Facility, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Emma Nye
- Experimental Histopathology Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Bradley Spencer-Dene
- Experimental Histopathology Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Markus E. Diefenbacher
- Mammalian Genetics Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Christopher Moore
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Madhu S. Kumar
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Miguel M. Murillo
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
- Lung Cancer Group, Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Eva Grönroos
- Translational Cancer Therapeutics Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Francois Lassailly
- In Vivo Imaging Facility, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Gordon Stamp
- Experimental Histopathology Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Julian Downward
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
- Lung Cancer Group, Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| |
Collapse
|
42
|
Fibroblast growth factor receptor 1 amplification is a common event in squamous cell carcinoma of the head and neck. Mod Pathol 2013; 26:1298-306. [PMID: 23619603 DOI: 10.1038/modpathol.2013.58] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 02/06/2013] [Indexed: 01/28/2023]
Abstract
Recently, we characterized fibroblast growth factor receptor 1 amplification as a target for a rational therapy in lung squamous cell carcinoma. Patients harboring this genetic event are currently eligible for treatment with antifibroblast growth factor receptor small-molecule inhibitors in phase I clinical trials. This has the potential to significantly improve standard therapy for lung squamous cell carcinoma patients. The aim of this study was to elucidate whether fibroblast growth factor receptor 1 amplification is also a common genetic event in head and neck squamous cell carcinoma. For this purpose, we assembled a cohort of 555 patients, including 264 with metastatic disease and 147 with recurrent disease. Formalin-fixed, paraffin-embedded material of primary tumors, metastases and recurrences were assessed for fibroblast growth factor receptor 1 copy number status using fluorescence in situ hybridization. Human papilloma virus status was detected by p16 immunohistochemistry staining and PCR-ELISA. Molecular parameters were correlated with each other and with clinicopathological data. We found 15% of primary head and neck squamous cell carcinoma to display a fibroblast growth factor receptor 1 amplification. In nearly all cases, metastatic and recurrent tumor samples shared the same amplification status as the corresponding primary tumor. Fibroblast growth factor receptor 1 amplification was associated with nicotine and alcohol consumption, but was mutually exclusive with human papilloma virus infection. Amplification of the gene was associated with parameters of worse outcome. Our data identify fibroblast growth factor receptor 1 amplification as a frequent event in primary and metastatic head and neck squamous cell carcinoma and represents a potential biomarker for more aggressive disease. Fibroblast growth factor receptor 1-amplified tumors could potentially comprise a subset of head and neck squamous cell carcinoma against which targeted small-molecule inhibitors hold therapeutic efficacy.
Collapse
|
43
|
Policastro LL, Ibañez IL, Notcovich C, Duran HA, Podhajcer OL. The tumor microenvironment: characterization, redox considerations, and novel approaches for reactive oxygen species-targeted gene therapy. Antioxid Redox Signal 2013; 19:854-95. [PMID: 22794113 DOI: 10.1089/ars.2011.4367] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The tumor microenvironment is a complex system that involves the interaction between malignant and neighbor stromal cells embedded in a mesh of extracellular matrix (ECM) components. Stromal cells (fibroblasts, endothelial, and inflammatory cells) are co-opted at different stages to help malignant cells invade the surrounding ECM and disseminate. Malignant cells have developed adaptive mechanisms to survive under the extreme conditions of the tumor microenvironment such as restricted oxygen supply (hypoxia), nutrient deprivation, and a prooxidant state among others. These conditions could be eventually used to target drugs that will be activated specifically in this microenvironment. Preclinical studies have shown that modulating cellular/tissue redox state by different gene therapy (GT) approaches was able to control tumor growth. In this review, we describe the most relevant features of the tumor microenvironment, addressing reactive oxygen species-generating sources that promote a prooxidative microenvironment inside the tumor mass. We describe different GT approaches that promote either a decreased or exacerbated prooxidative microenvironment, and those that make use of the differential levels of ROS between cancer and normal cells to achieve tumor growth inhibition.
Collapse
Affiliation(s)
- Lucia Laura Policastro
- Department of Micro and Nanotechnology, National Atomic Energy Commission, Buenos Aires 1650, Argentina.
| | | | | | | | | |
Collapse
|
44
|
Das K, Tan P. Molecular cytogenetics: recent developments and applications in cancer. Clin Genet 2013; 84:315-25. [DOI: 10.1111/cge.12229] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/02/2013] [Accepted: 07/02/2013] [Indexed: 12/13/2022]
Affiliation(s)
- K Das
- Cancer and Stem Cell Biology; Duke-NUS Graduate Medical School; Singapore Singapore
| | - P Tan
- Cancer and Stem Cell Biology; Duke-NUS Graduate Medical School; Singapore Singapore
- Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine; National University of Singapore; Singapore Singapore
- Genome Institute of Singapore; Singapore Singapore
- Cellular and Molecular Research; National Cancer Centre of Singapore; Singapore Singapore
| |
Collapse
|
45
|
Hallaj-Nezhadi S, Valizadeh H, Baradaran B, Dobakhti F, Lotfipour F. Preparation and characterization of gelatin nanoparticles containing pDNA encoding IL-12 and their expression in CT-26 carcinoma cells. Future Oncol 2013; 9:1195-206. [DOI: 10.2217/fon.13.82] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Gelatin as a biodegradable, nontoxic and biocompatible natural protein is a good candidate for gene delivery. In this study, pDNA-loaded gelatin nanoparticles were prepared and characterized for the expression of the cytokine IL-12 and anti-tumor effects. Materials & methods: Gelatin–pUMVC3–hIL-12 nanoparticles were prepared by the ethanol precipitation technique and evaluated for physicochemical characteristics, cytotoxiciy and transfection efficiency. Results: The prepared particles were spherical in shape with sizes varying from 344.27 to 826.23 nm, ζ-potentials between -944 and -165 mV, and greater than 97% encapsulation efficiency. The particles were nontoxic to CT-26 carcinoma cells. The nanoparticles prepared using 0.5% gelatin solution (G14) with a mean particle size of 816.87 nm (polydispersity index = 0.56 ± 0.01) demonstrated maximum transfection efficiency with 2.5-times higher expression compared with the naked plasmid. Conclusion: Gelatin–DNA nanoparticles using 0.5% gelatin solution had minimal cytotoxicity and can be used as a suitable candidate for further gene delivery studies and applications.
Collapse
Affiliation(s)
- Somayeh Hallaj-Nezhadi
- Drug Applied Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center & Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faramarz Dobakhti
- Faculty of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Farzaneh Lotfipour
- Hematology Oncology Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
46
|
Abstract
SIGNIFICANCE Diet exerts a major influence on the risk for developing cancer and heart disease. Food factors such as flavonoids are alleged to protect cells from premature aging and disease by shielding DNA, proteins, and lipids from oxidative damage. RECENT ADVANCES Our work has focused on clarifying the effects of dietary components on cancer cell proliferation and tumor growth, discovering mechanisms to explain the effects, and identifying the specific molecular targets of these compounds. Our strategy for identifying specific molecular targets of phytochemicals involves the use of supercomputer technology combined with protein crystallography, molecular biology, and experimental laboratory verification. CRITICAL ISSUES One of the greatest challenges for scientists is to reduce the accumulation of distortion and half truths reported in the popular media regarding the health benefits of certain foods or food supplements. The use of these is not new, but interest has increased dramatically because of perceived health benefits that are presumably acquired without unpleasant side effects. Flavonoids are touted to exert many beneficial effects in vitro. However, whether they can produce these effects in vivo is disputed. FUTURE DIRECTIONS The World Health Organization indicates that one third of all cancer deaths are preventable and that diet is closely linked to prevention. Based on this idea and epidemiological findings, attention has centered on dietary phytochemicals as an effective intervention in cancer development. However, an unequivocal link between diet and cancer has not been established. Thus, identifying cancer preventive dietary agents with specific molecular targets is essential to move forward toward successful cancer prevention.
Collapse
Affiliation(s)
- Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | | |
Collapse
|
47
|
Göke F, Franzen A, Menon R, Goltz D, Kirsten R, Boehm D, Vogel W, Göke A, Scheble V, Ellinger J, Gerigk U, Fend F, Wagner P, Schroeck A, Perner S. Rationale for treatment of metastatic squamous cell carcinoma of the lung using fibroblast growth factor receptor inhibitors. Chest 2013; 142:1020-1026. [PMID: 22499828 DOI: 10.1378/chest.11-2943] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND We previously identified amplification of the fibroblast growth factor receptor 1 gene (FGFR1) as a potential therapeutic target for small-molecule inhibitor therapy in squamous cell lung cancer (L-SCC). Currently, clinical phase I trials are underway to examine whether patients with FGFR1-amplified L-SCC benefit from a targeted therapy approach using small-molecule inhibitors. Because most patients with lung cancer present with metastatic disease, we investigated whether lymph node metastases in L-SCC share the FGFR1 amplification status of their corresponding primary tumor. METHODS The study cohort consisted of 72 patients with L-SCC, 39 with regional lymph node metastases. Tissue microarrays were constructed from formalin-fixed, paraffin-embedded tissue of the primary tumors and, where present, of the corresponding lymph node metastasis. A biotin-labeled target probe spanning the FGFR1 locus (8p11.22-23) was used to determine the FGFR1 amplification status by fluorescence in situ hybridization. RESULTS FGFR1 amplification was detected in 16% (12 of 72) of all primary L-SCCs. In metastatic tumors, 18% (seven of 39) of the lymph node metastases displayed FGFR1 amplification with an exact correlation of FGFR1 amplification status between tumor and metastatic tissue. CONCLUSIONS FGFR1 amplification is a common genetic event occurring at a frequency of 16% in L-SCCs. Moreover, lymph node metastases derived from FGFR1-amplified L-SCCs also exhibit FGFR1 amplification. Therefore, we suggest that the FGFR1 amplification is a clonal event in tumor progression. Beyond this biologically relevant observation, the findings carry potential therapeutic implications in that small-molecule inhibitors may be applicable to the treatment of a subset of patients with metastatic L-SCC.
Collapse
Affiliation(s)
- Friederike Göke
- Institute of Pathology, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany; Institute of Prostate Cancer Research, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Alina Franzen
- Institute of Pathology, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany; Institute of Prostate Cancer Research, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Roopika Menon
- Institute of Pathology, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany; Institute of Prostate Cancer Research, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Diane Goltz
- Institute of Pathology, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Robert Kirsten
- Institute of Prostate Cancer Research, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Diana Boehm
- Institute of Prostate Cancer Research, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Wenzel Vogel
- Institute of Prostate Cancer Research, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Antonia Göke
- Institute of Prostate Cancer Research, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Veit Scheble
- Department of Hematology and Oncology, University Hospital of Tuebingen, Tuebingen, Germany
| | - Joerg Ellinger
- Department of Urology, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Ulrich Gerigk
- Department of Thorax Surgery, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Falko Fend
- Institute of Pathology, University Hospital of Tuebingen, Tuebingen, Germany
| | - Patrick Wagner
- Division of Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Andreas Schroeck
- Institute of Prostate Cancer Research, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany; Department of Head and Neck Surgery, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany
| | - Sven Perner
- Institute of Pathology, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany; Institute of Prostate Cancer Research, Affiliated Malteser Hospital, University Hospital of Bonn, Bonn, Germany.
| |
Collapse
|
48
|
Ho SK, Thike AA, Cheok PY, Tse GMK, Tan PH. Phyllodes tumours of the breast: the role of CD34, vascular endothelial growth factor and β-catenin in histological grading and clinical outcome. Histopathology 2013; 63:393-406. [DOI: 10.1111/his.12177] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 04/28/2013] [Indexed: 12/15/2022]
Affiliation(s)
- Soo Keng Ho
- Department of Pathology; Singapore General Hospital; Singapore
| | - Aye Aye Thike
- Department of Pathology; Singapore General Hospital; Singapore
| | - Poh Yian Cheok
- Department of Pathology; Singapore General Hospital; Singapore
| | - Gary M-K Tse
- Department of Anatomical and Cellular Pathology; Chinese Univesity of Hong Kong
| | - Puay Hoon Tan
- Department of Pathology; Singapore General Hospital; Singapore
| |
Collapse
|
49
|
Liu X, Wang J, Chen L. Whole-exome sequencing reveals recurrent somatic mutation networks in cancer. Cancer Lett 2012; 340:270-6. [PMID: 23153794 DOI: 10.1016/j.canlet.2012.11.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/30/2012] [Accepted: 11/02/2012] [Indexed: 11/26/2022]
Abstract
The second-generation sequencing technologies have been extensively used to reveal the mechanism of tumorigenesis and find critical genes in cancer progression that can be potential targets of clinic treatment. Exome is a part of genome formed by exons which are the protein-coding portions of genes. The whole-exome sequencing information can reflect the mutations of the protein-coding region in the genome and depict the causal relationship between the mutations and phenotypes. Now, many network-based methods have been developed to identify cancer driver modules or pathways, which not only provide new insights into molecular mechanism of disease progression at network level but also can avoid low coverage or lowly recurrent on disease samples in contrast to individual driver genes. In this review, we focus on the recent advances on network-based methods for identifying cancer driver modules or pathways, including methods of whole-exome sequencing, somatic mutation detection, driver mutation identification, and mutation network reconstruction.
Collapse
Affiliation(s)
- Xiaoping Liu
- Key Laboratory of Systems Biology, SIBS-Novo Nordisk Translational Research Centre for PreDiabetes, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | |
Collapse
|
50
|
Wang W, Baladandayuthapani V, Morris JS, Broom BM, Manyam G, Do KA. iBAG: integrative Bayesian analysis of high-dimensional multiplatform genomics data. ACTA ACUST UNITED AC 2012; 29:149-59. [PMID: 23142963 PMCID: PMC3546799 DOI: 10.1093/bioinformatics/bts655] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Motivation: Analyzing data from multi-platform genomics experiments combined
with patients’ clinical outcomes helps us understand the complex biological
processes that characterize a disease, as well as how these processes relate to the
development of the disease. Current data integration approaches are limited in that they
do not consider the fundamental biological relationships that exist among the data
obtained from different platforms. Statistical Model: We propose an integrative Bayesian analysis of genomics
data (iBAG) framework for identifying important genes/biomarkers that are associated with
clinical outcome. This framework uses hierarchical modeling to combine the data obtained
from multiple platforms into one model. Results: We assess the performance of our methods using several synthetic
and real examples. Simulations show our integrative methods to have higher power to detect
disease-related genes than non-integrative methods. Using the Cancer Genome Atlas
glioblastoma dataset, we apply the iBAG model to integrate gene expression and methylation
data to study their associations with patient survival. Our proposed method discovers
multiple methylation-regulated genes that are related to patient survival, most of which
have important biological functions in other diseases but have not been previously studied
in glioblastoma. Availability:http://odin.mdacc.tmc.edu/∼vbaladan/. Contact:veera@mdanderson.org Supplementary information:Supplementary data are available at Bioinformatics
online.
Collapse
Affiliation(s)
- Wenting Wang
- Department of Biostatistics, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|