1
|
Webster AP, Thirlwell C. The Molecular Biology of Midgut Neuroendocrine Neoplasms. Endocr Rev 2024; 45:343-350. [PMID: 38123518 PMCID: PMC11074790 DOI: 10.1210/endrev/bnad034] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 10/12/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023]
Abstract
Midgut neuroendocrine neoplasms (NENs) are one of the most common subtypes of NEN, and their incidence is rising globally. Despite being the most frequently diagnosed malignancy of the small intestine, little is known about their underlying molecular biology. Their unusually low mutational burden compared to other solid tumors and the unexplained occurrence of multifocal tumors makes the molecular biology of midgut NENs a particularly fascinating field of research. This review provides an overview of recent advances in the understanding of the interplay of the genetic, epigenetic, and transcriptomic landscape in the development of midgut NENs, a topic that is critical to understanding their biology and improving treatment options and outcomes for patients.
Collapse
Affiliation(s)
- Amy P Webster
- Department of Clinical and Biomedical Science, University of Exeter College of Medicine and Health, Exeter, EX2 5DW, UK
| | - Chrissie Thirlwell
- Department of Clinical and Biomedical Science, University of Exeter College of Medicine and Health, Exeter, EX2 5DW, UK
- University of Bristol Medical School, University of Bristol, Bristol, BS8 1UD, UK
| |
Collapse
|
2
|
Mestre-Alagarda C, Srirajaskanthan R, Zen Y, Giwa M, Howard M, Ooft ML. Genetic and epigenetic prognosticators of neuroendocrine tumours of the GI tract, liver, biliary tract and pancreas: A systematic review and meta-analysis. Histopathology 2024; 84:255-265. [PMID: 37565289 DOI: 10.1111/his.15025] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/25/2023] [Accepted: 07/30/2023] [Indexed: 08/12/2023]
Abstract
Multiple recurrent genetic and epigenetic aberrations have been associated with worse prognosis in multiple studies of neuroendocrine tumours (NETs), but these have been mainly small cohorts and univariate analysis. This review and meta-analysis will focus upon the literature available on NETs of the gastrointestinal (GI) tract, liver, biliary tract and pancreas. PubMed and Embase were searched for publications that investigated the prognostic value of (epi)genetic changes of neuroendocrine tumours. A meta-analysis was performed assessing the association of the (epi)genetic alterations with overall survival (OS), disease-free survival (DFS) or locoregional control (LRC). In the pancreas DAXX/ATRX [hazard ratio (HR) = 3.29; 95% confidence interval (CI) = 2.28-4.74] and alternative lengthening telomeres (ALT) activation (HR = 8.20; 95% CI = 1.40-48.07) showed a pooled worse survival. In the small bowel NETs gains on chromosome 14 were associated with worse survival (HR 2.85; 95% CI = 1.40-5.81). NETs from different anatomical locations must be regarded as different biological entities with diverging molecular prognosticators, and epigenetic changes being important to the pathogenesis of these tumours. This review underpins the prognostic drivers of pancreatic NET which lie in mutations of DAXX/ATRX and ALT pathways. However, there is reaffirmation that prognostic molecular biomarkers of small bowel NETs should be sought in copy number variations (CNVs) rather than in single nucleotide variations (SNVs). This review also reveals how little is known about the prognostic significance of epigenetics in NETs.
Collapse
Affiliation(s)
| | | | - Yoh Zen
- Institute of Liver Studies, King's College Hospital and King's College London, London, UK
| | - Mojisola Giwa
- Department of Histopathology, King's College Hospital, King's College, London, UK
| | - Mark Howard
- Department of Histopathology, King's College Hospital, King's College, London, UK
| | - Marc Lucas Ooft
- Institute of Liver Studies, King's College Hospital, London, UK
- Pathology-DNA, Rijnstate Hospital, Arnhem, the Netherlands
| |
Collapse
|
3
|
Padwal MK, Basu S, Basu B. Application of Machine Learning in Predicting Hepatic Metastasis or Primary Site in Gastroenteropancreatic Neuroendocrine Tumors. Curr Oncol 2023; 30:9244-9261. [PMID: 37887568 PMCID: PMC10605255 DOI: 10.3390/curroncol30100668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) account for 80% of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs). GEP-NETs are well-differentiated tumors, highly heterogeneous in biology and origin, and are often diagnosed at the metastatic stage. Diagnosis is commonly through clinical symptoms, histopathology, and PET-CT imaging, while molecular markers for metastasis and the primary site are unknown. Here, we report the identification of multi-gene signatures for hepatic metastasis and primary sites through analyses on RNA-SEQ datasets of pancreatic and small intestinal NETs tissue samples. Relevant gene features, identified from the normalized RNA-SEQ data using the mRMRe algorithm, were used to develop seven Machine Learning models (LDA, RF, CART, k-NN, SVM, XGBOOST, GBM). Two multi-gene random forest (RF) models classified primary and metastatic samples with 100% accuracy in training and test cohorts and >90% accuracy in an independent validation cohort. Similarly, three multi-gene RF models identified the pancreas or small intestine as the primary site with 100% accuracy in training and test cohorts, and >95% accuracy in an independent cohort. Multi-label models for concurrent prediction of hepatic metastasis and primary site returned >98.42% and >87.42% accuracies on training and test cohorts, respectively. A robust molecular signature to predict liver metastasis or the primary site for GEP-NETs is reported for the first time and could complement the clinical management of GEP-NETs.
Collapse
Affiliation(s)
- Mahesh Kumar Padwal
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai 400085, India;
- Homi Bhabha National Institute, Mumbai 400094, India;
| | - Sandip Basu
- Homi Bhabha National Institute, Mumbai 400094, India;
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Mumbai 400012, India
| | - Bhakti Basu
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai 400085, India;
- Homi Bhabha National Institute, Mumbai 400094, India;
| |
Collapse
|
4
|
Klomp MJ, Refardt J, van Koetsveld PM, Campana C, Dalm SU, Dogan F, van Velthuysen MLF, Feelders RA, de Herder WW, Hofland J, Hofland LJ. Epigenetic regulation of SST 2 expression in small intestinal neuroendocrine tumors. Front Endocrinol (Lausanne) 2023; 14:1184436. [PMID: 37223009 PMCID: PMC10200989 DOI: 10.3389/fendo.2023.1184436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 04/11/2023] [Indexed: 05/25/2023] Open
Abstract
Background Somatostatin receptor type 2 (SST2) expression is critical for the diagnosis and treatment of neuroendocrine tumors and is associated with improved patient survival. Recent data suggest that epigenetic changes such as DNA methylation and histone modifications play an important role in regulating SST2 expression and tumorigenesis of NETs. However, there are limited data on the association between epigenetic marks and SST2 expression in small intestinal neuroendocrine tumors (SI-NETs). Methods Tissue samples from 16 patients diagnosed with SI-NETs and undergoing surgical resection of the primary tumor at Erasmus MC Rotterdam were analysed for SST2 expression levels and epigenetic marks surrounding the SST2 promoter region, i.e. DNA methylation and histone modifications H3K27me3 and H3K9ac. As a control, 13 normal SI-tissue samples were included. Results The SI-NET samples had high SST2 protein and mRNA expression levels; a median (IQR) of 80% (70-95) SST2-positive cells and 8.2 times elevated SST2 mRNA expression level compared to normal SI-tissue (p=0.0042). In comparison to normal SI-tissue, DNA methylation levels and H3K27me3 levels were significantly lower at five out of the eight targeted CpG positions and at two out of the three examined locations within the SST2 gene promoter region of the SI-NET samples, respectively. No differences in the level of activating histone mark H3K9ac were observed between matched samples. While no correlation was found between histone modification marks and SST2 expression, SST2 mRNA expression levels correlated negatively with DNA methylation within the SST2 promoter region in both normal SI-tissue and SI-NETs (p=0.006 and p=0.04, respectively). Conclusion SI-NETs have lower SST2 promoter methylation levels and lower H3K27me3 methylation levels compared to normal SI-tissue. Moreover, in contrast to the absence of a correlation with SST2 protein expression levels, significant negative correlations were found between SST2 mRNA expression level and the mean level of DNA methylation within the SST2 promoter region in both normal SI-tissue and SI-NET tissue. These results indicate that DNA methylation might be involved in regulating SST2 expression. However, the role of histone modifications in SI-NETs remains elusive.
Collapse
Affiliation(s)
- Maria J. Klomp
- ENETS Center of Excellence, Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center (MC) Cancer Institute, Rotterdam, Netherlands
- ENETS Center of Excellence, Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Julie Refardt
- ENETS Center of Excellence, Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center (MC) Cancer Institute, Rotterdam, Netherlands
- ENETS Center of Excellence, Department of Endocrinology, University Hospital Basel, Basel, Switzerland
| | - Peter M. van Koetsveld
- ENETS Center of Excellence, Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center (MC) Cancer Institute, Rotterdam, Netherlands
| | - Claudia Campana
- ENETS Center of Excellence, Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center (MC) Cancer Institute, Rotterdam, Netherlands
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties, School of Medical and Pharmaceutical Sciences, University of Genova, Genova, Italy
| | - Simone U. Dalm
- ENETS Center of Excellence, Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Fadime Dogan
- ENETS Center of Excellence, Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center (MC) Cancer Institute, Rotterdam, Netherlands
| | | | - Richard A. Feelders
- ENETS Center of Excellence, Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center (MC) Cancer Institute, Rotterdam, Netherlands
| | - Wouter W. de Herder
- ENETS Center of Excellence, Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center (MC) Cancer Institute, Rotterdam, Netherlands
| | - Johannes Hofland
- ENETS Center of Excellence, Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center (MC) Cancer Institute, Rotterdam, Netherlands
| | - Leo J. Hofland
- ENETS Center of Excellence, Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center (MC) Cancer Institute, Rotterdam, Netherlands
| |
Collapse
|
5
|
Smith J, Barnett E, Rodger EJ, Chatterjee A, Subramaniam RM. Neuroendocrine Neoplasms: Genetics and Epigenetics. PET Clin 2023; 18:169-187. [PMID: 36858744 DOI: 10.1016/j.cpet.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Neuroendocrine neoplasms (NENs) are a group of rare, heterogeneous tumors of neuroendocrine cell origin, affecting a range of different organs. The clinical management of NENs poses significant challenges, as tumors are often diagnosed at an advanced stage where overall survival remains poor with current treatment regimens. In addition, a host of complex and often unique molecular changes underpin the pathobiology of each NEN subtype. Exploitation of the unique genetic and epigenetic signatures driving each NEN subtype provides an opportunity to enhance the diagnosis, treatment, and monitoring of NEN in an emerging era of individualized medicine.
Collapse
Affiliation(s)
- Jim Smith
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Te Whatu Ora - Southern, Dunedin Public Hospital, 270 Great King Street, PO Box 913, Dunedin, New Zealand.
| | - Edward Barnett
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Euan J Rodger
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Rathan M Subramaniam
- Department of Medicine, Otago Medical School, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Department of Radiology, Duke University, 2301 Erwin Rd, BOX 3808, Durham, NC 27705, USA
| |
Collapse
|
6
|
Association of LAMA1 Single-Nucleotide Polymorphisms with Risk of Esophageal Squamous Cell Carcinoma among the Eastern Chinese Population. JOURNAL OF ONCOLOGY 2023; 2023:6922909. [PMID: 36824663 PMCID: PMC9943613 DOI: 10.1155/2023/6922909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 02/18/2023]
Abstract
Introduction LAMA1, also known as laminin subunit α1, is a member of the laminin family, which is widely reported to be a key basement membrane molecule that affects various biological activities and is associated with many kinds of diseases. We aimed to investigate the association between LAMA1single-nucleotide polymorphisms and the occurrence and progression of esophageal squamous cell carcinoma in the Chinese population. Method 2,186 participants were collected retrospectively between October 2008 and January 2017, including 1,043 ESCC patients and 1,143 noncancer patients. A 2 mL blood sample was obtained intravenously for the LDR for SNP analysis. The 6 SNP loci of LAMA1 were selected and examined. We analyzed the association of several genetic models of 6 LAMA1 SNP loci, sex, age, smoking and drinking status, and the occurrence of esophageal squamous cell carcinoma. Results In the rs62081531 G > A locus, genotype GA was a protective factor for ESCC compared with GG (OR: 0.830, P=0.046), especially among the younger and nondrinkers. At rs607230 T > C, genotype TC was linked with a lower risk of ESCC compared with TT. (OR: 0.613, P=0.034). Haplotype Frequencies revealed that Ars62081531Grs621993Ars539713Trs566655Ars73938538Crs607230 (OR: 0.803, P=0.028) and Grs62081531Grs621993Ars539713Trs566655Crs73938538Crs607230 (OR: 0.679, P=0.010) were strongly associated with lower susceptibility of ESCC. Conclusion The LAMA1 rs62081531, rs539713, rs566655, and rs607230 polymorphisms were demonstrated to be related to susceptibility to ESCC in the Chinese population. LAMA1 SNPs may have a significant impact on the occurrence of esophageal cancer and may serve as potential diagnostic biomarkers.
Collapse
|
7
|
Peculis R, Rovite V, Megnis K, Balcere I, Breiksa A, Nazarovs J, Stukens J, Konrade I, Sokolovska J, Pirags V, Klovins J. Whole exome sequencing reveals novel risk genes of pituitary neuroendocrine tumors. PLoS One 2022; 17:e0265306. [PMID: 36026497 PMCID: PMC9417189 DOI: 10.1371/journal.pone.0265306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 03/01/2022] [Indexed: 11/21/2022] Open
Abstract
Somatic genetic alterations in pituitary neuroendocrine tumors (PitNET) tissues have been identified in several studies, but detection of overlapping somatic PitNET candidate genes is rare. We sequenced and by employing multiple data analysis methods studied the exomes of 15 PitNET patients to improve discovery of novel factors involved in PitNET development. PitNET patients were recruited to the study before PitNET removal surgery. For each patient, two samples for DNA extraction were acquired: venous blood and PitNET tissue. Exome sequencing was performed using Illumina NexSeq 500 sequencer and data analyzed using two separate workflows and variant calling algorithms: GATK and Strelka2. A combination of two data analysis pipelines discovered 144 PitNET specific somatic variants (mean = 9.6, range 0–19 per PitNET) of which all were SNVs. Also, we detected previously known GNAS PitNET mutation and identified somatic variants in 11 genes, which have contained somatic variants in previous WES and WGS studies of PitNETs. Noteworthy, this is the third study detecting somatic variants in gene RYR1 in the exomes of PitNETs. In conclusion, we have identified two novel PitNET candidate genes (AC002519.6 and AHNAK) with recurrent somatic variants in our PitNET cohort and found 13 genes overlapping from previous PitNET studies that contain somatic variants. Our study demonstrated that the use of multiple sequencing data analysis pipelines can provide more accurate identification of somatic variants in PitNETs.
Collapse
Affiliation(s)
- Raitis Peculis
- Human Genetics and Molecular Medicine, Latvian Biomedical Research and Study Centre, Riga, Latvia
- * E-mail:
| | - Vita Rovite
- Human Genetics and Molecular Medicine, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Kaspars Megnis
- Human Genetics and Molecular Medicine, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Inga Balcere
- Department of Internal Medicine, Riga Stradins University, Riga, Latvia
| | - Austra Breiksa
- Institute of Pathology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Jurijs Nazarovs
- Institute of Pathology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Janis Stukens
- Department of Neurosurgery, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Ilze Konrade
- Human Genetics and Molecular Medicine, Latvian Biomedical Research and Study Centre, Riga, Latvia
- Department of Internal Medicine, Riga Stradins University, Riga, Latvia
| | | | - Valdis Pirags
- Human Genetics and Molecular Medicine, Latvian Biomedical Research and Study Centre, Riga, Latvia
- Faculty of Medicine, University of Latvia, Riga, Latvia
- Department of Endocrinology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Janis Klovins
- Human Genetics and Molecular Medicine, Latvian Biomedical Research and Study Centre, Riga, Latvia
| |
Collapse
|
8
|
Mettler E, Fottner C, Bakhshandeh N, Trenkler A, Kuchen R, Weber MM. Quantitative Analysis of Plasma Cell-Free DNA and Its DNA Integrity and Hypomethylation Status as Biomarkers for Tumor Burden and Disease Progression in Patients with Metastatic Neuroendocrine Neoplasias. Cancers (Basel) 2022; 14:cancers14041025. [PMID: 35205773 PMCID: PMC8870292 DOI: 10.3390/cancers14041025] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Neuroendocrine neoplasias (NEN) are a heterogeneous group of frequent slow-progressing malignant tumors for which a reliable marker for tumor relapse and progression is still lacking. Previously, circulating cell-free DNA and its global methylation status and fragmentation rate have been proposed to be valuable prognostic tumor markers in a variety of malignancies. In the current study, we compared plasma cell-free DNA (cfDNA) properties of NEN patients with a healthy control group and a group of surgically cured patients. Our results revealed significantly higher plasma cfDNA concentrations with increased fragmentation and hypomethylation in patients with advanced metastatic NEN, which was strongly associated with tumor load and could help to differentiate between metastasized disease and presumably cured patients. This suggests that the combined analysis of plasma cfDNA characteristics is a potent and sensitive prognostic and therapeutic biomarker for tumor burden and disease progression in patients with neuroendocrine neoplasias. Abstract Background: Neuroendocrine neoplasia (NEN) encompasses a diverse group of malignancies marked by histological heterogeneity and highly variable clinical outcomes. Apart from Chromogranin A, specific biomarkers predicting residual tumor disease, tumor burden, and disease progression in NEN are scant. Thus, there is a strong clinical need for new and minimally invasive biomarkers that allow for an evaluation of the prognosis, clinical course, and response to treatment of NEN patients, thereby helping implement individualized treatment decisions in this heterogeneous group of patients. In the current prospective study, we evaluated the role of plasma cell-free DNA concentration and its global hypomethylation and fragmentation as possible diagnostic and prognostic biomarkers in patients with neuroendocrine neoplasias. Methods: The plasma cfDNA concentration, cfDNA Alu hypomethylation, and LINE-1 cfDNA integrity were evaluated prospectively in 63 NEN patients with presumably cured or advanced metastatic disease. The cfDNA characteristics in NEN patients were compared to the results of a group of 29 healthy controls and correlated with clinical and histopathological data of the patients. Results: Patients with advanced NEN showed a significantly higher cfDNA concentration and percentage of Alu hypomethylation and a reduced LINE-1 cfDNA integrity as compared to the surgically cured NET patients and the healthy control group. The increased hypomethylation and concentration of cfDNA and the reduced cfDNA integrity in NEN patients were strongly associated with tumor burden and poor prognosis, while no correlation with tumor grading, differentiation, localization, or hormonal activity could be found. Multiparametric ROC analysis of plasma cfDNA characteristics was able to distinguish NEN patients with metastatic disease from the control group and the cured NEN patients with AUC values of 0.694 and 0.908, respectively. This was significant even for the group with only a low tumor burden. Conclusions: The present study, for the first time, demonstrates that the combination of plasma cfDNA concentration, global hypomethylation, and fragment length pattern has the potential to serve as a potent and sensitive prognostic and therapeutic “liquid biopsy” biomarker for tumor burden and disease progression in patients with neuroendocrine neoplasias.
Collapse
Affiliation(s)
- Esther Mettler
- Department of Endocrinology and Metabolism, I Medical Clinic, University Hospital, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany; (C.F.); (N.B.); (A.T.); (M.M.W.)
- Correspondence:
| | - Christian Fottner
- Department of Endocrinology and Metabolism, I Medical Clinic, University Hospital, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany; (C.F.); (N.B.); (A.T.); (M.M.W.)
| | - Neda Bakhshandeh
- Department of Endocrinology and Metabolism, I Medical Clinic, University Hospital, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany; (C.F.); (N.B.); (A.T.); (M.M.W.)
| | - Anja Trenkler
- Department of Endocrinology and Metabolism, I Medical Clinic, University Hospital, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany; (C.F.); (N.B.); (A.T.); (M.M.W.)
| | - Robert Kuchen
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany;
| | - Matthias M. Weber
- Department of Endocrinology and Metabolism, I Medical Clinic, University Hospital, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany; (C.F.); (N.B.); (A.T.); (M.M.W.)
| |
Collapse
|
9
|
EZH2 presents a therapeutic target for neuroendocrine tumors of the small intestine. Sci Rep 2021; 11:22733. [PMID: 34815475 PMCID: PMC8611048 DOI: 10.1038/s41598-021-02181-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 11/10/2021] [Indexed: 12/13/2022] Open
Abstract
Small intestinal neuroendocrine tumors (SI-NETs) are slow-growing tumors that seem genetically quite stable without highly recurrent mutations, but are epigenetically dysregulated. In contrast to the undetectable expression of the enhancer of zeste homolog 2 (EZH2) histone methyltransferase in the enterochromaffin cells of the small intestine, we found high and differential expression of EZH2 in primary SI-NETs and corresponding metastases. Silencing EZH2 in the SI-NET cell line CNDT2.5 reduced cell proliferation and induced apoptosis. Furthermore, EZH2 knockout inhibited tumor progression in a CNDT2.5 SI-NET xenograft mouse model, and treatment of SI-NET cell lines CNDT2.5 and GOT1 with the EZH2-specific inhibitor CPI-1205 decreased cell viability and promoted apoptosis. Moreover, CPI-1205 treatment reduced migration capacity of CNDT2.5 cells. The EZH2 inhibitor GSK126 also repressed proliferation of CNDT2.5 cells. Recently, metformin has received wide attention as a therapeutic option in diverse cancers. In CNDT2.5 and GOT1 cells, metformin suppressed EZH2 expression, and inhibited cell proliferation. Exposure of GOT1 three-dimensional cell spheroids to CPI-1205 or metformin arrested cell proliferation and decreased spheroid size. These novel findings support a possible role of EZH2 as a candidate oncogene in SI-NETs, and suggest that CPI-1205 and metformin should be further evaluated as therapeutic options for patients with SI-NETs.
Collapse
|
10
|
Carpizo DR, Harris CR. Genetic Drivers of Ileal Neuroendocrine Tumors. Cancers (Basel) 2021; 13:cancers13205070. [PMID: 34680217 PMCID: PMC8533727 DOI: 10.3390/cancers13205070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Although ileal neuroendocrine tumors are the most common tumors of the small intestine, they are not well-defined at the genetic level. Unlike most cancers, they have an unusually low number of mutations, and also lack recurrently mutated genes. Moreover ileal NETs have been difficult to study in the laboratory because there were no animal models and because cell lines were generally unavailable. But recent advances, including the first ileal NET mouse model as well as methods for culturing patient tumor samples, have been described and have already helped to identify IGF2 and CDK4 as two of the genetic drivers for this tumor type. These advances may help in the development of new treatments for patients. Abstract The genetic causes of ileal neuroendocrine tumors (ileal NETs, or I-NETs) have been a mystery. For most types of tumors, key genes were revealed by large scale genomic sequencing that demonstrated recurrent mutations of specific oncogenes or tumor suppressors. In contrast, genomic sequencing of ileal NETs demonstrated a distinct lack of recurrently mutated genes, suggesting that the mechanisms that drive the formation of I-NETs may be quite different than the cell-intrinsic mutations that drive the formation of other tumor types. However, recent mouse studies have identified the IGF2 and RB1 pathways in the formation of ileal NETs, which is supported by the subsequent analysis of patient samples. Thus, ileal NETs no longer appear to be a cancer without genetic causes.
Collapse
|
11
|
Sharma R, Lythgoe MP, Slaich B, Patel N. Exploring the Epigenome in Gastroenteropancreatic Neuroendocrine Neoplasias. Cancers (Basel) 2021; 13:4181. [PMID: 34439335 PMCID: PMC8394968 DOI: 10.3390/cancers13164181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/06/2021] [Accepted: 08/17/2021] [Indexed: 11/17/2022] Open
Abstract
Gastroenteropancreatic neuroendocrine neoplasias are a diverse group of neoplasms with different characteristics in terms of site, biological behaviour and metastatic potential. In comparison to other cancers, they are genetically quiet, harbouring relatively few somatic mutations. It is increasingly becoming evident that epigenetic changes are as relevant, if not more so, as somatic mutations in promoting oncogenesis. Despite significant tumour heterogeneity, it is obvious that DNA methylation, histone and chromatin modifications and microRNA expression profiles are distinctive for GEP-NEN subtypes and may correlate with clinical outcome. This review summarises existing knowledge on epigenetic changes, identifying potential contributions to pathogenesis and oncogenesis. In particular, we focus on epigenetic changes pertaining to well-differentiated neuroendocrine tumours, which make up the bulk of NENs. We also highlight both similarities and differences within the subtypes of GEP-NETs and how these relate and compare to other types of cancers. We relate epigenetic understanding to existing treatments and explore how this knowledge may be exploited in the development of novel treatment approaches, such as in theranostics and combining conventional treatment modalities. We consider potential barriers to epigenetic research in GEP-NENs and discuss strategies to optimise research and development of new therapies.
Collapse
Affiliation(s)
- Rohini Sharma
- Department of Surgery and Cancer, Imperial College London, London W12 ONN, UK;
| | - Mark P. Lythgoe
- Department of Surgery and Cancer, Imperial College London, London W12 ONN, UK;
| | - Bhavandeep Slaich
- Department of Medicine, University of Leicester, Leicester LE1 7RH, UK; (B.S.); (N.P.)
| | - Nishil Patel
- Department of Medicine, University of Leicester, Leicester LE1 7RH, UK; (B.S.); (N.P.)
| |
Collapse
|
12
|
Liu P, Liu JP, Sun SJ, Gao Y, Ai Y, Chen X, Sun Y, Zhou M, Liu Y, Xiong Y, Yuan HX. CBFB-MYH11 Fusion Sequesters RUNX1 in Cytoplasm to Prevent DNMT3A Recruitment to Target Genes in AML. Front Cell Dev Biol 2021; 9:675424. [PMID: 34336831 PMCID: PMC8321512 DOI: 10.3389/fcell.2021.675424] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/16/2021] [Indexed: 12/11/2022] Open
Abstract
A growing number of human diseases have been found to be associated with aberrant DNA methylation, including cancer. Mutations targeting genes encoding DNA methyltransferase (DNMT), TET family of DNA demethylases, and isocitrate dehydrogenase (IDH1, IDH2) that produce TET inhibitory metabolite, 2-hyoxyglutarate (2-HG), are found in more than half of acute myeloid leukemia (AML). To gain new insights into the regulation of DNA de/methylation and consequence of its alteration in cancer development, we searched for genes which are mutated in a manner that is linked with gene mutations involved in DNA de/methylation in multiple cancer types. We found that recurrent CBFB-MYH11 fusions, which result in the expression of fusion protein comprising core-binding factor β (CBFB) and myosin heavy chain 11 (MYH11) and are found in 6∼8% of AML patients, occur mutually exclusively with DNMT3A mutations. Tumors bearing CBFB-MYH11 fusion show DNA hypomethylation patterns similar to those with loss-of-function mutation of DNMT3A. Expression of CBFB-MYH11 fusion or inhibition of DNMT3A similarly impairs the methylation and expression of target genes of Runt related transcription factor 1 (RUNX1), a functional partner of CBFB. We demonstrate that RUNX1 directly interacts with DNMT3A and that CBFB-MYH11 fusion protein sequesters RUNX1 in the cytoplasm, thereby preventing RUNX1 from interacting with and recruiting DNMT3A to its target genes. Our results identify a novel regulation of DNA methylation and provide a molecular basis how CBFB-MYH11 fusion contributes to leukemogenesis.
Collapse
Affiliation(s)
- Peng Liu
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jin-Pin Liu
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Si-Jia Sun
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yun Gao
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yingjie Ai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Xiufei Chen
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yiping Sun
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Mengyu Zhou
- Ministry of Education (MOE) Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yun Liu
- Ministry of Education (MOE) Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yue Xiong
- Cullgen Inc., San Diego, CA, United States
| | - Hai-Xin Yuan
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Timmerman DM, Remmers TL, Hillenius S, Looijenga LHJ. Mechanisms of TP53 Pathway Inactivation in Embryonic and Somatic Cells-Relevance for Understanding (Germ Cell) Tumorigenesis. Int J Mol Sci 2021; 22:ijms22105377. [PMID: 34065345 PMCID: PMC8161298 DOI: 10.3390/ijms22105377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 01/10/2023] Open
Abstract
The P53 pathway is the most important cellular pathway to maintain genomic and cellular integrity, both in embryonic and non-embryonic cells. Stress signals induce its activation, initiating autophagy or cell cycle arrest to enable DNA repair. The persistence of these signals causes either senescence or apoptosis. Over 50% of all solid tumors harbor mutations in TP53 that inactivate the pathway. The remaining cancers are suggested to harbor mutations in genes that regulate the P53 pathway such as its inhibitors Mouse Double Minute 2 and 4 (MDM2 and MDM4, respectively). Many reviews have already been dedicated to P53, MDM2, and MDM4, while this review additionally focuses on the other factors that can deregulate P53 signaling. We discuss that P14ARF (ARF) functions as a negative regulator of MDM2, explaining the frequent loss of ARF detected in cancers. The long non-coding RNA Antisense Non-coding RNA in the INK4 Locus (ANRIL) is encoded on the same locus as ARF, inhibiting ARF expression, thus contributing to the process of tumorigenesis. Mutations in tripartite motif (TRIM) proteins deregulate P53 signaling through their ubiquitin ligase activity. Several microRNAs (miRNAs) inactivate the P53 pathway through inhibition of translation. CCCTC-binding factor (CTCF) maintains an open chromatin structure at the TP53 locus, explaining its inactivation of CTCF during tumorigenesis. P21, a downstream effector of P53, has been found to be deregulated in different tumor types. This review provides a comprehensive overview of these factors that are known to deregulate the P53 pathway in both somatic and embryonic cells, as well as their malignant counterparts (i.e., somatic and germ cell tumors). It provides insights into which aspects still need to be unraveled to grasp their contribution to tumorigenesis, putatively leading to novel targets for effective cancer therapies.
Collapse
|
14
|
Swellam M, Saad EA, Sabry S, Denewer A, Abdel Malak C, Abouzid A. Alterations of PTEN and SMAD4 methylation in diagnosis of breast cancer: implications of methyl II PCR assay. J Genet Eng Biotechnol 2021; 19:54. [PMID: 33825073 PMCID: PMC8024427 DOI: 10.1186/s43141-021-00154-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/26/2021] [Indexed: 12/22/2022]
Abstract
Background Diagnosis of breast cancer is more complicated due to lack of minimal invasive biomarker with sufficient precision. DNA methylation is a promising marker for cancer diagnosis. In this study, authors evaluated methylation patterns for PTEN and SMAD4 in blood samples using EpiTect Methyl II QPCR assay quantitative PCR technology. Results Methylation status for PTEN and SMAD4 were statistically significant as breast cancer patients reported hypermethylation compared to benign and control groups (77.1 ± 17.9 vs. 24.9 ± 4.5 and 15.1 ± 1.4 and 70.1 ± 14.4 vs. 28.2 ± 0.61 and 29.5 ± 3.6, respectively). ROC curve analysis revealed that both PTEN (AUC = 0.992) and SMAD4 (AUC = 0.853) had good discriminative power for differentiating BC from all non-cancer individuals (benign and healthy combined) compared to routine tumor markers CEA (AUC = 0.538) and CA15.3 (AUC = 0.686). High PTEN methylation degree was associated with late stages (84.2 ± 17.4), positive lymph node (84.2 ± 18.5), positive ER (81.3 ± 19.7), positive PgR (79.5 ± 19.1), and positive HER2 (80.7 ± 19.0) vs. 67.4 ± 13.8, 70.6 ± 14.8, 72.8 ± 14.9, 72.5 ± 14.7, and 70.2 ± 13.5 in early stages, negative lymph node, negative ER, negative PgR, and negative HER2, respectively. Similar results were obtained regarding SMAD4 methylation. Sensitivity, specificity, positive and negative predictive values, and accuracy for methylated PTEN were 100%, 95%, 99.1%, 100%, and 95%, respectively when differentiated BC from all-non cancer controls. Interestingly, PTEN could distinguish early BC stages with good sensitivity 84.4%, 51.4%, 69.1%, 72%, and 70%, respectively. Conclusion Methylation status of PTEN and SMAD4 is a promising blood marker for early detection of breast cancer. Future studies are needed for their role as prognostic markers.
Collapse
Affiliation(s)
- Menha Swellam
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division, High Throughput Molecular and Genetic Laboratory, Center for Excellences for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| | - Entsar A Saad
- Chemistry Department, Faculty of Science, Damietta University, Damietta, 34517, Egypt
| | - Shimaa Sabry
- Chemistry Department, Faculty of Science, Damietta University, Damietta, 34517, Egypt.
| | - Adel Denewer
- Surgical Oncology Department, Mansoura Oncology Centre, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Camelia Abdel Malak
- Chemistry Department, Faculty of Science, Damietta University, Damietta, 34517, Egypt
| | - Amr Abouzid
- Surgical Oncology Department, Mansoura Oncology Centre, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
15
|
Westrich JA, Vermeer DW, Colbert PL, Spanos WC, Pyeon D. The multifarious roles of the chemokine CXCL14 in cancer progression and immune responses. Mol Carcinog 2020; 59:794-806. [PMID: 32212206 DOI: 10.1002/mc.23188] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022]
Abstract
The chemokine CXCL14 is a highly conserved, homeostatic chemokine that is constitutively expressed in skin epithelia. Responsible for immune cell recruitment and maturation, as well as impacting epithelial cell motility, CXCL14 contributes to the establishment of immune surveillance within normal epithelial layers. Furthermore, CXCL14 is critical to upregulating major histocompatibility complex class I expression on tumor cells. Given these important roles, CXCL14 is often dysregulated in several types of carcinomas including cervical, colorectal, endometrial, and head and neck cancers. Its disruption has been shown to limit critical antitumor immune regulation and is correlated to poor patient prognosis. However, other studies have found that in certain cancers, namely pancreatic and some breast cancers, overexpression of stromal CXCL14 correlates with poor patient survival due to increased invasiveness. Contributing to the ambiguity CXCL14 plays in cancer is that the native CXCL14 receptor remains uncharacterized, although several candidate receptors have been proposed. Despite the complexity of CXCL14 functions, it remains clear that this chemokine is a key regulatory factor in cancer and represents a potential target for future cancer immunotherapies.
Collapse
Affiliation(s)
- Joseph A Westrich
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel W Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota
| | - Paul L Colbert
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota
| | - William C Spanos
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota
| | - Dohun Pyeon
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan
| |
Collapse
|
16
|
Colao A, de Nigris F, Modica R, Napoli C. Clinical Epigenetics of Neuroendocrine Tumors: The Road Ahead. Front Endocrinol (Lausanne) 2020; 11:604341. [PMID: 33384663 PMCID: PMC7770585 DOI: 10.3389/fendo.2020.604341] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/12/2020] [Indexed: 12/16/2022] Open
Abstract
Neuroendocrine tumors, or NETs, are cancer originating in neuroendocrine cells. They are mostly found in the gastrointestinal tract or lungs. Functional NETs are characterized by signs and symptoms caused by the oversecretion of hormones and other substances, but most NETs are non-functioning and diagnosis in advanced stages is common. Thus, novel diagnostic and therapeutic strategies are warranted. Epigenetics may contribute to refining the diagnosis, as well as to identify targeted therapy interfering with epigenetic-sensitive pathways. The goal of this review was to discuss the recent advancement in the epigenetic characterization of NETs highlighting their role in clinical findings.
Collapse
Affiliation(s)
- Annamaria Colao
- Department of Clinical Medicine and Surgery, Unesco Chair Health Education and Sustainable Development, Federico II University of Naples, Naples, Italy
| | - Filomena de Nigris
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Roberta Modica
- Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
- *Correspondence: Roberta Modica,
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
17
|
Nakagawa T, Wakui M, Hayashida T, Nishime C, Murata M. Intensive optimization and evaluation of global DNA methylation quantification using LC-MS/MS. Anal Bioanal Chem 2019; 411:7221-7231. [DOI: 10.1007/s00216-019-02115-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/09/2019] [Accepted: 08/26/2019] [Indexed: 01/22/2023]
|
18
|
Boons G, Vandamme T, Peeters M, Van Camp G, Op de Beeck K. Clinical applications of (epi)genetics in gastroenteropancreatic neuroendocrine neoplasms: Moving towards liquid biopsies. Rev Endocr Metab Disord 2019; 20:333-351. [PMID: 31368038 DOI: 10.1007/s11154-019-09508-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
High-throughput analysis, including next-generation sequencing and microarrays, have strongly improved our understanding of cancer biology. However, genomic data on rare cancer types, such as neuroendocrine neoplasms, has been lagging behind. Neuroendocrine neoplasms (NENs) develop from endocrine cells spread throughout the body and are highly heterogeneous in biological behavior. In this challenging disease, there is an urgent need for new therapies and new diagnostic, prognostic, follow-up and predictive biomarkers to aid patient management. The last decade, molecular data on neuroendocrine neoplasms of the gastrointestinal tract and pancreas, termed gastroenteropancreatic NENs (GEP-NENs), has strongly expanded. The aim of this review is to give an overview of the recent advances on (epi)genetic level and highlight their clinical applications to address the current needs in GEP-NENs. We illustrate how molecular alterations can be and are being used as therapeutic targets, how mutations in DAXX/ATRX and copy number variations could be used as prognostic biomarkers, how far we are in identifying predictive biomarkers and how genetics can contribute to GEP-NEN classification. Finally, we discuss recent studies on liquid biopsies in the field of GEP-NENs and illustrate how liquid biopsies can play a role in patient management. In conclusion, molecular studies have suggested multiple potential biomarkers, but further validation is ongoing.
Collapse
Affiliation(s)
- Gitta Boons
- Center for Oncological Research (CORE), University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650, Edegem, Belgium
| | - Timon Vandamme
- Center for Oncological Research (CORE), University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650, Edegem, Belgium
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Dr. Molewaterplein 50, 3015GE, Rotterdam, The Netherlands
| | - Marc Peeters
- Center for Oncological Research (CORE), University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Guy Van Camp
- Center for Oncological Research (CORE), University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610, Wilrijk, Belgium.
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650, Edegem, Belgium.
| | - Ken Op de Beeck
- Center for Oncological Research (CORE), University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650, Edegem, Belgium
| |
Collapse
|
19
|
Barazeghi E, Hellman P, Westin G, Stålberg P. PTPRM, a candidate tumor suppressor gene in small intestinal neuroendocrine tumors. Endocr Connect 2019; 8:1126-1135. [PMID: 31349215 PMCID: PMC6687034 DOI: 10.1530/ec-19-0279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 07/09/2019] [Indexed: 12/27/2022]
Abstract
Small intestinal neuroendocrine tumors (SI-NETs) are small, slow growing neoplasms with loss of one copy of chromosome 18 as a common event. Frequently mutated genes on chromosome 18 or elsewhere have not been found so far. The aim of this study was to investigate a possible tumor suppressor role of the transmembrane receptor type tyrosine phosphatase PTPµ (PTPRM at 18p11) in SI-NETs. Immunohistochemistry, quantitative RT-PCR, colony formation assay and quantitative CpG methylation analysis by pyrosequencing were performed. Undetectable/very low levels of PTPRM or aberrant pattern of immunostaining, with both negative and positive areas, were detected in the majority of tumors (33/40), and a significantly reduced mRNA expression in metastases compared to primary tumors was observed. Both the DNA methylation inhibitor 5-aza-2'-deoxycytidine and the S-adenosylhomocysteine hydrolase inhibitor 3-deazaneplanocin A (DZNep) induced PTPRM expression in CNDT2.5 and KRJ-I SI-NET cells. CpG methylation of upstream regulatory regions, the promoter region and the exon 1/intron 1 boundary was detected by pyrosequencing analysis of the two cell lines and not in the analyzed SI-NETs. Overexpression of PTPRM in the SI-NET cell lines reduced cell growth and cell proliferation and induced apoptosis. The tyrosine phosphatase activity of PTPRM was not involved in cell growth inhibition. The results support a role for PTPRM as a dysregulated candidate tumor suppressor gene in SI-NETs and further analyses of the involved mechanisms are warranted.
Collapse
Affiliation(s)
- Elham Barazeghi
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Rudbeck Laboratory, Uppsala, Sweden
- Correspondence should be addressed to E Barazeghi or P Stålberg: or
| | - Per Hellman
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Rudbeck Laboratory, Uppsala, Sweden
| | - Gunnar Westin
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Rudbeck Laboratory, Uppsala, Sweden
| | - Peter Stålberg
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Rudbeck Laboratory, Uppsala, Sweden
- Correspondence should be addressed to E Barazeghi or P Stålberg: or
| |
Collapse
|
20
|
Fotouhi O, Ghaderi M, Wang N, Zedenius J, Kjellman M, Xu D, Juhlin CC, Larsson C. Telomerase activation in small intestinal neuroendocrine tumours is associated with aberrant TERT promoter methylation, but not hot-spot mutations. Epigenetics 2019; 14:1224-1233. [PMID: 31322481 DOI: 10.1080/15592294.2019.1634987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Telomere maintenance is a critical requirement for enabling replicative immortality and tumour development. Here, telomerase expression and activity, telomere length (TL) and potential regulatory factors that can underlie telomerase machinery alterations in small intestinal neuroendocrine tumours (SI-NETs) were analyzed. Telomerase activity assessed by TRAP assay was increased in SI-NETs compared to normal ileum (P < 0.001). The telomerase reverse transcriptase gene (TERT) was over-expressed in SI-NETs vs. normal ileal samples (P = 0.01). Furthermore, relative TL assessed by qPCR was found shorter in tumours compared with normal ileum (P = 0.02) and in distant metastasis samples compared to primary tumours and local metastases (P= 0.02). TERT promoter hotspot mutations were not present and TERT copy number gain was only observed in 3/70 tumour samples. TERT or chromosome 18 copy number alterations were not associated with telomerase expression and activity or TL. However, hypermethylation of TERT promoter in Region B - in the proximity of the transcription start site - was inversely correlated with TERT expression and telomerase activity and positively correlated with TL. Global LINE1 methylation was positively correlated with TERT promoter Region B methylation and was inversely correlated with telomerase activity, TERT expression and the upstream Region A methylation. The results show that telomerase activation, TERT expression and shorter telomeres are commonly found in SI-NETs. Aberrant DNA methylation of TERT promoter and of LINE1 can be implicated in abnormal regulation of TERT in SI-NETs.
Collapse
Affiliation(s)
- Omid Fotouhi
- Department of Oncology-Pathology, Karolinska Institutet , Stockholm , Sweden
| | - Mehran Ghaderi
- Department of Oncology-Pathology, Karolinska Institutet , Stockholm , Sweden
| | - Na Wang
- Department of Oncology-Pathology, Karolinska Institutet , Stockholm , Sweden.,Cancer Center Karolinska,CCK
| | - Jan Zedenius
- Department of Molecular Medicine and Surgery, Karolinska Institutet , Stockholm , Sweden.,Department of Breast, Endocrine Tumours and Sarcoma, Karolinska University Hospital , Stockholm , Sweden
| | - Magnus Kjellman
- Department of Molecular Medicine and Surgery, Karolinska Institutet , Stockholm , Sweden.,Department of Breast, Endocrine Tumours and Sarcoma, Karolinska University Hospital , Stockholm , Sweden
| | - Dawei Xu
- Department of Medicine, Division of Hematology (D.X.), Karolinska University Hospital , Stockholm , Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet , Stockholm , Sweden.,Cancer Center Karolinska,CCK
| | - Catharina Larsson
- Department of Oncology-Pathology, Karolinska Institutet , Stockholm , Sweden.,Cancer Center Karolinska,CCK
| |
Collapse
|
21
|
Samsom KG, van Veenendaal LM, Valk GD, Vriens MR, Tesselaar MET, van den Berg JG. Molecular prognostic factors in small-intestinal neuroendocrine tumours. Endocr Connect 2019; 8:906-922. [PMID: 31189127 PMCID: PMC6599083 DOI: 10.1530/ec-19-0206] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/10/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Small-intestinal neuroendocrine tumours (SI-NETs) represent a heterogeneous group of rare tumours. In recent years, basic research in SI-NETs has attempted to unravel the molecular events underlying SI-NET tumorigenesis. AIM We aim to provide an overview of the current literature regarding prognostic and predictive molecular factors in patients with SI-NETs. METHOD A PubMed search was conducted on (epi)genetic prognostic factors in SI-NETs from 2000 until 2019. RESULTS The search yielded 1522 articles of which 20 reviews and 35 original studies were selected for further evaluation. SI-NETs are mutationally quiet tumours with a different genetic make-up compared to pancreatic NETs. Loss of heterozygosity at chromosome 18 is the most frequent genomic aberration (44-100%) followed by mutations of CDKN1B in 8%. Prognostic analyses were performed in 16 studies, of which 8 found a significant (epi)genetic association for survival or progression. Loss of heterozygosity at chromosome 18, gains of chromosome 4, 5, 7, 14 and 20p, copy gain of the SRC gene and low expression of RASSF1A and P16 were associated with poorer survival. In comparison with genetic mutations, epigenetic alterations are significantly more common in SI-NETs and may represent more promising targets in the treatment of SI-NETs. CONCLUSION SI-NETs are mutationally silent tumours. No biomarkers have been identified yet that can easily be adopted into current clinical decision making. SI-NETs may represent a heterogeneous disease and larger international studies are warranted to translate molecular findings into precision oncology.
Collapse
Affiliation(s)
- K G Samsom
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - L M van Veenendaal
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - G D Valk
- Department of Endocrine Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Correspondence should be addressed to G D Valk:
| | - M R Vriens
- Department of Surgical Oncology and Endocrine Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - M E T Tesselaar
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J G van den Berg
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Scarpa A. The landscape of molecular alterations in pancreatic and small intestinal neuroendocrine tumours. ANNALES D'ENDOCRINOLOGIE 2019; 80:153-158. [PMID: 31072588 DOI: 10.1016/j.ando.2019.04.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastroenteropancreatic Neuroendocrine Neoplasms (GEP-NENs) arise throughout the gut and feature varying biological behaviour and malignant potential. GEP-NENs include two genetically different entities, well-differentiated neuroendocrine tumours (NETs) and poorly differentiated neuroendocrine carcinomas (NEC). NECs are characterized by a dismal prognosis and by distinctive TP53 and RB1 inactivation which sets them apart from NETs. The latter, conversely, have a wide spectrum of aggressiveness and molecular alterations. Knowledge on their biology has recently expanded thanks to high-throughput studies focused on two important groups of well-differentiated neuroendocrine neoplasms: pancreatic (PanNETs) and small intestinal (SiNETs) tumours. PanNETs have been among the most studied also due to genetic syndromes featuring their onset. Research stemming from this observation has uncovered the inactivation of MEN1, VHL, TSC1/2, and the hyperactivation of the PI3K/mTOR pathway as distinctive biological features of these neoplasms. Next-Generation Sequencing added information on the role of telomere lengthening via ATRX/DAXX inactivation in a fraction of PanNETs, while other display shortened telomeres and recurrent chromosomal alterations. The data so far disclosed a heterogeneous combination of driver events, yet converging into four pathways including DNA damage repair, cell cycle regulation, PI3K/mTOR signalling and telomere maintenance. SiNETs showed a lesser relationship with mutational driver events, even in the case of familial cases. High throughput studies identified putative driver mutations in CDKN1 and APC which, however, were reported in a minor fraction (∼10%) of cases. Tumorigenesis of SiNETs seems to depend more on chromosomal alterations (loss of chromosome 8, gains at 4, 5 and 20) and epigenetic events, which converge to hyperactivate the PI3K/mTOR, MAPK and Wnt pathways. While calling for further integrative studies, these data lay previous and recent findings in a more defined frame and provide clinical research with several candidate markers for patient stratification and companion diagnostics.
Collapse
Affiliation(s)
- Aldo Scarpa
- RC-Net Centre for applied research on cancer, University and Hospital Trust of Verona, 37134 Verona, Italy; Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy.
| |
Collapse
|
23
|
Kerekes K, Bányai L, Trexler M, Patthy L. Structure, function and disease relevance of Wnt inhibitory factor 1, a secreted protein controlling the Wnt and hedgehog pathways. Growth Factors 2019; 37:29-52. [PMID: 31210071 DOI: 10.1080/08977194.2019.1626380] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Wnts and Hedgehogs (Hh) are large, lipid-modified extracellular morphogens that play key roles in embryonic development and stem cell proliferation of Metazoa. Both morphogens signal through heptahelical Frizzled-type receptors of the G-Protein Coupled Receptor family and there are several other similarities that suggest a common evolutionary origin of the Hh and Wnt pathways. There is evidence that the secreted protein, Wnt inhibitory factor 1 (WIF1) modulates the activity of both Wnts and Hhs and may thus contribute to the intertwining of these pathways. In this article, we review the structure, evolution, molecular interactions and functions of WIF1 with major emphasis on its role in carcinogenesis.
Collapse
Affiliation(s)
- Krisztina Kerekes
- a Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - László Bányai
- a Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - Mária Trexler
- a Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - László Patthy
- a Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| |
Collapse
|
24
|
Mafficini A, Scarpa A. Genetics and Epigenetics of Gastroenteropancreatic Neuroendocrine Neoplasms. Endocr Rev 2019; 40:506-536. [PMID: 30657883 PMCID: PMC6534496 DOI: 10.1210/er.2018-00160] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 12/27/2018] [Indexed: 12/11/2022]
Abstract
Gastroenteropancreatic (GEP) neuroendocrine neoplasms (NENs) are heterogeneous regarding site of origin, biological behavior, and malignant potential. There has been a rapid increase in data publication during the last 10 years, mainly driven by high-throughput studies on pancreatic and small intestinal neuroendocrine tumors (NETs). This review summarizes the present knowledge on genetic and epigenetic alterations. We integrated the available information from each compartment to give a pathway-based overview. This provided a summary of the critical alterations sustaining neoplastic cells. It also highlighted similarities and differences across anatomical locations and points that need further investigation. GEP-NENs include well-differentiated NETs and poorly differentiated neuroendocrine carcinomas (NECs). NENs are graded as G1, G2, or G3 based on mitotic count and/or Ki-67 labeling index, NECs are G3 by definition. The distinction between NETs and NECs is also linked to their genetic background, as TP53 and RB1 inactivation in NECs set them apart from NETs. A large number of genetic and epigenetic alterations have been reported. Recurrent changes have been traced back to a reduced number of core pathways, including DNA damage repair, cell cycle regulation, and phosphatidylinositol 3-kinase/mammalian target of rapamycin signaling. In pancreatic tumors, chromatin remodeling/histone methylation and telomere alteration are also affected. However, also owing to the paucity of disease models, further research is necessary to fully integrate and functionalize data on deregulated pathways to recapitulate the large heterogeneity of behaviors displayed by these tumors. This is expected to impact diagnostics, prognostic stratification, and planning of personalized therapy.
Collapse
Affiliation(s)
- Andrea Mafficini
- ARC-Net Center for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, Italy.,Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- ARC-Net Center for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, Italy.,Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| |
Collapse
|
25
|
Sahnane N, Ottini G, Turri-Zanoni M, Furlan D, Battaglia P, Karligkiotis A, Albeni C, Cerutti R, Mura E, Chiaravalli AM, Castelnuovo P, Sessa F, Facco C. Comprehensive analysis of HPV infection, EGFR exon 20 mutations and LINE1 hypomethylation as risk factors for malignant transformation of sinonasal-inverted papilloma to squamous cell carcinoma. Int J Cancer 2018; 144:1313-1320. [PMID: 30411788 DOI: 10.1002/ijc.31971] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/10/2018] [Accepted: 10/22/2018] [Indexed: 12/11/2022]
Abstract
Different risk factors are suspected to be involved in malignant transformation of sinonasal papillomas and include HPV infection, tobacco smoking, occupational exposure, EGFR/KRAS mutations and DNA methylation alterations. In our study, 25 inverted sinonasal papillomas (ISPs), 5 oncocytic sinonasal papillomas (OSP) and 35 squamous cell carcinomas (SCCs) from 54 patients were genotyped for 10 genes involved in EGFR signalling. HPV-DNA detection was performed by in-situ hybridisation and LINE-1 methylation was quantitatively determined by bisulphite-pyrosequencing. High-risk HPV was observed only in 13% of ISP-associated SCC and in 8% of de novo-SCC patients. EGFR mutations occurred in 72% of ISPs, 30% of ISP-associated SCCs and 17% of de novo-SCCs. At 5-year follow-up, SCC arose in only 30% (6/20) of patients with EGFR-mutated ISPs compared to 76% (13/17) of patients with EGFR-wild-type ISP (p = 0.0044). LINE-1 hypomethylation significantly increased from papilloma/early stage SCC to advanced stage SCC (p = 0.03) and was associated with occupational exposure (p = 0.01) and worse prognosis (p = 0.09). In conclusion, our results suggest that a small subset of these tumours could be related to HPV infection; EGFR mutations characterise those ISPs with a lower risk of developing into SCC; LINE-1 hypomethylation is associated with occupational exposure and could identify more aggressive nasal SCC.
Collapse
Affiliation(s)
- Nora Sahnane
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Giorgia Ottini
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Mario Turri-Zanoni
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Daniela Furlan
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Paolo Battaglia
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Apostolos Karligkiotis
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Chiara Albeni
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Roberta Cerutti
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Eleonora Mura
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Anna Maria Chiaravalli
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Paolo Castelnuovo
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Fausto Sessa
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria and ASST Sette-Laghi, Varese, Italy
| | - Carla Facco
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria and ASST Sette-Laghi, Varese, Italy
| |
Collapse
|
26
|
Arvidsson Y, Rehammar A, Bergström A, Andersson E, Altiparmak G, Swärd C, Wängberg B, Kristiansson E, Nilsson O. miRNA profiling of small intestinal neuroendocrine tumors defines novel molecular subtypes and identifies miR-375 as a biomarker of patient survival. Mod Pathol 2018; 31:1302-1317. [PMID: 29487354 DOI: 10.1038/s41379-018-0010-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 12/08/2017] [Accepted: 12/08/2017] [Indexed: 12/30/2022]
Abstract
The aim of this study was to define the miRNA profile of small intestinal neuroendocrine tumors and to search for novel molecular subgroups and prognostic biomarkers. miRNA profiling was conducted on 42 tumors from 37 patients who underwent surgery for small intestinal neuroendocrine tumors. Unsupervised hierarchical clustering analysis of miRNA profiles identified two groups of tumor metastases, denoted cluster M1 and M2. The smaller cluster M1 was associated with shorter overall survival and contained tumors with higher grade (WHO grade G2/3) and multiple chromosomal gains including gain of chromosome 14. Tumors of cluster M1 had elevated expression of miR-1246 and miR-663a, and reduced levels of miR-488-3p. Pathway analysis predicted Wnt signaling to be the most significantly altered signaling pathway between clusters M1 and M2. Analysis of miRNA expression in relation to tumor proliferation rate showed significant alterations including downregulation of miR-137 and miR-204-5p in tumors with Ki67 index above 3%. Similarly, tumor progression was associated with significant alterations in miRNA expression, e.g. higher expression of miR-95 and miR-210, and lower expression of miR-378a-3p in metastases. Pathway analysis predicted Wnt signaling to be altered during tumor progression, which was supported by decreased nuclear translocation of β-catenin in metastases. Survival analysis revealed that downregulation of miR-375 was associated with shorter overall survival. We performed in situ hybridization on biopsies from an independent cohort of small intestinal neuroendocrine tumors using tissue microarrays. Expression of miR-375 was found in 578/635 (91%) biopsies and survival analysis confirmed that there was a correlation between downregulation of miR-375 in tumor metastases and shorter patient survival. We conclude that miRNA profiling defines novel molecular subgroups of metastatic small intestinal neuroendocrine tumors and identifies miRNAs associated with tumor proliferation rate and progression. miR-375 is highly expressed in small intestinal neuroendocrine tumors and may be used as a prognostic biomarker.
Collapse
Affiliation(s)
- Yvonne Arvidsson
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Anna Rehammar
- Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Anders Bergström
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ellinor Andersson
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Gülay Altiparmak
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Christina Swärd
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Bo Wängberg
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Kristiansson
- Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Ola Nilsson
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
27
|
Barazeghi E, Prabhawa S, Norlén O, Hellman P, Stålberg P, Westin G. Decrease of 5-hydroxymethylcytosine and TET1 with nuclear exclusion of TET2 in small intestinal neuroendocrine tumors. BMC Cancer 2018; 18:764. [PMID: 30045709 PMCID: PMC6060499 DOI: 10.1186/s12885-018-4579-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 06/07/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Small intestinal neuroendocrine tumors (SI-NETs) originate from enterochromaffin cells scattered in the intestinal mucosa of the ileum and jejunum. Loss of one copy of chromosome 18 is the most frequent observed aberration in primary tumors and metastases. The aim of this study was to investigate possible involvement of 5-hydroxymethylcytosine (5hmC), TET1 and TET2 in SI-NETs. METHODS The analysis was conducted using 40 primary tumors and corresponding 47 metastases. The level of 5hmC, TET1 and TET2 was analyzed by DNA immune-dot blot assay and immunohistochemistry. Other methods included a colony forming assay, western blotting analysis, and quantitative bisulfite pyrosequencing analysis. The effect of the exportin-1 nuclear transport machinery inhibitors on cell proliferation and apoptosis was also explored using two SI-NET cell lines. RESULTS Variable levels of 5hmC and a mosaic staining appearance with a mixture of positive and negative cell nuclei, regardless of cell number and staining strength, was observed overall both in primary tumors and metastases. Similarly aberrant staining pattern was observed for TET1 and TET2. In a number of tumors (15/32) mosaic pattern together with areas of negative staining was also observed for TET1. Abolished expression of TET1 in the tumors did not seem to involve hypermethylation of the TET1 promoter region. Overexpression of TET1 in a colony forming assay supported a function as cell growth regulator. In contrast to 5hmC and TET1, TET2 was also observed in the cytoplasm of all the analyzed SI-NETs regardless of nuclear localization. Treatment of CNDT2.5 and KRJ-I cells with the exportin-1 (XPO1/CRM1) inhibitor, leptomycin B, induced reduction in the cytoplasm and nuclear retention of TET2. Aberrant partitioning of TET2 from the nucleus to the cytoplasm seemed therefore to involve the exportin-1 nuclear transport machinery. Reduced cell proliferation and induction of apoptosis were observed after treatment of CNDT2.5 and KRJ-I cells with leptomycin B or KPT-330 (selinexor). CONCLUSIONS SI-NETs are epigenetically dysregulated at the level of 5-hydroxymethylcytosine/ TET1/TET2. We suggest that KPT-330/selinexor or future developments should be considered and evaluated for single treatment of patients with SI-NET disease and also in combinations with somatostatin analogues, peptide receptor radiotherapy, or everolimus.
Collapse
Affiliation(s)
- Elham Barazeghi
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden
| | - Surendra Prabhawa
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden
| | - Olov Norlén
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden
| | - Per Hellman
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden
| | - Peter Stålberg
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden.
| | - Gunnar Westin
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
28
|
Hernandez-Cortes D, Alvarado-Cruz I, Solís-Heredia MJ, Quintanilla-Vega B. Epigenetic modulation of Nrf2 and Ogg1 gene expression in testicular germ cells by methyl parathion exposure. Toxicol Appl Pharmacol 2018. [PMID: 29540303 DOI: 10.1016/j.taap.2018.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Methyl parathion (Me-Pa) is an oxidizing organophosphate (OP) pesticide that generates reactive oxygen species (ROS) through its biotransformation. Some studies have also suggested that OP pesticides have the capacity to alkylate biomolecules, including DNA. In general, DNA methylation in gene promoters represses transcription. NRF2 is a key transcription factor that regulates the expression of antioxidant, metabolic and detoxifying genes through the antioxidant response element (ARE) situated in promoters of regulated genes. Furthermore, DNA repair genes, including 8-oxoguanine DNA glycosidase (OGG1), have been proposed as NRF2 target genes. Me-Pa exposure produces poor semen quality, genetic and oxidative damage in sperm cells, and reduced fertility. However, the Me-Pa effects on the methylation status and the expression of antioxidant (Nrf2) or DNA repair (Ogg1) genes in male germ cells have not been investigated. Therefore, mice were exposed to Me-Pa to evaluate the global (%5-mC) and specific methylation of Nrf2 and Ogg1 genes using pyrosequencing, gene expression, and total protein carbonylation in male germ cells. The results showed that Me-Pa significantly decreased the global DNA methylation pattern and significantly increased the methylation of two CpG sites within Ogg1 promoter and one CpG site within Nrf2 promoter. In addition, Ogg1 or Nrf2 expression did not change after Me-Pa exposure despite the oxidative damage produced. Altogether, our data suggest that Me-Pa toxicity alters Ogg1 and Nrf2 promoter methylation in male germ cells that may be modulating their gene expression.
Collapse
Affiliation(s)
| | - I Alvarado-Cruz
- Department of Toxicology, Cinvestav, Mexico City 07360, Mexico
| | | | | |
Collapse
|
29
|
Genetic and epigenetic background and protein expression profiles in relation to telomerase activation in medullary thyroid carcinoma. Oncotarget 2018; 7:21332-46. [PMID: 26870890 PMCID: PMC5008288 DOI: 10.18632/oncotarget.7237] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 01/17/2016] [Indexed: 01/08/2023] Open
Abstract
Medullary thyroid carcinomas (MTCs) exhibit telomerase activation in strong association with shorter patient survival. To understand the background of telomerase activation we quantified TERT copy numbers and TERT promoter methylation in 42 MTCs and normal thyroid references. Gain of TERT was demonstrated by quantitative PCR in 5/39 sporadic MTC. Increased methylation index (MetI) for CpG methylation at the TERT promoter was found in sporadic MTCs (P < 0.0001) and in MEN 2 associated MTCs (P = 0.011) vs. normal thyroid tissues. MetI correlated positively with TERT gene expression (r = 0.432, P = 0.006) and negatively with telomere length (r = −0.343, P = 0.032). MTC cases with MetI above the median of 52% had shorter survival as compared to cases with lower MetI (P = 0.005 for overall survival and P = 0.007 for disease-related survival). Protein expression profiles obtained by mass spectrometry were then studied in relation to telomerase activation in MTCs. Comparing protein levels between tumors defined by telomerase activity status, 240 proteins were associated with telomerase activity. Among telomerase activation positive cases a set of proteins was found to discriminate between MTCs with high and low TERT gene expression with enrichment for proteins involved in telomerase regulation. XRCC5 mRNA expression was found increased in MTCs vs. normal thyroid (P = 0.007). In conclusion the findings suggest a role for TERT copy number gain, TERT promoter methylation and XRCC5 expression in telomerase activation and telomere maintenance of MTC.
Collapse
|
30
|
Finnerty BM, Gray KD, Moore MD, Zarnegar R, Fahey III TJ. Epigenetics of gastroenteropancreatic neuroendocrine tumors: A clinicopathologic perspective. World J Gastrointest Oncol 2017; 9:341-353. [PMID: 28979716 PMCID: PMC5605334 DOI: 10.4251/wjgo.v9.i9.341] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 06/27/2017] [Accepted: 08/04/2017] [Indexed: 02/05/2023] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are a heterogeneous group of rare tumors whose site-specific tumor incidence and clinical behavior vary widely. Genetic alterations associated with familial inherited syndromes have been well defined; however, the genetic profile of sporadic tumors is less clear as their tumorigenesis does not appear to be controlled by classic oncogenes such as P53, RB, or KRAS. Even within GEP-NETs, there are no common oncogenic drivers; for example, DAXX/ATRX mutations are strongly implicated in the tumorigenesis of pancreatic but not small bowel NETs. Accordingly, the dysregulation of epigenetic mechanisms has been hypothesized as a potential regulator of GEP-NET tumorigenesis and has become a major focus of recent studies. Despite the heterogeneity of tumor cohorts evaluated in these studies, it is obvious that there are methylation patterns, chromatin remodeling alterations, and microRNA and long non-coding RNA (lncRNA) differential expression profiles that are distinctive of GEP-NETs, some of which are correlated with significant differences in clinical outcomes. Several translational studies have provided convincing data identifying potential prognostic biomarkers, and some of these have demonstrated preliminary success as serum biomarkers that can be used clinically. Nevertheless, there are many opportunities to further define the mechanisms by which these epigenetic modifications influence tumorigenesis, and this will provide better insight into their prognostic and therapeutic utility. Furthermore, these findings form the foundation for future studies evaluating the clinical efficacy of epigenetic modifications as prognostic biomarkers, as well as potential therapeutic targets.
Collapse
Affiliation(s)
- Brendan M Finnerty
- Department of Surgery, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, United States
| | - Katherine D Gray
- Department of Surgery, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, United States
| | - Maureen D Moore
- Department of Surgery, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, United States
| | - Rasa Zarnegar
- Department of Surgery, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, United States
| | - Thomas J Fahey III
- Department of Surgery, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, United States
| |
Collapse
|
31
|
Ren Y, Xiao L, Weng G, Shi B. Clinical significance of p16INK4A and p14ARF promoter methylation in renal cell carcinoma: a meta-analysis. Oncotarget 2017; 8:64385-64394. [PMID: 28969078 PMCID: PMC5610010 DOI: 10.18632/oncotarget.18826] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 06/02/2017] [Indexed: 11/25/2022] Open
Abstract
The inactivation of p16INK4A and p14ARF via promoter methylation has been investigated in various cancers. However, the clinical effects of p16INK4A and p14ARF promoter methylation on renal cell carcinoma (RCC) remain to be clarified. The pooled data were calculated and summarized. Finally, an investigation of 14 eligible studies with 1231 RCC patients and 689 control patients was performed. Methylated p16INK4A and p14ARF were observed to be significantly higher in RCC than in control subjects without malignancies (OR = 2.77, P = 0.005; OR = 11.73, P < 0.001, respectively). Methylated p16INK4A was significantly associated with the risk of RCC in the tissue subgroup, but not in the serum and urine subgroups. Methylated p16INK4A was significantly associated with tumor size. We did not find that p16INK4A promoter methylation was associated with sex, tumor grade, lymph node status, and tumor histology. Methylated p14ARF was significantly correlated with sex and tumor histology. Three studies reported that p16INK4A methylation was not significantly correlated with the prognosis of RCC. The results suggested that p16INK4A and p14ARF promoter methylation may be correlated with the carcinogenesis of RCC, and that methylated p14ARF , especially, can be a major susceptibility gene. We also found the different clinicopathological significance of 16INK4A and p14ARF in RCC. Additional studies with sufficient data are essential to further evaluate the clinical features and prognostic effect of p16INK4A and p14ARF promoter methylation in RCC.
Collapse
Affiliation(s)
- Yu Ren
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo 315000, People's Republic of China
| | - Li Xiao
- Department of Urologic Surgery, Chinese PLA General Hospital, The 309th Hospital of China People's Liberation Army, Beijing 100094, People's Republic of China
| | - Guobin Weng
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo 315000, People's Republic of China
| | - Bingyi Shi
- Department of Urologic Surgery, Chinese PLA General Hospital, The 309th Hospital of China People's Liberation Army, Beijing 100094, People's Republic of China
| |
Collapse
|
32
|
Di Domenico A, Wiedmer T, Marinoni I, Perren A. Genetic and epigenetic drivers of neuroendocrine tumours (NET). Endocr Relat Cancer 2017; 24:R315-R334. [PMID: 28710117 DOI: 10.1530/erc-17-0012] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 07/14/2017] [Indexed: 12/13/2022]
Abstract
Neuroendocrine tumours (NET) of the gastrointestinal tract and the lung are a rare and heterogeneous group of tumours. The molecular characterization and the clinical classification of these tumours have been evolving slowly and show differences according to organs of origin. Novel technologies such as next-generation sequencing revealed new molecular aspects of NET over the last years. Notably, whole-exome/genome sequencing (WES/WGS) approaches underlined the very low mutation rate of well-differentiated NET of all organs compared to other malignancies, while the engagement of epigenetic changes in driving NET evolution is emerging. Indeed, mutations in genes encoding for proteins directly involved in chromatin remodelling, such as DAXX and ATRX are a frequent event in NET. Epigenetic changes are reversible and targetable; therefore, an attractive target for treatment. The discovery of the mechanisms underlying the epigenetic changes and the implication on gene and miRNA expression in the different subgroups of NET may represent a crucial change in the diagnosis of this disease, reveal new therapy targets and identify predictive markers. Molecular profiles derived from omics data including DNA mutation, methylation, gene and miRNA expression have already shown promising results in distinguishing clinically and molecularly different subtypes of NET. In this review, we recapitulate the major genetic and epigenetic characteristics of pancreatic, lung and small intestinal NET and the affected pathways. We also discuss potential epigenetic mechanisms leading to NET development.
Collapse
Affiliation(s)
- Annunziata Di Domenico
- Institute of PathologyUniversity of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of Bern, Bern, Switzerland
| | - Tabea Wiedmer
- Institute of PathologyUniversity of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of Bern, Bern, Switzerland
| | | | - Aurel Perren
- Institute of PathologyUniversity of Bern, Bern, Switzerland
| |
Collapse
|
33
|
Wedge E, Hansen JW, Garde C, Asmar F, Tholstrup D, Kristensen SS, Munch-Petersen HD, Ralfkiaer E, Brown P, Grønbaek K, Kristensen LS. Global hypomethylation is an independent prognostic factor in diffuse large B cell lymphoma. Am J Hematol 2017; 92:689-694. [PMID: 28378885 DOI: 10.1002/ajh.24751] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/16/2017] [Accepted: 03/28/2017] [Indexed: 12/17/2022]
Abstract
Global hypomethylation has been linked to disease progression in several cancers, but has not been reported for Diffuse Large B Cell Lymphoma (DLBCL). This study aimed to assess global methylation in DLBCL and describe its prognostic value. Mean LINE1 methylation, a validated surrogate measure for global methylation, was measured in DNA from 67 tumor biopsies. Additionally, cell-free circulating DNA (cfDNA) in plasma samples from 74 patients was tested to assess the feasibility of global hypomethylation as a biomarker in liquid biopsies. LINE1 methylation was assessed using a commercially available kit, based on pyrosequencing of PCR amplified bisulfite-treated DNA. Global hypomethylation was detected in a subset of cases and was associated with poor overall survival in both tumor biopsies (P = .001) and cfDNA (P = .009). It was the strongest risk factor in multivariate analysis in both biopsies (HR: 10.65, CI: 2.03-55.81, P = .005) and cfDNA (HR: 11.87, CI: 2.80-50.20, P = .001), outperforming conventional clinical risk factors. Finally, hierarchical cluster analyses were performed for the cfDNA samples using previously published gene-specific methylation data. This analysis shows that global hypomethylation co-occurs with other epigenetic abnormalities, including DAPK1 promoter hypermethylation. In conclusion, we have shown that global hypomethylation is strongly associated with poor survival in DLBCL both when present in tumor biopsy DNA and when detected in plasma cfDNA, and has potential for clinical application as a prognostic biomarker.
Collapse
Affiliation(s)
- Eileen Wedge
- Department of Haematology; Rigshospitalet; Copenhagen Denmark
| | | | - Christian Garde
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen
| | - Fazila Asmar
- Department of Haematology; Rigshospitalet; Copenhagen Denmark
| | - Dorte Tholstrup
- Department of Haematology; Rigshospitalet; Copenhagen Denmark
| | | | - Helga D. Munch-Petersen
- Department of Haematology; Rigshospitalet; Copenhagen Denmark
- Department of Pathology; Rigshospitalet; Copenhagen Denmark
| | | | - Peter Brown
- Department of Haematology; Rigshospitalet; Copenhagen Denmark
| | | | - Lasse Sommer Kristensen
- Department of Haematology; Rigshospitalet; Copenhagen Denmark
- Department of Molecular Biology and Genetics; Aarhus University; Aarhus Denmark
| |
Collapse
|
34
|
Crabtree JS. Clinical and Preclinical Advances in Gastroenteropancreatic Neuroendocrine Tumor Therapy. Front Endocrinol (Lausanne) 2017; 8:341. [PMID: 29255447 PMCID: PMC5722794 DOI: 10.3389/fendo.2017.00341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022] Open
Abstract
The molecular events leading to gastroenteropancreatic neuroendocrine tumor (GEP-NET) formation are largely unknown. Over the past decades, systemic chemotherapies have been replaced by therapies directed at particular molecular targets such as the somatostatin receptors, mTOR complexes or proangiogenic molecules. These approaches have demonstrated some success in subtypes of this heterogeneous tumor group, but responses are still widely varied. This review highlights the clinical trials ongoing for neuroendocrine tumors (NETs) and includes emerging immunotherapy, which holds great promise for NETs based on successes in other tumor types. Current avenues of preclinical research, including Notch and PI3K/AKT, will lead to additional targeted therapies based on genome-wide studies that have cast a wide net in the search for driver mutations. Future preclinical and clinical investigations are required to identify those mutations predictive of therapeutic response or disease progression. Results of current clinical trials outlined here will better inform patient management with respect to agent selection, timing, duration and combination therapy in the treatment of NETs.
Collapse
Affiliation(s)
- Judy S. Crabtree
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- *Correspondence: Judy S. Crabtree,
| |
Collapse
|
35
|
Fotouhi O, Kjellin H, Larsson C, Hashemi J, Barriuso J, Juhlin CC, Lu M, Höög A, Pastrián LG, Lamarca A, Soto VH, Zedenius J, Mendiola M, Lehtiö J, Kjellman M. Proteomics Suggests a Role for APC-Survivin in Response to Somatostatin Analog Treatment of Neuroendocrine Tumors. J Clin Endocrinol Metab 2016; 101:3616-3627. [PMID: 27459532 PMCID: PMC5052342 DOI: 10.1210/jc.2016-2028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
CONTEXT Somatostatin analogs are established in the treatment of neuroendocrine tumors (NETs) including small intestinal NET; however, the molecular mechanisms are not well known. Here, we examined the direct effects of lanreotide in NET cell line models. SETTING AND DESIGN The cell lines HC45 and H727 were treated with 10nM lanreotide for different time periods and alterations of the proteome were analyzed by in-depth high-resolution isoelectric focusing tandem liquid chromatography-mass spectrometry. We next investigated whether the observed suppression of survivin was mediated by adenomatous polyposis coli (APC) and possible effects on tumor proliferation in vitro. Expression of survivin was assessed by immunohistochemistry in 112 NET cases and compared with patient outcome. RESULTS We quantified 6451 and 7801 proteins in HC45 and H727, respectively. After short time lanreotide treatment APC was increased and survivin reduced. Overexpression of APC in H727 cells decreased, and APC knock-down elevated the survivin level. The lanreotide regulation of APC-survivin could be suppressed by small interfering RNA against somatostatin receptor 2. Although lanreotide only gave slight inhibition of proliferation, targeting of survivin with the small molecule YM155 dramatically reduced proliferation. Moderate or high as compared with low or absent total survivin expression was associated with shorter progression-free survival, independent of tumor stage, grade, and localization. CONCLUSIONS We report a proteome-wide analysis of changes in response to lanreotide in NET cell lines. This analysis suggests a connection between somatostatin analog, APC, and survivin levels. Survivin is a possible prognostic factor and a new potential therapeutic target in NETs.
Collapse
Affiliation(s)
- Omid Fotouhi
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Hanna Kjellin
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Catharina Larsson
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Jamileh Hashemi
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Jorge Barriuso
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - C Christofer Juhlin
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Ming Lu
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Anders Höög
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Laura G Pastrián
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Angela Lamarca
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Victoria Heredia Soto
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Jan Zedenius
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Marta Mendiola
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Janne Lehtiö
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Magnus Kjellman
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| |
Collapse
|
36
|
Cives M, Simone V, Rizzo FM, Silvestris F. NETs: organ-related epigenetic derangements and potential clinical applications. Oncotarget 2016; 7:57414-57429. [PMID: 27418145 PMCID: PMC5302998 DOI: 10.18632/oncotarget.10598] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/30/2016] [Indexed: 12/15/2022] Open
Abstract
High-throughput next-generation sequencing methods have recently provided a detailed picture of the genetic landscape of neuroendocrine tumors (NETs), revealing recurrent mutations of chromatin-remodeling genes and little-to-no pathogenetic role for oncogenes commonly mutated in cancer. Concurrently, multiple epigenetic modifications have been described across the whole spectrum of NETs, and their putative function as tumorigenic drivers has been envisaged. As result, it is still unclear whether or not NETs are epigenetically-driven, rather than genetically-induced malignancies. Although the NET epigenome profiling has led to the identification of molecularly-distinct tumor subsets, validation studies in larger cohorts of patients are needed to translate the use of NET epitypes in clinical practice. In the precision medicine era, recognition of subpopulations of patients more likely to respond to therapeutic agents is critical, and future studies testing epigenetic biomarkers are therefore awaited. Restoration of the aberrant chromatin remodeling machinery is an attractive approach for future treatment of cancer and in several hematological malignancies a few epigenetic agents have been already approved. Although clinical outcomes of epigenetic therapies in NETs have been disappointing so far, further clinical trials are required to investigate the efficacy of these drugs. In this context, given the immune-stimulating effects of epidrugs, combination therapies with immune checkpoint inhibitors should be tested. In this review, we provide an overview of the epigenetic changes in both hereditary and sporadic NETs of the gastroenteropancreatic and bronchial tract, focusing on their diagnostic, prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Mauro Cives
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari “Aldo Moro”, Bari, Italy
| | - Valeria Simone
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari “Aldo Moro”, Bari, Italy
| | - Francesca Maria Rizzo
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari “Aldo Moro”, Bari, Italy
| | - Franco Silvestris
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
37
|
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) constitute a heterogeneous group of tumours associated with variable clinical presentations, growth rates, and prognoses. To improve the management of GEP-NENs, the WHO developed a classification system that enables tumours to be graded based on markers of cell proliferation in biopsy specimens. Indeed, histopathology has been a mainstay in the diagnosis of GEP-NENs, and the WHO grading system facilitates therapeutic decision-making; however, considerable intratumoural heterogeneity, predominantly comprising regional variations in proliferation rates, complicates the evaluation of tumour biology. The use of molecular imaging modalities to delineate the most-aggressive cell populations is becoming more widespread. In addition, molecular profiling is increasingly undertaken in the clinical setting, and genomic studies have revealed a number of chromosomal alterations in GEP-NENs, although the 'drivers' of neoplastic development have not been identified. Thus, our molecular understanding of GEP-NENs remains insufficient to inform on patient prognosis or selection for treatments, and the WHO classification continues to form the basis for management of this disease. Nevertheless, our increasing understanding of the molecular genetics and biology of GEP-NENs has begun to expose flaws in the WHO classification. We describe the current understanding of the molecular characteristics of GEP-NENs, and discuss how advances in molecular profiling measurements, including assays of circulating mRNAs, are likely to influence the management of these tumours.
Collapse
|
38
|
Legendre CR, Demeure MJ, Whitsett TG, Gooden GC, Bussey KJ, Jung S, Waibhav T, Kim S, Salhia B. Pathway Implications of Aberrant Global Methylation in Adrenocortical Cancer. PLoS One 2016; 11:e0150629. [PMID: 26963385 PMCID: PMC4786116 DOI: 10.1371/journal.pone.0150629] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 02/17/2016] [Indexed: 12/02/2022] Open
Abstract
Context Adrenocortical carcinomas (ACC) are a rare tumor type with a poor five-year survival rate and limited treatment options. Objective Understanding of the molecular pathogenesis of this disease has been aided by genomic analyses highlighting alterations in TP53, WNT, and IGF signaling pathways. Further elucidation is needed to reveal therapeutically actionable targets in ACC. Design In this study, global DNA methylation levels were assessed by the Infinium HumanMethylation450 BeadChip Array on 18 ACC tumors and 6 normal adrenal tissues. A new, non-linear correlation approach, the discretization method, assessed the relationship between DNA methylation/gene expression across ACC tumors. Results This correlation analysis revealed epigenetic regulation of genes known to modulate TP53, WNT, and IGF signaling, as well as silencing of the tumor suppressor MARCKS, previously unreported in ACC. Conclusions DNA methylation may regulate genes known to play a role in ACC pathogenesis as well as known tumor suppressors.
Collapse
Affiliation(s)
| | - Michael J. Demeure
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Timothy G. Whitsett
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Gerald C. Gooden
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Kimberly J. Bussey
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
- NantOmics, LLC, Phoenix, Arizona, United States of America
| | - Sungwon Jung
- Department of Genome Medicine and Science, Gachon University School of Medicine, Incheon, 21565, Republic of Korea
- Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, 21565, Republic of Korea
| | - Tembe Waibhav
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Seungchan Kim
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Bodour Salhia
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
- * E-mail:
| |
Collapse
|
39
|
Yang XY, Miyamoto C, Akasaka T, Izukuri K, Maehata Y, Ikoma T, Ozawa S, Hata RI. Chemokine CXCL14 is a multistep tumor suppressor. J Oral Biosci 2016. [DOI: 10.1016/j.job.2015.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
40
|
Karpathakis A, Dibra H, Pipinikas C, Feber A, Morris T, Francis J, Oukrif D, Mandair D, Pericleous M, Mohmaduvesh M, Serra S, Ogunbiyi O, Novelli M, Luong T, Asa SL, Kulke M, Toumpanakis C, Meyer T, Caplin M, Meyerson M, Beck S, Thirlwell C. Prognostic Impact of Novel Molecular Subtypes of Small Intestinal Neuroendocrine Tumor. Clin Cancer Res 2016; 22:250-8. [PMID: 26169971 DOI: 10.1158/1078-0432.ccr-15-0373] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 06/25/2015] [Indexed: 12/16/2022]
Abstract
PURPOSE Small intestinal neuroendocrine tumors (SINET) are the commonest malignancy of the small intestine; however, underlying pathogenic mechanisms remain poorly characterized. Whole-genome and -exome sequencing has demonstrated that SINETs are mutationally quiet, with the most frequent known mutation in the cyclin-dependent kinase inhibitor 1B gene (CDKN1B) occurring in only ∼8% of tumors, suggesting that alternative mechanisms may drive tumorigenesis. The aim of this study is to perform genome-wide molecular profiling of SINETs in order to identify pathogenic drivers based on molecular profiling. This study represents the largest unbiased integrated genomic, epigenomic, and transcriptomic analysis undertaken in this tumor type. EXPERIMENTAL DESIGN Here, we present data from integrated molecular analysis of SINETs (n = 97), including whole-exome or targeted CDKN1B sequencing (n = 29), HumanMethylation450 BeadChip (Illumina) array profiling (n = 69), methylated DNA immunoprecipitation sequencing (n = 16), copy-number variance analysis (n = 47), and Whole-Genome DASL (Illumina) expression array profiling (n = 43). RESULTS Based on molecular profiling, SINETs can be classified into three groups, which demonstrate significantly different progression-free survival after resection of primary tumor (not reached at 10 years vs. 56 months vs. 21 months, P = 0.04). Epimutations were found at a recurrence rate of up to 85%, and 21 epigenetically dysregulated genes were identified, including CDX1 (86%), CELSR3 (84%), FBP1 (84%), and GIPR (74%). CONCLUSIONS This is the first comprehensive integrated molecular analysis of SINETs. We have demonstrated that these tumors are highly epigenetically dysregulated. Furthermore, we have identified novel molecular subtypes with significant impact on progression-free survival.
Collapse
Affiliation(s)
- Anna Karpathakis
- University College London, London, United Kingdom. The Royal Free Hospital, London, United Kingdom
| | | | | | - Andrew Feber
- University College London, London, United Kingdom
| | | | - Joshua Francis
- The Broad Institute of Harvard and MIT, Cambridge, Massachusetts. Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Dalvinder Mandair
- University College London, London, United Kingdom. The Royal Free Hospital, London, United Kingdom
| | | | | | - Stefano Serra
- UHN Princess Margaret Cancer Centre, Toronto, Canada
| | | | | | | | - Sylvia L Asa
- UHN Princess Margaret Cancer Centre, Toronto, Canada
| | - Matthew Kulke
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Tim Meyer
- University College London, London, United Kingdom. The Royal Free Hospital, London, United Kingdom
| | | | - Matthew Meyerson
- The Broad Institute of Harvard and MIT, Cambridge, Massachusetts. Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Stephan Beck
- University College London, London, United Kingdom
| | - Christina Thirlwell
- University College London, London, United Kingdom. The Royal Free Hospital, London, United Kingdom.
| |
Collapse
|
41
|
Zheng P, Yang T, Ju L, Jiang B, Lou Y. Epigenetics in Legg-Calvé-Perthes disease: A study of global DNA methylation. J Int Med Res 2015; 43:758-64. [PMID: 26443715 DOI: 10.1177/0300060515591062] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To examine the global methylation status of DNA in blood cells of children with Legg-Calvé-Perthes disease (LCPD), since the aetiopathogenesis of LCPD remains unclear, and many factors closely associated with DNA methylation may be linked to the occurrence of LCPD. METHODS Children with LCPD and age-, sex- and body mass index-matched controls were evaluated. Methylation levels of the long interspersed nuclear element 1 (LINE-1), a biomarker of global DNA methylation, were quantified by methylation-specific polymerase chain reaction. RESULTS Of 82 children with LCPD (68 male/14 female) and 120 matched controls (98 male/22 female), methylation of the LINE-1 promoter was significantly lower in patients with LCPD compared with controls. Subgroup analyses showed that methylation of the LINE-1 promoter was significantly lower in male patients with LCPD compared with male controls. No significant between-group differences were observed in female participants. CONCLUSIONS Reduced global DNA methylation may be associated with increased risk of LCPD in male children. Further research is required to understand whether detection of global DNA methylation may provide a basis for clinical diagnosis and early intervention of LCPD.
Collapse
Affiliation(s)
- Pengfei Zheng
- Department of Paediatric Orthopaedics, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Tao Yang
- Department of Orthopaedics, People's Hospital of Jingjiang, Jingjiang, China
| | - Li Ju
- Department of Paediatric Orthopaedics, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Bo Jiang
- Department of Paediatric Orthopaedics, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yue Lou
- Department of Paediatric Orthopaedics, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Mapelli P, Aboagye EO, Stebbing J, Sharma R. Epigenetic changes in gastroenteropancreatic neuroendocrine tumours. Oncogene 2015; 34:4439-47. [PMID: 25435371 DOI: 10.1038/onc.2014.379] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/25/2014] [Accepted: 10/10/2014] [Indexed: 02/07/2023]
Abstract
An understanding of epigenetic drivers of tumorigenesis has developed rapidly during the last years. The identification of these changes including DNA methylation and histone modifications in gastroenteropancreatic neuroendocrine tumours (GEP-NETs) is a step forward in trying to define underlying biologic processes in this heterogeneous disease. The reversible nature of these changes represents a potential therapeutic target. We present an overview of the current knowledge of epigenetic alterations related to GEP-NETs, focusing on the influence and impact these changes have on pathogenesis and prognosis. The potential role of demethylating agents in the management of this patient population is discussed.
Collapse
Affiliation(s)
- P Mapelli
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - E O Aboagye
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - J Stebbing
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - R Sharma
- Department of Experimental Medicine, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
43
|
Bai JA, Hu YL, Tang QY. Advances in clinical and basic research of gastroentero-pancreatic neuroendocrine neoplasms. Shijie Huaren Xiaohua Zazhi 2015; 23:2913-2919. [DOI: 10.11569/wcjd.v23.i18.2913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastroentero-pancreatic neuroendocrine neoplasms (GEP-NENs) are a group of relatively rare tumors, which mainly originate from the peptidergic neuron and neuroendocrine cells of the gastroentero-pancreatic system. They are characterized by secretion of peptide hormones and neuroendocrine markers (such as synaptic vesicle proteins and chromaffin granule A). Surgery is the most effective therapy for GEP-NENs at early stages. For GEP-NENs at progressive stages, biological target therapies have aroused great interest. Current studies about the molecular basis of biological target therapies have focused on the GEP-NEN gene mutations and related signaling pathways. These studies have led to the clinical application with significant progress in GEP-NEN treatment. In this paper, we review the recent advances in the clinical and basic research of GEP-NENs.
Collapse
|
44
|
Miller HC, Kidd M, Castellano L, Frilling A. Molecular genetic findings in small bowel neuroendocrine neoplasms: a review of the literature. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2015. [DOI: 10.2217/ije.14.41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Small bowel neuroendocrine neoplasms (SBNEN) are the most common small bowel tumor and have an increasing incidence. Despite many treatment options, therapeutic strategy remains a key clinical challenge due to the paucity of large-scale, randomized controlled trials. The heterogeneity of SBNEN coupled with a lack of detailed information about the tumor biology, impedes patient stratification into groups based on tumor phenotypes or treatment response. More detailed analysis of the genetic and epigenetic characteristics of SBNEN, will allow treatment to move toward a more personalized medicine approach through the identification of novel biomarkers and therapeutic targets, with the aim to increase survival.
Collapse
Affiliation(s)
- Helen C Miller
- Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Mark Kidd
- Department of Surgery, Yale University, School of Medicine, 333 Cedar Street, New Haven, CT 06520–8062, USA
| | - Leandro Castellano
- Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Andrea Frilling
- Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| |
Collapse
|
45
|
Primary carcinoid tumor of medulla spinalis: case report and review of the literature. Eur J Med Res 2014; 19:71. [PMID: 25523133 PMCID: PMC4272791 DOI: 10.1186/s40001-014-0071-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 11/28/2014] [Indexed: 11/10/2022] Open
Abstract
Background Carcinoid tumors are slow growing neuroendocrine tumors which can originate from various sites within the body. A carcinoid tumor originating in the medulla spinalis has not previously been reported in the literature. Case report We report a case of a 33-year-old man, presenting with a five-month history of bilateral lower extremity pain, as well as paresthesia, and mild weakness in one lateral lower extremity. A lumbar laminectomy of L3 to L5 and en bloc resection of the tumor was performed. Postoperative histopathology and immunohistochemical analysis of the tumor were consistent with that of a carcinoid tumor. There were no clinical or radiological signs of tumor recurrence or metastasis at the patient’s two year postoperative follow-up. Conclusions During the differential diagnosis of medulla spinalis tumors, the possibility of a primary carcinoid tumor originating within the medulla spinalis should be considered. An accurate tumor classification is imperative to ensure that the most effective course of treatment is pursued. Electronic supplementary material The online version of this article (doi:10.1186/s40001-014-0071-7) contains supplementary material, which is available to authorized users.
Collapse
|