Skip to main content
Erschienen in: BMC Medicine 1/2011

Open Access 01.12.2011 | Minireview

Revisiting the technical validation of tumour biomarker assays: how to open a Pandora's box

verfasst von: Caterina Marchiò, Mitch Dowsett, Jorge S Reis-Filho

Erschienen in: BMC Medicine | Ausgabe 1/2011

Abstract

A tumour biomarker is a characteristic that is objectively measured and evaluated in tumour samples as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention. The development of a biomarker contemplates distinct phases, including discovery by hypothesis-generating preclinical or exploratory studies, development and qualification of the assay for the identification of the biomarker in clinical samples, and validation of its clinical significance. Although guidelines for the development and validation of biomarkers are available, their implementation is challenging, owing to the diversity of biomarkers being developed. The term 'validation' undoubtedly has several meanings; however, in the context of biomarker research, a test may be considered valid if it is 'fit for purpose'. In the process of validation of a biomarker assay, a key point is the validation of the methodology. Here we discuss the challenges for the technical validation of immunohistochemical and gene expression assays to detect tumour biomarkers and provide suggestions of pragmatic solutions to address these challenges.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1741-7015-9-41) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

CM drafted the manuscript. MD and JSR conceptually designed the manuscript and revised the draft. All authors read and approved the final version of the manuscript.
Abkürzungen
ER
oestrogen receptor
FFPE
formalin-fixed, paraffin-embedded
HER2
human epidermal growth factor receptor 2
IHC
immunohistochemistry
NEQAS
National External Quality Assessment Service
PR
progesterone receptor
siRNA
short interfering RNA
TMA
tissue microarray.

Introduction

Biomarkers are the defining facet of translational cancer research; however, there is a great deal of confusion about the actual definition of what a biomarker is and what its characteristics are. Arguably, the most widely accepted definition is the one put forward by the Biomarkers Definitions Working Group, which defines a biomarker as "a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention" [1]. As the definition suggests, biomarkers can be used for multiple purposes in cancer research and measured in distinct types of specimens, such as tissue samples as well as peripheral blood (see, for example, circulating tumour cells), by using several assays. Despite the existence of excellent guidelines for the development and validation of biomarkers [24], there is a great deal of confusion when it comes to determining the validity of an assay to detect a biomarker. The process of biomarker development is by no means trivial; for the purpose of simplicity, it can be broadly divided into four main phases: (1) discovery of a potential biomarker through hypothesis-generating preclinical or exploratory studies; (2) development and technical validation of the assay for the identification of the biomarker in clinical samples; (3) demonstration of the biomarker's potential clinical utility, first in retrospective analyses and subsequently in prospective studies; and (4) continued assessment of the validity of the biomarker in routine clinical practice (Table 1). The term 'validation' in the context of clinical studies has changed dramatically over the years; currently, perhaps the most adequate definition for a valid biomarker is a biomarker that is fit for purpose [5].
Table 1
Overview of the phases of biomarker development and validationa
Phase
Means/instruments
Main challenges and sources of bias
Discovery of a potential
biomarker
Hypothesis-generating preclinical
or exploratory studies
Selection of biomarker based on the
availability of antibodies on the market
Development and technical validation of
the assay for the identification of the
biomarker
Optimisation of IHC-based assays for
formalin-fixed, paraffin-embedded
samples
- Use of clinical samples not suitable for
the analysis (for example core biopsies instead
of surgical samples and TMA instead of
full sections)
- Lack of reliable positive and negative
controls
- Poor fixation of clinical samples
- Wrong antigen retrieval procedure
- Wrong detection method Misinterpretation
of the results
- Training/competency of the staff
- Suboptimal performance of the antibody
due to poor fixation of archival tissues
(in particular for retrospective studies)
Validation of the clinical significance
of the biomarker
First retrospective studies and
subsequent prospective studies
- Training/competency of the staff
- Use of small cohorts or large cohorts
that include series of cases in which
the biomarker has been previously validated
Continued assessment of the
validity of the biomarker in
routine practice
Internal and external quality
assurance program
- Poor participation/adhesion to the
programme
- Lack of competency of pathologists
participating in the program
- No action taken if failing quality
assurance
a Description of the phases of biomarker development and validation, and the main challenges and potential sources of bias, using immunohistochemistry-based
assays as a paradigm.
IHC: immunohistochemistry; TMA: tissue microarray.
Although great emphasis is given to the discovery and validation of the clinical significance of the biomarker, the technical validation of assays for novel biomarkers has not been embraced with the same enthusiasm, probably because of its more technical and apparently less rewarding nature. Nonetheless, the process of assay validation is critical for the introduction of a new biomarker in routine clinical practice. This minireview focuses on the technical issues related to validation of biomarkers analysed directly in human tumour tissue samples, with breast cancer pathology serving as a paradigm. It should be noted, however, that the concepts discussed in this review are applicable to biomarkers based on other types of samples (for example, circulating tumour cells, blood, serum, urine and other bodily fluids).

Validation: when and why?

A biomarker often fails to be incorporated into clinical practice, not because of flawed science underpinning its discovery but because of poor choice of the assay used for its detection and inadequate validation of its accuracy [6, 7]. For the successful use of a biomarker assay in clinical practice, it is of paramount importance that the testing of the assay employ robust reagents and be based on a reliable and robust technology. Several false dawns in translational research have stemmed from attempts to introduce a technology that was not sufficiently mature at the time and from the sources of technical bias not being entirely known (for example, mass spectrometry-based serum proteomic analysis for the diagnosis of ovarian cancer) [8, 9].
Methodological validation needs to be considered both in well-established and in new assays. In the first scenario, as the assay is already introduced into clinical practice, ongoing demonstration of validity through in-house and external quality assurance schemes allows false-positive and false-negative results to be minimised. The importance of this problem is placed in stark focus by recognising that these checks are necessary to ensure that potentially life-extending or lifesaving therapies are not denied to patients who may benefit from them and that patients are not unnecessarily exposed to toxicities or given false hope. Several initiatives have been introduced to provide guidelines for testing the adequacy of the routine biomarker tests (that is, oestrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) tests) in breast cancer (Table 2). In the United Kingdom, the National External Quality Assessment Service programme organises four assessments every year that include antibody testing on multi-tissue and cell line blocks, an expert review, a confidential report, online images and detailed analysis of data regarding methods [10]. Importantly, these approaches have led to a significant increase in the number of laboratories offering optimal quality immunohistochemical assessment of these markers [10]. External quality assurance does not, however, replace the need for internal quality control measures that enable individual batches of samples to be accepted with confidence or, alternatively, rejected. It should also be noted that internal controls in individual samples, represented by the presence of normal tissue adjacent to the tumour, can be of great support to define the validity of a test. For example, in the assessment of ER, PR and HER2, it is crucial to select a tumour block that, in addition to the tumour areas, also contains adjacent normal ducts or lobules which can be used as internal controls.
Table 2
Examples of external quality assurance schemes for routine biomarkers employed in breast cancer pathologya
QA scheme
Scope
Website
ASCO/CAP
To improve the accuracy of test results and ensure that
patients receive appropriate care
UK NEQAS
To promote optimal patient care by facilitating the
availability of reliable laboratory investigations,
through provision of objective information on
participant performance and professional advice
and assistance where appropriate
NordiQC
To promote the quality of IHC by arranging schemes
for pathology laboratories, assessing tissue stains,
giving recommendations for improvement and
providing good protocols
Canadian
external quality
assurance
program
To systematically monitor and improve the proficiency
of IHC testing laboratories and those involved with IHC
testing nationwide
The RCPA
Quality
Assurance
Program
To provide external proficiency testing for
histopathology laboratories in the areas of diagnostic
and technical expertise
a Names, scope and websites of the main external quality assurance schemes for breast cancer biomarkers based on immunohistochemical and in situ hybridisation assays.
QA: quality assurance; ASCO: American Society of Clinical Oncology; CAP: College of American Pathologists; UK NEQAS: United Kingdom
External Quality Assessment Service; IHC: immunohistochemistry; NordiQC: Nordic Immunohistochemical Quality Control; RCPA: Royal College
of Pathologists of Australasia.
The early technical validation of a new biomarker assay is critical to minimise waste of resources, generation of misleading results and possibly disrepute. In this context, however, full validity is often difficult to prove, given that these assays come directly from research assays for which a gold standard is usually not available.

Validation of assays for novel biomarkers

Immunohistochemistry, fluorescence and chromogenic in situ hybridisation, expression profiling, either microarray-based or performed by quantitative real-time reverse transcriptase-polymerase chain reaction (qRT-PCR), and mutation analysis represent the main techniques currently being introduced into everyday practice in pathology laboratories. Among these techniques, immunohistochemical tests remain the most widely used in routine practice and, importantly, in the assessment of biomarkers in translational research endeavours. We therefore focus primarily on their validation.
In the case of immunohistochemistry, a typical 'quick and easy' approach is illustrated in Figure 1. It should be noted, however, that such an approach carries with it far more pitfalls than many investigators assume. Although immunohistochemistry has been introduced in pathology laboratories for more than 25 years and is a relatively user-friendly technique, one should not forget that the results of immunohistochemical analysis may be affected by a series of preanalytical and analytical parameters [1115].
Preanalytical variables are the 'weakest link' of immunohistochemistry, as the properties of the tissues analysed and, consequently, the results, may be affected by several factors, including the time to collection (that is, the length of time that tissues are subjected to warm ischaemia between removal of the tissue at surgery and fixation), details of fixation (type of fixative agent used and length and conditions of fixation), dehydration steps, and conditions for paraffin-embedding (temperature of the paraffin). These preanalytical parameters are beyond the control of investigators, are most often unrecorded, and constitute a major potential source of bias, in particular in multicentre, retrospective studies. For example, the time to fixation and its length have been branded as the 'Achilles heel' of phosphoprotein assessment in clinical specimens [16] as recently shown by Pinhel and colleagues [16, 17], who found consistently significantly lower levels of the phosphoepitopes in surgical specimens compared with those found in core biopsies.
Four main analytical issues require special attention. Antigen retrieval (that is, a method that enables immunohistochemistry to be applied to formalin-fixed, paraffin-embedded (FFPE) samples), type of detection system, the choice of antibody, and the material to be used [1113]. Enzyme-based and heat-induced epitope retrieval are available, for which strict protocols should be followed to obtain accurate results [11]. Excellent reviews have described the pitfalls of antigen retrieval and how they can be overcome [1115]. Suffice it to say that in the absence of optimal positive and negative controls and a 'gold standard', changing antigen retrieval settings can render a given case positive or negative.
Arguably the most challenging aspect of any immunohistochemical assay is the choice of antibody. Before applying 'research-only' antibodies to FFPE tissues, their sensitivity and specificity need to be determined. It should be emphasised that this cannot be achieved merely by evaluating the sensitivity and specificity of the antibody by means of Western blot analysis; these parameters need to be assessed in FFPE samples and using optimal controls (Figure 2). It is always difficult to find the optimal balance between sensitivity and specificity, and, in this respect, the antigen retrieval method and antibody titration are critical, as is the availability of optimal controls to be used as a gold standard. One helpful and informative approach uses in-house or publicly available expression array data sets to identify cell lines that can be exploited as positive controls and then to validate their use as positive controls on the basis of qRT-PCR assay and Western blot analysis. Negative controls can be generated by siRNA (short interfering RNA) knockdown of the gene of interest in the cell line. Subsequently, antibody testing can be performed on FFPE cell pellets, and finally optimisation of immunohistochemistry on FFPE tissues will follow (Figure 2). In this process, the type of detected subcellular localisation may also help the researcher understand whether the antibody is specific. Finally, when dealing with biomarkers used in routine diagnostic practice, audit of the results from annual workload can provide supporting evidence of the actual performance of an optimised assay used in routine practice.
Another crucial aspect is the type of tissue to be used. Tissue microarrays (TMAs) have become very popular in studies aiming to determine the distribution of a given marker in a cohort of samples [18, 19]. Although TMAs have proven to be excellent tools, they should be employed for the testing of biomarkers whose expression is relatively homogeneous and should be used only if the concordance between the results of the analysis of a given marker on whole tissue sections and TMAs is close. Regrettably, assessing the latter appears not to be a common practice. It should be noted that this issue is applicable even to well-validated tests in breast cancer research and practice (for example, the discordant results in the analysis of PR expression in TMAs vs. whole tissue sections [20]).

Gene expression profiling studies: microarrays or micro-awry?

In recent years, microarray gene expression profiling and its derivatives have been widely applied to the molecular and biological classification of breast cancers, and several prognostic and predictive signatures have been reported, some of which have been introduced into clinical practice (for example, Oncotype DX Breast Cancer Assay (Genomic Health, Inc., Redwood City, CA, USA) and MammaPrint assay (Agendia BV, Amsterdam, the Netherlands); for reviews, please see Weigelt et al. [21], Sotiriou and Pusztai [22], and Reis-Filho et al. [23]). The types of analysis and the data they generate pose a major challenge for the translation of their findings into assays that can be used in routine clinical practice, as the reliability, reproducibility and stability of some have been called into question [2325]. In terms of preanalytical variables, most of the parameters that affect immunohistochemistry also affect gene expression profiling (for example, time to tissue fixation, time to freezing, length of fixation, type of fixative used and tissue storage). In addition, given that these technologies are based on nucleic acid extracts from tissue homogenates, the variable tumour content may also constitute a confounding factor. Cleator et al. [26] demonstrated that varying amounts of non-neoplastic cells in samples subjected to gene expression increases the error rates of multigene predictors, providing direct evidence that the non-tumour content of breast cancer samples has a significant effect on gene expression profiles.
Data analysis of these 'mega-parameter' profiles also poses huge challenges. Microarray technology and bioinformatics/statistics applied to microarray analysis have developed at disparate speeds and this may have led to inappropriate conclusions being drawn and contributed to the first wave of over-optimism and, then, to the subsequent wave of (over)scepticism with this type of technology experienced in the last 10 years [24]. Indeed, when these signatures were first described, there was little awareness of problems of data 'overfitting' and methods for power calculation for microarrays [21, 23]. This field has developed rapidly, however, and guidelines regulating how a therapeutically significant gene signature should be developed and validated are now available [27, 28]. The chances of success in developing and validating gene signatures will be significantly increased if these guidelines are strictly followed.

Conclusions

Technical validation and qualification of a biomarker assay may not be as glamorous as biomarker discovery; however, together they comprise the critical lynchpins of translational cancer research. Guidelines for the development and validation of biomarker assays are available. It should be noted, however, that not all aspects of these guidelines may be applicable to the assay to be developed. In these cases, imaginative approaches should be sought while erring on the side of caution. To avoid waste of resources and the use or publication of misleading data, full awareness of the technical challenges in assessment of the robustness of assays and careful evaluation of the context of the assay to be developed are required [29]. Finally, in the reporting of the results, uncertainties should be disclosed and all caveats ought to be voiced. While these issues are undoubtedly challenging and consideration of the numerous potential pitfalls may be discouraging, awareness and avoidance of these problems can allow clinically relevant work of great value to be conducted [29].

Acknowledgements

CM is funded by the University of Turin and by Ricerca Sanitaria Finalizzata 2009. MD is funded by Breakthrough Breast Cancer and The Mary-Jean Mitchell Green Foundation. We acknowledge National Health Service funding to the National Institute for Health Research Biomedical Research Centre. JSRF is funded in part by Breakthrough Breast Cancer.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

CM drafted the manuscript. MD and JSR conceptually designed the manuscript and revised the draft. All authors read and approved the final version of the manuscript.
Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
Zurück zum Zitat Biomarkers Definitions Working Group: Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001, 69: 89-95. 10.1067/mcp.2001.113989.CrossRef Biomarkers Definitions Working Group: Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001, 69: 89-95. 10.1067/mcp.2001.113989.CrossRef
2.
Zurück zum Zitat Diamandis EP, Hoffman BR, Sturgeon CM: National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines for the Use of Tumor Markers. Clin Chem. 2008, 54: 1935-1939. 10.1373/clinchem.2008.105494.CrossRef Diamandis EP, Hoffman BR, Sturgeon CM: National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines for the Use of Tumor Markers. Clin Chem. 2008, 54: 1935-1939. 10.1373/clinchem.2008.105494.CrossRef
3.
Zurück zum Zitat Sturgeon CM, Duffy MJ, Stenman UH, Lilja H, Brünner N, Chan DW, Babaian R, Bast RC, Dowell B, Esteva FJ, Haglund C, Harbeck N, Hayes DF, Holten-Andersen M, Klee GG, Lamerz R, Looijenga LH, Molina R, Nielsen HJ, Rittenhouse H, Semjonow A, Shih IeM, Sibley P, Sölétormos G, Stephan C, Sokoll L, Hoffman BR, Diamandis EP, National Academy of Clinical Biochemistry: National Academy of Clinical Biochemistry laboratory medicine practice guidelines for use of tumor markers in testicular, prostate, colorectal, breast, and ovarian cancers. Clin Chem. 2008, 54: e11-e79. 10.1373/clinchem.2008.105601.CrossRef Sturgeon CM, Duffy MJ, Stenman UH, Lilja H, Brünner N, Chan DW, Babaian R, Bast RC, Dowell B, Esteva FJ, Haglund C, Harbeck N, Hayes DF, Holten-Andersen M, Klee GG, Lamerz R, Looijenga LH, Molina R, Nielsen HJ, Rittenhouse H, Semjonow A, Shih IeM, Sibley P, Sölétormos G, Stephan C, Sokoll L, Hoffman BR, Diamandis EP, National Academy of Clinical Biochemistry: National Academy of Clinical Biochemistry laboratory medicine practice guidelines for use of tumor markers in testicular, prostate, colorectal, breast, and ovarian cancers. Clin Chem. 2008, 54: e11-e79. 10.1373/clinchem.2008.105601.CrossRef
4.
Zurück zum Zitat Sturgeon CM, Hoffman BR, Chan DW, Ch'ng SL, Hammond E, Hayes DF, Liotta LA, Petricoin EF, Schmitt M, Semmes OJ, Sölétormos G, van der Merwe E, Diamandis EP, National Academy of Clinical Biochemistry: National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines for use of tumor markers in clinical practice: quality requirements. Clin Chem. 2008, 54: e1-e10. 10.1373/clinchem.2007.094144.CrossRef Sturgeon CM, Hoffman BR, Chan DW, Ch'ng SL, Hammond E, Hayes DF, Liotta LA, Petricoin EF, Schmitt M, Semmes OJ, Sölétormos G, van der Merwe E, Diamandis EP, National Academy of Clinical Biochemistry: National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines for use of tumor markers in clinical practice: quality requirements. Clin Chem. 2008, 54: e1-e10. 10.1373/clinchem.2007.094144.CrossRef
5.
Zurück zum Zitat Ransohoff DF: Rules of evidence for cancer molecular-marker discovery and validation. Nat Rev Cancer. 2004, 4: 309-314. 10.1038/nrc1322.CrossRef Ransohoff DF: Rules of evidence for cancer molecular-marker discovery and validation. Nat Rev Cancer. 2004, 4: 309-314. 10.1038/nrc1322.CrossRef
6.
Zurück zum Zitat Carden CP, Sarker D, Postel-Vinay S, Yap TA, Attard G, Banerji U, Garrett MD, Thomas GV, Workman P, Kaye SB, de Bono JS: Can molecular biomarker-based patient selection in phase I trials accelerate anticancer drug development?. Drug Discov Today. 2010, 15: 88-97. 10.1016/j.drudis.2009.11.006.CrossRef Carden CP, Sarker D, Postel-Vinay S, Yap TA, Attard G, Banerji U, Garrett MD, Thomas GV, Workman P, Kaye SB, de Bono JS: Can molecular biomarker-based patient selection in phase I trials accelerate anticancer drug development?. Drug Discov Today. 2010, 15: 88-97. 10.1016/j.drudis.2009.11.006.CrossRef
7.
Zurück zum Zitat Cummings J, Raynaud F, Jones L, Sugar R, Dive C: Fit-for-purpose biomarker method validation for application in clinical trials of anticancer drugs. Br J Cancer. 2010, 103: 1313-1317. 10.1038/sj.bjc.6605910.CrossRefPubMedCentral Cummings J, Raynaud F, Jones L, Sugar R, Dive C: Fit-for-purpose biomarker method validation for application in clinical trials of anticancer drugs. Br J Cancer. 2010, 103: 1313-1317. 10.1038/sj.bjc.6605910.CrossRefPubMedCentral
8.
Zurück zum Zitat Ransohoff DF: Bias as a threat to the validity of cancer molecular-marker research. Nat Rev Cancer. 2005, 5: 142-149. 10.1038/nrc1550.CrossRef Ransohoff DF: Bias as a threat to the validity of cancer molecular-marker research. Nat Rev Cancer. 2005, 5: 142-149. 10.1038/nrc1550.CrossRef
9.
Zurück zum Zitat Diamandis EP: Re: Serum proteomic patterns for detection of prostate cancer. J Natl Cancer Inst. 2003, 95: 489-491. 10.1093/jnci/95.6.489.CrossRef Diamandis EP: Re: Serum proteomic patterns for detection of prostate cancer. J Natl Cancer Inst. 2003, 95: 489-491. 10.1093/jnci/95.6.489.CrossRef
10.
Zurück zum Zitat Miller K, Ibrahim M, Barnett S, Jasani B: Technical aspects of predictive and prognostic markers in breast cancer: what UK NEQAS data show. Curr Diagn Pathol. 2007, 13: 135-149. 10.1016/j.cdip.2006.12.003.CrossRef Miller K, Ibrahim M, Barnett S, Jasani B: Technical aspects of predictive and prognostic markers in breast cancer: what UK NEQAS data show. Curr Diagn Pathol. 2007, 13: 135-149. 10.1016/j.cdip.2006.12.003.CrossRef
11.
Zurück zum Zitat Bussolati G, Leonardo E: Technical pitfalls potentially affecting diagnoses in immunohistochemistry. J Clin Pathol. 2008, 61: 1184-1192. 10.1136/jcp.2007.047720.CrossRef Bussolati G, Leonardo E: Technical pitfalls potentially affecting diagnoses in immunohistochemistry. J Clin Pathol. 2008, 61: 1184-1192. 10.1136/jcp.2007.047720.CrossRef
12.
Zurück zum Zitat Yaziji H, Barry T: Diagnostic immunohistochemistry: what can go wrong?. Adv Anat Pathol. 2006, 13: 238-246. 10.1097/01.pap.0000213041.39070.2f.CrossRef Yaziji H, Barry T: Diagnostic immunohistochemistry: what can go wrong?. Adv Anat Pathol. 2006, 13: 238-246. 10.1097/01.pap.0000213041.39070.2f.CrossRef
13.
Zurück zum Zitat Leong AS: Pitfalls in diagnostic immunohistology. Adv Anat Pathol. 2004, 11: 86-93. 10.1097/00125480-200403000-00002.CrossRef Leong AS: Pitfalls in diagnostic immunohistology. Adv Anat Pathol. 2004, 11: 86-93. 10.1097/00125480-200403000-00002.CrossRef
14.
Zurück zum Zitat Leong TY, Cooper K, Leong AS: Immunohistology: past, present, and future. Adv Anat Pathol. 2010, 17: 404-418. 10.1097/PAP.0b013e3181f8957c.CrossRef Leong TY, Cooper K, Leong AS: Immunohistology: past, present, and future. Adv Anat Pathol. 2010, 17: 404-418. 10.1097/PAP.0b013e3181f8957c.CrossRef
15.
Zurück zum Zitat Leong TY, Leong AS: How does antigen retrieval work?. Adv Anat Pathol. 2007, 14: 129-131. 10.1097/PAP.0b013e31803250c7.CrossRef Leong TY, Leong AS: How does antigen retrieval work?. Adv Anat Pathol. 2007, 14: 129-131. 10.1097/PAP.0b013e31803250c7.CrossRef
16.
Zurück zum Zitat Siddiqui S, Rimm DL: Pre-analytic variables and phospho-specific antibodies: the Achilles heel of immunohistochemistry. Breast Cancer Res. 2010, 12: 113-10.1186/bcr2782.CrossRefPubMedCentral Siddiqui S, Rimm DL: Pre-analytic variables and phospho-specific antibodies: the Achilles heel of immunohistochemistry. Breast Cancer Res. 2010, 12: 113-10.1186/bcr2782.CrossRefPubMedCentral
17.
Zurück zum Zitat Pinhel IF, MacNeill FA, Hills MJ, Salter J, Detre S, A'Hern R, Nerurkar A, Osin P, Smith IE, Dowsett M: Extreme loss of immunoreactive p-Akt and p-Erk1/2 during routine fixation of primary breast cancer. Breast Cancer Res. 2010, 12: R76-10.1186/bcr2719.CrossRefPubMedCentral Pinhel IF, MacNeill FA, Hills MJ, Salter J, Detre S, A'Hern R, Nerurkar A, Osin P, Smith IE, Dowsett M: Extreme loss of immunoreactive p-Akt and p-Erk1/2 during routine fixation of primary breast cancer. Breast Cancer Res. 2010, 12: R76-10.1186/bcr2719.CrossRefPubMedCentral
18.
Zurück zum Zitat Kononen J, Bubendorf L, Kallioniemi A, Bärlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP: Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med. 1998, 4: 844-847. 10.1038/nm0798-844.CrossRef Kononen J, Bubendorf L, Kallioniemi A, Bärlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP: Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med. 1998, 4: 844-847. 10.1038/nm0798-844.CrossRef
19.
Zurück zum Zitat Camp RL, Charette LA, Rimm DL: Validation of tissue microarray technology in breast carcinoma. Lab Invest. 2000, 80: 1943-1949. 10.1038/labinvest.3780204.CrossRef Camp RL, Charette LA, Rimm DL: Validation of tissue microarray technology in breast carcinoma. Lab Invest. 2000, 80: 1943-1949. 10.1038/labinvest.3780204.CrossRef
20.
Zurück zum Zitat Nadji M, Gomez-Fernandez C, Ganjei-Azar P, Morales AR: Immunohistochemistry of estrogen and progesterone receptors reconsidered: experience with 5,993 breast cancers. Am J Clin Pathol. 2005, 123: 21-27. 10.1309/4WV79N2GHJ3X1841.CrossRef Nadji M, Gomez-Fernandez C, Ganjei-Azar P, Morales AR: Immunohistochemistry of estrogen and progesterone receptors reconsidered: experience with 5,993 breast cancers. Am J Clin Pathol. 2005, 123: 21-27. 10.1309/4WV79N2GHJ3X1841.CrossRef
21.
Zurück zum Zitat Weigelt B, Baehner FL, Reis-Filho JS: The contribution of gene expression profiling to breast cancer classification, prognostication and prediction: a retrospective of the last decade. J Pathol. 2010, 220: 263-280. 10.1002/path.2629.CrossRef Weigelt B, Baehner FL, Reis-Filho JS: The contribution of gene expression profiling to breast cancer classification, prognostication and prediction: a retrospective of the last decade. J Pathol. 2010, 220: 263-280. 10.1002/path.2629.CrossRef
22.
Zurück zum Zitat Sotiriou C, Pusztai L: Gene-expression signatures in breast cancer. N Engl J Med. 2009, 360: 790-800. 10.1056/NEJMra0801289.CrossRef Sotiriou C, Pusztai L: Gene-expression signatures in breast cancer. N Engl J Med. 2009, 360: 790-800. 10.1056/NEJMra0801289.CrossRef
23.
Zurück zum Zitat Reis-Filho JS, Weigelt B, Fumagalli D, Sotiriou C: Molecular profiling: moving away from tumor philately. Sci Transl Med. 2010, 2: 47ps43 Reis-Filho JS, Weigelt B, Fumagalli D, Sotiriou C: Molecular profiling: moving away from tumor philately. Sci Transl Med. 2010, 2: 47ps43
24.
Zurück zum Zitat Weigelt B, Reis-Filho JS: Molecular profiling currently offers no more than tumour morphology and basic immunohistochemistry. Breast Cancer Res. 2010, 12 (Suppl 4): S5-10.1186/bcr2734.CrossRefPubMedCentral Weigelt B, Reis-Filho JS: Molecular profiling currently offers no more than tumour morphology and basic immunohistochemistry. Breast Cancer Res. 2010, 12 (Suppl 4): S5-10.1186/bcr2734.CrossRefPubMedCentral
25.
Zurück zum Zitat Weigelt B, Mackay A, A'Hern R, Natrajan R, Tan DS, Dowsett M, Ashworth A, Reis-Filho JS: Breast cancer molecular profiling with single sample predictors: a retrospective analysis. Lancet Oncol. 2010, 11: 339-349. 10.1016/S1470-2045(10)70008-5.CrossRef Weigelt B, Mackay A, A'Hern R, Natrajan R, Tan DS, Dowsett M, Ashworth A, Reis-Filho JS: Breast cancer molecular profiling with single sample predictors: a retrospective analysis. Lancet Oncol. 2010, 11: 339-349. 10.1016/S1470-2045(10)70008-5.CrossRef
26.
Zurück zum Zitat Cleator SJ, Powles TJ, Dexter T, Fulford L, Mackay A, Smith IE, Valgeirsson H, Ashworth A, Dowsett M: The effect of the stromal component of breast tumours on prediction of clinical outcome using gene expression microarray analysis. Breast Cancer Res. 2006, 8: R32-10.1186/bcr1506.CrossRefPubMedCentral Cleator SJ, Powles TJ, Dexter T, Fulford L, Mackay A, Smith IE, Valgeirsson H, Ashworth A, Dowsett M: The effect of the stromal component of breast tumours on prediction of clinical outcome using gene expression microarray analysis. Breast Cancer Res. 2006, 8: R32-10.1186/bcr1506.CrossRefPubMedCentral
27.
Zurück zum Zitat Simon R: Roadmap for developing and validating therapeutically relevant genomic classifiers. J Clin Oncol. 2005, 23: 7332-7341. 10.1200/JCO.2005.02.8712.CrossRef Simon R: Roadmap for developing and validating therapeutically relevant genomic classifiers. J Clin Oncol. 2005, 23: 7332-7341. 10.1200/JCO.2005.02.8712.CrossRef
28.
Zurück zum Zitat Dupuy A, Simon RM: Critical review of published microarray studies for cancer outcome and guidelines on statistical analysis and reporting. J Natl Cancer Inst. 2007, 99: 147-157. 10.1093/jnci/djk018.CrossRef Dupuy A, Simon RM: Critical review of published microarray studies for cancer outcome and guidelines on statistical analysis and reporting. J Natl Cancer Inst. 2007, 99: 147-157. 10.1093/jnci/djk018.CrossRef
29.
Zurück zum Zitat Diamandis EP: Cancer biomarkers: can we turn recent failures into success?. J Natl Cancer Inst. 2010, 102: 1462-1467. 10.1093/jnci/djq306.CrossRefPubMedCentral Diamandis EP: Cancer biomarkers: can we turn recent failures into success?. J Natl Cancer Inst. 2010, 102: 1462-1467. 10.1093/jnci/djq306.CrossRefPubMedCentral
Metadaten
Titel
Revisiting the technical validation of tumour biomarker assays: how to open a Pandora's box
verfasst von
Caterina Marchiò
Mitch Dowsett
Jorge S Reis-Filho
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
BMC Medicine / Ausgabe 1/2011
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/1741-7015-9-41

Weitere Artikel der Ausgabe 1/2011

BMC Medicine 1/2011 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 24 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Update Allgemeinmedizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.