1
|
Lee B, Chern A, Fu AY, Zhang A, Sha MY. A Highly Sensitive XNA-Based RT-qPCR Assay for the Identification of ALK, RET, and ROS1 Fusions in Lung Cancer. Diagnostics (Basel) 2024; 14:488. [PMID: 38472960 DOI: 10.3390/diagnostics14050488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Lung cancer is often triggered by genetic alterations that result in the expression of oncogenic tyrosine kinases. Specifically, ALK, RET, and ROS1 chimeric receptor tyrosine kinases are observed in approximately 5-7%, 1-2%, and 1-2% of NSCLC patients, respectively. The presence of these fusion genes determines the response to tyrosine kinase inhibitors. Thus, accurate detection of these gene fusions is essential in cancer research and precision oncology. To address this need, we have developed a multiplexed RT-qPCR assay using xeno nucleic acid (XNA) molecular clamping technology to detect lung cancer fusions. This assay can quantitatively detect thirteen ALK, seven ROS1, and seven RET gene fusions in FFPE samples. The sensitivity of the assay was established at a limit of detection of 50 copies of the synthetic template. Our assay has successfully identified all fusion transcripts using 50 ng of RNA from both reference FFPE samples and cell lines. After validation, a total of 77 lung cancer patient FFPE samples were tested, demonstrating the effectiveness of the XNA-based fusion gene assay with clinical samples. Importantly, this assay is adaptable to highly degraded RNA samples with low input amounts. Future steps involve expanding the testing to include a broader range of clinical samples as well as cell-free RNAs to further validate its applicability and reliability.
Collapse
Affiliation(s)
- Bongyong Lee
- DiaCarta Inc., 4385 Hopyard Rd., Suite 100, Pleasanton, CA 94588, USA
| | - Andrew Chern
- DiaCarta Inc., 4385 Hopyard Rd., Suite 100, Pleasanton, CA 94588, USA
| | - Andrew Y Fu
- DiaCarta Inc., 4385 Hopyard Rd., Suite 100, Pleasanton, CA 94588, USA
| | - Aiguo Zhang
- DiaCarta Inc., 4385 Hopyard Rd., Suite 100, Pleasanton, CA 94588, USA
| | - Michael Y Sha
- DiaCarta Inc., 4385 Hopyard Rd., Suite 100, Pleasanton, CA 94588, USA
| |
Collapse
|
2
|
Cree IA, Lokuhetty D, Tan PH. The World Health Organization Classification of Tumors and External Quality Assurance for Immunohistochemistry and Molecular Pathology. Arch Pathol Lab Med 2022; 146:1303-1307. [PMID: 35671156 DOI: 10.5858/arpa.2021-0491-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— This article is based on a talk given by the lead author at the Eigth Annual Princeton Integrated Pathology Symposium: Breast Pathology, on Sunday, April 11, 2021. OBJECTIVE.— To show how the World Health Organization (WHO) Classification of Tumours links to the requirements for quality assurance in breast pathology, including both immunohistochemistry and molecular pathology. DATA SOURCES.— The WHO Classification of Tumours 5th edition Breast Tumours entries formed the basis of the talk, together with guidance published by the International Quality Network for Pathology. CONCLUSIONS.— The WHO Classification of Tumours provides a definitive set of international standards for tumor diagnosis contributed by experts, based on available clinical and research evidence. Techniques used in pathology need internal and external quality assurance to ensure accurate reports for patient management.
Collapse
Affiliation(s)
- Ian A Cree
- From the International Agency for Research on Cancer, World Health Organization, Lyon, France (Cree)
| | - Dilani Lokuhetty
- The Department of Pathology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka (Lokuhetty)
| | - Puay Hoon Tan
- The Division of Pathology, Singapore General Hospital, Singapore (Tan)
| |
Collapse
|
3
|
Normanno N, Apostolidis K, Wolf A, Al Dieri R, Deans Z, Fairley J, Maas J, Martinez A, Moch H, Nielsen S, Pilz T, Rouleau E, Patton S, Williams V. Access and quality of biomarker testing for precision oncology in Europe. Eur J Cancer 2022; 176:70-77. [PMID: 36194905 DOI: 10.1016/j.ejca.2022.09.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Predictive biomarkers are essential for selecting the best therapeutic strategy in patients with cancer. The International Quality Network for Pathology, the European Cancer Patient Coalition and the European Federation of Pharmaceuticals Industries and Associations evaluated the access to and quality of biomarker testing across Europe. METHODS Data sources included surveys of 141 laboratory managers and 1.665 patients, and 58 in-depth interviews with laboratory managers, physicians and payers. Four access metrics (laboratory access, test availability, test reimbursement, test order rate) and three quality metrics (quality scheme participation, laboratory accreditation, test turnaround time) were applied to rank the results. RESULTS The access to precision medicines is higher in countries with public national reimbursement processes in place. Lack of diagnostic laboratory infrastructure, inefficient organization and/or insufficient public reimbursement narrow the access to single biomarker tests in many European countries. In countries with limited public reimbursement, pharma and patients' out of pocket were the primary funding sources for testing. Uptake of multi-biomarker next generation sequencing (NGS) is highly varied, ranging from 0% to >50%. Financial constraints, a lack of NGS testing capabilities and the failure to include NGS testing in the guidelines represent the main barriers to NGS implementation. The quality of biomarker testing is highest in Western and Northern Europe, with more than 90% of laboratories participating in quality assurance schemes. CONCLUSIONS Our data clearly indicate the need for a call to action to ensure the clinical implementation of precision medicine in Europe.
Collapse
Affiliation(s)
- Nicola Normanno
- Istituto Nazionale Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy.
| | | | | | - Raed Al Dieri
- European Society of Pathology (ESP), Brussels, Belgium
| | | | | | - Jörg Maas
- Deutsche Gesellschaft für Pathologie E.V. (DGP), Berlin, Germany
| | | | - Holger Moch
- University Hospital and University, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
4
|
Shetty O, Shet T, Iyer R, Gogte P, Gurav M, Joshi P, Karnik N, Pai T, Epari S, Desai S. Impact of COVID-19 on quality checks of solid tumor molecular diagnostic testing-A surveillance by EQAS provider in India. PLoS One 2022; 17:e0274089. [PMID: 36137153 PMCID: PMC9498956 DOI: 10.1371/journal.pone.0274089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 08/22/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Molecular tests in solid tumours for targeted therapies call for the need to ensure precision testing. To accomplish this participation in the External Quality Assessment Program (EQAS) is required. This evaluates the consistency of diagnostic testing procedures and offers guidance for improving quality. Outbreak of COVID-19 pandemic led to worldwide lockdown and disruption of healthcare services including participation in EQAS.The present study describes the extended scope of EQAS offered byMPQAP (Molecular Pathology Quality Assurance Program), the first proficiency test provider for solid tumor diagnostics in India. The study surveys the preparedness of molecular testing laboratories in routine diagnostics and participation for quality assessment scheme. METHODS A documented guideline for measures and precautions to be carried by testing laboratories in performing routine diagnostic tests during the lockdown period were charted and distributed to all MPQAP participant centres. A survey was conducted for MPQAP participants to check whether laboratories were involved in COVID-19 testing and to evaluate the impact of lockdown on the operations of diagnostics procedures. From the acquired response of the survey, 2 cycles out of initially proposed 11 cycles were executed with transformed approach using digital tools and image interpretation modules. FINDINGS Out of 25 solid tumour testing laboratories registered as participants, 15 consented to participate in survey. The summary of survey conveyed the impact of COVID-19onroutine operations of diagnostics tests such as shortcomings in inventory and human resource management. Thirteen participants showed active willingness and consented to participate in EQAS test scheme. INTERPRETATIONS The survey findings and assessment of EQAS cycles endorsed the quality testing procedures carried by participating laboratories throughout the lockdown. It highlighted the utility of EQAS participation during pandemic along with emphasis on safety measures for continual improvement in quality of diagnostic services.
Collapse
Affiliation(s)
- Omshree Shetty
- Department of Pathology, Molecular Pathology Laboratory, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Tanuja Shet
- Department of Pathology, Molecular Pathology Laboratory, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Ramya Iyer
- Department of Pathology, Molecular Pathology Laboratory, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Prachi Gogte
- Department of Pathology, Molecular Pathology Laboratory, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Mamta Gurav
- Department of Pathology, Molecular Pathology Laboratory, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Pradnya Joshi
- Department of Pathology, Molecular Pathology Laboratory, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Nupur Karnik
- Department of Pathology, Molecular Pathology Laboratory, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Trupti Pai
- Department of Pathology, Molecular Pathology Laboratory, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Sridhar Epari
- Department of Pathology, Molecular Pathology Laboratory, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Sangeeta Desai
- Department of Pathology, Molecular Pathology Laboratory, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
5
|
Jöhrens K, Grassow M, Baretton G, Sperling F. [Interlaboratory comparisons-a central means of external quality assurance]. PATHOLOGIE (HEIDELBERG, GERMANY) 2022; 43:346-350. [PMID: 35948656 DOI: 10.1007/s00292-022-01102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
The precise performance of immunohistochemical and molecular examinations of diagnostic and predictive markers is essential for the further therapy of patients. Due to the increasing number of biomarkers and their detection at the immunohistochemical and molecular level in patient tissue, the pathology has a direct influence on the therapy of patients, which increases the value of external quality assurance (EQA). In pathology, various forms are available for this purpose, such as proficiency tests. The standards of both the certification and accreditation procedures of pathology require regular participation in EQA and a statement on the EQA policy of the institutes. The quality of an EQA depends on the scientific concept, the adequate selection of material, the evaluation concept, and the communication of results.
Collapse
Affiliation(s)
- Korinna Jöhrens
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.
- Qualitätssicherungs-Initiative Pathologie QuIP GmbH, Berlin, Deutschland.
| | - Maja Grassow
- Qualitätssicherungs-Initiative Pathologie QuIP GmbH, Berlin, Deutschland
| | - Gustavo Baretton
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland
| | - Florian Sperling
- Qualitätssicherungs-Initiative Pathologie QuIP GmbH, Berlin, Deutschland
| |
Collapse
|
6
|
The Significance of External Quality Assessment Schemes for Molecular Testing in Clinical Laboratories. Cancers (Basel) 2022; 14:cancers14153686. [PMID: 35954349 PMCID: PMC9367251 DOI: 10.3390/cancers14153686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/18/2022] Open
Abstract
Simple Summary Patients and clinicians often rely on the outcome of laboratory tests, but can we really trust these test results? Good quality management is key for laboratories to guarantee reliable test results. This review focusses on external quality assessment (EQA) schemes which are a tool for laboratories to examine and improve the quality of their testing routines. In this review, an overview of the role and importance of EQA schemes for clinical laboratories is given, and different types of EQA schemes and EQA providers available on the market are discussed, as well as recent developments in the EQA landscape. Abstract External quality assessment (EQA) schemes are a tool for clinical laboratories to evaluate and manage the quality of laboratory practice with the support of an independent party (i.e., an EQA provider). Depending on the context, there are different types of EQA schemes available, as well as various EQA providers, each with its own field of expertise. In this review, an overview of the general requirements for EQA schemes and EQA providers based on international guidelines is provided. The clinical and scientific value of these kinds of schemes for clinical laboratories, clinicians and patients are highlighted, in addition to the support EQA can provide to other types of laboratories, e.g., laboratories affiliated to biotech companies. Finally, recent developments and challenges in laboratory medicine and quality management, for example, the introduction of artificial intelligence in the laboratory and the shift to a more individual-approach instead of a laboratory-focused approach, are discussed. EQA schemes should represent current laboratory practice as much as possible, which poses the need for EQA providers to introduce latest laboratory innovations in their schemes and to apply up-to-date guidelines. By incorporating these state-of-the-art techniques, EQA aims to contribute to continuous learning.
Collapse
|
7
|
Dufraing K, Van Casteren K, Breyne J, D’Haene N, Van Campenhout C, Vander Borght S, Zwaenepoel K, Rouleau E, Schuuring E, von der Thüsen J, Dequeker E. Molecular pathology testing for non-small cell lung cancer: an observational study of elements currently present in request forms and result reports and the opinion of different stakeholders. BMC Cancer 2022; 22:736. [PMID: 35794532 PMCID: PMC9258204 DOI: 10.1186/s12885-022-09798-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND For patients with non-small cell lung cancer (NSCLC), targeted therapies are becoming part of the standard treatment. It is of question which information the clinicians provide on test requests and how the laboratories adapt test conclusions to this knowledge and regulations. METHODS This study consisted of two components; 1) checking the presence of pre-defined elements (administrative and key for therapy-choice) on completed requests and corresponding reports in Belgian laboratories, both for tissue- and liquid biopsy (LB)-testing and b) opinion analysis from Belgian pathologists/molecular biologists and clinicians during national pathology/oncology meetings. RESULTS Data from 4 out of 6 Belgian laboratories with ISO-accreditation for LB-testing were analyzed, of which 75% were university hospitals. On the scored requests (N = 4), 12 out of 19 ISO-required elements were present for tissue and 11 for LB-testing. Especially relevant patient history, such as line of therapy (for LB), tumor histology and the reason for testing were lacking. Similarly, 11 and 9 out of 18 elements were present in the reports (N = 4) for tissue and LB, respectively. Elements that pathologists/molecular biologists (N = 18) were missing on the request were the initial activating mutation, previous therapies, a clinical question and testing-related information. For reporting, an item considered important by both groups is the clinical interpretation of the test result. In addition, clinicians (N = 28) indicated that they also wish to read the percentage of neoplastic cells. CONCLUSIONS Communication flows between the laboratory and the clinician, together with possible pitfalls were identified. Based on the study results, templates for complete requesting and reporting were proposed.
Collapse
Affiliation(s)
- Kelly Dufraing
- Department of Public Health and Primary Care, University of Leuven, Biomedical Quality Assurance Research Unit, Leuven, Belgium
| | - Kaat Van Casteren
- Department of Public Health and Primary Care, University of Leuven, Biomedical Quality Assurance Research Unit, Leuven, Belgium
- Laboratory of Pathological Anatomy, Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research Antwerp (CORE), University of Antwerp, Wilrijk, Belgium
| | - Joke Breyne
- Department of Molecular Diagnostics, AZ Delta Roeselare Menen, Roeselare, Belgium
| | - Nicky D’Haene
- Department of Pathology, Erasme University Hospital, Brussels, Belgium
| | | | | | - Karen Zwaenepoel
- Laboratory of Pathological Anatomy, Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research Antwerp (CORE), University of Antwerp, Wilrijk, Belgium
| | - Etienne Rouleau
- Medical Biology and Pathology, Gustave Roussy, Paris, France
| | - Ed Schuuring
- Department of Pathology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Jan von der Thüsen
- Department of Pathology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Elisabeth Dequeker
- Department of Public Health and Primary Care, University of Leuven, Biomedical Quality Assurance Research Unit, Leuven, Belgium
| |
Collapse
|
8
|
Marchetti A, Barbareschi M, Barberis M, Buglioni S, Buttitta F, Fassan M, Fontanini G, Marchiò C, Papotti M, Pruneri G, Scarpa A, Stanta G, Tallini G, Troncone G, Veronese SM, Truini M, Sapino A. Real-World Data on NGS Diagnostics: a survey from the Italian Society of Pathology (SIAPeC) NGS Network. Pathologica 2021; 113:262-271. [PMID: 34463674 PMCID: PMC8488986 DOI: 10.32074/1591-951x-324] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 07/26/2021] [Indexed: 12/31/2022] Open
Abstract
Next Generation Sequencing (NGS) is increasingly used in diagnostic centers for the assessment of genomic alterations to select patients for precision oncology. The Italian Society of Anatomic Pathology and Diagnostic Cytopathology (SIAPEC) through the Molecular Pathology and Predictive Medicine Study Group (PMMP) has been following the progressive development of centers that have adopted NGS technology in diagnostics over time. In July 2017, a study network on massive parallel sequencing was activated in Italy and recognized as the NGS SIAPeC National Network by the SIAPeC Scientific Society Board. Since then, activities have been implemented within the network that provide for alignment of laboratories through diagnostic concordance analysis and monitoring of centers adhering to the Network. Recently, considering the growing need for extended genomic analyses, the PMMP distributed a national survey to assess activities related to the use of genomic diagnostics in oncology within the NGS SIAPEC National Network. Thirty centers participated in the survey. Eighty percent of the centers are laboratories within Pathology Departments. The distribution of laboratories in the country, the diagnostic laboratory/population ratio, the staff dedicated, the type and number of sequencing and mechatronics platforms available, the genomic panels utilized, and the type and number of diagnostic tests carried out in the last year in each center, are reported. The centers were also asked whether they participated in a multidisciplinary Molecular Tumor Board (MTB) for management of patients. Thirty percent of the centers had a MTB that was ratified by regional decree. The professionals most frequently involved in the core team of the MTB are the pathologist, oncologist, molecular biologist, geneticist, pharmacologist, and bioinformatician. The data from this survey indicate that NGS diagnostics in Italy is still heterogeneous in terms of geographical distribution and the characteristics of laboratories and diagnostic test performed. The implementation of activities that favors harmonization, the logistics and the convergence of biological material in reference centers for molecular analyses is a priority for the development of a functional laboratory network.
Collapse
Affiliation(s)
- Antonio Marchetti
- Laboratory of Molecular Diagnostics, Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, Italy
| | | | - Massimo Barberis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Lombardy, Italy
| | - Simonetta Buglioni
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fiamma Buttitta
- Laboratory of Molecular Diagnostics, Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, Italy
| | - Matteo Fassan
- Unit of Surgical Pathology, Department of Medicine (DIMED), University of Padua.,Veneto Institute of Oncology IRCCS, Padua, Italy
| | - Gabriella Fontanini
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Caterina Marchiò
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Mauro Papotti
- Pathology Unit, Department of Oncology, Città Della Salute e della Scienza Hospital, University of Turin, Turin, Italy
| | - Giancarlo Pruneri
- Department of Pathology and Laboratory Medicine, IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, and ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Giorgio Stanta
- DSM - Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Giovanni Tallini
- Department of Medicine (Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, DIMES), University of Bologna, Italy.,Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giancarlo Troncone
- Division of Pathology, Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | - Mauro Truini
- Pathological Anatomy Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Anna Sapino
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| |
Collapse
|
9
|
Keppens C, Boone E, Gameiro P, Tack V, Moreau E, Hodges E, Evans P, Brüggemann M, Carter I, Lenze D, Sarasquete ME, Möbs M, Liu H, Dequeker EMC, Groenen PJTA. Evaluation of a worldwide EQA scheme for complex clonality analysis of clinical lymphoproliferative cases demonstrates a learning effect. Virchows Arch 2021; 479:365-376. [PMID: 33686511 PMCID: PMC8364525 DOI: 10.1007/s00428-021-03046-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/18/2020] [Accepted: 01/28/2021] [Indexed: 11/26/2022]
Abstract
Clonality analysis of immunoglobulin (IG) or T-cell receptor (TR) gene rearrangements is routine practice to assist diagnosis of lymphoid malignancies. Participation in external quality assessment (EQA) aids laboratories in identifying systematic shortcomings. The aim of this study was to evaluate laboratories' improvement in IG/TR analysis and interpretation during five EQA rounds between 2014 and 2018. Each year, participants received a total of five cases for IG and five cases for TR testing. Paper-based cases were included for analysis of the final molecular conclusion that should be interpreted based on the integration of the individual PCR results. Wet cases were distributed for analysis of their routine protocol as well as evaluation of the final molecular conclusion. In total, 94.9% (506/533) of wet tests and 97.9% (829/847) of paper tests were correctly analyzed for IG, and 96.8% (507/524) wet tests and 93.2% (765/821) paper tests were correctly analyzed for TR. Analysis scores significantly improved when laboratories participated to more EQA rounds (p=0.001). Overall performance was significantly lower (p=0.008) for non-EuroClonality laboratories (95% for IG and 93% for TR) compared to EuroClonality laboratories (99% for IG and 97% for TR). The difference was not related to the EQA scheme year, anatomic origin of the sample, or final clinical diagnosis. This evaluation showed that repeated EQA participation helps to reduce performance differences between laboratories (EuroClonality versus non-EuroClonality) and between sample types (paper versus wet). The difficulties in interpreting oligoclonal cases highlighted the need for continued education by meetings and EQA schemes.
Collapse
Affiliation(s)
- Cleo Keppens
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Kapucijnenvoer 35 block d, 1st floor, box 7001, 3000 Leuven, Belgium
| | - Elke Boone
- AZ Delta vzw - Laboratorium Moleculaire Diagnostiek, Deltalaan 1, 8800 Roeselare, Belgium
| | - Paula Gameiro
- Laboratory of Hemato-Oncology, Portuguese Institute of Oncology of Lisbon, Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| | - Véronique Tack
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Kapucijnenvoer 35 block d, 1st floor, box 7001, 3000 Leuven, Belgium
| | - Elisabeth Moreau
- AZ Delta vzw - Laboratorium Moleculaire Diagnostiek, Deltalaan 1, 8800 Roeselare, Belgium
| | - Elizabeth Hodges
- Precision Medicine Centre, Queen’s University Belfast, Health Science Building, 97 Lisburn Road, Belfast, BT9 7AE UK
| | - Paul Evans
- HMDS, Leeds Institute of Oncology, St. James University Hospital, Level 3 Bexley Wing Leeds, Leeds, LS9 7TF UK
| | - Monika Brüggemann
- Department of Hematology, University Hospital Schleswig-Holstein, Langer Segen 8-10, 24105 Kiel, Germany
| | - Ian Carter
- Molecular Diagnostics, Histopathology, Nottingham University Hospitals NHS Trust, City Campus, Hucknall Rd., Nottingham, NG5 1PB UK
| | - Dido Lenze
- Institut für Pathologie, Molekularpathologie, Charité –Universitätsmedizin Berlin, Chariteplatz 1, 10117 Berlin, Germany
| | - Maria Eugenia Sarasquete
- Laboratorio Biología Molecular, Servicio de Hematología, Hospital Universitario de Salamanca, Paseo de San Vicente, 58-182, 37007 Salamanca, Spain
| | - Markus Möbs
- Institut für Pathologie, Molekularpathologie, Charité –Universitätsmedizin Berlin, Chariteplatz 1, 10117 Berlin, Germany
| | - Hongxiang Liu
- Molecular Malignancy Laboratory, Haematopathology and Oncology Diagnostic Service (HODS), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Box 234, Hills Road, Cambridge, CB2 0QQ UK
| | - Elisabeth M. C. Dequeker
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Kapucijnenvoer 35 block d, 1st floor, box 7001, 3000 Leuven, Belgium
| | - Patricia J. T. A. Groenen
- Department of Pathology, Radboud University Medical Centre Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
10
|
The evolving landscape of biomarker testing for non-small cell lung cancer in Europe. Lung Cancer 2021; 154:161-175. [PMID: 33690091 DOI: 10.1016/j.lungcan.2021.02.026] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 12/31/2022]
Abstract
The discovery of oncogenic driver mutations rendering non-small cell lung cancer (NSCLC) targetable by small-molecule inhibitors, and the development of immunotherapies, have revolutionised NSCLC treatment. Today, instead of non-selective chemotherapies, all patients with advanced NSCLC eligible for treatment (and increasing numbers with earlier, less extensive disease) require fast and comprehensive screening of biomarkers for first-line patient selection for targeted therapy, chemotherapy, or immunotherapy (with or without chemotherapy). To avoid unnecessary re-biopsies, biomarker screening before first-line treatment should also include markers that are actionable from second-line onwards; PD-L1 expression testing is also mandatory before initiating treatment. Population differences exist in the frequency of oncogenic driver mutations: EGFR mutations are more frequent in Asia than Europe, whereas the converse is true for KRAS mutations. In addition to approved first-line therapies, a number of emerging therapies are being investigated in clinical trials. Guidelines for biomarker testing vary by country, with the number of actionable targets and the requirement for extensive molecular screening strategies expected to increase. To meet diagnostic demands, rapid screening technologies for single-driver mutations have been implemented. Improvements in DNA- and RNA-based next-generation sequencing technologies enable analysis of a group of genes in one assay; however, turnaround times remain relatively long. Consequently, rapid screening technologies are being implemented alongside next-generation sequencing. Further challenges in the evolving landscape of biomarker testing in NSCLC are actionable primary and secondary resistance mechanisms to targeted therapies. Therefore, comprehensive testing on re-biopsies, collected at the time of disease progression, in combination with testing of circulating tumour DNA may provide important information to guide second- or third-line therapies. Furthermore, longitudinal biomarker testing can provide insights into tumour evolution and heterogeneity during the course of the disease. We summarise best practice strategies for Europe in the changing landscape of biomarker testing at diagnosis and during treatment.
Collapse
|
11
|
Cree IA, Indave Ruiz BI, Zavadil J, McKay J, Olivier M, Kozlakidis Z, Lazar AJ, Hyde C, Holdenrieder S, Hastings R, Rajpoot N, de la Fouchardiere A, Rous B, Zenklusen JC, Normanno N, Schilsky RL. The International Collaboration for Cancer Classification and Research. Int J Cancer 2021; 148:560-571. [PMID: 32818326 PMCID: PMC7756795 DOI: 10.1002/ijc.33260] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/21/2022]
Abstract
Gaps in the translation of research findings to clinical management have been recognized for decades. They exist for the diagnosis as well as the management of cancer. The international standards for cancer diagnosis are contained within the World Health Organization (WHO) Classification of Tumours, published by the International Agency for Research on Cancer (IARC) and known worldwide as the WHO Blue Books. In addition to their relevance to individual patients, these volumes provide a valuable contribution to cancer research and surveillance, fulfilling an important role in scientific evidence synthesis and international standard setting. However, the multidimensional nature of cancer classification, the way in which the WHO Classification of Tumours is constructed, and the scientific information overload in the field pose important challenges for the translation of research findings to tumour classification and hence cancer diagnosis. To help address these challenges, we have established the International Collaboration for Cancer Classification and Research (IC3 R) to provide a forum for the coordination of efforts in evidence generation, standard setting and best practice recommendations in the field of tumour classification. The first IC3 R meeting, held in Lyon, France, in February 2019, gathered representatives of major institutions involved in tumour classification and related fields to identify and discuss translational challenges in data comparability, standard setting, quality management, evidence evaluation and copyright, as well as to develop a collaborative plan for addressing these challenges.
Collapse
Affiliation(s)
- Ian A. Cree
- International Agency for Research on Cancer (IARC), World Health Organization (WHO)LyonFrance
| | | | - Jiri Zavadil
- International Agency for Research on Cancer (IARC), World Health Organization (WHO)LyonFrance
| | - James McKay
- International Agency for Research on Cancer (IARC), World Health Organization (WHO)LyonFrance
| | - Magali Olivier
- International Agency for Research on Cancer (IARC), World Health Organization (WHO)LyonFrance
| | - Zisis Kozlakidis
- International Agency for Research on Cancer (IARC), World Health Organization (WHO)LyonFrance
| | - Alexander J. Lazar
- Departments of Pathology, Genomic Medicine, and Translational Molecular PathologyThe University of Texas, MD Anderson Cancer CenterHoustonTexasUSA
| | - Chris Hyde
- Exeter Test GroupCollege of Medicine and Health, University of ExeterExeterUK
| | | | - Ros Hastings
- GenQA (Genomics External Quality Assessment)Women's Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation TrustOxfordUK
| | - Nasir Rajpoot
- Department of Computer ScienceUniversity of WarwickCoventryUK
- Alan Turing InstituteLondonUK
- Department of PathologyUniversity Hospitals Coventry & Warwickshire NHS TrustCoventryUK
| | | | - Brian Rous
- National Cancer Registration Service (Eastern Office), Public Health England, Victoria HouseCambridgeUK
| | - Jean Claude Zenklusen
- Center for Cancer GenomicsNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Nicola Normanno
- Cell Biology and Biotherapy UnitIstituto Nazionale Tumori—IRCCS—“Fondazione G. Pascale,” Via M. SemmolaNaplesItaly
| | | | | |
Collapse
|
12
|
Causes behind error rates for predictive biomarker testing: the utility of sending post-EQA surveys. Virchows Arch 2020; 478:995-1006. [PMID: 33225398 PMCID: PMC8099794 DOI: 10.1007/s00428-020-02966-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/14/2022]
Abstract
External quality assessment (EQA) schemes assess the performance of predictive biomarker testing in lung and colorectal cancer and have previously demonstrated variable error rates. No information is currently available on the underlying causes of incorrect EQA results in the laboratories. Participants in EQA schemes by the European Society of Pathology between 2014 and 2018 for lung and colorectal cancer were contacted to complete a survey if they had at least one analysis error or test failure in the provided cases. Of the 791 surveys that were sent, 325 were completed including data from 185 unique laboratories on 514 incorrectly analyzed or failed cases. For the digital cases and immunohistochemistry, the majority of errors were interpretation-related. For fluorescence in situ hybridization, problems with the EQA materials were reported frequently. For variant analysis, the causes were mainly methodological for lung cancer but variable for colorectal cancer. Post-analytical (clerical and interpretation) errors were more likely detected after release of the EQA results compared to pre-analytical and analytical issues. Accredited laboratories encountered fewer reagent problems and more often responded to the survey. A recent change in test methodology resulted in method-related problems. Testing more samples annually introduced personnel errors and lead to a lower performance in future schemes. Participation to quality improvement projects is important to reduce deviating test results in laboratories, as the different error causes differently affect the test performance. EQA providers could benefit from requesting root cause analyses behind errors to offer even more tailored feedback, subschemes, and cases.
Collapse
|
13
|
Delcourt T, Vanneste K, Soumali MR, Coucke W, Ghislain V, Hebrant A, Van Valckenborgh E, De Keersmaecker SCJ, Roosens NH, Van De Walle P, Van Den Bulcke M, Antoniou A. NGS for (Hemato-) Oncology in Belgium: Evaluation of Laboratory Performance and Feasibility of a National External Quality Assessment Program. Cancers (Basel) 2020; 12:E3180. [PMID: 33138022 PMCID: PMC7692129 DOI: 10.3390/cancers12113180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 01/08/2023] Open
Abstract
Next-generation sequencing (NGS) is being integrated into routine clinical practice in the field of (hemato-) oncology to search for variants with diagnostic, prognostic, or therapeutic value at potentially low allelic frequencies. The complex sequencing workflows used require careful validation and continuous quality control. Participation in external quality assessments (EQA) helps laboratories evaluate their performance and guarantee the validity of tests results with the ultimate goal of ensuring high-quality patient care. Here, we describe three benchmarking trials performed during the period 2017-2018 aiming firstly at establishing the state-of-the-art and secondly setting up a NGS-specific EQA program at the national level in the field of clinical (hemato-) oncology in Belgium. DNA samples derived from cell line mixes and artificially mutated cell lines, designed to carry variants of clinical relevance occurring in solid tumors, hematological malignancies, and BRCA1/BRCA2 genes, were sent to Belgian human genetics, anatomic pathology, and clinical biology laboratories, to be processed following routine practices, together with surveys covering technical aspects of the NGS workflows. Despite the wide variety of platforms and workflows currently applied in routine clinical practice, performance was satisfactory, since participating laboratories identified the targeted variants with success rates ranging between 93.06% and 97.63% depending on the benchmark, and few false negative or repeatability issues were identified. However, variant reporting and interpretation varied, underlining the need for further standardization. Our approach showcases the feasibility of developing and implementing EQA for routine clinical practice in the field of (hemato-) oncology, while highlighting the challenges faced.
Collapse
Affiliation(s)
- Thomas Delcourt
- Transversal activities in Applied Genomics, Sciensano, 1050 Brussels, Belgium; (T.D.); (K.V.); (S.C.J.D.K.); (N.H.R.)
| | - Kevin Vanneste
- Transversal activities in Applied Genomics, Sciensano, 1050 Brussels, Belgium; (T.D.); (K.V.); (S.C.J.D.K.); (N.H.R.)
| | - Mohamed Rida Soumali
- Quality of Laboratories, Sciensano, 1050 Brussels, Belgium; (M.R.S.); (W.C.); (V.G.); (P.V.D.W.)
| | - Wim Coucke
- Quality of Laboratories, Sciensano, 1050 Brussels, Belgium; (M.R.S.); (W.C.); (V.G.); (P.V.D.W.)
| | - Vanessa Ghislain
- Quality of Laboratories, Sciensano, 1050 Brussels, Belgium; (M.R.S.); (W.C.); (V.G.); (P.V.D.W.)
| | - Aline Hebrant
- Cancer Centre, Sciensano, 1050 Brussels, Belgium; (A.H.); (E.V.V.); (M.V.D.B.)
| | | | - Sigrid C. J. De Keersmaecker
- Transversal activities in Applied Genomics, Sciensano, 1050 Brussels, Belgium; (T.D.); (K.V.); (S.C.J.D.K.); (N.H.R.)
| | - Nancy H. Roosens
- Transversal activities in Applied Genomics, Sciensano, 1050 Brussels, Belgium; (T.D.); (K.V.); (S.C.J.D.K.); (N.H.R.)
| | - Philippe Van De Walle
- Quality of Laboratories, Sciensano, 1050 Brussels, Belgium; (M.R.S.); (W.C.); (V.G.); (P.V.D.W.)
| | - Marc Van Den Bulcke
- Cancer Centre, Sciensano, 1050 Brussels, Belgium; (A.H.); (E.V.V.); (M.V.D.B.)
| | - Aline Antoniou
- Quality of Laboratories, Sciensano, 1050 Brussels, Belgium; (M.R.S.); (W.C.); (V.G.); (P.V.D.W.)
| |
Collapse
|
14
|
Keppens C, Schuuring E, Dequeker EMC. Managing Deviating EQA Results: A Survey to Assess the Corrective and Preventive Actions of Medical Laboratories Testing for Oncological Biomarkers. Diagnostics (Basel) 2020; 10:E837. [PMID: 33080995 PMCID: PMC7603102 DOI: 10.3390/diagnostics10100837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/24/2022] Open
Abstract
Laboratories testing predictive biomarkers in lung and colorectal cancer are advised to participate in external quality assessment (EQA) schemes. This study aimed to investigate which corrective actions were taken by laboratories if predetermined performance criteria were not met, to ultimately improve current test practices. EQA participants from the European Society of Pathology between 2014 and 2018 for lung and colorectal cancer were contacted, if they had at least one analysis error or test failure in the provided cases, to complete a survey. For 72.4% of 514 deviating EQA results, an appropriate action was performed, most often including staff training (15.2%) and protocol revisions (14.6%). Main assigned persons were the molecular biologist (40.0%) and pathologist (46.5%). A change in test method or the use of complex techniques, such as next-generation sequencing, required more training and the involvement of dedicated personnel to reduce future test failures. The majority of participants adhered to ISO 15189 and implemented suitable actions by designated staff, not limited to accredited laboratories. However, for 27.6% of cases (by 20 laboratories) no corrective action was taken, especially for pre-analytic problems and complex techniques. The surveys were feasible to request information on results follow-up and further recommendations were provided.
Collapse
Affiliation(s)
- Cleo Keppens
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Kapucijnenvoer 35d, 3000 Leuven, Belgium;
| | - Ed Schuuring
- Department of Pathology, University Medical Center Groningen (UMCG), University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands;
| | - Elisabeth MC Dequeker
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Kapucijnenvoer 35d, 3000 Leuven, Belgium;
| |
Collapse
|
15
|
Dufraing K, Fenizia F, Torlakovic E, Wolstenholme N, Deans ZC, Rouleau E, Vyberg M, Parry S, Schuuring E, Dequeker EMC. Biomarker testing in oncology - Requirements for organizing external quality assessment programs to improve the performance of laboratory testing: revision of an expert opinion paper on behalf of IQNPath ABSL. Virchows Arch 2020; 478:553-565. [PMID: 33047156 PMCID: PMC7550230 DOI: 10.1007/s00428-020-02928-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/16/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022]
Abstract
In personalized medicine, predictive biomarker testing is the basis for an appropriate choice of therapy for patients with cancer. An important tool for laboratories to ensure accurate results is participation in external quality assurance (EQA) programs. Several providers offer predictive EQA programs for different cancer types, test methods, and sample types. In 2013, a guideline was published on the requirements for organizing high-quality EQA programs in molecular pathology. Now, after six years, steps were taken to further harmonize these EQA programs as an initiative by IQNPath ABSL, an umbrella organization founded by various EQA providers. This revision is based on current knowledge, adds recommendations for programs developed for predictive biomarkers by in situ methodologies (immunohistochemistry and in situ hybridization), and emphasized transparency and an evidence-based approach. In addition, this updated version also has the aim to give an overview of current practices from various EQA providers.
Collapse
Affiliation(s)
- K Dufraing
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35 blok d, 3000, Leuven, Belgium
| | - F Fenizia
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - E Torlakovic
- Department of Pathology and Laboratory Medicine, Royal University Hospital, College of Medicine, University of Saskatchewan and Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada
| | - N Wolstenholme
- European Molecular Quality Network (EMQN), Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester, M13 9WL, UK
| | - Z C Deans
- UK NEQAS for Molecular Genetics, Department of Laboratory Medicine, Royal Infirmary of Edinburgh, Little France Crescent, Edinburgh, EH16 4SA, UK
| | - E Rouleau
- Department of Medical Biology and Pathology, Gustave Roussy, Cancer Genetics Laboratory, Gustave Roussy, Villejuif, France
| | - M Vyberg
- NordiQC, Institute of Pathology, Aalborg University Hospital, Aalborg, Denmark
| | - S Parry
- UK NEQAS ICC & ISH, University College London Cancer Institute, London, UK
| | - E Schuuring
- Department of Pathology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30001, 9700, RB, Groningen, The Netherlands
| | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35 blok d, 3000, Leuven, Belgium.
| |
Collapse
|
16
|
Keppens C, Dequeker EMC, Rouleau E, 't Hart N, Bubendorf L, Dufraing K, Garrec C, Guéguen P, Lamy A, Marchetti A, Pauwels P, Ryska A, Tack V, Tornillo L, Van Casteren K, von der Thüsen JH, Zwaenepoel K, Lissenberg-Witte B, Thunnissen E, Schuuring E. Sensitive detection methods are key to identify secondary EGFR c.2369C>T p.(Thr790Met) in non-small cell lung cancer tissue samples. BMC Cancer 2020; 20:366. [PMID: 32357863 PMCID: PMC7193365 DOI: 10.1186/s12885-020-06831-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/06/2020] [Indexed: 01/15/2023] Open
Abstract
Background Correct identification of the EGFR c.2369C>T p.(Thr790Met) variant is key to decide on a targeted therapeutic strategy for patients with acquired EGFR TKI resistance in non-small cell lung cancer. The aim of this study was to evaluate the correct detection of this variant in 12 tumor tissue specimens tested by 324 laboratories participating in External Quality Assessment (EQA) schemes. Methods Data from EQA schemes were evaluated between 2013 and 2018 from cell lines (6) and resections (6) containing the EGFR c.2369C>T p.(Thr790Met) mutation. Adequate performance was defined as the percentage of tests for which an outcome was available and correct. Additional data on the used test method were collected from the participants. Chi-squared tests on contingency tables and a biserial rank correlation were applied by IBM SPSS Statistics version 25 (IBM, Armonk, NY, USA). Results In 26 of the 1190 tests (2.2%) a technical failure occurred. For the remaining 1164 results, 1008 (86.6%) were correct, 151 (12.9%) were false-negative and 5 (0.4%) included incorrect mutations. Correct p.(Thr790Met) detection improved over time and for repeated scheme participations. In-house non-next-generation sequencing (NGS) techniques performed worse (81.1%, n = 293) compared to non-NGS commercial kits (85.2%, n = 656) and NGS (97.0%, n = 239). Over time there was an increase in the users of NGS. Resection specimens performed worse (82.6%, n = 610 tests) compared to cell line material (90.9%, n = 578 tests), except for NGS (96.3%, n = 344 for resections and 98.6%, n = 312 for cell lines). Samples with multiple mutations were more difficult compared to samples with the single p.(Thr790Met) variant. A change of the test method was shown beneficial to reduce errors but introduced additional analysis failures. Conclusions A significant number of laboratories that offer p.(Thr790Met) testing did not detect this relevant mutation compared to the other EQA participants. However, correct identification of this variant is improving over time and was higher for NGS users. Revising the methodology might be useful to resolve errors, especially for resection specimens with low frequency or multiple variants. EQA providers should include challenging resections in the scheme.
Collapse
Affiliation(s)
- Cleo Keppens
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Elisabeth M C Dequeker
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Etienne Rouleau
- Service de Génétique des Tumeurs, Gustave Roussy, Villejuif Cedex, France
| | - Nils 't Hart
- Department of Pathology, University of Groningen, University Medical Center Groningen (UMCG), Hanzeplein 1, PO Box 30001, 9700, RB, Groningen, the Netherlands.,Department of Pathology, Isala, Zwolle, The Netherlands
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Kelly Dufraing
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Céline Garrec
- Institut de Biologie, CHU Hôtel Dieu, Laboratoire de Génétique Moléculaire, Nantes Cedex 1, France
| | - Paul Guéguen
- CHRU Brest/Hôpital Morvan, Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Brest, France
| | - Aude Lamy
- CHU de Rouen / Hôpital Charles Nicolle, laboratoire de génétique somatique des tumeurs, Rouen Cedex, France
| | - Antonio Marchetti
- Laboratory of Molecular Diagnostics, Center for Advanced Studies and Technology, University of Chieti, 66100, Chieti, Italy
| | - Patrick Pauwels
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium.,Centre for Oncological Research (CORE), University of Antwerp, Edegem, Belgium
| | - Ales Ryska
- Department of Pathology, Charles University Medical Faculty Hospital, Hradec Kralove, Czech Republic
| | - Véronique Tack
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Luigi Tornillo
- Institute of Pathology, University Hospital Basel, Basel, Switzerland.,GILAB, Allschwil, AG, Switzerland
| | - Kaat Van Casteren
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium.,Department of Pathology, University Hospital Antwerp, Edegem, Belgium.,Centre for Oncological Research (CORE), University of Antwerp, Edegem, Belgium
| | - Jan H von der Thüsen
- Department of pathology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Karen Zwaenepoel
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium.,Centre for Oncological Research (CORE), University of Antwerp, Edegem, Belgium
| | - Birgit Lissenberg-Witte
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Biostatistics, Amsterdam, The Netherlands
| | - Erik Thunnissen
- Department of pathology, VU University Medical Center (VUMC) Amsterdam, Amsterdam, the Netherlands
| | - Ed Schuuring
- Department of Pathology, University of Groningen, University Medical Center Groningen (UMCG), Hanzeplein 1, PO Box 30001, 9700, RB, Groningen, the Netherlands.
| |
Collapse
|
17
|
Peng R, Zhang R, Zhang J, Tan P, Han Y, Zhang K, Lin G, Xie J, Li J. Continual Improvement of the Reliability of EML4-ALK Rearrangement Detection in Non-Small-Cell Lung Cancer: A Long-Term Comparison of ALK Detection in China. J Mol Diagn 2020; 22:876-884. [PMID: 32302779 DOI: 10.1016/j.jmoldx.2020.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/24/2019] [Accepted: 03/30/2020] [Indexed: 12/01/2022] Open
Abstract
The results of EML4-ALK testing are critical to manage ALK tyrosine kinase receptor inhibitor treatment. Thus, the accurate detection of ALK rearrangement is increasingly becoming a matter of serious concern. To address this issue, a long-term EML4-ALK proficiency testing (PT) scheme was launched in China in 2015, serving as an educational tool for assessing and improving the testing quality of EML4-ALK fusion detection. Responses across 20 different PT samples interrogating three different variants and wild-type samples were collected between 2015 and 2019. Performance was analyzed by evaluating the detection methods, kits, and pre-analytic practices used to further display the landscape of changing conditions of the reliability of EML4-ALK testing. During the 5 years, 3224 results reported from 988 laboratories were evaluated, with an overall error rate of 5.36%. Along with an increasing number of participating laboratories, the error rate within each of the different methods showed a significantly downward trend over the years. No obvious differences in the error rates were found regarding the testing methods or kit manufacturers. Moreover, the individual performance of the laboratories improved when they participated in more PT scheme rounds. The data demonstrated that the performance of individual Chinese laboratories for EML4-ALK testing continuously improved over time by participating PT schemes, regardless of their method. However, care must be taken in standardized operations and validations.
Collapse
Affiliation(s)
- Rongxue Peng
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Rui Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Jiawei Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China; Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ping Tan
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China; Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanxi Han
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Kuo Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Guigao Lin
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Jiehong Xie
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China; Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
18
|
Castillo P, Marginet M, Jares P, García M, Gonzalvo E, Arance A, García A, Alos L, Teixido C. Implementation of an NGS panel for clinical practice in paraffin-embedded tissue samples from locally advanced and metastatic melanoma patients. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:101-108. [PMID: 36046072 PMCID: PMC9400780 DOI: 10.37349/etat.2020.00006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 12/22/2022] Open
Abstract
Aim: Single biomarker diagnostic test of BRAFV600 locus in metastatic melanoma is mandatory for treatment decision; however, multiple-gene based techniques, such as targeted next-generation sequencing (NGS) are being used to maximize the number of patients that can benefit from a targeted therapy. The main objective of this study is to investigate whether an NGS panel could be adopted in routine clinical care for advanced melanoma. Methods: Patients diagnosed with advanced melanoma at our center from 2017 to 2019 were included. Presence of genetic alterations was performed using two methods: real-time polymerase chain reaction-based Idylla test (Biocartis) and NGS with the oncomine solid tumor DNA kit (Thermo Fisher Scientific). Total genomic DNA was extracted from formalin-fixed and paraffin embedded samples for sequencing. Results: A total of 155 samples were evaluated for molecular analysis but 40 samples (25.8%) were inadequate for sequencing. The clinical utility of BRAFV600 real-time polymerase chain reaction and targeted-NGS was compared in 29 samples and a very good concordance was observed (Kappa = 0.89, 95% confidence interval 0.68 ± 1.05). An oncogenic mutation by NGS was found in 75 samples (65%)–53% of whom were candidates for personalized therapies. The most prevalent mutated genes were BRAF (39%), TP53 (23%), and NRAS (14%). Other genes identified at lower incidence (< 5%) were: PIK3CA, ERBB4, CTNNB1, STK11, FGFR1, SMAD4, KRAS, FGFR3, PTEN and AKT. Co-occurrence of oncogenic mutations was detected in 40% of the samples. Among the mutations identified, TP53 was significantly more prevalent in men (men 31.8% versus women 12.2%, P = 0.03) and NRAS in women (men 9.1% versus women 24.4%, P = 0.03). Conclusions: Targeted-NGS testing is a feasible technique to implement in the routine clinical practice. Based on our results, NGS has provided more information on target-genes than RT-PCR technique, maximizing the benefit for patients with advanced melanoma.
Collapse
Affiliation(s)
- Paola Castillo
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Marta Marginet
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Pedro Jares
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Mireia García
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Elena Gonzalvo
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Ana Arance
- Department of Medical Oncology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Adriana García
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Llucia Alos
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Cristina Teixido
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
19
|
External Quality Assessment Schemes for Biomarker Testing in Oncology: Comparison of Performance between Formalin-Fixed, Paraffin-Embedded-Tissue and Cell-Free Tumor DNA in Plasma. J Mol Diagn 2020; 22:736-747. [PMID: 32205291 DOI: 10.1016/j.jmoldx.2020.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 02/08/2020] [Accepted: 02/20/2020] [Indexed: 01/06/2023] Open
Abstract
Liquid biopsies have emerged as a useful addition to tissue biopsies in molecular pathology. Literature has shown lower laboratory performances when a new method of variant analysis is introduced. This study evaluated the differences in variant analysis between tissue and plasma samples after the introduction of liquid biopsy in molecular analysis. Data from a pilot external quality assessment scheme for the detection of molecular variants in plasma samples and from external quality assessment schemes for the detection of molecular variants in tissue samples were collected. Laboratory performance and error rates by sample were compared between matrices for variants present in both scheme types. Results showed lower overall performance [65.6% (n = 276) versus 89.2% (n = 1607)] and higher error rates [21.0% to 43.5% (n = 138) versus 8.7% to 16.7% (n = 234 to 689)] for the detection of variants in plasma compared to tissue, respectively. In the plasma samples, performance was decreased for variants with an allele frequency of 1% compared to 5% [56.5% (n = 138) versus 74.6% (n = 138)]. The implementation of liquid biopsy in the detection of circulating tumor DNA in plasma was associated with poor laboratory performance. It is important both to apply optimal detection methods and to extensively validate new methods for testing circulating tumor DNA before treatment decisions are made.
Collapse
|
20
|
Dufraing K, Keppens C, Tack V, Siebers AG, Kafatos G, Dube S, Demonty G, Lowe K, Kroeze LI, Ligtenberg M, Normanno N, Tembuyser L, Sara VB, van Krieken JH, C Dequeker EM. Evolution of RAS testing over time: factors influencing mutation rates in metastatic colorectal cancer patients. COLORECTAL CANCER 2020. [DOI: 10.2217/crc-2019-0013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Aim: Correct identification of RAS gene variants is key for targeted treatment decisions in patients with metastatic colorectal cancer. Published RAS mutation rates differ and could be influenced by several factors including testing methods. This study aimed to describe the performance of laboratories to correctly identify RAS variants over time and to understand how RAS testing has evolved in Europe. Materials & methods: Misclassification and test failure rates were calculated and related to the used test methodology for 239 unique laboratories participating in external quality assessment for metastatic colorectal cancer between 2013 and 2018. In addition, 33 laboratories completed a survey aiming to obtain more details on their routine testing strategies, number of samples analyzed and RAS mutation rates between 2013 and 2017. Results: The mutation status was correctly analyzed in 96.1% (N = 5471) RAS and BRAF tests. A total of 4.6% (N = 2860) RAS tests included false-negative results. In 1.6% (N = 5562) RAS and BRAF tests, an analysis failure occurred. Misclassifications and technical failures both decreased between 2013 and 2018. The number of next-generation sequencing users increased from 6.9% (N = 130) in 2013 to 44.6% (N = 112) in 2018. Over time, more codons were included in the methodologies, yet 23.2% (N = 112) did not offer full RAS testing (exon 2, 3, 4) in 2018. Based on the survey the overall RAS mutation rate was estimated as 45.2% (N = 27,325). Conclusion: This is the largest observational study reporting RAS mutation rates to-date. There was no trend of RAS mutation rates over time despite having a clear shift to more sensitive tests and increased quality of testing.
Collapse
Affiliation(s)
- Kelly Dufraing
- Biomedical Quality Assurance Research Unit, Department of Public Health & Primary Care, University of Leuven, Leuven, Belgium
| | - Cleo Keppens
- Biomedical Quality Assurance Research Unit, Department of Public Health & Primary Care, University of Leuven, Leuven, Belgium
| | - Véronique Tack
- Biomedical Quality Assurance Research Unit, Department of Public Health & Primary Care, University of Leuven, Leuven, Belgium
| | - Albert Gerrit Siebers
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | | | | | - Leonie Ilse Kroeze
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjolijn Ligtenberg
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicola Normanno
- Cell Biology & Biotherapy Unit, Istituto Nazionale Tumori – Fondazione Pascale, Naples, Italy
| | - Lien Tembuyser
- Biomedical Quality Assurance Research Unit, Department of Public Health & Primary Care, University of Leuven, Leuven, Belgium
| | - Vander Borght Sara
- Pathologische Ontleedkunde, Department of Pathology, University Hospitals Leuven, Belgium
| | | | - Elisabeth Marie C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health & Primary Care, University of Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Pu T, Shui R, Shi J, Liang Z, Yang W, Bu H, Li Q, Zhang Z. External quality assessment (EQA) program for the immunohistochemical detection of ER, PR and Ki-67 in breast cancer: results of an interlaboratory reproducibility ring study in China. BMC Cancer 2019; 19:978. [PMID: 31640622 PMCID: PMC6805628 DOI: 10.1186/s12885-019-6210-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 09/26/2019] [Indexed: 02/05/2023] Open
Abstract
Background An External Quality Assessment (EQA) program was developed to investigate the status of estrogen receptor (ER), progesterone receptor (PR), and Ki-67 immunohistochemical (IHC) detection in breast cancer and to evaluate the reproducibility of staining and interpretation in 44 pathology laboratories in China. Methods This program was implemented through three specific steps. In study I, three revising centres defined the reference value for 11 sections. In study II, 41 participating centres (PC) stained and interpreted 11 sections by their own daily practice IHC protocols. In study III, all cases received second interpretation opinions. Results The stained slides of 44 laboratories were up to the interpretation standard. The overall interpretation concordance rate of this study was over 90%. A perfect agreement was reached among the PCs for the cases with ER+ and PR+ > 50% and Ki-67 > 30%, whereas a moderate agreement was observed for intermediate categories. After second interpretations, the misclassification rates for ER were reduced by 12.20%, for PR were reduced by 17.07%, and for Ki-67 were reduced by 4.88%. Up to 31 PCs observed a benefit from the second opinion strategy. Conclusions This project is the first EQA study performed on a national scale for assessment of ER, PR and Ki-67 status by IHC in China. In the whole IHC evaluation process, the intermediate categories were less reproducible than those with high expression rates. Second opinions can significantly improve the diagnostic agreement of pathologists’ interpretations.
Collapse
Affiliation(s)
- Tianjie Pu
- Department of Pathology, West China Hospital, Sichuan University, Guo Xue Xiang 37#, Chengdu, 610041, Sichuan, China.,Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruohong Shui
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Jie Shi
- Department of Pathology, Peking Union Medical College Hospital, China Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhiyong Liang
- Department of Pathology, Peking Union Medical College Hospital, China Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wentao Yang
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Guo Xue Xiang 37#, Chengdu, 610041, Sichuan, China.,Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qin Li
- Department of Hospital Infection Control, Women's and Children's Hospital of Sichuan Province, Chengdu, China
| | - Zhang Zhang
- Department of Pathology, West China Hospital, Sichuan University, Guo Xue Xiang 37#, Chengdu, 610041, Sichuan, China.
| | | |
Collapse
|
22
|
Rajadurai P, Cheah PL, How SH, Liam CK, Annuar MAA, Omar N, Othman N, Marzuki NM, Pang YK, Bustamam RSA, Tho LM. Molecular testing for advanced non-small cell lung cancer in Malaysia: Consensus statement from the College of Pathologists, Academy of Medicine Malaysia, the Malaysian Thoracic Society, and the Malaysian Oncological Society. Lung Cancer 2019; 136:65-73. [DOI: 10.1016/j.lungcan.2019.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/13/2019] [Accepted: 08/02/2019] [Indexed: 12/18/2022]
|
23
|
Keppens C, Tack V, Dufraing K, Rouleau E, Ligtenberg MJL, Schuuring E, Dequeker EMC. Variation in nomenclature of somatic variants for selection of oncological therapies: Can we reach a consensus soon? Hum Mutat 2019; 41:7-16. [PMID: 31553104 PMCID: PMC6973115 DOI: 10.1002/humu.23926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 09/05/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022]
Abstract
A standardized nomenclature for reporting oncology biomarker variants is key to avoid misinterpretation of results and unambiguous registration in clinical databases. External quality assessment (EQA) schemes have revealed a need for more consistent nomenclature use in clinical genetics. We evaluated the propensity of EQA for improvement of compliance with Human Genome Variation Society (HGVS) recommendations for reporting of predictive somatic variants in lung and colorectal cancer. Variant entries between 2012 and 2018 were collected from written reports and electronic results sheets. In total, 4,053 variants were assessed, of which 12.1% complied with HGVS recommendations. Compliance improved over time from 2.1% (2012) to 22.3% (2018), especially when laboratories participated in multiple EQA schemes. Compliance was better for next-generation sequencing (20.9%) compared with targeted techniques (9.8%). In the 1792 reports, HGVS recommendations for reference sequences were met for 31.9% of reports, for 36.0% of noncommercial, and 26.5% of commercial test methods. Compliance improved from 16.7% (2012) to 33.1% (2018), and after repeated EQA participation. EQA participation improves compliance with HGVS recommendations. The residual percentage of errors in the most recent schemes suggests that laboratories, companies, and EQA providers need to collaborate for additional improvement of harmonization in clinical test reporting.
Collapse
Affiliation(s)
- Cleo Keppens
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Véronique Tack
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Kelly Dufraing
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Etienne Rouleau
- Gustave Roussy, Service de Génétique/Pathologie Moléculaire, Villejuif Cedex, France
| | - Marjolijn J L Ligtenberg
- Department of Pathology and Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ed Schuuring
- University Medical Center Groningen, Department of Pathology, University of Groningen, Groningen, The Netherlands
| | - Elisabeth M C Dequeker
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Fernandes Marques J, Pereira Reis J, Fernandes G, Hespanhol V, Machado JC, Costa JL. Circulating Tumor DNA: A Step into the Future of Cancer Management. Acta Cytol 2019; 63:456-465. [PMID: 30852572 DOI: 10.1159/000492917] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022]
Abstract
Liquid biopsy was introduced to the oncology field with the promise of revolutionizing the management of cancer patients, minimizing the exposure to invasive procedures such as tissue biopsy, and providing reliable information regarding therapy response and detection of disease relapse. Despite the significant increase in the number of published studies on circulating tumor DNA (ctDNA) in the past years, the emphasis of most studies is on the development of new technologies or on the clinical utility of ctDNA. This leaves a clear gap of knowledge concerning the biology of ctDNA, such as the fundamental mechanisms through which DNA from tumor cells is released into the circulation. Moreover, considering that ctDNA analysis is now currently being applied in clinical practice, the need for rigorous quality control is arising, and with it the necessity to standardize procedures, from sample collection to data analysis. This review focuses on the main aspects of ctDNA, including approaches currently available to evaluate tumor genetics, as well as the points that still require improvement in order to make liquid biopsy a key player in precision medicine.
Collapse
Affiliation(s)
- Joana Fernandes Marques
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Joana Pereira Reis
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Gabriela Fernandes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Department of Pulmonology, Hospital de São João, Porto, Portugal
| | - Venceslau Hespanhol
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Department of Pulmonology, Hospital de São João, Porto, Portugal
| | - José Carlos Machado
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - José Luís Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,
- Faculty of Medicine, University of Porto, Porto, Portugal,
| |
Collapse
|
25
|
Keppens C, Dufraing K, van Krieken HJ, Siebers AG, Kafatos G, Lowe K, Demonty G, Dequeker EMC. European follow-up of incorrect biomarker results for colorectal cancer demonstrates the importance of quality improvement projects. Virchows Arch 2019; 475:25-37. [PMID: 30719547 PMCID: PMC6611891 DOI: 10.1007/s00428-019-02525-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/05/2018] [Accepted: 01/10/2019] [Indexed: 01/09/2023]
Abstract
Biomarker analysis for colorectal cancer has been shown to be reliable in Europe with 97% of samples tested by EQA participants to be correctly classified. This study focuses on errors during the annual EQA assessment. The aim was to explore the causes and actions related to the observed errors and to provide feedback and assess any improvement between 2016 and 2017. An electronic survey was sent to all laboratories with minimum one genotyping error or technical failure on ten tumor samples. A workshop was organized based on 2016 survey responses. Improvement of performance in 2017 was assessed for returning participants (n = 76), survey respondents (n = 13) and workshop participants (n = 4). Survey respondents and workshop participants improved in terms of (maximum) analysis score, successful participation, and genotyping errors compared to all returning participants. In 2016, mostly pre- and post-analytical errors (both 25%) were observed caused by unsuitability of the tumor tissue for molecular analysis. In 2017, most errors were due to analytical problems (50.0%) caused by methodological problems. The most common actions taken (n = 58) were protocol revisions (34.5%) and staff training (15.5%). In 24.1% of issues identified no action was performed. Corrective actions were linked to an improved performance, especially if performed by the pathologist. Although biomarker testing has improved over time, error occurrence at different phases stresses the need for quality improvement throughout the test process. Participation to quality improvement projects and a close collaboration with the pathologist can have a positive influence on performance.
Collapse
Affiliation(s)
- Cleo Keppens
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Kapucijnenvoer 35 block d, 1st floor, box 7001, 3000 Leuven, Belgium
| | - Kelly Dufraing
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Kapucijnenvoer 35 block d, 1st floor, box 7001, 3000 Leuven, Belgium
| | - Han J. van Krieken
- Department of Pathology, Radboud University Medical Center, Geert Grooteplein 10 (route 812), P.O.Box 9101, 6500 HB Nijmegen (824), The Netherlands
| | - Albert G. Siebers
- Department of Pathology, Radboud University Medical Center, Geert Grooteplein 10 (route 812), P.O.Box 9101, 6500 HB Nijmegen (824), The Netherlands
| | - George Kafatos
- Amgen Ltd, 1 Uxbridge Business Park, Sanderson Road, Uxbridge, UB8 1DH UK
| | - Kimberly Lowe
- Amgen Inc, One Amgen Center Drive, MS 17-2-A, Thousand Oaks, CA 91320 USA
| | - Gaston Demonty
- Amgen Belgium S.A./N.V, Arianelaan 5, 1200 Brussels, Belgium
| | - Elisabeth M. C. Dequeker
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Kapucijnenvoer 35 block d, 1st floor, box 7001, 3000 Leuven, Belgium
| |
Collapse
|
26
|
Tack V, Schuuring E, Keppens C, Hart N', Pauwels P, van Krieken H, Dequeker EMC. Accreditation, setting and experience as indicators to assure quality in oncology biomarker testing laboratories. Br J Cancer 2018; 119:605-614. [PMID: 30140047 PMCID: PMC6162254 DOI: 10.1038/s41416-018-0204-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/15/2018] [Accepted: 07/09/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Predictive biomarkers allow clinicians to optimise cancer treatment decisions. Therefore, molecular biomarker test results need to be accurate and swiftly available. To ensure quality of oncology biomarker testing, external quality assessments (EQA) for somatic variant analyses were organised. This study hypothesised whether laboratory characteristics influence the performance of laboratories and whether these can be imposed before authorisation of biomarker testing. METHODS Longitudinal EQA data from the European Society of Pathology were available over six (metastatic colorectal cancer) and four years (non-small cell lung cancer), including the percentage of analysis errors and technical failures, and information on laboratory characteristics (accreditation status, laboratory setting, number of samples analysed and detection method). Statistical models for repeated measurements were used to analyse the association between the EQA results and the laboratory characteristics. RESULTS Laboratory accreditation was associated with fewer analysis errors in early stages of biomarker introduction into the laboratory. Analysing more samples, or university and research laboratories showed better performance. Changing the detection method did not have an effect. CONCLUSION The indicators support the clinicians in choosing molecular pathology laboratories by improving quality assurance and guaranteeing patient safety. Accreditation of laboratories, centralisation of biomarker testing or a university and research setting should be stimulated.
Collapse
Affiliation(s)
- Véronique Tack
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35, 3000, Leuven, Belgium
| | - Ed Schuuring
- Department of Pathology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Cleo Keppens
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35, 3000, Leuven, Belgium
| | - Nils 't Hart
- Department of Pathology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Patrick Pauwels
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Han van Krieken
- Department of Pathology, Radboud University Medical Center, Geert Grooteplein zuid 10, Huispost 824, 6525, Nijmegen, The Netherlands
| | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35, 3000, Leuven, Belgium.
| |
Collapse
|
27
|
Tack V, Spans L, Schuuring E, Keppens C, Zwaenepoel K, Pauwels P, Van Houdt J, Dequeker EMC. Describing the Reportable Range Is Important for Reliable Treatment Decisions: A Multiple Laboratory Study for Molecular Tumor Profiling Using Next-Generation Sequencing. J Mol Diagn 2018; 20:743-753. [PMID: 30055348 DOI: 10.1016/j.jmoldx.2018.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 01/04/2023] Open
Abstract
Because interpretation of next-generation sequencing (NGS) data remains challenging, optimization of the NGS process is needed to obtain correct sequencing results. Therefore, extensive validation and continuous monitoring of the quality is essential. NGS performance was compared with traditional detection methods and technical quality of nine NGS technologies was assessed. First, nine formalin-fixed, paraffin-embedded patient samples were analyzed by 114 laboratories by using different detection methods. No significant differences in performance were observed between analyses with NGS and traditional techniques. Second, two DNA control samples were analyzed for a selected number of variants by 26 participants with the use of nine different NGS technologies. Quality control metrics were analyzed from raw data files and a survey about routine procedures. Results showed large differences in coverages, but observed variant allele frequencies in raw data files were in line with predefined variant allele frequencies. Many false negative results were found because of low-quality regions, which were not reported as such. It is recommended to disclose the reportable range, the fraction of targeted genomic regions for which calls of acceptable quality can be generated, to avoid any errors in therapy decisions. NGS can be a reliable technique, only if essential quality control during analysis is applied and reported.
Collapse
Affiliation(s)
- Véronique Tack
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Lien Spans
- Center for Human Genetics, University of Leuven, Leuven, Belgium
| | - Ed Schuuring
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cleo Keppens
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Karen Zwaenepoel
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium
| | - Patrick Pauwels
- Center for Oncologic Research (CORE), University of Antwerp, Antwerp, Belgium
| | | | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium; Department of Medical Diagnostics, University Hospital Leuven, Leuven, Belgium.
| |
Collapse
|
28
|
Lin G, Zhang X, Zhang K, Han Y, Tan L, Li J. Evaluation of tacrolimus-related CYP3A5 genotyping in China: Results from the First External Quality Assessment Exercise. J Clin Lab Anal 2018; 32:e22563. [PMID: 29708622 DOI: 10.1002/jcla.22563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 04/08/2018] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Tacrolimus is the most widely used immunosuppressant in solid organ transplant patients. The cytochrome P450 3A5 (CYP3A5) has been proved to be associated with tacrolimus dose requirement. Molecular detection for CYP3A5 genotyping is demanded for the optimization of treatments of tacrolimus. METHODS To achieve the consistency and accuracy of the testing results, the Chinese National Center for Clinical Laboratories (NCCL) organized a national external quality assessment(EQA) program to evaluate the performance of laboratories providing CYP3A5 genotyping. Ten validated DNA samples covering the common genetic polymorphisms of CYP3A5 were delivered to 33 voluntary laboratories, and their detecting results and clinical written reports were evaluated. RESULTS Thirty-three datasets were received. The corresponding analytical sensitivity was 95.9% (285/297 challenges; 95% confidence interval: 93.0%-97.9%), and the analytical specificity was 95.3% (346/363; 95% confidence interval: 92.6%-97.2%). Thirty of the participating laboratories correctly identified the CYP3A5 allele status for all EQA samples. Three laboratories made genotyping errors, and 2 of them failed to detect any of the homozygotes such as *1/*1 and *3/*3. Twenty-eight CYP3A5*3 tests reports were submitted, but many reports showed a shortage of essential information. No reports fulfilled all the consensus recommendations for pharmacogenetic test result reporting. CONCLUSION The EQA program highlighted the necessity for an improvement in the accuracy of genotyping for some of the laboratories and a greater education on the reporting of CYP3A5 genotyping results.
Collapse
Affiliation(s)
- Guigao Lin
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing, China
| | - Xiao Zhang
- Department of Laboratory, The People's Hospital of Hunan Province, First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Kuo Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing, China
| | - Yanxi Han
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing, China
| | - Liming Tan
- Department of Laboratory, The People's Hospital of Hunan Province, First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing, China
| |
Collapse
|
29
|
Keppens C, Tack V, Hart N', Tembuyser L, Ryska A, Pauwels P, Zwaenepoel K, Schuuring E, Cabillic F, Tornillo L, Warth A, Weichert W, Dequeker E. A stitch in time saves nine: external quality assessment rounds demonstrate improved quality of biomarker analysis in lung cancer. Oncotarget 2018; 9:20524-20538. [PMID: 29755669 PMCID: PMC5945546 DOI: 10.18632/oncotarget.24980] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/26/2018] [Indexed: 12/23/2022] Open
Abstract
Biomarker analysis has become routine practice in the treatment of non-small cell lung cancer (NSCLC). To ensure high quality testing, participation to external quality assessment (EQA) schemes is essential. This article provides a longitudinal overview of the EQA performance for EGFR, ALK, and ROS1 analyses in NSCLC between 2012 and 2015. The four scheme years were organized by the European Society of Pathology according to the ISO 17043 standard. Participants were asked to analyze the provided tissue using their routine procedures. Analysis scores improved for individual laboratories upon participation to more EQA schemes, except for ROS1 immunohistochemistry (IHC). For EGFR analysis, scheme error rates were 18.8%, 14.1% and 7.5% in 2013, 2014 and 2015 respectively. For ALK testing, error rates decreased between 2012 and 2015 by 5.2%, 3.2% and 11.8% for the fluorescence in situ hybridization (FISH), FISH digital, and IHC subschemes, respectively. In contrast, for ROS1 error rates increased between 2014 and 2015 for FISH and IHC by 3.2% and 9.3%. Technical failures decreased over the years for all three markers. Results show that EQA contributes to an ameliorated performance for most predictive biomarkers in NSCLC. Room for improvement is still present, especially for ROS1 analysis.
Collapse
Affiliation(s)
- Cleo Keppens
- University of Leuven, Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, Leuven, Belgium
| | - Véronique Tack
- University of Leuven, Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, Leuven, Belgium
| | - Nils 't Hart
- University Medical Center Groningen, Department of Pathology, Groningen, The Netherlands
| | - Lien Tembuyser
- University of Leuven, Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, Leuven, Belgium
| | - Ales Ryska
- Charles University Medical Faculty and University Hospital, Department of Pathology, Hradec Kralove, Czech Republic
| | - Patrick Pauwels
- Center for Oncologic Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Karen Zwaenepoel
- University Hospital Antwerp, Department of Pathology, Edegem, Belgium
| | - Ed Schuuring
- University Medical Center Groningen, Department of Pathology, Groningen, The Netherlands
| | - Florian Cabillic
- Cytogenetics and Cellular Biology Department, CHU de Rennes, Rennes, France.,INSERM, INRA, Université Rennes 1, Université Bretagne Loire, Nutrition Metabolisms and Cancer, Rennes, France
| | - Luigi Tornillo
- University of Basel, Basel, Switzerland.,GILAB AG, Allschwil, Switzerland
| | - Arne Warth
- University Hospital Heidelberg, Heidelberg, Germany
| | | | - Elisabeth Dequeker
- University of Leuven, Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, Leuven, Belgium
| | | |
Collapse
|
30
|
Susman S, Berindan-Neagoe I, Petrushev B, Pirlog R, Florian IS, Mihu CM, Berce C, Craciun L, Grewal R, Tomuleasa C. The role of the pathology department in the preanalytical phase of molecular analyses. Cancer Manag Res 2018; 10:745-753. [PMID: 29695931 PMCID: PMC5903845 DOI: 10.2147/cmar.s150851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
After introducing the new molecules for the treatment of patients with tumoral pathology, the therapeutical decision will be taken depending on the molecular profile performed upon the harvested tissues. This major modification makes the molecular and morphological analysis an essential part in the clinical management of patients and the pathologist plays an important role in this process. The quality and reproducibility of the results are imperative today and they depend on both the reliability of the molecular techniques and the quality of the tissue we use in the process. Also, the genomics and proteomics techniques, used increasingly often, require high-quality tissues, and pathology laboratories play a very significant role in the management of all phases of this process. In this paper the parameters which must be followed in order to obtain optimal results within the techniques which analyze nucleic acids and proteins were reviewed.
Collapse
Affiliation(s)
- Sergiu Susman
- Department of Pathology, Imogen Research Center.,Department of Morphological Sciences
| | | | - Bobe Petrushev
- Research Center for Functional Genomics and Translational Medicine
| | | | - Ioan-Stefan Florian
- Department of Neurosurgery, Iuliu Hatieganu University of Medicine and Pharmacy
| | | | - Cristian Berce
- Research Center for Functional Genomics and Translational Medicine
| | | | - Ravnit Grewal
- Department of Hematology, Ion Chiricuta Oncology Institute
| | - Ciprian Tomuleasa
- Research Center for Functional Genomics and Translational Medicine.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Haematopathology, Tygerberg Academic Hospital, Tygerberg, South Africa
| |
Collapse
|
31
|
Dufraing K, De Hertogh G, Tack V, Keppens C, Dequeker EMC, van Krieken JH. External Quality Assessment Identifies Training Needs to Determine the Neoplastic Cell Content for Biomarker Testing. J Mol Diagn 2018; 20:455-464. [PMID: 29625250 DOI: 10.1016/j.jmoldx.2018.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/14/2018] [Accepted: 03/05/2018] [Indexed: 10/17/2022] Open
Abstract
Neoplastic cell content determination is crucial for biomarker testing. It is known that interobserver variation exists, but largescale data are missing about variation in tumor delineation and cell content determination. Results were obtained from the external quality assessment program for metastatic colorectal cancer from the European Society of Pathology (N = 5776 observations). The study included three parts: current practices were surveyed, neoplastic cell content estimations and delineations were retrieved from stained slides, and clinical reports were analyzed. Seventeen of 43 pathologists determined the neoplastic cell content in a tumor-rich area for DNA extraction and took immune cells (n = 37), tumor cell distribution (n = 33), desmoplastic stroma (n = 30), necrosis (n = 29), and mucus (n = 23) into account. The selected area was highly variable, and the average difference between the highest and lowest estimation ranged between 51% and 78% (2011 to 2017). The number of overestimations was alarmingly high in samples containing <30% tumor cells. Of concern is that 33 of 105 laboratories reported a wild-type result in a sample without tumor in 2017. Standardization of neoplastic cell content determination is needed for test outcome interpretation. The authors' data show variation in estimation practices, tumor delineations and estimations, and interpretation problems (n = 226 reports). Further training for selecting the most suitable block and creating clear reports is urgently needed.
Collapse
Affiliation(s)
- Kelly Dufraing
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gert De Hertogh
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Véronique Tack
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Cleo Keppens
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium.
| | - J Han van Krieken
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
32
|
RAS testing for colorectal cancer patients is reliable in European laboratories that pass external quality assessment. Virchows Arch 2018; 472:717-725. [DOI: 10.1007/s00428-017-2291-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/11/2017] [Accepted: 12/28/2017] [Indexed: 12/22/2022]
|
33
|
Review of the implementation of plasma ctDNA testing on behalf of IQN Path ASBL: a perspective from an EQA providers' survey. VIRCHOWS ARCHIV : AN INTERNATIONAL JOURNAL OF PATHOLOGY 2017. [PMID: 28840321 DOI: 10.1007/s00428-017-2222-z] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
|
34
|
Review of the implementation of plasma ctDNA testing on behalf of IQN Path ASBL: a perspective from an EQA providers' survey. Virchows Arch 2017; 471:809-813. [PMID: 28840321 PMCID: PMC5711978 DOI: 10.1007/s00428-017-2222-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/06/2017] [Accepted: 08/14/2017] [Indexed: 01/28/2023]
|
35
|
Han Y, Zhang R, Lin G, Zhang K, Xie J, Li J. Quality Assessment of Reporting Performance for EGFR Molecular Diagnosis in Non-Small Cell Lung Cancer. Oncologist 2017; 22:1325-1332. [PMID: 28701573 DOI: 10.1634/theoncologist.2017-0042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/08/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Reports serve as a bridge between laboratories and clinicians, help synthesize an overwhelming amount of raw data into evidence-based medicine, and play a significant role in designing clinical treatments. In an effort to guarantee high-quality epidermal growth factor receptor (EGFR) gene mutation testing and reporting performance, the National Center for Clinical Laboratories launched a proficiency testing (PT) scheme reflecting clinical practices in China since 2014. This study focuses on the quality assessment of gene mutation reports. MATERIALS AND METHODS Fifty-three laboratories that submitted reports in both 2014 and 2016 EGFR gene mutation PT schemes were selected for report analysis and comparison according to predefined evaluation criteria. RESULTS The average score for reports from 2014 was 14 out of 30 points. The overall scores for reports from 2016 improved substantially, yielding an average score of 20 out of 30 points. Among the evaluation criteria, general items were well documented in the reports. However, items specific to molecular diagnosis were far from satisfactory, and some items were even missing. CONCLUSION The quality assessment of clinical written reports from 2014 and 2016 demonstrates that substantial improvements have been made in overall reporting performance. However, not all statements pertaining to important elements met expectations. To continue education, repeated PT schemes need to be executed in a timely fashion to expose and address existing shortcomings in clinical reports. There remains ample room for improvement towards generating concise, comprehensive, and readable reports. IMPLICATIONS FOR PRACTICE This article compares the quality of clinical gene mutation reports submitted in 2014 to those submitted in 2016 epidermal growth factor receptor proficiency testing schemes, exposes the existing shortcomings, and discusses ways to communicate results more effectively in the future. The findings demonstrate that notable progress was observed in the overall reporting performance. However, key points specific to molecular diagnosis were far from expectation, and some items were even missing. Standardization needs to be emphasized to improve the report format and content. This article provides a reference that laboratories can use to write concise, comprehensive, and readily accessible clinical reports.
Collapse
Affiliation(s)
- Yanxi Han
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Rui Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Guigao Lin
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Kuo Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Jiehong Xie
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| |
Collapse
|
36
|
Thunnissen E, Allen TC, Adam J, Aisner DL, Beasley MB, Borczuk AC, Cagle PT, Capelozzi VL, Cooper W, Hariri LP, Kern I, Lantuejoul S, Miller R, Mino-Kenudson M, Radonic T, Raparia K, Rekhtman N, Roy-Chowdhuri S, Russell P, Schneider F, Sholl LM, Tsao MS, Vivero M, Yatabe Y. Immunohistochemistry of Pulmonary Biomarkers: A Perspective From Members of the Pulmonary Pathology Society. Arch Pathol Lab Med 2017; 142:408-419. [PMID: 28686497 DOI: 10.5858/arpa.2017-0106-sa] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The use of immunohistochemistry for the determination of pulmonary carcinoma biomarkers is a well-established and powerful technique. Immunohistochemisty is readily available in pathology laboratories, is relatively easy to perform and assess, can provide clinically meaningful results very quickly, and is relatively inexpensive. Pulmonary predictive biomarkers provide results essential for timely and accurate therapeutic decision making; for patients with metastatic non-small cell lung cancer, predictive immunohistochemistry includes ALK and programmed death ligand-1 (PD-L1) (ROS1, EGFR in Europe) testing. Handling along proper methodologic lines is needed to ensure patients receive the most accurate and representative test outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yasushi Yatabe
- From the Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands (Drs Thunnissen and Radonic); the Department of Pathology, The University of Texas Medical Branch, Galveston (Dr Allen); the Department of Pathology, Gustave Roussy, Villejuif, France (Dr Adam); the Department of Pathology, University of Colorado, Aurora (Dr Aisner); the Department of Pathology, Mount Sinai Medical Center, New York, New York (Dr Beasley); the Department of Pathology, Weill Cornell University Medical Center, New York, New York (Dr Borczuk); the Department of Pathology & Genomic Medicine, Houston Methodist Hospital, Houston, Texas (Drs Cagle and Miller); the Department of Pathology, University of São Paulo, São Paulo, Brazil (Dr Capelozzi); the Department of Pathology, Royal Prince Alfred Hospital, Sydney, Australia (Dr Cooper); the Department of Pathology, Massachusetts General Hospital, Boston (Drs Hariri and Mino-Kenudson); the Department of Pathology, University Clinic Golnik, Golnik, Slovenia (Dr Kern); the Department of Pathology, INSERM U578, CHU A Michallon, Centre Léon Bérard, Lyon, Université Joseph Fourier INSERM U 823, Institut A. Bonniot, Grenoble, France (Dr Lantuejoul); the Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Dr Raparia); the Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (Dr Rekhtman); the Department of Pathology, The University Of Texas MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); the Department of Pathology, St. Vincent's Pathology, Fitzroy, Australia (Ms Russell); the Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (Dr Schneider); the Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Drs Sholl and Vivero); the Department of Pathology, University of Toronto, University Health Network, Toronto, Ontario, Canada (Dr Tsao); and the Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan (Dr Yatabe)
| |
Collapse
|
37
|
Normanno N, Fenizia F, Castiglione F, Barberis M, Taddei GL, Truini M, De Rosa G, Pinto C, Marchetti A. External quality assessment for EGFR mutations in Italy: improvements in performances over the time. ESMO Open 2017; 2:e000160. [PMID: 29181190 PMCID: PMC5699165 DOI: 10.1136/esmoopen-2017-000160] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 12/19/2022] Open
Abstract
External quality assessment (EQA) schemes are essential procedures to assess the quality level of laboratories performing molecular testing of the epidermal growth factor receptor (EGFR) gene in non-small cell lung cancer. The Italian Association of Medical Oncology (AIOM) and the Italian Society of Pathology (SIAPEC-IAP) organise EGFR EQA programmes to ensure that the Italian laboratories achieve the quality standard levels required. Comparing the 2011, 2013 and 2015 EGFR EQA schemes, it was possible to observe improvements in the methodologies used and the outcomes. The use of direct sequencing was reduced from 78.7% in 2011 to only 14.1% in 2015, whereas the use of pyrosequencing and real-time PCR increased. The number of rounds in which centres using direct sequencing failed was significantly higher than the number of rounds that failed using other methods, both when analysing each single scheme and when combining the three EQAs together. In 2011 and 2013, about 29% of the participants failed the first phase of the programmes, compared with the 13% of centres failing in 2015, suggesting that the switch to more sensitive and robust methods could allow to increase the percentage of good performers. Although the molecular analyses are performed with good quality in Italy, the continuous education carried out by AIOM and SIAPEC-IAP remains a fundamental tool to maintain this quality level.
Collapse
Affiliation(s)
- Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale'-IRCCS, Naples, Italy
| | - Francesca Fenizia
- Laboratory of Pharmacogenomics, Centro di Ricerche Oncologiche di Mercogliano (CROM), Istituto Nazionale Tumori 'Fondazione Giovanni Pascale' IRCCS, Naples, Italy
| | | | - Massimo Barberis
- Department of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Gian Luigi Taddei
- Department of Human Pathology and Oncology, University of Florence, Florence, Italy
| | - Mauro Truini
- Pathological Anatomy Histology and Cytogenetics, Niguarda Cancer Center, Niguarda Ca' Granda Hospital, Milan, Italy
| | - Gaetano De Rosa
- Pathology Section, Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Carmine Pinto
- Department of Medical Oncology, S. Maria Hospital - IRCCS, Reggio Emilia, Italy
| | - Antonio Marchetti
- Center of Predictive Molecular Medicine, "G. d'Annunzio" University, Chieti, Italy
| |
Collapse
|
38
|
Tack V, Dufraing K, Deans ZC, van Krieken HJ, Dequeker EMC. The ins and outs of molecular pathology reporting. Virchows Arch 2017; 471:199-207. [PMID: 28343306 DOI: 10.1007/s00428-017-2108-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/07/2017] [Accepted: 03/12/2017] [Indexed: 01/15/2023]
Abstract
The raid evolution in molecular pathology resulting in an increasing complexity requires careful reporting. The need for standardisation is clearer than ever. While synoptic reporting was first used for reporting hereditary genetic diseases, it is becoming more frequent in pathology, especially molecular pathology reports too. The narrative approach is no longer feasible with the growing amount of essential data present on the report, although narrative components are still necessary for interpretation in molecular pathology. On the way towards standardisation of reports, guidelines can be a helpful tool. There are several guidelines that focus on reporting in the field of hereditary diseases, but it is not always feasible to extrapolate these to the reporting of somatic variants in molecular pathology. The rise of multi-gene testing causes challenges for the laboratories. In order to provide a continuous optimisation of the laboratory testing process, including reporting, external quality assessment is essential and has already proven to improve the quality of reports. In general, a clear and concise report for molecular pathology can be created by including elements deemed important by different guidelines, adapting the report to the process flows of the laboratory and integrating the report with the laboratory information management system and the patient record.
Collapse
Affiliation(s)
- Véronique Tack
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35 Blok D, 3000, Leuven, Belgium
| | - Kelly Dufraing
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35 Blok D, 3000, Leuven, Belgium
| | - Zandra C Deans
- Department of Laboratory Medicine, UK NEQAS for Molecular Genetics, UK NEQAS Edinburgh, The Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Han J van Krieken
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Kapucijnenvoer 35 Blok D, 3000, Leuven, Belgium.
| |
Collapse
|
39
|
Reiman A, Kikuchi H, Scocchia D, Smith P, Tsang YW, Snead D, Cree IA. Validation of an NGS mutation detection panel for melanoma. BMC Cancer 2017; 17:150. [PMID: 28228113 PMCID: PMC5322598 DOI: 10.1186/s12885-017-3149-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 02/18/2017] [Indexed: 11/10/2022] Open
Abstract
Background Knowledge of the genotype of melanoma is important to guide patient management. Identification of mutations in BRAF and c-KIT lead directly to targeted treatment, but it is also helpful to know if there are driver oncogene mutations in NRAS, GNAQ or GNA11 as these patients may benefit from alternative strategies such as immunotherapy. Methods While polymerase chain reaction (PCR) methods are often used to detect BRAF mutations, next generation sequencing (NGS) is able to determine all of the necessary information on several genes at once, with potential advantages in turnaround time. We describe here an Ampliseq hotspot panel for melanoma for use with the IonTorrent Personal Genome Machine (PGM) which covers the mutations currently of most clinical interest. Results We have validated this in 151 cases of skin and uveal melanoma from our files, and correlated the data with PCR based assessment of BRAF status. There was excellent agreement, with few discrepancies, though NGS does have greater coverage and picks up some mutations that would be missed by PCR. However, these are often rare and of unknown significance for treatment. Conclusions PCR methods are rapid, less time-consuming and less expensive than NGS, and could be used as triage for patients requiring more extensive diagnostic workup. The NGS panel described here is suitable for clinical use with formalin-fixed paraffin-embedded (FFPE) samples. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3149-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne Reiman
- Department of Pathology - Coventry and Warwickshire Pathology Services (CWPS), University Hospitals Coventry and Warwickshire, Coventry, CV2 2DX, UK.,Centre for Research in Applied Biological and Exercise Sciences, Coventry University, Coventry, CV1 5FB, UK
| | - Hugh Kikuchi
- Department of Pathology - Coventry and Warwickshire Pathology Services (CWPS), University Hospitals Coventry and Warwickshire, Coventry, CV2 2DX, UK.,Centre for Research in Applied Biological and Exercise Sciences, Coventry University, Coventry, CV1 5FB, UK
| | - Daniela Scocchia
- Department of Pathology - Coventry and Warwickshire Pathology Services (CWPS), University Hospitals Coventry and Warwickshire, Coventry, CV2 2DX, UK
| | - Peter Smith
- Department of Pathology - Coventry and Warwickshire Pathology Services (CWPS), University Hospitals Coventry and Warwickshire, Coventry, CV2 2DX, UK
| | - Yee Wah Tsang
- Department of Pathology - Coventry and Warwickshire Pathology Services (CWPS), University Hospitals Coventry and Warwickshire, Coventry, CV2 2DX, UK
| | - David Snead
- Department of Pathology - Coventry and Warwickshire Pathology Services (CWPS), University Hospitals Coventry and Warwickshire, Coventry, CV2 2DX, UK
| | - Ian A Cree
- Department of Pathology - Coventry and Warwickshire Pathology Services (CWPS), University Hospitals Coventry and Warwickshire, Coventry, CV2 2DX, UK. .,Institute of Ophthalmology, University College London, Bath Street, London, EC1V 9EL, UK. .,Centre for Technology Enabled Health Research (CTEHR), Faculty of Health & Life Sciences, Coventry University, Coventry, CV1 5FB, UK.
| |
Collapse
|
40
|
van Krieken H, Deans S, Hall JA, Normanno N, Ciardiello F, Douillard JY. Quality to rely on: meeting report of the 5th Meeting of External Quality Assessment, Naples 2016. ESMO Open 2016; 1:e000114. [PMID: 27933215 PMCID: PMC5133380 DOI: 10.1136/esmoopen-2016-000114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 10/24/2016] [Indexed: 01/01/2023] Open
Affiliation(s)
- Han van Krieken
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sandi Deans
- Department of Laboratory Medicine , UK NEQAS for Molecular Genetics, Royal Infirmary of Edinburgh , Edinburgh , UK
| | - Jacqueline A Hall
- IQN Path A.S.B.L, Luxembourg, Luxembourg; Faculty Medicine, Division of Cancer, Department of Surgery & Cancer, Imperial College London, London, UK
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit , Istituto Nazionale Tumouri "Fondazione Giovanni Pascale" IRCCS , Napoli , Italy
| | | | | |
Collapse
|
41
|
Diagnostic RAS mutation analysis by polymerase chain reaction (PCR). BIOMOLECULAR DETECTION AND QUANTIFICATION 2016; 8:29-32. [PMID: 27335808 PMCID: PMC4906127 DOI: 10.1016/j.bdq.2016.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/26/2016] [Accepted: 05/17/2016] [Indexed: 02/07/2023]
Abstract
RAS mutation analysis is an important companion diagnostic test. Treatment of colorectal cancer with anti-Epidermal Growth Factor Receptor (EGFR) therapy requires demonstration of RAS mutation status (both KRAS and NRAS), and it is good practice to include BRAF. In Non-Small Cell Lung Cancer (NSCLC) and melanoma, assessment of RAS mutation status can be helpful in triaging patient samples for more extensive testing. This mini-review will discuss the role of PCR methods in providing rapid diagnostic information for cancer patients.
Collapse
|
42
|
Van Krieken JHJM, Rouleau E, Ligtenberg MJL, Normanno N, Patterson SD, Jung A. RAS testing in metastatic colorectal cancer: advances in Europe. Virchows Arch 2016; 468:383-96. [PMID: 26573425 PMCID: PMC4830882 DOI: 10.1007/s00428-015-1876-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 10/20/2015] [Accepted: 10/22/2015] [Indexed: 12/22/2022]
Abstract
Personalized medicine shows promise for maximizing efficacy and minimizing toxicity of anti-cancer treatment. KRAS exon 2 mutations are predictive of resistance to epidermal growth factor receptor-directed monoclonal antibodies in patients with metastatic colorectal cancer. Recent studies have shown that broader RAS testing (KRAS and NRAS) is needed to select patients for treatment. While Sanger sequencing is still used, approaches based on various methodologies are available. Few CE-approved kits, however, detect the full spectrum of RAS mutations. More recently, "next-generation" sequencing has been developed for research use, including parallel semiconductor sequencing and reversible termination. These techniques have high technical sensitivities for detecting mutations, although the ideal threshold is currently unknown. Finally, liquid biopsy has the potential to become an additional tool to assess tumor-derived DNA. For accurate and timely RAS testing, appropriate sampling and prompt delivery of material is critical. Processes to ensure efficient turnaround from sample request to RAS evaluation must be implemented so that patients receive the most appropriate treatment. Given the variety of methodologies, external quality assurance programs are important to ensure a high standard of RAS testing. Here, we review technical and practical aspects of RAS testing for pathologists working with metastatic colorectal cancer tumor samples. The extension of markers from KRAS to RAS testing is the new paradigm for biomarker testing in colorectal cancer.
Collapse
Affiliation(s)
- J Han J M Van Krieken
- Department of Pathology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, Netherlands.
| | | | - Marjolijn J L Ligtenberg
- Department of Pathology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, Netherlands
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, INT-Fondazione Pascale, Naples, Italy
| | - Scott D Patterson
- Amgen Inc., Thousand Oaks, CA, USA
- Gilead Sciences, Inc., Foster City, CA, USA
| | - Andreas Jung
- Institute of Pathology, University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
43
|
Tack V, Deans ZC, Wolstenholme N, Patton S, Dequeker EMC. What's in a Name? A Coordinated Approach toward the Correct Use of a Uniform Nomenclature to Improve Patient Reports and Databases. Hum Mutat 2016; 37:570-5. [DOI: 10.1002/humu.22975] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/04/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Véronique Tack
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit; KU Leuven; Leuven Belgium
| | - Zandra C. Deans
- Department of Laboratory Medicine, UK NEQAS for Molecular Genetics, UK NEQAS Edinburgh; The Royal Infirmary of Edinburgh; Edinburgh UK
| | - Nicola Wolstenholme
- EMQN, Manchester Centre for Genomic Medicine; St Mary's Hospital; Manchester M13 9WL UK
| | - Simon Patton
- EMQN, Manchester Centre for Genomic Medicine; St Mary's Hospital; Manchester M13 9WL UK
| | - Elisabeth M. C. Dequeker
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit; KU Leuven; Leuven Belgium
| |
Collapse
|
44
|
Dequeker EM, Keppens C, Egele C, Delen S, Lamy A, Lemoine A, Sabourin JC, Andrieu C, Ligtenberg M, Fetique D, Tops B, Descarpentries C, Blons H, Denoux Y, Aube C, Penault-Llorca F, Hofman P, Leroy K, Le Marechal C, Doucet L, Duranton-Tanneur V, Pedeutour F, Soubeyran I, Côté JF, Emile JF, Vignaud JM, Monhoven N, Haddad V, Laurent-Puig P, van Krieken H, Nowak F, Lonchamp E, Bellocq JP, Rouleau E. Three Rounds of External Quality Assessment in France to Evaluate the Performance of 28 Platforms for Multiparametric Molecular Testing in Metastatic Colorectal and Non-Small Cell Lung Cancer. J Mol Diagn 2016; 18:205-14. [DOI: 10.1016/j.jmoldx.2015.09.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/29/2015] [Accepted: 09/23/2015] [Indexed: 12/31/2022] Open
|
45
|
Dietel M, Bubendorf L, Dingemans AMC, Dooms C, Elmberger G, García RC, Kerr KM, Lim E, López-Ríos F, Thunnissen E, Van Schil PE, von Laffert M. Diagnostic procedures for non-small-cell lung cancer (NSCLC): recommendations of the European Expert Group. Thorax 2015; 71:177-84. [PMID: 26530085 PMCID: PMC4752623 DOI: 10.1136/thoraxjnl-2014-206677] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 07/21/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND There is currently no Europe-wide consensus on the appropriate preanalytical measures and workflow to optimise procedures for tissue-based molecular testing of non-small-cell lung cancer (NSCLC). To address this, a group of lung cancer experts (see list of authors) convened to discuss and propose standard operating procedures (SOPs) for NSCLC. METHODS Based on earlier meetings and scientific expertise on lung cancer, a multidisciplinary group meeting was aligned. The aim was to include all relevant aspects concerning NSCLC diagnosis. After careful consideration, the following topics were selected and each was reviewed by the experts: surgical resection and sampling; biopsy procedures for analysis; preanalytical and other variables affecting quality of tissue; tissue conservation; testing procedures for epidermal growth factor receptor, anaplastic lymphoma kinase and ROS proto-oncogene 1, receptor tyrosine kinase (ROS1) in lung tissue and cytological specimens; as well as standardised reporting and quality control (QC). Finally, an optimal workflow was described. RESULTS Suggested optimal procedures and workflows are discussed in detail. The broad consensus was that the complex workflow presented can only be executed effectively by an interdisciplinary approach using a well-trained team. CONCLUSIONS To optimise diagnosis and treatment of patients with NSCLC, it is essential to establish SOPs that are adaptable to the local situation. In addition, a continuous QC system and a local multidisciplinary tumour-type-oriented board are essential.
Collapse
Affiliation(s)
- Manfred Dietel
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Anne-Marie C Dingemans
- Department of Respiratory Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Christophe Dooms
- Respiratory Division, University Hospitals KU Leuven, Leuven, Belgium
| | - Göran Elmberger
- Department of Laboratory Medicine, Pathology, Örebro University Hospital, Örebro, Sweden
| | - Rosa Calero García
- Department of Radiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Keith M Kerr
- Aberdeen University Medical School, Aberdeen, UK
| | - Eric Lim
- Academic Division of Thoracic Surgery, The Royal Brompton Hospital and Imperial College, London, UK
| | - Fernando López-Ríos
- Laboratorio de Dianas Terapéuticas, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul E Van Schil
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital, Antwerp, Belgium
| | | |
Collapse
|
46
|
Dietel M, Jöhrens K, Laffert MV, Hummel M, Bläker H, Pfitzner BM, Lehmann A, Denkert C, Darb-Esfahani S, Lenze D, Heppner FL, Koch A, Sers C, Klauschen F, Anagnostopoulos I. A 2015 update on predictive molecular pathology and its role in targeted cancer therapy: a review focussing on clinical relevance. Cancer Gene Ther 2015; 22:417-30. [PMID: 26358176 DOI: 10.1038/cgt.2015.39] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/15/2022]
Abstract
In April 2013 our group published a review on predictive molecular pathology in this journal. Although only 2 years have passed many new facts and stimulating developments have happened in diagnostic molecular pathology rendering it worthwhile to present an up-date on this topic. A major technical improvement is certainly given by the introduction of next-generation sequencing (NGS; amplicon, whole exome, whole genome) and its application to formalin-fixed paraffin-embedded (FFPE) tissue in routine diagnostics. Based on this 'revolution' the analyses of numerous genetic alterations in parallel has become a routine approach opening the chance to characterize patients' malignant tumors much more deeply without increasing turn-around time and costs. In the near future this will open new strategies to apply 'off-label' targeted therapies, e.g. for rare tumors, otherwise resistant tumors etc. The clinically relevant genetic aberrations described in this review include mutation analyses of RAS (KRAS and NRAS), BRAF and PI3K in colorectal cancer, KIT or PDGFR alpha as well as BRAF, NRAS and KIT in malignant melanoma. Moreover, we present several recent advances in the molecular characterization of malignant lymphoma. Beside the well-known mutations in NSCLC (EGFR, ALK) a number of chromosomal aberrations (KRAS, ROS1, MET) have become relevant. Only very recently has the clinical need for analysis of BRCA1/2 come up and proven as a true challenge for routine diagnostics because of the genes' special structure and hot-spot-free mutational distribution. The genetic alterations are discussed in connection with their increasingly important role in companion diagnostics to apply targeted drugs as efficient as possible. As another aspect of the increasing number of druggable mutations, we discuss the challenges personalized therapies pose for the design of clinical studies to prove optimal efficacy particularly with respect to combination therapies of multiple targeted drugs and conventional chemotherapy. Such combinations would lead to an extremely high complexity that would hardly be manageable by applying conventional study designs for approval, e.g. by the FDA or EMA. Up-coming challenges such as the application of methylation assays and proteomic analyses on FFPE tissue will also be discussed briefly to open the door towards the ultimate goal of reading a patients' tissue as 'deeply' as possible. Although it is yet to be shown, which levels of biological information are most informative for predictive pathology, an integrated molecular characterization of tumors will likely offer the most comprehensive view for individualized therapy approaches. To optimize cancer treatment we need to understand tumor biology in much more detail on morphological, genetic, proteomic as well as epigenetic grounds. Finally, the complex challenges on the level of drug design, molecular diagnostics, and clinical trials make necessary a close collaboration among academic institutions, regulatory authorities and pharmaceutical companies.
Collapse
Affiliation(s)
- M Dietel
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - K Jöhrens
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - M V Laffert
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - M Hummel
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - H Bläker
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - B M Pfitzner
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - A Lehmann
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - C Denkert
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - S Darb-Esfahani
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - D Lenze
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - F L Heppner
- Institute of Neuropathology, Charité, University Medicine Berlin, Berlin, Germany
| | - A Koch
- Institute of Neuropathology, Charité, University Medicine Berlin, Berlin, Germany
| | - C Sers
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - F Klauschen
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| | - I Anagnostopoulos
- Institute of Pathology, Charité, University Medicine Berlin, Berlin, Germany
| |
Collapse
|
47
|
Saridaki Z, Saegart X, De Vriendt V, Hatzidaki D, Palmans S, De Smedt L, De Hertogh G, Tejpar S. KRAS, NRAS, BRAF mutation comparison of endoscopic and surgically removed primary CRC paired samples: is endoscopy biopsy material adequate for molecular evaluation? Br J Cancer 2015; 113:914-20. [PMID: 26325103 PMCID: PMC4578093 DOI: 10.1038/bjc.2015.307] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/23/2015] [Accepted: 07/30/2015] [Indexed: 12/31/2022] Open
Abstract
Background: An everyday clinical practice dilemma in the 20–30% of metastatic colorectal cancer (CRC) patients that have not been operated on their primary tumour, is, under which specific histopathology and molecular circumstances, an endoscopic biopsy could be considered adequate to provide a representative RAS/BRAF molecular status to guide treatment. Methods: A consecutive series of 193 paired biopsy and primary CRC tumour samples between August 2008 and 2010 available in the Department of Pathology archives, University Hospitals, KU Leuven were retrieved. For a pair to be included, in the endoscopic biopsy, 20% of invasive adenocarcinoma cells should be present and enough slides to yield an extracted DNA concentration of ⩾5 ng μl−1, and no <2 ng μl−1 should be available for cutting. Exons 2–4 KRAS/NRAS, BRAF, PIK3CA molecular evaluation was performed with RT–PCR and Sequenom. Results: From 165 deemed adequate by the pathologist pairs, 85 (51.5%) were concordantly mutated in at least one of the tested genes, 70 (42.5%) were wt and 10 (6%) were discordant, harbouring a mutation in the primary and not in the endoscopic biopsy. In the re-evaluation, when more slides were cut per discordant pair, mutational status changed in two of the six discordantly KRAS-mutated pairs. A strong strength of agreement for both runs was observed (Cohen's kappa, k=0.877, P<0.001 and k=0.901, P<0.001, respectively) between the surgically acquired and the endoscopic biopsy specimens' evaluation. Conclusions: Based on our results, an endoscopic biopsy could provide an accurate mutational profile and become a justified alternative to a surgically removed primary tumour specimen, as long as specific histopathology criteria are met.
Collapse
Affiliation(s)
- Z Saridaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Voutes 71110, Heraklion, Greece.,Department of Oncology, Laboratory of Molecular Digestive Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - X Saegart
- Center for Translational Cell and Tissue Research KU Leuven, Minderbroederstraat 12 3000 Leuven, Belgium
| | - V De Vriendt
- Department of Oncology, Laboratory of Molecular Digestive Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - D Hatzidaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Voutes 71110, Heraklion, Greece
| | - S Palmans
- Center for Translational Cell and Tissue Research KU Leuven, Minderbroederstraat 12 3000 Leuven, Belgium
| | - L De Smedt
- Center for Translational Cell and Tissue Research KU Leuven, Minderbroederstraat 12 3000 Leuven, Belgium
| | - G De Hertogh
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium
| | - S Tejpar
- Department of Oncology, Laboratory of Molecular Digestive Oncology, Katholieke Universiteit Leuven, Leuven, Belgium.,University Hospitals Leuven and KU Leuven, Leuven, Belgium
| |
Collapse
|
48
|
Endorsing good quality assurance practices in molecular pathology: risks and recommendations for diagnostic laboratories and external quality assessment providers. Virchows Arch 2015; 468:31-41. [PMID: 26306715 DOI: 10.1007/s00428-015-1839-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/06/2015] [Accepted: 08/17/2015] [Indexed: 12/13/2022]
Abstract
Quality assurance is an indispensable element in a molecular diagnostic laboratory. The ultimate goal is to warrant patient safety. Several risks that can compromise high quality procedures are at stake, from sample collection to the test performed by the laboratory, the reporting of test results to clinicians, and the organization of effective external quality assessment schemes. Quality assurance should therefore be safeguarded at each level and should imply a holistic multidisciplinary approach. This review aims to provide an overview of good quality assurance practices and discusses certain risks and recommendations to promote and improve quality assurance for both diagnostic laboratories and for external quality assessment providers. The number of molecular targets is continuously rising, and new technologies are evolving. As this poses challenges for clinical implementation and increases the demand for external quality assessment, the formation of an international association for improving quality assurance in molecular pathology is called for.
Collapse
|
49
|
Tack V, Ligtenberg MJL, Tembuyser L, Normanno N, Vander Borght S, Han van Krieken J, Dequeker EMC. External quality assessment unravels interlaboratory differences in quality of RAS testing for anti-EGFR therapy in colorectal cancer. Oncologist 2015; 20:257-62. [PMID: 25657200 PMCID: PMC4350801 DOI: 10.1634/theoncologist.2014-0382] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/02/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Regulations for the selection of patients with metastatic colorectal cancer for anti-EGFR treatment changed at the end of 2013. The set of mutations to be tested extended from KRAS codons 12 and 13 to KRAS and NRAS exons 2, 3, and 4. A European external quality assessment scheme monitored the performance of laboratories and evaluated the implementation of the new regulations. MATERIALS AND METHODS The 131 participating laboratories received 10 samples of formalin-fixed paraffin-embedded material, including RAS (exon 2, 3, 4) and BRAF mutations. Mock clinical data were provided for three cases. Using their routine methods, laboratories determined the genotypes and submitted three written reports. Assessors scored the results according to predefined evaluation criteria. RESULTS Half of the participants (49.3%) had completely implemented the new test requirements (codons 12, 13, 59, 61, 117, and 146 of KRAS and NRAS), and 96 laboratories (73.3%) made no genotype mistakes. Correct nomenclature, according to the Human Genome Variation Society, was used by 82 laboratories (62.6%). CONCLUSION Although regulations were effective for several months, many laboratories were not ready for full RAS testing in the context of anti-EGFR therapy. Nevertheless, in each participating country, there are laboratories that provide complete and correct testing. External quality assessments can be used to monitor implementation of new test regulations and to stimulate the laboratories to improve their testing procedures. Because the results of this program are available on the website of the European Society of Pathology, patients and clinicians can refer test samples to a reliable laboratory.
Collapse
Affiliation(s)
- Véronique Tack
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands; Cell Biology and Biotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Naples, Italy; Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Marjolijn J L Ligtenberg
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands; Cell Biology and Biotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Naples, Italy; Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Lien Tembuyser
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands; Cell Biology and Biotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Naples, Italy; Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Nicola Normanno
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands; Cell Biology and Biotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Naples, Italy; Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Sara Vander Borght
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands; Cell Biology and Biotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Naples, Italy; Department of Pathology, UZ Leuven, Leuven, Belgium
| | - J Han van Krieken
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands; Cell Biology and Biotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Naples, Italy; Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands; Cell Biology and Biotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Naples, Italy; Department of Pathology, UZ Leuven, Leuven, Belgium
| |
Collapse
|
50
|
Bolton L, Reiman A, Lucas K, Timms J, Cree IA. KRAS mutation analysis by PCR: a comparison of two methods. PLoS One 2015; 10:e0115672. [PMID: 25568935 PMCID: PMC4287618 DOI: 10.1371/journal.pone.0115672] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 11/26/2014] [Indexed: 01/04/2023] Open
Abstract
Background KRAS mutation assays are important companion diagnostic tests to guide anti-EGFR antibody treatment of metastatic colorectal cancer. Direct comparison of newer diagnostic methods with existing methods is an important part of validation of any new technique. In this this study, we have compared the Therascreen (Qiagen) ARMS assay with Competitive Allele-Specific TaqMan PCR (castPCR, Life Technologies) to determine equivalence for KRAS mutation analysis. Methods DNA was extracted by Maxwell (Promega) from 99 colorectal cancers. The ARMS-based Therascreen and a customized castPCR assay were performed according to the manufacturer’s instructions. All assays were performed on either an Applied Biosystems 7500 Fast Dx or a ViiA7 real-time PCR machine (both from Life Technologies). The data were collected and discrepant results re-tested with newly extracted DNA from the same blocks in both assay types. Results Of the 99 tumors included, Therascreen showed 62 tumors to be wild-type (WT) for KRAS, while 37 had KRAS mutations on initial testing. CastPCR showed 61 tumors to be wild-type (WT) for KRAS, while 38 had KRAS mutations. Thirteen tumors showed BRAF mutation in castPCR and in one of these there was also a KRAS mutation. The custom castPCR plate included several other KRAS mutations and BRAF V600E, not included in Therascreen, explaining the higher number of mutations detected by castPCR. Re-testing of discrepant results was required in three tumors, all of which then achieved concordance for KRAS. CastPCR assay Ct values were on average 2 cycles lower than Therascreen. Conclusion There was excellent correlation between the two methods. Although castPCR assay shows lower Ct values than Therascreen, this is unlikely to be clinically significant.
Collapse
Affiliation(s)
- Louise Bolton
- Department of Pathology, Queen Alexandra Hospital, Portsmouth, United Kingdom
| | - Anne Reiman
- Department of Pathology and Warwick Medical School, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | - Katie Lucas
- Department of Pathology and Warwick Medical School, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | - Judith Timms
- Department of Pathology and Warwick Medical School, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | - Ian A. Cree
- Department of Pathology and Warwick Medical School, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
- * E-mail:
| |
Collapse
|