63 results on '"COPY-NUMBER VARIATION"'
Search Results
2. Organ Specific Copy Number Variations in Visceral Metastases of Human Melanoma
- Author
-
Sarolta Kárpáti, Jeovanis Gil, József Tímár, Viktória Doma, Orsolya Papp, György Marko-Varga, and Laura Vízkeleti
- Subjects
distant organ metastasis ,DDR deficiency ,HGF/MET autocrine activation ,immunogenic mimicry ,BRAF and NRAS mutant allele frequency ,Cancer Research ,Lung ,business.industry ,Melanoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Locus (genetics) ,medicine.disease ,Article ,Lymphatic system ,medicine.anatomical_structure ,Oncology ,Cancer research ,Medicine ,Ectopic expression ,Copy-number variation ,business ,Autocrine signalling ,Gene ,RC254-282 - Abstract
Simple Summary Malignant melanoma is a highly metastatic disease disseminating to several distant sites. This potential is also of great clinical impact for patient survival and therapeutic success. Knowledge about melanoma genomics is mainly based on lymphatic or skin metastases derived data, whereas data from distant sites is limited. Therefore, an autopsy-based visceral metastasis biobank was established, and an array-based copy number variation (CNV) analysis was performed, focusing primarily on major organs (brain, lung, and liver) and completed partly by proteomic analysis. A unique picture emerged about organ-specific CNV-type distributions or gene alterations, including the frequent loss of DNA damage error genes in brain metastases, the presence of HGF/MET autocrine loop in brain and lung metastases, the traces of immunogenic mimicry exclusive for lung metastases or the correlation of BRAF copy number and mutant allele frequency, especially in lung metastases. All these above phenomena have a great influence on therapy efficacy or resistance. Abstract Malignant melanoma is one of the most aggressive skin cancers with high potential of visceral dissemination. Since the information about melanoma genomics is mainly based on primary tumors and lymphatic or skin metastases, an autopsy-based visceral metastasis biobank was established. We used copy number variation arrays (N = 38 samples) to reveal organ specific alterations. Results were partly completed by proteomic analysis. A significant increase of high-copy number gains was found in an organ-specific manner, whereas copy number losses were predominant in brain metastases, including the loss of numerous DNA damage response genes. Amplification of many immune genes was also observed, several of them are novel in melanoma, suggesting that their ectopic expression is possibly underestimated. This “immunogenic mimicry” was exclusive for lung metastasis. We also provided evidence for the possible autocrine activation of c-MET, especially in brain and lung metastases. Furthermore, frequent loss of 9p21 locus in brain metastases may predict higher metastatic potential to this organ. Finally, a significant correlation was observed between BRAF gene copy number and mutant allele frequency, mainly in lung metastases. All of these events may influence therapy efficacy in an organ specific manner, which knowledge may help in alleviating difficulties caused by resistance.
- Published
- 2021
3. MET Amplification in Non-Small Cell Lung Cancer (NSCLC)—A Consecutive Evaluation Using Next-Generation Sequencing (NGS) in a Real-World Setting
- Author
-
Christoph Seidl, Arndt Hartmann, Rumo Leistner, Christoph Schubart, Wolfgang Hohenforst-Schmidt, Gerhard Seitz, Michael Vieth, Horia Sirbu, Lars Tögel, Florian S. Fuchs, William Sterlacci, Ruth Seggewiss-Bernhardt, Robert Stöhr, Markus Kapp, Ramona Erber, Michael Mugler, and Florian Haller
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,precision medicine ,non-small cell lung cancer (NSCLC) ,Context (language use) ,Biology ,amplification ,NSCLC ,DNA sequencing ,Article ,Internal medicine ,medicine ,Copy-number variation ,ddc:610 ,MET ,Gene ,fluorescence in situ hybridization ,MET inhibitor ,RC254-282 ,medicine.diagnostic_test ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Gold standard (test) ,Amplicon ,medicine.disease ,next-generation sequencing ,Fluorescence in situ hybridization ,resistance mechanism - Abstract
In non-small cell lung cancer (NSCLC), approximately 1–3% of cases harbor an increased gene copy number (GCN) of the MET gene. This alteration can be due to de novo amplification of the MET gene or can represent a secondary resistance mechanism in response to targeted therapies. To date, the gold standard method to evaluate the GCN of MET is fluorescence in situ hybridization (FISH). However, next-generation sequencing (NGS) is becoming more relevant to optimize therapy by revealing the mutational profile of each NSCLC. Using evaluable n = 205 NSCLC cases of a consecutive cohort, this study addressed the question of whether an amplicon based NGS assay can completely replace the FISH method regarding the classification of MET GCN status. Out of the 205 evaluable cases, only n = 9 cases (43.7%) of n = 16 high-level MET amplified cases assessed by FISH were classified as amplified by NGS. Cases harboring a MET GCN >, 10 showed the best concordance when comparing FISH versus NGS (80%). This study confirms that an amplicon-based NGS assessment of the MET GCN detects high-level MET amplified cases harboring a MET GCN >, 10 but fails to detect the various facets of MET gene amplification in the context of a therapy-induced resistance mechanism.
- Published
- 2021
- Full Text
- View/download PDF
4. Genomic Determinants of Homologous Recombination Deficiency across Human Cancers
- Author
-
Kuan-lin Huang, Lajos Pusztai, Tao Qing, Li Ding, Tomi Jun, and Xinfeng Wang
- Subjects
Cancer Research ,Candidate gene ,Somatic cell ,PALB2 ,copy number variation ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,homologous recombination ,Synthetic lethality ,Biology ,medicine.disease ,Phenotype ,Germline ,Article ,Oncology ,medicine ,Cancer research ,DNA damage repair ,germline and somatic ,Copy-number variation ,RC254-282 - Abstract
Simple Summary Tumors with homologous recombination deficiency (HRD) respond to Poly-ADP ribose polymerase inhibitor (PARPi) therapy in breast, ovarian, prostate, and pancreatic cancers. However, in addition to a handful of known pathogenic variants including those affecting BRCA1/2, it remains unclear what other genomic events can cause HRD. Our study systematically examined the germline and somatic genome of over 9000 cancers. We identified alterations associated with HRD, including mutations in ATM and ATR genes; specific deletions in stomach, bladder, and lung cancer; and BRCA-wild type breast, ovarian, and pancreatic cancers. These findings suggest a potentially broader utility for PARPi for cancers harboring a wide range of genomic alterations. Abstract Germline BRCA1/2 mutations associated with HRD are clinical biomarkers for sensitivity to poly-ADP ribose polymerase inhibitors (PARPi) treatment in breast, ovarian, pancreatic, and prostate cancers. However, it remains unclear whether other mutations may also lead to HRD and PARPi sensitivity across a broader range of cancer types. Our goal was to determine the germline or somatic alterations associated with the HRD phenotype that might therefore confer PARPi sensitivity. Using germline and somatic genomic data from over 9000 tumors representing 32 cancer types, we examined associations between HRD scores and pathogenic germline variants, somatic driver mutations, and copy number deletions in 30 candidate genes involved in homologous recombination. We identified several germline and somatic mutations (e.g., BRCA1/2, PALB2, ATM, and ATR mutations) associated with HRD phenotype in ovarian, breast, pancreatic, stomach, bladder, and lung cancer. The co-occurrence of germline BRCA1 variants and somatic TP53 mutations was significantly associated with increasing HRD in breast cancer. Notably, we also identified multiple somatic copy number deletions associated with HRD. Our study suggests that multiple cancer types include tumor subsets that show HRD phenotype and should be considered in the future clinical studies of PARPi and synthetic lethality strategies exploiting HRD, which can be caused by a large number of genomic alterations.
- Published
- 2021
5. The Clinical Utility of Optical Genome Mapping for the Assessment of Genomic Aberrations in Acute Lymphoblastic Leukemia
- Author
-
Martin Stanulla, Martin Schrappe, Brigitte Schlegelberger, Marie Wolter, Jana Lentes, Marie Stelter, Markéta Žaliová, Maximilian Schieck, Doris Steinemann, Anke K. Bergmann, Gunnar Cario, Jonathan Lukas Lühmann, Anja Möricke, Josephine Kater, and Gudrun Göhring
- Subjects
Cancer Research ,copy number alteration ,Lymphoblastic Leukemia ,Cancer ,risk assessment ,gene fusion ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Karyotype ,Chromosomal translocation ,Computational biology ,molecular karyotyping ,Biology ,Optical Genome Mapping ,medicine.disease ,Article ,Fusion gene ,Oncology ,Gene mapping ,medicine ,Copy-number variation ,Hyperdiploidy ,ALL ,prognostic marker ,RC254-282 - Abstract
Acute lymphoblastic leukemia (ALL) is the most prevalent type of cancer occurring in children. ALL is characterized by structural and numeric genomic aberrations that strongly correlate with prognosis and clinical outcome. Usually, a combination of cyto- and molecular genetic methods (karyotyping, array-CGH, FISH, RT-PCR, RNA-Seq) is needed to identify all aberrations relevant for risk stratification. We investigated the feasibility of optical genome mapping (OGM), a DNA-based method, to detect these aberrations in an all-in-one approach. As proof of principle, twelve pediatric ALL samples were analyzed by OGM, and results were validated by comparing OGM data to results obtained from routine diagnostics. All genomic aberrations including translocations (e.g., dic(9, 12)), aneuploidies (e.g., high hyperdiploidy) and copy number variations (e.g., IKZF1, PAX5) known from other techniques were also detected by OGM. Moreover, OGM was superior to well-established techniques for resolution of the more complex structure of a translocation t(12, 21) and had a higher sensitivity for detection of copy number alterations. Importantly, a new and unknown gene fusion of JAK2 and NPAT due to a translocation t(9, 11) was detected. We demonstrate the feasibility of OGM to detect well-established as well as new putative prognostic markers in an all-in-one approach in ALL. We hope that these limited results will be confirmed with testing of more samples in the future.
- Published
- 2021
6. Development and Validation of Targeted Gene Sequencing Panel Based Companion Diagnostic for Korean Patients with Solid Tumors
- Author
-
Ju Han Kim, Seung-Yong Jeong, Kye Hwa Lee, Byung Joo Min, Sang Won Shin, Ja-Lok Ku, Myung Eui Seo, Yeul Hong Kim, and Woo Seung Lee
- Subjects
pharmacogenomics ,Cancer Research ,business.industry ,precision medicine ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,Computational biology ,Precision medicine ,medicine.disease ,Tumor tissue ,DNA sequencing ,Article ,Oncology ,Pharmacogenomics ,custom panel ,medicine ,Copy-number variation ,targeted sequencing ,Gene screening ,business ,RC254-282 ,Companion diagnostic - Abstract
Simple Summary We have developed and analytically validated the Korean Pan-cancer Companion Diagnostic (CDX) Panel to apply targeted anticancer drugs to Korean patients based on the molecular characteristics of tumors using tumor samples without matched patient normal samples. The panel included 31 genes with reported single nucleotide variants, 9 genes with reported copy number variations, and 15 genes with predictive responses to targeted drugs under clinical testing, enabling the panel to be analyzed for the targets of 30 targeted anticancer drugs. It is cost-effective and optimized for cancer type-specific therapy in Korean cancer patients across solid cancer types while minimizing the limitations of existing approaches. This gene screening method is expected to reduce test turnaround time and cost, making it a balanced approach to investigate solid cancer-related gene regions. Abstract Recently, several panels using two representative targeting methods have been developed but they do not reflect racial specificity, especially for Asians. We have developed and analytically validated the Korean Pan-cancer Companion Diagnostic (CDX) Panel to apply targeted anticancer drugs to Korean patients based on the molecular characteristics of tumors using tumor samples without matched patient normal samples. The panel included 31 genes with reported single nucleotide variants, 9 genes with reported copy number variations, and 15 genes with predictive responses to targeted drugs under clinical testing, enabling the panel to be analyzed for the targets of 30 targeted anticancer drugs. It is cost-effective and optimized for cancer type-specific therapy in Korean cancer patients across solid cancer types while minimizing the limitations of existing approaches. In addition, the optimized filtering protocol for somatic variants from tumor-only samples enables researchers to use this panel without matched normal samples. To verify the panel, 241 frozen tumor tissues and 71 formalin-fixed paraffin-embedded (FFPE) samples from several institutes were registered. This gene screening method is expected to reduce test turnaround time and cost, making it a balanced approach to investigate solid cancer-related gene regions.
- Published
- 2021
7. TCR Clonality and Genomic Instability Signatures as Prognostic Biomarkers in High Grade Serous Ovarian Cancer
- Author
-
François Ghiringhelli, Isabelle Ray Coquard, Hugo Mananet, Caroline Truntzer, Zoé Tharin, Jean-David Fumet, Julie Lecuelle, Juliette Albuisson, Valentin Derangère, Laurent Arnould, Vincent Goussot, and Romain Boidot
- Subjects
Genome instability ,Cancer Research ,Tumor microenvironment ,medicine.medical_treatment ,T cell ,T-cell receptor ,TCR clonality ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,biomarkers ,Immunotherapy ,Biology ,HGSC ,Article ,Serous fluid ,Immune system ,medicine.anatomical_structure ,Oncology ,HRD ,medicine ,Cancer research ,Copy-number variation ,prognostic ,RC254-282 - Abstract
Simple Summary High-grade serous ovarian carcinoma (HGSC) could be analyzed with a molecular stratification defined by different genomic instability signatures associated with specific mutational process and prognostic biomarkers. Immune infiltrate is known to be a robust biomarker in HGSC. We aimed to investigate immune parameters according to genomic instability signatures. We observed that homologous recombination deficiency positive, copy cumber variant signature 7 and TCR (T cells receptor) clonality are good prognostic biomarkers in HGSC. Combining TCR clonality and genomic instability signature or T cell infiltration improved the prognostic value compared to each variable taken alone. We provided a description of immune parameters according to different genomic instability signatures. We identified TCR clonality, alone or combined with genomic instability, as a promising prognostic biomarker in HGSC. Abstract Purpose: Immune infiltration is a prognostic factor in high-grade serous ovarian carcinoma (HGSC) but immunotherapy efficacy is disappointing. Genomic instability is now used to guide the therapeutic value of PARP inhibitors. We aimed to investigate exome-derived parameters to assess the tumor microenvironment according to genomic instability profile. Methods: We used the HGSC TCGA (the cancer genome atlas) dataset with genomic characteristics, including homologous recombination deficiency (HRD), copy number variant (CNV) signatures, TCR (T cell receptor) clonality and abundance of tissue-infiltrating immune and stromal cell populations. We then investigated the relationship with survival data. Results: In 578 HGSC patients, HRD status, CNV signature 7 and TCR clonality were associated with longer survival. The combination of high CNV signature 7 expression and HRD status or high CNV signature 3 expression and high TCR clonality was associated with a trend towards longer survival compared to each variable alone. Combining T cell infiltrate and TCR clonality improved the prognostic value compared to T cells infiltration alone. Prognostic value of TCR clonality was confirmed in an independent cohort. Conclusions: TCR clonality is an emerging prognostic biomarker that improves T cell infiltrate information. Analysis of TCR clonality combined with genomic instability could be an interesting prognostic biomarker.
- Published
- 2021
8. Accurate Prognosis Prediction of Pancreatic Ductal Adenocarcinoma Using Integrated Clinico-Genomic Data of Endoscopic Ultrasound-Guided Fine Needle Biopsy
- Author
-
Minseob Cho, Kyu Taek Lee, Kwang Hyuck Lee, Dong Hyo Noh, Young Kyung Sung, Kee-Taek Jang, Kim Hye-Min, Jong Kyun Lee, Chung Lee, Woong-Yang Park, Joo Kyung Park, Dae-Soon Son, Nayoung Kim, and Se-Hoon Lee
- Subjects
0301 basic medicine ,Oncology ,Endoscopic ultrasound ,Cancer Research ,medicine.medical_specialty ,endocrine system diseases ,pancreatic ductal adenocarcinoma ,endoscopic ultrasound-guided fine needle core biopsy ,medicine.disease_cause ,Article ,Deep sequencing ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,CDKN2A ,Internal medicine ,Biopsy ,medicine ,Copy-number variation ,Stage (cooking) ,RC254-282 ,medicine.diagnostic_test ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,digestive system diseases ,030104 developmental biology ,030220 oncology & carcinogenesis ,clinico-genomic model ,KRAS ,prognosis prediction ,business ,targeted deep sequencing - Abstract
The aim of this study was to investigate the clinical utility of minimal specimens acquired from endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB) and perform targeted deep sequencing as a prognosis prediction tool for pancreatic ductal adenocarcinoma (PDAC). A total of 116 specimens with pathologically confirmed PDAC via EUS-FNB were tested using CancerSCAN® panel for a customized targeted deep sequencing. Clinical prognostic factors significantly associated with survival in PDACs were as follows: stage, tumor mass size, tumor location, metastasis, chemotherapy, and initial CA19-9 level. A total of 114 patients (98.3%) had at least a single genetic alteration, and no mutations were detected in two patients, although they were qualified for the targeted deep sequencing. The frequencies of major gene mutations responsible for PDACs were KRAS 90%, CDKN2A 31%, TP53 77%, and SMAD4 29%. A somatic point mutation of NF1, copy number alteration of SMAD4, and loss-of-function of CDKN2A were significantly associated genetic factors for overall survival. Moreover, BRCA2 point mutation was related to liver metastasis. Finally, a clinico-genomic model was developed to estimate the prognosis of patients with PDAC based on clinical parameters and genetic alterations affecting survival in patients, 20 single nucleotide variants and three copy number variations were selected. Targeted deep sequencing on minimal specimens of PDACs was performed, and it was applied to establish a clinico-genomic model for prognosis prediction.
- Published
- 2021
9. Whole Exome Sequencing of Biliary Tubulopapillary Neoplasms Reveals Common Mutations in Chromatin Remodeling Genes
- Author
-
Günter Klöppel, Julia Weber, Thomas Engleitner, Roland Rad, Gisela Keller, Björn Konukiewitz, Sebastian Lange, Alexander Muckenhuber, Irene Esposito, Katja Steiger, Wilko Weichert, Claudia Gross, Nicole Pfarr, Nazmi Volkan Adsay, Benjamin Goeppert, Moritz Jesinghaus, Anna Melissa Schlitter, Adsay, Volkan (ORCID 0000-0002-1308-3701 & YÖK ID286248), Gross, Claudia, Engleitner, Thomas, Lange, Sebastian, Weber, Julia, Jesinghaus, Moritz, Konukiewitz, Bjoern, Muckenhuber, Alexander, Steiger, Katja, Pfarr, Nicole, Goeppert, Benjamin, Keller, Gisela, Weichert, Wilko, Kloeppel, Gunter, Rad, Roland, Esposito, Irene, Schlitter, Anna Melissa, and School of Medicine
- Subjects
0301 basic medicine ,Cancer Research ,Biology ,medicine.disease_cause ,Article ,Chromatin remodeling ,whole exome sequencing ,03 medical and health sciences ,0302 clinical medicine ,CDKN2A ,bile duct ,medicine ,GNAS complex locus ,Copy-number variation ,pancreas ,Exome sequencing ,RC254-282 ,Genetics ,intraductal tubulopapillary neoplasms ,Pancreas ,Bile duct ,Intraductal tubulopapillary neoplasms ,Whole exome sequencing ,Chromatin remodeling genes ,chromatin remodeling genes ,Genetic heterogeneity ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,KRAS ,Carcinogenesis - Abstract
Simple summary: Intraductal tubulopapillary neoplasms (ITPN) have recently been described as rare precursor lesions of pancreatic ductal adenocarcinoma and cholangiocarcinoma. Despite a high number of associated invasive adenocarcinomas at the time of diagnosis, patients with ITPN tend to have a much better clinical outcome than those with classical pancreato-biliary adenocarcinoma. Furthermore, rare molecular studies of ITPN show an unexpected lack of hotspot mutations in common driver genes of pancreato-biliary adenocarcinoma, including KRAS. This article reports the first large comprehensive and comparative molecular study of pancreato-biliary ITPN. In the absence of KRAS mutations, we found a high genetic heterogeneity with enrichment in core signaling pathways, including putative actionable genomic targets in one-third of the cases. Whereas, pancreatic ITPN demonstrates a highly distinct genetic profile, differing from classical pancreatic carcinogenesis, biliary ITPN and classical cholangiocarcinoma share common alterations in key genes of the chromatin remodeling pathway, and therefore, appear more closely related than pancreatic ITPN and classical pancreatic ductal adenocarcinoma PDAC. The molecular carcinogenesis of intraductal tubulopapillary neoplasms (ITPN), recently described as rare neoplasms in the pancreato-biliary tract with a favorable prognosis despite a high incidence of associated pancreato-biliary adenocarcinoma, is still poorly understood. To identify driver genes, chromosomal gains and losses, mutational signatures, key signaling pathways, and potential therapeutic targets, the molecular profile of 11 biliary and 6 pancreatic ITPNs, associated with invasive adenocarcinoma in 14/17 cases, are studied by whole exome sequencing (WES). The WES of 17 ITPNs reveals common copy number variants (CNVs) broadly distributed across the genome, with recurrent chromosomal deletions primarily in 1p36 and 9p21 affecting the tumor suppressors CHD5 and CDKN2A, respectively, and gains in 1q affecting the prominent oncogene AKT3. The identified somatic nucleotide variants (SNVs) involve few core signaling pathways despite high genetic heterogeneity with diverse mutational spectra: Chromatin remodeling, the cell cycle, and DNA damage/repair. An OncoKB search identifies putative actionable genomic targets in 35% of the cases (6/17), including recurrent missense mutations of the FGFR2 gene in biliary ITPNs (2/11, 18%). Our results show that somatic SNV in classical cancer genes, typically associated with pancreato-biliary carcinogenesis, were absent (KRAS, IDH1/2, GNAS, and others) to rare (TP53 and SMAD4, 6%, respectively) in ITPNs. Mutational signature pattern analysis reveals a predominance of an age-related pattern. Our findings highlight that biliary ITPN and classical cholangiocarcinoma display commonalities, in particular mutations in genes of the chromatin remodeling pathway, and appear, therefore, more closely related than pancreatic ITPN and classical pancreatic ductal adenocarcinoma., German Research Foundation (DFG)
- Published
- 2021
10. Prognostic Factors for Wilms Tumor Recurrence: A Review of the Literature
- Author
-
Alissa Groenendijk, Rajkumar Venkatramani, Ronald R. de Krijger, Norbert Graf, Jesper Brok, James I. Geller, Marry M. van den Heuvel-Eibrink, Jarno Drost, Annelies M. C. Mavinkurve-Groothuis, Filippo Spreafico, Christian Rübe, J. Godzinski, Daniela Perotti, and Harm van Tinteren
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,Prognostic variable ,medicine.medical_specialty ,recurrence ,Review ,Loss of heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Overall survival ,medicine ,Copy-number variation ,RC254-282 ,Treatment regimen ,business.industry ,Advanced stage ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Wilms tumor ,Wilms' tumor ,medicine.disease ,Tumor recurrence ,030104 developmental biology ,pediatric ,030220 oncology & carcinogenesis ,prognosis ,business - Abstract
Simple Summary A Wilms tumor is a childhood kidney tumor. In high-income countries, 90% of patients with this tumor survive. However, the tumor recurs in 15% of patients. It is important to identify the patients at risk of recurrence in order to adjust treatment in such a way that recurrence may potentially be prevented. However, we are currently unable to determine precisely which patients are at risk of recurrence. Therefore, we present an overview of factors that influence the risk of recurrence, also known as prognostic factors. These factors range from patient-, tumor- and treatment-related characteristics to geographic and socioeconomic factors. In addition to these factors, biological markers, such as genetic alterations, should be studied more intensively as these markers may be able to better identify patients at risk of tumor recurrence. Abstract In high-income countries, the overall survival of children with Wilms tumors (WT) is ~90%. However, overall, 15% of patients experience tumor recurrence. The adverse prognostic factors currently used for risk stratification (advanced stage, high risk histology, and combined loss of heterozygosity at 1p and 16q in chemotherapy-naïve WTs) are present in only one third of these cases, and the significance of these factors is prone to change with advancing knowledge and improved treatment regimens. Therefore, we present a comprehensive, updated overview of the published prognostic variables for WT recurrence, ranging from patient-, tumor- and treatment-related characteristics to geographic and socioeconomic factors. Improved first-line treatment regimens based on clinicopathological characteristics and advancing knowledge on copy number variations unveil the importance of further investigating the significance of biological markers for WT recurrence in international collaborations.
- Published
- 2021
11. Next Generation Cytogenetics in Myeloid Hematological Neoplasms: Detection of CNVs and Translocations
- Author
-
Patricia Font, Mariana Bastos-Oreiro, Paula Muñiz, Ismael Buño, Carolina Martínez-Laperche, Javier Anguita, Julia Suárez-González, Mónica Ballesteros, María Chicano, Cristina Andrés-Zayas, Mi Kwon, Diego Carbonell, Mercedes Ballesteros-Culebras, Gabriela Rodríguez-Macías, José Luis Díez-Martín, and Sergio Lois
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Chromosomal translocation ,Biology ,Trisomy 8 ,Article ,cytogenetics ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Copy-number variation ,Multiplex ligation-dependent probe amplification ,RC254-282 ,Genetics ,Cytogenetics ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,myeloid neoplasms ,Mutational analysis ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,NGS ,Hematological neoplasm - Abstract
Conventional cytogenetics are the gold standard for the identification of chromosomal alterations recurrent in myeloid neoplasms. Some next-generation sequencing (NGS) panels are designed for the detection of copy number variations (CNV) or translocations, however, their use is far from being widespread. Here we report on the results of a commercial panel including frequent mutations, CNVs and translocations in myeloid neoplasms. Frequent chromosomal alterations were analyzed by NGS in 135 patients with myeloid neoplasms and three with acute lymphoblastic leukemia. NGS analysis was performed using the enrichment-capture Myeloid Neoplasm-GeneSGKit (Sistemas Genómicos, Spain) gene panel including 35 genes for mutational analysis and frequent CNVs and translocations. NGS results were validated with cytogenetics and/or MLPA when possible. A total of 66 frequent alterations included in NGS panel were detected, 48 of them detected by NGS and cytogenetics. Ten of them were observed only by cytogenetics (mainly trisomy 8), and another eight only by NGS (mainly deletion of 12p). Aside from this, 38 secondary CNVs were detected in any of the genes included mainly for mutational analysis. NGS represents a reliable complementary source of information for the analysis of CNVs and translocations. Moreover, NGS could be a useful tool for the detection of alterations not observed by conventional cytogenetics.
- Published
- 2021
12. Mutation Profile of Aggressive Pheochromocytoma and Paraganglioma with Comparison of TCGA Data
- Author
-
Tae-Yon Sung, Eun-Gyoung Hong, Won Gu Kim, Yun Mi Choi, Dong Eun Song, Se Jin Jang, Jinyeong Lim, Min Ji Jeon, Sung-Min Chun, Ji-Young Lee, and Yu-Mi Lee
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,SDHB ,Article ,Germline ,Pheochromocytoma ,03 medical and health sciences ,paraganglioma ,0302 clinical medicine ,Germline mutation ,Paraganglioma ,Internal medicine ,medicine ,HRAS ,Copy-number variation ,DNA mutation analysis ,high-throughput nucleotide sequencing ,RC254-282 ,business.industry ,aggressive pheochromocytoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation (genetic algorithm) ,prognosis ,business - Abstract
Simple Summary Pheochromocytomas and paragangliomas (PPGLs) are neuroendocrine tumors arising from chromaffin cells of the adrenal medulla, or extra-adrenal paraganglia, respectively. In PPGLs, germline or somatic mutations in one of the known susceptibility genes are identified in up to 60% patients. Recent WHO classification defines that all PPGLs can have metastatic potential. The term, ‘malignant’ is replaced with ‘metastatic’ in this group of tumors. However, the peculiar genetic events that drive the aggressive behavior, including metastasis in PPGLs are yet poorly understood. We performed targeted next-generation sequencing analysis to characterize the mutation profile in fifteen aggressive PPGL patients and compared accessible data of aggressive PPGLs from The Cancer Genome Atlas (TCGA) with findings of our cohort. This targeted mutational analysis might expand the mutation profile of aggressive PPGLs, and may also be useful in detecting the possible experimental therapeutic options or predicting poor prognosis. Abstract In pheochromocytoma and paraganglioma (PPGL), germline or somatic mutations in one of the known susceptibility genes are identified in up to 60% patients. However, the peculiar genetic events that drive the aggressive behavior including metastasis in PPGL are poorly understood. We performed targeted next-generation sequencing analysis to characterize the mutation profile in fifteen aggressive PPGL patients and compared accessible data of aggressive PPGLs from The Cancer Genome Atlas (TCGA) with findings of our cohort. A total of 115 germline and 34 somatic variants were identified with a median 0.58 per megabase tumor mutation burden in our cohort. The most frequent mutation was SDHB germline mutation (27%) and the second frequent mutations were somatic mutations for SETD2, NF1, and HRAS (13%, respectively). Patients were subtyped into three categories based on the kind of mutated genes: pseudohypoxia (n = 5), kinase (n = 5), and unknown (n = 5) group. In copy number variation analysis, deletion of chromosome arm 1p harboring SDHB gene was the most frequently observed. In our cohort, SDHB mutation and pseudohypoxia subtype were significantly associated with poor overall survival. In conclusion, subtyping of mutation profile can be helpful in aggressive PPGL patients with heterogeneous prognosis to make relevant follow-up plan and achieve proper treatment.
- Published
- 2021
13. Cell-Free DNA Analysis by Whole-Exome Sequencing for Hepatocellular Carcinoma: A Pilot Study in Thailand
- Author
-
Pongserath Sirichindakul, Pattapon Kunadirek, Intawat Nookaew, Thidathip Wongsurawat, Nutcha Pinjaroen, Pisit Tangkijvanich, Natthaya Chuaypen, and Piroon Jenjaroenpun
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.disease_cause ,Germline ,Article ,cell-free DNA ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Internal medicine ,Medicine ,Copy-number variation ,whole-exome sequencing ,neoplasms ,Exome sequencing ,RC254-282 ,Oxford Nanopore Technologies ,Mutation ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,hepatocellular carcinoma ,medicine.disease ,Thailand ,digestive system diseases ,030104 developmental biology ,copy-number variants ,Cell-free fetal DNA ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Biomarker (medicine) ,biomarker ,business - Abstract
Cell-free DNA (cfDNA) has been used as a non-invasive biomarker for detecting cancer-specific mutations. However, the mutational profile of cfDNA in Thai patients with hepatocellular carcinoma (HCC) has not been investigated. Here, we demonstrated the utility of using whole-exome sequencing (WES) of cfDNA to define the somatic mutation profiles of HCC in Thai patients. The comprehensive profile of cfDNA was determined with WES to identify variants in matched cfDNA and germline DNA from 30 HCC patients in Thailand who underwent nonoperative therapies. The level of cfDNA was higher in HCC patients compared with chronic hepatitis patients (p-value <, 0.001). Single nucleotide variants were present in somatic genes in cfDNA, including in ZNF814 (27%), HRNR (20%), ZNF492 (20%), ADAMTS12 (17%), FLG (17%), OBSCN (17%), TP53 (17%), and TTN (17%). These same mutations were matched to HCC mutation data from The Cancer Genome Atlas (TCGA) and a previous Thai HCC study. The co-occurrence of HRNR and TTN mutations in cfDNA was associated with shorter overall survival in HCC patients (hazard ratio = 1.60, p-value = 0.0196). These findings indicate that the mutational profile of cfDNA accurately reflected that of HCC tissue and suggest that cfDNA could serve as a useful biomarker for diagnosis and prognosis in Thai HCC patients. In addition, we demonstrated the use of the pocket-sized sequencer of Oxford Nanopore Technology to detect copy-number variants in HCC tissues that could be applied for onsite clinical detection/monitoring of HCC.
- Published
- 2021
14. Rare Germline Variants in Chordoma-Related Genes and Chordoma Susceptibility
- Author
-
Brian D. Carter, Yanzi Xiao, Amy Hutchinson, Xiaohong R. Yang, Mary L. McMaster, Chuzhong Li, Songbai Gui, Laura Beane-Freeman, Jiwei Bai, Alisa M. Goldstein, Nirav N Shah, Sally Yepes, Belynda Hicks, Dilys M. Parry, Mingyi Wang, Hela Koka, Kristine Jones, Neal D. Freedman, Meredith Yeager, Yazhuo Zhang, Aurelie Vogt, Bin Zhu, and Stephen J. Chanock
- Subjects
0301 basic medicine ,musculoskeletal diseases ,Cancer Research ,notochord development ,Population ,Biology ,Germline ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Genetic predisposition ,Missense mutation ,Copy-number variation ,education ,chordoma ,Exome sequencing ,RC254-282 ,Genetics ,education.field_of_study ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,cancer susceptibility ,Skull Base Chordoma ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,germline variants ,WES ,Chordoma ,WGS - Abstract
Background: Chordoma is a rare bone cancer with an unknown etiology. TBXT is the only chordoma susceptibility gene identified to date, germline single nucleotide variants and copy number variants in TBXT have been associated with chordoma susceptibility in familial and sporadic chordoma. However, the genetic susceptibility of chordoma remains largely unknown. In this study, we investigated rare germline genetic variants in genes involved in TBXT/chordoma-related signaling pathways and other biological processes in chordoma patients from North America and China. Methods: We identified variants that were very rare in general population and internal control datasets and showed evidence for pathogenicity in 265 genes in a whole exome sequencing (WES) dataset of 138 chordoma patients of European ancestry and in a whole genome sequencing (WGS) dataset of 80 Chinese patients with skull base chordoma. Results: Rare and likely pathogenic variants were identified in 32 of 138 European ancestry patients (23%), including genes that are part of notochord development, PI3K/AKT/mTOR, Sonic Hedgehog, SWI/SNF complex and mesoderm development pathways. Rare pathogenic variants in COL2A1, EXT1, PDK1, LRP2, TBXT and TSC2, among others, were also observed in Chinese patients. Conclusion: We identified several rare loss-of-function and predicted deleterious missense variants in germline DNA from patients with chordoma, which may influence chordoma predisposition and reflect a complex susceptibility, warranting further investigation in large studies.
- Published
- 2021
15. Genotype-Phenotype Correlations in Neurofibromatosis Type 1: A Single-Center Cohort Study
- Author
-
Guido Morcaldi, Cristina Chelleri, Michele Iacomino, Marco Pavanello, Federico Zara, Maria Cristina Diana, Claudia Milanaccio, Antonio Verrico, Patrizia De Marco, Valeria Capra, Renata Bocciardi, Paolo Scudieri, Carlo Minetti, Maria Luisa Garrè, Marco Di Duca, Pasquale Striano, Francesca Madia, M. Traverso, Irene Schiavetti, Antonella Riva, Gianluca Piccolo, Gianluca Piatelli, Gianmaria Viglizzo, Vincenzo Salpietro, Marcello Scala, Francesco Caroli, Andrea Accogli, and Simona Baldassari
- Subjects
0301 basic medicine ,Cancer Research ,cDNA sequencing ,030105 genetics & heredity ,lcsh:RC254-282 ,Article ,Frameshift mutation ,genotype–phenotype correlations ,03 medical and health sciences ,splicing ,neurofibromatosis type I ,medicine ,stop-gain ,Missense mutation ,Brain tumor ,CDNA sequencing ,Genotype–phenotype correlations ,MLPA ,Neurofibromatosis type I ,NF1 ,NGS ,Splicing ,Stop-gain ,Multiplex ligation-dependent probe amplification ,Copy-number variation ,Neurofibromatosis ,Genetic testing ,Genetics ,medicine.diagnostic_test ,business.industry ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,business ,brain tumor ,Cohort study - Abstract
Neurofibromatosis type 1 (NF1) is a proteiform genetic condition caused by pathogenic variants in NF1 and characterized by a heterogeneous phenotypic presentation. Relevant genotype–phenotype correlations have recently emerged, but only few pertinent studies are available. We retrospectively reviewed clinical, instrumental, and genetic data from a cohort of 583 individuals meeting at least 1 diagnostic National Institutes of Health (NIH) criterion for NF1. Of these, 365 subjects fulfilled ≥2 NIH criteria, including 235 pediatric patients. Genetic testing was performed through cDNA-based sequencing, Next Generation Sequencing (NGS), and Multiplex Ligation-dependent Probe Amplification (MLPA). Uni- and multivariate statistical analysis was used to investigate genotype–phenotype correlations. Among patients fulfilling ≥ 2 NIH criteria, causative single nucleotide variants (SNVs) and copy number variations (CNVs) were detected in 267/365 (73.2%) and 20/365 (5.5%) cases. Missense variants negatively correlated with neurofibromas (p = 0.005). Skeletal abnormalities were associated with whole gene deletions (p = 0.05) and frameshift variants (p = 0.006). The c.3721C>, T, p.(R1241*) variant positively correlated with structural brain alterations (p = 0.031), whereas Lisch nodules (p = 0.05) and endocrinological disorders (p = 0.043) were associated with the c.6855C>, A, p.(Y2285*) variant. We identified novel NF1 genotype–phenotype correlations and provided an overview of known associations, supporting their potential relevance in the implementation of patient management.
- Published
- 2021
- Full Text
- View/download PDF
16. A systematic review of the use of circulating cell-free DNA dynamics to monitor response to treatment in metastatic breast cancer patients
- Author
-
Teoman Deger, Saskia M. Wilting, Stefan Sleijfer, Elisabeth M. Jongbloed, Agnes Jager, and John W.M. Martens
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Treatment response ,lcsh:RC254-282 ,03 medical and health sciences ,liquid biopsies ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Internal medicine ,medicine ,Copy-number variation ,circulating tumor DNA ,business.industry ,Cancer ,treatment response ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Response to treatment ,Metastatic breast cancer ,Circulating Cell-Free DNA ,030104 developmental biology ,Circulating tumor DNA ,030220 oncology & carcinogenesis ,Clinical validity ,Systematic Review ,metastatic breast cancer ,business - Abstract
Simple Summary Currently, the most commonly used method to monitor response to treatment in metastatic breast cancer patients is by radiological imaging. However, these imaging techniques are relatively insensitive and give little to no insight into biological tumor characteristics that might be relevant for the choice of treatment. Circulating tumor DNA (ctDNA), released by tumor cells into the blood of cancer patients, can be used to overcome these shortcomings. Besides the fact that specific alterations are known to predict response to treatment and development of resistance, the total amount of ctDNA is believed to reflect the proliferation rate of the tumor, suggesting ctDNA levels can be used as a general tool to evaluate treatment response. Different methods are available to measure ctDNA primarily based on detection of cancer-specific somatic mutations, DNA methylation, and copy number variations. In this review we have critically analyzed recently published studies using blood-derived ctDNA of metastatic breast cancer patients on multiple time points to monitor disease response in respect to analytical validity and clinical utility. Abstract Monitoring treatment response in metastatic breast cancer currently consists mainly of radiological and clinical assessments. These methods have high inter-observer variation, suboptimal sensitivity to determine response to treatment and give little insight into the biological characteristics of the tumor. Assessing circulating tumor DNA (ctDNA) over time could be employed to address these limitations. Several ways to quantify and characterize ctDNA exist, based on somatic mutations, copy number variations, methylation, and global circulating cell-free DNA (cfDNA) fragment sizes and concentrations. These methods are being explored and technically validated, but to date none of these methods are applied clinically. We systematically reviewed the literature on the use of quantitative ctDNA measurements over time to monitor response to systemic therapy in patients with metastatic breast cancer. Cochrane, Embase, PubMed and Google Scholar databases were searched to find studies focusing on the use of cfDNA to longitudinally monitor treatment response in advanced breast cancer patients until October 2020. This resulted in a total of 33 studies which met the inclusion criteria. These studies were heterogeneous in (pre-)processing procedures, applied techniques and design. An association between ctDNA and treatment response was found in most of the included studies, independent of the applied assay. To implement ctDNA-based response monitoring into daily clinical practice for metastatic breast cancer patients, sample (pre-) processing procedures need to be standardized and large prospectively collected sample cohorts with well annotated clinical follow-up are required to establish its clinical validity.
- Published
- 2021
17. Identification and Validation of an Immune-Associated RNA-Binding Proteins Signature to Predict Clinical Outcomes and Therapeutic Responses in Glioma Patients
- Author
-
Yimin Li, Qian Liu, Minfeng Shu, and Ruotong Tian
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,RNA-binding protein ,immune microenvironment ,Human leukocyte antigen ,risk score ,chemotherapy ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Germline mutation ,tumor-infiltrating immune cells ,Glioma ,glioma ,Medicine ,prognostic model ,Copy-number variation ,Chemotherapy ,business.industry ,Immunotherapy ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,immunotherapy ,business - Abstract
Simple Summary Both of tumor-infiltrating immune cells and the RNA-binding proteins (RBPs) that are able to mediate immune infiltration contribute to the prognosis of patients with glioma. However, immune-associated RBPs in glioma remain unexplored. In this study, we developed a method to identify RBPs associated with immune infiltration in glioma and 216 RBPs were defined as immune-associated RBPs. Among them, eight RBPs were selected to construct a risk signature that proved to be a novel and independent prognostic factor. Higher risk scores meant worse overall survival and higher expression of human leukocyte antigen and immune checkpoints. Additionally, analyses of pathway enrichment, somatic mutation, copy number variations, and immuno-/chemotherapeutic response prediction were performed to evaluate the differences between high- and low-risk groups. Generally, we demonstrated an eight immune-associated RBPs prognostic signature that was valuable in predicting the survival of glioma patients and directing immunotherapy and chemotherapy. Abstract The prognosis of patients with glioma is largely related to both the tumor-infiltrating immune cells and the expression of RNA-binding proteins (RBPs) that are able to regulate various pro-inflammatory and oncogenic mediators. However, immune-associated RBPs in glioma remain unexplored. In this study, we captured patient data from The Cancer Genome Atlas (TCGA) and divided them into two immune subtype groups according to the difference in infiltration of immune cells. After differential expression and co-expression analysis, we identified 216 RBPs defined as immune-associated RBPs. After narrowing down processes, eight RBPs were selected out to construct a risk signature that proven to be a novel and independent prognostic factor. The patients were divided into high- and low-risk groups on the basis of risk score. Higher risk scores meant worse overall survival and higher expression of human leukocyte antigen and immune checkpoints such as PD1 and CTLA4. In addition, analyses of pathway enrichment, somatic mutation, copy number variations and immuno-/chemotherapeutic response prediction were performed in high- and low-risk groups and compared with each other. For the first time, we demonstrated a novel signature composed of eight immune-associated RBPs that was valuable in predicting the survival of glioma patients and directing immunotherapy and chemotherapy.
- Published
- 2021
18. Genomic Signature of Oral Squamous Cell Carcinomas from Non-Smoking Non-Drinking Patients
- Author
-
Antony W. Burgess, Michael McCullough, Oliver M. Sieber, Kendrick Koo, Tim A. Iseli, Christopher Angel, David Wiesenfeld, and Dmitri Mouradov
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lymphovascular invasion ,EGFR ,Perineural invasion ,PIK3CA ,medicine.disease_cause ,lcsh:RC254-282 ,tobacco ,Article ,DNA copy number ,CDKN2A ,03 medical and health sciences ,0302 clinical medicine ,human papilloma virus ,Internal medicine ,medicine ,TP53 ,Copy-number variation ,targeted sequencing ,neoplasms ,Univariate analysis ,Mutation ,business.industry ,alcohol ,Cancer ,Genomic signature ,oral cancer ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,stomatognathic diseases ,030104 developmental biology ,030220 oncology & carcinogenesis ,business - Abstract
Molecular alterations in 176 patients with oral squamous cell carcinomas (OSCC) were evaluated to delineate differences in non-smoking non-drinking (NSND) patients. Somatic mutations and DNA copy number variations (CNVs) in a 68-gene panel and human papilloma virus (HPV) status were interrogated using targeted next-generation sequencing. In the entire cohort, TP53 (60%) and CDKN2A (24%) were most frequently mutated, and the most common CNVs were EGFR amplifications (9%) and deletions of BRCA2 (5%) and CDKN2A (4%). Significant associations were found for TP53 mutation and nodal disease, lymphovascular invasion and extracapsular spread, CDKN2A mutation or deletion with advanced tumour stage, and EGFR amplification with perineural invasion and extracapsular spread. PIK3CA mutation, CDKN2A deletion, and EGFR amplification were associated with worse survival in univariate analyses (p <, 0.05 for all comparisons). There were 59 NSND patients who tended to be female and older than patients who smoke and/or drink, and showed enrichment of CDKN2A mutations, EGFR amplifications, and BRCA2 deletions (p <, 0.05 for all comparisons), with a younger subset showing higher mutation burden. HPV was detected in three OSCC patients and not associated with smoking and drinking habits. NSND OSCC exhibits distinct genomic profiles and further exploration to elucidate the molecular aetiology in these patients is warranted.
- Published
- 2021
- Full Text
- View/download PDF
19. Distinct Signatures of Genomic Copy Number Variants Define Subgroups of Merkel Cell Carcinoma Tumors
- Author
-
Clayton Green, David H. Kim, Isaac Brownell, Natasha T. Hill, Klaus J. Busam, and Emily Y. Chu
- Subjects
0301 basic medicine ,Cancer Research ,virus negative Merkel cell carcinoma and copy number variant ,Merkel cell polyomavirus ,virus positive Merkel cell carcinoma ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Merkel cell carcinoma ,medicine ,Copy-number variation ,Gene ,Genetics ,biology ,Cancer ,food and beverages ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,biology.organism_classification ,medicine.disease ,genomic DNA ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Skin cancer - Abstract
Simple Summary Cancer results from genetic changes in cells. These changes are often mutations that alter the DNA sequence of critical genes. However, duplications and deletions in cancer-related genes can also contribute to malignant transformation. In this study we use Nanostring technology to assess DNA copy number changes in samples of Merkel cell carcinoma (MCC), a rare and aggressive neuroendocrine skin tumor. We were able to identify recurrent amplifications and deletions in cancer-related genes. We also found that MCC tumors grouped into three distinct copy number variant profiles. The first group consisted of tumors with multiple deletions. The second group contained tumors with low levels of genomic structural alterations. The last group comprised tumors containing multiple amplifications. Our study suggests that most MCC tumors are associated with deletions in cancer-related genes or are lacking in copy number changes, whereas a small percentage of tumors are associated with genomic amplifications. Abstract Merkel cell carcinoma (MCC) is a rare, aggressive neuroendocrine skin cancer. Most MCC tumors contain integrated Merkel cell polyomavirus DNA (virus-positive MCC, VP-MCC) and carry a low somatic mutation burden whereas virus-negative MCC (VN-MCC) possess numerous ultraviolet-signature mutations. In contrast to viral oncogenes and sequence mutations, little is known about genomic structural variants in MCC. To identify copy number variants in commonly altered genes, we analyzed genomic DNA from 31 tumor samples using the Nanostring nCounter copy number cancer panel. Unsupervised clustering revealed three tumor groups with distinct genomic structural variant signatures. The first cluster was characterized by multiple recurrent deletions in genes such as RB1 and WT1. The second cluster contained eight VP-MCC and displayed very few structural variations. The final cluster contained one VP-MCC and four VN-MCC with predominantly genomic amplifications in genes like MDM4, SKP2, and KIT and deletions in TP53. Overall, VN-MCC contained more structure variation than VP-MCC but did not cluster separately from VP-MCC. The observation that most MCC tumors demonstrate a deletion-dominated structural group signature, independent of virus status, suggests a shared pathophysiology among most VP-MCC and VN-MCC tumors.
- Published
- 2021
20. Spatial Heterogeneity in Large Resected Diffuse Large B-Cell Lymphoma Bulks Analysed by Massively Parallel Sequencing of Multiple Synchronous Biopsies
- Author
-
Sandro M. Wagner, Daniel Neureiter, Teresa Magnes, Thomas Melchardt, Markus Steiner, Alexander Egle, Eckhard Klieser, Gabriel Rinnerthaler, Richard Greil, Florian Huemer, Nadja Zaborsky, Aaron R. Thorner, Konstantin Schlick, and Lukas Weiss
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Lymph node biopsy ,diffuse large B-cell lymphoma ,clonal evolution ,Biology ,Somatic evolution in cancer ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,lymphomagenesis ,hemic and lymphatic diseases ,medicine ,PTEN ,Copy-number variation ,Massive parallel sequencing ,medicine.diagnostic_test ,massively parallel sequencing ,spatial heterogeneity ,CD79B ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Lymphoma ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Diffuse large B-cell lymphoma - Abstract
Simple Summary Substantial genetic heterogeneity was described within large tumour masses of several cancer entities. However, this topic has not been addressed in patients with diffuse large B-cell lymphoma (DLBCL). Therefore, we collected multiple biopsies of twelve patients who had diagnostic or therapeutic resections of large lymphoma bulks and analysed 213 genes known to be important for lymphoma biology. The biopsies of each patient were compared to investigate the spatial heterogeneity in DLBCL. Ten out of twelve patients had discordant mutations which were not present in all of their biopsies and similar results were seen by the analysis of copy number variants. Some of the involved genes have a known prognostic and therapeutic relevance in DLBCL. This shows that single biopsies underestimate the complexity of the disease and might overlook possible mechanisms of resistance and therapeutic targets. Abstract Diffuse large B-cell lymphoma (DLBCL) usually needs to be treated immediately after diagnosis from a single lymph node biopsy. However, several reports in other malignancies have shown substantial spatial heterogeneity within large tumours. Therefore, we collected multiple synchronous biopsies of twelve patients that had diagnostic or therapeutic resections of large lymphoma masses and performed next-generation sequencing of 213 genes known to be important for lymphoma biology. Due to the high tumour cell content in the biopsies, we were able to detect several mutations which were present with a stable allelic frequency across all the biopsies of each patient. However, ten out of twelve patients had spatially discordant mutations and similar results were found by the analysis of copy number variants. The median Jaccard similarity coefficient, a measure of the similarity of a sample set was 0.77 (range 0.47–1), and some of the involved genes such as CARD11, CD79B, TP53, and PTEN have a known prognostic or therapeutic relevance in DLBCL. This shows that single biopsies underestimate the complexity of the disease and might overlook possible mechanisms of resistance and therapeutic targets. In the future, the broader application of liquid biopsies will have to overcome these obstacles.
- Published
- 2021
21. Genetic Analysis of Multiple Myeloma Identifies Cytogenetic Alterations Implicated in Disease Complexity and Progression
- Author
-
Guido Tricot, Fangping Chen, Hongwei Xu, Benjamin W. Darbro, John D. Shaughnessy, Erik Wendlandt, Fenghuang Zhan, Gregory S. Thomas, and Can Li
- Subjects
0301 basic medicine ,Cancer Research ,Computational biology ,Disease ,Biology ,Genetic analysis ,Genome ,lcsh:RC254-282 ,Article ,cytogenetics ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Copy-number variation ,protein network signatures ,Multiple myeloma ,Genetic heterogeneity ,gene expression profiles ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Gene expression profiling ,multiple myeloma ,030104 developmental biology ,copy number variations ,Oncology ,030220 oncology & carcinogenesis ,DNA microarray - Abstract
Simple Summary Multiple myeloma (MM) is the second most common hematological neoplasia with a high incidence in elderly populations. The disease is characterized by a severe chaos of genomic abnormality. Comprehensive examinations of myeloma cytogenetics are needed for better understanding of MM and potential application to the development of novel therapeutic regiments. Here we utilized gene expression profiling and CytoScan HD genomic arrays to investigate molecular alterations in myeloma leading to disease progression and poor clinical outcomes. We demonstrates that genetic abnormalities within MM patients exhibit unique protein network signatures that can be exploited for implementation of existing therapies targeting key pathways and the development of novel therapeutics. Abstract Multiple myeloma (MM) is a genetically heterogeneous disease characterized by genomic chaos making it difficult to distinguish driver from passenger mutations. In this study, we integrated data from whole genome gene expression profiling (GEP) microarrays and CytoScan HD high-resolution genomic arrays to integrate GEP with copy number variations (CNV) to more precisely define molecular alterations in MM important for disease initiation, progression and poor clinical outcome. We utilized gene expression arrays from 351 MM samples and CytoScan HD arrays from 97 MM samples to identify eight CNV events that represent possible MM drivers. By integrating GEP and CNV data we divided the MM into eight unique subgroups and demonstrated that patients within one of the eight distinct subgroups exhibited common and unique protein network signatures that can be utilized to identify new therapeutic interventions based on pathway dysregulation. Data also point to the central role of 1q gains and the upregulated expression of ANP32E, DTL, IFI16, UBE2Q1, and UBE2T as potential drivers of MM aggressiveness. The data presented here utilized a novel approach to identify potential driver CNV events in MM, the creation of an improved definition of the molecular basis of MM and the identification of potential new points of therapeutic intervention.
- Published
- 2021
22. Molecular Changes in Retinoblastoma beyond RB1: Findings from Next-Generation Sequencing
- Author
-
Michael F. Berger, Diana Mandelker, David H. Abramson, Michael Walsh, Allison Richards, Mark T.A. Donoghue, Ira J. Dunkel, and Jasmine H. Francis
- Subjects
0301 basic medicine ,Cancer Research ,Somatic cell ,Biology ,lcsh:RC254-282 ,DNA sequencing ,Germline ,Article ,retinoblastoma ,Loss of heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,medicine ,Copy-number variation ,BCOR ,Gene ,Genetics ,Retinoblastoma ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,eye diseases ,030104 developmental biology ,copy number variations ,Oncology ,030220 oncology & carcinogenesis ,vitreous seeds ,next-generation sequencing - Abstract
This investigation uses hybridization capture-based next-generation sequencing to deepen our understanding of genetics that underlie retinoblastoma. Eighty-three enucleated retinoblastoma specimens were evaluated using a MSK-IMPACT clinical next-generation sequencing panel to evaluate both somatic and germline alterations. Somatic copy number variations (CNVs) were also identified. Genetic profiles were correlated to clinicopathologic characteristics. RB1 inactivation was found in 79 (97.5%) patients. All specimens had additional molecular alterations. The most common non-RB1 gene alteration was BCOR in 19 (22.9%). Five (11.0%) had pathogenic germline mutations in other non-RB1 cancer predisposition genes. Significant clinicopathologic correlations included: vitreous seeds associated with 1q gains and 16q loss of heterozygosity (BH-corrected p-value = 0.008, 0.004, OR = 12.6, 26.7, respectively). BCOR mutations were associated with poor prognosis, specifically metastases-free survival (MFS) (nominal p-value 0.03). Furthermore, retinoblastoma patients can have non-RB1 germline mutations in other cancer-associated genes. No two specimens had the identical genetic profile, emphasizing the individuality of tumors with the same clinical diagnosis.
- Published
- 2021
23. Homologous Recombination Repair Mechanisms in Serous Endometrial Cancer
- Author
-
Jenny-Maria Jönsson, Maria Bååth, Ida Björnheden, Irem Durmaz Sahin, Anna Måsbäck, and Ingrid Hedenfalk
- Subjects
copy-number variation ,PARP inhibition ,DNA repair ,serous endometrial cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,homologous recombination repair deficiency ,lcsh:RC254-282 ,Article - Abstract
Serous endometrial cancer (SEC) resembles high-grade serous ovarian cancer (HGSOC) genetically and clinically, with recurrent copy number alterations, TP53 mutations and a poor prognosis. Thus, SEC patients may benefit from targeted treatments used in HGSOC, e.g., PARP inhibitors. However, the preclinical and clinical knowledge about SEC is scarce, and the exact role of defective DNA repair in this tumor subgroup is largely unknown. We aimed to outline the prevalence of homologous recombination repair deficiency (HRD), copy-number alterations, and somatic mutations in SEC. OncoScan SNP arrays were applied to 19 tumors in a consecutive SEC series to calculate HRD scores and explore global copy-number profiles and genomic aberrations. Copy-number signatures were established and targeted sequencing of 27 HRD-associated genes was performed. All factors were examined in relation to HRD scores to investigate potential drivers of the HRD phenotype. Ten of the 19 SEC tumors (53%) had an HRD score >, 42, considered to reflect an HRD phenotype. Higher HRD score was associated with loss of heterozygosity in key HRD genes, and copy-number signatures associated with non-BRCA1/2 dependent HRD in HGSOC. A high number of SECs display an HRD phenotype. It remains to be elucidated whether this also confers PARP inhibitor sensitivity.
- Published
- 2021
24. Performance of In Silico Prediction Tools for the Detection of Germline Copy Number Variations in Cancer Predisposition Genes in 4208 Female Index Patients with Familial Breast and Ovarian Cancer
- Author
-
Julika Borde, Sandra Schmidt, Malwina Suszynska, Piotr Kozlowski, Jonas Weber, Katarzyna Klonowska, Corinna Ernst, Jan Hauke, Louisa Lepkes, Rita K. Schmutzler, Barbara Wappenschmidt, Mohamad Kayali, Eric Hahnen, and Britta Blümcke
- Subjects
0301 basic medicine ,Cancer Research ,congenital, hereditary, and neonatal diseases and abnormalities ,endocrine system diseases ,PALB2 ,In silico ,CNV ,Biology ,MLH1 ,breast/ovarian cancer susceptibility genes ,lcsh:RC254-282 ,Germline ,Article ,03 medical and health sciences ,0302 clinical medicine ,mental disorders ,PMS2 ,Copy-number variation ,CHEK2 ,multigene panel sequencing ,Genetics ,HBOC ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,MSH2 ,030220 oncology & carcinogenesis - Abstract
The identification of germline copy number variants (CNVs) by targeted next-generation sequencing (NGS) frequently relies on in silico CNV prediction tools with unknown sensitivities. We investigated the performances of four in silico CNV prediction tools, including one commercial (Sophia Genetics DDM) and three non-commercial tools (ExomeDepth, GATK gCNV, panelcn.MOPS) in 17 cancer predisposition genes in 4208 female index patients with familial breast and/or ovarian cancer (BC/OC). CNV predictions were verified via multiplex ligation-dependent probe amplification. We identified 77 CNVs in 76 out of 4208 patients (1.81%), 33 CNVs were identified in genes other than BRCA1/2, mostly in ATM, CHEK2, and RAD51C and less frequently in BARD1, MLH1, MSH2, PALB2, PMS2, RAD51D, and TP53. The Sophia Genetics DDM software showed the highest sensitivity, six CNVs were missed by at least one of the non-commercial tools. The positive predictive values ranged from 5.9% (74/1249) for panelcn.MOPS to 79.1% (72/91) for ExomeDepth. Verification of in silico predicted CNVs is required due to high frequencies of false positive predictions, particularly affecting target regions at the extremes of the GC content or target length distributions. CNV detection should not be restricted to BRCA1/2 due to the relevant proportion of CNVs in further BC/OC predisposition genes.
- Published
- 2021
- Full Text
- View/download PDF
25. Assessment of HER2 Protein Overexpression and Gene Amplification in Renal Collecting Duct Carcinoma: Therapeutic Implication
- Author
-
Giuseppe Simone, Vito Michele Fazio, Rui Henrique, M. T. Landi, Steno Sentinelli, Enrico Munari, Nazareno Suardi, Manuela Costantini, Michele Gallucci, Umberto Anceschi, Umberto Capitanio, Aldo Brassetti, Roberta Lucianò, Carmen Jerónimo, Liborio Torregrossa, Carla Azzurra Amoreo, Sara Petronilho, Greta Alì, and Maria Luana Poeta
- Subjects
0301 basic medicine ,Cancer Research ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,Collecting duct carcinoma ,0302 clinical medicine ,Biomarker ,FISH ,HER2 ,IHC ,Renal collecting duct carcinoma ,Gene duplication ,medicine ,Copy-number variation ,skin and connective tissue diseases ,Kidney ,business.industry ,Cancer ,Histology ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,renal collecting duct carcinoma ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Immunohistochemistry ,Biomarker (medicine) ,biomarker ,business - Abstract
Collecting duct carcinoma (CDC) is rare and aggressive histology of kidney cancers. Although different therapeutic approaches have been tested, the 2-year survival remains very poor. Since CDC exhibits overlapping features with urothelial carcinoma, the analysis of shared molecular alterations could provide new insights into the understanding of this rare disease and also therapeutic options. We collected 26 CDC cases, and we assessed HER2 protein expression by immunohistochemistry (IHC) and gene amplification by fluorescence in-situ hybridization (FISH) according to 2018 ASCO/CAP HER2-testing recommendations. Six out of twenty-six (23%) tumors showed HER2 positive staining. In particular, 3+ score was present in 2/6 cases (33%), 2+ in 3/6 cases (50%) and 1+ in 1/6 cases (17%). The 6 HER2+ tumors were also analyzed by FISH to assess gene copy number. One out of six CDC with IHC 3+ was also HER2 amplified, showing an average HER2 copy number &ge, 4.0 (10.85) and a HER2/CEP17 ratio &ge, (5.63), while the 5/6 cases were HER2 negative. Based on the 2018 ASCO/CAP guidelines overall, 2/26 CDC cases (8%) were HER2+. The present study provides evidence for testing, in future studies, HER2 to assess its clinical value as a novel target for the treatment of this highly malignant cancer.
- Published
- 2020
26. Systems Biology Approach Identifies Prognostic Signatures of Poor Overall Survival and Guides the Prioritization of Novel BET-CHK1 Combination Therapy for Osteosarcoma
- Author
-
L. Daniel Wurtz, Savannah Dyer, Erika A. Dobrota, Jamie L. Renbarger, Pankita H. Pandya, Anthony L. Sinn, Michael J. Ferguson, Mary E. Murray, Farinaz Barghi, Khadijeh Bijangi-Vishehsaraei, Lijun Cheng, Harlan E. Shannon, Barbara J. Bailey, Kathryn L. Coy, Sandeep Batra, Courtney N. Young, Adily N. Elmi, Lang Li, Jeremiah Shultz, Quinton Thompson, Todd E. Bertrand, Eric A. Albright, M. Reza Saadatzadeh, Melissa A. Trowbridge, Karen E. Pollok, Shan Tang, Jixin Ding, and Mark S. Marshall
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Combination therapy ,Systems biology ,CHK1 ,Disease ,MYC ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,precision genomics ,Internal medicine ,osteosarcoma ,Gene expression ,Medicine ,Copy-number variation ,BETs ,Young adult ,RAD21 ,business.industry ,biomarkers ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,molecular signature ,Bromodomain ,030104 developmental biology ,030220 oncology & carcinogenesis ,Osteosarcoma ,business - Abstract
Osteosarcoma (OS) patients exhibit poor overall survival, partly due to copy number variations (CNVs) resulting in dysregulated gene expression and therapeutic resistance. To identify actionable prognostic signatures of poor overall survival, we employed a systems biology approach using public databases to integrate CNVs, gene expression, and survival outcomes in pediatric, adolescent, and young adult OS patients. Chromosome 8 was a hotspot for poor prognostic signatures. The MYC-RAD21 copy number gain (8q24) correlated with increased gene expression and poor overall survival in 90% of the patients (n = 85). MYC and RAD21 play a role in replication-stress, which is a therapeutically actionable network. We prioritized replication-stress regulators, bromodomain and extra-terminal proteins (BETs), and CHK1, in order to test the hypothesis that the inhibition of BET + CHK1 in MYC-RAD21+ pediatric OS models would be efficacious and safe. We demonstrate that MYC-RAD21+ pediatric OS cell lines were sensitive to the inhibition of BET (BETi) and CHK1 (CHK1i) at clinically achievable concentrations. While the potentiation of CHK1i-mediated effects by BETi was BET-BRD4-dependent, MYC expression was BET-BRD4-independent. In MYC-RAD21+ pediatric OS xenografts, BETi + CHK1i significantly decreased tumor growth, increased survival, and was well tolerated. Therefore, targeting replication stress is a promising strategy to pursue as a therapeutic option for this devastating disease.
- Published
- 2020
27. Whole genome analysis of ovarian granulosa cell tumors reveals tumor heterogeneity and a high- grade tp53-specific subgroup
- Author
-
Hannah S. van Meurs, Joachim Kutzera, Gijs van Haaften, Ronald P. Zweemer, S. T. Paijens, Glen R. Monroe, Petronella O. Witteveen, Christianne A.R. Lok, Ellen Stelloo, Luc R.C.W. van Lonkhuijzen, J. W. Groeneweg, René H.M. Verheijen, Jurgen M.J. Piek, Hans W. Nijman, Geertruida N. Jonges, Joline F. Roze, Obstetrics and Gynaecology, Targeted Gynaecologic Oncology (TARGON), and Translational Immunology Groningen (TRIGR)
- Subjects
0301 basic medicine ,endocrine system ,Cancer Research ,FOXL2 ,Tumor heterogeneity ,Ovarian Granulosa Cell ,Somatic cell ,Biology ,medicine.disease_cause ,Genome ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Ovarian cancer ,medicine ,Copy-number variation ,Chromosome 12 ,Whole genome sequencing ,Mutation ,urogenital system ,FRAMEWORK ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,CANCER ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Granulosa cell tumors ,MONOSOMY 22 ,Cancer research - Abstract
Adult granulosa cell tumors (AGCTs) harbor a somatic FOXL2 c.402C>, G mutation in ~95% of cases and are mainly surgically removed due to limited systemic treatment effect. In this study, potentially targetable genomic alterations in AGCTs were investigated by whole genome sequencing on 46 tumor samples and matched normal DNA. Copy number variant (CNV) analysis confirmed gain of chromosome 12 and 14, and loss of 22. Pathogenic TP53 mutations were identified in three patients with highest tumor mutational burden and mitotic activity, defining a high-grade AGCT subgroup. Within-patient tumor comparisons showed 29&ndash, 80% unique somatic mutations per sample, suggesting tumor heterogeneity. A higher mutational burden was found in recurrent tumors, as compared to primary AGCTs. FOXL2-wildtype AGCTs harbored DICER1, TERT(C228T) and TP53 mutations and similar CNV profiles as FOXL2-mutant tumors. Our study confirms that absence of the FOXL2 c.402C>, G mutation does not exclude AGCT diagnosis. The lack of overlapping variants in targetable cancer genes indicates the need for personalized treatment for AGCT patients.
- Published
- 2020
28. Immune-Omics Networks of CD27, PD1, and PDL1 in Non-Small Cell Lung Cancer
- Author
-
Salvi Singh, Nancy Lan Guo, Qing Ye, and Peter R Qian
- Subjects
RNA interference (RNAi) ,Cancer Research ,medicine.medical_treatment ,DNA copy number variation ,non-small cell lung cancer (NSCLC) ,chemotherapy ,Article ,RNA interference ,Boolean implication networks ,medicine ,Copy-number variation ,Lung cancer ,Gene ,radiotherapy ,RC254-282 ,Chemotherapy ,business.industry ,biomarkers ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,repurposing drugs ,Immunotherapy ,medicine.disease ,respiratory tract diseases ,Radiation therapy ,Oncology ,Cancer research ,immunotherapy ,CRISPR-Cas9 ,business - Abstract
To date, there are no prognostic/predictive biomarkers to select chemotherapy, immunotherapy, and radiotherapy in individual non-small cell lung cancer (NSCLC) patients. Major immune-checkpoint inhibitors (ICIs) have more DNA copy number variations (CNV) than mutations in The Cancer Genome Atlas (TCGA) NSCLC tumors. Nevertheless, CNV-mediated dysregulated gene expression in NSCLC is not well understood. Integrated CNV and transcriptional profiles in NSCLC tumors (n = 371) were analyzed using Boolean implication networks for the identification of a multi-omics CD27, PD1, and PDL1 network, containing novel prognostic genes and proliferation genes. A 5-gene (EIF2AK3, F2RL3, FOSL1, SLC25A26, and SPP1) prognostic model was developed and validated for patient stratification (p <, 0.02, Kaplan–Meier analyses) in NSCLC tumors (n = 1,163). A total of 13 genes (COPA, CSE1L, EIF2B3, LSM3, MCM5, PMPCB, POLR1B, POLR2F, PSMC3, PSMD11, RPL32, RPS18, and SNRPE) had a significant impact on proliferation in 100% of the NSCLC cell lines in both CRISPR-Cas9 (n = 78) and RNA interference (RNAi) assays (n = 92). Multiple identified genes were associated with chemoresponse and radiotherapy response in NSCLC cell lines (n = 117) and patient tumors (n = 966). Repurposing drugs were discovered based on this immune-omics network to improve NSCLC treatment.
- Published
- 2021
- Full Text
- View/download PDF
29. Integrated Analysis of Structural Variation and RNA Expression of FGFR2 and Its Splicing Modulator ESRP1 Highlight the ESRP1amp-FGFR2norm-FGFR2-IIIchigh Axis in Diffuse Gastric Cancer
- Author
-
Marta Ferreira, Joana Carvalho, Carla Oliveira, Sara Pinto Teles, Patrícia Oliveira, Pedro G. Ferreira, and Instituto de Investigação e Inovação em Saúde
- Subjects
0301 basic medicine ,musculoskeletal diseases ,Cancer Research ,congenital, hereditary, and neonatal diseases and abnormalities ,diffuse ,fgfr2-iiic ,fgfr2-iiib ,Biology ,lcsh:RC254-282 ,Article ,Structural variation ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Epigenetics ,Copy-number variation ,Gene ,integumentary system ,gastric cancer ,fgfr2 ,Cancer ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Biomarker (cell) ,esrp1 ,stomatognathic diseases ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,DNA methylation ,RNA splicing ,Cancer research - Abstract
Gastric Cancer (GC) is one of the most common and deadliest types of cancer in the world. To improve GC prognosis, increasing efforts are being made to develop new targeted therapies. Although FGFR2 genetic amplification and protein overexpression in GC have been targeted in clinical trials, so far no improvement in patient overall survival has been found. To address this issue, we studied genetic and epigenetic events affecting FGFR2 and its splicing regulator ESRP1 in GC that could be used as new therapeutic targets or predictive biomarkers. We performed copy number variation (CNV), DNA methylation, and RNA expression analyses of FGFR2/ESRP1 across several cohorts. We discovered that both genes were frequently amplified and demethylated in GC, resulting in increased ESRP1 expression and of a specific FGFR2 isoform: FGFR2-IIIb. We also showed that ESRP1 amplification in GC correlated with a significant decreased expression of FGFR2-IIIc, an alternative FGFR2 splicing isoform. Furthermore, when we performed a survival analysis, we observed that patients harboring diffuse-type tumors with low FGFR2-IIIc expression revealed a better overall survival than patients with FGFR2-IIIc high-expressing diffuse tumors. Our results encourage further studies on the role of ESRP1 in GC and support FGFR2-IIIc as a relevant biomarker in GC. Ipatimup integrate the i3S Research Unit, which is partially supported by FCT, the Portuguese Foundation for Science and Technology. This work was supported by: (1) FEDER?Fundo Europeu de Desenvolvimento Regional funds through the COMPETE 2020?Operacional Programme for Competitiveness and Internationalisation (POCI), Portugal 2020, and by Portuguese funds through Portuguese Science & Technology Foundation (FCT)/Minist?rio da Ci?ncia, Tecnologia e Inova??o in the framework of the project ?Institute for Research and Innovation in Health Sciences? (POCI-01-0145-FEDER-007274); (2) Project NORTE-01-0145-FEDER-000029 and NORTE-01-0145-FEDER-000003, supported by Norte Portugal Regional Programme (NORTE 2020), under the PORTUGAL 2020 Partnership Agreement, through FEDER; (3) FCT with the Grant References PTDC/BBB-ECT/2518/2014 and POCI-01-0145-FEDER-30164; (4) Project GenomePT Ref. POCI-01-0145-FEDER-022184, supported by COMPETE 2020?POCI, Lisboa Portugal Regional Operational Programme Lisboa2020, Algarve Portugal Regional Operational Programme (CRESC Algarve2020), under the PORTUGAL 2020 Partnership Agreement, through FEDER and FCT; (5) FEDER-POCI-FCT Grant Ref. POCI-01-0145-FEDER-016390; (6) Salary support to M.F. by GenomePT Project ref POCI-01-0145-FEDER-022184; Fellowship to S.P.T. by Grant References PTDC/BBB-ECT/2518/2014. Funding: Ipatimup integrate the i3S Research Unit, which is partially supported by FCT, the Portuguese Foundation for Science and Technology. This work was supported by: (1) FEDER—Fundo Europeu de Desenvolvimento Regional funds through the COMPETE 2020—Operacional Programme for Competitiveness and Internationalisation (POCI), Portugal 2020, and by Portuguese funds through Portuguese Science & Technology Foundation (FCT)/Ministério da Ciência, Tecnologia e Inovação in the framework of the project “Institute for Research and Innovation in Health Sciences” (POCI-01-0145-FEDER-007274); (2) Project NORTE-01-0145-FEDER-000029 and NORTE-01-0145-FEDER-000003, supported by Norte Portugal Regional Programme (NORTE 2020), under the PORTUGAL 2020 Partnership Agreement, through FEDER; (3) FCT with the Grant References PTDC/BBB-ECT/2518/2014 and POCI-01-0145-FEDER-30164; (4) Project GenomePT Ref. POCI-01-0145-FEDER-022184, supported by COMPETE 2020—POCI, Lisboa Portugal Regional Operational Programme Lisboa2020, Algarve Portugal Regional Operational Programme (CRESC Algarve2020), under the PORTUGAL 2020 Partnership Agreement, through FEDER and FCT; (5) FEDER-POCI-FCT Grant Ref. POCI-01-0145-FEDER-016390; (6) Salary support to M.F. by GenomePT Project ref POCI-01-0145-FEDER-022184; Fellowship to S.P.T. by Grant References PTDC/BBB-ECT/2518/2014.
- Published
- 2019
30. A Network Analysis of Multiple Myeloma Related Gene Signatures
- Author
-
Li Wang, Alessandro Laganà, Jun Zhu, Seungyeul Yoo, Eric E. Schadt, Yu Liu, Haocheng Yu, Eunjee Lee, and Samir Parekh
- Subjects
0301 basic medicine ,Cancer Research ,Disease ,Computational biology ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,gene signature ,Article ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,medicine ,Copy-number variation ,Gene ,Subnetwork ,Multiple myeloma ,treatment response ,Gene signature ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,3. Good health ,multiple myeloma ,030104 developmental biology ,Bayesian network ,Oncology ,030220 oncology & carcinogenesis ,Carcinogenesis ,prognostic - Abstract
Multiple myeloma (MM) is the second most prevalent hematological cancer. MM is a complex and heterogeneous disease, and thus, it is essential to leverage omics data from large MM cohorts to understand the molecular mechanisms underlying MM tumorigenesis, progression, and drug responses, which may aid in the development of better treatments. In this study, we analyzed gene expression, copy number variation, and clinical data from the Multiple Myeloma Research Consortium (MMRC) dataset and constructed a multiple myeloma molecular causal network (M3CN). The M3CN was used to unify eight prognostic gene signatures in the literature that shared very few genes between them, resulting in a prognostic subnetwork of the M3CN, consisting of 178 genes that were enriched for genes involved in cell cycle (fold enrichment = 8.4, p value = 6.1 ×, 10&minus, 26). The M3CN was further used to characterize immunomodulators and proteasome inhibitors for MM, demonstrating the pleiotropic effects of these drugs, with drug-response signature genes enriched across multiple M3CN subnetworks. Network analyses indicated potential links between these drug-response subnetworks and the prognostic subnetwork. To elucidate the structure of these important MM subnetworks, we identified putative key regulators predicted to modulate the state of these subnetworks. Finally, to assess the predictive power of our network-based models, we stratified MM patients in an independent cohort, the MMRF-CoMMpass study, based on the prognostic subnetwork, and compared the performance of this subnetwork against other signatures in the literature. We show that the M3CN-derived prognostic subnetwork achieved the best separation between different risk groups in terms of log-rank test p-values and hazard ratios. In summary, this work demonstrates the power of a probabilistic causal network approach to understanding molecular mechanisms underlying the different MM signatures.
- Published
- 2019
- Full Text
- View/download PDF
31. Next-Generation Sequencing Improves Diagnosis, Prognosis and Clinical Management of Myeloid Neoplasms
- Author
-
José Luis Díez-Martín, Patricia Font, Javier Anguita, Paula Muñiz, Pascual Balsalobre, Gabriela Rodríguez-Macías, Mariana Bastos-Oreiro, Julia Suárez-González, Diego Carbonell, Cristina Andrés-Zayas, Mónica Ballesteros, Juan Carlos Triviño, Mi Kwon, Carolina Martínez-Laperche, Ismael Buño, and María Chicano
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,routine diagnosis ,myeloid neoplasm ,Context (language use) ,Disease ,acute myeloid leukemia ,lcsh:RC254-282 ,Article ,Myeloid Neoplasm ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Medicine ,Copy-number variation ,business.industry ,Cytogenetics ,Myeloid leukemia ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Clinical trial ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Next-generation sequencing ,business - Abstract
Molecular diagnosis of myeloid neoplasms (MN) is based on the detection of multiple genetic alterations using various techniques. Next-generation sequencing (NGS) has been proved as a useful method for analyzing many genes simultaneously. In this context, we analyzed diagnostic samples from 121 patients affected by MN and ten relapse samples from a subset of acute myeloid leukemia patients using two enrichment-capture NGS gene panels. Pathogenicity classification of variants was enhanced by the development and application of a custom onco-hematology score. A total of 278 pathogenic variants were detected in 84% of patients. For structural alterations, 82% of those identified by cytogenetics were detected by NGS, 25 of 31 copy number variants and three out of three translocations. The detection of variants using NGS changed the diagnosis of seven patients and the prognosis of 15 patients and enabled us to identify 44 suitable candidates for clinical trials. Regarding AML, six of the ten relapsed patients lost or gained variants, comparing with diagnostic samples. In conclusion, the use of NGS panels in MN improves genetic characterization of the disease compared with conventional methods, thus demonstrating its potential clinical utility in routine clinical testing. This approach leads to better-adjusted treatments for each patient.
- Published
- 2019
32. Molecular Basis of Cisplatin Resistance in Testicular Germ Cell Tumors
- Author
-
Václav Eis, Eva Kindlová, Ludmila Boublikova, Michael Svaton, Tomas Buchler, Marketa Zaliova, Blanka Rosova, Alena Dobiasova, Katerina Rejlova, Martina Slamova, Jan Trka, Violeta Bakardjieva-Mihaylova, Petr Klézl, Marek Grega, Karel Fiser, Jan Stuchly, Karolina Skvarova Kramarzova, and Roman Zachoval
- Subjects
Regulation of gene expression ,next generation sequencing ,Cancer Research ,Communication ,Testicular Germ Cell Tumor ,Biology ,Cell cycle ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Oncology ,Histone methyltransferase ,Cancer research ,testicular germ cell tumor ,molecular aberrations ,cell cycle ,Copy-number variation ,Epigenetics ,cisplatin resistance ,Gene ,ATRX - Abstract
The emergence of cisplatin (CDDP) resistance is the main cause of treatment failure and death in patients with testicular germ cell tumors (TGCT), but its biologic background is poorly understood. To study the molecular basis of CDDP resistance in TGCT we prepared and sequenced CDDP-exposed TGCT cell lines as well as 31 primary patients’ samples. Long-term exposure to CDDP increased the CDDP resistance 10 times in the NCCIT cell line, while no major resistance was achieved in Tera-2. Development of CDDP resistance was accompanied by changes in the cell cycle (increase in G1 and decrease in S-fraction), increased number of acquired mutations, of which 3 were present within ATRX gene, as well as changes in gene expression pattern. Copy number variation analysis showed, apart from obligatory gain of 12p, several other large-scale gains (chr 1, 17, 20, 21) and losses (chr X), with additional more CNVs found in CDDP-resistant cells (e.g., further losses on chr 1, 4, 18, and gain on chr 8). In the patients’ samples, those who developed CDDP resistance and died of TGCT (2/31) showed high numbers of acquired aberrations, both SNPs and CNVs, and harbored mutations in genes potentially relevant to TGCT development (e.g., TRERF1, TFAP2C in one patient, MAP2K1 and NSD1 in another one). Among all primary tumor samples, the most commonly mutated gene was NSD1, affected in 9/31 patients. This gene encoding histone methyl transferase was also downregulated and identified among the 50 most differentially expressed genes in CDDP-resistant NCCIT cell line. Interestingly, 2/31 TGCT patients harbored mutations in the ATRX gene encoding a chromatin modifier that has been shown to have a critical function in sexual differentiation. Our research newly highlights its probable involvement also in testicular tumors. Both findings support the emerging role of altered epigenetic gene regulation in TGCT and CDDP resistance development.
- Published
- 2019
33. Etiology-Specific Analysis of Hepatocellular Carcinoma Transcriptome Reveals Genetic Dysregulation in Pathways Implicated in Immunotherapy Efficacy
- Author
-
Xiao Qi Wang, Weg M. Ongkeko, Hao Zheng, Wei Tse Li, Eric Y. Chang, Tatiana Kisseleva, Angela E. Zou, and Christine O. Honda
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Biology ,lcsh:RC254-282 ,Genome ,Article ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,microRNA ,medicine ,Copy-number variation ,Gene ,cancer immunotherapy ,Immunotherapy ,TCGA ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,mutations ,microRNAs ,030104 developmental biology ,copy number variations ,Oncology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Cancer research - Abstract
Immunotherapy has emerged in recent years as arguably the most effective treatment for advanced hepatocellular carcinoma (HCC), but the failure of a large percentage of patients to respond to immunotherapy remains as the ultimate obstacle to successful treatment. Etiology-associated dysregulation of immune-associated (IA) genes may be central to the development of this differential clinical response. We identified immune-associated genes potentially dysregulated by alcohol or viral hepatitis B in HCC and validated alcohol-induced dysregulations in vitro while using large-scale RNA-sequencing data from The Cancer Genome Atlas (TCGA). Thirty-four clinically relevant dysregulated IA genes were identified. We profiled the correlation of all genomic alterations in HCC patients to IA gene expression while using the information theory-based algorithm REVEALER to investigate the molecular mechanism for their dysregulation and explore the possibility of genome-based patient stratification. We also studied gene expression regulators and identified multiple microRNAs that were implicated in HCC pathogenesis that can potentially regulate these IA genes&rsquo, expression. Our study identified potential key pathways, including the IL-7 signaling pathway and TNFRSF4 (OX40)- NF-&kappa, B pathway, to target in immunotherapy treatments and presents microRNAs as promising therapeutic targets for dysregulated IA genes because of their extensive regulatory roles in the cancer immune landscape.
- Published
- 2019
- Full Text
- View/download PDF
34. Design and Validation of a Gene-Targeted, Next-Generation Sequencing Panel for Routine Diagnosis in Gliomas
- Author
-
Oriane Blanchard, Nancy De Nève, Isabelle Salmon, Bárbara Meléndez, Claude Van Campenhout, Laetitia Lebrun, and Nicky D'Haene
- Subjects
Cancer Research ,Concordance ,Computational biology ,lcsh:RC254-282 ,DNA sequencing ,Article ,03 medical and health sciences ,0302 clinical medicine ,molecular pathology ,Glioma ,glioma ,medicine ,Copy-number variation ,Gene ,1p/19q codeletion ,business.industry ,Molecular pathology ,Sciences bio-médicales et agricoles ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Oncology ,030220 oncology & carcinogenesis ,Who criteria ,next-generation sequencing ,Differential diagnosis ,business ,030217 neurology & neurosurgery - Abstract
The updated 2016 World Health Organization (WHO) classification system for gliomas integrates molecular alterations and histology to provide a greater diagnostic and prognostic utility than the previous, histology-based classification. The increasing number of markers that are tested in a correct diagnostic procedure makes gene-targeted, next-generation sequencing (NGS) a powerful tool in routine pathology practice. We designed a 14-gene NGS panel specifically aimed at the diagnosis of glioma, which allows simultaneous detection of mutations and copy number variations, including the 1p/19q-codeletion and Epidermal Growth Factor Receptor (EGFR) amplification. To validate this panel, we used reference mutated DNAs, nontumor and non-glioma samples, and 52 glioma samples that were previously characterized. The panel was then prospectively applied to 91 brain lesions. A specificity of 100% and sensitivity of 99.4% was achieved for mutation detection. Orthogonal methods, such as in situ hybridization and immunohistochemical techniques, were used for validation, which showed high concordance. The molecular alterations that were identified allowed diagnosis according to the updated WHO criteria, and helped in the differential diagnosis of difficult cases. This NGS panel is an accurate and sensitive method, which could replace multiple tests for the same sample. Moreover, it is a rapid and cost-effective approach that can be easily implemented in the routine diagnosis of gliomas., info:eu-repo/semantics/published
- Published
- 2019
- Full Text
- View/download PDF
35. Bioinformatics Analysis for Circulating Cell-Free DNA in Cancer
- Author
-
Liang Wang, Meijun Du, and Chiang Ching Huang
- Subjects
0301 basic medicine ,Cancer Research ,Computational biology ,Review ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,DNA sequencing ,cell-free DNA ,03 medical and health sciences ,0302 clinical medicine ,Cancer screening ,medicine ,Epigenetics ,Copy-number variation ,Liquid biopsy ,next generation sequencing ,Mutation ,copy number variation ,bioinformatics ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Circulating Cell-Free DNA ,3. Good health ,030104 developmental biology ,Oncology ,Cell-free fetal DNA ,030220 oncology & carcinogenesis ,methylation ,mutation - Abstract
Molecular analysis of cell-free DNA (cfDNA) that circulates in plasma and other body fluids represents a “liquid biopsy” approach for non-invasive cancer screening or monitoring. The rapid development of sequencing technologies has made cfDNA a promising source to study cancer development and progression. Specific genetic and epigenetic alterations have been found in plasma, serum, and urine cfDNA and could potentially be used as diagnostic or prognostic biomarkers in various cancer types. In this review, we will discuss the molecular characteristics of cancer cfDNA and major bioinformatics approaches involved in the analysis of cfDNA sequencing data for detecting genetic mutation, copy number alteration, methylation change, and nucleosome positioning variation. We highlight specific challenges in sensitivity to detect genetic aberrations and robustness of statistical analysis. Finally, we provide perspectives regarding the standard and continuing development of bioinformatics analysis to move this promising screening tool into clinical practice.
- Published
- 2019
36. Genomic Profiling of KRAS/NRAS/BRAF/PIK3CA Wild-Type Metastatic Colorectal Cancer Patients Reveals Novel Mutations in Genes Potentially Associated with Resistance to Anti-EGFR Agents
- Author
-
Alessia Iannaccone, Daniela Frezzetti, Anna Maria Rachiglio, Marianeve Carotenuto, Evaristo Maiello, Antonella De Luca, Cristin Roma, Matilde Lambiase, Francesca Fenizia, Nicola Normanno, Fortunato Ciardiello, E. Martinelli, Claudia Cardone, Rachiglio, A. M., Lambiase, M., Fenizia, F., Roma, C., Cardone, C., Iannaccone, A., De Luca, A., Carotenuto, M., Frezzetti, D., Martinelli, E., Maiello, E., Ciardiello, F., and Normanno, N.
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,Colorectal cancer ,anti-EGFR monoclonal antibodies ,colorectal cancer ,medicine.disease_cause ,lcsh:RC254-282 ,resistance ,03 medical and health sciences ,0302 clinical medicine ,MAP2K1 ,Anti-EGFR monoclonal antibodie ,medicine ,Progression-free survival ,Copy-number variation ,neoplasms ,Cetuximab ,business.industry ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,digestive system diseases ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,FOLFIRI ,KRAS ,genomic profiling ,business ,medicine.drug - Abstract
Previous findings suggest that metastatic colorectal carcinoma (mCRC) patients with KRAS/NRAS/BRAF/PIK3CA wild-type (quadruple-wt) tumors are highly sensitive to anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (MoAbs). However, additional molecular alterations might be involved in the de novo resistance to these drugs. We performed a comprehensive molecular profiling of 21 quadruple-wt tumors from mCRC patients enrolled in the &ldquo, Cetuximab After Progression in KRAS wild-type colorectal cancer patients&rdquo, (CAPRI-GOIM) trial of first line FOLFIRI plus cetuximab. Tumor samples were analyzed with a targeted sequencing panel covering single nucleotide variants (SNVs), insertions/deletions (Indels), copy number variations (CNVs), and gene fusions in 143 cancer-related genes. The analysis revealed in all 21 patients the presence of at least one SNV/Indel and in 10/21 cases (48%) the presence of at least one CNV. Furthermore, 17/21 (81%) patients had co-existing SNVs/Indels in different genes. Quadruple-wt mCRC from patients with the shorter progression free survival (PFS) were enriched with peculiar genetic alterations in KRAS, FBXW7, MAP2K1, and NF1 genes as compared with patients with longer PFS. These data suggest that a wide genetic profiling of quadruple-wt mCRC patients might help to identify novel markers of de novo resistance to anti-EGFR MoAbs.
- Published
- 2019
37. Integrative Analysis Reveals Subtype-Specific Regulatory Determinants in Triple Negative Breast Cancer
- Author
-
Ted M. Lakowski, Pingzhao Hu, Shujun Huang, and Wayne Xu
- Subjects
0301 basic medicine ,Cancer Research ,Biology ,lcsh:RC254-282 ,Article ,triple negative ,03 medical and health sciences ,0302 clinical medicine ,breast cancer ,microRNA ,Gene expression ,Copy-number variation ,lasso ,Transcription factor ,Gene ,Triple-negative breast cancer ,FOXM1 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,humanities ,3. Good health ,030104 developmental biology ,Oncology ,regulator ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer research ,PPARA - Abstract
Different breast cancer (BC) subtypes have unique gene expression patterns, but their regulatory mechanisms have yet to be fully elucidated. We hypothesized that the top upregulated (Yin) and downregulated (Yang) genes determine the fate of cancer cells. To reveal the regulatory determinants of these Yin and Yang genes in different BC subtypes, we developed a lasso regression model integrating DNA methylation (DM), copy number variation (CNV) and microRNA (miRNA) expression of 391 BC patients, coupled with miRNA&ndash, target interactions and transcription factor (TF) binding sites. A total of 25, 20, 15 and 24 key regulators were identified for luminal A, luminal B, Her2-enriched, and triple negative (TN) subtypes, respectively. Many of the 24 TN regulators were found to regulate the PPARA and FOXM1 pathways. The Yin Yang gene expression mean ratio (YMR) and combined risk score (CRS) signatures built with either the targets of or the TN regulators were associated with the BC patients&rsquo, survival. Previously, we identified FOXM1 and PPARA as the top Yin and Yang pathways in TN, respectively. These two pathways and their regulators could be further explored experimentally, which might help to identify potential therapeutic targets for TN.
- Published
- 2019
- Full Text
- View/download PDF
38. Gliosarcoma Is Driven by Alterations in PI3K/Akt, RAS/MAPK Pathways and Characterized by Collagen Gene Expression Signature
- Author
-
Kamil Wojnicki, Wiesława Grajkowska, Bartosz Wojtas, Paweł Nauman, Bozena Kaminska, Marta Maleszewska, Ulrich Schüller, Rainer Glass, Shamba S Mondal, Christel Herold-Mende, and Bartłomiej Gielniewski
- Subjects
0301 basic medicine ,collagen ,Cancer Research ,gliosarcoma ,Gliosarcoma ,Somatic cell ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,medicine ,PTEN ,Copy-number variation ,Gene ,PI3K/AKT/mTOR pathway ,RAS/MAPK ,Mutation ,PI3K/Akt ,glioblastoma ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,mutation ,transcriptome - Abstract
Gliosarcoma is a very rare brain tumor reported to be a variant of glioblastoma (GBM), IDH-wildtype. While differences in molecular and histological features between gliosarcoma and GBM were reported, detailed information on the genetic background of this tumor is lacking. We intend to fill in this knowledge gap by the complex analysis of somatic mutations, indels, copy number variations, translocations and gene expression patterns in gliosarcomas. Using next generation sequencing, we determined somatic mutations, copy number variations (CNVs) and translocations in 10 gliosarcomas. Six tumors have been further subjected to RNA sequencing analysis and gene expression patterns have been compared to those of GBMs. We demonstrate that gliosarcoma bears somatic alterations in gene coding for PI3K/Akt (PTEN, PI3K) and RAS/MAPK (NF1, BRAF) signaling pathways that are crucial for tumor growth. Interestingly, the frequency of PTEN alterations in gliosarcomas was much higher than in GBMs. Aberrations of PTEN were the most frequent and occurred in 70% of samples. We identified genes differentially expressed in gliosarcoma compared to GBM (including collagen signature) and confirmed a difference in the protein level by immunohistochemistry. We found several novel translocations (including translocations in the RABGEF1 gene) creating potentially unfavorable combinations. Collected results on genetic alterations and transcriptomic profiles offer new insights into gliosarcoma pathobiology, highlight differences in gliosarcoma and GBM genetic backgrounds and point out to distinct molecular cues for targeted treatment.
- Published
- 2019
39. Reverse Engineering Cancer: Inferring Transcriptional Gene Signatures from Copy Number Aberrations with ICAro
- Author
-
Maurizio Fanciulli, Matteo Pallocca, and Davide Angeli
- Subjects
0301 basic medicine ,Reverse engineering ,Cancer Research ,transcriptional signatures ,Computer science ,Computational biology ,computer.software_genre ,lcsh:RC254-282 ,Article ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,biomarker discovery ,Copy-number variation ,Biomarker discovery ,Gene ,gene inactivation biomarkers ,copy number variation ,Robustness (evolution) ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Random forest ,copy number aberration ,030104 developmental biology ,Workflow ,Oncology ,030220 oncology & carcinogenesis ,gene signature extraction ,firehose ,computer ,TCGA mining ,cancer CRISPR ,gene loss biomarkers - Abstract
The characterization of a gene product function is a process that involves multiple laboratory techniques in order to silence the gene itself and to understand the resulting cellular phenotype via several omics profiling. When it comes to tumor cells, usually the translation process from in vitro characterization results to human validation is a difficult journey. Here, we present a simple algorithm to extract mRNA signatures from cancer datasets, where a particular gene has been deleted at the genomic level, ICAro. The process is implemented as a two-step workflow. The first one employs several filters in order to select the two patient subsets: the inactivated one, where the target gene is deleted, and the control one, where large genomic rearrangements should be absent. The second step performs a signature extraction via a Differential Expression analysis and a complementary Random Forest approach to provide an additional gene ranking in terms of information loss. We benchmarked the system robustness on a panel of genes frequently deleted in cancers, where we validated the downregulation of target genes and found a correlation with signatures extracted with the L1000 tool, outperforming random sampling for two out of six L1000 classes. Furthermore, we present a use case correlation with a published transcriptomic experiment. In conclusion, deciphering the complex interactions of the tumor environment is a challenge that requires the integration of several experimental techniques in order to create reproducible results. We implemented a tool which could be of use when trying to find mRNA signatures related to a gene loss event to better understand its function or for a gene-loss associated biomarker research.
- Published
- 2019
40. Multiregional Sequencing of IDH-WT Glioblastoma Reveals High Genetic Heterogeneity and a Dynamic Evolutionary History
- Author
-
Riccardo Vannozzi, Martina Modena, Mariangela Morelli, Geoffrey J. Pilkington, Sara Franceschi, Prospero Civita, Serena Barachini, Paolo Aretini, Orazio Santonocito, Francesco Pasqualetti, Francesca Lessi, Chiara Maria Mazzanti, Valerio Ortenzi, and Antonio Giuseppe Naccarato
- Subjects
0301 basic medicine ,Cancer Research ,clonal evolution ,tumor progression ,Computational biology ,Biology ,Somatic evolution in cancer ,Article ,03 medical and health sciences ,Therapeutic approach ,0302 clinical medicine ,multiregional sequencing ,temporal heterogeneity ,Clonal evolution ,Glioblastoma ,Multiregional sequencing ,Spatial heterogeneity ,Temporal heterogeneity ,Tumor phylogeny ,Tumor progression ,Copy-number variation ,Evolutionary dynamics ,RC254-282 ,Exome sequencing ,Genetic heterogeneity ,glioblastoma ,spatial heterogeneity ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Precision medicine ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,tumor phylogeny - Abstract
Simple Summary Glioblastoma is the most common and aggressive primary brain malignancy in adults. In addition to extensive inter-patient heterogeneity, glioblastoma shows intra-tumor extensive cellular and molecular heterogeneity, both spatially and temporally. This heterogeneity is one of the main reasons for the poor prognosis and overall survival. Moreover, it raises the important question of whether the molecular characterization of a single biopsy sample, as performed in standard diagnostics, actually represents the entire lesion. In this study, we sequenced the whole exome of nine spatially different cancer regions of three primary glioblastomas. We characterized their mutational profiles and copy number alterations, with implications for our understanding of tumor biology in relation to clonal architecture and evolutionary dynamics, as well as therapeutically relevant alterations. Abstract Glioblastoma is one of the most common and lethal primary neoplasms of the brain. Patient survival has not improved significantly over the past three decades and the patient median survival is just over one year. Tumor heterogeneity is thought to be a major determinant of therapeutic failure and a major reason for poor overall survival. This work aims to comprehensively define intra- and inter-tumor heterogeneity by mapping the genomic and mutational landscape of multiple areas of three primary IDH wild-type (IDH-WT) glioblastomas. Using whole exome sequencing, we explored how copy number variation, chromosomal and single loci amplifications/deletions, and mutational burden are spatially distributed across nine different tumor regions. The results show that all tumors exhibit a different signature despite the same diagnosis. Above all, a high inter-tumor heterogeneity emerges. The evolutionary dynamics of all identified mutations within each region underline the questionable value of a single biopsy and thus the therapeutic approach for the patient. Multiregional collection and subsequent sequencing are essential to try to address the clinical challenge of precision medicine. Especially in glioblastoma, this approach could provide powerful support to pathologists and oncologists in evaluating the diagnosis and defining the best treatment option.
- Published
- 2021
- Full Text
- View/download PDF
41. Candidate Markers of Olaparib Response from Genomic Data Analyses of Human Cancer Cell Lines
- Author
-
Euridice Carmona, Anne-Marie Mes-Masson, Celia M. T. Greenwood, Patricia N. Tonin, Jiannis Ragoussis, and Setor Amuzu
- Subjects
0301 basic medicine ,Cancer Research ,Population ,Biology ,medicine.disease_cause ,olaparib ,PARP1 ,lcsh:RC254-282 ,Article ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,genomic markers ,medicine ,Copy-number variation ,education ,Exome sequencing ,education.field_of_study ,Mutation ,cancer cell lines ,Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,PARP inhibitor ,Cancer research - Abstract
Simple Summary Olaparib is an oral medication typically used to treat certain advanced ovarian and breast cancers with mutations in BRCA1 or BRCA2 genes. Mutations in these genes can increase the risk of developing breast, ovarian, and other types of cancer. Olaparib is the first clinically approved drug that specifically targets a vulnerability of cancers with these mutations. Genetic alterations in cancer tumors can affect response to treatment in cancer patients. Cancer models such as cell lines, which are cancer cells derived from patients and have been grown in the laboratory over time, can be used to identify these alterations which may contribute to sensitivity or resistance to treatment. We analyzed data from two independent groups of cancer cell lines and identified alterations in additional genes (PUM3, EEF1A1 and ELP4) that potentially increase sensitivity to olaparib. Further experimental and clinical investigations are required to validate our findings. Abstract The benefit of PARP inhibitor olaparib in relapsed and advanced high-grade serous ovarian carcinoma (HGSOC) is well established especially in BRCA1/2 mutation carriers. Identification of additional biomarkers can help expand the population of patients most likely to benefit from olaparib treatment. To identify candidate markers of olaparib response we analyzed genomic and in vitro olaparib response data from two independent groups of cancer cell lines. Using pan-cancer cell lines (n = 896) from the Genomics of Drug Sensitivity in Cancer database, we applied linear regression methods to identify statistically significant gene predictors of olaparib response based on mRNA expression. We then analyzed whole exome sequencing and mRNA gene expression data from our collection of 18 HGSOC cell lines previously classified as sensitive, intermediate, or resistant based on in vitro olaparib response for mutations, copy number variation and differential expression of candidate olaparib response genes. We identify genes previously associated with olaparib response (SLFN11, ABCB1), and discover novel candidate olaparib sensitivity genes with known functions including interaction with PARP1 (PUM3, EEF1A1) and involvement in homologous recombination DNA repair (ELP4). Further investigations at experimental and clinical levels are required to validate novel candidates, and ultimately determine their efficacy as potential biomarkers of olaparib sensitivity.
- Published
- 2021
- Full Text
- View/download PDF
42. Melanotic Neuroectodermal Tumor of Infancy (MNTI) and Pineal Anlage Tumor (PAT) Harbor A Medulloblastoma Signature by DNA Methylation Profiling
- Author
-
Giuseppe Cinalli, Vittoria Donofrio, Rita Alaggio, Sabrina Rossi, Lea F. Surrey, Ivy John, Andrea Ciolfi, Francesca Gianno, Angela Mastronuzzi, Oscar Lopez-Nunez, Lucia Pedace, Franco Locatelli, Evelina Miele, Felice Giangaspero, and Marco Tartaglia
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Brain tumor ,dna methylation profiling ,pineal anlage tumor ,Biology ,Histogenesis ,medulloblastoma ,lcsh:RC254-282 ,Article ,Subclass ,pineal an-lage tumor ,03 medical and health sciences ,0302 clinical medicine ,melanotic neuroectodermal tumor ,melanotic progonoma ,DNA methylation ,copy number variation ,medicine ,Copy-number variation ,Medulloblastoma ,Melanotic neuroectodermal tumor of infancy ,Methylation ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,Settore MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,Oncology ,030217 neurology & neurosurgery - Abstract
Simple Summary Melanotic neuroectodermal tumor of infancy (MNTI) is a rare tumor of uncertain origin, morphologically overlapping other rare neoplasms such as pineal anlage tumor (PAT) and a subset of medulloblastomas (i.e., melanotic medulloblastoma). Despite the similarities with MNTI, their possible histogenetic relationship has been traditionally disregarded based on their aggressive behavior and dismal prognosis. The aim of this study was to further characterize the molecular features of MNTI and PAT based on DNA-methylation and copy number variation profiling analysis. We found that MNTI shares a methylation profile with group 3 high-risk medulloblastoma, and potentially with PAT, suggesting a common histogenesis. Most MNTIs in our series lacked copy number variation alterations, whereas their presence in the one PAT deserves further study in larger cohorts to better determine their impact in prognosis and biologic behavior. Abstract MNTI is a rare tumor of indeterminate histogenesis and molecular signature. We performed methylation and copy number variation (CNV) profiles in patients with MNTI (n = 7) and PAT (n = 1) compared to the methylation brain tumor classifier v11b4 (BT-C) and the medulloblastoma (MB) classifier group 3/4 v1.0 (MB3/4-C). The patients’ mean age was 8 months (range: 4–48). The BT-C classified five MNTIs and one PAT (relapse) as class family MB-G3/G4, subclass group 3 (score: >0.9). The remaining two MNTIs and PAT (primary) were classified as class family plexus tumor, subclass pediatric (scores: >0.45). The MB3/4-C classified all MNTIs as high-risk MB-G3, Subtype II (score: >0.45). The primary PAT was classified as subtype III (score: 0.99) and its relapse as subtype II/III. MNTI and PAT clustered close to MB-G3. CNV analysis showed multiple rearrangements in one PAT and two MNTIs. The median follow-up was 54 months (four MNTIs in remission, one PAT died). In conclusion, we demonstrated that MNTI shares a homogenous methylation profile with MB-G3, and possibly with PAT. The role of a multipotent progenitor cell (i.e., early cranial neural crest cell) in their histogenesis and the influence of the anatomical site, tumor microenvironment, and other cytogenetic events in their divergent biologic behavior deserve further investigation.
- Published
- 2021
- Full Text
- View/download PDF
43. Mutational Landscape of Virus- and UV-Associated Merkel Cell Carcinoma Cell Lines Is Comparable to Tumor Tissue
- Author
-
Angela Hebel-Cherouny, Kai Horny, Jürgen C. Becker, Jochen Utikal, Corinna Wülbeck, and Patricia Gerhardt
- Subjects
0301 basic medicine ,Cancer Research ,DNA damage ,Merkel cell polyomavirus ,MYC ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,Article ,Virus ,merkel cell carcinoma ,03 medical and health sciences ,0302 clinical medicine ,medicine ,whole-exome ,TP53 ,Copy-number variation ,Exome sequencing ,significantly mutated genes ,Merkel cell carcinoma ,copy number variation ,food and beverages ,cell line ,merkel cell polyoma virus ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,biology.organism_classification ,medicine.disease ,UV ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,RB1 ,Carcinogenesis - Abstract
Simple Summary Merkel cell carcinoma (MCC) is an aggressive, rare skin cancer which is caused either by a virus or chronic UV exposure. For both forms, distinct genetic alterations have been described; however, these observations were mostly made in tumor tissue. Since cancer cell lines are frequently used as preclinical models to investigate biological function, we considered it necessary to establish the genomic landscape of MCC cell lines by whole-exome sequencing. We confirmed the presence of UV-induced DNA damage, a high number of mutations and several coding mutations in virus-negative cell lines which were absent in virus-positive cell lines; these, however, harbored characteristic copy number variations, suggesting some virally caused genetic instability. Knowing the genomic features of MCC cell lines validates previous, and facilitates upcoming, experimental studies to discover their biological and translational relevance. Abstract Merkel cell carcinoma (MCC) is a rare, highly aggressive cutaneous malignancy that is either associated with the integration of the Merkel cell polyomavirus or chronic UV exposure. These two types of carcinogenesis are reflected in characteristic mutational features present in MCC tumor lesions. However, the genomic characteristics of MCC cell lines used as preclinical models are not well established. Thus, we analyzed the exomes of three virus-negative and six virus-positive MCC cell lines, all showing a classical neuroendocrine growth pattern. Virus-negative cell lines are characterized by a high tumor mutational burden (TMB), UV-light-induced DNA damage, functionally relevant coding mutations, e.g., in RB1 and TP53, and large amounts of copy number variations (CNVs). In contrast, virus-positive cell lines have a low TMB with few coding mutations and lack prominent mutational signatures, but harbor characteristic CNVs. One of the virus-negative cell lines has a local MYC amplification associated with high MYC mRNA expression. In conclusion, virus-positive and -negative MCC cell lines with a neuroendocrine growth pattern resemble mutational features observed in MCC tissue samples, which strengthens their utility for functional studies.
- Published
- 2021
- Full Text
- View/download PDF
44. T-Cell Lymphoma Clonality by Copy Number Variation Analysis of T-Cell Receptor Genes
- Author
-
Zi Wei Wong, Chee-Leong Cheng, Soo Yong Tan, Zhenhua Li, Tae-Hoon Chung, Jing Quan Lim, Shangying Chen, Allen Eng Juh Yeoh, Joseph D. Khoury, Shigeo Nakamura, Evelyn Huizi Lim, Ming Liang Oon, Shih-Sung Chuang, Wee Joo Chng, Gwyneth Soon, Siok Bian Ng, Soon Thye Lim, Olaf Rötzschke, Kenneth Hon Kim Ban, Bernett Lee, Yong-Howe Ho, Seiichi Kato, Sai Mun Leong, Emiko Takahashi, Choon Kiat Ong, Rex Au-Yeung, Claudio Tripodo, Oon M.L., Lim J.Q., Lee B., Leong S.M., Soon G.S., Wong Z.W., Lim E.H., Li Z., Juh Yeoh A.E., Chen S., Kim Ban K.H., Chung T.-H., Tan S.-Y., Chuang S.-S., Kato S., Nakamura S., Takahashi E., Ho Y.-H., Khoury J.D., Au-Yeung R.K.H., Cheng C.-L., Lim S.-T., Chng W.-J., Tripodo C., Rotzschke O., Ong C.K., and Ng S.-B.
- Subjects
0301 basic medicine ,clone (Java method) ,Cancer Research ,clonality ,Biology ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,T-cell lymphoma ,T-cell receptor ,Copy-number variation ,copy number variation analysis ,Gene ,Whole genome sequencing ,whole genome sequencing ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Molecular biology ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,T-Cell Receptor Gene ,Monoclonal ,Clonality, Copy number variation analysis, T-cell lymphoma, T-cell receptor, Whole genome sequencing - Abstract
Simple Summary T-cells defend the human body from pathogenic invasion via specific recognition by T-cell receptors (TCRs). The TCR genes undergo recombination (rearrangement) in a myriad of possible ways to generate different TCRs that can recognize a wide diversity of foreign antigens. However, in patients with T-cell lymphoma (TCL), a particular T-cell becomes malignant and proliferates, resulting in a population of genetically identical cells with same TCR rearrangement pattern. To help diagnose patients with TCL, a polymerase chain reaction (PCR)-based assay is currently used to determine if neoplastic cells in patient samples are of T-cell origin and bear identical (monoclonal) TCR rearrangement pattern. Herein, we report the application of a novel segmentation and copy number computation algorithm to accurately identify different TCR rearrangement patterns using data from the whole genome sequencing of patient materials. Our approach may improve the diagnostic accuracy of TCLs and can be similarly applied to the diagnosis of B-cell lymphomas. Abstract T-cell lymphomas arise from a single neoplastic clone and exhibit identical patterns of deletions in T-cell receptor (TCR) genes. Whole genome sequencing (WGS) data represent a treasure trove of information for the development of novel clinical applications. However, the use of WGS to identify clonal T-cell proliferations has not been systematically studied. In this study, based on WGS data, we identified monoclonal rearrangements (MRs) of T-cell receptors (TCR) genes using a novel segmentation algorithm and copy number computation. We evaluated the feasibility of this technique as a marker of T-cell clonality using T-cell lymphomas (TCL, n = 44) and extranodal NK/T-cell lymphomas (ENKTLs, n = 20), and identified 98% of TCLs with one or more TCR gene MRs, against 91% detected using PCR. TCR MRs were absent in all ENKTLs and NK cell lines. Sensitivity-wise, this platform is sufficiently competent, with MRs detected in the majority of samples with tumor content under 25% and it can also distinguish monoallelic from biallelic MRs. Understanding the copy number landscape of TCR using WGS data may engender new diagnostic applications in hematolymphoid pathology, which can be readily adapted to the analysis of B-cell receptor loci for B-cell clonality determination.
- Published
- 2021
- Full Text
- View/download PDF
45. Comprehensive Cohort Analysis of Mutational Spectrum in Early Onset Breast Cancer Patients
- Author
-
Yu-Feng Huang, Nai-Chuan Chang, Mohit K. Midha, Alice L. Yu, Chen-Yang Shen, Kuo Ping Chiu, King-Jen Chang, Tan-Chi Fan, Yu-Tai Wang, Chien-Jen Chen, Tzu-Han Chen, Hsiao-Hsiang Yang, and Wen-Hung Kuo
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Oncology and Carcinogenesis ,early onset breast cancer ,lcsh:RC254-282 ,Article ,Germline ,early onset breast cancer (EOBC) ,03 medical and health sciences ,missense mutations ,nonsynonymous mutations ,0302 clinical medicine ,Breast cancer ,Germline mutation ,Clinical Research ,Internal medicine ,Breast Cancer ,Genetics ,2.1 Biological and endogenous factors ,Medicine ,Missense mutation ,Copy-number variation ,Aetiology ,Cancer ,business.industry ,Prevention ,Mortality rate ,Human Genome ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cohort ,somatic mutations ,germline mutations ,business ,Biotechnology ,Cohort study - Abstract
Early onset breast cancer (EOBC), diagnosed at age ~40 or younger, is associated with a poorer prognosis and higher mortality rate compared to breast cancer diagnosed at age 50 or older. EOBC poses a serious threat to public health and requires in-depth investigation. We studied a cohort comprising 90 Taiwanese female patients, aiming to unravel the underlying mechanisms of EOBC etiopathogenesis. Sequence data generated by whole-exome sequencing (WES) and whole-genome sequencing (WGS) from white blood cell (WBC)&ndash, tumor pairs were analyzed to identify somatic missense mutations, copy number variations (CNVs) and germline missense mutations. Similar to regular breast cancer, the key somatic mutation-susceptibility genes of EOBC include TP53 (40% prevalence), PIK3CA (37%), GATA3 (17%) and KMT2C (17%), which are frequently reported in breast cancer, however, the structural protein-coding genes MUC17 (19%), FLG (16%) and NEBL (11%) show a significantly higher prevalence in EOBC. Furthermore, the top 2 genes harboring EOBC germline mutations, MUC16 (19%) and KRT18 (19%), encode structural proteins. Compared to conventional breast cancer, an unexpectedly higher number of EOBC susceptibility genes encode structural proteins. We suspect that mutations in structural proteins may increase physical permeability to environmental hormones and carcinogens and cause breast cancer to occur at a young age.
- Published
- 2020
- Full Text
- View/download PDF
46. Molecular Features of Metaplastic Breast Carcinoma: An Infrequent Subtype of Triple Negative Breast Carcinoma
- Author
-
Maria Luisa Palacios-Berraquero, Silvia González-Martínez, Irene Carretero-Barrio, Javier Cortes, Jose Perez-Garcia, Belén Pérez-Mies, José Palacios, Institut Català de la Salut, [González-Martínez S] Clinical Researcher, Hospital Universitario Ramón y Cajal, Madrid, Spain. [Pérez-Mies B] Pathology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain. Instituto Ramón y Cajal for Health Research (IRYCIS), Madrid, Spain. CIBER-ONC, Instituto de Salud Carlos III, Madrid, Spain. Faculty of Medicine, University of Alcalá de Henares, Alcalá de Henares, Madrid, Spain. Breast Pathology Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain. [Carretero-Barrio I] Pathology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain. [Palacios-Berraquero ML] Hematology and Hemotherapy Department, Clínica Universidad de Navarra, Pamplona, Spain. [Perez-García J] IOB Institute of Oncology, Quironsalud Group, Hospital Quiron, Barcelona, Spain. [Cortés J] CIBER-ONC, Instituto de Salud Carlos III, Madrid, Spain. IOB Institute of Oncology, Quironsalud Group, Hospital Quiron, Barcelona, Spain. IOB Institute of Oncology, Quironsalud Group, Madrid, Spain. Medica Scientia Innovation Research, 08018 Barcelona, Spain. Medica Scientia Innovation Research, Ridgewood, USA. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
0301 basic medicine ,MBC ,Cancer Research ,Review ,Biology ,lcsh:RC254-282 ,Pathogenesis ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Mama - Càncer ,CDKN2A ,CDKN2B ,polycyclic compounds ,Neoplasms::Neoplasms by Histologic Type::Neoplasms, Glandular and Epithelial::Carcinoma::Carcinoma, Squamous Cell [DISEASES] ,Molecular alterations ,Metaplastic breast carcinoma ,neoplasias::neoplasias por tipo histológico::neoplasias glandulares y epiteliales::carcinoma::carcinoma de células escamosas [ENFERMEDADES] ,molecular alterations ,Copy-number variation ,skin and connective tissue diseases ,neoplasms ,metaplastic breast carcinoma ,PI3K/AKT/mTOR pathway ,Resistència als medicaments ,treatment ,Metaplastic Breast Carcinoma ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Prognosis ,bacterial infections and mycoses ,neoplasias::neoplasias por localización::neoplasias de la mama::neoplasias de mama triple negativos [ENFERMEDADES] ,Treatment ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Neoplasms::Neoplasms by Site::Breast Neoplasms::Triple Negative Breast Neoplasms [DISEASES] ,Cancer research ,bacteria ,Triple-Negative Breast Carcinoma ,prognosis - Abstract
Carcinoma de mama metaplàsic; Alteracions moleculars; Pronòstic Carcinoma de mama metaplásico; Alteraciones moleculares; Pronóstico Metaplastic breast carcinoma; Molecular alterations; Prognosis Metaplastic breast carcinoma (MBC) is a heterogeneous group of infrequent invasive carcinomas that display differentiation of the neoplastic epithelium towards squamous cells and/or mesenchymal-type elements. Most MBC have a triple negative phenotype and poor prognosis. Thus, MBC have worse survival rates than other invasive breast carcinomas, including other triple negative breast carcinomas (TNBC). In this study, we reviewed the molecular features of MBC, pointing out the differences among subtypes. The most frequently mutated genes in MBC were TP53 and PIK3CA. Additionally, mutations in the other genes of the PI3K/AKT pathway indicated its importance in the pathogenesis of MBC. Regarding copy number variations (CNVs), MYC was the most frequently amplified gene, and the most frequent gene loss affected the CDKN2A/CDKN2B locus. Furthermore, the pattern of mutations and CNVs of MBC differed from those reported in other TNBC. However, the molecular profile of MBC was not homogeneous among histological subtypes, being the alterations in the PI3K pathway most frequent in spindle cell carcinomas. Transcriptomic studies have demonstrated an epithelial to mesenchymal program activation and the enrichment of stemness genes in most MBC. In addition, current studies are attempting to define the immune microenvironment of these tumors. In conclusion, due to specific molecular features, MBC have a different clinical behavior from other types of TNBC, being more resistant to standard chemotherapy. For this reason, new therapeutic approaches based on tumor molecular characteristics are needed to treat MBC. This review was funded by grants from the Instituto de Salud Carlos III (ISCIII) (PI19/01331) and CIBERONC (CB16/12/00316 and CB16/12/00449), co-financed by the European Development Regional Fund. ‘A way to achieve Europe’ (FEDER), and by the Spanish Association Against Cancer Scientific Foundation (Grupos Coordinados Traslacionales aecc 2018).
- Published
- 2020
- Full Text
- View/download PDF
47. The Implementation of DNA Methylation Profiling into a Multistep Diagnostic Process in Pediatric Neuropathology: A 2-Year Real-World Experience by the French Neuropathology Network.
- Author
-
Pages, Melanie, Uro-Coste, Emmanuelle, Colin, Carole, Meyronet, David, Gauchotte, Guillaume, Maurage, Claude-Alain, Rousseau, Audrey, Godfraind, Catherine, Mokhtari, Karima, Silva, Karen, Figarella-Branger, Dominique, Varlet, Pascale, and Giangaspero, Felice
- Subjects
- *
NEUROLOGISTS , *UNCERTAINTY , *MOLECULAR pathology , *DNA methylation , *TUMORS in children , *GENE expression profiling , *DESCRIPTIVE statistics , *LONGITUDINAL method , *CHILDREN ,DIAGNOSIS of tumors in children ,CENTRAL nervous system tumors - Abstract
Simple Summary: Supported by the "easy-to-use" and free DKFZ central nervous system (CNS) tumor classification web tool, DNA methylation profiling is a method changing the routine diagnostic practice in neuro-oncology. This work depicts a real-world practice experience by the French neuropathology network of incorporating DNA methylation profiling into the diagnostic process of challenging pediatric CNS tumors. After two rounds of histopathological review by neuropathology experts—including morphology, neuroimaging, immunohistochemistry, panel sequencing and FISH—62 tumors still presenting diagnostic uncertainty were selected for DNA methylation profiling. Using the DKFZ "classifier" and combining all additional information obtained from DNA methylation array, we observed significant diagnostic refinements and amendments. DNA methylation was successful in a significant number of cases (71%) despite the complex specificities of the cohort. Our study evaluates how DNA methylation testing would impact diagnosis and presents illustrative and representative cases. DNA methylation profiling has recently emerged as a powerful tool to help establish diagnosis in neuro-oncology. Here we present our national diagnostic strategy as the French neuropathology network (RENOCLIP-LOC) and our current approach of integrating DNA methylation profiling into our multistep diagnostic process for challenging pediatric CNS tumors. The tumors with diagnostic uncertainty were prospectively selected for DNA methylation after two rounds of review by neuropathology experts. We first integrated the classifier score into the histopathological findings. Subsequent analyses using t-SNE (t-Distributed Stochastic Neighbor Embedding) representation were performed. An additional step consisted of analyzing copy-number variation data (CNV). Finally, we combined all data to establish diagnoses and evaluated the impact of DNA methylation profiling on diagnostic and grading changes that would affect patient management. Over two years, 62 pediatric tumors were profiled. (1) Integrating the classifier score to the histopathological findings impacted the diagnosis in 33 cases (53%). (2) t-SNE analysis provided arguments for diagnosis in 26/35 cases with calibrated scores <0.84 (74.3%). (3) CNV investigations also evidenced alterations used for diagnosis and prognostication. (4) A diagnosis was finally established for 44 tumors (71%). Our results support the use of DNA methylation for challenging pediatric tumors. We demonstrated how additional methylation-based analyses complement the classifier score to support conventional histopathological diagnosis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
48. Homologous Recombination Repair Mechanisms in Serous Endometrial Cancer.
- Author
-
Jönsson, Jenny-Maria, Bååth, Maria, Björnheden, Ida, Sahin, Irem Durmaz, Måsbäck, Anna, and Hedenfalk, Ingrid
- Subjects
- *
THERAPEUTIC use of antineoplastic agents , *CHROMOSOME abnormalities , *DNA repair , *ENZYME inhibitors , *METABOLIC disorders , *GENETIC mutation , *TUMORS , *PHENOTYPES , *ENDOMETRIAL tumors , *GENOMICS , *SINGLE nucleotide polymorphisms , *DESCRIPTIVE statistics , *SEQUENCE analysis ,TUMOR genetics - Abstract
Simple Summary: Serous endometrial cancer is an unusual and aggressive endometrial cancer subtype, conferring the highest mortality of all endometrial cancers. In many ways, it resembles the more common tumor entity high-grade serous ovarian cancer. Thus, there is an urgent need for better treatment options for serous endometrial cancer patients. It is crucial for all dividing cells that the DNA repair is functioning correctly. Our aim was to investigate deficiencies in DNA repair in serous endometrial cancer, in particular the presence of homologous recombination repair deficiency. This kind of DNA repair defect may indicate that a specific targeted therapy, so-called PARP inhibitors, which are already in use for the treatment of ovarian cancer, may be useful also in serous endometrial cancer. This study contributes to the largely unexplored field of DNA repair deficiencies in serous endometrial cancer, and may hence contribute to future improved prognosis for these patients. Serous endometrial cancer (SEC) resembles high-grade serous ovarian cancer (HGSOC) genetically and clinically, with recurrent copy number alterations, TP53 mutations and a poor prognosis. Thus, SEC patients may benefit from targeted treatments used in HGSOC, e.g., PARP inhibitors. However, the preclinical and clinical knowledge about SEC is scarce, and the exact role of defective DNA repair in this tumor subgroup is largely unknown. We aimed to outline the prevalence of homologous recombination repair deficiency (HRD), copy-number alterations, and somatic mutations in SEC. OncoScan SNP arrays were applied to 19 tumors in a consecutive SEC series to calculate HRD scores and explore global copy-number profiles and genomic aberrations. Copy-number signatures were established and targeted sequencing of 27 HRD-associated genes was performed. All factors were examined in relation to HRD scores to investigate potential drivers of the HRD phenotype. Ten of the 19 SEC tumors (53%) had an HRD score > 42, considered to reflect an HRD phenotype. Higher HRD score was associated with loss of heterozygosity in key HRD genes, and copy-number signatures associated with non-BRCA1/2 dependent HRD in HGSOC. A high number of SECs display an HRD phenotype. It remains to be elucidated whether this also confers PARP inhibitor sensitivity. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
49. Identification of Circulating Genomic and Metabolic Biomarkers in Intrahepatic Cholangiocarcinoma
- Author
-
Joe Harvey, Matteo P. Ferla, Ricky A. Sharma, Jenny C. Taylor, E Giacopuzzi, Pamela J. Kaisaki, McCullagh Jso., H Winter, Melissa M. Pentony, and Knight Sjl.
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,IDH1 ,orotic acid ,Metabolite ,2-hydroxyglutarate ,Gene mutation ,Biology ,medicine.disease_cause ,digestive system ,TYMS ,IDH2 ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,CAD ,Copy-number variation ,Biomarker discovery ,neoplasms ,UMPS ,Intrahepatic cholangiocarcinoma ,DHODH ,circulating DNA ,metabolomics ,digestive system diseases ,3. Good health ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,KRAS - Abstract
Intrahepatic cholangiocarcinoma (ICC) is an aggressive cancer arising from the bile ducts with a need for earlier diagnosis and a greater range of treatment options. KRAS/NRAS mutations are common in ICC tumours and 6&ndash, 32% of patients also have isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) gene mutations associated with metabolic changes. This feasibility study investigated sequencing circulating tumour DNA (ctDNA) combined with metabolite profiling of plasma as a method for biomarker discovery in ICC patients. Plasma was collected from four ICC patients receiving radio-embolisation and healthy controls at multiple time points. ctDNA was sequenced using Ampliseq cancer hotspot panel-v2 on Ion Torrent PGM for single nucleotide variants (SNV) detection and with Illumina whole genome sequencing for copy number variants (CNV) and further targeted examination for SNVs. Untargeted analysis of metabolites from patient and control plasma was performed using liquid chromatography coupled with high-resolution tandem mass spectrometry (LC-MS/MS). Metabolite identification was performed using multi-parameter comparisons with analysis of authentic standards, and univariate statistical analysis was performed to identify differences in metabolite abundance between patient and control samples. Recurrent somatic SNVs and CNVs were identified in ctDNA from three out of four patients that included both NRAS and IDH1 mutations linked to ICC. Plasma metabolite analysis revealed biomarker metabolites associated with ICC and in particular 2-hydroxyglutarate (2-HG) levels were elevated in both samples from the only patient showing a variant allele in IDH1. A reduction in the number of CNVs was observed with treatment. This study demonstrates that ctDNA and metabolite levels can be identified and correlated in ICC patient blood samples and differentiated from healthy controls. We conclude that combining genomic and metabolic analysis of plasma offers an effective approach to biomarker identification with potential for disease stratification and early detection studies.
- Published
- 2019
- Full Text
- View/download PDF
50. Comprehensive Outline of Whole Exome Sequencing Data Analysis Tools Available in Clinical Oncology
- Author
-
Áron Bartha and Balázs Győrffy
- Subjects
0301 basic medicine ,Clinical Oncology ,Cancer Research ,Computer science ,Druggability ,Review ,bioinformatics ,Computational biology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,whole exome sequencing ,03 medical and health sciences ,Identification (information) ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Mutation (genetic algorithm) ,cancer ,Human genome ,Copy-number variation ,Analysis tools ,Exome sequencing - Abstract
Whole exome sequencing (WES) enables the analysis of all protein coding sequences in the human genome. This technology enables the investigation of cancer-related genetic aberrations that are predominantly located in the exonic regions. WES delivers high-throughput results at a reasonable price. Here, we review analysis tools enabling utilization of WES data in clinical and research settings. Technically, WES initially allows the detection of single nucleotide variants (SNVs) and copy number variations (CNVs), and data obtained through these methods can be combined and further utilized. Variant calling algorithms for SNVs range from standalone tools to machine learning-based combined pipelines. Tools for CNV detection compare the number of reads aligned to a dedicated segment. Both SNVs and CNVs help to identify mutations resulting in pharmacologically druggable alterations. The identification of homologous recombination deficiency enables the use of PARP inhibitors. Determining microsatellite instability and tumor mutation burden helps to select patients eligible for immunotherapy. To pave the way for clinical applications, we have to recognize some limitations of WES, including its restricted ability to detect CNVs, low coverage compared to targeted sequencing, and the missing consensus regarding references and minimal application requirements. Recently, Galaxy became the leading platform in non-command line-based WES data processing. The maturation of next-generation sequencing is reinforced by Food and Drug Administration (FDA)-approved methods for cancer screening, detection, and follow-up. WES is on the verge of becoming an affordable and sufficiently evolved technology for everyday clinical use.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.