1
|
Haas JS, Phillips KA, Liang SY, Hassett MJ, Keohane C, Elkin EB, Armstrong J, Toscano M. Genomic testing and therapies for breast cancer in clinical practice. J Oncol Pract 2011; 7:e1s-7s. [PMID: 21886507 PMCID: PMC3092459 DOI: 10.1200/jop.2011.000299] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2011] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Given the likely proliferation of targeted testing and treatment strategies for cancer, a better understanding of the utilization patterns of human epidermal growth factor receptor 2 (HER2) testing and trastuzumab and newer gene expression profiling (GEP) for risk stratification and chemotherapy decision making are important. STUDY DESIGN Cross-sectional. METHODS We performed a medical record review of women age 35 to 65 years diagnosed between 2006 and 2007 with invasive localized breast cancer, identified using claims from a large national health plan (N = 775). RESULTS Almost all women received HER2 testing (96.9%), and 24.9% of women with an accepted indication received GEP. Unexplained socioeconomic differences in GEP use were apparent after adjusting for age and clinical characteristics; specifically, GEP use increased with income. For example, those in the lowest income category (< $40,000) were less likely than those with an income of $125,000 or more to receive GEP (odds ratio, 0.34; 95% CI, 0.16 to 0.73). A majority of women (57.7%) with HER2-positive disease received trastuzumab; among these women, differences in age and clinical characteristics were not apparent, although surprisingly, those in the lowest income category were more likely than those in the high-income category to receive trastuzumab (P = .02). Among women who did not have a positive HER2 test, 3.9% still received trastuzumab. Receipt of adjuvant chemotherapy increased as GEP score indicated greater risk of recurrence. CONCLUSION Identifying and eliminating unnecessary variation in the use of these expensive tests and treatments should be part of quality improvement and efficiency programs.
Collapse
Affiliation(s)
- Jennifer S. Haas
- Brigham and Women's Hospital; Dana-Farber Cancer Institute, Boston, MA; University of California, San Francisco, San Francisco, CA; Memorial Sloan-Kettering Cancer Center, New York, NY; and Aetna, Hartford, CT
| | - Kathryn A. Phillips
- Brigham and Women's Hospital; Dana-Farber Cancer Institute, Boston, MA; University of California, San Francisco, San Francisco, CA; Memorial Sloan-Kettering Cancer Center, New York, NY; and Aetna, Hartford, CT
| | - Su-Ying Liang
- Brigham and Women's Hospital; Dana-Farber Cancer Institute, Boston, MA; University of California, San Francisco, San Francisco, CA; Memorial Sloan-Kettering Cancer Center, New York, NY; and Aetna, Hartford, CT
| | - Michael J. Hassett
- Brigham and Women's Hospital; Dana-Farber Cancer Institute, Boston, MA; University of California, San Francisco, San Francisco, CA; Memorial Sloan-Kettering Cancer Center, New York, NY; and Aetna, Hartford, CT
| | - Carol Keohane
- Brigham and Women's Hospital; Dana-Farber Cancer Institute, Boston, MA; University of California, San Francisco, San Francisco, CA; Memorial Sloan-Kettering Cancer Center, New York, NY; and Aetna, Hartford, CT
| | - Elena B. Elkin
- Brigham and Women's Hospital; Dana-Farber Cancer Institute, Boston, MA; University of California, San Francisco, San Francisco, CA; Memorial Sloan-Kettering Cancer Center, New York, NY; and Aetna, Hartford, CT
| | - Joanne Armstrong
- Brigham and Women's Hospital; Dana-Farber Cancer Institute, Boston, MA; University of California, San Francisco, San Francisco, CA; Memorial Sloan-Kettering Cancer Center, New York, NY; and Aetna, Hartford, CT
| | - Michele Toscano
- Brigham and Women's Hospital; Dana-Farber Cancer Institute, Boston, MA; University of California, San Francisco, San Francisco, CA; Memorial Sloan-Kettering Cancer Center, New York, NY; and Aetna, Hartford, CT
| |
Collapse
|
2
|
Becker F, van El CG, Ibarreta D, Zika E, Hogarth S, Borry P, Cambon-Thomsen A, Cassiman JJ, Evers-Kiebooms G, Hodgson S, Janssens ACJW, Kaariainen H, Krawczak M, Kristoffersson U, Lubinski J, Patch C, Penchaszadeh VB, Read A, Rogowski W, Sequeiros J, Tranebjaerg L, van Langen IM, Wallace H, Zimmern R, Schmidtke J, Cornel MC. Genetic testing and common disorders in a public health framework: how to assess relevance and possibilities. Background Document to the ESHG recommendations on genetic testing and common disorders. Eur J Hum Genet 2011; 19 Suppl 1:S6-44. [PMID: 21412252 PMCID: PMC3327518 DOI: 10.1038/ejhg.2010.249] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Frauke Becker
- Hannover Medical School, Department of Human Genetics, Hannover, Germany
| | - Carla G van El
- Department of Clinical Genetics and EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Dolores Ibarreta
- IPTS Institute for Prospective Technological Studies, Joint Research Centre, European Commission, Seville, Spain
| | - Eleni Zika
- IPTS Institute for Prospective Technological Studies, Joint Research Centre, European Commission, Seville, Spain
| | - Stuart Hogarth
- Department of Social Sciences, Loughborough University, Loughborough, UK
| | - Pascal Borry
- Department of Clinical Genetics and EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
- Centre for Biomedical Ethics and Law, Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Medical Humanities and EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Anne Cambon-Thomsen
- Inserm, U 558, Department of Epidemiology, Health Economics and Public Health, University Paul Sabatier, Toulouse, France
| | | | - Gerry Evers-Kiebooms
- Psychosocial Genetics Unit University Hospitals, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Shirley Hodgson
- Department of Clinical Genetics, St George's University of London, London, UK
| | - A Cécile J W Janssens
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Michael Krawczak
- Institute of Medical Informatics and Statistics, Christian-Albrechts-Universität, Kiel, Germany
| | | | - Jan Lubinski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | | | | | - Andrew Read
- Division of Human Development, School of Clinical Sciences, University of Nottingham, Nottingham, UK
| | - Wolf Rogowski
- Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, Clinical Center, Ludwig Maximilians University, Munich, Germany
| | - Jorge Sequeiros
- IBMC – Institute for Molecular and Cell Biology, and ICBAS, University of Porto, Porto, Portugal
| | - Lisbeth Tranebjaerg
- Department of Audiology, H:S Bispebjerg Hospital and Wilhelm Johannsen Centre of Functional Genomics, University of Copenhagen, Copenhagen, Denmark
| | - Irene M van Langen
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Helen Wallace
- GeneWatch UK, The Mill House, Tideswell, Derbyshire, UK
| | - Ron Zimmern
- PHG Foundation, Worts Causeway, Cambridge, UK
| | - Jörg Schmidtke
- Hannover Medical School, Department of Human Genetics, Hannover, Germany
| | - Martina C Cornel
- Department of Clinical Genetics and EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Bartlett J, Campbell FM, Ibrahim M, O’Grady A, Kay E, Faulkes C, Collins N, Starczynski J, Morgan JM, Jasani B, Miller K. A UK NEQAS ISH multicenter ring study using the Ventana HER2 dual-color ISH assay. Am J Clin Pathol 2011; 135:157-62. [PMID: 21173138 DOI: 10.1309/ajcpvprkk1enedgq] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
We performed a multicenter assessment of a new HER2 dual-color chromogenic in situ hybridization (CISH) test and herein report on concordance of CISH data with fluorescence in situ hybridization (FISH) data and intraobserver and interlaboratory scoring consistency. HER2 results were evaluated using duplicate cores from 30 breast cancers in 5 laboratories using the Ventana HER2 dual-color ISH assay (Ventana Medical Systems, Cambridgeshire, England) and in 1 central laboratory using a standard FISH assay. Overall 93.3% of cases were successfully analyzed by CISH across the 5 participating laboratories. There was excellent concordance (98.0% overall) for diagnosis of HER2 amplification by CISH compared with FISH. Intraobserver variability (7.7%) and intersite variability (9.1%) of absolute HER2/chromosome enumeration probe 17 ratios were tightly controlled across all participating laboratories. The Ventana HER2 dual-color ISH assay is robust and reproducible, shows good concordance with a standard FISH assay, and complies with requirements in national and international guidelines for performance of ISH-based diagnostic tests.
Collapse
|
4
|
Amoureux MC, Coulibaly B, Chinot O, Loundou A, Metellus P, Rougon G, Figarella-Branger D. Polysialic acid neural cell adhesion molecule (PSA-NCAM) is an adverse prognosis factor in glioblastoma, and regulates olig2 expression in glioma cell lines. BMC Cancer 2010; 10:91. [PMID: 20219118 PMCID: PMC2854115 DOI: 10.1186/1471-2407-10-91] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 03/10/2010] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most aggressive and frequent brain tumor, albeit without cure. Although patient survival is limited to one year on average, significant variability in outcome is observed. The assessment of biomarkers is needed to gain better knowledge of this type of tumor, help prognosis, design and evaluate therapies. The neurodevelopmental polysialic acid neural cell adhesion molecule (PSA-NCAM) protein is overexpressed in various cancers. Here, we studied its expression in GBM and evaluated its prognosis value for overall survival (OS) and disease free survival (DFS). METHODS We set up a specific and sensitive enzyme linked immunosorbent assay (ELISA) test for PSA-NCAM quantification, which correlated well with PSA-NCAM semi quantitative analysis by immunohistochemistry, and thus provides an accurate quantitative measurement of PSA-NCAM content for the 56 GBM biopsies analyzed. For statistics, the Spearman correlation coefficient was used to evaluate the consistency between the immunohistochemistry and ELISA data. Patients' survival was estimated by using the Kaplan-Meier method, and curves were compared using the log-rank test. On multivariate analysis, the effect of potential risk factors on the DFS and OS were evaluated using the cox regression proportional hazard models. The threshold for statistical significance was p = 0.05. RESULTS We showed that PSA-NCAM was expressed by approximately two thirds of the GBM at variable levels. On univariate analysis, PSA-NCAM content was an adverse prognosis factor for both OS (p = 0.04) and DFS (p = 0.0017). On multivariate analysis, PSA-NCAM expression was an independent negative predictor of OS (p = 0.046) and DFS (p = 0.007). Furthermore, in glioma cell lines, PSA-NCAM level expression was correlated to the one of olig2, a transcription factor required for gliomagenesis. CONCLUSION PSA-NCAM represents a valuable biomarker for the prognosis of GBM patients.
Collapse
Affiliation(s)
- Marie-Claude Amoureux
- Université de la Méditerranée CNRS UMR6216, Institut de Biologie du Développement de Marseille Luminy, Marseille, France.
| | | | | | | | | | | | | |
Collapse
|
5
|
Bartlett JMS, Campbell FM, Ibrahim M, Thomas J, Wencyk P, Ellis I, Kay E, Connolly Y, O’Grady A, Barnett S, Starczynski J, Cunningham P, Miller K. A UK NEQAS ICC and ISH multicentre study using the Kreatech PoseidonHER2FISH probe: intersite variation can be rigorously controlled using FISH. Histopathology 2010; 56:297-304. [DOI: 10.1111/j.1365-2559.2010.03493.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
6
|
Pommier SJ, Quan GG, Christante D, Muller P, Newell AEH, Olson SB, Diggs B, Muldoon L, Neuwelt E, Pommier RF. Characterizing the HER2/neu status and metastatic potential of breast cancer stem/progenitor cells. Ann Surg Oncol 2009; 17:613-23. [PMID: 19838757 DOI: 10.1245/s10434-009-0730-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Treatment resistance, long latency, and high recurrence rates suggest that breast cancers arise from defective breast stem cells. HYPOTHESIS Within cancers, subpopulations of cells will demonstrate differences in stem/progenitor potential, HER2/neu amplification, and gene expression. Related cells will be found in normal breast tissue. METHODS ER-/PR-/HER2/neu + breast cancer cells were flow-sorted into subpopulations: (A) CD49f(+) CD24(-), (B) CD49f(+)CD24(+), (C) CD49f CD24(-), and (D) CD49f(-)CD24(+). Gel matrix cell invasion, fluorescence in situ hybridization (FISH) HER2/neu amplification, and qRT-PCR gene expression were measured in all groups. Cells from sorted groups were implanted into rat brains. Resultant tumors were analyzed by immunohistochemistry (IHC) and FISH. Normal breast tissue was examined by IHC. RESULTS Tumor development varied among sorted groups (25-75%), but was highest in group A. Tumor cells were mostly CD49f(-)CD24(-), with variable fractions of other stem/progenitor cells. Tumors showed HER2/neu amplification, but fewer chromosome 17 per cell than inoculates. Group A tumors exhibited cells with normal chromosome 17 copy number and near normal HER2/neu amplification. Cell invasion was 61% higher in unsorted cells and 34-42% in sorted groups compared with controls. Sorted groups showed significantly different expression of development, proliferation, and invasion associated genes. In normal breast tissue, CD49f(+) cells were identified in CD14(+) CK19(-) basal epithelial layers of mammary glands; these were 95% CD24(+) and 60% CD44(+). CONCLUSIONS Breast cancer stem/progenitor cell populations differ in tumor-initiating potential but are not solely responsible for metastasis. Cancer stem/progenitor cells are less polyploid than cancer cells in general and may not be HER2/neu amplified. In normal breast tissue, breast stem/progenitor cell-like populations are present.
Collapse
Affiliation(s)
- SuEllen J Pommier
- Division of Surgical Oncology, Department of General Surgery, Oregon Health & Science University, Portland, OR, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Bartlett J, Campbell FM, Ibrahim M, Wencyk P, Ellis I, Kay E, Connolly Y, O’Grady A, Di Palma S, Starczynski J, Morgan JM, Jasani B, Miller K. Chromogenic in situ hybridization: a multicenter study comparing silver in situ hybridization with FISH. Am J Clin Pathol 2009; 132:514-20. [PMID: 19762528 DOI: 10.1309/ajcpxy3mj6gsrcyp] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Our purposes were to perform a robust assessment of a new HER2 chromogenic in situ hybridization test and report on concordance of silver in situ hybridization (SISH) data with fluorescence in situ hybridization (FISH) data and on intraobserver and interlaboratory scoring consistency. HER2 results were scored from 45 breast cancers in 7 laboratories using the Ventana (Tucson, AZ) INFORM HER-2 SISH assay and in 1 central laboratory using a standard FISH assay. Overall, 94.8% of cases were successfully analyzed by SISH across the 6 participating laboratories that reported data. Concordance for diagnosis of HER2 amplification by SISH compared with FISH was high (96.0% overall). Intraobserver variability (8.0%) and intersite variability (12.66%) of absolute HER2/chromosome 17 ratios appear to be tightly controlled across all 6 participating laboratories. The Ventana INFORM HER-2 SISH assay is robust and reproducible, shows good concordance with a standard FISH assay, and complies with requirements in national guidelines for performance of diagnostic tests.
Collapse
Affiliation(s)
- J.M.S. Bartlett
- Endocrine Cancer Group, Edinburgh, Scotland
- UK National External Quality Assessment Scheme, University College London, London, England
| | | | - Merdol Ibrahim
- UK National External Quality Assessment Scheme, University College London, London, England
| | - Peter Wencyk
- Department of Histopathology, Nottingham City Hospital, Nottingham, England
| | - Ian Ellis
- Department of Histopathology, Nottingham City Hospital, Nottingham, England
| | - Elaine Kay
- Department of Histopathology, Beaumont Hospital, Dublin, Ireland
| | | | - Anthony O’Grady
- Department of Histopathology, Beaumont Hospital, Dublin, Ireland
| | - Silvana Di Palma
- Department of Histopathology, the RSCH, University of Surrey, Guildford, England
| | | | - John M. Morgan
- Department of Histopathology, Cardiff & Vale NHS Trust, Cardiff, Wales
| | - Bharat Jasani
- Department of Pathology, School of Medicine, Cardiff University, Cardiff, Wales
| | - Keith Miller
- UK National External Quality Assessment Scheme, University College London, London, England
| |
Collapse
|
8
|
Gustavson MD, Bourke-Martin B, Reilly D, Cregger M, Williams C, Mayotte J, Zerkowski M, Tedeschi G, Pinard R, Christiansen J. Standardization of HER2 immunohistochemistry in breast cancer by automated quantitative analysis. Arch Pathol Lab Med 2009; 133:1413-9. [PMID: 19722747 DOI: 10.5858/133.9.1413] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2008] [Indexed: 11/06/2022]
Abstract
CONTEXT There is critical need for standardization of HER2 immunohistochemistry testing in the clinical laboratory setting. Recently, the American Society of Clinical Oncology and the College of American Pathologists have submitted guidelines recommending that laboratories achieve 95% concordance between assays and observers for HER2 testing. OBJECTIVE As a potential aid to pathologists for achieving these new guidelines, we have conducted an examination using automated quantitative analysis (AQUA analysis) to provide a standardized HER2 immunohistochemistry expression score across instruments (sites), operators, and staining runs. DESIGN We analyzed HER2 expression by immunohistochemistry in a cohort (n = 669) of invasive breast cancers in tissue microarray format across different instruments (n = 3), operators (n = 3), and staining runs (n = 3). Using light source, instrument calibration techniques, and a new generation of image analysis software, we produced normalized AQUA scores for each parameter and examined their reproducibility. RESULTS The average percent coefficients of variation across instruments, operators, and staining runs were 1.8%, 2.0%, and 5.1%, respectively. For positive/negative classification between parameters, concordance rates ranged from 94.5% to 99.3% for all cases. Differentially classified cases only occurred around the determined cut point, not over the entire distribution. CONCLUSIONS These data demonstrate that AQUA analysis can provide a standardized HER2 immunohistochemistry test that can meet current guidelines by the American Society of Clinical Oncology/College of American Pathologists. The use of AQUA analysis could allow for standardized and objective HER2 testing in clinical laboratories.
Collapse
Affiliation(s)
- Mark D Gustavson
- Department of Development, HistoRx Inc, New Haven, CT 06511, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Bartlett JM, Ibrahim M, Jasani B, Morgan JM, Ellis I, Kay E, Connolly Y, Campbell F, O’Grady A, Barnett S, Miller K. External quality assurance of HER2 FISH and ISH testing: three years of the UK national external quality assurance scheme. Am J Clin Pathol 2009; 131:106-11. [PMID: 19095573 DOI: 10.1309/ajcpln78zqxemnma] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The American Society of Clinical Oncology/College of American Pathologists guidelines highlighted the critical importance of quality assurance in diagnostic testing for HER2. Unstained formalin-fixed, paraffin-embedded human breast carcinoma cell line sections were circulated to scheme participants on 9 occasions. "Reference laboratories" reported results for the HER2/chromosome 17 ratio and HER2 copy number for 3 years for each cell line, including 418 sets of results (1,671 results total). The number of participants was 62 laboratories in the final analysis. The mean and SD of results from reference laboratories demonstrated consistency during the 3-year period. The percentage of laboratories achieving "appropriate" results ranged from 45% to 88%, and the percentage achieving "inappropriate" results ranged from 5% to 29%. No consistent effect of the HER2 in situ hybridization testing method was demonstrated. Participation in external quality assurance schemes is a valuable mechanism for demonstrating and acquiring consistency for HER2 testing by in situ hybridization. Poor performance can be corrected via assistance and advice.
Collapse
Affiliation(s)
- John M.S. Bartlett
- Endocrine Cancer Group, Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh, Scotland
- UK National External Quality Assessment Service, Cardiff University, Cardiff, Wales
| | - Merdol Ibrahim
- UK National External Quality Assessment Service, Cardiff University, Cardiff, Wales
| | - Bharat Jasani
- Department of Pathology, School of Medicine, Cardiff University, Cardiff, Wales
| | - John M. Morgan
- Department of Pathology, School of Medicine, Cardiff University, Cardiff, Wales
| | - Ian Ellis
- University of Nottingham, Molecular Medical Sciences, Department of Histopathology, Nottingham City Hospital, Nottingham, England
| | - Elaine Kay
- Department of Histopathology, Beaumont Hospital and the Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, University College London, London, England
| | - Yvonne Connolly
- Adelaide and Meath Hospital, incorporating the National Children’s Hospital, Dublin
| | - Fiona Campbell
- Endocrine Cancer Group, Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh, Scotland
| | - Anthony O’Grady
- Department of Histopathology, Beaumont Hospital and the Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, University College London, London, England
| | - Sarah Barnett
- Advanced Diagnostics, University College London, London, England
| | - Keith Miller
- UK National External Quality Assessment Service, Cardiff University, Cardiff, Wales
| |
Collapse
|
10
|
Giltnane JM, Molinaro A, Cheng H, Robinson A, Turbin D, Gelmon K, Huntsman D, Rimm DL. Comparison of quantitative immunofluorescence with conventional methods for HER2/neu testing with respect to response to trastuzumab therapy in metastatic breast cancer. Arch Pathol Lab Med 2008; 132:1635-47. [PMID: 18834223 DOI: 10.5858/2008-132-1635-coqiwc] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2008] [Indexed: 11/06/2022]
Abstract
CONTEXT Selection for trastuzumab therapy depends on a companion diagnostic assessment of HER2 by either immunohistochemistry (IHC) for protein overexpression or fluorescence in situ hybridization (FISH) to detect gene amplification. Although many studies have compared IHC to FISH, few have compared the tests to the true gold standard, tumor response. OBJECTIVE To compare HER2 testing by FISH and IHC along with a third immunofluorescence-based assay (automated quantitative analysis-tissue microarray [AQUA-TMA]) and to assess the value of each test for prediction of response to trastuzumab. DESIGN Immunohistochemistry and FISH assays were done on both whole slides (IHC-WS and FISH-WS) and on TMAs (IHC-TMA and FISH-TMA). AQUA was only done on TMAs (AQUA-TMA). Response was assessed according to modified Response Evaluation Criteria in Solid Tumors. RESULTS AQUA-TMA scores showed a significant linear relationship to both the FISH signal ratio and IHC scores on whole sections and TMAs. Assay assessment by outcome showed no association between response and FISH-WS ratio (P = .96), FISH-TMA (P = .55), IHC-WS (P = .75), or IHC-TMA (P = .06), but a significant relationship between AQUA score and categoric response was observed (P = .01). Assessed as a function of outcome using models of logistic regression, both AQUA-TMA and IHC-TMA were equally significant (P = .01). FISH-WS was the most sensitive assay, with a significantly higher true-positive fraction than all other tests except AQUA-TMA, although it was the least specific. IHC-TMA was the most specific assay. The lowest misclassification rate was achieved using AQUA-TMA (0.30). CONCLUSIONS Both AQUA-TMA and IHC-TMA were substantially more predictive than the FISH or IHC-WS tests. Although these results are derived from a small retrospective series, they suggest that accurate measurement of protein expression and unbiased selection of tissue for measurement may be key factors in prediction of response.
Collapse
Affiliation(s)
- Jennifer M Giltnane
- Departments of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520-8023 , USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
The human genome project and related research initiatives have enabled the identification of a significant number of genetic variants that are predictive of drug response and outcome (pharmacogenomic biomarkers). As yet, incorporation of routine pharmacogenomic testing into clinical practice has been relatively modest. Potential barriers to adoption include a relative lack of prospective controlled trials establishing the benefits of such testing, economic constraints, and ethical concerns, among others. Clinicians considering the use of pharmacogenomic testing in their practice also may be unfamiliar with the concepts and principles underlying this rapidly evolving discipline. Consequently, the purpose of this review is to provide the clinical pharmacologist with a primer on the principles and molecular mechanisms underlying pharmacogenomics. In addition, the methods currently being used to discover novel pharmacogenomic biomarkers and then apply these to clinical practice will be described.
Collapse
Affiliation(s)
- Michael H Court
- Comparative and Molecular Pharmacogenomics Laboratory, Department of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
12
|
Dancey JE. Early Clinical Trial Design Issues: Patient Populations, End Points, and Barriers. Lung Cancer 2007. [DOI: 10.3109/9781420020359.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
13
|
Kummar S, Kinders R, Rubinstein L, Parchment RE, Murgo AJ, Collins J, Pickeral O, Low J, Steinberg SM, Gutierrez M, Yang S, Helman L, Wiltrout R, Tomaszewski JE, Doroshow JH. Compressing drug development timelines in oncology using phase '0' trials. Nat Rev Cancer 2007; 7:131-9. [PMID: 17251919 DOI: 10.1038/nrc2066] [Citation(s) in RCA: 170] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The optimal evaluation of molecularly targeted anticancer agents requires the integration of pharmacodynamic assays into early clinical investigations. Phase '0' trials conducted under the new Exploratory Investigational New Drug Guidance from the US Food and Drug Administration can provide a platform to establish the feasibility of assays for target modulation in human samples, evaluate biomarkers for drug effects and provide pharmacokinetic data. Phase 0 trials could facilitate rational drug selection, identify therapeutic failures early, and might compress timelines for anticancer drug development. We expect that such trials will become a routine part of early-phase oncological drug development in the future.
Collapse
Affiliation(s)
- Shivaani Kummar
- Center for Cancer Research, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Haab BB, Paulovich AG, Anderson NL, Clark AM, Downing GJ, Hermjakob H, Labaer J, Uhlen M. A Reagent Resource to Identify Proteins and Peptides of Interest for the Cancer Community. Mol Cell Proteomics 2006; 5:1996-2007. [PMID: 16867976 DOI: 10.1074/mcp.t600020-mcp200] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
On the basis of discussions with representatives from all sectors of the cancer research community, the National Cancer Institute (NCI) recognizes the immense opportunities to apply proteomics technologies to further cancer research. Validated and well characterized affinity capture reagents (e.g. antibodies, aptamers, and affibodies) will play a key role in proteomics research platforms for the prevention, early detection, treatment, and monitoring of cancer. To discuss ways to develop new resources and optimize current opportunities in this area, the NCI convened the "Proteomic Technologies Reagents Resource Workshop" in Chicago, IL on December 12-13, 2005. The workshop brought together leading scientists in proteomics research to discuss model systems for evaluating and delivering resources for reagents to support MS and affinity capture platforms. Speakers discussed issues and identified action items related to an overall vision for and proposed models for a shared proteomics reagents resource, applications of affinity capture methods in cancer research, quality control and validation of affinity capture reagents, considerations for target selection, and construction of a reagents database. The meeting also featured presentations and discussion from leading private sector investigators on state-of-the-art technologies and capabilities to meet the user community's needs. This workshop was developed as a component of the NCI's Clinical Proteomics Technologies Initiative for Cancer, a coordinated initiative that includes the establishment of reagent resources for the scientific community. This workshop report explores various approaches to develop a framework that will most effectively fulfill the needs of the NCI and the cancer research community.
Collapse
Affiliation(s)
- Brian B Haab
- Van Andel Research Institute, Grand Rapids, Michigan 49503, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Kozlowski S, Swann P. Current and future issues in the manufacturing and development of monoclonal antibodies. Adv Drug Deliv Rev 2006; 58:707-22. [PMID: 16828921 DOI: 10.1016/j.addr.2006.05.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2005] [Accepted: 05/06/2006] [Indexed: 11/20/2022]
Abstract
Despite a slow beginning, monoclonal antibodies have had many successes over the past decade. It is important that these successes continue, bringing more products for more indications to market. Although manufacturing is not the most common cause of product failure, product quality issues can delay antibody development. Manufacturing has depended on the triad of process validation, process control and product testing. Applying product knowledge proactively to manufacturing (quality by design) may allow greater flexibility and maintain or improve product quality. An integrated approach to biological characterization is an important aspect of product knowledge. Greater product knowledge also facilitates development in other disciplines. Independent of manufacturing strategy, there are a number of regulatory hurdles in initial and ongoing antibody development. These are described to help prevent unnecessary delays.
Collapse
Affiliation(s)
- Steven Kozlowski
- Office of Biotechnology Products, Office of Pharmaceutical Science, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, MD, USA.
| | | |
Collapse
|
16
|
Marrer E, Baty F, Kehren J, Chibout SD, Brutsche M. Past, present and future of gene expression-tailored therapy for lung cancer. Per Med 2006; 3:165-175. [DOI: 10.2217/17410541.3.2.165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
“Variability is the law of life, and as no two faces are the same, so no two bodies are alike, and no two individuals react alike and behave alike under the abnormal conditions which we know as disease.” Sir William Osler (1849–1919). All human beings are different and some of these differences are the variations in response to xenobiotics. Personalized medicine means: the right patient population, the right drug, the right dose, the right indication, and administration at the right time. This review provides an update on concepts of personalized therapy for lung cancer.
Collapse
Affiliation(s)
| | - Florent Baty
- University Hospital Basel, Pulmonary Gene Research, Basel, Switzerland
| | | | | | - Martin Brutsche
- University Hospital Basel, Pulmonary Gene Research, Basel, Switzerland
| |
Collapse
|
17
|
Abstract
Are new concepts in biomedical ethics required to keep pace with the developments in post-Human Genome Project (HGP) genomics? This paper traces the place of ethics in the post-HGP landscape. The need for a revision of the approach taken by biomedical ethics toward questions in genomics has been appreciated for years. Traditional biomedical ethics, led by the protection paradigm, was devised to serve a very different context. Today, compelling ethical questions arise from the tension between individual and collective interests in the context of population-based data collection and research. The collection of phenotype data, and the development of new sequencing technologies, raises burning questions that call for innovative tools and models in ethics. Future developments that will likely include the routine availability of personal genome information, and the advent of systems biology as a framework for interpretation, will require ongoing flexibility and a creative approach.
Collapse
Affiliation(s)
- Jeantine E Lunshof
- VU university medical center, Department of Clinical Genetics & Human Genetics, Section Community Genetics, EMGO Institute, Van der Boechorststraat 7, PO Box 7057, 1007 MB Amsterdam, The Netherlands.
| |
Collapse
|
18
|
Taube SE, Jacobson JW, Lively TG. Cancer diagnostics: decision criteria for marker utilization in the clinic. ACTA ACUST UNITED AC 2006; 5:357-64. [PMID: 16336001 DOI: 10.2165/00129785-200505060-00003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
A new diagnostic tool must pass three major tests before it is adopted for routine clinical use. First, the tool must be robust and reproducible; second, the clinical value of the tool must be proven, i.e. the tool should reliably trigger a clinical decision that results in patient benefit; and, third, the clinical community has to be convinced of the need for this tool and the benefits it affords. Another factor that can influence the adoption of new tools relates to the cost and the vagaries of insurance reimbursement. The Cancer Diagnosis Program (CDP) of the US National Cancer Institute (NCI) launched the Program for the Assessment of Clinical Cancer Tests (PACCT) in 2000 to develop a process for moving the results of new technologies and new understanding of cancer biology more efficiently and effectively into clinical practice. PACCT has developed an algorithm that incorporates the iterative nature of assay development into an evaluation process that includes developers and end users. The effective introduction of new tests into clinical practice has been hampered by a series of common problems that are best described using examples of successes and failures. The successful application of the PACCT algorithm is described in the discussion of the recent development of the OncotypeDX assay and plan for a prospective trial of this assay by the NCI-supported Clinical Trials Cooperative Groups. The assay uses reverse transcription (RT)-PCR evaluation of a set of 16 genes that were shown to strongly associate with the risk of recurrence of breast cancer in women who presented with early stage disease (hormone responsive, and no involvement of the auxiliary lymph nodes). The test is highly reproducible. It provides information to aid the physician and patient in making important clinical decisions, including the aggressiveness of the therapy that should be recommended. A trial is planned to test whether OncotypeDX can be used as a standalone trigger for specific treatment decisions. The problems that have been encountered and have delayed the development of other diagnostic tools are exemplified in the development of tests for human epidermal growth factor receptor (HER2) overexpression, for predictors of response to epidermal growth factor receptor inhibitors, and for the detection of residual disease following chemotherapy.
Collapse
MESH Headings
- Algorithms
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/standards
- Bone Marrow Examination
- Breast Neoplasms/diagnosis
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Carcinoma, Non-Small-Cell Lung/diagnosis
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Clinical Trials as Topic
- ErbB Receptors/analysis
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/genetics
- Female
- Gefitinib
- Genetic Testing
- Humans
- Immunohistochemistry/economics
- Immunohistochemistry/standards
- Lung Neoplasms/diagnosis
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lymphatic Metastasis
- Neoplasm, Residual
- Neoplasms/diagnosis
- Neoplastic Cells, Circulating
- Patient Selection
- Practice Guidelines as Topic
- Predictive Value of Tests
- Protein Kinase Inhibitors/therapeutic use
- Quinazolines/therapeutic use
- Receptor, ErbB-2/analysis
- Receptor, ErbB-2/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Sensitivity and Specificity
- Technology Assessment, Biomedical/standards
- Technology Assessment, Biomedical/trends
- Trastuzumab
Collapse
Affiliation(s)
- Sheila E Taube
- Cancer Diagnosis Program, Division of Cancer Treatment & Diagnosis, National Cancer Institute, Rockville, Maryland 20952, USA.
| | | | | |
Collapse
|