1
|
Pirosanto Y, Laseca N, Valera M, Molina A, Moreno-Millán M, Bugno-Poniewierska M, Ross P, Azor P, Demyda-Peyrás S. Screening and detection of chromosomal copy number alterations in the domestic horse using SNP-array genotyping data. Anim Genet 2021; 52:431-439. [PMID: 34013628 DOI: 10.1111/age.13077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/25/2021] [Accepted: 04/22/2021] [Indexed: 12/27/2022]
Abstract
Chromosomal abnormalities are a common cause of infertility in horses. However, they are difficult to detect using automated methods. Here, we propose a simple methodology based on single nucleotide polymorphism (SNP)-array data that allows us to detect the main chromosomal abnormalities in horses in a single procedure. As proof of concept, we were able to detect chromosomal abnormalities in 33 out of 268 individuals, including monosomies, chimerisms, and male and female sex-reversions, by analyzing the raw signal intensity produced by an SNP array-based genotyping platform. We also demonstrated that the procedure is not affected by the SNP density of the array employed or by the inbreeding level of the individuals. Finally, the methodology proposed in this study could be performed in an open bioinformatic environment, thus permitting its integration as a flexible screening tool in diagnostic laboratories and genomic breeding programs.
Collapse
Affiliation(s)
- Y Pirosanto
- Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, Calle 60 y 118 s/n, La Plata, 1900, Argentina.,IGEVET (UNLP-CONICET LA PLATA), Facultad de Ciencias Veterinarias, UNLP, Calle 60 y 118 s/n, La Plata, 1900, Argentina
| | - N Laseca
- Laboratorio de Diagnóstico Genético Veterinario, Departamento de Genética, Universidad de Córdoba, CN IV KM 396, Edificio Gregor Mendel, Campus Rabanales, Córdoba, 14071, España
| | - M Valera
- Departamento de Agronomía, Escuela Técnica Superior de Ingeniería Agronómica, Universidad de Sevilla, Ctra. de Utrera km 1, Sevilla, 41013, España
| | - A Molina
- Laboratorio de Diagnóstico Genético Veterinario, Departamento de Genética, Universidad de Córdoba, CN IV KM 396, Edificio Gregor Mendel, Campus Rabanales, Córdoba, 14071, España
| | - M Moreno-Millán
- Laboratorio de Diagnóstico Genético Veterinario, Departamento de Genética, Universidad de Córdoba, CN IV KM 396, Edificio Gregor Mendel, Campus Rabanales, Córdoba, 14071, España
| | - M Bugno-Poniewierska
- Katedra Rozrodu, Anatomii i Genomiki Zwierząt Wydział Hodowli i Biologii Zwierząt, Uniwersytet Rolniczy im. Hugona Kołłątaja w Krakowie, al. Mickiewicza 24/28, Krakow, 30-059, Poland
| | - P Ross
- Department of Animal Science, University of California, Davis, One Shields Ave., Davis, CA, 95616, USA
| | - P Azor
- Asociación Nacional de Criadores de Caballos de Pura Raza Española (ANCCE), Edif. Indotorre · Avda. del Reino Unido 11, pl. 3ª 2, Sevilla, 41012, España
| | - S Demyda-Peyrás
- Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, Calle 60 y 118 s/n, La Plata, 1900, Argentina.,IGEVET (UNLP-CONICET LA PLATA), Facultad de Ciencias Veterinarias, UNLP, Calle 60 y 118 s/n, La Plata, 1900, Argentina
| |
Collapse
|
2
|
Tang Z, Kanagal-Shamanna R, Tang G, Patel K, Medeiros LJ, Toruner GA. Analytical and clinical performance of chromosomal microarrays compared with FISH panel and conventional karyotyping in patients with chronic lymphocytic leukemia. Leuk Res 2021; 108:106616. [PMID: 34022744 DOI: 10.1016/j.leukres.2021.106616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/26/2021] [Accepted: 05/10/2021] [Indexed: 11/19/2022]
Abstract
In this single center retrospective analysis on 102 CLL patients, we assessed analytical and clinical performance of CMA against a targeted FISH panel (ATM, TP53, CEP12, D13S319 and LAMP1 loci) and karyotyping. CMA yielded additional information compared to karyotype in 39 cases (38 %). On the other hand, while CMA detected aberrations were also detected by FISH in all 31 cases (30 %), aberrations with low clonal size (<30 %) detected by FISH were missed by CMA. When evaluated with National Cancer Center Network (NCCN) guidelines, the capture rate of prognostic relevant cytogenetic information for FISH only, FISH + Chromosomes and FISH + CMA analyses were 95, 96 and 100 % respectively. With Cancer Cytogenomics Consortium (CGC) Criteria, these figures for FISH only, FISH + Chromosomes and FISH + CMA were 88 %, 92 and 100 % respectively. In conclusion, CMA provides additional analytical information to FISH and karyotyping, but this information has a clinical utility only in a small number of patients. Limit of detection (LOD) issues preclude replacement of FISH by CMA, but CMA may be a viable alternative to karyotyping. Further research is warranted.
Collapse
Affiliation(s)
- Zhenya Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Keyur Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Gokce A Toruner
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States.
| |
Collapse
|
3
|
Assessing copy number aberrations and copy-neutral loss-of-heterozygosity across the genome as best practice: An evidence-based review from the Cancer Genomics Consortium (CGC) working group for chronic lymphocytic leukemia. Cancer Genet 2018; 228-229:236-250. [DOI: 10.1016/j.cancergen.2018.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/29/2018] [Accepted: 07/01/2018] [Indexed: 01/18/2023]
|
4
|
Vetro C, Haferlach T, Jeromin S, Stengel A, Zenger M, Nadarajah N, Baer C, Weissmann S, Kern W, Meggendorfer M, Haferlach C. Identification of prognostic parameters in CLL with no abnormalities detected by chromosome banding and FISH analyses. Br J Haematol 2018; 183:47-59. [PMID: 30022491 DOI: 10.1111/bjh.15498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/07/2018] [Indexed: 01/09/2023]
Abstract
Chronic Lymphocytic Leukaemia (CLL) is a heterogeneous disease with a clinical course dependent on cytogenetic features. However, in 15-20% of cases both chromosome banding and fluorescence in situ hybridisation analyses do not show any kind of abnormality. With the aim to identify dependable molecular prognostic factors in this subgroup, we performed a comprehensive analysis on 171 patients including genomic arrays (comparative genomic hybridisation and single nucleotide polymorphism), immunoglobulin heavy chain variable region genes (IGHV) status, flow cytometry and targeted sequencing. Genomic arrays detected 73 aberrations in 39 patients (23%). Most frequently, patients had 1 aberration (25/171; 15%), while 14 patients (8%) had at least 2 aberrations. IGHV status was unmutated in 53/171 (31%) patients. SF3B1 was the most frequently mutated gene (26/171 patients; 15%), followed by NOTCH1 (15/171; 9%). At univariate analysis, an adverse impact on time to treatment (TTT) was evident for SF3B1 mutations, higher white blood cell count, higher CLL cells percentage by flow cytometry, CD38 positivity, IGHV unmutated status and at least 2 genomic array abnormalities. Of these, SF3B1 mutations, CLL cells percentage, IGHV unmutated status and number of genomic array aberrations maintained their impact in multivariate analysis. In conclusion, by integrating genomic and molecular data, we identified patients at higher risk for treatment need.
Collapse
Affiliation(s)
| | | | | | - Anna Stengel
- MLL Munich Leukaemia Laboratory, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
5
|
The mutational signature of chronic lymphocytic leukemia. Biochem J 2016; 473:3725-3740. [DOI: 10.1042/bcj20160256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/23/2016] [Indexed: 01/14/2023]
Abstract
Advances in next-generation sequencing technologies continue to unravel the cancer genome, identifying key biological pathways important for disease pathogenesis and clinically relevant genetic lesions. These studies have provided unprecedented resolution of the cancer genome, facilitating significant advances in the ability to detect many cancers, and predict patients who will develop an aggressive disease or respond poorly to treatment. The mature B-cell neoplasm chronic lymphocytic leukaemia remains at the forefront of these genomic analyses, largely due its protracted natural history and the accessibility to suitable material for study. We now possess a comprehensive view of the genomic copy number mutational landscape of the disease, as well as a detail description of clonal evolution, and the molecular mechanisms that drive the acquisition of genomic lesions and more broadly, genomic complexity. Here, recent genomic insights with associated biological and clinical implications will be reviewed.
Collapse
|
6
|
Sutphin BS, Boczek NJ, Barajas-Martínez H, Hu D, Ye D, Tester DJ, Antzelevitch C, Ackerman MJ. Molecular and Functional Characterization of Rare CACNA1C Variants in Sudden Unexplained Death in the Young. CONGENIT HEART DIS 2016; 11:683-692. [PMID: 27218670 DOI: 10.1111/chd.12371] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 02/03/2016] [Accepted: 03/18/2016] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Perturbations in the CACNA1C-encoded L-type calcium channel α-subunit have been linked recently to heritable arrhythmia syndromes, including Timothy syndrome, Brugada syndrome, early repolarization syndrome, and long QT syndrome. These heritable arrhythmia syndromes may serve as a pathogenic basis for autopsy-negative sudden unexplained death in the young (SUDY). However, the contribution of CACNA1C mutations to SUDY is unknown. OBJECTIVE We set out to determine the spectrum, prevalence, and pathophysiology of rare CACNA1C variants in SUDY. METHODS Mutational analysis of CACNA1C was conducted in 82 SUDY cases using polymerase chain reaction, denaturing high-performance liquid chromatography, and direct sequencing. Identified variants were engineered using site-directed mutagenesis, and heterologously expressed in TSA-201 or HEK293 cells. RESULTS Two SUDY cases (2.4%) harbored functional variants in CACNA1C. The E850del and N2091S variants involve highly conserved residues and localize to the II-III linker and C-terminus, respectively. Although observed in publically available exome databases, both variants confer abnormal CaV 1.2 electrophysiological characteristics. Examination of the electrophysiological properties revealed the E850del mutation in CACNA1C led to a 95% loss-of-function in ICa , and the N2091S variant led to a 105% gain-of-function in ICa. Additionally, N2091S led to minor kinetic alterations including a -3.4 mV shift in V1/2 of activation. CONCLUSION This study provides molecular and functional evidence that rare CACNA1C genetic variants may contribute to the underlying pathogenic basis for some cases of SUDY in either a gain or loss-of-function mechanism.
Collapse
Affiliation(s)
- Brittan S Sutphin
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland, Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minn, USA
| | - Nicole J Boczek
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland, Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minn, USA
| | | | - Dan Hu
- Department of Molecular Genetics, Masonic Medical Research Laboratory, Utica, N.Y., USA
| | - Dan Ye
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland, Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minn, USA
| | - David J Tester
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland, Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minn, USA
| | - Charles Antzelevitch
- Department of Molecular Genetics, Masonic Medical Research Laboratory, Utica, N.Y., USA
| | - Michael J Ackerman
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland, Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minn, USA.,Departments of Medicine (Division of Cardiovascular Diseases) and Pediatrics (Division of Pediatric Cardiology), Mayo Clinic, Rochester, Minn, USA
| |
Collapse
|
7
|
Bhoi S, Ljungström V, Baliakas P, Mattsson M, Smedby KE, Juliusson G, Rosenquist R, Mansouri L. Prognostic impact of epigenetic classification in chronic lymphocytic leukemia: The case of subset #2. Epigenetics 2016; 11:449-55. [PMID: 27128508 DOI: 10.1080/15592294.2016.1178432] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Based on the methylation status of 5 single CpG sites, a novel epigenetic classification of chronic lymphocytic leukemia (CLL) was recently proposed, classifying CLL patients into 3 clinico-biological subgroups with different outcome, termed memory like CLL (m-CLL), naïve like CLL (n-CLL), and a third intermediate CLL subgroup (i-CLL). While m-CLL and n-CLL patients at large corresponded to patients carrying mutated and unmutated IGHV genes, respectively, limited information exists regarding the less defined i-CLL group. Using pyrosequencing, we investigated the prognostic impact of the proposed 5 CpG signature in a well-characterized CLL cohort (135 cases), including IGHV-mutated and unmutated patients as well as clinically aggressive stereotyped subset #2 patients. Overall, we confirmed the signature's association with established prognostic markers. Moreover, in the presence of the IGHV mutational status, the epigenetic signature remained independently associated with both time-to-first-treatment and overall survival in multivariate analyses. As a prime finding, we observed that subset #2 patients were predominantly classified as i-CLL, probably reflecting their borderline IGHV mutational status (97-99% germline identity), though having a similarly poor prognosis as n-CLL patients. In summary, we validated the epigenetic classifier as an independent factor in CLL prognostication and provide further evidence that subset #2 is a member of the i-CLL group, hence supporting the existence of a third, intermediate epigenetic subgroup.
Collapse
Affiliation(s)
- Sujata Bhoi
- a Department of Immunology , Genetics and Pathology, Science for Life Laboratory, Uppsala University , Sweden
| | - Viktor Ljungström
- a Department of Immunology , Genetics and Pathology, Science for Life Laboratory, Uppsala University , Sweden
| | - Panagiotis Baliakas
- a Department of Immunology , Genetics and Pathology, Science for Life Laboratory, Uppsala University , Sweden
| | - Mattias Mattsson
- a Department of Immunology , Genetics and Pathology, Science for Life Laboratory, Uppsala University , Sweden.,b Department of Medical Sciences , Uppsala University , Uppsala , Sweden
| | - Karin E Smedby
- c Department of Medicine Solna , Clinical Epidemiology Unit, Karolinska Institutet , Stockholm , Sweden
| | - Gunnar Juliusson
- d Department of Laboratory Medicine , Stem Cell Center, Hematology and Transplantation, Lund University , Lund , Sweden
| | - Richard Rosenquist
- a Department of Immunology , Genetics and Pathology, Science for Life Laboratory, Uppsala University , Sweden
| | - Larry Mansouri
- a Department of Immunology , Genetics and Pathology, Science for Life Laboratory, Uppsala University , Sweden
| |
Collapse
|
8
|
La-Touche S, Lemetre C, Lambros M, Stankiewicz E, Ng CKY, Weigelt B, Rajab R, Tinwell B, Corbishley C, Watkin N, Berney D, Reis-Filho JS. DNA Copy Number Aberrations, and Human Papillomavirus Status in Penile Carcinoma. Clinico-Pathological Correlations and Potential Driver Genes. PLoS One 2016; 11:e0146740. [PMID: 26901676 PMCID: PMC4763861 DOI: 10.1371/journal.pone.0146740] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/21/2015] [Indexed: 12/11/2022] Open
Abstract
Penile squamous cell carcinoma is a rare disease, in which somatic genetic aberrations have yet to be characterized. We hypothesized that gene copy aberrations might correlate with human papillomavirus status and clinico-pathological features. We sought to determine the spectrum of gene copy number aberrations in a large series of PSCCs and to define their correlations with human papillomavirus, histopathological subtype, and tumor grade, stage and lymph node status. Seventy formalin-fixed, paraffin embedded penile squamous cell carcinomas were centrally reviewed by expert uropathologists. DNA was extracted from micro-dissected samples, subjected to PCR-based human papillomavirus assessment and genotyping (INNO-LiPA human papillomavirus Genotyping Extra Assay) and microarray-based comparative genomic hybridization using a 32K Bacterial Artificial Chromosome array platform. Sixty-four samples yielded interpretable results. Recurrent gains were observed in chromosomes 1p13.3-q44 (88%), 3p12.3-q29 (86%), 5p15.33-p11 (67%) and 8p12-q24.3 (84%). Amplifications of 5p15.33-p11 and 11p14.1-p12 were found in seven (11%) and four (6%) cases, respectively. Losses were observed in chromosomes 2q33-q37.3 (86%), 3p26.3-q11.1 (83%) and 11q12.2-q25 (81%). Although many losses and gains were similar throughout the cohort, there were small significant differences observed at specific loci, between human papillomavirus positive and negative tumors, between tumor types, and tumor grade and nodal status. These results demonstrate that despite the diversity of genetic aberrations in penile squamous cell carcinomas, there are significant correlations between the clinico-pathological data and the genetic changes that may play a role in disease natural history and progression and highlight potential driver genes, which may feature in molecular pathways for existing therapeutic agents.
Collapse
Affiliation(s)
- Susannah La-Touche
- Bart's Cancer Institute, Centre for Molecular Oncology, Queen Mary University of London, John Vane Science Centre, Charterhouse square, London, United Kingdom
- * E-mail:
| | - Christophe Lemetre
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Maryou Lambros
- Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Elzbieta Stankiewicz
- Bart's Cancer Institute, Centre for Molecular Oncology, Queen Mary University of London, John Vane Science Centre, Charterhouse square, London, United Kingdom
| | - Charlotte K. Y. Ng
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Ramzi Rajab
- St George’s Hospital, Tooting, London, United Kingdom
| | | | | | - Nick Watkin
- St George’s Hospital, Tooting, London, United Kingdom
| | - Dan Berney
- Bart's Cancer Institute, Centre for Molecular Oncology, Queen Mary University of London, John Vane Science Centre, Charterhouse square, London, United Kingdom
| | - Jorge S. Reis-Filho
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| |
Collapse
|
9
|
Sellmann L, Scholtysik R, de Beer D, Eisele L, Klein-Hitpass L, Nückel H, Dührsen U, Dürig J, Röth A, Baerlocher GM. Shorter telomeres correlate with an increase in the number of uniparental disomies in patients with chronic lymphocytic leukemia. Leuk Lymphoma 2015; 57:590-5. [DOI: 10.3109/10428194.2015.1076929] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Ludger Sellmann
- Department of Haematology, Medical Faculty, University of Duisburg-Essen, Essen, Germany,
- Institute of Cell Biology (Cancer Research), Medical Faculty, University of Duisburg-Essen, Essen, Germany,
| | - Rene Scholtysik
- Institute of Cell Biology (Cancer Research), Medical Faculty, University of Duisburg-Essen, Essen, Germany,
| | - Dirk de Beer
- Experimental Haematology, Department of Clinical Research, University Bern, Switzerland, and
| | - Lewin Eisele
- Department of Haematology, Medical Faculty, University of Duisburg-Essen, Essen, Germany,
| | - Ludger Klein-Hitpass
- Institute of Cell Biology (Cancer Research), Medical Faculty, University of Duisburg-Essen, Essen, Germany,
| | - Holger Nückel
- Department of Haematology, Medical Faculty, University of Duisburg-Essen, Essen, Germany,
| | - Ulrich Dührsen
- Department of Haematology, Medical Faculty, University of Duisburg-Essen, Essen, Germany,
| | - Jan Dürig
- Department of Haematology, Medical Faculty, University of Duisburg-Essen, Essen, Germany,
| | - Alexander Röth
- Department of Haematology, Medical Faculty, University of Duisburg-Essen, Essen, Germany,
| | - Gabriela M. Baerlocher
- Experimental Haematology, Department of Clinical Research, University Bern, Switzerland, and
- Department of Haematology, University Hospital, Bern, Switzerland
| |
Collapse
|
10
|
Sutton LA, Rosenquist R. The complex interplay between cell-intrinsic and cell-extrinsic factors driving the evolution of chronic lymphocytic leukemia. Semin Cancer Biol 2015; 34:22-35. [DOI: 10.1016/j.semcancer.2015.04.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 01/08/2023]
|
11
|
Ng CKY, Martelotto LG, Gauthier A, Wen HC, Piscuoglio S, Lim RS, Cowell CF, Wilkerson PM, Wai P, Rodrigues DN, Arnould L, Geyer FC, Bromberg SE, Lacroix-Triki M, Penault-Llorca F, Giard S, Sastre-Garau X, Natrajan R, Norton L, Cottu PH, Weigelt B, Vincent-Salomon A, Reis-Filho JS. Intra-tumor genetic heterogeneity and alternative driver genetic alterations in breast cancers with heterogeneous HER2 gene amplification. Genome Biol 2015; 16:107. [PMID: 25994018 PMCID: PMC4440518 DOI: 10.1186/s13059-015-0657-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 04/20/2015] [Indexed: 01/08/2023] Open
Abstract
Background HER2 is overexpressed and amplified in approximately 15% of invasive breast cancers, and is the molecular target and predictive marker of response to anti-HER2 agents. In a subset of these cases, heterogeneous distribution of HER2 gene amplification can be found, which creates clinically challenging scenarios. Currently, breast cancers with HER2 amplification/overexpression in just over 10% of cancer cells are considered HER2-positive for clinical purposes; however, it is unclear as to whether the HER2-negative components of such tumors would be driven by distinct genetic alterations. Here we sought to characterize the pathologic and genetic features of the HER2-positive and HER2-negative components of breast cancers with heterogeneous HER2 gene amplification and to define the repertoire of potential driver genetic alterations in the HER2-negative components of these cases. Results We separately analyzed the HER2-negative and HER2-positive components of 12 HER2 heterogeneous breast cancers using gene copy number profiling and massively parallel sequencing, and identified potential driver genetic alterations restricted to the HER2-negative cells in each case. In vitro experiments provided functional evidence to suggest that BRF2 and DSN1 overexpression/amplification, and the HER2 I767M mutation may be alterations that compensate for the lack of HER2 amplification in the HER2-negative components of HER2 heterogeneous breast cancers. Conclusions Our results indicate that even driver genetic alterations, such as HER2 gene amplification, can be heterogeneously distributed within a cancer, and that the HER2-negative components are likely driven by genetic alterations not present in the HER2-positive components, including BRF2 and DSN1 amplification and HER2 somatic mutations. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0657-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charlotte K Y Ng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Luciano G Martelotto
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Arnaud Gauthier
- Department of Tumor Biology, Institut Curie, 75248, Paris, France.
| | - Huei-Chi Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Salvatore Piscuoglio
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Raymond S Lim
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Catherine F Cowell
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Paul M Wilkerson
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, SW3 6JB, UK.
| | - Patty Wai
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, SW3 6JB, UK.
| | - Daniel N Rodrigues
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, SW3 6JB, UK.
| | - Laurent Arnould
- Department of Pathology and CRB Ferdinand Cabanne, Centre Georges Francois Leclerc, 21000, Dijon, France.
| | - Felipe C Geyer
- Departments of Anatomic Pathology and Oncology, Hospital Israelita Albert Einstein, São Paulo, 05652-900, Brazil.
| | - Silvio E Bromberg
- Departments of Anatomic Pathology and Oncology, Hospital Israelita Albert Einstein, São Paulo, 05652-900, Brazil.
| | - Magali Lacroix-Triki
- Department of Pathology, Institut Claudius Regaud, IUCT-Oncopole, 31059, Toulouse, France.
| | - Frederique Penault-Llorca
- Department of Pathology, Centre Jean Perrin, and University of Auvergne, 63000, Clermont Ferrand, France.
| | - Sylvia Giard
- Department of Pathology, Centre Oscar Lambret, 59000, Lille, France.
| | | | - Rachael Natrajan
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, SW3 6JB, UK.
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Paul H Cottu
- Department of Medical Oncology, Institut Curie, 75248, Paris, France.
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | | | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA. .,Affiliate Member, Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA. .,Affiliate Member, Computational Biology Center, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
12
|
Vujkovic M, Attiyeh EF, Ries RE, Horn M, Goodman EK, Ding Y, Kavcic M, Alonzo TA, Gerbing RB, Hirsch B, Raimondi S, Gamis AS, Meshinchi S, Aplenc R. Concordance of copy number alterations using a common analytic pipeline for genome-wide analysis of Illumina and Affymetrix genotyping data: a report from the Children's Oncology Group. Cancer Genet 2015; 208:408-13. [PMID: 26163103 DOI: 10.1016/j.cancergen.2015.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 12/30/2022]
Abstract
Copy number alterations (CNAs) are a hallmark of pediatric cancer genomes. An increasing number of research groups use multiple platforms and software packages to detect and analyze CNAs. However, different platforms have experimental and analysis-specific biases that may yield different results. We sought to estimate the concordance of CNAs in children with de novo acute myeloid leukemia between two experimental platforms: Affymetrix SNP 6.0 array and Illumina OmniQuad 2.5 BeadChip. Forty-five paired tumor-remission samples were genotyped on both platforms, and CNAs were estimated from total signal intensity and allelic contrast values using the allele-specific copy number analysis of tumors (ASCAT) algorithm. The two platforms were comparable in detection of CNAs, each missing only two segments from a total of 42 CNAs (4.6%). Overall, there was an interplatform agreement of 96% for allele-specific tumor profiles. However, poor quality samples with low signal/noise ratios showed a high rate of false-positive segments independent of the genotyping platform. These results demonstrate that a common analytic pipeline can be utilized for SNP array data from these two platforms. The customized programming template for the preprocessing, data integration, and analysis is publicly available at https://github.com/AplenCHOP/affyLumCNA.
Collapse
Affiliation(s)
- Marijana Vujkovic
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Edward F Attiyeh
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rhonda E Ries
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Michelle Horn
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth K Goodman
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yang Ding
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Todd A Alonzo
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | | | - Susana Raimondi
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alan S Gamis
- Division of Hematology/Oncology/Bone Marrow Transplantation, Children's Mercy Hospitals and Clinics, Kansas City, MO, USA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Richard Aplenc
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
13
|
Strefford JC. The genomic landscape of chronic lymphocytic leukaemia: biological and clinical implications. Br J Haematol 2014; 169:14-31. [PMID: 25496136 DOI: 10.1111/bjh.13254] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic lymphocytic leukaemia (CLL) remains at the forefront of the genetic analysis of human tumours, principally due its prevalence, protracted natural history and accessibility to suitable material for analysis. With the application of high-throughput genetic technologies, we have an unbridled view of the architecture of the CLL genome, including a comprehensive description of the copy number and mutational landscape of the disease, a detailed picture of clonal evolution during pathogenesis, and the molecular mechanisms that drive genomic instability and therapeutic resistance. This work has nuanced the prognostic importance of established copy number alterations, and identified novel prognostically relevant gene mutations that function within biological pathways that are attractive treatment targets. Herein, an overview of recent genomic discoveries will be reviewed, with associated biological and clinical implications, and a view into how clinical implementation may be facilitated.
Collapse
Affiliation(s)
- Jonathan C Strefford
- Cancer Genomics, Academic Unit of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
14
|
Sutton LA, Rosenquist R. Clonal evolution in chronic lymphocytic leukemia: impact of subclonality on disease progression. Expert Rev Hematol 2014; 8:71-8. [PMID: 25345442 DOI: 10.1586/17474086.2015.972930] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In recent years, next-generation sequencing has unraveled the molecular landscape in chronic lymphocytic leukemia with the discovery of a number of recurrently mutated genes. Mutations in several of these genes, such as NOTCH1, SF3B1 and BIRC3, are linked to a more aggressive disease with early disease progression, short time-to-first-treatment and even chemorefractoriness. Although in its infancy, we have also begun to understand the complex dynamics of subclonal diversity and its impact on disease outcome. From pioneering studies, we know that certain genetic events are found in the majority of chronic lymphocytic leukemia cells and are considered as 'clonal driver mutations' (e.g., +12, 13q-), whereas others, present only in a fraction of the tumor, are deemed to be 'subclonal driver mutations' for example, TP53 and SF3B1. Over the coming years, we need to gain a deeper insight into the dynamics of this subclonal architecture to understand how, at an individual level, chronic lymphocytic leukemia patients should be followed, which will be particularly relevant as novel targeted therapies begin to emerge.
Collapse
Affiliation(s)
- Lesley-Ann Sutton
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751-85, Uppsala, Sweden
| | | |
Collapse
|
15
|
Natrajan R, Wilkerson PM, Marchiò C, Piscuoglio S, Ng CKY, Wai P, Lambros MB, Samartzis EP, Dedes KJ, Frankum J, Bajrami I, Kopec A, Mackay A, A'hern R, Fenwick K, Kozarewa I, Hakas J, Mitsopoulos C, Hardisson D, Lord CJ, Kumar-Sinha C, Ashworth A, Weigelt B, Sapino A, Chinnaiyan AM, Maher CA, Reis-Filho JS. Characterization of the genomic features and expressed fusion genes in micropapillary carcinomas of the breast. J Pathol 2014; 232:553-65. [PMID: 24395524 PMCID: PMC4013428 DOI: 10.1002/path.4325] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 12/04/2013] [Accepted: 12/29/2013] [Indexed: 12/30/2022]
Abstract
Micropapillary carcinoma (MPC) is a rare histological special type of breast cancer, characterized by an aggressive clinical behaviour and a pattern of copy number aberrations (CNAs) distinct from that of grade- and oestrogen receptor (ER)-matched invasive carcinomas of no special type (IC-NSTs). The aims of this study were to determine whether MPCs are underpinned by a recurrent fusion gene(s) or mutations in 273 genes recurrently mutated in breast cancer. Sixteen MPCs were subjected to microarray-based comparative genomic hybridization (aCGH) analysis and Sequenom OncoCarta mutation analysis. Eight and five MPCs were subjected to targeted capture and RNA sequencing, respectively. aCGH analysis confirmed our previous observations about the repertoire of CNAs of MPCs. Sequencing analysis revealed a spectrum of mutations similar to those of luminal B IC-NSTs, and recurrent mutations affecting mitogen-activated protein kinase family genes and NBPF10. RNA-sequencing analysis identified 17 high-confidence fusion genes, eight of which were validated and two of which were in-frame. No recurrent fusions were identified in an independent series of MPCs and IC-NSTs. Forced expression of in-frame fusion genes (SLC2A1-FAF1 and BCAS4-AURKA) resulted in increased viability of breast cancer cells. In addition, genomic disruption of CDK12 caused by out-of-frame rearrangements was found in one MPC and in 13% of HER2-positive breast cancers, identified through a re-analysis of publicly available massively parallel sequencing data. In vitro analyses revealed that CDK12 gene disruption results in sensitivity to PARP inhibition, and forced expression of wild-type CDK12 in a CDK12-null cell line model resulted in relative resistance to PARP inhibition. Our findings demonstrate that MPCs are neither defined by highly recurrent mutations in the 273 genes tested, nor underpinned by a recurrent fusion gene. Although seemingly private genetic events, some of the fusion transcripts found in MPCs may play a role in maintenance of a malignant phenotype and potentially offer therapeutic opportunities.
Collapse
Affiliation(s)
- Rachael Natrajan
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Paul M Wilkerson
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | | | - Salvatore Piscuoglio
- Department of Pathology, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
| | - Charlotte KY Ng
- Department of Pathology, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
| | - Patty Wai
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Maryou B Lambros
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | | | | | - Jessica Frankum
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Ilirjana Bajrami
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Alicja Kopec
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Alan Mackay
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Roger A'hern
- Cancer Research UK Clinical Trials Unit, The Institute of Cancer ResearchSutton, UK
| | - Kerry Fenwick
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Iwanka Kozarewa
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Jarle Hakas
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Costas Mitsopoulos
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - David Hardisson
- Department of Pathology, Hospital Universitario La Paz, Universidad Autonoma de Madrid, Hospital La Paz Institute for Health Research (IdiPAZ)Madrid, Spain
| | - Christopher J Lord
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Chandan Kumar-Sinha
- Michigan Center for Translational Pathology (MCTP), Department of Pathology, University of MichiganAnn Arbor, MI, USA
| | - Alan Ashworth
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
| | - Anna Sapino
- Department of Medical Sciences, University of TurinTurin, Italy
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology (MCTP), Department of Pathology, University of MichiganAnn Arbor, MI, USA
| | - Christopher A Maher
- Washington University Genome Institute, Washington UniversitySt Louis, MO, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
| |
Collapse
|
16
|
Forsberg LA, Absher D, Dumanski JP. Republished: Non-heritable genetics of human disease: spotlight on post-zygotic genetic variation acquired during lifetime. Postgrad Med J 2014; 89:417-26. [PMID: 23781115 PMCID: PMC3711362 DOI: 10.1136/postgradmedj-2012-101322rep] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The heritability of most common, multifactorial diseases is rather modest and known genetic effects account for a small part of it. The remaining portion of disease aetiology has been conventionally ascribed to environmental effects, with an unknown part being stochastic. This review focuses on recent studies highlighting stochastic events of potentially great importance in human disease—the accumulation of post-zygotic structural aberrations with age in phenotypically normal humans. These findings are in agreement with a substantial mutational load predicted to occur during lifetime within the human soma. A major consequence of these results is that the genetic profile of a single tissue collected at one time point should be used with caution as a faithful portrait of other tissues from the same subject or the same tissue throughout life. Thus, the design of studies in human genetics interrogating a single sample per subject or applying lymphoblastoid cell lines may come into question. Sporadic disorders are common in medicine. We wish to stress the non-heritable genetic variation as a potentially important factor behind the development of sporadic diseases. Moreover, associations between post-zygotic mutations, clonal cell expansions and their relation to cancer predisposition are central in this context. Post-zygotic mutations are amenable to robust examination and are likely to explain a sizable part of non-heritable disease causality, which has routinely been thought of as synonymous with environmental factors. In view of the widespread accumulation of genetic aberrations with age and strong predictions of disease risk from such analyses, studies of post-zygotic mutations may be a fruitful approach for delineation of variants that are causative for common human disorders.
Collapse
Affiliation(s)
- Lars Anders Forsberg
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, Uppsala, Sweden
| | | | | |
Collapse
|
17
|
Genomic profiling of histological special types of breast cancer. Breast Cancer Res Treat 2013; 142:257-69. [PMID: 24162157 DOI: 10.1007/s10549-013-2740-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 10/16/2013] [Indexed: 12/30/2022]
Abstract
Histological special types of breast cancer have distinctive morphological features and account for up to 25 % of all invasive breast cancers. We sought to determine whether at the genomic level, histological special types of breast cancer are distinct from grade- and estrogen receptor (ER)-matched invasive carcinomas of no special type (IC-NSTs), and to define genes whose expression correlates with gene copy number in histological special types of breast cancer. We characterized 59 breast cancers of ten histological special types using array-based comparative genomic hybridization (aCGH). Hierarchical clustering revealed that the patterns of gene copy number aberrations segregated with ER-status and histological grade, and that samples from each of the breast cancer histological special types preferentially clustered together. We confirmed the patterns of gene copy number aberrations previously reported for lobular, micropapillary, metaplastic, and mucinous carcinomas. On the other hand, metaplastic and medullary carcinomas were found to have genomic profiles similar to those of grade- and ER-matched IC-NSTs. The genomic aberrations observed in invasive carcinomas with osteoclast-like stromal giant cells support its classification as IC-NST variant. Integrative aCGH and gene expression analysis led to the identification of 145 transcripts that were significantly overexpressed when amplified in histological special types of breast cancer. Our results illustrate that together with histological grade and ER-status, histological type is also associated with the patterns and complexity of gene copy number aberrations in breast cancer, with adenoid cystic and mucinous carcinomas being examples of ER-negative and ER-positive breast cancers with distinctive repertoires of gene copy number aberrations.
Collapse
|
18
|
Improgo MR, Brown JR. Genomic approaches to chronic lymphocytic leukemia. Hematol Oncol Clin North Am 2013; 27:157-71. [PMID: 23561468 DOI: 10.1016/j.hoc.2013.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
This article discusses recent advances in genomic approaches used to understand chronic lymphocytic leukemia. Tools for analyzing DNA-level lesions are described, data obtained from these various platforms summarized, and the clinical relevance of these findings discussed.
Collapse
Affiliation(s)
- Ma Reina Improgo
- Department of Medical Oncology, Dana-Farber Cancer Institute, CLL Center, Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|
19
|
Mansouri L, Grabowski P, Degerman S, Svenson U, Gunnarsson R, Cahill N, Smedby KE, Geisler C, Juliusson G, Roos G, Rosenquist R. Short telomere length is associated with NOTCH1/SF3B1/TP53 aberrations and poor outcome in newly diagnosed chronic lymphocytic leukemia patients. Am J Hematol 2013; 88:647-51. [PMID: 23620080 DOI: 10.1002/ajh.23466] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 03/28/2013] [Accepted: 04/18/2013] [Indexed: 12/21/2022]
Abstract
Most previous studies on telomere length (TL) in chronic lymphocytic leukemia (CLL) are based on referral cohorts including a high proportion of aggressive cases. Here, the impact of TL was analyzed in a population-based cohort of newly diagnosed CLL (n = 265) and in relation to other prognostic markers. Short telomeres were particularly associated with high-risk genetic markers, such as NOTCH1, SF3B1, or TP53 aberrations, and predicted a short time to treatment (TTT) and overall survival (OS) (both P < 0.0001). TL was an independent prognostic factor and subdivided patients with otherwise good-prognostic features (e.g., mutated IGHV genes, favorable cytogenetics) into subgroups with different outcome. Furthermore, in follow-up samples (n = 119) taken 5-8 years after diagnosis, TL correlated well with TL at diagnosis and remained unaffected by treatment. Altogether, these novel data indicate that short TL already at diagnosis is associated with poor outcome in CLL and that TL can be measured at later stages of the disease.
Collapse
Affiliation(s)
- Larry Mansouri
- Department of Immunology; Genetics; and Pathology; Rudbeck Laboratory; Uppsala University; Uppsala; Sweden
| | - Pawel Grabowski
- Department of Medical Biosciences; Umeå University; Umeå; Sweden
| | - Sofie Degerman
- Department of Medical Biosciences; Umeå University; Umeå; Sweden
| | - Ulrika Svenson
- Department of Medical Biosciences; Umeå University; Umeå; Sweden
| | - Rebeqa Gunnarsson
- Department of Immunology; Genetics; and Pathology; Rudbeck Laboratory; Uppsala University; Uppsala; Sweden
| | - Nicola Cahill
- Department of Immunology; Genetics; and Pathology; Rudbeck Laboratory; Uppsala University; Uppsala; Sweden
| | - Karin Ekström Smedby
- Department of Medicine; Clinical Epidemiology Unit; Karolinska Institutet; Stockholm; Sweden
| | | | - Gunnar Juliusson
- Department of Laboratory Medicine; Stem Cell Center; Hematology and Transplantation; Lund University; Lund; Sweden
| | - Göran Roos
- Department of Medical Biosciences; Umeå University; Umeå; Sweden
| | - Richard Rosenquist
- Department of Immunology; Genetics; and Pathology; Rudbeck Laboratory; Uppsala University; Uppsala; Sweden
| |
Collapse
|
20
|
Alsolami R, Knight SJL, Schuh A. Clinical application of targeted and genome-wide technologies: can we predict treatment responses in chronic lymphocytic leukemia? Per Med 2013; 10:361-376. [PMID: 24611071 PMCID: PMC3943176 DOI: 10.2217/pme.13.33] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is low-grade lymphoma of mature B cells and it is considered to be the most common type of hematological malignancy in the western world. CLL is characterized by a chronically relapsing course and clinical and biological heterogeneity. Many patients do not require any treatment for years. Although important progress has been made in the treatment of CLL, none of the conventional treatment options are curative. Recurrent chromosomal abnormalities have been identified and are associated with prognosis and pathogenesis of the disease. More recently, unbiased genome-wide technologies have identified multiple additional recurrent aberrations. The precise predictive value of these has not been established, but it is likely that the genetic heterogeneity observed at least partly reflects the clinical variability. The present article reviews our current knowledge of predictive markers in CLL using whole-genome technologies.
Collapse
Affiliation(s)
- Reem Alsolami
- Oxford National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
- King Abdulaziz University, Faculty of Applied Medical Sciences, Jeddah, Saudi Arabia
| | - Samantha JL Knight
- Oxford National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Anna Schuh
- Oxford National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
| |
Collapse
|
21
|
Cooley LD, Lebo M, Li MM, Slovak ML, Wolff DJ. American College of Medical Genetics and Genomics technical standards and guidelines: microarray analysis for chromosome abnormalities in neoplastic disorders. Genet Med 2013; 15:484-94. [DOI: 10.1038/gim.2013.49] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
22
|
Gunnarsson R, Mansouri L, Rosenquist R. Exploring the genetic landscape in chronic lymphocytic leukemia using high-resolution technologies. Leuk Lymphoma 2013; 54:1583-90. [PMID: 23167608 DOI: 10.3109/10428194.2012.751530] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abstract During recent years, microarray-based technologies and next-generation sequencing (NGS) have been applied in chronic lymphocytic leukemia (CLL) in order to identify novel genomic aberrations that may contribute to the pathogenesis of the disease. Even though high-resolution microarray studies have confirmed the importance of the known recurrent aberrations, i.e. del(11q), trisomy 12, del(13q) and del(17p), and have more precisely delineated the genomic borders of these aberrations, only a few novel aberrations, found at a low frequency, have been detected with these techniques. In contrast to this, the application of NGS technology of the coding genome (exome sequencing) or the entire genome (whole-genome sequencing) has unveiled a number of novel recurrent mutations in e.g. the NOTCH1, SF3B1 and BIRC3 genes. Importantly, mutations in these latter genes were reported to be associated with a particularly poor outcome, similar to TP53 aberrations, and may play key roles in tumor development, treatment resistance and prognosis. In this review, we not only summarize the latest achievements using array-based or NGS technologies, but also point to new directions for research aiming to unravel the complex genetic "map" in CLL and its prognostic subsets.
Collapse
Affiliation(s)
- Rebeqa Gunnarsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|
23
|
Shiu KK, Wetterskog D, Mackay A, Natrajan R, Lambros M, Sims D, Bajrami I, Brough R, Frankum J, Sharpe R, Marchio C, Horlings H, Reyal F, van der Vijver M, Turner N, Reis-Filho JS, Lord CJ, Ashworth A. Integrative molecular and functional profiling of ERBB2-amplified breast cancers identifies new genetic dependencies. Oncogene 2013; 33:619-31. [PMID: 23334330 DOI: 10.1038/onc.2012.625] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 11/04/2012] [Accepted: 11/14/2012] [Indexed: 12/30/2022]
Abstract
Overexpression of the receptor tyrosine kinase ERBB2 (also known as HER2) occurs in around 15% of breast cancers and is driven by amplification of the ERBB2 gene. ERBB2 amplification is a marker of poor prognosis, and although anti-ERBB2-targeted therapies have shown significant clinical benefit, de novo and acquired resistance remains an important problem. Genomic profiling has demonstrated that ERBB2+ve breast cancers are distinguished from ER+ve and 'triple-negative' breast cancers by harbouring not only the ERBB2 amplification on 17q12, but also a number of co-amplified genes on 17q12 and amplification events on other chromosomes. Some of these genes may have important roles in influencing clinical outcome, and could represent genetic dependencies in ERBB2+ve cancers and therefore potential therapeutic targets. Here, we describe an integrated genomic, gene expression and functional analysis to determine whether the genes present within amplicons are critical for the survival of ERBB2+ve breast tumour cells. We show that only a fraction of the ERBB2-amplified breast tumour lines are truly addicted to the ERBB2 oncogene at the mRNA level and display a heterogeneous set of additional genetic dependencies. These include an addiction to the transcription factor gene TFAP2C when it is amplified and overexpressed, suggesting that TFAP2C represents a genetic dependency in some ERBB2+ve breast cancer cells.
Collapse
Affiliation(s)
- K-K Shiu
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - D Wetterskog
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - A Mackay
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - R Natrajan
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - M Lambros
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - D Sims
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - I Bajrami
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - R Brough
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - J Frankum
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - R Sharpe
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - C Marchio
- Department of Biomedical Sciences and Human Oncology, University of Turin, Turin, Italy
| | - H Horlings
- Department of Pathology, Academic Medical Centre, Amsterdam, The Netherlands
| | - F Reyal
- Department of Pathology, Academic Medical Centre, Amsterdam, The Netherlands
| | - M van der Vijver
- Department of Pathology, Academic Medical Centre, Amsterdam, The Netherlands
| | - N Turner
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - J S Reis-Filho
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - C J Lord
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - A Ashworth
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| |
Collapse
|
24
|
Forsberg LA, Absher D, Dumanski JP. Non-heritable genetics of human disease: spotlight on post-zygotic genetic variation acquired during lifetime. J Med Genet 2013; 50:1-10. [PMID: 23172682 PMCID: PMC3534255 DOI: 10.1136/jmedgenet-2012-101322] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 10/18/2012] [Accepted: 10/19/2012] [Indexed: 01/06/2023]
Abstract
The heritability of most common, multifactorial diseases is rather modest and known genetic effects account for a small part of it. The remaining portion of disease aetiology has been conventionally ascribed to environmental effects, with an unknown part being stochastic. This review focuses on recent studies highlighting stochastic events of potentially great importance in human disease-the accumulation of post-zygotic structural aberrations with age in phenotypically normal humans. These findings are in agreement with a substantial mutational load predicted to occur during lifetime within the human soma. A major consequence of these results is that the genetic profile of a single tissue collected at one time point should be used with caution as a faithful portrait of other tissues from the same subject or the same tissue throughout life. Thus, the design of studies in human genetics interrogating a single sample per subject or applying lymphoblastoid cell lines may come into question. Sporadic disorders are common in medicine. We wish to stress the non-heritable genetic variation as a potentially important factor behind the development of sporadic diseases. Moreover, associations between post-zygotic mutations, clonal cell expansions and their relation to cancer predisposition are central in this context. Post-zygotic mutations are amenable to robust examination and are likely to explain a sizable part of non-heritable disease causality, which has routinely been thought of as synonymous with environmental factors. In view of the widespread accumulation of genetic aberrations with age and strong predictions of disease risk from such analyses, studies of post-zygotic mutations may be a fruitful approach for delineation of variants that are causative for common human disorders.
Collapse
Affiliation(s)
- Lars Anders Forsberg
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|
25
|
Abstract
This chapter summarizes the current knowledge on gene copy number changes found in lung tumors, and their application in the diagnosis, prognostication, and prediction of response to chemotherapy. Examples of the identification of specific "driver" oncogenes within amplified DNA segments are described. A model of how array-CGH could be integrated clinically into the routine workup of lung cancers in clinical laboratory is proposed.
Collapse
Affiliation(s)
- Kenneth J Craddock
- Department of Pathology, Toronto General Hospital University Health Network, Toronto, ON, Canada.
| | | | | |
Collapse
|
26
|
Schweighofer CD, Coombes KR, Majewski T, Barron LL, Lerner S, Sargent RL, O'Brien S, Ferrajoli A, Wierda WG, Czerniak BA, Medeiros LJ, Keating MJ, Abruzzo LV. Genomic variation by whole-genome SNP mapping arrays predicts time-to-event outcome in patients with chronic lymphocytic leukemia: a comparison of CLL and HapMap genotypes. J Mol Diagn 2012; 15:196-209. [PMID: 23273604 DOI: 10.1016/j.jmoldx.2012.09.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 09/20/2012] [Accepted: 09/27/2012] [Indexed: 02/06/2023] Open
Abstract
Genomic abnormalities, such as deletions in 11q22 or 17p13, are associated with poorer prognosis in patients with chronic lymphocytic leukemia (CLL). We hypothesized that unknown regions of copy number variation (CNV) affect clinical outcome and can be detected by array-based single-nucleotide polymorphism (SNP) genotyping. We compared SNP genotypes from 168 untreated patients with CLL with genotypes from 73 white HapMap controls. We identified 322 regions of recurrent CNV, 82 of which occurred significantly more often in CLL than in HapMap (CLL-specific CNV), including regions typically aberrant in CLL: deletions in 6q21, 11q22, 13q14, and 17p13 and trisomy 12. In univariate analyses, 35 of total and 11 of CLL-specific CNVs were associated with unfavorable time-to-event outcomes, including gains or losses in chromosomes 2p, 4p, 4q, 6p, 6q, 7q, 11p, 11q, and 17p. In multivariate analyses, six CNVs (ie, CLL-specific variations in 11p15.1-15.4 or 6q27) predicted time-to-treatment or overall survival independently of established markers of prognosis. Moreover, genotypic complexity (ie, the number of independent CNVs per patient) significantly predicted prognosis, with a median time-to-treatment of 64 months versus 23 months in patients with zero to one versus two or more CNVs, respectively (P = 3.3 × 10(-8)). In summary, a comparison of SNP genotypes from patients with CLL with HapMap controls allowed us to identify known and unknown recurrent CNVs and to determine regions and rates of CNV that predict poorer prognosis in patients with CLL.
Collapse
Affiliation(s)
- Carmen D Schweighofer
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Staaf J, Isaksson S, Karlsson A, Jönsson M, Johansson L, Jönsson P, Botling J, Micke P, Baldetorp B, Planck M. Landscape of somatic allelic imbalances and copy number alterations in human lung carcinoma. Int J Cancer 2012; 132:2020-31. [PMID: 23023297 DOI: 10.1002/ijc.27879] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 09/13/2012] [Indexed: 12/27/2022]
Abstract
Lung cancer is the worldwide leading cause of death from cancer and has been shown to be a heterogeneous disease at the genomic level. To delineate the genomic landscape of copy number alterations, amplifications, loss-of-heterozygosity (LOH), tumor ploidy and copy-neutral allelic imbalance in lung cancer, microarray-based genomic profiles from 2,141 tumors and cell lines including adenocarcinomas (AC, n = 1,206), squamous cell carcinomas (SqCC, n = 467), large cell carcinomas (n = 37) and small cell lung carcinomas (SCLC, n = 88) were assembled from different repositories. Copy number alteration differences between lung cancer histologies were confirmed in 285 unrelated tumors analyzed by BAC array comparative genomic hybridization. Tumor ploidy patterns were validated by DNA flow cytometry analysis of 129 unrelated cases. Eighty-nine recurrent copy number alterations (55 gains, 34 losses) were identified harboring genes with gene expression putatively driven by gene dosage through integration with gene expression data for 496 cases. Thirteen and 26 of identified regions discriminated AC/SqCC and AC/SqCC/SCLC, respectively, while 48 regions harbored recurrent (n > 15) high-level amplifications comprising established and putative oncogenes, differing in frequency and coamplification patterns between histologies. Lung cancer histologies displayed differences in patterns/frequency of copy number alterations, genomic architecture, LOH, copy-neutral allelic imbalance and tumor ploidy, with AC generally displaying less copy number alterations and allelic imbalance. Moreover, a strong association was demonstrated between different types of copy number alterations and allelic imbalances with tumor aneuploidy. In summary, these analyses provide a comprehensive overview of the landscape of genomic alterations in lung cancer, highlighting differences but also similarities between subgroups of the disease.
Collapse
Affiliation(s)
- Johan Staaf
- Department of Oncology, Clinical Sciences, Lund University and Skåne University Hospital, Barngatan 2B, Lund, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Puiggros A, Puigdecanet E, Salido M, Ferrer A, Abella E, Gimeno E, Nonell L, Herranz MJ, Galván AB, Rodríguez-Rivera M, Melero C, Pairet S, Bellosillo B, Serrano S, Florensa L, Solé F, Espinet B. Genomic arrays in chronic lymphocytic leukemia routine clinical practice: are we ready to substitute conventional cytogenetics and fluorescence in situ hybridization techniques? Leuk Lymphoma 2012; 54:986-95. [PMID: 22994157 DOI: 10.3109/10428194.2012.731598] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by a highly variable clinical course. Del(11q) and del(17p), routinely studied by conventional G-banding cytogenetics (CGC) and fluorescence in situ hybridization (FISH), have been related to progression and shorter overall survival. Recently, array-based karyotyping has gained acceptance as a high-resolution new tool for detecting genomic imbalances. The aim of the present study was to compare genomic arrays with CGC and FISH to ascertain whether the current techniques could be substituted in routine procedures. We analyzed 70 patients with CLL using the Cytogenetics Whole-Genome 2.7M Array and CytoScan HD Array (Affymetrix), CGC and FISH with the classical CLL panel. Whereas 31.4% and 68.6% of patients presented abnormalities when studied by CGC and FISH, respectively, these rates increased when arrays were also analyzed (78.6% and 80%). Although abnormality detection is higher when arrays are applied, one case with del(11q) and three with del(17p) were missed by genomic arrays due to their limited sensitivity. We consider that the complete substitution of CGC and FISH by genomic arrays in routine laboratories could negatively affect the management of some patients harboring 11q or 17p deletions. In conclusion, genomic arrays are valid to detect known and novel genomic imbalances in CLL, but should be maintained as a complementary tool to the current techniques.
Collapse
Affiliation(s)
- Anna Puiggros
- Programa de Recerca en Càncer, Grup de Recerca Translacional en Neoplàsies Hematològiques (GRETNHE), IMIM-Hospital del Mar, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Microarray, gene sequencing, and reverse transcriptase–polymerase chain reaction analyses of a cryptic PML-RARA translocation. Cancer Genet 2012; 205:537-40. [DOI: 10.1016/j.cancergen.2012.07.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 07/06/2012] [Accepted: 07/31/2012] [Indexed: 11/22/2022]
|
30
|
Cahill N, Bergh AC, Kanduri M, Göransson-Kultima H, Mansouri L, Isaksson A, Ryan F, Smedby KE, Juliusson G, Sundström C, Rosén A, Rosenquist R. 450K-array analysis of chronic lymphocytic leukemia cells reveals global DNA methylation to be relatively stable over time and similar in resting and proliferative compartments. Leukemia 2012; 27:150-8. [DOI: 10.1038/leu.2012.245] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
31
|
Hernandez L, Wilkerson PM, Lambros MB, Campion-Flora A, Rodrigues DN, Gauthier A, Cabral C, Pawar V, Mackay A, A’Hern R, Marchiò C, Palacios J, Natrajan R, Weigelt B, Reis-Filho JS. Genomic and mutational profiling of ductal carcinomas in situ and matched adjacent invasive breast cancers reveals intra-tumour genetic heterogeneity and clonal selection. J Pathol 2012; 227:42-52. [PMID: 22252965 PMCID: PMC4975517 DOI: 10.1002/path.3990] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 01/11/2012] [Accepted: 01/12/2012] [Indexed: 12/21/2022]
Abstract
The mechanisms underlying the progression from ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) of the breast are yet to be fully elucidated. Several hypotheses have been put forward to explain the progression from DCIS to IDC, including the selection of a subpopulation of cancer cells with specific genetic aberrations, and the acquisition of new genetic aberrations or non-genetic mechanisms mediated by the tumour microenvironment. To determine whether synchronously diagnosed ipsilateral DCI and IDCs have modal populations with distinct repertoires of gene copy number aberrations and mutations in common oncogenes, matched frozen samples of DCIS and IDC were retrieved from 13 patients and subjected to microarray-based comparative genomic hybridization (aCGH) and Sequenom MassARRAY (Oncocarta v 1.0 panel). Fluorescence in situ hybridization and Sanger sequencing were employed to validate the aCGH and Sequenom findings, respectively. Although the genomic profiles of matched DCI and IDCs were similar, in three of 13 matched pairs amplification of distinct loci (ie 1q41, 2q24.2, 6q22.31, 7q11.21, 8q21.2 and 9p13.3) was either restricted to, or more prevalent in, the modal population of cancer cells of one of the components. Sequenom MassARRAY identified PIK3CA mutations restricted to the DCIS component in two cases, and in a third case the frequency of the PIK3CA mutant allele reduced from 49% in the DCIS to 25% in the IDC component. Despite the genomic similarities between synchronous DCIS and IDC, our data provide strong circumstantial evidence to suggest that in some cases the progression from DCIS to IDC is driven by the selection of non-modal clones that harbour a specific repertoire of genetic aberrations.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Class I Phosphatidylinositol 3-Kinases
- Clonal Evolution
- Clone Cells
- Comparative Genomic Hybridization
- DNA Mutational Analysis
- DNA, Neoplasm/analysis
- Disease Progression
- Female
- Gene Expression Profiling
- Genetic Heterogeneity
- Genomics/methods
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Mutation
- Neoplasms, Multiple Primary
- Phosphatidylinositol 3-Kinases/genetics
Collapse
Affiliation(s)
- Lucia Hernandez
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- Department of Anatomic Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Paul M Wilkerson
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Maryou B Lambros
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Adriana Campion-Flora
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Daniel Nava Rodrigues
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Arnaud Gauthier
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- Department of Tumour Biology, Institut Curie, 75005 Paris, France
| | - Cecilia Cabral
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Vidya Pawar
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Alan Mackay
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Roger A’Hern
- Cancer Research UK Clinical Trials Unit, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Caterina Marchiò
- Department of Biomedical Sciences and Human Oncology, University of Turin, Turin, Italy
| | - Jose Palacios
- Hospital Universitario Virgen del Rocio, Seville, Spain
| | - Rachael Natrajan
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Britta Weigelt
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, London WC2A 3LY, UK
| | - Jorge S Reis-Filho
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
32
|
Geyer FC, Lacroix-Triki M, Colombo PE, Patani N, Gauthier A, Natrajan R, Lambros MBK, Khalifeh I, Albarracin C, Orru S, Marchiò C, Sapino A, Mackay A, Weigelt B, Schmitt FC, Wesseling J, Sneige N, Reis-Filho JS. Molecular evidence in support of the neoplastic and precursor nature of microglandular adenosis. Histopathology 2012; 60:E115-30. [PMID: 22486256 DOI: 10.1111/j.1365-2559.2012.04207.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIMS Microglandular adenosis (MGA) is a proliferative breast lesion, which has been proposed to be a potential precursor of triple-negative breast cancers. The aims of this study were to determine whether MGAs harbour genetic alterations and if any such genetic aberrations found in MGAs are similar to those found in matched invasive carcinomas. METHODS AND RESULTS Twelve cases of MGA and/or atypical MGA (AMGA), 10 of which were associated with invasive carcinoma, were evaluated. Immunohistochemical profiling revealed that all invasive carcinomas were of triple-negative phenotype and expressed S100, cytokeratins 8/18 and 'basal' markers. The morphologically distinct components of each case (MGA, AMGA and/or invasive carcinoma) were microdissected and subjected to microarray comparative genomic hybridization. Apart from three typical MGAs, all samples harboured genetic alterations. The percentage of the genome affected by copy number aberrations in MGA/AMGA ranged from 0.5 to 61.9%, indicating varying levels of genetic instability. In three cases, MGA/AMGA displayed copy number aberrations similar to those found in matched invasive components, providing strong circumstantial evidence that MGA may constitute the substrate for the invasive carcinoma development. CONCLUSIONS Our results support the contention that MGA can be a clonal lesion and non-obligate precursor of triple-negative breast cancer.
Collapse
Affiliation(s)
- Felipe C Geyer
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Natrajan R, Mackay A, Wilkerson PM, Lambros MB, Wetterskog D, Arnedos M, Shiu KK, Geyer FC, Langerød A, Kreike B, Reyal F, Horlings HM, van de Vijver MJ, Palacios J, Weigelt B, Reis-Filho JS. Functional characterization of the 19q12 amplicon in grade III breast cancers. Breast Cancer Res 2012; 14:R53. [PMID: 22433433 PMCID: PMC3446387 DOI: 10.1186/bcr3154] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 03/04/2012] [Accepted: 03/20/2012] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION The 19q12 locus is amplified in a subgroup of oestrogen receptor (ER)-negative grade III breast cancers. This amplicon comprises nine genes, including cyclin E1 (CCNE1), which has been proposed as its 'driver'. The aim of this study was to identify the genes within the 19q12 amplicon whose expression is required for the survival of cancer cells harbouring their amplification. METHODS We investigated the presence of 19q12 amplification in a series of 313 frozen primary breast cancers and 56 breast cancer cell lines using microarray comparative genomic hybridisation (aCGH). The nine genes mapping to the smallest region of amplification on 19q12 were silenced using RNA interference in phenotypically matched breast cancer cell lines with (MDA-MB-157 and HCC1569) and without (Hs578T, MCF7, MDA-MB-231, ZR75.1, JIMT1 and BT474) amplification of this locus. Genes whose silencing was selectively lethal in amplified cells were taken forward for further validation. The effects of cyclin-dependent kinase 2 (CDK2) silencing and chemical inhibition were tested in cancer cells with and without CCNE1 amplification. RESULTS 19q12 amplification was identified in 7.8% of ER-negative grade III breast cancer. Of the nine genes mapping to this amplicon, UQCRFS1, POP4, PLEKHF1, C19ORF12, CCNE1 and C19ORF2 were significantly over-expressed when amplified in primary breast cancers and/or breast cancer cell lines. Silencing of POP4, PLEKHF1, CCNE1 and TSZH3 selectively reduced cell viability in cancer cells harbouring their amplification. Cancer cells with CCNE1 amplification were shown to be dependent on CDK2 expression and kinase activity for their survival. CONCLUSIONS The 19q12 amplicon may harbour more than a single 'driver', given that expression of POP4, PLEKHF1, CCNE1 and TSZH3 is required for the survival of cancer cells displaying their amplification. The observation that cancer cells harbouring CCNE1 gene amplification are sensitive to CDK2 inhibitors provides a rationale for the testing of these chemical inhibitors in a subgroup of patients with ER-negative grade III breast cancers.
Collapse
Affiliation(s)
- Rachael Natrajan
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Alan Mackay
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Paul M Wilkerson
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Maryou B Lambros
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Daniel Wetterskog
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Monica Arnedos
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Kai-Keen Shiu
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Felipe C Geyer
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Anita Langerød
- Department of Genetics, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Ullernchausèen 70, Montebello, Oslo, 0310, Norway
| | - Bas Kreike
- Institute for Radiation Oncology Arnhem, Wagnerlaan 47, Arnhem 6815 AD, The Netherlands
| | - Fabien Reyal
- Department of Surgery, Institut Curie, 26 rue d'Ulm, Paris, 75005, France
| | - Hugo M Horlings
- Department of Pathology, Academic Medical Center, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Marc J van de Vijver
- Department of Pathology, Academic Medical Center, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Jose Palacios
- Servicio de Anatomia Patologica, HHUU Virgen del Rocío, Avda. Manuel Siurot, s/n, Seville, 41013, Spain
| | - Britta Weigelt
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Jorge S Reis-Filho
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| |
Collapse
|
34
|
Abstract
Whole-genome genetic diagnostics has changed the clinical landscape of pediatric and adolescent medicine. In this article, we review the history of clinical cytogenetics as the field has progressed from studying chromosomes prepared from cells squashed between 2 slides to the high-resolution, whole-genome technology in use today, which has allowed for the identification of numerous previously unrecognized microdeletion and microduplication syndromes. Types of arrays and the data they collect are addressed, as are the types of results that array comparative genomic hybridization studies may generate. Throughout the review, we present case stories to illustrate the familiar (Down syndrome) and the new (a never-before reported microdeletion on the long arm of chromosome 12).
Collapse
|
35
|
Li Z, Fang ZY, Ding Y, Yao WT, Yang Y, Zhu ZQ, Wang W, Zhang QX. Amplifications of NCOA3 gene in colorectal cancers in a Chinese population. World J Gastroenterol 2012; 18:855-60. [PMID: 22371647 PMCID: PMC3286150 DOI: 10.3748/wjg.v18.i8.855] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2011] [Revised: 08/08/2011] [Accepted: 08/31/2011] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the copy number variation of NACO3 gene in colorectal cancer (CRC) and its correlation with tumor progression.
METHODS: A total of 142 samples of case-matched CRC tissues and adjacent normal tissues were obtained from patients undergoing bowel resection. Quantitative real-time polymerase chain reaction method was used to investigate the copy number variations of NCOA3 as well as gene expression in the collected tissues.
RESULTS: Copy number gains of NCOA3 were detected in 39 CRC samples (27.5%) and were correlated with tumor progression (χ2 = 6.42, P = 0.0112). Moreover, there was a positive correlation between copy number gain and mRNA over-expression of NCOA3 in CRCs (P = 0.0023). Expression level of NCOA3 mRNA was also enhanced in the CRC samples with unaltered copy numbers (3.85 ± 1.23 vs 2.71 ± 0.64, P < 0.01).
CONCLUSION: Sporadic colorectal cancers exhibit different mechanisms of NCOA3 regulation.
Collapse
|
36
|
Sellmann L, Carpinteiro A, Nückel H, Scholtysik R, Siemer D, Klein-Hipass L, Kube D, Dürig J, Dührsen U, Stanelle J, Küppers R. p53 protein expression in chronic lymphocytic leukemia. Leuk Lymphoma 2012; 53:1282-8. [PMID: 22220854 DOI: 10.3109/10428194.2011.654115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Alterations in the function of the p53 pathway are frequently described in chronic lymphocytic leukemia (CLL), mostly associated with deletion of 17p13 and/or mutations of the TP53 gene. In the present study, we investigated 103 CLLs for the impact of protein expression of full-length p53 and its isoforms β and γ. A strong correlation between deletions of 17p13 and an accumulation of full-length p53 protein was found and was associated with a worse outcome compared to CLL with normal p53 (treatment-free survival p < 0.001, overall survival p = 0.04). Interestingly, the relative expression levels between full-length p53 protein and its isoforms β and γ were significantly altered in CLL even without deletions of 17p13, compared to normal B-cells (p = 0.005). Furthermore, CLLs with higher p53 protein ratios showed worse clinical courses compared to CLLs with lower p53 protein ratios. Taken together, the differential expression of p53 isoforms could disrupt the p53 response and contribute to CLL pathogenesis.
Collapse
Affiliation(s)
- Ludger Sellmann
- Department of Hematology, Medical Faculty, University of Duisburg-Essen, Essen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Age-related somatic structural changes in the nuclear genome of human blood cells. Am J Hum Genet 2012; 90:217-28. [PMID: 22305530 DOI: 10.1016/j.ajhg.2011.12.009] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 12/06/2011] [Accepted: 12/14/2011] [Indexed: 01/10/2023] Open
Abstract
Structural variations are among the most frequent interindividual genetic differences in the human genome. The frequency and distribution of de novo somatic structural variants in normal cells is, however, poorly explored. Using age-stratified cohorts of 318 monozygotic (MZ) twins and 296 single-born subjects, we describe age-related accumulation of copy-number variation in the nuclear genomes in vivo and frequency changes for both megabase- and kilobase-range variants. Megabase-range aberrations were found in 3.4% (9 of 264) of subjects ≥60 years old; these subjects included 78 MZ twin pairs and 108 single-born individuals. No such findings were observed in 81 MZ pairs or 180 single-born subjects who were ≤55 years old. Recurrent region- and gene-specific mutations, mostly deletions, were observed. Longitudinal analyses of 43 subjects whose data were collected 7-19 years apart suggest considerable variation in the rate of accumulation of clones carrying structural changes. Furthermore, the longitudinal analysis of individuals with structural aberrations suggests that there is a natural self-removal of aberrant cell clones from peripheral blood. In three healthy subjects, we detected somatic aberrations characteristic of patients with myelodysplastic syndrome. The recurrent rearrangements uncovered here are candidates for common age-related defects in human blood cells. We anticipate that extension of these results will allow determination of the genetic age of different somatic-cell lineages and estimation of possible individual differences between genetic and chronological age. Our work might also help to explain the cause of an age-related reduction in the number of cell clones in the blood; such a reduction is one of the hallmarks of immunosenescence.
Collapse
|
38
|
Duprez R, Wilkerson PM, Lacroix-Triki M, Lambros MB, MacKay A, A’Hern R, Gauthier A, Pawar V, Colombo PE, Daley F, Natrajan R, Ward E, MacGrogan G, Arbion F, Michenet P, Weigelt B, Vincent-Salomon A, Reis-Filho JS. Immunophenotypic and genomic characterization of papillary carcinomas of the breast. J Pathol 2012; 226:427-441. [PMID: 22025283 PMCID: PMC4962905 DOI: 10.1002/path.3032] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/21/2011] [Accepted: 10/12/2011] [Indexed: 12/20/2022]
Abstract
Papillary carcinomas are a special histological type of breast cancer and have a relatively good outcome. We characterized the genomic and phenotypic characteristics of papillary carcinomas to determine whether they would constitute an entity distinct from grade- and oestrogen receptor (ER)-matched invasive ductal carcinomas of no special type (IDC-NSTs). The phenotype of 63 papillary carcinomas of the breast and grade- and ER-matched IDC-NSTs was determined by immunohistochemistry. DNA of sufficient quality was extracted from 49 microdissected papillary carcinomas and 49 microdissected grade- and ER-matched IDC-NSTs. These samples were subjected to high-resolution microarray-based comparative genomic hybridization (aCGH) and Sequenom MassARRAY sequencing analysis of 19 known oncogenes. Papillary carcinomas were predominantly of low histological grade, expressed immunohistochemical markers consistent with a luminal phenotype, and a lower rate of lymph node metastasis and p53 expression than grade- and ER-matched IDC-NSTs. Papillary carcinomas displayed less genomic aberrations than grade- and ER-matched IDC-NSTs; however, the patterns of gene copy number aberrations found in papillary carcinomas were similar to those of ER- and grade-matched IDC-NSTs, including 16q losses. Furthermore, PIK3CA mutations were found in 43% and 29% of papillary carcinomas and grade- and ER-matched IDC-NSTs, respectively. The genomic profiles of encapsulated, solid and invasive papillary carcinomas, the three morphological subtypes, were remarkably similar. Our results demonstrate that papillary carcinomas are a homogeneous special histological type of breast cancer. The similarities in the genomic profiles of papillary carcinomas and grade- and ER-matched IDC-NSTs suggest that papillary carcinomas may be best positioned as part of the spectrum of ER-positive breast cancers, rather than as a distinct entity. Furthermore, the good prognosis of papillary carcinomas may stem from the low rates of lymph node metastasis and p53 expression, low number of gene copy number aberrations and high prevalence of PIK3CA mutations.
Collapse
Affiliation(s)
- Raphaëlle Duprez
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Paul M Wilkerson
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Magali Lacroix-Triki
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
- Institut Claudius Regaud, 31052 Toulouse, France
| | - Maryou B Lambros
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Alan MacKay
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Roger A’Hern
- CRUK Clinical Trials Unit, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Arnaud Gauthier
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
- Institut Curie, 75005 Paris, France
| | - Vidya Pawar
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Pierre-Emanuel Colombo
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Frances Daley
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Rachael Natrajan
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Eric Ward
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | | | - Flavie Arbion
- Centre Hospitalier Universitaire, 37044 Tours, France
| | | | - Britta Weigelt
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, London WC2A 3LY, UK
| | | | - Jorge S Reis-Filho
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| |
Collapse
|
39
|
Geyer FC, de Biase D, Lambros MBK, Ragazzi M, Lopez-Garcia MA, Natrajan R, Mackay A, Kurelac I, Gasparre G, Ashworth A, Eusebi V, Reis-Filho JS, Tallini G. Genomic profiling of mitochondrion-rich breast carcinoma: chromosomal changes may be relevant for mitochondria accumulation and tumour biology. Breast Cancer Res Treat 2012; 132:15-28. [PMID: 21509527 DOI: 10.1007/s10549-011-1504-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 04/04/2011] [Indexed: 02/08/2023]
Abstract
Oncocytic carcinomas are composed of mitochondrion-rich cells. Though recognised by the WHO classification as a histological special type of breast cancer, their status as a discrete pathological entity remains a matter of contention. Given that oncocytic tumours of other anatomical sites display distinct clinico-pathological and molecular features, we sought to define the molecular genetic features of mitochondrion-rich breast tumours and to compare them with a series of histological grade- and oestrogen receptor status-matched invasive ductal carcinomas of no special type. Seventeen mitochondrion-rich breast carcinomas, including nine bona fide oncocytic carcinomas, were profiled with antibodies against oestrogen, progesterone and androgen receptors, HER2, Ki67, GCDFP-15, chromogranin, epithelial membrane antigen, cytokeratin 7, cytokeratin 14, CD68 and mitochondria antigen. These tumours were microdissected and DNA extracted from samples with >70% of tumour cells. Fourteen cases yielded DNA of sufficient quality/quantity and were subjected to high-resolution microarray comparative genomic hybridisation analysis. The genomic profiles were compared to those of 28 grade- and oestrogen receptor status-matched invasive ductal carcinomas of no special type. Oncocytic and other mitochondrion-rich tumours did not differ significantly between themselves. As a group, mitochondrion-rich carcinomas were immunophenotypically heterogenous. Recurrent copy number changes were similar to those described in unselected breast cancers. However, unsupervised and supervised analysis identified a subset of mitochondrion-rich cancers, which often displayed gains of 11q13.1-q13.2 and 19p13. Changes in the latter two chromosomal regions have been shown to be associated with oncocytic tumours of the kidney and thyroid, respectively, and host several nuclear genes with specific mitochondrial function. Our results indicate that in a way akin to oncocytic tumours of other anatomical sites, at least a subset of mitochondrion-rich breast carcinomas may be underpinned by a distinct pattern of chromosomal changes potentially relevant for mitochondria accumulation and constitute a discrete molecular entity.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma/genetics
- Carcinoma/metabolism
- Carcinoma/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Chromosome Aberrations
- Chromosomes, Human/genetics
- Chromosomes, Human/metabolism
- Cluster Analysis
- Comparative Genomic Hybridization
- Female
- Humans
- Middle Aged
- Mitochondria/pathology
- Mitochondrial Proteins/genetics
- Neoplasm Grading
- Phenotype
- Receptors, Estrogen/metabolism
Collapse
Affiliation(s)
- Felipe C Geyer
- The Breakthrough Breast Cancer Research Centre, ICR, 237 Fulham Road, London SW3 6JB, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lindgren D, Höglund M, Vallon-Christersson J. Genotyping techniques to address diversity in tumors. Adv Cancer Res 2012; 112:151-82. [PMID: 21925304 DOI: 10.1016/b978-0-12-387688-1.00006-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Array-based genotyping platforms have during recent years been established as a valuable tool for the characterization of genomic alterations in cancer. The analysis of tumor samples, however, presents challenges for data analysis and interpretation. For example, tumor samples are often admixed with nonaberrant cells that define the tumor microenvironment, such as infiltrating lymphocytes and fibroblasts, or vasculature. Furthermore, tumors often comprise subclones harboring divergent aberrations that are acquired subsequent to the tumor-initiating event. The combined analysis of both genotype and copy number status obtained by array-based genotyping platforms provide opportunities to address these challenges. In this chapter, we present the basic principles for current array-based genotyping platforms and how they can be used to infer genotype and copy number for acquired genomic alterations. We describe how these techniques can be used to resolve tumor ploidy, normal cell admixture, and subclonality. We also exemplify how genotyping techniques can be applied in tumor studies to elucidate the hierarchy among tumor clones, and thus, provide means to study clonal expansion and tumor evolution.
Collapse
Affiliation(s)
- David Lindgren
- Center for Molecular Pathology, Department of Laboratory Medicine, Lund University, SUS Malmö, Malmö, Sweden
| | | | | |
Collapse
|
41
|
Staaf J, Jönsson G, Ringnér M, Baldetorp B, Borg A. Landscape of somatic allelic imbalances and copy number alterations in HER2-amplified breast cancer. Breast Cancer Res 2011; 13:R129. [PMID: 22169037 PMCID: PMC3326571 DOI: 10.1186/bcr3075] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 11/21/2011] [Accepted: 12/14/2011] [Indexed: 01/20/2023] Open
Abstract
Introduction Human epidermal growth factor receptor 2 (HER2)-amplified breast cancer represents a clinically well-defined subgroup due to availability of targeted treatment. However, HER2-amplified tumors have been shown to be heterogeneous at the genomic level by genome-wide microarray analyses, pointing towards a need of further investigations for identification of recurrent copy number alterations and delineation of patterns of allelic imbalance. Methods High-density whole genome array-based comparative genomic hybridization (aCGH) and single nucleotide polymorphism (SNP) array data from 260 HER2-amplified breast tumors or cell lines, and 346 HER2-negative breast cancers with molecular subtype information were assembled from different repositories. Copy number alteration (CNA), loss-of-heterozygosity (LOH), copy number neutral allelic imbalance (CNN-AI), subclonal CNA and patterns of tumor DNA ploidy were analyzed using bioinformatical methods such as genomic identification of significant targets in cancer (GISTIC) and genome alteration print (GAP). The patterns of tumor ploidy were confirmed in 338 unrelated breast cancers analyzed by DNA flow cytometry with concurrent BAC aCGH and gene expression data. Results A core set of 36 genomic regions commonly affected by copy number gain or loss was identified by integrating results with a previous study, together comprising > 400 HER2-amplified tumors. While CNN-AI frequency appeared evenly distributed over chromosomes in HER2-amplified tumors, not targeting specific regions and often < 20% in frequency, the occurrence of LOH was strongly associated with regions of copy number loss. HER2-amplified and HER2-negative tumors stratified by molecular subtypes displayed different patterns of LOH and CNN-AI, with basal-like tumors showing highest frequencies followed by HER2-amplified and luminal B cases. Tumor aneuploidy was strongly associated with increasing levels of LOH, CNN-AI, CNAs and occurrence of subclonal copy number events, irrespective of subtype. Finally, SNP data from individual tumors indicated that genomic amplification in general appears as monoallelic, that is, it preferentially targets one parental chromosome in HER2-amplified tumors. Conclusions We have delineated the genomic landscape of CNAs, amplifications, LOH, and CNN-AI in HER2-amplified breast cancer, but also demonstrated a strong association between different types of genomic aberrations and tumor aneuploidy irrespective of molecular subtype.
Collapse
Affiliation(s)
- Johan Staaf
- Department of Oncology, Clinical Sciences, Lund University and Skåne University Hospital, Barngatan 2B, SE 22185 Lund, Sweden.
| | | | | | | | | |
Collapse
|
42
|
Wetterskog D, Lopez-Garcia MA, Lambros MB, A'Hern R, Geyer FC, Milanezi F, Cabral MC, Natrajan R, Gauthier A, Shiu KK, Orr N, Shousha S, Gatalica Z, Mackay A, Palacios J, Reis-Filho JS, Weigelt B. Adenoid cystic carcinomas constitute a genomically distinct subgroup of triple-negative and basal-like breast cancers. J Pathol 2011; 226:84-96. [PMID: 22015727 DOI: 10.1002/path.2974] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 07/17/2011] [Accepted: 07/18/2011] [Indexed: 12/20/2022]
Abstract
Adenoid cystic carcinoma (AdCC) is a rare form of triple-negative and basal-like breast cancer that has an indolent clinical behaviour. Four breast AdCCs were recently shown to harbour the recurrent chromosomal translocation t(6;9)(q22-23;p23-24), which leads to the formation of the MYB-NFIB fusion gene. Our aims were (i) to determine the prevalence of the MYB-NFIB fusion gene in AdCCs of the breast; (ii) to characterize the gene copy number aberrations found in AdCCs; and (iii) to determine whether AdCCs are genomically distinct from histological grade-matched or triple-negative and basal-like invasive ductal carcinomas of no special type (IDC-NSTs). The presence of the MYB-NFIB fusion gene was investigated in 13 AdCCs of the breast by fluorescence in situ hybridization (FISH) and reverse transcriptase-PCR (RT-PCR), and MYB and BRCA1 RNA expression was determined by quantitative RT-PCR. Fourteen AdCCs, 14 histological grade-matched IDC-NSTs, and 14 IDC-NSTs of triple-negative and basal-like phenotype were microdissected and subjected to high-resolution microarray-based comparative genomic hybridization (aCGH). The MYB-NFIB fusion gene was detected in all but one AdCC. aCGH analysis demonstrated a relatively low number of copy number aberrations and a lack of recurrent amplifications in breast AdCCs. Contrary to grade-matched IDC-NSTs, AdCCs lacked 1q gains and 16q losses, and in contrast with basal-like IDC-NSTs, AdCCs displayed fewer gene copy number aberrations and expressed MYB and BRCA1 at significantly higher levels. Breast AdCCs constitute an entity distinct from grade-matched and triple-negative and basal-like IDC-NSTs, emphasizing the importance of histological subtyping of triple-negative and basal-like breast carcinomas.
Collapse
Affiliation(s)
- Daniel Wetterskog
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Rajan N, Elliott R, Clewes O, Mackay A, Reis-Filho JS, Burn J, Langtry J, Sieber-Blum M, Lord CJ, Ashworth A. Dysregulated TRK signalling is a therapeutic target in CYLD defective tumours. Oncogene 2011; 30:4243-60. [PMID: 21552290 PMCID: PMC3175103 DOI: 10.1038/onc.2011.133] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 02/17/2011] [Accepted: 02/24/2011] [Indexed: 12/22/2022]
Abstract
Individuals with germline mutations in the tumour-suppressor gene CYLD are at high risk of developing disfiguring cutaneous appendageal tumours, the defining tumour being the highly organised cylindroma. Here, we analysed CYLD mutant tumour genomes by array comparative genomic hybridisation and gene expression microarray analysis. CYLD mutant tumours were characterised by an absence of copy-number aberrations apart from LOH chromosome 16q, the genomic location of the CYLD gene. Gene expression profiling of CYLD mutant tumours showed dysregulated tropomyosin kinase (TRK) signalling, with overexpression of TRKB and TRKC in tumours when compared with perilesional skin. Immunohistochemical analysis of a tumour microarray showed strong membranous TRKB and TRKC staining in cylindromas, as well as elevated levels of ERK phosphorylation and BCL2 expression. Membranous TRKC overexpression was also observed in 70% of sporadic BCCs. RNA interference-mediated silencing of TRKB and TRKC, as well as treatment with the small-molecule TRK inhibitor lestaurtinib, reduced colony formation and proliferation in 3D primary cell cultures established from CYLD mutant tumours. These results suggest that TRK inhibition could be used as a strategy to treat tumours with loss of functional CYLD.
Collapse
MESH Headings
- Adenoma, Sweat Gland/genetics
- Adenoma, Sweat Gland/metabolism
- Adenoma, Sweat Gland/pathology
- Carbazoles/pharmacology
- Carcinoma, Adenoid Cystic/genetics
- Carcinoma, Adenoid Cystic/metabolism
- Carcinoma, Adenoid Cystic/pathology
- Cluster Analysis
- Comparative Genomic Hybridization
- Deubiquitinating Enzyme CYLD
- Furans
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoblotting
- Immunohistochemistry
- Mutation
- Neoplasms/genetics
- Neoplasms/metabolism
- Neoplasms/pathology
- Neoplasms, Basal Cell/genetics
- Neoplasms, Basal Cell/metabolism
- Neoplasms, Basal Cell/pathology
- Oligonucleotide Array Sequence Analysis
- Primary Cell Culture
- Protein Kinases/genetics
- Protein Kinases/metabolism
- RNA Interference
- Receptor, trkB/antagonists & inhibitors
- Receptor, trkB/genetics
- Receptor, trkB/metabolism
- Receptor, trkC/antagonists & inhibitors
- Receptor, trkC/genetics
- Receptor, trkC/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/genetics
- Sweat Gland Neoplasms/genetics
- Sweat Gland Neoplasms/metabolism
- Sweat Gland Neoplasms/pathology
- Tissue Array Analysis
- Tumor Cells, Cultured
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- N Rajan
- Institute of Human Genetics, University of Newcastle upon Tyne, Newcastle upon Tyne, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
STUDY DESIGN Comparative genomic hybridization (CGH) microarrays. OBJECTIVE To identify genomic copy number variations (CNVs) in degenerative lumbar scoliosis (DLS) patients, and investigate the possibility of genetic predisposition in DLS. SUMMARY OF BACKGROUND DATA Genome scanning technology enables search for presence of CNVs. CGH microarray is a useful procedure in a genome-wide study. METHODS Among 45 consecutive patients who were diagnosed as DLS, 15 patients who manifested greatest Cobb's angle were selected for the array-CGH based CNV analysis. Control group was blood samples from 58 individuals without DLS. Oligonucleotide CGH microarray was utilized to analyze the CNV. Gene searches were performed for CNV DNA with significant gene-dosage difference. Validation qualitative PCR(qPCR) was performed at 3 genetic loci: at chromosome 2--TMEM163 gene, at chromosome 16--ANKRD 11 gene, and at chromosome 18--NFATC1 gene. RESULTS Genomic gains and losses were observed using the oligonucleotide CGH microarray. Identified CNVs were 446 ± 129 per individual. Gain- and loss-CNVs were identified as 196 ± 24 and 250 ± 110, respectively. The length of total CNV per individual was 30,946,730 ± 31,658,175 bp, and mean CNV-length was 61,017 ± 40,620 (median length 6411 ± 1994). Comparison with control group revealed 260 CNVs, which were significant (P < 10(-3)). Validation qPCR for gene-dosage comparison of DLS group DNA versus control group DNA in TMEM163 (P < 0.001); ANKRD 11 (P = 0.000); and NFATC1 (P = 0.000) gene showed significant difference. CONCLUSION Various whole-genome CNVs specific to DLS patients were observed. Validation qPCR confirmed significantly different gene-dosages for TMEM163, ANKRD 11, and NFATC1 genes. We consider that the expression of DLS is supported by various typical CNV-associated structural variants of the genome.
Collapse
|
45
|
Wilkerson PM, Dedes KJ, Wetterskog D, Mackay A, Lambros MB, Mansour M, Frankum J, Lord CJ, Natrajan R, Ashworth A, Reis-Filho JS. Functional characterization of EMSY gene amplification in human cancers. J Pathol 2011; 225:29-42. [PMID: 21735447 DOI: 10.1002/path.2944] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 05/18/2011] [Accepted: 05/18/2011] [Indexed: 11/10/2022]
Abstract
The 11q13-q14 locus is frequently amplified in human cancers, with a complex structure harbouring multiple potential oncogenic drivers. The EMSY gene has been proposed as a driver of the third core of the 11q13-q14 amplicon. This gene encodes a protein reported to be a BRCA2-binding partner, which when over-expressed would lead to impairment of BRCA2 functions and could constitute a mechanism for BRCA2 inactivation in non-hereditary breast and ovarian cancers. We hypothesized that if EMSY amplification abrogates BRCA2 functions, cells harbouring this aberration would be unable to elicit competent homologous recombination DNA repair and, therefore, may have increased sensitivity to genotoxic therapies and potent PARP inhibitors. Microarray-based comparative genomic hybridization of cell lines from distinct tumour sites, including breast, ovary, pancreas, oesophagus, lung and the oral cavity, led to the identification of 10 cell lines with EMSY amplification and 18 without. EMSY amplification was shown to correlate with EMSY mRNA levels, although not all cell lines harbouring EMSY amplification displayed EMSY mRNA or protein over-expression. RNA interference-mediated silencing of EMSY did not lead to a reduction in cell viability in tumour models harbouring EMSY amplification. Cell lines with and without EMSY amplification displayed a similar ability to elicit RAD51 foci in response to DNA damaging agents, and similar sensitivity to cisplatin and olaparib. Taken together, this suggests that EMSY is unlikely to be a driver of the 11q13-q14 amplicon and does not have a dominant role in modulating the response to agents targeting cells with defective homologous recombination.
Collapse
Affiliation(s)
- Paul M Wilkerson
- Molecular Pathology Team, Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Guan M, Liu L, Zhao X, Wu Q, Yu B, Shao Y, Yang H, Fu X, Wan J, Zhang W. Copy number variations of EphA3 are associated with multiple types of hematologic malignancies. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2011; 11:50-3. [PMID: 21454190 DOI: 10.3816/clml.2011.n.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND EphA3 is a component of the Eph receptor family, the largest subgroup of the receptor tyrosine kinase (RTK) family. A recent array-based study implicated the presence of copy-number variations (CNVs) of EphA3 in the genomes of acute myelogenous leukemia. CNVs are present in the general population at varying degrees, and have been found to associate with various types of diseases including hematologic malignancies. However, most of the current studies focused on the genome-wide screening of CNVs, and the functional impact of such regions needs to be extensively investigated in large number of clinical samples. PATIENTS AND METHODS In our study, we collected 617 bone marrow samples from multiple types of hematologic malignancies as well as healthy controls. DNA copy numbers and mRNA levels of EphA3 in these samples were examined. RESULTS We found significant association between the CNVs of EphA3 and these hematologic malignancies including acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), multiple myeloma (MM), and myelodysplastic syndrome (MDS). We also observed a positive correlation between the relative mRNA level and gene dosage of EphA3. CONCLUSION The CNVs of EphA3 were associated with multiple types of hematologic malignancies including ALL, AML, CLL, CML, MM, and MDS.
Collapse
Affiliation(s)
- Ming Guan
- Department of Clinical Laboratory, Huashan Hospital, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
van der Veken LT, Buijs A. Array CGH in human leukemia: from somatics to genetics. Cytogenet Genome Res 2011; 135:260-70. [PMID: 21893961 DOI: 10.1159/000330629] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
During the past decade, array CGH has been applied to study copy number alterations in the genome in human leukemia in relation to prediction of prognosis or responsiveness to therapy. In the first segment of this review, we will focus on the identification of acquired mutations by array CGH, followed by studies on the pathogenesis of leukemia associated with germline genetic variants, phenotypic presentation and response to treatment. In the last section, we will discuss constitutional genomic aberrations causally related to myeloid leukemogenesis.
Collapse
Affiliation(s)
- L T van der Veken
- Section of Genome Diagnostics, Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
48
|
Kaderi MA, Kanduri M, Buhl AM, Sevov M, Cahill N, Gunnarsson R, Jansson M, Smedby KE, Hjalgrim H, Jurlander J, Juliusson G, Mansouri L, Rosenquist R. LPL is the strongest prognostic factor in a comparative analysis of RNA-based markers in early chronic lymphocytic leukemia. Haematologica 2011; 96:1153-60. [PMID: 21508119 PMCID: PMC3148909 DOI: 10.3324/haematol.2010.039396] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 03/25/2011] [Accepted: 04/20/2011] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The expression levels of LPL, ZAP70, TCL1A, CLLU1 and MCL1 have recently been proposed as prognostic factors in chronic lymphocytic leukemia. However, few studies have systematically compared these different RNA-based markers. DESIGN AND METHODS Using real-time quantitative PCR, we measured the mRNA expression levels of these genes in unsorted samples from 252 newly diagnosed chronic lymphocytic leukemia patients and correlated our data with established prognostic markers (for example Binet stage, CD38, IGHV gene mutational status and genomic aberrations) and clinical outcome. RESULTS High expression levels of all RNA-based markers, except MCL1, predicted shorter overall survival and time to treatment, with LPL being the most significant. In multivariate analysis including the RNA-based markers, LPL expression was the only independent prognostic marker for overall survival and time to treatment. When studying LPL expression and the established markers, LPL expression retained its independent prognostic strength for overall survival. All of the RNA-based markers, albeit with varying ability, added prognostic information to established markers, with LPL expression giving the most significant results. Notably, high LPL expression predicted a worse outcome in good-prognosis subgroups, such as patients with mutated IGHV genes, Binet stage A, CD38 negativity or favorable cytogenetics. In particular, the combination of LPL expression and CD38 could further stratify Binet stage A patients. CONCLUSIONS LPL expression is the strongest RNA-based prognostic marker in chronic lymphocytic leukemia that could potentially be applied to predict outcome in the clinical setting, particularly in the large group of patients with favorable prognosis.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Female
- Humans
- Immunoglobulin Heavy Chains/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Lipoprotein Lipase/genetics
- Lipoprotein Lipase/metabolism
- Male
- Middle Aged
- Multivariate Analysis
- Mutation/genetics
- Prognosis
- RNA, Messenger/metabolism
- Survival Analysis
- Treatment Outcome
Collapse
Affiliation(s)
- Mohd Arifin Kaderi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Basic Health Sciences, KAHS IIUM Kuantan Campus, Pahang, Malaysia
| | - Meena Kanduri
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anne Mette Buhl
- Department of Hematology, Leukemia Laboratory, Rigshospitalet, Copenhagen, Denmark
| | - Marie Sevov
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Nicola Cahill
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rebeqa Gunnarsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Laboratory Medicine, Stem Cell Center, Hematology and Transplantation, Lund University, Lund, Sweden
| | - Mattias Jansson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Karin Ekström Smedby
- Department of Medicine, Clinical Epidemiology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Hjalgrim
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Jesper Jurlander
- Department of Hematology, Leukemia Laboratory, Rigshospitalet, Copenhagen, Denmark
| | - Gunnar Juliusson
- Department of Laboratory Medicine, Stem Cell Center, Hematology and Transplantation, Lund University, Lund, Sweden
| | - Larry Mansouri
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Richard Rosenquist
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
49
|
Hungermann D, Schmidt H, Natrajan R, Tidow N, Poos K, Reis-Filho JS, Brandt B, Buerger H, Korsching E. Influence of whole arm loss of chromosome 16q on gene expression patterns in oestrogen receptor-positive, invasive breast cancer. J Pathol 2011; 224:517-28. [PMID: 21706489 DOI: 10.1002/path.2938] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 04/15/2011] [Accepted: 05/09/2011] [Indexed: 01/05/2023]
Abstract
A whole chromosome arm loss of 16q belongs to the most frequent and earliest chromosomal alterations in invasive and in situ breast cancers of all common subtypes. Besides E-cadherin, several putative tumour suppressor genes residing on 16q in breast cancer have been investigated. However, the significance of these findings has remained unclear. Thus, other mechanisms leading to gene loss of function (eg haploinsufficiency, or distortion of multiple regulative subnetworks) remain to be tested as a hypothesis. To define the effect on gene expression of whole-arm loss of chromosome 16q in invasive breast cancer, we performed global gene expression analysis on a series of 18 genetically extensively characterized invasive ductal breast carcinomas and verified the results by quantitative real-time PCR (qRT-PCR). The distribution of the differential genes across the genome and their expression status was studied. A second approach by qRT-PCR in an independent series of 30 breast carcinomas helped to narrow down the observed effect. Whole-arm chromosome 16q losses, irrespective of other chromosomal changes, are associated with decreased expression of a number of candidate genes located on 16q (eg CDA08, CGI-128, SNTB2, NQO1, SF3B3, KIAA0174, ATBF1, GABARAPL2, KARS, GCSH, MBTPS1 and ZDHHC7) in breast carcinomas with a low degree of genetic instability. qRT-PCR provided evidence to suggest that the expression of these genes was reduced in a gene dosage-dependent manner. The differential expression of the candidate genes according to the chromosomal 16q-status vanished in genetically advanced breast cancer cases and changed ER status. These results corroborate previous reports about the importance of whole-arm loss of chromosome 16q in breast carcinogenesis and give evidence for the first time that haploinsufficiency, in the sense of a gene dosage effect, might be an important contributing factor in the early steps of breast carcinogenesis.
Collapse
MESH Headings
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Chromosome Deletion
- Chromosomes, Human, Pair 16/genetics
- Comparative Genomic Hybridization/methods
- Female
- Gene Dosage
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic
- Humans
- Neoplasm Invasiveness
- Neoplasm Proteins/metabolism
- Oligonucleotide Array Sequence Analysis/methods
- Receptors, Estrogen/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
Collapse
|
50
|
Wan J, Gao Y, Zhao X, Wu Q, Fu X, Shao Y, Yang H, Guan M, Yu B, Zhang W. The association between the copy-number variations of ZMAT4 and hematological malignancy. ACTA ACUST UNITED AC 2011; 16:20-3. [PMID: 21269563 DOI: 10.1179/102453311x12902908411751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Copy-number variations (CNVs) have been found in association with various types of diseases, including hematological malignancies. A recent array-based study implicated the presence of CNVs of ZMAT4 in the genome of acute myelogenous leukemia. In our study, we collected 617 bone marrow samples from multitypes of hematological malignancies as well as healthy controls. We found significant association between the CNVs of ZMAT4 and these hematological malignancies, including acute lymphoblastic leukemia, acute myelogenous leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, multiple myeloma, and myelodysplastic syndrome. We also examined the expression of ZMAT4 mRNA in the samples with 1 or 2 copies of DNA, and observed a weak yet positive correlation between the relative expression level and gene dosage. In conclusion, the CNVs of ZMAT4 have the potential to serve as a diagnostic indicator, alone or in combination with other markers, for hematological malignancies.
Collapse
Affiliation(s)
- Jun Wan
- Biomedical Research Institute, Shenzhen-PKU-HKUST Medical Center, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|