101
|
De Mattos-Arruda L, Weigelt B, Cortes J, Won HH, Ng CKY, Nuciforo P, Bidard FC, Aura C, Saura C, Peg V, Piscuoglio S, Oliveira M, Smolders Y, Patel P, Norton L, Tabernero J, Berger MF, Seoane J, Reis-Filho JS. Capturing intra-tumor genetic heterogeneity by de novo mutation profiling of circulating cell-free tumor DNA: a proof-of-principle. Ann Oncol 2014; 25:1729-1735. [PMID: 25009010 PMCID: PMC6276937 DOI: 10.1093/annonc/mdu239] [Citation(s) in RCA: 277] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 06/20/2014] [Accepted: 06/27/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Plasma-derived cell-free tumor DNA (ctDNA) constitutes a potential surrogate for tumor DNA obtained from tissue biopsies. We posit that massively parallel sequencing (MPS) analysis of ctDNA may help define the repertoire of mutations in breast cancer and monitor tumor somatic alterations during the course of targeted therapy. PATIENT AND METHODS A 66-year-old patient presented with synchronous estrogen receptor-positive/HER2-negative, highly proliferative, grade 2, mixed invasive ductal-lobular carcinoma with bone and liver metastases at diagnosis. DNA extracted from archival tumor material, plasma and peripheral blood leukocytes was subjected to targeted MPS using a platform comprising 300 cancer genes known to harbor actionable mutations. Multiple plasma samples were collected during the fourth line of treatment with an AKT inhibitor. RESULTS Average read depths of 287x were obtained from the archival primary tumor, 139x from the liver metastasis and between 200x and 900x from ctDNA samples. Sixteen somatic non-synonymous mutations were detected in the liver metastasis, of which 9 (CDKN2A, AKT1, TP53, JAK3, TSC1, NF1, CDH1, MML3 and CTNNB1) were also detected in >5% of the alleles found in the primary tumor sample. Not all mutations identified in the metastasis were reliably identified in the primary tumor (e.g. FLT4). Analysis of ctDNA, nevertheless, captured all mutations present in the primary tumor and/or liver metastasis. In the longitudinal monitoring of the patient, the mutant allele fractions identified in ctDNA samples varied over time and mirrored the pharmacodynamic response to the targeted therapy as assessed by positron emission tomography-computed tomography. CONCLUSIONS This proof-of-principle study is one of the first to demonstrate that high-depth targeted MPS of plasma-derived ctDNA constitutes a potential tool for de novo mutation identification and monitoring of somatic genetic alterations during the course of targeted therapy, and may be employed to overcome the challenges posed by intra-tumor genetic heterogeneity. REGISTERED CLINICAL TRIAL www.clinicaltrials.gov, NCT01090960.
Collapse
Affiliation(s)
- L De Mattos-Arruda
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona; Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - B Weigelt
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - J Cortes
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona
| | - H H Won
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - C K Y Ng
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - P Nuciforo
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona
| | - F-C Bidard
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA; Department of Medical Oncology, Institut Curie, Paris, France
| | - C Aura
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona
| | - C Saura
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona
| | - V Peg
- Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - S Piscuoglio
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - M Oliveira
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona
| | - Y Smolders
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - P Patel
- Genentech, Inc., San Francisco
| | - L Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - J Tabernero
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona; Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M F Berger
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - J Seoane
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona; Universitat Autònoma de Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - J S Reis-Filho
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA.
| |
Collapse
|
102
|
Kim TM, Jung SH, Kim MS, Baek IP, Park SW, Lee SH, Lee HH, Kim SS, Chung YJ, Lee SH. The mutational burdens and evolutionary ages of early gastric cancers are comparable to those of advanced gastric cancers. J Pathol 2014; 234:365-74. [PMID: 25042771 DOI: 10.1002/path.4401] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 06/22/2014] [Accepted: 06/27/2014] [Indexed: 12/15/2022]
Abstract
Early gastric cancers (EGCs) precede advanced gastric cancers (AGCs), with a favourable prognosis compared to AGC. To understand the progression mechanism of EGC to AGC, it is required to disclose EGC and AGC genomes in mutational and evolutionary perspectives. We performed whole-exome sequencing and copy number profiling of nine microsatellite (MS)-unstable (MSI-H) (five EGCs and four AGCs) and eight MS-stable (MSS) gastric cancers (four EGCs and four AGCs). In the cancers, we observed well-known driver mutations (TP53, APC, PIK3CA, ARID1A, and KRAS) that were enriched in cancer-related pathways, including chromatin remodelling and tyrosine kinase activity. The MSI-H genomes harboured ten times more mutations, but were largely depleted of copy number alterations (CNAs) compared to the MSS cancers. Interestingly, EGC genomes showed a comparable level of mutations to AGC in terms of the number, sequence composition, and functional consequences (potential driver mutations and affected pathways) of mutations. Furthermore, the CNAs between EGC and AGC genomes were not significantly different in either MSI-H and MSS. Evolutionary analyses using somatic mutations and MSI as molecular clocks further identified that EGC genomes were as old as AGC genomes in both MSS and MSI-H cancers. Our results suggest that the genetic makeup for gastric cancer may already be achieved in EGC genomes and that the time required for transition to AGC may be relatively short. Also, the data suggest a possibility that the mutational profiles obtained from early biopsies may be useful in the clinical settings for the molecular diagnosis and therapeutics of gastric cancer patients.
Collapse
Affiliation(s)
- Tae-Min Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Piscuoglio S, Ng CKY, Martelotto LG, Eberle CA, Cowell CF, Natrajan R, Bidard FC, De Mattos-Arruda L, Wilkerson PM, Mariani O, Vincent-Salomon A, Weigelt B, Reis-Filho JS. Integrative genomic and transcriptomic characterization of papillary carcinomas of the breast. Mol Oncol 2014; 8:1588-602. [PMID: 25041824 PMCID: PMC5037246 DOI: 10.1016/j.molonc.2014.06.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 06/08/2014] [Accepted: 06/17/2014] [Indexed: 02/06/2023] Open
Abstract
Papillary carcinoma (PC) is a rare type of breast cancer, which comprises three histologic subtypes: encapsulated PC (EPC), solid PC (SPC) and invasive PC (IPC). Microarray‐based gene expression and Affymetrix SNP 6.0 gene copy number profiling, and RNA‐sequencing revealed that PCs are luminal breast cancers that display transcriptomic profiles distinct from those of grade‐ and estrogen receptor (ER)‐matched invasive ductal carcinomas of no special type (IDC‐NSTs), and that the papillary histologic pattern is unlikely to be underpinned by a highly recurrent expressed fusion gene or a highly recurrent expressed mutation. Despite displaying similar patterns of gene copy number alterations, significant differences in the transcriptomic profiles of EPCs, SPCs and IPCs were found, and may account for their different histologic features. Papillary carcinomas of the breast display distinctive transcriptomic profiles. Proliferation‐related genes are expressed at low levels in papillary carcinomas. Papillary carcinomas are unlikely to be underpinned by a highly recurrent fusion gene. Papillary carcinomas are unlikely to be underpinned by a highly recurrent expressed mutation.
Collapse
Affiliation(s)
- Salvatore Piscuoglio
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York 10065, NY, USA
| | - Charlotte K Y Ng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York 10065, NY, USA
| | - Luciano G Martelotto
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York 10065, NY, USA
| | - Carey A Eberle
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York 10065, NY, USA
| | - Catherine F Cowell
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York 10065, NY, USA
| | - Rachael Natrajan
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | - François-Clement Bidard
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York 10065, NY, USA; Institut Curie, Department of Biopathology and INSERM U934, Paris, France
| | - Leticia De Mattos-Arruda
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York 10065, NY, USA
| | - Paul M Wilkerson
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | - Odette Mariani
- Institut Curie, Department of Biopathology and INSERM U934, Paris, France
| | | | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York 10065, NY, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York 10065, NY, USA.
| |
Collapse
|
104
|
Narod SA, Rakovitch E. A comparison of the risks of in-breast recurrence after a diagnosis of dcis or early invasive breast cancer. ACTA ACUST UNITED AC 2014; 21:119-24. [PMID: 24940092 DOI: 10.3747/co.21.1892] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND It is controversial whether ductal carcinoma in situ (dcis) is a preinvasive marker of breast cancer or if it is part of a spectrum of small cancers with malignant potential. Comparing clinical outcomes in women with invasive and noninvasive breast lesions might help to resolve the issue. METHODS From a database of 2641 patients with breast cancer, we selected women who had been treated with breast-conserving surgery for a cancer that was 2.0 cm or less in size, node-negative, and nonpalpable. No subject received chemotherapy. Cancers were categorized as noninvasive (stage 0, n = 172) or invasive (stage 1, n = 401) based on a review of the pathology records. We compared the actuarial risks of in-breast recurrence after invasive and noninvasive breast lesions before and after adjusting for tamoxifen and radiotherapy. RESULTS The 18-year cumulative risk of in-breast recurrence was 35.2% for patients with dcis and 12.8% for patients with small invasive cancers (hazard ratio: 2.4; 95% confidence interval: 1.5 to 3.8; p < 0.0003). After adjustment for radiotherapy and tamoxifen treatment, the difference was small and nonsignificant (hazard ratio: 1.4; 95% confidence interval: 0.9 to 2.4; p = 0.22). CONCLUSIONS For women with small, nonpalpable, node-negative breast cancers, the likelihood of experiencing an in-breast recurrence was associated with radiotherapy and with tamoxifen, but not with the presence of cancer cells invading beyond the basement membrane.
Collapse
Affiliation(s)
- S A Narod
- Women's College Research Institute, Women's College Hospital, and the Dalla Lana School of Public Health University of Toronto, Toronto, ON
| | - E Rakovitch
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON
| |
Collapse
|
105
|
Li CF, Liu CX, Li BC, Shen YY, Cui XB, Liu W, Dong HC, Pang LJ, Liang WH, Li F. Recurrent inflammatory myofibroblastic tumors harboring PIK3CA and KIT mutations. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:3673-3683. [PMID: 25120743 PMCID: PMC4128978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 06/27/2014] [Indexed: 06/03/2023]
Abstract
Inflammatory myofibroblastic tumour (IMT) is a relatively rare soft tissue malignancy. It exhibits locally aggressive behavior with a tendency for local recurrence and rare metastasis, and rare recurrent IMTs may show histological progression. The genetic hallmark of IMT is ALK rearrangement from chromosome arm 2p, but gene mutations involved in IMT remain poorly understood. The aim of the present study was to perform a pairwise comparison of the gene mutations occurring in primary and recurrent IMT from the same patient. We conducted a high-throughput analysis of 238 known mutations of 19 oncogenes in pairwise comparison primary and recurrent samples from 2 patients of IMT using Sequenom MassARRAY technology. Our results revealed 2 mutations in 2 recurrent lesion samples, including one in exon 11 of the KIT gene, resulting in a T-C substitution at position 1727 (L576P), the recurrent sample underwent histologic progression with "pleomorphic undifferentiated sarcoma-like" transformation; the other mutation was in exon 19 of the phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA) gene, resulting in a G-A substitution at position 1624 (E542K). Moreover, no any mutation was found in the primary lesion samples from 2 patients. Our findings suggest that variable genome changes might be present in IMT, especially during the progression from a primary tumour to recurrence. To the best of our knowledge, no such longitudinal study of IMT has been undertaken previously.
Collapse
Affiliation(s)
- Cheng-Fang Li
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
| | - Chun-Xia Liu
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
| | - Bing-Cheng Li
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
| | - Yao-Yuan Shen
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
| | - Xiao-Bin Cui
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
| | - Wei Liu
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
| | - Hong-Chao Dong
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
| | - Li-Juan Pang
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
| | - Wei-Hua Liang
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
| | - Feng Li
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of MedicineShihezi, Xinjiang 832002, China
| |
Collapse
|
106
|
Jiang YZ, Yu KD, Bao J, Peng WT, Shao ZM. Favorable Prognostic Impact in Loss of TP53 and PIK3CA Mutations after Neoadjuvant Chemotherapy in Breast Cancer. Cancer Res 2014; 74:3399-407. [DOI: 10.1158/0008-5472.can-14-0092] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
107
|
Norum JH, Andersen K, Sørlie T. Lessons learned from the intrinsic subtypes of breast cancer in the quest for precision therapy. Br J Surg 2014; 101:925-38. [PMID: 24849143 DOI: 10.1002/bjs.9562] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 04/16/2014] [Indexed: 01/06/2023]
Abstract
BACKGROUND Wide variability in breast cancer, between patients and within each individual neoplasm, adds confounding complexity to the treatment of the disease. In clinical practice, hormone receptor status has been used to classify breast tumours and to guide treatment. Modern classification systems should take the wide tumour heterogeneity into account to improve patient outcome. METHODS This article reviews the identification of the intrinsic molecular subtypes of breast cancer, their prognostic and therapeutic implications, and the impact of tumour heterogeneity on cancer progression and treatment. The possibility of functionally addressing tumour-specific characteristics in in vivo models to inform decisions for precision therapies is also discussed. RESULTS Despite the robust breast tumour classification system provided by gene expression profiling, heterogeneity is also evident within these molecular portraits. A complicating factor in breast cancer classification is the process of selective clonality within developing neoplasms. Phenotypically and functionally distinct clones representing the intratumour heterogeneity might confuse molecular classification. Molecular portraits of the heterogeneous primary tumour might not necessarily reflect the subclone of cancer cells that causes the disease to relapse. Studies of reciprocal relationships between cancer cell subpopulations within developing tumours are therefore needed, and are possible only in genetically engineered mouse models or patient-derived xenograft models, in which the treatment-induced selection pressure on individual cell clones can be mimicked. CONCLUSION In the future, more refined classifications, based on integration of information at several molecular levels, are required to improve treatment guidelines. Large-scale translational research efforts paved the way for identification of the intrinsic subtypes, and are still fundamental for ensuring future progress in cancer care.
Collapse
Affiliation(s)
- J H Norum
- Department of Genetics, Institute of Cancer Research, Oslo, Norway; Cancer Stem Cell Innovation Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | | | | |
Collapse
|
108
|
Abstract
In recent years it has become clear that cancer cells within a single tumor can display striking morphological, genetic and behavioral variability. Burgeoning genetic, epigenetic and phenomenological data support the existence of intra-tumor genetic heterogeneity in breast cancers; however, its basis is yet to be fully defined. Two of the most widely evoked concepts to explain the origin of heterogeneity within tumors are the cancer stem cell hypothesis and the clonal evolution model. Although the cancer stem cell model appeared to provide an explanation for the variability among the neoplastic cells within a given cancer, advances in massively parallel sequencing have provided several lines of evidence to suggest that intra-tumor genetic heterogeneity likely plays a fundamental role in the phenotypic heterogeneity observed in cancers. Many challenges remain, however, in the interpretation of the next generation sequencing results obtained so far. Here we review the models that explain tumor heterogeneity, the causes of intra-tumor genetic diversity and their impact on our understanding and management of breast cancer, methods to study intra-tumor heterogeneity and the assessment of intra-tumor genetic heterogeneity in the clinic.
Collapse
|
109
|
Sakr RA, Weigelt B, Chandarlapaty S, Andrade VP, Guerini-Rocco E, Giri D, Ng CKY, Cowell CF, Rosen N, Reis-Filho JS, King TA. PI3K pathway activation in high-grade ductal carcinoma in situ--implications for progression to invasive breast carcinoma. Clin Cancer Res 2014; 20:2326-37. [PMID: 24634376 PMCID: PMC4015460 DOI: 10.1158/1078-0432.ccr-13-2267] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE To assess the prevalence of phosphoinositide 3-kinase (PI3K) pathway alterations in pure high-grade ductal carcinoma in situ (DCIS) and DCIS associated with invasive breast cancer (IBC), and to determine whether DCIS and adjacent IBCs harbor distinct PI3K pathway aberrations. EXPERIMENTAL DESIGN Eighty-nine cases of pure high-grade DCIS and 119 cases of high-grade DCIS associated with IBC were characterized according to estrogen receptor (ER) and HER2 status, subjected to immunohistochemical analysis of PTEN, INPP4B, phosphorylated (p)AKT and pS6 expression, and to microdissection followed by Sequenom genotyping of PIK3CA and AKT1 hotspot mutations. RESULTS Alterations affecting the PI3K pathway were found in a subset of pure DCIS and DCIS adjacent to IBC. A subtype-matched comparison of pure DCIS and DCIS adjacent to IBC revealed that PIK3CA hotspot mutations and pAKT expression were significantly more prevalent in ER-positive/HER2-negative DCIS adjacent to IBC (P values, 0.005 and 0.043, respectively), and that in ER-negative/HER2-positive cases INPP4B loss of expression was more frequently observed in pure DCIS (a P value of 0.013). No differences in the parameters analyzed were observed in a pairwise comparison of the in situ and invasive components of cases of DCIS and adjacent IBC. Analysis of the PIK3CA-mutant allelic frequencies in DCIS and synchronous IBC revealed cases in which PIK3CA mutations were either restricted to the DCIS or to the invasive components. CONCLUSION Molecular aberrations affecting the PI3K pathway may play a role in the progression from high-grade DCIS to IBC in a subset of cases (e.g., a subgroup of ER-positive/HER2-negative lesions).
Collapse
Affiliation(s)
- Rita A. Sakr
- Breast Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Sarat Chandarlapaty
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Victor P. Andrade
- Breast Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Elena Guerini-Rocco
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Dilip Giri
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Charlotte K. Y. Ng
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Catherine F. Cowell
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Neal Rosen
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Jorge S. Reis-Filho
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Tari A. King
- Breast Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
110
|
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
|
111
|
De Mattos-Arruda L, Bidard FC, Won HH, Cortes J, Ng CKY, Peg V, Nuciforo P, Jungbluth AA, Weigelt B, Berger MF, Seoane J, Reis-Filho JS. Establishing the origin of metastatic deposits in the setting of multiple primary malignancies: the role of massively parallel sequencing. Mol Oncol 2014; 8:150-8. [PMID: 24220311 PMCID: PMC5528499 DOI: 10.1016/j.molonc.2013.10.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/08/2013] [Accepted: 10/11/2013] [Indexed: 01/03/2023] Open
Abstract
In this proof-of-principle study, we sought to define whether targeted capture massively parallel sequencing can be employed to determine the origin of metastatic deposits in cases of synchronous primary malignancies and metastases in distinct anatomical sites. DNA samples extracted from synchronous tumor masses in the breast, adnexal, and pelvic-peritoneal regions from a 62-year-old BRCA1 germline mutation carrier were subjected to targeted massively parallel sequencing using a platform comprising 300 cancer genes known to harbor actionable mutations. In addition to BRCA1 germline mutations, all lesions harbored somatic loss of the BRCA1 wild-type allele and TP53 somatic mutations. The primary breast cancer displayed a TP53 frameshift (p.Q317fs) mutation, whereas and the adnexal lesion harbored a TP53 nonsense (p.R213*) mutation, consistent with a diagnosis of two independent primary tumors (i.e. breast and ovarian cancer). The adnexal tumor and all pelvic-peritoneal implants harbored identical TP53 (p.R213*) and NCOA2 (p.G952R) somatic mutations. Evidence of genetic heterogeneity within and between lesions was observed, both in terms of somatic mutations and copy number aberrations. The repertoires of somatic genetic aberrations found in the breast, ovarian, and pelvic-peritoneal lesions provided direct evidence in support of the distinct origin of the breast and ovarian cancers, and established that the pelvic-peritoneal implants were clonally related to the ovarian lesion. These observations were consistent with those obtained with immunohistochemical analyses employing markers to differentiate between carcinomas of the breast and ovary, including WT1 and PAX8. Our results on this case of a patient with BRCA1-mutant breast and ovarian cancer demonstrate that massively parallel sequencing may constitute a useful tool to define the relationship, clonality and intra-tumor genetic heterogeneity between primary tumor masses and their metastatic deposits in patients with multiple primary malignancies and synchronous metastases.
Collapse
Affiliation(s)
- Leticia De Mattos-Arruda
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francois-Clement Bidard
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Department of Medical Oncology, Institut Curie, Paris, France
| | - Helen H Won
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Javier Cortes
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; Medica Scientia Innovation Research (MedSIR), Barcelona, Spain
| | - Charlotte K Y Ng
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Vicente Peg
- Universitat Autònoma de Barcelona, Barcelona, Spain; Pathology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Paolo Nuciforo
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Achim A Jungbluth
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Michael F Berger
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Joan Seoane
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
112
|
Carraro DM, Elias EV, Andrade VP. Ductal carcinoma in situ of the breast: morphological and molecular features implicated in progression. Biosci Rep 2014; 34:e00090. [PMID: 27919043 PMCID: PMC3894794 DOI: 10.1042/bsr20130077] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 10/29/2013] [Accepted: 11/20/2013] [Indexed: 12/21/2022] Open
Abstract
The spread of mammographic screening programmes around the world, including in developing countries, has substantially contributed to the diagnosis of small non-palpable lesions, which has increased the detection rate of DCIS (ductal carcinoma in situ). DCIS is heterogeneous in several ways, such as its clinical presentation, morphology and genomic profile. Excellent outcomes have been reported; however, many questions remain unanswered. For example, which patients groups are overtreated and could instead benefit from minimal intervention and which patient groups require a more traditional multidisciplinary approach. The development of a comprehensive integrated analysis that includes the radiological, morphological and genetic aspects of DCIS is necessary to answer these questions. This review focuses on discussing the significant findings about the morphological and molecular features of DCIS and its progression that have helped to uncover the biological and genetic heterogeneity of this disease. The knowledge gained in recent years might allow the development of tailored clinical management for women with DCIS in the future.
Collapse
Affiliation(s)
- Dirce M Carraro
- Laboratory of Genomics and Molecular Biology, International Center of Research, A.C. Camargo Cancer Center, São Paulo, SP 01509-900, Brazil
- National Institute of Science and Technology in Oncogenomics (INCITO), São Paulo, SP 01509-900, Brazil
| | - Eliana V Elias
- Laboratory of Genomics and Molecular Biology, International Center of Research, A.C. Camargo Cancer Center, São Paulo, SP 01509-900, Brazil
| | - Victor P Andrade
- Department of Anatomical Pathology, A.C. Camargo Cancer Center, São Paulo, S.P. 01509-900, Brazil
| |
Collapse
|
113
|
Almendro V, Cheng YK, Randles A, Itzkovitz S, Marusyk A, Ametller E, Gonzalez-Farre X, Muñoz M, Russnes HG, Helland A, Rye IH, Borresen-Dale AL, Maruyama R, van Oudenaarden A, Dowsett M, Jones RL, Reis-Filho J, Gascon P, Gönen M, Michor F, Polyak K. Inference of tumor evolution during chemotherapy by computational modeling and in situ analysis of genetic and phenotypic cellular diversity. Cell Rep 2014; 6:514-27. [PMID: 24462293 DOI: 10.1016/j.celrep.2013.12.041] [Citation(s) in RCA: 210] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 11/14/2013] [Accepted: 12/30/2013] [Indexed: 01/10/2023] Open
Abstract
Cancer therapy exerts a strong selection pressure that shapes tumor evolution, yet our knowledge of how tumors change during treatment is limited. Here, we report the analysis of cellular heterogeneity for genetic and phenotypic features and their spatial distribution in breast tumors pre- and post-neoadjuvant chemotherapy. We found that intratumor genetic diversity was tumor-subtype specific, and it did not change during treatment in tumors with partial or no response. However, lower pretreatment genetic diversity was significantly associated with pathologic complete response. In contrast, phenotypic diversity was different between pre- and posttreatment samples. We also observed significant changes in the spatial distribution of cells with distinct genetic and phenotypic features. We used these experimental data to develop a stochastic computational model to infer tumor growth patterns and evolutionary dynamics. Our results highlight the importance of integrated analysis of genotypes and phenotypes of single cells in intact tissues to predict tumor evolution.
Collapse
Affiliation(s)
- Vanessa Almendro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain
| | - Yu-Kang Cheng
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA
| | - Amanda Randles
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA; Center for Applied Scientific Computing, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Shalev Itzkovitz
- Departments of Physics and Biology and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Andriy Marusyk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Elisabet Ametller
- Department of Medical Oncology, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain
| | - Xavier Gonzalez-Farre
- Department of Medical Oncology, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain
| | - Montse Muñoz
- Department of Medical Oncology, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain
| | - Hege G Russnes
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo 0424, Norway; K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo 0316, Norway; Department of Pathology, Oslo University Hospital, Oslo 0424, Norway
| | - Aslaug Helland
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo 0424, Norway; Department of Oncology, Oslo University Hospital, Oslo 0424, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo 0316, Norway
| | - Inga H Rye
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo 0424, Norway; K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo 0316, Norway
| | - Anne-Lise Borresen-Dale
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo 0424, Norway; K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo 0316, Norway
| | - Reo Maruyama
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Alexander van Oudenaarden
- Departments of Physics and Biology and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, the Netherlands
| | - Mitchell Dowsett
- The Royal Marsden Hospital, The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London SW3 6JJ, UK
| | - Robin L Jones
- The Royal Marsden Hospital, The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London SW3 6JJ, UK; Seattle Cancer Care Alliance, Seattle, WA 98109-1023, USA
| | - Jorge Reis-Filho
- The Royal Marsden Hospital, The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London SW3 6JJ, UK; Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Pere Gascon
- Department of Medical Oncology, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Franziska Michor
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA.
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA.
| |
Collapse
|
114
|
Tennstedt P, Bölch C, Strobel G, Minner S, Burkhardt L, Grob T, Masser S, Sauter G, Schlomm T, Simon R. Patterns of TPD52 overexpression in multiple human solid tumor types analyzed by quantitative PCR. Int J Oncol 2013; 44:609-15. [PMID: 24317684 DOI: 10.3892/ijo.2013.2200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 10/29/2013] [Indexed: 11/06/2022] Open
Abstract
Tumor protein D52 (TPD52) is located at chromosome 8q21, a region that is frequently gained or amplified in multiple human cancer types. TPD52 has been suggested as a potential target for new anticancer therapies. In order to analyze TPD52 expression in the most prevalent human cancer types, we employed quantitative PCR to measure TPD52 mRNA levels in formalin-fixed tissue samples from more than 900 cancer tissues obtained from 29 different human cancer types. TPD52 was expressed at varying levels in all tested normal tissues, including skin, lymph node, lung, oral mucosa, breast, endometrium, ovary, vulva, myometrium, liver, pancreas, stomach, kidney, prostate, testis, urinary bladder, thyroid gland, brain, muscle and fat tissue. TPD52 was upregulated in 18/29 (62%) tested cancer types. Strongest expression was found in non-seminoma (56-fold overexpression compared to corresponding normal tissue), seminoma (42-fold), ductal (28-fold) and lobular breast cancer (14-fold). In these tumor types, TPD52 upregulation was found in the vast majority (>80%) of tested samples. Downregulation was found in 11 (38%) tumor types, most strongly in papillary renal cell cancer (-8-fold), leiomyosarcoma (-6-fold), clear cell renal cell cancer (-5-fold), liposarcoma (-5-fold) and lung cancer (-4-fold). These results demonstrate that TPD52 is frequently and strongly upregulated in many human cancer types, which may represent candidate tumor types for potential anti-TPD52 therapies.
Collapse
Affiliation(s)
- Pierre Tennstedt
- Martini-Clinic, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Charlotte Bölch
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gundula Strobel
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lia Burkhardt
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Grob
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sawinee Masser
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schlomm
- Martini-Clinic, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
115
|
Hanker AB, Cook RS, Arteaga CL. Mouse models and anti-HER2 therapies. Oncotarget 2013; 4:1866-7. [PMID: 24272818 PMCID: PMC3875754 DOI: 10.18632/oncotarget.1481] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/13/2013] [Indexed: 11/25/2022] Open
Affiliation(s)
- Ariella B Hanker
- Departments of Medicine and Cancer Biology, Breast Cancer Research Program, Vanderbilt- Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | | | | |
Collapse
|
116
|
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
|
117
|
Cowell CF, Weigelt B, Sakr RA, Ng CKY, Hicks J, King TA, Reis-Filho JS. Progression from ductal carcinoma in situ to invasive breast cancer: revisited. Mol Oncol 2013; 7:859-69. [PMID: 23890733 DOI: 10.1016/j.molonc.2013.07.005] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 07/04/2013] [Indexed: 12/21/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is an intraductal neoplastic proliferation of epithelial cells that is separated from the breast stroma by an intact layer of basement membrane and myoepithelial cells. DCIS is a non-obligate precursor of invasive breast cancer, and up to 40% of these lesions progress to invasive disease if untreated. Currently, it is not possible to predict accurately which DCIS would be more likely to progress to invasive breast cancer as neither the significant drivers of the invasive transition have been identified, nor has the clinical utility of tests predicting the likelihood of progression been demonstrated. Although molecular studies have shown that qualitatively, synchronous DCIS and invasive breast cancers are remarkably similar, there is burgeoning evidence to demonstrate that intra-tumor genetic heterogeneity is observed in a subset of DCIS, and that the process of progression to invasive disease may constitute an 'evolutionary bottleneck', resulting in the selection of subsets of tumor cells with specific genetic and/or epigenetic aberrations. Here we review the clinical challenge posed by DCIS, the contribution of the microenvironment and genetic aberrations to the progression from in situ to invasive breast cancer, the emerging evidence of the impact of intra-tumor genetic heterogeneity on this process, and strategies to combat this heterogeneity.
Collapse
Affiliation(s)
- Catherine F Cowell
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | | | | | | | | | | | | |
Collapse
|
118
|
Ng CKY, Pemberton HN, Reis-Filho JS. Breast cancer intratumor genetic heterogeneity: causes and implications. Expert Rev Anticancer Ther 2013; 12:1021-32. [PMID: 23030222 DOI: 10.1586/era.12.85] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There is burgeoning evidence to suggest that tumor evolution follows the laws of Darwinian evolution, whereby individual tumor cell clones harbor private genetic aberrations in addition to the founder mutations, and that these distinct populations of cancer cells interact in competitive and mutualistic manners. The combined effect of genetic and epigenetic instability, and differential selective pressures according to the microenvironment and therapeutic interventions, create many different evolutionary routes such that intratumor heterogeneity is inevitable. Numerous cytogenetic, comparative genomic hybridization and, more recently, massively parallel sequencing studies have generated indisputable evidence of this phenomenon. The impact of intratumor heterogeneity on response and resistance to therapy is beginning to be understood; this information may prove crucial for the potentials of personalized medicine to be realized. In this review, the evidence of intratumor heterogeneity in breast cancer, its potential causes and implications for the clinical management of breast cancer patients are discussed.
Collapse
Affiliation(s)
- Charlotte K Y Ng
- Molecular Pathology Team, Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, SW3 6JB, UK
| | | | | |
Collapse
|
119
|
Saieg MA, Geddie WR, Boerner SL, Bailey D, Crump M, da Cunha Santos G. EZH2 and CD79B mutational status over time in B-cell non-Hodgkin lymphomas detected by high-throughput sequencing using minimal samples. Cancer Cytopathol 2013; 121:377-86. [PMID: 23361872 PMCID: PMC3799012 DOI: 10.1002/cncy.21262] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/18/2012] [Accepted: 10/24/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND: Numerous genomic abnormalities in B-cell non-Hodgkin lymphomas (NHLs) have been revealed by novel high-throughput technologies, including recurrent mutations in EZH2 (enhancer of zeste homolog 2) and CD79B (B cell antigen receptor complex-associated protein beta chain) genes. This study sought to determine the evolution of the mutational status of EZH2 and CD79B over time in different samples from the same patient in a cohort of B-cell NHLs, through use of a customized multiplex mutation assay. METHODS: DNA that was extracted from cytological material stored on FTA cards as well as from additional specimens, including archived frozen and formalin-fixed histological specimens, archived stained smears, and cytospin preparations, were submitted to a multiplex mutation assay specifically designed for the detection of point mutations involving EZH2 and CD79B, using MassARRAY spectrometry followed by Sanger sequencing. RESULTS: All 121 samples from 80 B-cell NHL cases were successfully analyzed. Mutations in EZH2 (Y646) and CD79B (Y196) were detected in 13.2% and 8% of the samples, respectively, almost exclusively in follicular lymphomas and diffuse large B-cell lymphomas. In one-third of the positive cases, a wild type was detected in a different sample from the same patient during follow-up. CONCLUSIONS: Testing multiple minimal tissue samples using a high-throughput multiplex platform exponentially increases tissue availability for molecular analysis and might facilitate future studies of tumor progression and the related molecular events. Mutational status of EZH2 and CD79B may vary in B-cell NHL samples over time and support the concept that individualized therapy should be based on molecular findings at the time of treatment, rather than on results obtained from previous specimens. Cancer (Cancer Cytopathol) 2013;121:377–386. © 2013 American Cancer Society.
Collapse
Affiliation(s)
- Mauro Ajaj Saieg
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
120
|
Dhodapkar MV. Personalized immune-interception of cancer and the battle of two adaptive systems--when is the time right? Cancer Prev Res (Phila) 2013; 6:173-6. [PMID: 23341571 DOI: 10.1158/1940-6207.capr-13-0011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A growing body of evidence points to a coevolutionary model of cancer, wherein the cross-talk between tumor cells (or their subclones) and the host determine the malignant potential of individual tumors. Most of this natural history is clinically invisible and includes preneoplastic states. The capacity of the immune system to recognize these incipient lesions provides the basis for targeting them immunologically to arrest the development of preneoplasia toward clinical cancer. Kimura and colleagues provide evidence of immunogenicity of a potential cancer vaccine in patients with a history of advanced colon adenomas. These studies provide proof-of-principle or feasibility of such an approach in the clinic. Here, we discuss emerging opportunities and challenges in harnessing the immune system to "intercept" the precursor or preneoplastic lesions. Both cancer cells as well as the immune system represent independent and complex systems with plasticity and adaptive potential. It is therefore likely that specific aspects of the cross-talk between tumor cells and host may differ between individual tumors and determine the evolution of both tumors and the host response. We try to make the case to consider individualized approaches based on the genetic make-up of tumor cells and properties of the host response. Such strategies may be needed to optimally position the immune system to prevent cancers.
Collapse
Affiliation(s)
- Madhav V Dhodapkar
- Yale University School of Medicine, 333 Cedar Street, Box 208028, New Haven, CT 06510, USA.
| |
Collapse
|
121
|
Ruark E, Snape K, Humburg P, Loveday C, Bajrami I, Brough R, Rodrigues DN, Renwick A, Seal S, Ramsay E, Duarte SDV, Rivas MA, Warren-Perry M, Zachariou A, Campion-Flora A, Hanks S, Murray A, Pour NA, Douglas J, Gregory L, Rimmer A, Walker NM, Yang TP, Adlard JW, Barwell J, Berg J, Brady AF, Brewer C, Brice G, Chapman C, Cook J, Davidson R, Donaldson A, Douglas F, Eccles D, Evans DG, Greenhalgh L, Henderson A, Izatt L, Kumar A, Lalloo F, Miedzybrodzka Z, Morrison PJ, Paterson J, Porteous M, Rogers MT, Shanley S, Walker L, Gore M, Houlston R, Brown MA, Caufield MJ, Deloukas P, McCarthy MI, Todd JA, The Breast and Ovarian Cancer Susceptibility Collaboration (BOCS), Wellcome Trust Case Control Consortium, Turnbull C, Reis-Filho JS, Ashworth A, Antoniou AC, Lord CJ, Donnelly P, Rahman N. Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer. Nature 2013; 493:406-10. [PMID: 23242139 PMCID: PMC3759028 DOI: 10.1038/nature11725] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 10/26/2012] [Indexed: 02/06/2023]
Abstract
Improved sequencing technologies offer unprecedented opportunities for investigating the role of rare genetic variation in common disease. However, there are considerable challenges with respect to study design, data analysis and replication. Using pooled next-generation sequencing of 507 genes implicated in the repair of DNA in 1,150 samples, an analytical strategy focused on protein-truncating variants (PTVs) and a large-scale sequencing case-control replication experiment in 13,642 individuals, here we show that rare PTVs in the p53-inducible protein phosphatase PPM1D are associated with predisposition to breast cancer and ovarian cancer. PPM1D PTV mutations were present in 25 out of 7,781 cases versus 1 out of 5,861 controls (P = 1.12 × 10(-5)), including 18 mutations in 6,912 individuals with breast cancer (P = 2.42 × 10(-4)) and 12 mutations in 1,121 individuals with ovarian cancer (P = 3.10 × 10(-9)). Notably, all of the identified PPM1D PTVs were mosaic in lymphocyte DNA and clustered within a 370-base-pair region in the final exon of the gene, carboxy-terminal to the phosphatase catalytic domain. Functional studies demonstrate that the mutations result in enhanced suppression of p53 in response to ionizing radiation exposure, suggesting that the mutant alleles encode hyperactive PPM1D isoforms. Thus, although the mutations cause premature protein truncation, they do not result in the simple loss-of-function effect typically associated with this class of variant, but instead probably have a gain-of-function effect. Our results have implications for the detection and management of breast and ovarian cancer risk. More generally, these data provide new insights into the role of rare and of mosaic genetic variants in common conditions, and the use of sequencing in their identification.
Collapse
Affiliation(s)
- Elise Ruark
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Katie Snape
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Peter Humburg
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Chey Loveday
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Ilirjana Bajrami
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Rachel Brough
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
- Cancer Research UK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Daniel Nava Rodrigues
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Anthony Renwick
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Sheila Seal
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Emma Ramsay
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | | | - Manuel A. Rivas
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7LD, UK
| | - Margaret Warren-Perry
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Anna Zachariou
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Adriana Campion-Flora
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Sandra Hanks
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Anne Murray
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Naser Ansari Pour
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Jenny Douglas
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Lorna Gregory
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Andrew Rimmer
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Neil M. Walker
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY, UK
| | - Tsun-Po Yang
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Julian W. Adlard
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds, LS7 4SA, UK
| | - Julian Barwell
- Leicestershire Genetics Centre, University Hospitals of Leicester NHS Trust, LE1 5WW, UK
| | - Jonathan Berg
- Human genetics, Division of Medical Sciences, University of Dundee, DD1 9SY, UK
| | - Angela F. Brady
- NW Thames Regional Genetics Service, Kennedy Galton Centre, London, HA1 3UJ, UK
| | - Carole Brewer
- Peninsula Regional Genetics Service, Royal Devon & Exeter Hospital, Exeter, EX1 2ED, UK
| | - Glen Brice
- SW Thames Regional Genetics Service, St George’s Hospital, London, SW17 0RE, UK
| | - Cyril Chapman
- West Midlands Regional Genetics Service, Birmingham Women’s Hospital, Birmingham, B15 2TG, UK
| | - Jackie Cook
- Sheffield Regional Genetics Service, Sheffield Children’s NHS Foundation Trust, S10 2TH, UK
| | - Rosemarie Davidson
- West of Scotland Regional Genetics Service, Laboratory Medicine, Southern General Hospital, Glasgow, G51 4TF, UK
| | - Alan Donaldson
- South Western Regional Genetics Service, University Hospitals of Bristol NHS Foundation Trust, BS2 8EG, UK
| | - Fiona Douglas
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, NE1 3BZ, UK
| | - Diana Eccles
- Faculty of Medicine, University of Southampton, Southampton University Hospitals NHS Trust, SO16 5YA, UK
| | - D. Gareth Evans
- Genetic Medicine, Manchester Academic Health Science Centre, St. Mary’s Hospital, Manchester M13 9WL, UK
| | - Lynn Greenhalgh
- Merseyside and Cheshire Clinical Genetics Service, Liverpool Women’s NHS Foundation Trust, Liverpool, L8 7SS, UK
| | - Alex Henderson
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, NE1 3BZ, UK
| | - Louise Izatt
- SE Thames Regional Genetics Service, Guy’s and St Thomas NHS Foundation Trust, London, SE1 9RT, UK
| | - Ajith Kumar
- NE Thames Regional Genetics Service, Great Ormond St Hospital, London, WC1N 3JH, UK
| | - Fiona Lalloo
- University Dept of Medical Genetics & Regional Genetics Service, St Mary’s Hospital, Manchester, M13 9WL, UK
| | - Zosia Miedzybrodzka
- University of Aberdeen and North of Scotland Clinical Genetics Service, Aberdeen Royal Infirmary, AB25 2ZA, UK
| | - Patrick J. Morrison
- Northern Ireland Regional Genetics Service, Belfast HSC Trust, Department of Medical Genetics, Queen’s University Belfast, BT9 7AB, UK
| | - Joan Paterson
- East Anglian Regional Genetics Service, Cambridge University Hospitals NHS Foundation Trust, CB2 0QQ, UK
| | - Mary Porteous
- South East of Scotland Clinical Genetics Service, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Mark T. Rogers
- All Wales Medical Genetics Service, University Hospital of Wales, Cardiff, CF14 4XW, UK
| | - Susan Shanley
- Dept of Cancer Genetics, Royal Marsden NHS Foundation Trust, Sutton, SM2 5PT, UK
| | - Lisa Walker
- Oxford Regional Genetics Service, Oxford University Hospitals NHS Trust, Oxford, OX3 7LJ, UK
| | - Martin Gore
- Dept of Gynaecologic Oncology, Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Richard Houlston
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Matthew A. Brown
- University of Queensland Diamantina Institute, University of Queensland, Princess Alexandra Hospital, Woolloongabba, Brisbane, 4102, Australia
| | - Mark J. Caufield
- Clinical Pharmacology and Barts and The London Genome Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Panagiotis Deloukas
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Mark I. McCarthy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LI, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, OX3 7LI, UK
| | - John A. Todd
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY, UK
| | | | | | - Clare Turnbull
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
- Dept of Cancer Genetics, Royal Marsden NHS Foundation Trust, Sutton, SM2 5PT, UK
| | - Jorge S. Reis-Filho
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Alan Ashworth
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Antonis C. Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Christopher J. Lord
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Peter Donnelly
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Department of Statistics, University of Oxford, Oxford, OX1 3TG, UK
| | - Nazneen Rahman
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
- Dept of Cancer Genetics, Royal Marsden NHS Foundation Trust, Sutton, SM2 5PT, UK
| |
Collapse
|
122
|
Greer LT, Rosman M, Mylander WC, Hooke J, Kovatich A, Sawyer K, Buras RR, Shriver CD, Tafra L. Does breast tumor heterogeneity necessitate further immunohistochemical staining on surgical specimens? J Am Coll Surg 2012; 216:239-51. [PMID: 23141136 DOI: 10.1016/j.jamcollsurg.2012.09.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Revised: 09/11/2012] [Accepted: 09/12/2012] [Indexed: 01/24/2023]
Abstract
BACKGROUND Prognostic and predictive tumor markers in breast cancer are most commonly performed on core needle biopsies (CNB) of the primary tumor. Because treatment recommendations are influenced by these markers, it is imperative to verify strong concordance between tumor markers on CNB specimens and the corresponding surgical specimens (SS). STUDY DESIGN A prospective study was performed on 165 women (205 samples) with breast cancer diagnosed from January 2009 to July 2011. Tumor type, grade, estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor 2 (HER2), and Ki67 expression by immunohistochemical (IHC) testing were retrospectively analyzed in the CNB and SS. Contingency tables and agreement modeling were performed. RESULTS There was substantial agreement between the CNB and SS for PR% and HER2; moderate agreement for tumor type, grade, and ER%; and fair agreement for Ki67%. In 8% of patients (n = 13), tumor heterogeneity was seen. In heterogeneous tumors the overall concordance between the CNB and SS was worse, especially for HER2. Six of these patients had areas of tumor that were positive for HER2, which were not detected in their CNBs. Nine patients had multiple distinct molecular subtypes within their tumor(s). CONCLUSIONS The heterogeneous distribution of antigens in breast cancer tumors raises concern that the CNB may not adequately represent the true biologic profile in all patients. There is strong concordance for tumor type, ER, and PR between CNB and SS (although a quantitative decline was noted from CNB to SS); however, HER2 activity does not appear to be adequately detected on CNB in patients with heterogeneous tumors. These data suggest that IHC testing on the CNB alone may not be adequate to tailor targeted therapy in all patients.
Collapse
Affiliation(s)
- Lauren T Greer
- Clinical Breast Care Project, Walter Reed National Military Medical Center, Bethesda, MD 20889-5600, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Kurelac I, MacKay A, Lambros MBK, Di Cesare E, Cenacchi G, Ceccarelli C, Morra I, Melcarne A, Morandi L, Calabrese FM, Attimonelli M, Tallini G, Reis-Filho JS, Gasparre G. Somatic complex I disruptive mitochondrial DNA mutations are modifiers of tumorigenesis that correlate with low genomic instability in pituitary adenomas. Hum Mol Genet 2012; 22:226-38. [PMID: 23049073 DOI: 10.1093/hmg/dds422] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mitochondrial DNA (mtDNA) mutations leading to the disruption of respiratory complex I (CI) have been shown to exhibit anti-tumorigenic effects, at variance with those impairing only the function but not the assembly of the complex, which appear to contribute positively to cancer development. Owing to the challenges in the analysis of the multi-copy mitochondrial genome, it is yet to be determined whether tumour-associated mtDNA lesions occur as somatic modifying factors or as germ-line predisposing elements. Here we investigated the whole mitochondrial genome sequence of 20 pituitary adenomas with oncocytic phenotype and identified pathogenic and/or novel mtDNA mutations in 60% of the cases. Using highly sensitive techniques, namely fluorescent PCR and allele-specific locked nucleic acid quantitative PCR, we identified the most likely somatic nature of these mutations in our sample set, since none of the mutations was detected in the corresponding blood tissue of the patients analysed. Furthermore, we have subjected a series of 48 pituitary adenomas to a high-resolution array comparative genomic hybridization analysis, which revealed that CI disruptive mutations, and the oncocytic phenotype, significantly correlate with low number of chromosomal aberrations in the nuclear genome. We conclude that CI disruptive mutations in pituitary adenomas are somatic modifiers of tumorigenesis most likely contributing not only to the development of oncocytic change, but also to a less aggressive tumour phenotype, as indicated by a stable karyotype.
Collapse
Affiliation(s)
- Ivana Kurelac
- Dip. di Scienze Mediche e Chirurgiche, U.O. Genetica Medica, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Heselmeyer-Haddad K, Berroa Garcia LY, Bradley A, Ortiz-Melendez C, Lee WJ, Christensen R, Prindiville SA, Calzone KA, Soballe PW, Hu Y, Chowdhury SA, Schwartz R, Schäffer AA, Ried T. Single-cell genetic analysis of ductal carcinoma in situ and invasive breast cancer reveals enormous tumor heterogeneity yet conserved genomic imbalances and gain of MYC during progression. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1807-22. [PMID: 23062488 DOI: 10.1016/j.ajpath.2012.07.012] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 07/16/2012] [Accepted: 07/25/2012] [Indexed: 10/27/2022]
Abstract
Ductal carcinoma in situ (DCIS) is a precursor lesion of invasive ductal carcinoma (IDC) of the breast. To understand the dynamics of genomic alterations in this progression, we used four multicolor fluorescence in situ hybridization probe panels consisting of the oncogenes COX2, MYC, HER2, CCND1, and ZNF217 and the tumor suppressor genes DBC2, CDH1, and TP53 to visualize copy number changes in 13 cases of synchronous DCIS and IDC based on single-cell analyses. The DCIS had a lower degree of chromosomal instability than the IDC. Despite enormous intercellular heterogeneity in DCIS and IDC, we observed signal patterns consistent with a nonrandom distribution of genomic imbalances. CDH1 was most commonly lost, and gain of MYC emerged during progression from DCIS to IDC. Four of 13 DCISs showed identical clonal imbalances in the IDCs. Six cases revealed a switch, and in four of those, the IDC had acquired a gain of MYC. In one case, the major clone in the IDC was one of several clones in the DCIS, and in another case, the major clone in the DCIS became one of the two major clones in the IDC. Despite considerable chromosomal instability, in most cases the evolution from DCIS to IDC is determined by recurrent patterns of genomic imbalances, consistent with a biological continuum.
Collapse
Affiliation(s)
- Kerstin Heselmeyer-Haddad
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Weigelt B, Reis-Filho J, Swanton C. Genomic analyses to select patients for adjuvant chemotherapy: trials and tribulations. Ann Oncol 2012; 23 Suppl 10:x211-8. [DOI: 10.1093/annonc/mds323] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
126
|
Jang M, Kim E, Choi Y, Lee H, Kim Y, Kim J, Kang E, Kim SW, Kim I, Park S. FGFR1 is amplified during the progression of in situ to invasive breast carcinoma. Breast Cancer Res 2012; 14:R115. [PMID: 22863309 PMCID: PMC3680930 DOI: 10.1186/bcr3239] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 08/03/2012] [Indexed: 12/17/2022] Open
Abstract
Introduction Gene amplification is an important mechanism for activating oncogenes in malignant tumors. Although amplification of HER2, C-MYC, CCND1 and FGFR1 has been reported in breast cancers, their role in the progression of in situ to invasive breast carcinoma is unclear. To investigate this question we compared the amplification frequencies of these genes in pure ductal carcinoma in situ (DCIS), DCIS associated with invasive carcinoma, and invasive carcinoma. Methods We performed fluorescence in situ hybridization of the selected genes on tissue microarrays composed of 179 pure DCIS and 438 invasive carcinomas. Two hundred and sixteen of the latter had DCIS components, and in those cases we compared gene amplification in the intraductal and invasive components of each carcinoma. Results The rate of amplification of FGFR1 was higher in invasive carcinomas than in the pure DCIS, but the opposite was true for HER2 amplification. These findings applied consistently to high-grade tumors, but not to low/intermediate-grade tumors. The amplification status of HER2, C-MYC, CCND1 and FGFR1 was generally similar in the matched invasive and DCIS components of the same tumors. However, FGFR1 amplification was more common in the invasive components than in the DCIS components. In survival analyses, FGFR1 amplification was found to be an independent prognostic factor for poor disease-free survival for all patients with invasive carcinoma and for the hormone receptor-positive subgroup. Conclusion Amplification of HER2, C-MYC and CCND1 seems to play a role in the early development of breast cancer, but not in its progression. However, the increased frequency of FGFR1 amplification in invasive carcinomas compared with pure DCIS and in the invasive components of individual tumors, and its association with decreased disease-free survival, suggests a role for FGFR1 amplification in the progression of breast cancer including in situ-to-invasive transition, as well as initiation.
Collapse
|