1
|
Ida CM, Johnson DR, Nair AA, Davila J, Kollmeyer TM, Minn K, Fadra NM, Balcom JR, Fung KMA, Kim DK, Kaufmann TJ, Kipp BR, Halling KC, Jenkins RB, Giannini C. Polymorphous Low-Grade Neuroepithelial Tumor of the Young (PLNTY): Molecular Profiling Confirms Frequent MAPK Pathway Activation. J Neuropathol Exp Neurol 2021; 80:821-829. [PMID: 34363682 DOI: 10.1093/jnen/nlab075] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Polymorphous low-grade neuroepithelial tumor of the young (PLNTY) is a recently described epileptogenic tumor characterized by oligodendroglioma-like components, aberrant CD34 expression, and frequent mitogen-activated protein kinase (MAPK) pathway activation. We molecularly profiled 13 cases with diagnostic histopathological features of PLNTY (10 female; median age, 16 years; range, 5-52). Patients frequently presented with seizures (9 of 12 with available history) and temporal lobe tumors (9 of 13). MAPK pathway activating alterations were identified in all 13 cases. Fusions were present in the 7 youngest patients: FGFR2-CTNNA3 (n = 2), FGFR2-KIAA1598 (FGFR2-SHTN1) (n = 1), FGFR2-INA (n = 1), FGFR2-MPRIP (n = 1), QKI-NTRK2 (n = 1), and KIAA1549-BRAF (n = 1). BRAF V600E mutation was present in 6 patients (17 years or older). Two fusion-positive cases additionally harbored TP53/RB1 abnormalities suggesting biallelic inactivation. Copy number changes predominantly involving whole chromosomes were observed in all 10 evaluated cases, with losses of chromosome 10q occurring with FGFR2-KIAA1598 (SHTN1)/CTNNA3 fusions. The KIAA1549-BRAF and QKI-NTRK2 fusions were associated respectively with a 7q34 deletion and 9q21 duplication. This study shows that despite its name, PLNTY also occurs in older adults, who frequently show BRAF V600E mutation. It also expands the spectrum of the MAPK pathway activating alterations associated with PLNTY and demonstrates recurrent chromosomal copy number changes consistent with chromosomal instability.
Collapse
Affiliation(s)
- Cristiane M Ida
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA (CMI, TMK, KM, JRB, BRK, KCH, RBJ, CG)
| | - Derek R Johnson
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA (DRJ, DKK, TJK)
| | - Asha A Nair
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA (AAN, JD, NMF)
| | - Jaime Davila
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA (AAN, JD, NMF).,Department of Mathematics, Statistics and Computer Science, St Olaf College, Northfield, Minnesota, USA (JD)
| | - Thomas M Kollmeyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA (CMI, TMK, KM, JRB, BRK, KCH, RBJ, CG)
| | - Kay Minn
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA (CMI, TMK, KM, JRB, BRK, KCH, RBJ, CG)
| | - Numrah M Fadra
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA (AAN, JD, NMF)
| | - Jessica R Balcom
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA (CMI, TMK, KM, JRB, BRK, KCH, RBJ, CG)
| | - Kar-Ming A Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA (KMAF)
| | - Dong Kun Kim
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA (DRJ, DKK, TJK)
| | - Timothy J Kaufmann
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA (DRJ, DKK, TJK)
| | - Benjamin R Kipp
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA (CMI, TMK, KM, JRB, BRK, KCH, RBJ, CG)
| | - Kevin C Halling
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA (CMI, TMK, KM, JRB, BRK, KCH, RBJ, CG)
| | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA (CMI, TMK, KM, JRB, BRK, KCH, RBJ, CG)
| | - Caterina Giannini
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA (CMI, TMK, KM, JRB, BRK, KCH, RBJ, CG)
| |
Collapse
|
2
|
Mokánszki A, Bádon ES, Mónus A, Tóth L, Bittner N, Méhes G. Cell-free DNA From Pleural Effusion Samples: Is It Right for Molecular Testing in Lung Adenocarcinoma? Pathol Oncol Res 2021; 27:613071. [PMID: 34257581 PMCID: PMC8262152 DOI: 10.3389/pore.2021.613071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/19/2021] [Indexed: 12/03/2022]
Abstract
Pathogenic molecular features gained specific significance in therapeutic decisions in lung carcinoma in the past decade. Initial and follow up genetic testing requres appropriate amounts and quality of tumor derived DNA, but tumor sampling, especially for disease monitoring is generally limited. Further to the peripheral blood (PB), samples from pleural fluid, accumulating in diverse lung processes might serve as an alternative source for cell-free DNA (cfDNA) for genetic profiling. In our study, cfDNA isolated from the pleural effusion and from the PB, and genomic DNA (gDNA) obtained from tissue/cellular samples were analyzed and compared from altogether 65 patients with pulmonary disease, including 36 lung adenocarcinomas. The quantity of effusion cfDNA yield appeared to be significantly higher compared to that from simultaneously collected PB plasma (23.2 vs. 4.8 ng/μl, p < 0.05). Gene mutations could be safely demonstrated from the effusion cfDNA fraction obtained from adenocarcinoma patients, 3/36 EGFR, 9/36 KRAS and 1/36 BRAF gene variants were detected. In this series, 9/13 samples showed an effusion+/plasma-mutational status, while only 1/13 samples presented with the opposite findings (effusion-/plasma+). gDNA analysis from sediment cell blocks from the identical effusion sample was surprisingly ineffective for lung adenocarcinoma profiling due to the low DNA yield. In conclusion, the cell free supernatant of pleural effusions appears to concentrate cancer derived cfDNA and seems to be particularly suitable for serial genotyping of pulmonary adenocarcinoma.
Collapse
Affiliation(s)
- Attila Mokánszki
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Emese Sarolta Bádon
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anikó Mónus
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Tóth
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nóra Bittner
- Department of Pulmonology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
3
|
Low-coverage whole-genome sequencing of extracellular vesicle-associated DNA in patients with metastatic cancer. Sci Rep 2021; 11:4016. [PMID: 33597619 PMCID: PMC7889887 DOI: 10.1038/s41598-021-83436-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
Low-coverage whole-genome sequencing (LC-WGS) can provide insight into oncogenic molecular changes. Serum extracellular vesicles (EV) represent a novel liquid biopsy source of tumoral DNA. This study compared copy number alteration (CNA) profiles generated from LC-WGS of formalin-fixed paraffin-embedded (FFPE) tumoral DNA and EV-DNA obtained from cancer patients. Patients with squamous cell carcinoma of the base of tongue (n = 3) and cutaneous squamous cell carcinoma (n = 2) were included. LC-WGS (0.5-1X coverage) was performed on FFPE-DNA and serum EV-DNA. Similarity between CNA profiles was analysed using QDNAseq. FFPE samples had a mean CNA of 31 (range 17–50) over 1.9 × 109 (range 1.0–2.6 × 109) bp in length, and EV samples had a mean CNA value of 17 (range 7–19) over 7.6 × 108 (range 2.9–15 × 108) bp in length. A mean of 8 (range 0–21) CNA over 5.9 × 108 (range 1.6–14 × 108) bp in length was found to overlap between EV and FFPE-derived samples per patient. Although the mean correlation efficient between samples was r = 0.34 (range − .08 to 0.99), this was not statistically significant (p > 0.05). Regions of highest deletion and duplication in FFPE samples were not well reflected in the EV-DNA. Selected CNA regions in EV-associated DNA were reflective of the primary tumor, however appreciation of global CNA and areas of most significant change was lost. The utility of LC-WGS of EV-derived DNA is likely limited to molecular alterations of known interest.
Collapse
|
4
|
Through the looking glass and what you find there: making sense of comparative genomic hybridization and fluorescence in situ hybridization for melanoma diagnosis. Mod Pathol 2020; 33:1318-1330. [PMID: 32066861 DOI: 10.1038/s41379-020-0490-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/11/2022]
Abstract
Melanoma is the leading cause of death among cutaneous neoplasms. Best outcome relies on early detection and accurate pathologic diagnosis. For the great majority of melanocytic tumors, histopathologic examination can reliably distinguish nevi from melanomas. However, there is a subset of melanocytic tumors that cannot be definitively classified as benign or malignant using histopathological criteria alone. These tumors are usually diagnosed using terms that imply various degrees of uncertainty in regards to their malignant potential and create the possibility for over or undertreatment. For such tumors, additional ancillary tests would be beneficial in adjudicating a more definitive diagnosis. In recent years, DNA-based molecular ancillary tests, specifically comparative genomic hybridization and fluorescence in situ hybridization, have been developed to help guide the diagnosis of ambiguous melanocytic proliferations. This study will present an updated overview of these two major ancillary tests, which are currently being used in clinical practice to assist in the diagnosis of challenging melanocytic neoplasms.
Collapse
|
5
|
Yu MC, Wu TH, Lee CW, Lee YS, Lian JH, Tsai CL, Hsieh SY, Tsai CN. Percentage genome change and chromosome 7q amplification predict sorafenib response in advanced hepatocellular carcinoma. Biomed J 2020; 44:S73-S83. [PMID: 35747997 PMCID: PMC9038951 DOI: 10.1016/j.bj.2020.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 12/24/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) may arise from genomic instability and has dismal outcome. Sorafenib is the first-line treatment for advanced stage HCC, but its therapeutic efficacy is less than 50%. Biomarkers for predicting the therapeutic efficacy of sorafenib administration to patients with advanced HCC are required. Here, we evaluated the role of chromosomal copy number aberrations (CNAs) in patients with advanced HCC who were treated with sorafenib along with their drug response. Methods The response to sorafenib treatment of twenty-three HCC patients who developed advanced recurrence after partial hepatectomy was analyzed using the modified Response Evaluation Criteria in Solid Tumors (mRECIST). Formalin fixed paraffin embedded (FFPE) tissue specimens obtained after tumor resection were analyzed using the Affymetrix OncoScan® FFPE assay. Results From the 23 patients analyzed in this study, 7 (30.4%) had complete/partial response to sorafenib (CR/PR), 7 (30.4%) had stable disease (SD), and 9 (39.1%) had progressive disease (PD). The mean genome-wide percentage of genome change acquisition via the OncoScan platform was 19.8% for patients with CR/PR/SD and 50.02% in the PD group (p = 0.055). Percentage of genome change above 33% was associated with adverse outcomes for sorafenib treatment in the time-to-progression analysis (p = 0.007) and overall survival (p = 0.096). Among these CNAs, amplification of chromosome 7q, containing the multidrug resistance gene ATP Binding Cassette Subfamily B Member 1 (ACBC1), significantly associated with poor overall survival (p = 0.004) and time-to-progression (p < 0.001). Conclusions Higher percentage genome change and amplification of chromosome 7q in advanced HCC is associated with sorafenib resistance.
Collapse
|
6
|
Chao J, Bedell V, Lee J, Li MS, Chu P, Yuan YC, Zhao D, Klempner SJ, Lin RJ. Association Between Spatial Heterogeneity Within Nonmetastatic Gastroesophageal Adenocarcinomas and Survival. JAMA Netw Open 2020; 3:e203652. [PMID: 32338752 PMCID: PMC7186861 DOI: 10.1001/jamanetworkopen.2020.3652] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IMPORTANCE Intratumoral heterogeneity has been recognized as a significant barrier in successfully developing targetable biomarkers for gastroesophageal adenocarcinoma (GEA) and may affect neoadjuvant precision medicine approaches. OBJECTIVE To describe intratumoral spatial heterogeneity of tumor cell populations in nonmetastatic GEA and its association with survival. DESIGN, SETTING, AND PARTICIPANTS This case series retrospectively identified 41 patients with GEA who underwent up-front surgical resection at a tertiary referral cancer center from January 1, 1989, through December 31, 2013. Survival was calculated from date of surgery to date of death through June 1, 2017. Data were analyzed from June 2, 2017, to March 1, 2019. MAIN OUTCOMES AND MEASURES Overall survival, intratumoral clonal composition determined by genomic single-nucleotide variation array and bioinformatic analysis, and intercellular tumoral distances determined by multiprobe fluorescence in situ hybridization. RESULTS Among the 41 patients included in the analysis (22 men [54%]; mean [SD] age, 63 [12] years), a high proportion (19 [46%]) presented with tumors possessing high intratumoral heterogeneity. Kaplan-Meier analysis demonstrated that cases with an intratumoral clonal composition count of at least 2 exhibited worse survival compared with cases with a clonal composition count of 0 to 1 (univariate hazard ratio, 3.92; 95% CI, 1.27-12.08; P = .02). This finding remained significant on multivariate analysis controlling for stage, Lauren histologic subtype, receipt of adjuvant therapy, and age (multivariate hazard ratio, 4.55; 95% CI, 1.09-19.04; P = .04). Multiprobe fluorescence in situ hybridization demonstrated intratumoral clonal populations coexisting at submillimeter distances with differing relevant oncogenic copy number alterations, such as EGFR, JAK2, FGFR2, MET, CCND1, KRAS, MYC, PIK3CA, CD274, and PDCD1LG2. CONCLUSIONS AND RELEVANCE This study found that spatial intratumoral heterogeneity of oncogenic copy number alterations exists before metastatic dissemination, and increased heterogeneity was associated with worse outcomes in resected GEA. Baseline heterogeneity illustrates the challenges in GEA targeted therapy. Further study may offer insight into strategies on combinatorial and/or sequential targeted and immunotherapeutic approaches.
Collapse
Affiliation(s)
- Joseph Chao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Victoria Bedell
- Cytogenetics Core, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Min Sierra Li
- Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Peiguo Chu
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Yate-Ching Yuan
- Bioinformatics Core, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Dan Zhao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Samuel J. Klempner
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston
- Harvard Medical School, Boston, Massachusetts
| | - Ren-Jang Lin
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, California
| |
Collapse
|
7
|
|
8
|
Jünger ST, Mynarek M, Wohlers I, Dörner E, Mühlen AZ, Velez-Char N, von Hoff K, Rutkowski S, Warmuth-Metz M, Kortmann RD, Timmermann B, Rahmann S, Klein-Hitpass L, von Bueren AO, Pietsch T. Improved risk-stratification for posterior fossa ependymoma of childhood considering clinical, histological and genetic features - a retrospective analysis of the HIT ependymoma trial cohort. Acta Neuropathol Commun 2019; 7:181. [PMID: 31727173 PMCID: PMC6857225 DOI: 10.1186/s40478-019-0820-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 09/29/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Risk stratification of children with ependymomas of the posterior fossa in current therapeutic protocols is mainly based on clinical criteria. We aimed to identify independent outcome predictors for this disease entity by a systematic integrated analysis of clinical, histological and genetic information in a defined cohort of patients treated according to the German HIT protocols. METHODS Tumor samples of 134 patients aged 0.2-15.9 years treated between 1999 and 2010 according to HIT protocols were analyzed for histological features including mitotic activity, necrosis and vascular proliferation and genomic alterations by SNP and molecular inversion probe analysis. Survival analysis was performed by Kaplan-Meier method with log rank test and multivariate Cox regression analysis. RESULTS Residual tumor after surgery, chromosome 1q gain and structural genomic alterations were identified as predictors of significantly shorter event-free (EFS) and overall survival (OS). Furthermore, specific histological features including vascular proliferation, necrosis and high mitotic activity were predictive for shorter OS. Multivariate Cox regression revealed residual tumor, chromosome 1q gain and mitotic activity as independent predictors of both EFS and OS. Using these independent predictors of outcome, we were able to build a 3-tiered risk stratification model that separates patients with standard, intermediate and high risk, and which outperforms current stratification procedures. CONCLUSION The integration of defined clinical, histological and genetic parameters led to an improved risk-stratification model for posterior fossa ependymoma of childhood. After validation in independent cohorts this model may provide the basis for risk-adapted treatment of children with ependymomas of the posterior fossa.
Collapse
Affiliation(s)
- Stephanie T Jünger
- Department of Neuropathology, Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn, Sigmund-Freud-St. 25, D-53127, Bonn, Germany
- Present address: Department of Neurosurgery, University of Cologne Medical Center, Cologne, Germany
| | - Martin Mynarek
- Department of Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Inken Wohlers
- Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Present address: Group for Medical Systems Biology, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Evelyn Dörner
- Department of Neuropathology, Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn, Sigmund-Freud-St. 25, D-53127, Bonn, Germany
| | - Anja Zur Mühlen
- Department of Neuropathology, Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn, Sigmund-Freud-St. 25, D-53127, Bonn, Germany
| | - Natalia Velez-Char
- Department of Neuropathology, Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn, Sigmund-Freud-St. 25, D-53127, Bonn, Germany
| | - Katja von Hoff
- Department of Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Rutkowski
- Department of Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Monika Warmuth-Metz
- Institute of Diagnostic and Interventional Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | | | | | - Sven Rahmann
- Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ludger Klein-Hitpass
- Department of Cell Biology (Tumor Research), University of Duisburg-Essen Medical Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Andre O von Bueren
- Department of Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Present address: Division of Pediatric Hematology and Oncology, Department of Pediatrics, Obstetrics and Gynecology, University Hospital of Geneva, Geneva, Switzerland
| | - Torsten Pietsch
- Department of Neuropathology, Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn, Sigmund-Freud-St. 25, D-53127, Bonn, Germany.
| |
Collapse
|
9
|
Blessing MM, Blackburn PR, Krishnan C, Harrod VL, Barr Fritcher EG, Zysk CD, Jackson RA, Milosevic D, Nair AA, Davila JI, Balcom JR, Jenkins RB, Halling KC, Kipp BR, Nageswara Rao AA, Laack NN, Daniels DJ, Macon WR, Ida CM. Desmoplastic Infantile Ganglioglioma: A MAPK Pathway-Driven and Microglia/Macrophage-Rich Neuroepithelial Tumor. J Neuropathol Exp Neurol 2019; 78:1011-1021. [DOI: 10.1093/jnen/nlz086] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Abstract
MAPK pathway activation has been recurrently observed in desmoplastic infantile ganglioglioma/astrocytoma (DIG/DIA) with reported disproportionally low mutation allele frequencies relative to the apparent high tumor content, suggesting that MAPK pathway alterations may be subclonal. We sought to expand the number of molecularly profiled cases and investigate if tumor cell composition could account for the observed low mutation allele frequencies. Molecular (targeted neuro-oncology next-generation sequencing/RNA sequencing and OncoScan microarray) and immunohistochemical (CD68-PGM1/CD163/CD14/CD11c/lysozyme/CD3/CD20/CD34/PD-L1) studies were performed in 7 DIG. Activating MAPK pathway alterations were identified in 4 (57%) cases: 3 had a BRAF mutation (V600E/V600D/V600_W604delinsDQTDG, at 8%–27% variant allele frequency) and 1 showed a TPM3-NTRK1 fusion. Copy number changes were infrequent and nonrecurrent. All tumors had at least 30% of cells morphologically and immunophenotypically consistent with microglial/macrophage lineage. Two subtotally resected tumors regrew; 1 was re-excised and received adjuvant treatment (chemotherapy/targeted therapy), with clinical response to targeted therapy only. Even with residual tumor, all patients are alive (median follow-up, 83 months; 19–139). This study further supports DIG as another MAPK pathway-driven neuroepithelial tumor, thus expanding potential treatment options for tumors not amenable to surgical cure, and suggests that DIG is a microglia/macrophage-rich neuroepithelial tumor with frequent low driver mutation allele frequencies.
Collapse
Affiliation(s)
- Melissa M Blessing
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Patrick R Blackburn
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Chandra Krishnan
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Virginia L Harrod
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Emily G Barr Fritcher
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Christopher D Zysk
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Rory A Jackson
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Dragana Milosevic
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Asha A Nair
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Jaime I Davila
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Jessica R Balcom
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Robert B Jenkins
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Kevin C Halling
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Benjamin R Kipp
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Amulya A Nageswara Rao
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Nadia N Laack
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - David J Daniels
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - William R Macon
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Cristiane M Ida
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| |
Collapse
|
10
|
Wang CI, Kao HK, Chen TW, Huang Y, Cheng HW, Yi JS, Hung SY, Wu CS, Lee YS, Chang KP. Characterization of Copy Number Variations in Oral Cavity Squamous Cell Carcinoma Reveals a Novel Role for MLLT3 in Cell Invasiveness. Oncologist 2019; 24:e1388-e1400. [PMID: 31273053 DOI: 10.1634/theoncologist.2019-0063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/25/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND DNA copy number variations (CNVs) are a hallmark of cancer, and the current study aimed to demonstrate the profile of the CNVs for oral cavity squamous cell carcinoma (OSCC) and elucidate the clinicopathological associations and molecular mechanisms of a potential marker derived from CNVs, mixed-lineage leukemia translocated to chromosome 3 protein (MLLT3), in OSCC carcinogenesis. MATERIALS AND METHODS CNVs in 37 OSCC tissue specimens were analyzed using a high-resolution microarray, the OncoScan array. Gene expression was analyzed by real-time polymerase chain reaction in 127 OSCC and normal tissue samples. Cell function assays included cell cycle, migration, invasion and chromatin immunoprecipitation assays. RESULTS We found a novel copy number amplified region, chromosome 9p, encompassing MLLT3 via the comparison of our data set with six other OSCC genome-wide CNV data sets. MLLT3 overexpression was associated with poorer overall survival in patients with OSCC (p = .048). MLLT3 knockdown reduced cell migration and invasion. The reduced invasion ability in MLLT3-knockdown cells was rescued with double knockdown of MLLT3 and CBP/p300-interacting transactivator with ED rich carboxy-terminal domain 4 (CITED4; 21.0% vs. 61.5%). Knockdown of MLLT3 impaired disruptor of telomeric silencing-1-like (Dot1L)-associated hypermethylation in the promoter of the tumor suppressor, CITED4 (p < .001), and hence dysregulated HIF-1α-mediated genes (TWIST, MMP1, MMP2, VIM, and CDH1) in OSCC cells. CONCLUSION We identified unique CNVs in tumors of Taiwanese patients with OSCC. Notably, MLLT3 overexpression is related to the poorer prognosis of patients with OSCC and is required for Dot1L-mediated transcriptional repression of CITED4, leading to dysregulation of HIF-1α-mediated genes. IMPLICATIONS FOR PRACTICE This article reports unique copy number variations in oral cavity squamous cell carcinoma (OSCC) tumors of Taiwanese patients. Notably, MLLT3 overexpression is related to the poorer prognosis of patients with OSCC and is required for Dot1L-mediated transcriptional repression of CITED4, leading to dysregulation of HIF-1α-mediated genes.
Collapse
Affiliation(s)
- Chun-I Wang
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Huang-Kai Kao
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Wen Chen
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
- Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), National Chiao Tung University, Hsinchu, Taiwan
| | - Yenlin Huang
- Department of Pathology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hsing-Wen Cheng
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jui-Shan Yi
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Shao-Yu Hung
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chi-Sheng Wu
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Yun-Shien Lee
- Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan
| | - Kai-Ping Chang
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
11
|
Ji X, Chen F, Zhou Y, Li J, Yuan Y, Mo Y, Liu Q, Tseng JY, Shih-Chieh Lin D, Shen SH, Liu Y, Ye W, Cheung YN, Yuen KY, Lin S, Fu M, Zhang H, Liu N, Wang J, Yang H, Wang Y, Li S, Fan S, Jin X, Mao M, Sung PL. Copy number variation profile in noninvasive prenatal testing (NIPT) can identify co-existing maternal malignancies: Case reports and a literature review. Taiwan J Obstet Gynecol 2019; 57:871-877. [PMID: 30545544 DOI: 10.1016/j.tjog.2018.10.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2018] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE The coexistence of maternal malignancy and pregnancy has received increasing attention in Noninvasive prenatal testing (NIPT) studies. Malignancy in pregnant women potentially affects the copy number variation (CNV) profile in NIPT results. Only one case of hematologic cancer has been reported in a Hong-Kong pregnant women, and solid tumors have never been reported in pregnant Chinese women. CASE REPORT The patients with dysgerminoma and cervical cancer showed aberrant chromosomal aneuploidies in NIPT and concordant patterns of genome disruption in tumor tissues. The genomic aberrations in the gastric cancer patient had similar copy number variation pattern of gastric cancer. CONCLUSION The findings in this study and the literature review further validate the effect of maternal malignancy on the copy number variation profile in NIPT data and strengthen the possibility of detecting malignant tumors with NIPT in the future.
Collapse
Affiliation(s)
- Xing Ji
- Department of Prenatal Diagnosis, Xin Hua Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai, China; Department of Genetics, Shanghai Institute for Pediatric Research, Shanghai, China
| | - Fang Chen
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | - Jia Li
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | - Yu Mo
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Qiang Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Jen-Yu Tseng
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Diego Shih-Chieh Lin
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shu-Huei Shen
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu Liu
- Department of Prenatal Diagnosis, Xin Hua Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai, China; Department of Genetics, Shanghai Institute for Pediatric Research, Shanghai, China
| | - Weiping Ye
- Department of Obstetrics, Xin Hua Hospital Affiliated with Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | | | - Ka Yiu Yuen
- BGI Health Company Limited, Hong Kong SAR, China
| | - Siyuan Lin
- BGI Health Company Limited, Hong Kong SAR, China
| | - Meili Fu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | - Na Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, Guangdong, China; James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, Guangdong, China; James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | | | - Shen Li
- Department of Pathology, Shanghai Punan Hospital, Shanghai, China.
| | - Shushu Fan
- The Genetic Diagnosis Center and Reproductive Center, Yue Bei People's Hospital, Shao Guan, Guangdong, China.
| | - Xin Jin
- BGI-Shenzhen, Shenzhen, Guangdong, China; School of Medicine, South China University of Technology, Guangzhou, China.
| | - Mao Mao
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Pi-Lin Sung
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
12
|
Martin-Guerrero I, Salaverria I, Burkhardt B, Chassagne-Clement C, Szczepanowski M, Bens S, Klapper W, Zimmermann M, Kabickova E, Bertrand Y, Reiter A, Siebert R, Oschlies I. Non-leukemic pediatric mixed phenotype acute leukemia/lymphoma: Genomic characterization and clinical outcome in a prospective trial for pediatric lymphoblastic lymphoma. Genes Chromosomes Cancer 2018; 58:365-372. [PMID: 30578714 DOI: 10.1002/gcc.22726] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 12/18/2018] [Accepted: 12/18/2018] [Indexed: 11/08/2022] Open
Abstract
Rare cases of hematological precursor neoplasms fulfill the diagnostic criteria of mixed phenotype acute leukemia (MPAL), characterized by expression patterns of at least two hematopoietic lineages, for which a highly aggressive behavior was reported. We present a series of 11 pediatric non-leukemic MPAL identified among 146 precursor lymphoblastic lymphomas included in the prospective trial Euro-LBL 02. Paraffin-embedded biopsies of 10 cases were suitable for molecular analyses using OncoScan assay (n = 7), fluorescence in situ hybridization (FISH; n = 7) or both (n = 5). Except for one case with biallelic KMT2A (MLL) breaks, all cases analyzed by FISH lacked the most common translocations defining molecular subsets of lymphoblastic leukemia/lymphomas. Two non-leukemic B-myeloid MPALs showed the typical genomic profile of hyperdiploid precursor B-cell lymphoblastic leukemia with gains of chromosomes 4, 6, 10, 14, 18, and 21. One B-T MPAL showed typical aberrations of T-cell lymphoblastic lymphoma, such as copy number neutral loss of heterozygosity (CNN-LOH) at 9p targeting a 9p21.3 deletion of CDKN2A and 11q12.2-qter affecting the ATM gene. ATM was also mutated in a T-myeloid MPAL case with additional loss at 7q21.2-q36.3 and mutation of NRAS, two alterations common in myeloid disorders. No recurrent regions of CNN-LOH were observed. The outcome under treatment was good with all patients being alive in first complete remission after treatment according to a protocol for precursor lymphoblastic lymphoma (follow-up 3-10 years, median: 4.9 years). In summary, the present series of non-leukemic MPALs widely lacked recurrently reported translocations in lymphoid/myeloid neoplasias and showed heterogeneous spectrum of chromosomal imbalances.
Collapse
Affiliation(s)
- Idoia Martin-Guerrero
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Campus Kiel/Christian-Albrechts University, Kiel, Germany.,Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country, Leioa, Spain
| | - Itziar Salaverria
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Campus Kiel/Christian-Albrechts University, Kiel, Germany.,Department of Pathology, Hematopathology Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBERONC, University of Barcelona, Barcelona, Spain
| | - Birgit Burkhardt
- NHL-BFM Study Center and Department of Pediatric Hematology and Oncology, University Children's Hospital, Münster, Germany
| | | | - Monika Szczepanowski
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel/Christian-Albrechts University, Kiel, Germany
| | - Susanne Bens
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Campus Kiel/Christian-Albrechts University, Kiel, Germany.,Institute of Human Genetics, Ulm University & Ulm University Medical Center, Ulm, Germany
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel/Christian-Albrechts University, Kiel, Germany
| | - Martin Zimmermann
- Department of Pediatric Hematology and Oncology, Medical School Hannover, Hannover, Germany
| | - Edita Kabickova
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University Prague and University Hospital Motol, Motol, Czech Republic
| | - Yves Bertrand
- Department of Pediatric Hematology, IHOP and Claude Bernard University, Lyon, France
| | - Alfred Reiter
- Department of Pediatric Hematology and Oncology, NHL-BFM-Study Center Justus Liebig University, Germany
| | - Reiner Siebert
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Campus Kiel/Christian-Albrechts University, Kiel, Germany.,Institute of Human Genetics, Ulm University & Ulm University Medical Center, Ulm, Germany
| | - Ilske Oschlies
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel/Christian-Albrechts University, Kiel, Germany
| |
Collapse
|
13
|
Kanagal-Shamanna R, Hodge JC, Tucker T, Shetty S, Yenamandra A, Dixon-McIver A, Bryke C, Huxley E, Lennon PA, Raca G, Xu X, Jeffries S, Quintero-Rivera F, Greipp PT, Slovak ML, Iqbal MA, Fang M. Assessing copy number aberrations and copy neutral loss of heterozygosity across the genome as best practice: An evidence based review of clinical utility from the cancer genomics consortium (CGC) working group for myelodysplastic syndrome, myelodysplastic/myeloproliferative and myeloproliferative neoplasms. Cancer Genet 2018; 228-229:197-217. [PMID: 30377088 DOI: 10.1016/j.cancergen.2018.07.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 12/16/2022]
Abstract
Multiple studies have demonstrated the utility of chromosomal microarray (CMA) testing to identify clinically significant copy number alterations (CNAs) and copy-neutral loss-of-heterozygosity (CN-LOH) in myeloid malignancies. However, guidelines for integrating CMA as a standard practice for diagnostic evaluation, assessment of prognosis and predicting treatment response are still lacking. CMA has not been recommended for clinical work-up of myeloid malignancies by the WHO 2016 or the NCCN 2017 guidelines but is a suggested test by the European LeukaemiaNet 2013 for the diagnosis of primary myelodysplastic syndrome (MDS). The Cancer Genomics Consortium (CGC) Working Group for Myeloid Neoplasms systematically reviewed peer-reviewed literature to determine the power of CMA in (1) improving diagnostic yield, (2) refining risk stratification, and (3) providing additional genomic information to guide therapy. In this manuscript, we summarize the evidence base for the clinical utility of array testing in the workup of MDS, myelodysplastic/myeloproliferative neoplasms (MDS/MPN) and myeloproliferative neoplasms (MPN). This review provides a list of recurrent CNAs and CN-LOH noted in this disease spectrum and describes the clinical significance of the aberrations and how they complement gene mutation findings by sequencing. Furthermore, for new or suspected diagnosis of MDS or MPN, we present suggestions for integrating genomic testing methods (CMA and mutation testing by next generation sequencing) into the current standard-of-care clinical laboratory testing (karyotype, FISH, morphology, and flow).
Collapse
Affiliation(s)
- Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston TX, USA.
| | - Jennelle C Hodge
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, USA; Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Tracy Tucker
- Department of Pathology and Laboratory Medicine, Cancer Genetics Laboratory, British Columbia Cancer Agency, Vancouver, BC Canada
| | - Shashi Shetty
- Department of Pathology, UHCMC, University Hospitals and Case Western Reserve University, Cleveland, OH, USA
| | - Ashwini Yenamandra
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Christine Bryke
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Emma Huxley
- West Midlands Regional Genetics Laboratory, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | | | - Gordana Raca
- Department of Pathology and Laboratory Medicine, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - Xinjie Xu
- ARUP Laboratories, University of Utah, Salt Lake City, UT, USA
| | - Sally Jeffries
- West Midlands Regional Genetics Laboratory, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Fabiola Quintero-Rivera
- Department of Pathology and Laboratory Medicine, UCLA Clinical Genomics Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Patricia T Greipp
- Department of Laboratory Medicine and Pathology, Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Marilyn L Slovak
- TriCore Reference Laboratories, University of New Mexico, Albuquerque, NM, USA
| | - M Anwar Iqbal
- University of Rochester Medical Center, Rochester, NY, USA
| | - Min Fang
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA, USA.
| |
Collapse
|
14
|
Emanuel PO, Andea AA, Vidal CI, Missall TA, Novoa RA, Bohlke AK, Hughes SR, Hurley MY, Kim J. Evidence behind the use of molecular tests in melanocytic lesions and practice patterns of these tests by dermatopathologists. J Cutan Pathol 2018; 45:839-846. [DOI: 10.1111/cup.13327] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 06/24/2018] [Accepted: 07/05/2018] [Indexed: 12/21/2022]
Affiliation(s)
| | - Aleodor A. Andea
- Departments of Dermatology and Pathology; University of Michigan Medical Center; Ann Arbor Michigan
| | - Claudia I. Vidal
- Departments of Dermatology and Pathology; Saint Louis University School of Medicine; St. Louis Missouri
| | - Tricia A. Missall
- Departments of Dermatology and Pathology; Saint Louis University School of Medicine; St. Louis Missouri
| | - Roberto A. Novoa
- Departments of Dermatology and Pathology; Stanford University School of Medicine; Stanford California
| | | | - Sarah R. Hughes
- Department of Pathology; Gundersen Health System; La Crosse Wisconsin
| | - Maria Y. Hurley
- Departments of Dermatology and Pathology; Saint Louis University School of Medicine; St. Louis Missouri
| | - Jinah Kim
- Departments of Dermatology and Pathology; Stanford University School of Medicine; Stanford California
| |
Collapse
|
15
|
Quality and concordance of genotyping array data of 12,064 samples from 5840 cancer patients. Genomics 2018; 111:950-957. [PMID: 29902512 DOI: 10.1016/j.ygeno.2018.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/21/2018] [Accepted: 06/05/2018] [Indexed: 12/21/2022]
Abstract
Genotyping arrays characterize genome-wide SNPs for a study cohort and were the primary technology behind genome wide association studies over the last decade. The Cancer Genome Atlas (TCGA) is one of the largest cancer consortium studies, and it collected genotyping data for all of its participants. Using TCGA SNP data genotyped using the Affymetrix 6.0 SNP array from 12,064 samples, we conducted a comprehensive comparisons across DNA sources (tumor tissue, normal tissue, and blood) and sample storage protocols (formalin-fixed paraffin-embedded (FFPE) vs. freshly frozen (FF)), examining genotypes, transition/transversion ratios, and mutation catalogues. During the analysis, we made important observations in relevance to the data quality issues. SNP concordance was excellent between blood and normal tissues, and slightly lower between blood and tumor tissue due to potential somatic mutations in the tumors. The observed poor SNP concordance between FFPE and FF samples suggested a batch effect. The transition/transversion ratio, a metric commonly used for quality control purpose in exome sequencing projects, appeared less applicable for genotyping array data due to the whole-genome coverage built into the array design. Moreover, there were substantially more loss of heterozygosity events than gain of heterozygosity when comparing tumors relative to normal tissues and blood. This might be a consequence of extensive copy number deletions in tumors. In summary, our thorough evaluation calls for more adequate quality control practices and provides guidelines for improved application of TCGA genotyping data.
Collapse
|
16
|
Rotolo F, Zhu CQ, Brambilla E, Graziano SL, Olaussen K, Le-Chevalier T, Pignon JP, Kratzke R, Soria JC, Shepherd FA, Seymour L, Michiels S, Tsao MS. Genome-wide copy number analyses of samples from LACE-Bio project identify novel prognostic and predictive markers in early stage non-small cell lung cancer. Transl Lung Cancer Res 2018; 7:416-427. [PMID: 30050779 DOI: 10.21037/tlcr.2018.05.01] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Adjuvant chemotherapy (ACT) provides modest benefit in resected non-small cell lung cancer (NSCLC) patients. Genome-wide studies have identified gene copy number aberrations (CNA), but their prognostic implication is unknown. Methods DNA from 1,013 FFPE tumor samples from three pivotal multicenter randomized trials (ACT vs. control) in the LACE-Bio consortium (median follow-up: 5.2 years) was successfully extracted, profiled using a molecular inversion probe SNP assay, normalized relative to a pool of normal tissues and segmented. Minimally recurrent regions were identified. P values were adjusted to control the false discovery rate (Q values). Results A total of 976 samples successfully profiled, 414 (42%) adenocarcinoma (ADC), 430 (44%) squamous cell carcinoma (SCC) and 132 (14%) other NSCLC; 710 (73%) males. We identified 431 recurrent regions, with on average 51 gains and 43 losses; 253 regions (59%) were ≤3 Mb. Most frequent gains (up to 48%) were on chr1, 3q, 5p, 6p, 8q, 22q; most frequent losses (up to 40%) on chr3p, 8p, 9p. CNA frequency of 195 regions was significantly different (Q≤0.05) between ADC and SCC. Fourteen regions (7p11-12, 9p21, 18q12, and 19p11-13) were associated with disease-free survival (DFS) (univariate P≤0.005, Q<0.142), with poorer DFS for losses of regions including CDKN2A/B [hazard ratio (HR) for 2-fold lower CN: 1.5 (95% CI: 1.2-1.9), P<0.001, Q=0.020] and STK11 [HR =2.4 (1.3-4.3), P=0.005, Q=0.15]. Chromosomal instability was associated with poorer DFS (HR =1.5, P=0.015), OS (HR =1.2, P=0.189) and lung-cancer specific survival (HR =1.7, P=0.003). Conclusions These large-scale genome-wide analyses of gene CNA provide new candidate prognostic markers for stage I-III NSCLC.
Collapse
Affiliation(s)
- Federico Rotolo
- Gustave Roussy, Université Paris-Saclay, Service de Biostatistique et d'Epidémiologie, Villejuif, France.,Université Paris-Saclay, Univ. Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France.,Ligue Nationale Contre le Cancer Meta-Analysis Platform, Gustave Roussy, Villejuif, France
| | - Chang-Qi Zhu
- University Health Network, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Elisabeth Brambilla
- Department of Pathology, Institut Albert Bonniot, Hopital Albert Michallon, Grenoble, France
| | | | - Ken Olaussen
- INSERM U981, Université Paris-Sud, Université Paris-Saclay and Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Jean-Pierre Pignon
- Gustave Roussy, Université Paris-Saclay, Service de Biostatistique et d'Epidémiologie, Villejuif, France.,Université Paris-Saclay, Univ. Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France.,Ligue Nationale Contre le Cancer Meta-Analysis Platform, Gustave Roussy, Villejuif, France
| | - Robert Kratzke
- Department of Medical Oncology, University of Minnesota, Minneapolis, MN, USA
| | - Jean-Charles Soria
- INSERM U981, Université Paris-Sud, Université Paris-Saclay and Gustave Roussy Cancer Campus, Villejuif, France.,Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Frances A Shepherd
- University Health Network, Princess Margaret Cancer Centre, Toronto, ON, Canada.,Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, ON, Canada
| | - Lesley Seymour
- Canadian Cancer Trials Group and Queen's University, Kingston, ON, Canada
| | - Stefan Michiels
- Gustave Roussy, Université Paris-Saclay, Service de Biostatistique et d'Epidémiologie, Villejuif, France.,Université Paris-Saclay, Univ. Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France.,Ligue Nationale Contre le Cancer Meta-Analysis Platform, Gustave Roussy, Villejuif, France
| | - Ming-Sound Tsao
- University Health Network, Princess Margaret Cancer Centre, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Gray PN, Vuong H, Tsai P, Lu HM, Mu W, Hsuan V, Hoo J, Shah S, Uyeda L, Fox S, Patel H, Janicek M, Brown S, Dobrea L, Wagman L, Plimack E, Mehra R, Golemis EA, Bilusic M, Zibelman M, Elliott A. TumorNext: A comprehensive tumor profiling assay that incorporates high resolution copy number analysis and germline status to improve testing accuracy. Oncotarget 2018; 7:68206-68228. [PMID: 27626691 PMCID: PMC5356550 DOI: 10.18632/oncotarget.11910] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/26/2016] [Indexed: 01/08/2023] Open
Abstract
The development of targeted therapies for both germline and somatic DNA mutations has increased the need for molecular profiling assays to determine the mutational status of specific genes. Moreover, the potential of off-label prescription of targeted therapies favors classifying tumors based on DNA alterations rather than traditional tissue pathology. Here we describe the analytical validation of a custom probe-based NGS tumor panel, TumorNext, which can detect single nucleotide variants, small insertions and deletions in 142 genes that are frequently mutated in somatic and/or germline cancers. TumorNext also detects gene fusions and structural variants, such as tandem duplications and inversions, in 15 frequently disrupted oncogenes and tumor suppressors. The assay uses a matched control and custom bioinformatics pipeline to differentiate between somatic and germline mutations, allowing precise variant classification. We tested 170 previously characterized samples, of which > 95% were formalin-fixed paraffin embedded tissue from 8 different cancer types, and highlight examples where lack of germline status may have led to the inappropriate prescription of therapy. We also describe the validation of the Affymetrix OncoScan platform, an array technology for high resolution copy number variant detection for use in parallel with the NGS panel that can detect single copy amplifications and hemizygous deletions. We analyzed 80 previously characterized formalin-fixed paraffin-embedded specimens and provide examples of hemizygous deletion detection in samples with known pathogenic germline mutations. Thus, the TumorNext combined approach of NGS and OncoScan potentially allows for the identification of the “second hit” in hereditary cancer patients.
Collapse
Affiliation(s)
| | - Huy Vuong
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | - Pei Tsai
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | | | - Wenbo Mu
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | | | - Jayne Hoo
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | - Swati Shah
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | - Lisa Uyeda
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | | | | | | | | | | | | | | | - Ranee Mehra
- Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | | | | | | | | |
Collapse
|
18
|
Husain H, Nykin D, Bui N, Quan D, Gomez G, Woodward B, Venkatapathy S, Duttagupta R, Fung E, Lippman SM, Kurzrock R. Cell-Free DNA from Ascites and Pleural Effusions: Molecular Insights into Genomic Aberrations and Disease Biology. Mol Cancer Ther 2018; 16:948-955. [PMID: 28468865 DOI: 10.1158/1535-7163.mct-16-0436] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/21/2016] [Accepted: 12/07/2016] [Indexed: 11/16/2022]
Abstract
Collection of cell-free DNA (cfDNA) from the blood of individuals with cancer has permitted noninvasive tumor genome analysis. Detection and characterization of cfDNA in ascites and pleural effusions have not yet been reported. Herein, we analyzed cfDNA in the ascites and pleural effusions from six individuals with metastatic cancer. In all cases, cfDNA copy number variations (CNV) were discovered within the effusate. One individual had a relevant alteration with a high copy amplification in EGFR in a never smoker with lung cancer, who showed only MDM2 and CDK4 amplification in a prior tissue biopsy. Another subject with metastatic breast cancer had cytology-positive ascites and an activating PIK3CA mutation identified in the tissue, blood, and ascites collectively. This individual had tumor regression after the administration of the mTOR inhibitor everolimus and had evidence of chromotripsis from chromosomal rearrangements noted in the cell-free ascitic fluid. These results indicate that cfDNA from ascites and pleural effusions may provide additional information not detected with tumor and plasma cell-free DNA molecular characterization, and a context for important insights into tumor biology and clonal dynamic change within primary tumor and metastatic deposits. Mol Cancer Ther; 16(5); 948-55. ©2017 AACR.
Collapse
Affiliation(s)
- Hatim Husain
- Division of Hematology and Oncology, University of California San Diego, San Diego, California.
| | - David Nykin
- Division of Hematology and Oncology, University of California San Diego, San Diego, California
| | - Nam Bui
- Division of Hematology and Oncology, University of California San Diego, San Diego, California
| | - Daniel Quan
- Division of Hematology and Oncology, University of California San Diego, San Diego, California
| | - German Gomez
- Division of Hematology and Oncology, University of California San Diego, San Diego, California
| | - Brian Woodward
- Division of Hematology and Oncology, University of California San Diego, San Diego, California
| | | | | | - Eric Fung
- ThermoFisher Scientific, Inc, Santa Clara, California
| | - Scott M Lippman
- Division of Hematology and Oncology, University of California San Diego, San Diego, California
| | - Razelle Kurzrock
- Division of Hematology and Oncology, University of California San Diego, San Diego, California
| |
Collapse
|
19
|
Yu MC, Lee CW, Lee YS, Lian JH, Tsai CL, Liu YP, Wu CH, Tsai CN. Prediction of early-stage hepatocellular carcinoma using OncoScan chromosomal copy number aberration data. World J Gastroenterol 2017; 23:7818-7829. [PMID: 29209123 PMCID: PMC5703911 DOI: 10.3748/wjg.v23.i44.7818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/26/2017] [Accepted: 11/07/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To identify chromosomal copy number aberrations (CNAs) in early-stage hepatocellular carcinoma (HCC) and analyze whether they are correlated with patient prognosis.
METHODS One hundred and twenty patients with early-stage HCC were enrolled in our study, with the collection of formalin fixed, paraffin-embedded (FFPE) specimens and clinicopathological data. Tumor areas were marked by certified pathologists on a hematoxylin and eosin-stained slide, and cancer and adjacent non-cancerous tissues underwent extraction of DNA, which was analyzed with the Affymetrix OncoScan platform to assess CNAs and loss of heterozygosity (LOH). Ten individuals with nonmalignant disease were used as the control group. Another cohort consisting of 40 patients with stage I/II HCC were enrolled to analyze gene expression and to correlate findings with the OncoScan data.
RESULTS Copy number amplifications occurred at chromosomes 1q21.1-q44 and 8q12.3-24.3 and deletions were found at 4q13.1-q35.2, 8p 23.2-21.1, 16q23.3-24.3, and 17p13.3-12, while LOH commonly occurred at 1p32.3, 3p21.31, 8p23.2-21.1, 16q22.1-24.3, and 17p 13.3-11 in early-stage HCC. Using Cox regression analysis, we also found that a higher percentage of genome change (≥ 60%) was an independent factor for worse prognosis in early-stage HCC (P = 0.031). Among the 875 genes in the OncoScan GeneChip, six were independent predictors of worse disease-free survival, of which three were amplified (MYC, ELAC2, and SYK) and three were deleted (GAK, MECOM, and WRN). Further, patients with HCC who exhibited ≥ 3 CNAs involving these six genes have worse outcomes compared to those who had < 3 CNAs (P < 0.001). Similarly, Asian patients with stage I HCC from The Cancer Genome Atlas harboring CNAs with these genes were also predicted to have poorer outcomes.
CONCLUSION Patients with early-stage HCC and increased genome change or CNAs involving MYC, ELAC2, SYK, GAK, MECOM, or WRN are at risk for poorer outcome after resection.
Collapse
Affiliation(s)
- Ming-Chin Yu
- Department of Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department of Surgery, Xiamen Chang Gung Hospital, Xiamen 361028, China
| | - Chao-Wei Lee
- Department of Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yun-Shien Lee
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department of Biotechnology, Ming-Chuan University, Taoyuan 33348, Taiwan
| | - Jang-Hau Lian
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Chia-Lung Tsai
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yi-Ping Liu
- Department of Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Chun-Hsing Wu
- Department of Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Chi-Neu Tsai
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| |
Collapse
|
20
|
Gliem TJ, Aypar U. Development of a Chromosomal Microarray Test for the Detection of Abnormalities in Formalin-Fixed, Paraffin-Embedded Products of Conception Specimens. J Mol Diagn 2017; 19:843-847. [DOI: 10.1016/j.jmoldx.2017.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/26/2017] [Accepted: 07/10/2017] [Indexed: 10/19/2022] Open
|
21
|
Hoekstra AS, Hensen EF, Jordanova ES, Korpershoek E, van der Horst-Schrivers AN, Cornelisse C, Corssmit EPM, Hes FJ, Jansen JC, Kunst HPM, Timmers HJLM, Bateman A, Eccles D, Bovée JVMG, Devilee P, Bayley JP. Loss of maternal chromosome 11 is a signature event in SDHAF2, SDHD, and VHL-related paragangliomas, but less significant in SDHB-related paragangliomas. Oncotarget 2017; 8:14525-14536. [PMID: 28099933 PMCID: PMC5362423 DOI: 10.18632/oncotarget.14649] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 01/04/2017] [Indexed: 12/28/2022] Open
Abstract
Germline mutations in the succinate dehydrogenase (SDHA, SDHB, SDHC, SDHD, SDHAF2) or Von Hippel-Lindau (VHL) genes cause hereditary paraganglioma/pheochromocytoma. While SDHB (1p36) and VHL (3p25) are associated with autosomal dominant disease, SDHD (11q23) and SDHAF2 (11q13) show a remarkable parent-of-origin effect whereby tumor formation is almost completely dependent on paternal transmission of the mutant allele. Loss of the entire maternal copy of chromosome 11 occurs frequently in SDHD-linked tumors, and has been suggested to be the basis for this typical inheritance pattern.Using fluorescent in situ hybridization, microsatellite marker and SNP array analysis, we demonstrate that loss of the entire copy of chromosome 11 is also frequent in SDHAF2-related PGLs, occurring in 89% of tumors. Analysis of two imprinted differentially methylated regions (DMR) in 11p15, H19-DMR and KvDMR, showed that this loss always affected the maternal copy of chromosome 11. Likewise, loss of maternal chromosome 11p15 was demonstrated in 85% of SDHD and 75% of VHL-related PGLs/PCCs. By contrast, both copies of chromosome 11 were found to be retained in 62% of SDHB-mutated PGLs/PCCs, while only 31% showed loss of maternal chromosome 11p15. Genome-wide copy number analysis revealed frequent loss of 1p in SDHB mutant tumors and show greater genomic instability compared to SDHD and SDHAF2.These results show that loss of the entire copy of maternal chromosome 11 is a highly specific and statistically significant event in SDHAF2, SDHD and VHL-related PGLs/PCCs, but is less significant in SDHB-mutated tumors, suggesting that these tumors have a distinct genetic etiology.
Collapse
Affiliation(s)
- Attje S Hoekstra
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik F Hensen
- Department of Otolaryngology/Head and Neck Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Esther Korpershoek
- Department of Pathology, Josephine Nefkens Institute, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Cees Cornelisse
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eleonora P M Corssmit
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Frederik J Hes
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen C Jansen
- Department of Otorhinolaryngology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henricus P M Kunst
- Department of Otorhinolaryngology, Head and Neck Surgery, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Henri J L M Timmers
- Department of Medicine, Division of Endocrinology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Adrian Bateman
- Department of Cellular Pathology, University Hospital Southampton, Southampton, UK
| | - Diana Eccles
- University of Southampton School of Medicine, Cancer Sciences Division, Somers Cancer Research Building, Southampton, UK
| | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Devilee
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jean-Pierre Bayley
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
22
|
Genetic evaluation of juvenile xanthogranuloma: genomic abnormalities are uncommon in solitary lesions, advanced cases may show more complexity. Mod Pathol 2017; 30:1234-1240. [PMID: 28752840 DOI: 10.1038/modpathol.2017.50] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 04/10/2017] [Accepted: 04/19/2017] [Indexed: 12/14/2022]
Abstract
Juvenile xanthogranuloma is a rare histiocytic proliferation primarily affecting infants and young children, characterized by aberrant infiltration of histiocyte-derived cells in the skin, soft tissues and more rarely, visceral organs. Juvenile xanthogranuloma is generally considered to be a benign disorder; most lesions are solitary cutaneous nodules that resolve spontaneously without treatment. However, cases with extracutaneous involvement, multiple lesions, and/or systemic disease often require aggressive therapy. Though molecular studies have provided evidence of clonality in juvenile xanthogranuloma, in support of a neoplastic process, little is known about the genetic profile of juvenile xanthogranuloma. We used molecular inversion probe array technology to evaluate the genomic characteristics (copy number alterations or copy neutral-loss of heterozygosity) of 21 archived cases of juvenile xanthogranuloma (19 solitary, 1 diffuse cutaneous, 1 systemic). Four cases (19%) showed acquired, clonal alterations. Two lesions from a case of diffuse cutaneous juvenile xanthogranuloma showed distinct profiles: JXG-1a contained trisomy 5 and 17 and JXG-1b contained loss of heterozygosity in 5q. The systemic juvenile xanthogranuloma (JXG-2) showed multiple genomic alterations. Only two of 19 solitary juvenile xanthogranulomas showed abnormal genomic profiles: JXG-3 showed gains on 1q and 11q and JXG-4 showed a 7.2 Mb loss in 3p. No recurrent abnormalities were observed among these cases. The presence of non-recurrent copy number alterations in a subset of samples implies that copy number changes are unlikely driving pathogenesis in juvenile xanthogranuloma, but may be acquired during disease progression. The presence of genomic abnormalities in more advanced cases (ie, systemic and diffuse cutaneous juvenile xanthogranuloma) supports this notion, particularly as the advanced cases of juvenile xanthogranuloma presented more genomic complexity.
Collapse
|
23
|
Iddawela M, Rueda O, Eremin J, Eremin O, Cowley J, Earl HM, Caldas C. Integrative analysis of copy number and gene expression in breast cancer using formalin-fixed paraffin-embedded core biopsy tissue: a feasibility study. BMC Genomics 2017; 18:526. [PMID: 28697743 PMCID: PMC5506605 DOI: 10.1186/s12864-017-3867-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/16/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND An absence of reliable molecular markers has hampered individualised breast cancer treatments, and a major limitation for translational research is the lack of fresh tissue. There are, however, abundant banks of formalin-fixed paraffin-embedded (FFPE) tissue. This study evaluated two platforms available for the analysis of DNA copy number and gene expression using FFPE samples. METHODS The cDNA-mediated annealing, selection, extension, and ligation assay (DASL™) has been developed for gene expression analysis and the Molecular Inversion Probes assay (Oncoscan™), were used for copy number analysis using FFPE tissues. Gene expression and copy number were evaluated in core-biopsy samples from patients with breast cancer undergoing neoadjuvant chemotherapy (NAC). RESULTS Forty-three core-biopsies were evaluated and characteristic copy number changes in breast cancers, gains in 1q, 8q, 11q, 17q and 20q and losses in 6q, 8p, 13q and 16q, were confirmed. Regions that frequently exhibited gains in tumours showing a pathological complete response (pCR) to NAC were 1q (55%), 8q (40%) and 17q (40%), whereas 11q11 (37%) gain was the most frequent change in non-pCR tumours. Gains associated with poor survival were 11q13 (62%), 8q24 (54%) and 20q (47%). Gene expression assessed by DASL correlated with immunohistochemistry (IHC) analysis for oestrogen receptor (ER) [area under the curve (AUC) = 0.95], progesterone receptor (PR)(AUC = 0.90) and human epidermal growth factor type-2 receptor (HER-2) (AUC = 0.96). Differential expression analysis between ER+ and ER- cancers identified over-expression of TTF1, LAF-4 and C-MYB (p ≤ 0.05), and between pCR vs non-pCRs, over-expression of CXCL9, AREG, B-MYB and under-expression of ABCG2. CONCLUSION This study was an integrative analysis of copy number and gene expression using FFPE core biopsies and showed that molecular marker data from FFPE tissues were consistent with those in previous studies using fresh-frozen samples. FFPE tissue can provide reliable information and will be a useful tool in molecular marker studies. TRIAL REGISTRATION Trial registration number ISRCTN09184069 and registered retrospectively on 02/06/2010.
Collapse
Affiliation(s)
- Mahesh Iddawela
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge, UK
- Cambridge Breast Unit, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre, Cambridge, UK
- Department of Anatomy & Developmental Biology, Monash University, Clayton, VIC 3800 Australia
- School of Clinical Sciences, Monash University, Clayton, Australia
| | - Oscar Rueda
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| | - Jenny Eremin
- Research and Development, Lincoln Breast Unit, Lincoln County Hospital, Lincoln, UK
- Nottingham Digestive Disease Centre, Faculty of Medicine and Health Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, UK
| | - Oleg Eremin
- Research and Development, Lincoln Breast Unit, Lincoln County Hospital, Lincoln, UK
- Nottingham Digestive Disease Centre, Faculty of Medicine and Health Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, UK
| | - Jed Cowley
- PathLinks, Lincoln County Hospital, Lincoln, UK
| | - Helena M. Earl
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge, UK
- Cambridge Breast Unit, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre, Cambridge, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge, UK
- Cambridge Breast Unit, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre, Cambridge, UK
| |
Collapse
|
24
|
Emami NC, Leong L, Wan E, Van Blarigan EL, Cooperberg MR, Tenggara I, Carroll PR, Chan JM, Witte JS, Simko JP. Tissue Sources for Accurate Measurement of Germline DNA Genotypes in Prostate Cancer Patients Treated With Radical Prostatectomy. Prostate 2017; 77:425-434. [PMID: 27900799 PMCID: PMC5479703 DOI: 10.1002/pros.23283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/03/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND Benign tissue from a tumor-containing organ is commonly the only available source for obtaining a patient's unmutated genome for use in cancer research. While it is critical to identify histologically normal tissue that is independent of the tumor lineage, few additional considerations are applied to the choice of this material for such measurements. METHODS Normal formalin-fixed, paraffin-embedded seminal vesicle, and urethral tissues, in addition to whole blood, were collected from 31 prostate cancer patients having undergone radical prostatectomy. Genotype concordance was evaluated for DNA from each tissue source in relation to whole blood. RESULTS Overall, there was a greater genotype call rate for DNA derived from urethral tissue (97.0%) in comparison with patient-matched seminal vesicle tissues (95.9%, P = 0.0015). Furthermore, with reference to patient-matched whole blood, urethral samples exhibited higher genotype concordance (94.1%) than that of seminal vesicle samples (92.5%, P = 0.035). CONCLUSIONS These findings highlight the heterogeneity between diverse sources of DNA in genotype measurement and motivate the consideration of normal tissue biases in tumor-normal analyses. Prostate 77: 425-434, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nima C. Emami
- Program in Biological and Medical Informatics, University of California, San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Lancelote Leong
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Eunice Wan
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| | - Erin L. Van Blarigan
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Matthew R. Cooperberg
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Imelda Tenggara
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Peter R. Carroll
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - June M. Chan
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - John S. Witte
- Program in Biological and Medical Informatics, University of California, San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Correspondence to: Jeffry P. Simko, 1825 4th St., Room M2360, San Francisco, CA 94158, 415-353-7171 (Phone), 415-353-7094 (Fax), . John S. Witte, 1450 3rd St., San Francisco, CA 94158, 415-502-6882 (Phone), 415-476-1356 (Fax),
| | - Jeffry P. Simko
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Anatomic Pathology, University of California, San Francisco, San Francisco, California
- Correspondence to: Jeffry P. Simko, 1825 4th St., Room M2360, San Francisco, CA 94158, 415-353-7171 (Phone), 415-353-7094 (Fax), . John S. Witte, 1450 3rd St., San Francisco, CA 94158, 415-502-6882 (Phone), 415-476-1356 (Fax),
| |
Collapse
|
25
|
Genomic characteristics of trastuzumab-resistant Her2-positive metastatic breast cancer. J Cancer Res Clin Oncol 2017; 143:1255-1262. [PMID: 28247034 PMCID: PMC5486569 DOI: 10.1007/s00432-017-2358-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 01/27/2017] [Indexed: 12/22/2022]
Abstract
Purpose Resistance to trastuzumab therapy is linked to phosphoinositol 3-kinase (PI3K) pathway activation. One key downstream effector and regulator of this pathway is the mechanistic target of rapamycin (mTOR). In 2011, a phase I/II study evaluated the combination of trastuzumab and everolimus (a mTOR inhibitor) for treatment of Her2-positive metastatic breast cancer (MBC) for patients who had progressed on trastuzumab-based therapy. Methods We retrospectively analyzed GeneChip microarray data from 22 of 47 patients included in the study. Results Using an unbiased approach, we found that mutations in BRAF, EGFR and KIT are significantly more common in this heavily treated population when compared with the cohort of invasive breast carcinoma patients in The Cancer Genome Atlas (TCGA). Furthermore, 10 out of 22 patients had PIK3CA mutations (45.4%) but PI3KCA status was not predictive of PFS in our cohort. Finally, the use of OncoScantm has allowed us to detected mutations in five genes that have not been shown to be mutated in TCGA subset of Her-2 overexpressing breast cancer: CTNNB1, HRAS, KRAS, NF2 and SMARCB1. Conclusion Mutational burden in heavily treated trastuzumab-resistant Her2-positive metastatic breast cancer is highly variable and not directly correlated with outcome. Activation of the MAPK/ERK pathway through mutations in EGFR, BRAF or KIT may mediate resistance to trastuzumab.
Collapse
|
26
|
Chen H, Singh RR, Lu X, Huo L, Yao H, Aldape K, Abraham R, Virani S, Mehrotra M, Mishra BM, Bousamra A, Albarracin C, Wu Y, Roy-Chowdhuri S, Shamanna RK, Routbort MJ, Medeiros LJ, Patel KP, Broaddus R, Sahin A, Luthra R. Genome-wide copy number aberrations and HER2 and FGFR1 alterations in primary breast cancer by molecular inversion probe microarray. Oncotarget 2017; 8:10845-10857. [PMID: 28125801 PMCID: PMC5355228 DOI: 10.18632/oncotarget.14802] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/10/2017] [Indexed: 11/25/2022] Open
Abstract
Breast cancer remains the second leading cause of cancer-related death in women despite stratification based on standard hormonal receptor (HR) and HER2 testing. Additional prognostic markers are needed to improve breast cancer treatment. Chromothripsis, a catastrophic genome rearrangement, has been described recently in various cancer genomes and affects cancer progression and prognosis. However, little is known about chromothripsis in breast cancer. To identify novel prognostic biomarkers in breast cancer, we used molecular inversion probe (MIP) microarray to explore genome-wide copy number aberrations (CNA) and breast cancer-related gene alterations in DNA extracted from formalin-fixed paraffin-embedded tissue. We examined 42 primary breast cancers with known HR and HER2 status assessed via immunohistochemistry and FISH and analyzed MIP microarray results for correlation with standard tests and survival outcomes. Global genome-wide CNA ranged from 0.2% to 65.7%. Chromothripsis-like patterns were observed in 23/38 (61%) cases and were more prevalent in cases with ≥10% CNA (20/26, 77%) than in cases with <10% CNA (3/12, 25%; p<0.01). Most frequently involved chromosomal segment was 17q12-q21, the HER2 locus. Chromothripsis-like patterns involving 17q12 were observed in 8/19 (42%) of HER2-amplified tumors but not in any of the tumors without HER2 amplification (0/19; p<0.01). HER2 amplification detected by MIP microarray was 95% concordant with conventional testing (39/41). Interestingly, 21% of patients (9/42) had fibroblast growth factor receptor 1 (FGFR1)amplification and had a 460% higher risk for mortality than those without FGFR1 amplification (p<0.01). In summary, MIP microarray provided a robust assessment of genomic CNA of breast cancer.
Collapse
MESH Headings
- Adult
- Aged
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Breast Neoplasms, Male/genetics
- Breast Neoplasms, Male/metabolism
- Breast Neoplasms, Male/pathology
- Chromosomes, Human, Pair 17/genetics
- Chromothripsis
- DNA Copy Number Variations
- Female
- Gene Amplification
- Genome-Wide Association Study/methods
- High-Throughput Nucleotide Sequencing
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Male
- Microarray Analysis/methods
- Middle Aged
- Molecular Probes/genetics
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Survival Analysis
Collapse
Affiliation(s)
- Hui Chen
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rajesh R. Singh
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xinyan Lu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lei Huo
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hui Yao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth Aldape
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Anatomic Pathology, Laboratory Medicine Program, University Health Network, Toronto, Canada
| | - Ronald Abraham
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shumaila Virani
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meenakshi Mehrotra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bal Mukund Mishra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alex Bousamra
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Allegheny Health Network, Pittsburgh, PA, USA
| | - Constance Albarracin
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yun Wu
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sinchita Roy-Chowdhuri
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rashmi Kanagal Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark J. Routbort
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L. Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur P. Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Russell Broaddus
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aysegul Sahin
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
27
|
The Utilization of Formalin Fixed-Paraffin-Embedded Specimens in High Throughput Genomic Studies. Int J Genomics 2017; 2017:1926304. [PMID: 28246590 PMCID: PMC5299160 DOI: 10.1155/2017/1926304] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/09/2017] [Indexed: 01/09/2023] Open
Abstract
High throughput genomic assays empower us to study the entire human genome in short time with reasonable cost. Formalin fixed-paraffin-embedded (FFPE) tissue processing remains the most economical approach for longitudinal tissue specimen storage. Therefore, the ability to apply high throughput genomic applications to FFPE specimens can expand clinical assays and discovery. Many studies have measured the accuracy and repeatability of data generated from FFPE specimens using high throughput genomic assays. Together, these studies demonstrate feasibility and provide crucial guidance for future studies using FFPE specimens. Here, we summarize the findings of these studies and discuss the limitations of high throughput data generated from FFPE specimens across several platforms that include microarray, high throughput sequencing, and NanoString.
Collapse
|
28
|
Jung HS, Lefferts JA, Tsongalis GJ. Utilization of the oncoscan microarray assay in cancer diagnostics. ACTA ACUST UNITED AC 2017. [DOI: 10.1186/s41241-016-0007-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
29
|
Kader T, Goode DL, Wong SQ, Connaughton J, Rowley SM, Devereux L, Byrne D, Fox SB, Mir Arnau G, Tothill RW, Campbell IG, Gorringe KL. Copy number analysis by low coverage whole genome sequencing using ultra low-input DNA from formalin-fixed paraffin embedded tumor tissue. Genome Med 2016; 8:121. [PMID: 27846907 PMCID: PMC5111221 DOI: 10.1186/s13073-016-0375-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 10/26/2016] [Indexed: 11/10/2022] Open
Abstract
Unlocking clinically translatable genomic information, including copy number alterations (CNA), from formalin-fixed paraffin-embedded (FFPE) tissue is challenging due to low yields and degraded DNA. We describe a robust, cost-effective low-coverage whole genome sequencing (LC WGS) method for CNA detection using 5 ng of FFPE-derived DNA. CN profiles using 100 ng or 5 ng input DNA were highly concordant and comparable with molecular inversion probe (MIP) array profiles. LC WGS improved CN profiles of samples that performed poorly using MIP arrays. Our technique enables identification of driver and prognostic CNAs in archival patient samples previously deemed unsuitable for genomic analysis due to DNA limitations.
Collapse
Affiliation(s)
- Tanjina Kader
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - David L Goode
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Bioinformatics and Cancer Genomics Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia
| | - Stephen Q Wong
- Molecular Biomarkers and Translational Genomics Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia
| | - Jacquie Connaughton
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia
| | - Simone M Rowley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia
| | - Lisa Devereux
- LifePool, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia
| | - David Byrne
- Pathology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia
| | - Stephen B Fox
- Pathology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia
| | - Gisela Mir Arnau
- Molecular Genomics Core Facility, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia
| | - Richard W Tothill
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Molecular Imaging and Targeted Therapeutics Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia.,Department of Pathology, University of Melbourne, Parkville, VIC, Australia
| | - Ian G Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Department of Pathology, University of Melbourne, Parkville, VIC, Australia
| | - Kylie L Gorringe
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia. .,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia. .,Department of Pathology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
30
|
Prognostic and Predictive Value of RAS Gene Mutations in Colorectal Cancer: Moving Beyond KRAS Exon 2. Drugs 2016; 75:1739-56. [PMID: 26347132 DOI: 10.1007/s40265-015-0459-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The advent of anti-EGFR (epidermal growth factor receptor) therapy resulted in significant progress in the treatment of metastatic colorectal cancer patients. However, many patients do not respond to this therapy or develop acquired resistance within a few months after the start of treatment. Since 2008, anti-EGFR therapy is restricted to KRAS wild-type patients as it has been shown that KRAS exon 2-mutated patients do not respond to this therapy. Still, up to 60 % of KRAS exon 2 wild-type patients show primary resistance to this treatment. Recently, several studies investigating the predictive and prognostic role of RAS mutations other than in KRAS exon 2 demonstrated that patients with these mutations are not responding to therapy. However, the role of these mutations has long been questioned as The National Comprehensive Cancer Network Guidelines in Oncology and the European Medicines Agency indications had already been changed in order to restrict anti-EGFR therapy to all RAS wild-type colorectal cancer patients, while the Food and Drug Administration guidelines remained unchanged. Recently, the Food and Drug Administration guidelines have also been changed, which implies the importance of RAS mutations beyond KRAS exon 2 in colorectal cancer. In this review, we discuss the most important studies regarding the predictive and prognostic role of RAS mutations other than in KRAS exon 2 in order to demonstrate the importance of these RAS mutations in patients with metastatic colorectal cancer treated with anti-EGFR therapy.
Collapse
|
31
|
Eosinophilic, Solid, and Cystic Renal Cell Carcinoma: Clinicopathologic Study of 16 Unique, Sporadic Neoplasms Occurring in Women. Am J Surg Pathol 2016; 40:60-71. [PMID: 26414221 DOI: 10.1097/pas.0000000000000508] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A unique renal neoplasm characterized by eosinophilic cytoplasm and solid and cystic growth was recently reported in patients with tuberous sclerosis complex (TSC). We searched multiple institutional archives and consult files in an attempt to identify a sporadic counterpart. We identified 16 morphologically identical cases, all in women, without clinical features of TSC. The median age was 57 years (range, 31 to 75 y). Macroscopically, tumors were tan and had a solid and macrocystic (12) or only solid appearance (4). Average tumor size was 50 mm (median, 38.5 mm; range, 15 to 135 mm). Microscopically, the tumors showed solid areas admixed with variably sized macrocysts and microcysts that were lined by cells with a pronounced hobnail arrangement. The cells had voluminous eosinophilic cytoplasm with prominent granular cytoplasmic stippling and round to oval nuclei with prominent nucleoli. Scattered histiocytes and lymphocytes were invariably present. Thirteen of 16 patients were stage pT1; 2 were pT2, and 1 was pT3a. The cells demonstrated a distinct immunoprofile: nuclear PAX8 expression, predominant CK20-positive/CK7-negative phenotype, patchy AMACR staining, but no CD117 reactivity. Thirteen of 14 patients with follow-up were alive and without disease progression after 2 to 138 months (mean: 53 mo; median: 37.5 mo); 1 patient died of other causes. Although similar to a subset of renal cell carcinomas (RCCs) seen in TSC, we propose that sporadic "eosinophilic, solid, and cystic RCC," which occurs predominantly in female individuals and is characterized by distinct morphologic features, predominant CK20-positive/CK7-negative immunophenotype, and indolent behavior, represents a novel subtype of RCC.
Collapse
|
32
|
Genomic copy number analysis of a spectrum of blue nevi identifies recurrent aberrations of entire chromosomal arms in melanoma ex blue nevus. Mod Pathol 2016; 29:227-39. [PMID: 26743478 DOI: 10.1038/modpathol.2015.153] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 11/22/2015] [Accepted: 11/23/2015] [Indexed: 12/14/2022]
Abstract
Blue nevi may display significant atypia or undergo malignant transformation. Morphologic diagnosis of this spectrum of lesions is notoriously difficult, and molecular tools are increasingly used to improve diagnostic accuracy. We studied copy number aberrations in a cohort of cellular blue nevi, atypical cellular blue nevi, and melanomas ex blue nevi using Affymetrix's OncoScan platform. Cases with sufficient DNA were analyzed for GNAQ, GNA11, and HRAS mutations. Copy number aberrations were detected in 0 of 5 (0%) cellular blue nevi, 3 of 12 (25%) atypical cellular blue nevi, and 6 of 9 (67%) melanomas ex blue nevi. None of the atypical cellular blue nevi displayed more than one aberration, whereas complex aberrations involving four or more regions were seen exclusively in melanomas ex blue nevi. Gains and losses of entire chromosomal arms were identified in four of five melanomas ex blue nevi with copy number aberrations. In particular, gains of 1q, 4p, 6p, and 8q, and losses of 1p and 4q were each found in at least two melanomas. Whole chromosome aberrations were also common, and represented the sole finding in one atypical cellular blue nevus. When seen in melanomas, however, whole chromosome aberrations were invariably accompanied by partial aberrations of other chromosomes. Three melanomas ex blue nevi harbored aberrations, which were absent or negligible in their precursor components, suggesting progression in tumor biology. Gene mutations involving GNAQ and GNA11 were each detected in two of eight melanomas ex blue nevi. In conclusion, copy number aberrations are more common and often complex in melanomas ex blue nevi compared with cellular and atypical cellular blue nevi. Identification of recurrent gains and losses of entire chromosomal arms in melanomas ex blue nevi suggests that development of new probes targeting these regions may improve detection and risk stratification of these lesions.
Collapse
|
33
|
Sebastián E, Alcoceba M, Martín-García D, Blanco Ó, Sanchez-Barba M, Balanzategui A, Marín L, Montes-Moreno S, González-Barca E, Pardal E, Jiménez C, García-Álvarez M, Clot G, Carracedo Á, Gutiérrez NC, Sarasquete ME, Chillón C, Corral R, Prieto-Conde MI, Caballero MD, Salaverria I, García-Sanz R, González M. High-resolution copy number analysis of paired normal-tumor samples from diffuse large B cell lymphoma. Ann Hematol 2015; 95:253-62. [PMID: 26573278 DOI: 10.1007/s00277-015-2552-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/06/2015] [Indexed: 12/23/2022]
Abstract
Copy number analysis can be useful for assessing prognosis in diffuse large B cell lymphoma (DLBCL). We analyzed copy number data from tumor samples of 60 patients diagnosed with DLBCL de novo and their matched normal samples. We detected 63 recurrent copy number alterations (CNAs), including 33 gains, 30 losses, and nine recurrent acquired copy number neutral loss of heterozygosity (CNN-LOH). Interestingly, 20 % of cases acquired CNN-LOH of 6p21 locus, which involves the HLA region. In normal cells, there were no CNAs but we observed CNN-LOH involving some key lymphoma regions such as 6p21 and 9p24.1 (5 %) and 17p13.1 (2.5 %) in DLBCL patients. Furthermore, a model with some specific CNA was able to predict the subtype of DLBCL, 1p36.32 and 10q23.31 losses being restricted to germinal center B cell-like (GCB) DLBCL. In contrast, 8p23.3 losses and 11q24.3 gains were strongly associated with the non-GCB subtype. A poor prognosis was associated with biallelic inactivation of TP53 or 18p11.32 losses, while prognosis was better in cases carrying 11q24.3 gains. In summary, CNA abnormalities identify specific DLBCL groups, and we describe CNN-LOH in germline cells from DLBCL patients that are associated with genes that probably play a key role in DLBCL development.
Collapse
Affiliation(s)
- Elena Sebastián
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Spanish Lymphoma/Autologous Bone Marrow Transplant Study Group (GELTAMO), Salamanca, Spain
| | - Miguel Alcoceba
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Spanish Lymphoma/Autologous Bone Marrow Transplant Study Group (GELTAMO), Salamanca, Spain
| | - David Martín-García
- Hematopathology Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Óscar Blanco
- Department of Pathology, University Hospital of Salamanca, Salamanca, Spain
| | | | - Ana Balanzategui
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Luis Marín
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Santiago Montes-Moreno
- Spanish Lymphoma/Autologous Bone Marrow Transplant Study Group (GELTAMO), Salamanca, Spain
- Department of Pathology, University Hospital of Marqués de Valdecilla/IFIMAV, Santander, Spain
| | - Eva González-Barca
- Spanish Lymphoma/Autologous Bone Marrow Transplant Study Group (GELTAMO), Salamanca, Spain
| | - Emilia Pardal
- Spanish Lymphoma/Autologous Bone Marrow Transplant Study Group (GELTAMO), Salamanca, Spain
| | - Cristina Jiménez
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
| | - María García-Álvarez
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
| | - Guillem Clot
- Hematopathology Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ángel Carracedo
- Fundación Pública Galega de Medicina Xenómica, IDIS, SERGAS, Santiago de Compostela, Spain
- Grupo de Medicina Xenómica, CIBERER, Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Norma C Gutiérrez
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - M Eugenia Sarasquete
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Carmen Chillón
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Rocío Corral
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - M Isabel Prieto-Conde
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
| | - M Dolores Caballero
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Spanish Lymphoma/Autologous Bone Marrow Transplant Study Group (GELTAMO), Salamanca, Spain
| | - Itziar Salaverria
- Hematopathology Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ramón García-Sanz
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.
- Spanish Lymphoma/Autologous Bone Marrow Transplant Study Group (GELTAMO), Salamanca, Spain.
- Center for Cancer Research (CIC, IBMCC-USAL-CSIC), Salamanca, Spain.
| | - Marcos González
- Molecular Biology & Histocompatibility Unit, Department of Hematology, IBSAL - University Hospital of Salamanca, Paseo de San Vicente, 58-182, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Center for Cancer Research (CIC, IBMCC-USAL-CSIC), Salamanca, Spain
| |
Collapse
|
34
|
High-quality genotyping data from formalin-fixed, paraffin-embedded tissue on the drug metabolizing enzymes and transporters plus array. J Mol Diagn 2015; 17:4-9. [PMID: 25528187 DOI: 10.1016/j.jmoldx.2014.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/16/2014] [Accepted: 08/28/2014] [Indexed: 01/19/2023] Open
Abstract
The Affymetrix Drug Metabolizing Enzymes and Transporters (DMET) Plus array covers 1936 markers in 231 genes involved in drug metabolism and transport. Blood- and saliva-derived DNA works well on the DMET array, but the utility of DNA from FFPE tissue has not been reported for this array. As the ability to use DNA from FFPE tissue on the array could open the potential for large retrospective sample collections, we examined the performance and reliability of FFPE-derived DNA on the DMET Plus array. Germline DNA isolated from archived normal FFPE tissue blocks stored for 3 to 19 years and matched blood or saliva from 16 patients with osteosarcoma were genotyped on the DMET Plus array. Concordance was assessed by calculating agreement and the κ-statistic. We observed high call rates for both the blood- or saliva-derived DNA samples (99.4%) and the FFPE-derived DNA samples (98.9%). Moreover, the concordance among the 16 blood- or saliva-derived DNA and FFPE DNA pairs was high (97.4%, κ = 0.915). This is the first study showing that DNA from normal FFPE tissue provides accurate and reliable genotypes on the DMET Plus array compared with blood- or saliva-derived DNA. This finding provides an opportunity for pharmacogenetic studies in diseases with high mortality rates and prevents a bias in studies where otherwise only alive patients can be included.
Collapse
|
35
|
Not All Next Generation Sequencing Diagnostics are Created Equal: Understanding the Nuances of Solid Tumor Assay Design for Somatic Mutation Detection. Cancers (Basel) 2015; 7:1313-32. [PMID: 26193321 PMCID: PMC4586770 DOI: 10.3390/cancers7030837] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 06/30/2015] [Accepted: 07/10/2015] [Indexed: 01/15/2023] Open
Abstract
The molecular characterization of tumors using next generation sequencing (NGS) is an emerging diagnostic tool that is quickly becoming an integral part of clinical decision making. Cancer genomic profiling involves significant challenges including DNA quality and quantity, tumor heterogeneity, and the need to detect a wide variety of complex genetic mutations. Most available comprehensive diagnostic tests rely on primer based amplification or probe based capture methods coupled with NGS to detect hotspot mutation sites or whole regions implicated in disease. These tumor panels utilize highly customized bioinformatics pipelines to perform the difficult task of accurately calling cancer relevant alterations such as single nucleotide variations, small indels or large genomic alterations from the NGS data. In this review, we will discuss the challenges of solid tumor assay design/analysis and report a case study that highlights the need to include complementary technologies (i.e., arrays) and germline analysis in tumor testing to reliably identify copy number alterations and actionable variants.
Collapse
|
36
|
Bambury RM, Bhatt AS, Riester M, Pedamallu CS, Duke F, Bellmunt J, Stack EC, Werner L, Park R, Iyer G, Loda M, Kantoff PW, Michor F, Meyerson M, Rosenberg JE. DNA copy number analysis of metastatic urothelial carcinoma with comparison to primary tumors. BMC Cancer 2015; 15:242. [PMID: 25886454 PMCID: PMC4392457 DOI: 10.1186/s12885-015-1192-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 03/16/2015] [Indexed: 01/10/2023] Open
Abstract
Background To date, there have been no reports characterizing the genome-wide somatic DNA chromosomal copy-number alteration landscape in metastatic urothelial carcinoma. We sought to characterize the DNA copy-number profile in a cohort of metastatic samples and compare them to a cohort of primary urothelial carcinoma samples in order to identify changes that are associated with progression from primary to metastatic disease. Methods Using molecular inversion probe array analysis we compared genome-wide chromosomal copy-number alterations between 30 metastatic and 29 primary UC samples. Whole transcriptome RNA-Seq analysis was also performed in primary and matched metastatic samples which was available for 9 patients. Results Based on a focused analysis of 32 genes in which alterations may be clinically actionable, there were significantly more amplifications/deletions in metastases (8.6% vs 4.5%, p < 0.001). In particular, there was a higher frequency of E2F3 amplification in metastases (30% vs 7%, p = 0.046). Paired primary and metastatic tissue was available for 11 patients and 3 of these had amplifications of potential clinical relevance in metastases that were not in the primary tumor including ERBB2, CDK4, CCND1, E2F3, and AKT1. The transcriptional activity of these amplifications was supported by RNA expression data. Conclusions The discordance in alterations between primary and metastatic tissue may be of clinical relevance in the era of genomically directed precision cancer medicine. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1192-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Richard M Bambury
- Memorial Sloan Kettering Cancer Center/Weill Cornell Medical College, New York, USA.
| | - Ami S Bhatt
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA. .,The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Markus Riester
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA.
| | - Chandra Sekhar Pedamallu
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA. .,The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Fujiko Duke
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA. .,The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Joaquim Bellmunt
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA.
| | - Edward C Stack
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA.
| | - Lillian Werner
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA.
| | - Rachel Park
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA.
| | - Gopa Iyer
- Memorial Sloan Kettering Cancer Center/Weill Cornell Medical College, New York, USA.
| | - Massimo Loda
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA. .,The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Philip W Kantoff
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA.
| | - Franziska Michor
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA.
| | - Matthew Meyerson
- Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, USA. .,The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Jonathan E Rosenberg
- Memorial Sloan Kettering Cancer Center/Weill Cornell Medical College, New York, USA.
| |
Collapse
|
37
|
Greytak SR, Engel KB, Bass BP, Moore HM. Accuracy of Molecular Data Generated with FFPE Biospecimens: Lessons from the Literature. Cancer Res 2015; 75:1541-7. [PMID: 25836717 DOI: 10.1158/0008-5472.can-14-2378] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 12/22/2014] [Indexed: 12/15/2022]
Abstract
Formalin-fixed and paraffin-embedded (FFPE) tissue biospecimens are a valuable resource for molecular cancer research. Although much can be gained from their use, it remains unclear whether the genomic and expression profiles obtained from FFPE biospecimens accurately reflect the physiologic condition of the patient from which they were procured, or if such profiles are confounded by biologic effects from formalin fixation and processing. To assess the physiologic accuracy of genomic and expression data generated with FFPE specimens, we surveyed the literature for articles investigating genomic and expression endpoints in case-matched FFPE and fresh or frozen human biospecimens using the National Cancer Institute's Biospecimen Research Database (http://biospecimens.cancer.gov/brd). Results of the survey revealed that the level of concordance between differentially preserved biospecimens varied among analytical parameters and platforms but also among reports, genes/transcripts of interest, and tumor status. The identified analytical techniques and parameters that resulted in strong correlations between FFPE and frozen biospecimens may provide guidance when optimizing molecular protocols for FFPE use; however, discrepancies reported for similar assays also illustrate the importance of validating protocols optimized for use with FFPE specimens with a case-matched fresh or frozen cohort for each platform, gene or transcript, and FFPE processing regime. On the basis of evidence published to date, validation of analytical parameters with a properly handled frozen cohort is necessary to ensure a high degree of concordance and confidence in the results obtained with FFPE biospecimens.
Collapse
Affiliation(s)
| | | | | | - Helen M Moore
- Biorepositories and Biospecimen Research Branch, Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
38
|
Takano EA, Hunter SM, Campbell IG, Fox SB. Low-grade fibromatosis-like spindle cell carcinomas of the breast are molecularly exiguous. J Clin Pathol 2015; 68:362-7. [PMID: 25713418 DOI: 10.1136/jclinpath-2014-202824] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/29/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Low-grade fibromatosis-like spindle cell carcinomas are very rare breast carcinomas comprising <0.5% of all breast cancers. They demonstrate immunohistochemical (IHC) features of basal-like/metaplastic breast carcinomas, but the underlying molecular characteristics are unknown. We hypothesised that, as with IHC similarities, there may be common genomic alterations between spindle cell and basal-like/metaplastic carcinomas. METHODS AND RESULTS Genomic mutational profile and genomic copy number aberration (CNA) analyses were performed on three cases of this unusual entity, and findings were compared with that reported for basal-like/metaplastic breast carcinomas. Copy number analyses by molecular inversion probe assays of the three spindle cell carcinoma samples revealed little overall genomic CNAs with only minor changes identified (fraction of the genome altered; 1.3%-6.4%), but with a common 9p21.3 loss in 2 out of 3 samples, with CDKN2A (p16) being a likely candidate. No areas of commonality were identified in an in silico analysis compared with publically available basal-like/metaplastic carcinoma copy number data. CONCLUSIONS These tumours are characterised by low genomic instability, and share no CNAs with other metaplastic carcinomas. These findings favour this entity being a unique group genotype and belie their apparent homogeneous morphology and phenotype.
Collapse
Affiliation(s)
- Elena A Takano
- Department of Pathology, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia
| | - Sally M Hunter
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia
| | - Ian G Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
39
|
Chao A, Lai CH, Lee YS, Ueng SH, Lin CY, Wang TH. Molecular characteristics of endometrial cancer coexisting with peritoneal malignant mesothelioma in Li-Fraumeni-like syndrome. BMC Cancer 2015; 15:8. [PMID: 25588929 PMCID: PMC4312462 DOI: 10.1186/s12885-015-1010-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 01/06/2015] [Indexed: 11/22/2022] Open
Abstract
Background Endometrial cancer that occurs concurrently with peritoneal malignant mesothelioma (PMM) is difficult to diagnose preoperatively. Case presentation A postmenopausal woman had endometrial cancer extending to the cervix, vagina and pelvic lymph nodes, and PMM in bilateral ovaries, cul-de-sac, and multiple peritoneal sites. Adjuvant therapies included chemotherapy and radiotherapy. Targeted, massively parallel DNA sequencing and molecular inversion probe microarray analysis revealed a germline TP53 mutation compatible with Li-Fraumeni-like syndrome, somatic mutations of PIK3CA in the endometrial cancer, and a somatic mutation of GNA11 and JAK3 in the PMM. Large-scale genomic amplifications and some deletions were found in the endometrial cancer. The patient has been stable for 24 months after therapy. One of her four children was also found to carry the germline TP53 mutation. Conclusions Molecular characterization of the coexistent tumors not only helps us make the definite diagnosis, but also provides information to select targeted therapies if needed in the future. Identification of germline TP53 mutation further urged us to monitor future development of malignancies in this patient and encourage cancer screening in her family. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1010-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Angel Chao
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.
| | - Chyong-Huey Lai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.
| | - Yun-Shien Lee
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan. .,Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan.
| | - Shir-Hwa Ueng
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.
| | - Chiao-Yun Lin
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.
| | - Tzu-Hao Wang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan. .,Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
40
|
Anti-EGFR Resistance in Colorectal Cancer: Current Knowledge and Future Perspectives. CURRENT COLORECTAL CANCER REPORTS 2014. [DOI: 10.1007/s11888-014-0242-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
41
|
Alexiev BA, Zou YS. Clear cell papillary renal cell carcinoma: A chromosomal microarray analysis of two cases using a novel Molecular Inversion Probe (MIP) technology. Pathol Res Pract 2014; 210:1049-53. [DOI: 10.1016/j.prp.2014.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 09/29/2014] [Accepted: 10/13/2014] [Indexed: 10/24/2022]
|
42
|
Küsters-Vandevelde HVN, van Engen- van Grunsven IACH, Coupland SE, Lake SL, Rijntjes J, Pfundt R, Küsters B, Wesseling P, Blokx WAM, Groenen PJTA. Mutations in G Protein Encoding Genes and Chromosomal Alterations in Primary Leptomeningeal Melanocytic Neoplasms. Pathol Oncol Res 2014; 21:439-47. [DOI: 10.1007/s12253-014-9841-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 09/11/2014] [Indexed: 10/24/2022]
|
43
|
Guancial EA, Werner L, Bellmunt J, Bamias A, Choueiri TK, Ross R, Schutz FA, Park RS, O'Brien RJ, Hirsch MS, Barletta JA, Berman DM, Lis R, Loda M, Stack EC, Garraway LA, Riester M, Michor F, Kantoff PW, Rosenberg JE. FGFR3 expression in primary and metastatic urothelial carcinoma of the bladder. Cancer Med 2014; 3:835-44. [PMID: 24846059 PMCID: PMC4303151 DOI: 10.1002/cam4.262] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 02/24/2014] [Accepted: 03/25/2014] [Indexed: 01/03/2023] Open
Abstract
While fibroblast growth factor receptor 3 (FGFR3) is frequently mutated or overexpressed in nonmuscle-invasive urothelial carcinoma (UC), the prevalence of FGFR3 protein expression and mutation remains unknown in muscle-invasive disease. FGFR3 protein and mRNA expression, mutational status, and copy number variation were retrospectively analyzed in 231 patients with formalin-fixed paraffin-embedded primary UCs, 33 metastases, and 14 paired primary and metastatic tumors using the following methods: immunohistochemistry, NanoString nCounterTM, OncoMap or Affymetrix OncoScanTM array, and Gain and Loss of Analysis of DNA and Genomic Identification of Significant Targets in Cancer software. FGFR3 immunohistochemistry staining was present in 29% of primary UCs and 49% of metastases and did not impact overall survival (P = 0.89, primary tumors; P = 0.78, metastases). FGFR3 mutations were observed in 2% of primary tumors and 9% of metastases. Mutant tumors expressed higher levels of FGFR3 mRNA than wild-type tumors (P < 0.001). FGFR3 copy number gain and loss were rare events in primary and metastatic tumors (0.8% each; 3.0% and 12.3%, respectively). FGFR3 immunohistochemistry staining is present in one third of primary muscle-invasive UCs and half of metastases, while FGFR3 mutations and copy number changes are relatively uncommon.
Collapse
Affiliation(s)
- Elizabeth A Guancial
- Dana-Farber Cancer Institute, Boston, Massachusetts; University of Rochester, Rochester, New York
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chen G, Chakravarti N, Aardalen K, Lazar AJ, Tetzlaff MT, Wubbenhorst B, Kim SB, Kopetz S, Ledoux AA, Gopal YNV, Pereira CG, Deng W, Lee JS, Nathanson KL, Aldape KD, Prieto VG, Stuart D, Davies MA. Molecular profiling of patient-matched brain and extracranial melanoma metastases implicates the PI3K pathway as a therapeutic target. Clin Cancer Res 2014; 20:5537-46. [PMID: 24803579 DOI: 10.1158/1078-0432.ccr-13-3003] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE An improved understanding of the molecular pathogenesis of brain metastases, one of the most common and devastating complications of advanced melanoma, may identify and prioritize rational therapeutic approaches for this disease. In particular, the identification of molecular differences between brain and extracranial metastases would support the need for the development of organ-specific therapeutic approaches. EXPERIMENTAL DESIGN Hotspot mutations, copy number variations (CNV), global mRNA expression patterns, and quantitative analysis of protein expression and activation by reverse-phase protein array (RPPA) analysis were evaluated in pairs of melanoma brain metastases and extracranial metastases from patients who had undergone surgical resection for both types of tumors. RESULTS The status of 154 previously reported hotspot mutations, including driver mutations in BRAF and NRAS, were concordant in all evaluable patient-matched pairs of tumors. Overall patterns of CNV, mRNA expression, and protein expression were largely similar between the paired samples for individual patients. However, brain metastases demonstrated increased expression of several activation-specific protein markers in the PI3K/AKT pathway compared with the extracranial metastases. CONCLUSIONS These results add to the understanding of the molecular characteristics of melanoma brain metastases and support the rationale for additional testing of the PI3K/AKT pathway as a therapeutic target in these highly aggressive tumors.
Collapse
Affiliation(s)
- Guo Chen
- Departments of Melanoma Medical Oncology,
| | | | - Kimberly Aardalen
- Novartis Institutes for Biomedical Research, Emeryville, California; and
| | | | | | - Bradley Wubbenhorst
- Division of Medical Genetics, Department of Medicine, The University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | - Scott Kopetz
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | | - Katherine L Nathanson
- Division of Medical Genetics, Department of Medicine, The University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | | | - Darrin Stuart
- Novartis Institutes for Biomedical Research, Emeryville, California; and
| | | |
Collapse
|
45
|
Genomic aberrations of BRCA1-mutated fallopian tube carcinomas. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1871-6. [PMID: 24726640 DOI: 10.1016/j.ajpath.2014.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/20/2014] [Accepted: 02/20/2014] [Indexed: 01/30/2023]
Abstract
Intraepithelial carcinomas of the fallopian tube are putative precursors to high-grade serous carcinomas of the ovary and peritoneum. Molecular characterization of these early precursors is limited but could be the key to identifying tumor biomarkers for early detection. This study presents a genome-wide copy number analysis of occult fallopian tube carcinomas identified through risk-reducing prophylactic oophorectomy from three women with germline BRCA1 mutations, demonstrating that extensive genomic aberrations are already established at this early stage. We found no indication of a difference in the level of genomic aberration observed in fallopian tube carcinomas compared with high-grade serous ovarian carcinomas. These findings suggest that spread to the peritoneal cavity may require no or very little further tumor evolution, which raises the question of what is the real window of opportunity to detect high-grade serous peritoneal carcinoma arising from the fallopian tube before it spreads. Nonetheless, the similarity of the genomic aberrations to those observed in high-grade serous ovarian carcinomas suggests that genetic biomarkers identified in late-stage disease may be relevant for early detection.
Collapse
|
46
|
Intratumoral genetic heterogeneity in metastatic melanoma is accompanied by variation in malignant behaviors. BMC Med Genomics 2013; 6:40. [PMID: 24119551 PMCID: PMC3852494 DOI: 10.1186/1755-8794-6-40] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Accepted: 09/19/2013] [Indexed: 02/06/2023] Open
Abstract
Background Intratumoral heterogeneity is a major obstacle for the treatment of cancer, as the presence of even minor populations that are insensitive to therapy can lead to disease relapse. Increased clonal diversity has been correlated with a poor prognosis for cancer patients, and we therefore examined genetic, transcriptional, and functional diversity in metastatic melanoma. Methods Amplicon sequencing and SNP microarrays were used to profile somatic mutations and DNA copy number changes in multiple regions from metastatic lesions. Clonal genetic and transcriptional heterogeneity was also assessed in single cell clones from early passage cell lines, which were then subjected to clonogenicity and drug sensitivity assays. Results MAPK pathway and tumor suppressor mutations were identified in all regions of the melanoma metastases analyzed. In contrast, we identified copy number abnormalities present in only some regions in addition to homogeneously present changes, suggesting ongoing genetic evolution following metastatic spread. Copy number heterogeneity from a tumor was represented in matched cell line clones, which also varied in their clonogenicity and drug sensitivity. Minor clones were identified based on dissimilarity to the parental cell line, and these clones were the most clonogenic and least sensitive to drugs. Finally, treatment of a polyclonal cell line with paclitaxel to enrich for drug-resistant cells resulted in the adoption of a gene expression profile with features of one of the minor clones, supporting the idea that these populations can mediate disease relapse. Conclusion Our results support the hypothesis that minor clones might have major consequences for patient outcomes in melanoma.
Collapse
|
47
|
Lemech CR, Arkenau HT. The future is here: tumour profiling in day-to-day clinical practice. Drug Discov Today 2013; 18:689-91. [DOI: 10.1016/j.drudis.2013.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 06/12/2013] [Accepted: 06/14/2013] [Indexed: 10/26/2022]
|
48
|
Abstract
Cancer is associated with mutated genes, and analysis of tumour-linked genetic alterations is increasingly used for diagnostic, prognostic and treatment purposes. The genetic profile of solid tumours is currently obtained from surgical or biopsy specimens; however, the latter procedure cannot always be performed routinely owing to its invasive nature. Information acquired from a single biopsy provides a spatially and temporally limited snap-shot of a tumour and might fail to reflect its heterogeneity. Tumour cells release circulating free DNA (cfDNA) into the blood, but the majority of circulating DNA is often not of cancerous origin, and detection of cancer-associated alleles in the blood has long been impossible to achieve. Technological advances have overcome these restrictions, making it possible to identify both genetic and epigenetic aberrations. A liquid biopsy, or blood sample, can provide the genetic landscape of all cancerous lesions (primary and metastases) as well as offering the opportunity to systematically track genomic evolution. This Review will explore how tumour-associated mutations detectable in the blood can be used in the clinic after diagnosis, including the assessment of prognosis, early detection of disease recurrence, and as surrogates for traditional biopsies with the purpose of predicting response to treatments and the development of acquired resistance.
Collapse
|
49
|
Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 2013; 10:472-84. [PMID: 23836314 DOI: 10.1038/nrclinonc.2013.110] [Citation(s) in RCA: 1255] [Impact Index Per Article: 114.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cancer is associated with mutated genes, and analysis of tumour-linked genetic alterations is increasingly used for diagnostic, prognostic and treatment purposes. The genetic profile of solid tumours is currently obtained from surgical or biopsy specimens; however, the latter procedure cannot always be performed routinely owing to its invasive nature. Information acquired from a single biopsy provides a spatially and temporally limited snap-shot of a tumour and might fail to reflect its heterogeneity. Tumour cells release circulating free DNA (cfDNA) into the blood, but the majority of circulating DNA is often not of cancerous origin, and detection of cancer-associated alleles in the blood has long been impossible to achieve. Technological advances have overcome these restrictions, making it possible to identify both genetic and epigenetic aberrations. A liquid biopsy, or blood sample, can provide the genetic landscape of all cancerous lesions (primary and metastases) as well as offering the opportunity to systematically track genomic evolution. This Review will explore how tumour-associated mutations detectable in the blood can be used in the clinic after diagnosis, including the assessment of prognosis, early detection of disease recurrence, and as surrogates for traditional biopsies with the purpose of predicting response to treatments and the development of acquired resistance.
Collapse
Affiliation(s)
- Emily Crowley
- Department of Oncology, University of Turin, Institute for Cancer Research and Treatment, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Turin, Italy
| | | | | | | |
Collapse
|
50
|
Rowe LR, Thaker HM, Opitz JM, Schiffman JD, Haddadin ZM, Erickson LK, South ST. Molecular Inversion Probe Array for the Genetic Evaluation of Stillbirth Using Formalin-Fixed, Paraffin-Embedded Tissue. J Mol Diagn 2013; 15:466-72. [DOI: 10.1016/j.jmoldx.2013.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 02/28/2013] [Accepted: 03/15/2013] [Indexed: 11/25/2022] Open
|