1
|
Ntzifa A, Lianidou E. Pre-analytical conditions and implementation of quality control steps in liquid biopsy analysis. Crit Rev Clin Lab Sci 2023; 60:573-594. [PMID: 37518938 DOI: 10.1080/10408363.2023.2230290] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/23/2023] [Indexed: 08/01/2023]
Abstract
Over the last decade, great advancements have been made in the field of liquid biopsy through extensive research and the development of new technologies that facilitate the use of liquid biopsy for cancer patients. This is shown by the numerous liquid biopsy tests that gained clearance by the US Food and Drug Administration (FDA) in recent years. Liquid biopsy has significantly altered cancer treatment by providing clinicians with powerful and immediate information about therapeutic decisions. However, the clinical integration of liquid biopsy is still challenging and there are many critical factors to consider prior to its implementation into routine clinical practice. Lack of standardization due to technical challenges and the definition of the clinical utility of specific assays further complicates the establishment of Standard Operating Procedures (SOPs) in liquid biopsy. Harmonization of laboratories to established guidelines is of major importance to overcome inter-lab variabilities observed. Quality control assessment in diagnostic laboratories that offer liquid biopsy testing will ensure that clinicians can base their therapeutic decisions on robust results. The regular participation of laboratories in external quality assessment schemes for liquid biopsy testing aims to promptly pinpoint deficiencies and efficiently educate laboratories to improve their quality of services. Accreditation of liquid biopsy diagnostic laboratories based on the ISO15189 standard in Europe or by CLIA/CAP accreditation procedures in the US is the best way to achieve the adaptation of liquid biopsy into the clinical setting by assuring reliable results for the clinicians and their cancer patients. Nowadays, various organizations from academia, industry, and regulatory agencies collaborate to set a framework that will include all procedures from the pre-analytical phase and the analytical process to the final interpretation of results. In this review, we underline several challenges in the analysis of circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs) concerning standardization of protocols, quality control assessment, harmonization of laboratories, and compliance to specific guidelines that need to be thoroughly considered before liquid biopsy enters the clinic.
Collapse
Affiliation(s)
- Aliki Ntzifa
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
2
|
Hofman P, Calabrese F, Kern I, Adam J, Alarcão A, Alborelli I, Anton NT, Arndt A, Avdalyan A, Barberis M, Bégueret H, Bisig B, Blons H, Boström P, Brcic L, Bubanovic G, Buisson A, Caliò A, Cannone M, Carvalho L, Caumont C, Cayre A, Chalabreysse L, Chenard MP, Conde E, Copin MC, Côté JF, D'Haene N, Dai HY, de Leval L, Delongova P, Denčić-Fekete M, Fabre A, Ferenc F, Forest F, de Fraipont F, Garcia-Martos M, Gauchotte G, Geraghty R, Guerin E, Guerrero D, Hernandez S, Hurník P, Jean-Jacques B, Kashofer K, Kazdal D, Lantuejoul S, Leonce C, Lupo A, Malapelle U, Matej R, Merlin JL, Mertz KD, Morel A, Mutka A, Normanno N, Ovidiu P, Panizo A, Papotti MG, Parobkova E, Pasello G, Pauwels P, Pelosi G, Penault-Llorca F, Picot T, Piton N, Pittaro A, Planchard G, Poté N, Radonic T, Rapa I, Rappa A, Roma C, Rot M, Sabourin JC, Salmon I, Prince SS, Scarpa A, Schuuring E, Serre I, Siozopoulou V, Sizaret D, Smojver-Ježek S, Solassol J, Steinestel K, Stojšić J, Syrykh C, Timofeev S, Troncone G, Uguen A, Valmary-Degano S, Vigier A, Volante M, Wahl SGF, Stenzinger A, Ilié M. Real-world EGFR testing practices for non-small-cell lung cancer by thoracic pathology laboratories across Europe. ESMO Open 2023; 8:101628. [PMID: 37713929 PMCID: PMC10594022 DOI: 10.1016/j.esmoop.2023.101628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/14/2023] [Accepted: 08/02/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Testing for epidermal growth factor receptor (EGFR) mutations is an essential recommendation in guidelines for metastatic non-squamous non-small-cell lung cancer, and is considered mandatory in European countries. However, in practice, challenges are often faced when carrying out routine biomarker testing, including access to testing, inadequate tissue samples and long turnaround times (TATs). MATERIALS AND METHODS To evaluate the real-world EGFR testing practices of European pathology laboratories, an online survey was set up and validated by the Pulmonary Pathology Working Group of the European Society of Pathology and distributed to 64 expert testing laboratories. The retrospective survey focussed on laboratory organisation and daily EGFR testing practice of pathologists and molecular biologists between 2018 and 2021. RESULTS TATs varied greatly both between and within countries. These discrepancies may be partly due to reflex testing practices, as 20.8% of laboratories carried out EGFR testing only at the request of the clinician. Many laboratories across Europe still favour single-test sequencing as a primary method of EGFR mutation identification; 32.7% indicated that they only used targeted techniques and 45.1% used single-gene testing followed by next-generation sequencing (NGS), depending on the case. Reported testing rates were consistent over time with no significant decrease in the number of EGFR tests carried out in 2020, despite the increased pressure faced by testing facilities during the COVID-19 pandemic. ISO 15189 accreditation was reported by 42.0% of molecular biology laboratories for single-test sequencing, and by 42.3% for NGS. 92.5% of laboratories indicated they regularly participate in an external quality assessment scheme. CONCLUSIONS These results highlight the strong heterogeneity of EGFR testing that still occurs within thoracic pathology and molecular biology laboratories across Europe. Even among expert testing facilities there is variability in testing capabilities, TAT, reflex testing practice and laboratory accreditation, stressing the need to harmonise reimbursement technologies and decision-making algorithms in Europe.
Collapse
Affiliation(s)
- P Hofman
- Laboratory of Clinical and Experimental Pathology, FHU OncoAge, Biobank Côte d'Azur BB-0033-00025, Louis Pasteur Hospital, IRCAN, Université Côte d'Azur, Nice, France.
| | - F Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - I Kern
- Department of Pathology, University Clinic Golnik, Golnik, Slovenia
| | - J Adam
- Department of Pathology, Groupe Hospitalier Paris Saint-Joseph, Paris, France
| | - A Alarcão
- IAP-PM, Institute of Anatomical and Molecular Pathology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - I Alborelli
- Department of Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - N T Anton
- Department of Genetics, University Hospital Bichat-Claude Bernard, Paris University, Paris, France
| | - A Arndt
- Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081 Ulm, Germany
| | - A Avdalyan
- Multidisciplinary Clinical Center "Kommunarka" of the Moscow Health Department, Moscow, Russia
| | - M Barberis
- Oncogenomics Unit, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - H Bégueret
- Department of Pathology, University Hospital of Bordeaux, Hôpital Haut-Lévêque, Pessac, France
| | - B Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - H Blons
- Pharmacogenomics and Molecular Oncology Unit, Biochemistry Department, Assistance Publique-Hopitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - P Boström
- Department of Pathology, Turku University Hospital, Turku, Finland
| | - L Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - G Bubanovic
- Laboratory for Molecular Pathology, Department of Pathology, University of Zagreb School of Medicine and University Hospital Centre Zagreb, Zagreb, Croatia
| | - A Buisson
- Department of Biopathology, Centre Léon Bérard, Lyon, France
| | - A Caliò
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - M Cannone
- Inter-Hospital Pathology Division, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - L Carvalho
- IAP-PM, Institute of Anatomical and Molecular Pathology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - C Caumont
- Department of Tumor Biology, University Hospital of Bordeaux, Hospital Haut-Lévêque, Pessac, France
| | - A Cayre
- Department of Biopathology, Jean Perrin Centre, Clermont-Ferrand, France
| | - L Chalabreysse
- Department of Pathology, Groupement Hospitalier Est, Bron, France
| | - M P Chenard
- Department of Pathology, University Hospital of Strasbourg, 67098 Strasbourg, France
| | - E Conde
- Department of Pathology, 12 de Octubre University Hospital, Universidad Complutense de Madrid, Research Institute 12 de Octubre University Hospital (i+12), CIBERONC, Madrid, Spain
| | - M C Copin
- Department of Pathology, Université d'Angers, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - J F Côté
- Department of Pathology, Institut Mutualiste Montsouris, Paris, France
| | - N D'Haene
- Department of Pathology, Erasme Hospital, HUB ULB, Brussels, Belgium
| | - H Y Dai
- Department of Pathology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - L de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - P Delongova
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University Hospital Ostrava, Ostrava, Czech Republic
| | | | - A Fabre
- Department of Histopathology, St. Vincent's University Hospital, University College Dublin School of Medicine, Dublin, Ireland
| | - F Ferenc
- Department of Pathology, University of Oradea, Oradea, Romania
| | - F Forest
- Department of Pathology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - F de Fraipont
- Medical Unit of Molecular Genetic (Hereditary Diseases and Oncology), Grenoble University Hospital, Grenoble, France
| | - M Garcia-Martos
- Department of Pathology, Gregorio Marañón General University Hospital, Madrid, Spain
| | - G Gauchotte
- Department of Biopathology, CHRU-ICL, CHRU Nancy, Vandoeuvre-lès-Nancy, France
| | - R Geraghty
- Department of Histopathology, St. Vincent's University Hospital, University College Dublin School of Medicine, Dublin, Ireland
| | - E Guerin
- Department of Molecular Cancer Genetics, Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, Strasbourg, France
| | - D Guerrero
- Biomedical Research Centre, Navarra Health Service, Pamplona, Navarra, Spain
| | - S Hernandez
- Department of Pathology, 12 de Octubre University Hospital, Universidad Complutense de Madrid, Research Institute 12 de Octubre University Hospital (i+12), CIBERONC, Madrid, Spain
| | - P Hurník
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University Hospital Ostrava, Ostrava, Czech Republic
| | - B Jean-Jacques
- Department of Pathology, CHU de Caen Côte de Nacre, Caen, France
| | - K Kashofer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - D Kazdal
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - S Lantuejoul
- Department of Biopathology, Centre Leon Berard Unicancer and Pathology Research Platform, Cancer Research Center of Lyon (CRCL), Lyon, France
| | - C Leonce
- Department of Pathology, Groupement Hospitalier Est, Bron, France
| | - A Lupo
- Department of Pathology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, France
| | - U Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - R Matej
- Department of Pathology and Molecular Medicine, Thomayer University Hospital, Prague, Czech Republic
| | - J L Merlin
- Department of Biopathology, Institut de Cancérologie de Lorraine, University of Lorraine, Vandoeuvre-Les-Nancy, France
| | - K D Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - A Morel
- Department of Innate Immunity and Immunotherapy, Institut de Cancérologie de l'Ouest - Centre Paul Papin, Angers, France
| | - A Mutka
- HUSLAB, Department of Pathology, Helsinki University Hospital, Helsinki, Finland
| | - N Normanno
- Cell Biology and Biotherapy Unit, INT-Fondazione Pascale, Via M. Semmola, Naples, Italy
| | - P Ovidiu
- Department of Pathology, University of Oradea, Oradea, Romania
| | - A Panizo
- Department of Pathology, Complejo Hospitalario de Navarra, Pamplona, Navarra, Spain
| | - M G Papotti
- Division of Pathology, University Hospital Città Della Salute, Turin, Italy
| | - E Parobkova
- Department of Pathology and Molecular Medicine, Thomayer University Hospital, Prague, Czech Republic
| | - G Pasello
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - P Pauwels
- Department of Pathology, University Hospital Antwerp and University of Antwerp, Antwerp, Belgium
| | - G Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - F Penault-Llorca
- Department of Pathology, Clermont Auvergne University, "Molecular Imaging and Theranostic Strategies", Center Jean Perrin, Montalembert, Clermont-Ferrand, France
| | - T Picot
- Department of Pathology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - N Piton
- Department of Pathology, Rouen University Hospital, France and Normandie University, UNIROUEN, Inserm U1245, Rouen, France
| | - A Pittaro
- Division of Pathology, University Hospital Città Della Salute, Turin, Italy
| | - G Planchard
- Department of Pathology, CHU de Caen Côte de Nacre, Caen, France
| | - N Poté
- Department of Pathology, Hospital Bichat Bichat, Assistance Publique Hôpitaux de Paris; Université Paris Cité, Paris, France
| | - T Radonic
- Department of Pathology, Amsterdam University Medical Center, VUMC, University of Amsterdam, Amsterdam, Netherlands
| | - I Rapa
- Pathology Unit, San Luigi Hospital, Orbassano Turin, Italy
| | - A Rappa
- Oncogenomics Unit, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - C Roma
- Cell Biology and Biotherapy Unit, INT-Fondazione Pascale, Via M. Semmola, Naples, Italy
| | - M Rot
- Department of Pathology, University Clinic Golnik, Golnik, Slovenia
| | - J C Sabourin
- Department of Pathology, Rouen University Hospital, France and Normandie University, UNIROUEN, Inserm U1245, Rouen, France
| | - I Salmon
- Department of Pathology, Erasme Hospital, HUB ULB, Brussels, Belgium; CurePath, Jumet, Belgium
| | - S Savic Prince
- Department of Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - A Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - E Schuuring
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - I Serre
- Department of Pathology, Gui de Chauliac Hospital, Montpellier University Medical Center, University of Montpellier, 80 Avenue Augustin Fliche, Montpellier, France
| | - V Siozopoulou
- Department of Pathology, University Hospital Antwerp and University of Antwerp, Antwerp, Belgium
| | - D Sizaret
- Department of Pathology, CHRU Tours - Hôpital Trousseau, Chambray-lès-Tours, France
| | - S Smojver-Ježek
- Division for Pulmonary Cytology, Department of Pathology and Cytology, University Hospital Centre Zagreb, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - J Solassol
- Solid Tumour Laboratory, Pathology and Oncobiology Department, CHU Montpellier, University of Montpellier, Montpellier, France
| | - K Steinestel
- Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081 Ulm, Germany
| | - J Stojšić
- Department of Thoracic Pathology, Section of Pathology, University Clinical Centre of Serbia, Belgrade, Serbia
| | - C Syrykh
- Department of Pathology, IUC-T-Oncopole, Toulouse, France
| | - S Timofeev
- Multidisciplinary Clinical Center "Kommunarka" of the Moscow Health Department, Moscow, Russia
| | - G Troncone
- Department of Pathology, University of Oradea, Oradea, Romania
| | - A Uguen
- Department of Pathological Anatomy and Cytology, CHRU de Brest, Brest, France; LBAI, UMR1227, INSERM, University of Brest, CHU de Brest, Brest, France
| | - S Valmary-Degano
- Department of Pathology, Institute for Advanced Biosciences, CHU Grenoble Alpes, Université Grenoble Alpes, Grenoble, France
| | - A Vigier
- Department of Pathology, IUC-T-Oncopole, Toulouse, France
| | - M Volante
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - S G F Wahl
- Department of Pathology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - A Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - M Ilié
- Laboratory of Clinical and Experimental Pathology, FHU OncoAge, Biobank Côte d'Azur BB-0033-00025, Louis Pasteur Hospital, IRCAN, Université Côte d'Azur, Nice, France
| |
Collapse
|
3
|
Nauwelaers I, Laudus N, Peeters D, Acs B, Denkert C, Michiels S, Horlings H, Siziopikou KP, Ely S, Zardavas D, Mustimbo R, Bartlett J, Floris G, Hartman J, van Deurzen CHM, Ceusters D, Dequeker E, Salgado R. External Quality Assessment 2.0: The Importance of a Standardized Implementation of TILs for Daily and Trial Practices. Cancers (Basel) 2022; 14:cancers14153762. [PMID: 35954426 PMCID: PMC9367276 DOI: 10.3390/cancers14153762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
New assays are developed regularly to improve health care for patients. It is important to ensure that assays are performed correctly. Therefore, it is advised to participate in training and proficiency (competence assessment) programs. Tumor infiltrating lymphocytes (TILs) might improve the estimates of response to therapy and prognosis. Herewith, we propose a new training and proficiency program in which each pathologist can train and test themselves regarding TILs (and PDL1) scoring.
Collapse
Affiliation(s)
- Inne Nauwelaers
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Kapucijnenvoer 35d, 3000 Leuven, Belgium; (I.N.); (N.L.); (D.C.); (E.D.)
| | - Nele Laudus
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Kapucijnenvoer 35d, 3000 Leuven, Belgium; (I.N.); (N.L.); (D.C.); (E.D.)
| | - Dieter Peeters
- CellCarta NV, 2610 Antwerp, Belgium;
- Department of Pathology, AZ Sint-Maarten, 2800 Mechelen, Belgium
| | - Balazs Acs
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institute, 171 64 Stockholm, Sweden; (B.A.); (J.H.)
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Carsten Denkert
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg (UKGM), Baldingerstr. 1, 35043 Marburg, Germany;
| | - Stefan Michiels
- Department of Biostatistics and Epidemiology, Gustave Roussy, University Paris-Saclay, 94800 Villejuif, France;
- Oncostat U1018, Inserm, Labeled Ligue Contre le Cancer, University Paris-Saclay, 94800 Villejuif, France
| | - Hugo Horlings
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
- Department of Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Kalliopi P. Siziopikou
- Department of Pathology, Section of Breast Pathology, Northwestern University, Chicago, IL 60611, USA;
| | - Scott Ely
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ 08540, USA; (S.E.); (R.M.)
| | - Dimitrios Zardavas
- Oncology Clinical Development, Bristol-Myers Squibb, Princeton, NJ 08540, USA;
| | - Roberts Mustimbo
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ 08540, USA; (S.E.); (R.M.)
| | - John Bartlett
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Edinburgh EH4 2XR, UK;
| | - Giuseppe Floris
- Laboratory of Translational Cell & Tissue Research, Department of Imaging and Pathology, University of Leuven, 3000 Leuven, Belgium;
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Johan Hartman
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institute, 171 64 Stockholm, Sweden; (B.A.); (J.H.)
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | | | - Dorien Ceusters
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Kapucijnenvoer 35d, 3000 Leuven, Belgium; (I.N.); (N.L.); (D.C.); (E.D.)
| | - Els Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Kapucijnenvoer 35d, 3000 Leuven, Belgium; (I.N.); (N.L.); (D.C.); (E.D.)
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, 2610 Antwerp, Belgium
- Division of Research, Peter Mac Callum Cancer Centre, Melbourne, VIC 300, Australia
- Correspondence:
| |
Collapse
|
4
|
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
|
5
|
Fairley JA, Cheetham MH, Patton SJ, Rouleau E, Denis M, Dequeker EMC, Schuuring E, van Casteren K, Fenizia F, Normanno N, Deans ZC. Results of a worldwide external quality assessment of cfDNA testing in lung Cancer. BMC Cancer 2022; 22:759. [PMID: 35820813 PMCID: PMC9275131 DOI: 10.1186/s12885-022-09849-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/01/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Circulating cell free DNA (cfDNA) testing of plasma for EGFR somatic variants in lung cancer patients is being widely implemented and with any new service, external quality assessment (EQA) is required to ensure patient safety. An international consortium, International Quality Network for Pathology (IQNPath), has delivered a second round of assessment to measure the accuracy of cfDNA testing for lung cancer and the interpretation of the results. METHODS A collaboration of five EQA provider organisations, all members of IQNPath, have delivered the assessment during 2018-19 to a total of 264 laboratories from 45 countries. Bespoke plasma reference material containing a range of EGFR mutations at varying allelic frequencies were supplied to laboratories for testing and reporting according to routine procedures. The genotyping accuracy and clinical reporting was reviewed against standardised criteria and feedback was provided to participants. RESULTS The overall genotyping error rate in the EQA was found to be 11.1%. Low allelic frequency samples were the most challenging and were not detected by some testing methods, resulting in critical genotyping errors. This was reflected in higher false negative rates for samples with variant allele frequencies (VAF) rates less than 1.5% compared to higher frequencies. A sample with two different EGFR mutations gave inconsistent detection of both mutations. However, for one sample, where two variants were present at a VAF of less than 1% then both mutations were correctly detected in 145/263 laboratories. Reports often did not address the risk that tumour DNA may have not been tested and limitations of the methodologies provided by participants were insufficient. This was reflected in the average interpretation score for the EQA being 1.49 out of a maximum of 2. CONCLUSIONS The variability in the standard of genotyping and reporting highlighted the need for EQA and educational guidance in this field to ensure the delivery of high-quality clinical services where testing of cfDNA is the only option for clinical management.
Collapse
Affiliation(s)
- Jennifer A Fairley
- GenQA, Nine, Edinburgh Bioquarter, 9 Little France Road, Edinburgh, EH16 4SA, UK.
| | - Melanie H Cheetham
- EMQN CIC, Unit 4, Enterprise House, Pencroft Way, Manchester Science Park, Manchester, M15 6SE, UK
| | - Simon J Patton
- EMQN CIC, Unit 4, Enterprise House, Pencroft Way, Manchester Science Park, Manchester, M15 6SE, UK
| | - Etienne Rouleau
- Medical Biology and Pathology Department, Gustave Roussy, Villejuif, France
| | - Marc Denis
- Department of Biochemistry and INSERM U1232, Centre Hospitalier Universitaire de Nantes, 9 quai Moncousu, F-44093, Nantes Cedex, France
| | - Elisabeth M C Dequeker
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, KU Leuven, Kapucijnenvoer 35d, 3000, Leuven, Belgium
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Kaat van Casteren
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, KU Leuven, Kapucijnenvoer 35d, 3000, Leuven, Belgium
| | | | - Nicola Normanno
- Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Napoli, Italy
| | - Zandra C Deans
- GenQA, Nine, Edinburgh Bioquarter, 9 Little France Road, Edinburgh, EH16 4SA, UK
| |
Collapse
|
6
|
Keppens C, Dequeker EM, Pauwels P, Ryska A, 't Hart N, von der Thüsen JH. PD-L1 immunohistochemistry in non-small-cell lung cancer: unraveling differences in staining concordance and interpretation. Virchows Arch 2020; 478:827-839. [PMID: 33275169 PMCID: PMC8099807 DOI: 10.1007/s00428-020-02976-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 11/03/2020] [Accepted: 11/22/2020] [Indexed: 12/30/2022]
Abstract
Programmed death ligand 1 (PD-L1) immunohistochemistry (IHC) is accepted as a predictive biomarker for the selection of immune checkpoint inhibitors. We evaluated the staining quality and estimation of the tumor proportion score (TPS) in non-small-cell lung cancer during two external quality assessment (EQA) schemes by the European Society of Pathology. Participants received two tissue micro-arrays with three (2017) and four (2018) cases for PD-L1 IHC and a positive tonsil control, for staining by their routine protocol. After the participants returned stained slides to the EQA coordination center, three pathologists assessed each slide and awarded an expert staining score from 1 to 5 points based on the staining concordance. Expert scores significantly (p < 0.01) improved between EQA schemes from 3.8 (n = 67) to 4.3 (n = 74) on 5 points. Participants used 32 different protocols: the majority applied the 22C3 (56.7%) (Dako), SP263 (19.1%) (Ventana), and E1L3N (Cell Signaling) (7.1%) clones. Staining artifacts consisted mainly of very weak or weak antigen demonstration (63.0%) or excessive background staining (19.8%). Participants using CE-IVD kits reached a higher score compared with those using laboratory-developed tests (LDTs) (p < 0.05), mainly attributed to a better concordance of SP263. The TPS was under- and over-estimated in 20/423 (4.7%) and 24/423 (5.7%) cases, respectively, correlating to a lower expert score. Additional research is needed on the concordance of less common protocols, and on reasons for lower LDT concordance. Laboratories should carefully validate all test methods and regularly verify their performance. EQA participation should focus on both staining concordance and interpretation of PD-L1 IHC.
Collapse
Affiliation(s)
- Cleo Keppens
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Elisabeth Mc Dequeker
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Patrick Pauwels
- Center for Oncologic Research (CORE), University of Antwerp, Antwerp, Belgium.,Department of Pathology, University Hospital Antwerp, Edegem, Belgium
| | - Ales Ryska
- Department of Pathology, Charles University Medical Faculty and University Hospital, Hradec Kralove, Czech Republic
| | - Nils 't Hart
- Department of Pathology, University Medical Center Groningen, Groningen, The Netherlands.,Department of Pathology, Isala Klinieken, Zwolle, The Netherlands
| | - Jan H von der Thüsen
- Department of Pathology, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
Keppens C, von der Thüsen J, Pauwels P, Ryska A, 't Hart N, Schuuring E, Miller K, Thunnissen E, Zwaenepoel K, Dequeker EMC. Staining Performance of ALK and ROS1 Immunohistochemistry and Influence on Interpretation in Non-Small-Cell Lung Cancer. J Mol Diagn 2020; 22:1438-1452. [PMID: 33011443 DOI: 10.1016/j.jmoldx.2020.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/15/2020] [Accepted: 09/16/2020] [Indexed: 01/02/2023] Open
Abstract
Selection of non-small-cell lung cancer patients for treatment relies on the detection of expression of anaplastic lymphoma kinase (ALK) and ROS proto-oncogene 1 (ROS1) protein by immunohistochemistry (IHC). We evaluated staining performance for different IHC protocols and laboratory characteristics, and their influence on ALK and ROS1 interpretation during external quality assessment schemes between 2015 and 2018. Participants received five formalin-fixed, paraffin-embedded cases for staining by their routine protocol, whereafter at least two pathologists scored them simultaneously under a multihead microscope and awarded a graded expert staining score (ESS) from 1 to 5 points based on staining quality. European Conformity in Vitro Diagnostic kits (such as D5F3) revealed a better ALK ESS compared with laboratory-developed tests. ESS was indifferent to the applied antibody dilution or a recent protocol change. Lower ESSs were observed for higher antibody incubation times and temperatures. ESS for various ROS1 protocols were largely similar. Overall, for both markers, ESS improved over time and for repeated external quality assessment participation but was independent of laboratory setting or experience. Except for ROS1, ESS positively correlated with laboratory accreditation. IHC stains with lower ESS correlated with increased error rates in ALK and ROS1 interpretation and analysis failures. Laboratory characteristics differently affected staining quality and interpretation, and laboratories should assess both aspects, and less common protocols need improvement in staining performance.
Collapse
Affiliation(s)
- Cleo Keppens
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Jan von der Thüsen
- Department of Pathology, Erasmus Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Patrick Pauwels
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium; Centre for Oncological Research, University of Antwerp, Edegem, Belgium
| | - Ales Ryska
- Department of Pathology, Charles University Medical Faculty Hospital, Hradec Kralove, Czech Republic
| | - Nils 't Hart
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Pathology, Isala Klinieken, Zwolle, the Netherlands
| | - Ed Schuuring
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Keith Miller
- UK National External Quality Assessment Scheme for Immunocytochemistry and in Situ Hybridisation, University College London Cancer Institute, London, United Kingdom
| | - Erik Thunnissen
- Department of Pathology, Vrije Universiteit Amsterdam Medical Center, Amsterdam, the Netherlands
| | - Karen Zwaenepoel
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium; Centre for Oncological Research, University of Antwerp, Edegem, Belgium
| | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium.
| |
Collapse
|
11
|
Brcic L, Kern I. Outsourcing predictive biomarker testing in non-small cell carcinoma: a personal view of pathologists. Transl Lung Cancer Res 2020; 9:2194-2198. [PMID: 33209643 PMCID: PMC7653138 DOI: 10.21037/tlcr-20-297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Izidor Kern
- Cytology and Pathology Laboratory, University Clinic of Respiratory and Allergic Diseases, Golnik, Slovenia
| |
Collapse
|
12
|
Laudus N, Audrézet MP, Girodon E, Morris MA, Radojkovic D, Raynal C, Seia M, Štambergová A, Torkler H, Yamamoto R, Dequeker EMC. Laboratory reporting on the clinical spectrum of CFTR p.Arg117His: Still room for improvement. J Cyst Fibros 2020; 19:969-974. [PMID: 32505523 DOI: 10.1016/j.jcf.2020.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND The clinical spectrum associated with cystic fibrosis transmembrane conductance regulator (CFTR) variant p.Arg117His is highly variable, ranging from full-blown cystic fibrosis (CF) in a small number of cases to CFTR-related disorders (CFTR-RDs) or no symptoms at all. Therefore, taking into account phenotype variability is essential for interpretation. External quality assessment (EQA) schemes can help laboratories to objectively assess the quality of genotyping and reporting by the laboratory. METHODS We performed a retrospective longitudinal data analysis on laboratory performance regarding the interpretation of p.Arg117His during CF EQA scheme participation. Completeness and accuracy of reporting on two mock clinical cases were each compared over time (case 1: 2005, 2007 and 2012; case 2: 2015 and 2018). These cases concerned subjects compound heterozygous for p.Phe508del and p.Arg117His in cis with 7T, but with different clinical backgrounds (family planning (case 1) versus diagnostic testing for a child (case 2)). Furthermore, we analyzed the influence of previous participations, annual test volume, accreditation status and laboratory setting on overall performance. RESULTS Overall performance improved over time, except during the 2007 CF EQA scheme. In addition, previous participations had a beneficial effect on laboratory performance. Accreditation status, annual test volume and laboratory setting did not significantly influence total interpretation scores. CONCLUSIONS In general, laboratories performed well on both cases, although reporting on the variable clinical spectrum of p.Arg117His in cis with 7T and on the disease liability of individual CFTR variants can still improve. Moreover, this study underlined the educational role of CF EQA schemes.
Collapse
Affiliation(s)
- Nele Laudus
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | | | - Emmanuelle Girodon
- Laboratoire de Génétique et Biologie Moléculaires, AP-HP.Centre-Université de Paris, Hôpital Cochin, Paris, France
| | | | - Dragica Radojkovic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Caroline Raynal
- Laboratoire de Génétique Moléculaire, CHU de Montpellier, Montpellier, France
| | - Manuela Seia
- Laboratorio di Genetica Medica - Settore di Genetica Molecolare, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alexandra Štambergová
- Department of Biology and Medical Genetics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - Heike Torkler
- MVZ Dr. Eberhard & Partner Dortmund, Dortmund, Germany
| | | | - 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 Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
van den Broek D, Hiltermann TJN, Biesma B, Dinjens WNM, 't Hart NA, Hinrichs JWJ, Leers MPG, Monkhorst K, van Oosterhout M, Scharnhorst V, Schuuring E, Speel EJM, van den Heuvel MM, van Schaik RHN, von der Thüsen J, Willems SM, de Visser L, Ligtenberg MJL. Implementation of Novel Molecular Biomarkers for Non-small Cell Lung Cancer in the Netherlands: How to Deal With Increasing Complexity. Front Oncol 2020; 9:1521. [PMID: 32039011 PMCID: PMC6987414 DOI: 10.3389/fonc.2019.01521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/17/2019] [Indexed: 12/30/2022] Open
Abstract
The diagnostic landscape of non-small cell lung cancer (NSCLC) is changing rapidly with the availability of novel treatments. Despite high-level healthcare in the Netherlands, not all patients with NSCLC are tested with the currently relevant predictive tumor markers that are necessary for optimal decision-making for today's available targeted or immunotherapy. An expert workshop on the molecular diagnosis of NSCLC involving pulmonary oncologists, clinical chemists, pathologists, and clinical scientists in molecular pathology was held in the Netherlands on December 10, 2018. The aims of the workshop were to facilitate cross-disciplinary discussions regarding standards of practice, and address recent developments and associated challenges that impact future practice. This paper presents a summary of the discussions and consensus opinions of the workshop participants on the initial challenges of harmonization of the detection and clinical use of predictive markers of NSCLC. A key theme identified was the need for broader and active participation of all stakeholders involved in molecular diagnostic services for NSCLC, including healthcare professionals across all disciplines, the hospitals and clinics involved in service delivery, healthcare insurers, and industry groups involved in diagnostic and treatment innovations. Such collaboration is essential to integrate different technologies into molecular diagnostics practice, to increase nationwide patient access to novel technologies, and to ensure consensus-preferred biomarkers are tested.
Collapse
Affiliation(s)
- Daan van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - T. Jeroen N. Hiltermann
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bonne Biesma
- Department of Pulmonary Diseases, Jeroen Bosch Hospital, 's-Hertogenbosch, Netherlands
| | - Winand N. M. Dinjens
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Nils A. 't Hart
- Department of Pathology, Isala Klinieken, Zwolle, Netherlands
| | - John W. J. Hinrichs
- Symbiant Pathology Expert Centre, Alkmaar, Netherlands
- Department of Pathology, University Medical Center, Utrecht, Netherlands
| | - Mathie P. G. Leers
- Department of Clinical Chemistry, Zuyderland Medical Center, Sittard-Geleen, Netherlands
| | - Kim Monkhorst
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | - Ed Schuuring
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ernst-Jan M. Speel
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | | | - Ron H. N. van Schaik
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jan von der Thüsen
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Stefan M. Willems
- Department of Pathology, University Medical Center, Utrecht, Netherlands
| | | | - Marjolijn J. L. Ligtenberg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Pathology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
17
|
Aysal A, Pehlivanoğlu B, Ekmekci S, Gündoğdu B. How to Set Up a Molecular Pathology Lab: A Guide for Pathologists. Turk Patoloji Derg 2020; 36:179-187. [PMID: 32525209 PMCID: PMC10510618 DOI: 10.5146/tjpath.2020.01488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/09/2020] [Indexed: 11/18/2022] Open
Abstract
In today's pathology practice, pathologists combine molecular tests with conventional histopathological methods. Pathology laboratories should therefore be designed and operated in accordance with the requirements of molecular testing procedures. While the specifics of the requirements may vary depending on the spectrum of the tests that will be performed, there are several basic criteria that need to be fulfilled for standardization. Adequate space, appropriate equipment and qualified personnel are required to establish a molecular pathology laboratory. One of the most important points that should be taken into consideration while designing a molecular pathology laboratory is to create a plan to prevent contamination. As molecular diagnosis has a major role in treatment decisions, the management of the molecular pathology laboratory is of utmost importance. In this review, the criteria required to establish an optimal molecular pathology laboratory will be reviewed.
Collapse
Affiliation(s)
- Anıl Aysal
- Department of Molecular Pathology, Dokuz Eylul University, Graduate School of Health Sciences, Izmir, Turkey
| | - Burçin Pehlivanoğlu
- Department of Molecular Pathology, Dokuz Eylul University, Graduate School of Health Sciences, Izmir, Turkey
| | - Sümeyye Ekmekci
- Department of Molecular Pathology, Dokuz Eylul University, Graduate School of Health Sciences, Izmir, Turkey
| | - Betül Gündoğdu
- Department of Molecular Pathology, Dokuz Eylul University, Graduate School of Health Sciences, Izmir, Turkey
| |
Collapse
|
18
|
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
|