1
|
van Opijnen MP, Broekman MLD, Cuppen E, Dubbink HJ, Ter Elst A, van Eijk R, Mühlebner A, Jansen C, van der Geize R, Speel EJM, Groenen PJTA, de Vos FYF, Wesseling P, de Leng WWJ, Maas SLN. Next generation sequencing of high-grade adult-type diffuse glioma in the Netherlands: interlaboratory variation in the primary diagnostic and recurrent setting. J Neurooncol 2024; 166:485-492. [PMID: 38285243 PMCID: PMC10876806 DOI: 10.1007/s11060-024-04568-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/09/2024] [Indexed: 01/30/2024]
Abstract
PURPOSE Next generation sequencing (NGS) is an important tool used in clinical practice to obtain the required molecular information for accurate diagnostics of high-grade adult-type diffuse glioma (HGG). Since individual centers use either in-house produced or standardized panels, interlaboratory variation could play a role in the practice of HGG diagnosis and treatment. This study aimed to investigate the current practice in NGS application for both primary and recurrent HGG. METHODS This nationwide Dutch survey used the expertise of (neuro)pathologists and clinical scientists in molecular pathology (CSMPs) by sending online questionnaires on clinical and technical aspects. Primary outcome was an overview of panel composition in the different centers for diagnostic practice of HGG. Secondary outcomes included practice for recurrent HGG and future perspectives. RESULTS Out of twelve neuro-oncology centers, the survey was filled out by eleven (neuro)pathologists and seven CSMPs. The composition of the diagnostic NGS panels differed in each center with numbers of genes ranging from 12 to 523. Differences are more pronounced when tests are performed to find therapeutic targets in the case of recurrent disease: about half of the centers test for gene fusions (60%) and tumor mutational burden (40%). CONCLUSION Current notable interlaboratory variations as illustrated in this study should be reduced in order to refine diagnostics and improve precision oncology. In-house developed tests, standardized panels and routine application of broad gene panels all have their own advantages and disadvantages. Future research would be of interest to study the clinical impact of variation in diagnostic approaches.
Collapse
Affiliation(s)
- Mark P van Opijnen
- Department of Neurosurgery, Haaglanden Medical Center, The Hague, The Netherlands.
- Department of Neurosurgery, Leiden University Medical Center, Leiden, The Netherlands.
| | - Marike L D Broekman
- Department of Neurosurgery, Haaglanden Medical Center, The Hague, The Netherlands
- Department of Neurosurgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Edwin Cuppen
- Hartwig Medical Foundation, Amsterdam, The Netherlands
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hendrikus J Dubbink
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Arja Ter Elst
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Ronald van Eijk
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Angelika Mühlebner
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Casper Jansen
- Laboratorium Pathologie Oost-Nederland, Hengelo, The Netherlands
| | | | - Ernst-Jan M Speel
- Department of Pathology, GROW-School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Filip Y F de Vos
- Department of Medical Oncology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Wendy W J de Leng
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sybren L N Maas
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Learning mechanisms and outcomes of an interprofessional molecular pathology workshop for residents. Acad Pathol 2022; 9:100056. [PMID: 36281273 PMCID: PMC9587361 DOI: 10.1016/j.acpath.2022.100056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/10/2022] [Accepted: 09/03/2022] [Indexed: 11/06/2022] Open
Abstract
The developments in targeted therapies and molecular pathology have changed the classification of tumors and precision oncology. Pathologists and clinical scientists in molecular pathology and oncologists have regular multidisciplinary meetings and are responsible for translating molecular results into an appropriate treatment plan. This requires expertise and skills to be effective team players. Interprofessional collaboration (IPC) is essential for professionals in medicine; however, learning opportunities in current resident training are limited. This narrative study explores the collaborative output and learning mechanisms of interprofessional learning (IPL) of residents of different disciplines in the Morphology & MolecularPLUS workshop and its preparation. Topics that were discussed in the workshop were technologies for the detection of mutations, copy number variations, tumor mutational burden, and circulating tumor DNA (ctDNA) analysis in the context of differential diagnosis and precision oncology. Data were collected by analyzing pre- and post-workshop questionnaires and interviews. An interprofessional team of three residents of each hospital had to be formed by one of the residents, which was challenging as not all residents from a hospital knew each other. Residents reported to have got to know each other and have learned about each other's roles and perspectives. They gained knowledge of molecular pathology and the added value of IPC, in particular, for residents early in their training. Enabling meetings for medical residents of different disciplines to get acquainted was perceived as the most facilitating factor for IPL. Time constraints as the biggest barrier in daily practice. We recommend offering IPL activities as early as possible in residency programs.
Collapse
|
3
|
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
|
4
|
"The leading role of pathology in assessing the somatic molecular alterations of cancer: Position Paper of the European Society of Pathology": letter to the Editor. Virchows Arch 2020; 478:379-380. [PMID: 32761392 PMCID: PMC7969541 DOI: 10.1007/s00428-020-02898-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
5
|
Gullo I, Marques A, Pinto R, Cirnes L, Schmitt F. Morphological control for molecular testing: a practical approach. J Clin Pathol 2020; 74:331-333. [PMID: 32763918 DOI: 10.1136/jclinpath-2020-206890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 11/04/2022]
Abstract
The determination of molecular aberrations within tumours is important for diagnostic, prognostic and predictive purposes. Pathologists play a critical role in the workflow of molecular diagnostics, by assuring accurate pathological diagnosis, requesting appropriate molecular testing, selecting the adequate tissue section for molecular analysis, enriching tumour cell content by manual macrodissection and estimating the tumour cellularity. Particularly, the assessment of the malignant cell fraction within a tumour section is a key determinant for an appropriate interpretation of the molecular findings. Several factors may impact the estimation of tumour cellularity and constitute a potential pitfall for the final interpretation of the molecular analysis. Evidence suggests that the reliability of morphological control could be improved by training. The scope of this commentary is to provide the training morpho-molecular pathologists with the practical tools necessary to master microscopic morphological control for solid tumours, as well as a set of images that could serve as a training set.
Collapse
Affiliation(s)
- Irene Gullo
- Department of Pathology, CHUSJ - Centro Hospitalar Universitário de São João EPE, Porto, Portugal.,Department of Pathology, FMUP - Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Ana Marques
- Department of Pathology, CHUSJ - Centro Hospitalar Universitário de São João EPE, Porto, Portugal.,Department of Pathology, FMUP - Faculty of Medicine of the University of Porto, Porto, Portugal.,IPATIMUP Diagnostics, IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
| | - Regina Pinto
- IPATIMUP Diagnostics, IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
| | - Luis Cirnes
- IPATIMUP Diagnostics, IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
| | - Fernando Schmitt
- Department of Pathology, FMUP - Faculty of Medicine of the University of Porto, Porto, Portugal .,IPATIMUP Diagnostics, IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
| |
Collapse
|
6
|
Albano L, Losa M, Barzaghi LR, Spatola G, Panni P, Terreni MR, Mortini P. Primary sellar melanocytoma: pathological, clinical and treatment review. J Endocrinol Invest 2020; 43:575-585. [PMID: 31797309 DOI: 10.1007/s40618-019-01158-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/28/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND Sellar melanocytomas represent a small subgroup of primary melanocytic tumors. They arise from melanocytes located in the meningeal lining of the sellar floor or in the diaphragma sellae and this location is very uncommon. Usually, sellar melanocytomas are benign and slow-growing tumors with a high likelihood of recurrence. PURPOSE To our knowledge, due to the rarity of this condition, there are no guidelines regarding their diagnosis and treatment in the medical literature to date. We have developed a narrative review, analyzing the available studies regarding primary sellar melanocytomas reported in the medical literature. We have found ten papers on this topic and all of them are case reports. In all patients, tumor diagnosis was performed after the occurrence of neurological symptoms, in particular progressive visual loss or endocrinological disorders. The diagnosis is difficult, and it requires several preoperative and postoperative investigations, but histological examination is crucial. CONCLUSIONS Transsphenoidal surgery is the first-choice treatment. In case of tumor's recurrence or regrowth, the role of radiation therapy and chemotherapy is not entirely clear.
Collapse
Affiliation(s)
- L Albano
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20132, Milan, Italy.
| | - M Losa
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20132, Milan, Italy
| | - L R Barzaghi
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20132, Milan, Italy
| | - G Spatola
- Department of Neurosurgery, Assistance Publique Hopitaux de Marseille, Marseille, France
| | - P Panni
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20132, Milan, Italy
| | - M R Terreni
- Department of Pathology, I.R.C.C.S. San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy
| | - P Mortini
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
7
|
Matias-Guiu X, Stanta G, Carneiro F, Ryska A, Hoefler G, Moch H. The leading role of pathology in assessing the somatic molecular alterations of cancer: Position Paper of the European Society of Pathology. Virchows Arch 2020; 476:491-497. [PMID: 32124002 PMCID: PMC7156353 DOI: 10.1007/s00428-020-02757-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 01/05/2023]
Abstract
Molecular pathology is an essential part of pathology complementing conventional morphological tools to obtain a correct integrated diagnosis with appropriate assessment of prognosis and prediction of response to therapy, particularly in cancer. There is a concern about the situation of molecular pathology in some areas of Europe, namely, regarding the central role of pathologists in assessing somatic genomic alterations in cancer. In some countries, there are attempts that other laboratory medicine specialists perform the molecular analysis of somatic alterations in cancer, particularly now when next generation sequencing (NGS) is incorporated into clinical practice. In this scenario, pathologists may play just the role of “tissue providers,” and other specialists may take the lead in molecular analysis. Geneticists and laboratory medicine specialists have all background and skills to perform genetic analysis of germline alterations in hereditary disorders, including familial forms of cancers. However, interpretation of somatic alterations of cancer belongs to the specific scientific domain of pathology. Pathologists are necessary to guarantee the quality of the results, for several reasons: (1) The identified molecular alterations should be interpreted in the appropriate morphologic context, since most of them are context-specific; (2) pre-analytical issues must be taken into consideration; (3) it is crucial to check the proportion of tumor cells in the sample subjected to analysis and presence of inflammatory infiltrate and necrosis should be monitored; and 4) the role of pathologists is crucial to select the most appropriate methods and to control the turnaround time in which the molecular results are delivered in the context of an integrated diagnosis. Obviously, there is the possibility of having core facilities for NGS in a hospital to perform the sequence analysis that are open to other specialties (microbiologists, geneticists), but also in this scenario, pathologists should have the lead in assessing somatic alterations of cancer. In this article, we emphasize the importance of interpreting somatic molecular alterations of the tumors in the context of morphology. In this Position Paper of the European Society of Pathology, we strongly support a central role of pathology departments in the process of analysis and interpretation of somatic molecular alterations in cancer.
Collapse
Affiliation(s)
- Xavier Matias-Guiu
- Hospital Universitari Arnau de Vilanova. Universitat de Lleida, IRBLleida. CIBERONC, Hospital U de Bellvitge. IDIBELL, University of Barcelona, Av Rovira Roure, 80, 25198, Lleida, Spain.
| | - Giorgio Stanta
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Fátima Carneiro
- Department of Pathology, Medical Faculty of the University of Porto/Centro Hospitalar Universitário São João and Ipatimup/i3S, Porto, Portugal
| | - Ales Ryska
- The Fingerland Department of Pathology, Charles University Medical Faculty and University Hospital, Hradec Kralove, Czech Republic
| | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, D&R Center of Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Holger Moch
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
8
|
Simarro J, Murria R, Pérez-Simó G, Llop M, Mancheño N, Ramos D, Juan ID, Barragán E, Laiz B, Cases E, Ansótegui E, Gómez-Codina J, Aparicio J, Salvador C, Juan Ó, Palanca S. Development, Implementation and Assessment of Molecular Diagnostics by Next Generation Sequencing in Personalized Treatment of Cancer: Experience of a Public Reference Healthcare Hospital. Cancers (Basel) 2019; 11:E1196. [PMID: 31426418 PMCID: PMC6721584 DOI: 10.3390/cancers11081196] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023] Open
Abstract
The establishment of precision medicine in cancer patients requires the study of several biomarkers. Single-gene testing approaches are limited by sample availability and turnaround time. Next generation sequencing (NGS) provides an alternative for detecting genetic alterations in several genes with low sample requirements. Here we show the implementation to routine diagnostics of a NGS assay under International Organization for Standardization (UNE-EN ISO 15189:2013) accreditation. For this purpose, 106 non-small cell lung cancer (NSCLC) and 102 metastatic colorectal cancer (mCRC) specimens were selected for NGS analysis with Oncomine Solid Tumor (ThermoFisher). In NSCLC the most prevalently mutated gene was TP53 (49%), followed by KRAS (31%) and EGFR (13%); in mCRC, TP53 (50%), KRAS (48%) and PIK3CA (16%) were the most frequently mutated genes. Moreover, NGS identified actionable genetic alterations in 58% of NSCLC patients, and 49% of mCRC patients did not harbor primary resistance mechanisms to anti-EGFR treatment. Validation with conventional approaches showed an overall agreement >90%. Turnaround time and cost analysis revealed that NGS implementation is feasible in the public healthcare context. Therefore, NGS is a multiplexed molecular diagnostic tool able to overcome the limitations of current molecular diagnosis in advanced cancer, allowing an improved and economically sustainable molecular profiling.
Collapse
Affiliation(s)
- Javier Simarro
- Molecular Biology Unit, Service of Clinical Analysis, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
- Clinical and Translational Cancer Research Group, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Rosa Murria
- Molecular Biology Unit, Service of Clinical Analysis, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
- Clinical and Translational Cancer Research Group, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Gema Pérez-Simó
- Molecular Biology Unit, Service of Clinical Analysis, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
- Clinical and Translational Cancer Research Group, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Marta Llop
- Molecular Biology Unit, Service of Clinical Analysis, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - Nuria Mancheño
- Department of Pathology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - David Ramos
- Department of Pathology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - Inmaculada de Juan
- Molecular Biology Unit, Service of Clinical Analysis, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
- Clinical and Translational Cancer Research Group, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Eva Barragán
- Molecular Biology Unit, Service of Clinical Analysis, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - Begoña Laiz
- Molecular Biology Unit, Service of Clinical Analysis, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - Enrique Cases
- Department of Pulmonology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - Emilio Ansótegui
- Department of Pulmonology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - José Gómez-Codina
- Clinical and Translational Cancer Research Group, Health Research Institute La Fe, 46026 Valencia, Spain
- Department of Medical Oncology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - Jorge Aparicio
- Clinical and Translational Cancer Research Group, Health Research Institute La Fe, 46026 Valencia, Spain
- Department of Medical Oncology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - Carmen Salvador
- Clinical and Translational Cancer Research Group, Health Research Institute La Fe, 46026 Valencia, Spain
- Department of Medical Oncology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - Óscar Juan
- Department of Medical Oncology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | - Sarai Palanca
- Molecular Biology Unit, Service of Clinical Analysis, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain.
- Clinical and Translational Cancer Research Group, Health Research Institute La Fe, 46026 Valencia, Spain.
| |
Collapse
|
9
|
Crisafulli C, Romeo PD, Calabrò M, Epasto LM, Alberti S. Pharmacogenetic and pharmacogenomic discovery strategies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:225-241. [PMID: 35582724 PMCID: PMC8992635 DOI: 10.20517/cdr.2018.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 11/12/2022]
Abstract
Genetic/genomic profiling at a single-patient level is expected to provide critical information for determining inter-individual drug toxicity and potential efficacy in cancer therapy. A better definition of cancer subtypes at a molecular level, may correspondingly complement such pharmacogenetic and pharmacogenomic approaches, for more effective personalized treatments. Current pharmacogenetic/pharmacogenomic strategies are largely based on the identification of known polymorphisms, thus limiting the discovery of novel or rarer genetic variants. Recent improvements in cost and throughput of next generation sequencing (NGS) are now making whole-genome profiling a plausible alternative for clinical procedures. Beyond classical pharmacogenetic/pharmacogenomic traits for drug metabolism, NGS screening programs of cancer genomes may lead to the identification of novel cancer-driving mutations. These may not only constitute novel therapeutic targets, but also effector determinants for metabolic pathways linked to drug metabolism. An additional advantage is that cancer NGS profiling is now leading to discovering targetable mutations, e.g., in glioblastomas and pancreatic cancers, which were originally discovered in other tumor types, thus allowing for effective repurposing of active drugs already on the market.
Collapse
Affiliation(s)
- Concetta Crisafulli
- Department of Biomedical Sciences - BIOMORF, University of Messina, via Consolare Valeria, 98125 Messina, Italy
| | | | - Marco Calabrò
- Department of Biomedical Sciences - BIOMORF, University of Messina, via Consolare Valeria, 98125 Messina, Italy
| | - Ludovica Martina Epasto
- Unit of Medical Genetics, University of Messina, via Consolare Valeria, 98125 Messina, Italy
| | - Saverio Alberti
- Department of Biomedical Sciences - BIOMORF, University of Messina, via Consolare Valeria, 98125 Messina, Italy.,Unit of Medical Genetics, University of Messina, via Consolare Valeria, 98125 Messina, Italy.,Correspondence Address: Prof. Saverio Alberti, Unit of Medical Genetics, BIOMORF Department of Biomedical Sciences, University of Messina, via Consolare Valeria, 98125 Messina, Italy. E-mail:
| |
Collapse
|
10
|
van der Werf MJ, Ködmön C. Whole-Genome Sequencing as Tool for Investigating International Tuberculosis Outbreaks: A Systematic Review. Front Public Health 2019; 7:87. [PMID: 31058125 PMCID: PMC6478655 DOI: 10.3389/fpubh.2019.00087] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/01/2019] [Indexed: 12/31/2022] Open
Abstract
Background: Whole-genome sequencing (WGS) can support the investigation of tuberculosis (TB) outbreaks. The technique has been applied to estimate the timing and directionality of transmission and to exclude cases from an investigation. This review assesses how WGS was applied in international outbreak investigations and discusses the advantages and challenges of the application of WGS. Methods: Databases were searched for reports on international TB outbreak investigations. Information was extracted on: Why was WGS applied?; How was WGS applied?; Organizational issues; WGS methodology; What was learned/what were the implications of the WGS investigation?; and challenges and lessons learned. Results: Three studies reporting on international outbreak investigations were identified. Retrospective WGS sequencing was performed in all studies and prospective typing in two to study TB transmission. In one study, WGS data were produced centrally (i.e., in one laboratory) and analysis was done centrally. In two studies, WGS data production was done in a decentralized manner, and analysis was centralized in one laboratory. Three groups of professionals were involved in the international outbreak investigation: public health authorities, laboratory experts, and clinicians. The reported WGS methodology applied differed between the studies in some aspects, e.g., sequencing platform; quality measures, percentage of the reference genome covered, and the mean genomic coverage; analysis, use of a reference genome or de novo assembly; and software used for alignment and analysis. In all three studies, in-house scripts were used for variance calling, and the single nucleotide polymorphism (SNP) approach was used for analysis. All outbreak investigation reports stated that WGS refuted suspected transmission events and provided supporting evidence for epidemiological data. Several challenges were reported of which most were not related to WGS. The only challenge related to WGS was the timeframe of getting WGS data if WGS is not routinely performed. Conclusions: WGS was considered a useful addition in international TB outbreak investigations. Further standardization of the WGS methodology and good structures for international collaboration and coordination are needed to take full advantage of this new technology. Whether the use of WGS results in earlier detection of cases and thus limits transmission still needs to be determined.
Collapse
Affiliation(s)
| | - Csaba Ködmön
- European Centre for Disease Prevention and Control, Stockholm, Sweden
| |
Collapse
|
11
|
Neagu AN. Proteome Imaging: From Classic to Modern Mass Spectrometry-Based Molecular Histology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:55-98. [PMID: 31347042 DOI: 10.1007/978-3-030-15950-4_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In order to overcome the limitations of classic imaging in Histology during the actually era of multiomics, the multi-color "molecular microscope" by its emerging "molecular pictures" offers quantitative and spatial information about thousands of molecular profiles without labeling of potential targets. Healthy and diseased human tissues, as well as those of diverse invertebrate and vertebrate animal models, including genetically engineered species and cultured cells, can be easily analyzed by histology-directed MALDI imaging mass spectrometry. The aims of this review are to discuss a range of proteomic information emerging from MALDI mass spectrometry imaging comparative to classic histology, histochemistry and immunohistochemistry, with applications in biology and medicine, concerning the detection and distribution of structural proteins and biological active molecules, such as antimicrobial peptides and proteins, allergens, neurotransmitters and hormones, enzymes, growth factors, toxins and others. The molecular imaging is very well suited for discovery and validation of candidate protein biomarkers in neuroproteomics, oncoproteomics, aging and age-related diseases, parasitoproteomics, forensic, and ecotoxicology. Additionally, in situ proteome imaging may help to elucidate the physiological and pathological mechanisms involved in developmental biology, reproductive research, amyloidogenesis, tumorigenesis, wound healing, neural network regeneration, matrix mineralization, apoptosis and oxidative stress, pain tolerance, cell cycle and transformation under oncogenic stress, tumor heterogeneity, behavior and aggressiveness, drugs bioaccumulation and biotransformation, organism's reaction against environmental penetrating xenobiotics, immune signaling, assessment of integrity and functionality of tissue barriers, behavioral biology, and molecular origins of diseases. MALDI MSI is certainly a valuable tool for personalized medicine and "Eco-Evo-Devo" integrative biology in the current context of global environmental challenges.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, "Alexandru Ioan Cuza" University of Iasi, Iasi, Romania.
| |
Collapse
|
12
|
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
|
13
|
Fassan M. Molecular Diagnostics in Pathology: Time for a Next-Generation Pathologist? Arch Pathol Lab Med 2018; 142:313-320. [DOI: 10.5858/arpa.2017-0269-ra] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Context.—Comprehensive molecular investigations of mainstream carcinogenic processes have led to the use of effective molecular targeted agents in most cases of solid tumors in clinical settings.Objective.—To update readers regarding the evolving role of the pathologist in the therapeutic decision-making process and the introduction of next-generation technologies into pathology practice.Data Sources.—Current literature on the topic, primarily sourced from the PubMed (National Center for Biotechnology Information, Bethesda, Maryland) database, were reviewed.Conclusions.—Adequate evaluation of cytologic-based and tissue-based predictive diagnostic biomarkers largely depends on both proper pathologic characterization and customized processing of biospecimens. Moreover, increased requests for molecular testing have paralleled the recent, sharp decrease in tumor material to be analyzed—material that currently comprises cytology specimens or, at minimum, small biopsies in most cases of metastatic/advanced disease. Traditional diagnostic pathology has been completely revolutionized by the introduction of next-generation technologies, which provide multigene, targeted mutational profiling, even in the most complex of clinical cases. Combining traditional and molecular knowledge, pathologists integrate the morphological, clinical, and molecular dimensions of a disease, leading to a proper diagnosis and, therefore, the most-appropriate tailored therapy.
Collapse
Affiliation(s)
- Matteo Fassan
- From the Department of Medicine, Surgical Pathology and Cytopathology Unit, University of Padua, Padua, Italy
| |
Collapse
|
14
|
Haynes HR, Killick-Cole CL, Hares KM, Redondo J, Kemp KC, Moutasim KA, Faulkner C, Wilkins A, Kurian KM. Evaluation of the quality of RNA extracted from archival FFPE glioblastoma and epilepsy surgical samples for gene expression assays. J Clin Pathol 2018; 71:695-701. [DOI: 10.1136/jclinpath-2017-204969] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 01/05/2023]
Abstract
AimsHistopathological tissue samples are being increasingly used as sources of nucleic acids in molecular pathology translational research. This study investigated the suitability of glioblastoma and control central nervous system (CNS) formalin-fixed paraffin embedded (FFPE) tissue-derived RNA for gene expression analyses.MethodsTotal RNA was extracted from control (temporal lobe resection tissue) and glioblastoma FFPE tissue samples. RNA purity (260/280 ratios) was determined and RNA integrity number (RIN) analysis was performed. RNA was subsequently used for RT-qPCR for two reference genes,18SandGAPDH.ResultsReference gene expression was equivalent between control and glioblastoma tissue when using RNA extracted from FFPE tissue, which has key implications for biological normalisation for CNS gene expression studies. There was a significant difference between the mean RIN values of control and glioblastoma FFPE tissue. There was no significant correlation between 260/280 or RIN values versus total RNA yield. The age of the tissue blocks did not influence RNA yield, fragmentation or purity. There was no significant correlation between RIN or 260/280 ratios and mean qPCR cycle threshold for either reference gene.ConclusionsThis study showed that routinely available CNS FFPE tissue is suitable for RNA extraction and downstream gene expression studies, even after 60 months of storage. Substantial RNA fragmentation associated with glioblastoma and control FFPE tissue blocks did not preclude downstream RT-qPCR gene expression analyses. Cross validation with both archival and prospectively collated FFPE specimens is required to further demonstrate that CNS tissue blocks can be used in novel translational molecular biomarker studies.
Collapse
|
15
|
Deshpande A, Lang W, McDowell T, Sivakumar S, Zhang J, Wang J, San Lucas FA, Fowler J, Kadara H, Scheet P. Strategies for identification of somatic variants using the Ion Torrent deep targeted sequencing platform. BMC Bioinformatics 2018; 19:5. [PMID: 29301485 PMCID: PMC5753459 DOI: 10.1186/s12859-017-1991-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 12/06/2017] [Indexed: 01/10/2023] Open
Abstract
Background ‘Next-generation’ (NGS) sequencing has wide application in medical genetics, including the detection of somatic variation in cancer. The Ion Torrent-based (IONT) platform is among NGS technologies employed in clinical, research and diagnostic settings. However, identifying mutations from IONT deep sequencing with high confidence has remained a challenge. We compared various computational variant-calling methods to derive a variant identification pipeline that may improve the molecular diagnostic and research utility of IONT. Results Using IONT, we surveyed variants from the 409-gene Comprehensive Cancer Panel in whole-section tumors, intra-tumoral biopsies and matched normal samples obtained from frozen tissues and blood from four early-stage non-small cell lung cancer (NSCLC) patients. We used MuTect, Varscan2, IONT’s proprietary Ion Reporter, and a simple subtraction we called “Poor Man’s Caller.” Together these produced calls at 637 loci across all samples. Visual validation of 434 called variants was performed, and performance of the methods assessed individually and in combination. Of the subset of inspected putative variant calls (n=223) in genomic regions that were not intronic or intergenic, 68 variants (30%) were deemed valid after visual inspection. Among the individual methods, the Ion Reporter method offered perhaps the most reasonable tradeoffs. Ion Reporter captured 83% of all discovered variants; 50% of its variants were visually validated. Aggregating results from multiple packages offered varied improvements in performance. Conclusions Overall, Ion Reporter offered the most attractive performance among the individual callers. This study suggests combined strategies to maximize sensitivity and positive predictive value in variant calling using IONT deep sequencing. Electronic supplementary material The online version of this article (doi:10.1186/s12859-017-1991-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aditya Deshpande
- Departments of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,University of Texas School of Public Health, Houston, TX, USA
| | - Wenhua Lang
- Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina McDowell
- Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Smruthy Sivakumar
- Departments of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jiexin Zhang
- Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - F Anthony San Lucas
- Departments of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jerry Fowler
- Departments of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Humam Kadara
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | - Paul Scheet
- Departments of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,University of Texas School of Public Health, Houston, TX, USA. .,University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
16
|
Nandakumar P, Mansouri A, Das S. The Role of ATRX in Glioma Biology. Front Oncol 2017; 7:236. [PMID: 29034211 PMCID: PMC5626857 DOI: 10.3389/fonc.2017.00236] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/14/2017] [Indexed: 02/03/2023] Open
Abstract
The current World Health Organization classification of CNS tumors has made a tremendous leap from past editions by incorporating molecular criteria in addition to the pre-existing histological parameters. The revised version has had a particular impact on the classification of diffuse low-grade gliomas and their high-grade variants. The ATRX status is one of the critical markers that define the molecular classification of gliomas. In this review, we will first provide an overview of the role of ATRX in regular cell biology. Furthermore, the role of ATRX in tumorigenesis, specifically gliomas, is comprehensively elucidated. The possible correlation of ATRX status with other genetic/epigenetic modifications is also presented. We conclude by discussing some of the challenges associated with incorporating ATRX status assessment into routine clinical practice while also exploring opportunities for future diagnostics/therapeutics in gliomas based on ATRX status.
Collapse
Affiliation(s)
- Pravanya Nandakumar
- Division of Neurosurgery, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Alireza Mansouri
- Center for Cancer Research, Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.,Division of Neuro-Oncology, Johns Hopkins University, Baltimore, MD, United States
| | - Sunit Das
- Division of Neurosurgery, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada.,The Arthur and Sonia Labatt Brain Tumour Centre, Hospital for Sick Kids, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
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]
|
18
|
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]
|
19
|
Asor E, Stav MY, Simon E, Fahoum I, Sabo E, Ben-Izhak O, Hershkovitz D. Risk for molecular contamination of tissue samples evaluated for targeted anti-cancer therapy. PLoS One 2017; 12:e0173760. [PMID: 28288198 PMCID: PMC5348008 DOI: 10.1371/journal.pone.0173760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/27/2017] [Indexed: 01/26/2023] Open
Abstract
With the increasing usage of sensitive PCR technology for pharmacogenetics, cross contamination becomes a significant concern. Researchers employed techniques which basically include replacing laboratory equipment after each sample preparation; however, there are no recommended guidelines. In the present work we wanted to evaluate the risk of cross contamination during tissue processing using the routine precaution measures. Twenty-one surgical samples of lung adenocarcinoma were used, of which 7 contained EGFR exon 19 mutation, 7 contained EGFR exon 21 mutation (p.L858R) and 7 were EGFR wild-type. The samples were ordered by alternating the mutation group to maximize the potential for cross contamination and underwent tissue sectioning and de-paraffinization. The entire process was performed using the same tools. Following DNA extraction all samples underwent PCR amplification and were scrutinized for small fractions of EGFR mutation using deep sequencing with the Ion torrent PGM technology. Twenty samples yielded results. The fraction of mutated copies was 41 ± 23% (range 11–66) for the cases with known exon 19 mutation and 48±24% (range 0–65) for the cases with known exon 21 mutations. No in-frame exon 19 deletion mutations were identified in the wild-type (WT) and exon 21 groups. The fraction of EGFR exon 21 (codon 858) mutations was 0.018±0.014% (range 0–0.05%) in the WT and exon 19 groups, which was not statistically different than the background sequencing artifact noise for the same base-pair alteration (p = 0.21). Our results suggest that standard precautions are sufficient for molecular pathology diagnosis of surgical samples and are not associated with increased risk of cross contamination.
Collapse
Affiliation(s)
- Eyal Asor
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Institute of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Michael Y. Stav
- Institute of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Einav Simon
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ibrahim Fahoum
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Edmond Sabo
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Institute of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Ofer Ben-Izhak
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Institute of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Dov Hershkovitz
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- * E-mail:
| |
Collapse
|
20
|
Hamblin A, Wordsworth S, Fermont JM, Page S, Kaur K, Camps C, Kaisaki P, Gupta A, Talbot D, Middleton M, Henderson S, Cutts A, Vavoulis DV, Housby N, Tomlinson I, Taylor JC, Schuh A. Clinical applicability and cost of a 46-gene panel for genomic analysis of solid tumours: Retrospective validation and prospective audit in the UK National Health Service. PLoS Med 2017; 14:e1002230. [PMID: 28196074 PMCID: PMC5308858 DOI: 10.1371/journal.pmed.1002230] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 01/04/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Single gene tests to predict whether cancers respond to specific targeted therapies are performed increasingly often. Advances in sequencing technology, collectively referred to as next generation sequencing (NGS), mean the entire cancer genome or parts of it can now be sequenced at speed with increased depth and sensitivity. However, translation of NGS into routine cancer care has been slow. Healthcare stakeholders are unclear about the clinical utility of NGS and are concerned it could be an expensive addition to cancer diagnostics, rather than an affordable alternative to single gene testing. METHODS AND FINDINGS We validated a 46-gene hotspot cancer panel assay allowing multiple gene testing from small diagnostic biopsies. From 1 January 2013 to 31 December 2013, solid tumour samples (including non-small-cell lung carcinoma [NSCLC], colorectal carcinoma, and melanoma) were sequenced in the context of the UK National Health Service from 351 consecutively submitted prospective cases for which treating clinicians thought the patient had potential to benefit from more extensive genetic analysis. Following histological assessment, tumour-rich regions of formalin-fixed paraffin-embedded (FFPE) sections underwent macrodissection, DNA extraction, NGS, and analysis using a pipeline centred on Torrent Suite software. With a median turnaround time of seven working days, an integrated clinical report was produced indicating the variants detected, including those with potential diagnostic, prognostic, therapeutic, or clinical trial entry implications. Accompanying phenotypic data were collected, and a detailed cost analysis of the panel compared with single gene testing was undertaken to assess affordability for routine patient care. Panel sequencing was successful for 97% (342/351) of tumour samples in the prospective cohort and showed 100% concordance with known mutations (detected using cobas assays). At least one mutation was identified in 87% (296/342) of tumours. A locally actionable mutation (i.e., available targeted treatment or clinical trial) was identified in 122/351 patients (35%). Forty patients received targeted treatment, in 22/40 (55%) cases solely due to use of the panel. Examination of published data on the potential efficacy of targeted therapies showed theoretically actionable mutations (i.e., mutations for which targeted treatment was potentially appropriate) in 66% (71/107) and 39% (41/105) of melanoma and NSCLC patients, respectively. At a cost of £339 (US$449) per patient, the panel was less expensive locally than performing more than two or three single gene tests. Study limitations include the use of FFPE samples, which do not always provide high-quality DNA, and the use of "real world" data: submission of cases for sequencing did not always follow clinical guidelines, meaning that when mutations were detected, patients were not always eligible for targeted treatments on clinical grounds. CONCLUSIONS This study demonstrates that more extensive tumour sequencing can identify mutations that could improve clinical decision-making in routine cancer care, potentially improving patient outcomes, at an affordable level for healthcare providers.
Collapse
Affiliation(s)
- Angela Hamblin
- Molecular Diagnostics Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Sarah Wordsworth
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Jilles M. Fermont
- Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Suzanne Page
- Molecular Diagnostics Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Kulvinder Kaur
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Carme Camps
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Pamela Kaisaki
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Avinash Gupta
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Denis Talbot
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Mark Middleton
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Shirley Henderson
- Molecular Diagnostics Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Anthony Cutts
- Molecular Diagnostics Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Dimitrios V. Vavoulis
- Molecular Diagnostics Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Nick Housby
- Molecular Diagnostics Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Ian Tomlinson
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Jenny C. Taylor
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Schuh
- Molecular Diagnostics Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
21
|
Basic Molecular Pathology and Cytogenetics for Practicing Pathologists: Correlation With Morphology and With a Focus on Aspects of Diagnostic or Therapeutic Utility. Adv Anat Pathol 2016; 23:368-380. [PMID: 27740961 DOI: 10.1097/pap.0000000000000124] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Morphology, as confronted in the everyday practice, often correlates with specific molecular features, which have important implications not only in pathogenesis and in diagnosis but also in prognosis and therapy. Thus, it is important that the classical pathology includes a sound knowledge of molecular aspects of disease. These molecular concepts are complex and not easily understood by all engaged in the routine practice of histopathology. Thus, the aim of this review is to present a summary of most of the necessary concepts for pathologists involving molecular pathology and genetics, beginning from basic definitions and mechanisms to major abnormalities and the methodology to detect them, correlating at the same time, the specific morphologic features associated with every abnormality.
Collapse
|
22
|
Dubbink HJ, Atmodimedjo PN, van Marion R, Krol NMG, Riegman PHJ, Kros JM, van den Bent MJ, Dinjens WNM. Diagnostic Detection of Allelic Losses and Imbalances by Next-Generation Sequencing: 1p/19q Co-Deletion Analysis of Gliomas. J Mol Diagn 2016; 18:775-786. [PMID: 27461031 DOI: 10.1016/j.jmoldx.2016.06.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 05/06/2016] [Accepted: 06/01/2016] [Indexed: 02/05/2023] Open
Abstract
Cancer cells are genomically unstable and accumulate tumor type-specific molecular aberrations, which may represent hallmarks for predicting prognosis and targets for therapy. Co-deletion of chromosomes 1p and 19q marks gliomas with an oligodendroglioma component and predicts a better prognosis and response to chemotherapy. In the current study, we present a novel method to detect chromosome 1p/19q co-deletion or loss of heterozygosity (LOH) in a diagnostic setting, based on single-nucleotide polymorphism (SNP) analysis and next-generation sequencing (NGS). We selected highly polymorphic SNPs distributed evenly over both chromosome arms. To experimentally determine the sensitivity and specificity of targeted SNP analysis, we used DNAs extracted from 49 routine formalin-fixed, paraffin-embedded glioma tissues and compared the outcome with diagnostic microsatellite-based LOH analysis and calculated estimates. We show that targeted SNP analysis by NGS allows reliable detection of 1p and/or 19q deletion in a background of 70% of normal cells according to calculated outcomes, is more sensitive than microsatellite-based LOH analysis, and requires much less DNA. This specific and sensitive SNP assay is broadly applicable for simultaneous allelic imbalance analysis of multiple genomic regions and can be incorporated easily into NGS mutation analyses. The combined mutation and chromosomal imbalance analysis in a single NGS assay is suited perfectly for routine glioma diagnostics and other diagnostic molecular pathology applications.
Collapse
Affiliation(s)
- Hendrikus J Dubbink
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Peggy N Atmodimedjo
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ronald van Marion
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Niels M G Krol
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Peter H J Riegman
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Johan M Kros
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Martin J van den Bent
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Winand N M Dinjens
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
23
|
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
|
24
|
Dong L, Wang W, Li A, Kansal R, Chen Y, Chen H, Li X. Clinical Next Generation Sequencing for Precision Medicine in Cancer. Curr Genomics 2016; 16:253-63. [PMID: 27006629 PMCID: PMC4765520 DOI: 10.2174/1389202915666150511205313] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 05/05/2015] [Accepted: 05/08/2015] [Indexed: 12/23/2022] Open
Abstract
Rapid adoption of next generation sequencing (NGS) in genomic medicine has been driven
by low cost, high throughput sequencing and rapid advances in our understanding of the genetic bases
of human diseases. Today, the NGS method has dominated sequencing space in genomic research, and
quickly entered clinical practice. Because unique features of NGS perfectly meet the clinical reality
(need to do more with less), the NGS technology is becoming a driving force to realize the dream of precision medicine.
This article describes the strengths of NGS, NGS panels used in precision medicine, current applications of NGS in cytology,
and its challenges and future directions for routine clinical use.
Collapse
Affiliation(s)
- Ling Dong
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, 650 Charles Young Dr., Los Angeles, CA 90095, USA
| | - Wanheng Wang
- Shanxi Guoxin Caregeno Medical Laboratories (MedLab), 7 Jiahua St., Taiyuan, Shanxi Province, China 030006
| | - Alvin Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, 650 Charles Young Dr., Los Angeles, CA 90095, USA
| | - Rina Kansal
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, 650 Charles Young Dr., Los Angeles, CA 90095, USA
| | - Yuhan Chen
- Heilongjiang University of Chinese Medicine, No. 24 Heping Road, Harbin, China 150040
| | - Hong Chen
- Qiqihaer First Hospital, No.30, Gongyuan Road, Qiqihar, Heilongjiang Province, China 161005
| | - Xinmin Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, 650 Charles Young Dr., Los Angeles, CA 90095, USA
| |
Collapse
|
25
|
HPV testing for primary cervical screening: Laboratory issues and evolving requirements for robust quality assurance. J Clin Virol 2016; 76 Suppl 1:S22-S28. [DOI: 10.1016/j.jcv.2015.10.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/13/2015] [Accepted: 10/31/2015] [Indexed: 11/22/2022]
|
26
|
Goswami RS, Luthra R, Singh RR, Patel KP, Routbort MJ, Aldape KD, Yao H, Dang HD, Barkoh BA, Manekia J, Medeiros LJ, Roy-Chowdhuri S, Stewart J, Broaddus RR, Chen H. Identification of Factors Affecting the Success of Next-Generation Sequencing Testing in Solid Tumors. Am J Clin Pathol 2016; 145:222-37. [PMID: 27124905 DOI: 10.1093/ajcp/aqv023] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Clinical laboratories are rapidly implementing next-generation sequencing (NGS) tests for mutation analysis, but there are few guidelines regarding sample quality for successful results. METHODS We aimed to establish tissue quality parameters for successful NGS in solid tumors and to improve NGS performance. RESULTS Analysis of 614 clinical cases tested in 2013 using a 50-gene hotspot mutation panel identified the major cause for unsuccessful NGS analysis was DNA less than 10 ng (91%, 67/74) associated with extremely small and low cellularity samples. High success rates were associated with resection procedures (333/342, 97%) and biopsied tumor larger than 10 mm(2) (77/77, 100%). NGS can be successfully performed on bone specimens processed with formic acid-based decalcification procedures (8/11, 73%). Tumor type and paraffin block age did not affect success. We demonstrated that NGS can be carried out on samples with less than 10 ng DNA. Analysis of 408 cases tested in 2014 using an optimized workflow showed improved NGS success rates from 88% to 95% (387/408) with pronounced improvement among tiny (<10 mm(2)) samples (from 76% to 94%) as well as cytology samples (from 58% to 87%). CONCLUSIONS Identifying preanalytical tissue factors allows us to improve NGS performance and to successfully test tumors obtained from minimally invasive procedures.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hui Yao
- Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Masui K, Mischel PS, Reifenberger G. Molecular classification of gliomas. HANDBOOK OF CLINICAL NEUROLOGY 2016; 134:97-120. [PMID: 26948350 DOI: 10.1016/b978-0-12-802997-8.00006-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The identification of distinct genetic and epigenetic profiles in different types of gliomas has revealed novel diagnostic, prognostic, and predictive molecular biomarkers for refinement of glioma classification and improved prediction of therapy response and outcome. Therefore, the new (2016) World Health Organization (WHO) classification of tumors of the central nervous system breaks with the traditional principle of diagnosis based on histologic criteria only and incorporates molecular markers. This will involve a multilayered approach combining histologic features and molecular information in an "integrated diagnosis". We review the current state of diagnostic molecular markers for gliomas, focusing on isocitrate dehydrogenase 1 or 2 (IDH1/IDH2) gene mutation, α-thalassemia/mental retardation syndrome X-linked (ATRX) gene mutation, 1p/19q co-deletion and telomerase reverse transcriptase (TERT) promoter mutation in adult tumors, as well as v-raf murine sarcoma viral oncogene homolog B1 (BRAF) and H3 histone family 3A (H3F3A) aberrations in pediatric gliomas. We also outline prognostic and predictive molecular markers, including O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation, and discuss the potential clinical relevance of biologic glioblastoma subtypes defined by integration of multiomics data. Commonly used methods for individual marker detection as well as novel large-scale DNA methylation profiling and next-generation sequencing approaches are discussed. Finally, we illustrate how advances in molecular diagnostics affect novel strategies of targeted therapy, thereby raising new challenges and identifying new leads for personalized treatment of glioma patients.
Collapse
Affiliation(s)
- Kenta Masui
- Department of Pathology, Tokyo Women's Medical University, Shinjku-ku, Tokyo, Japan; Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Guido Reifenberger
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
28
|
Le Tourneau C, Kamal M, Tsimberidou AM, Bedard P, Pierron G, Callens C, Rouleau E, Vincent-Salomon A, Servant N, Alt M, Rouzier R, Paoletti X, Delattre O, Bièche I. Treatment Algorithms Based on Tumor Molecular Profiling: The Essence of Precision Medicine Trials. J Natl Cancer Inst 2015; 108:djv362. [PMID: 26598514 PMCID: PMC4830395 DOI: 10.1093/jnci/djv362] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/26/2015] [Indexed: 12/13/2022] Open
Abstract
With the advent of high-throughput molecular technologies, several precision medicine (PM) studies are currently ongoing that include molecular screening programs and PM clinical trials. Molecular profiling programs establish the molecular profile of patients' tumors with the aim to guide therapy based on identified molecular alterations. The aim of prospective PM clinical trials is to assess the clinical utility of tumor molecular profiling and to determine whether treatment selection based on molecular alterations produces superior outcomes compared with unselected treatment. These trials use treatment algorithms to assign patients to specific targeted therapies based on tumor molecular alterations. These algorithms should be governed by fixed rules to ensure standardization and reproducibility. Here, we summarize key molecular, biological, and technical criteria that, in our view, should be addressed when establishing treatment algorithms based on tumor molecular profiling for PM trials.
Collapse
Affiliation(s)
- Christophe Le Tourneau
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Maud Kamal
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Apostolia-Maria Tsimberidou
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Philippe Bedard
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Gaëlle Pierron
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Céline Callens
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Etienne Rouleau
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Anne Vincent-Salomon
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Nicolas Servant
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Marie Alt
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Roman Rouzier
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Xavier Paoletti
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Olivier Delattre
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| | - Ivan Bièche
- Affiliations of authors:Department of Medical Oncology, Institut Curie , Paris & Saint-Cloud , France (CLT, MK, MA); EA7285 Versailles-St-Quentin-en-Yvelines University , France (CLT, RR); Investigational Cancer Therapeutics, M. D. Anderson Cancer Center , Houston, TX (AMT); Drug Development Program, Department of Medical Oncology and Hematology, Princess Margaret Hospital , Toronto , Canada (PB); Department of Genetics, Institut Curie , Paris , France (GP, CC, ER, IB); Department of Pathology, Institut Curie , Paris , France (AVS); Institut Curie / INSERM U900 , Paris , France (NS, XP); Department of Surgery, Institut Curie , Paris & Saint-Cloud , France (RR); Institut Curie, INSERM U830 , Paris , France (OD); EA7331, University of Paris-Descartes , Paris , France (IB)
| |
Collapse
|
29
|
Küsters-Vandevelde HVN, Küsters B, van Engen-van Grunsven ACH, Groenen PJTA, Wesseling P, Blokx WAM. Primary melanocytic tumors of the central nervous system: a review with focus on molecular aspects. Brain Pathol 2015; 25:209-26. [PMID: 25534128 DOI: 10.1111/bpa.12241] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 12/16/2014] [Indexed: 02/06/2023] Open
Abstract
Primary melanocytic tumors of the central nervous system (CNS) represent a spectrum of rare tumors. They can be benign or malignant and occur in adults as well as in children, the latter often in the context of neurocutaneous melanosis. Until recently, the genetic alterations in these tumors were largely unknown. This is in contrast with cutaneous and uveal melanomas, which are known to harbor distinct oncogenic mutations that can be used as targets for treatment with small-molecule inhibitors in the advanced setting. Recently, novel insights in the molecular alterations underlying primary melanocytic tumors of the CNS were obtained, including different oncogenic mutations in tumors in adult patients (especially GNAQ, GNA11) vs. children (especially NRAS). In this review, the focus is on molecular characteristics of primary melanocytic tumors of the CNS. We summarize what is known about their genetic alterations and discuss implications for pathogenesis and differential diagnosis with other pigmented tumors in or around the CNS. Finally, new therapeutic options with targeted therapy are discussed.
Collapse
|
30
|
Röcken C. Quality assurance in clinical trials—the role of pathology. Virchows Arch 2015; 468:83-92. [DOI: 10.1007/s00428-015-1857-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 09/23/2015] [Indexed: 02/06/2023]
|
31
|
Li T, He J, Mao X, Bi Y, Luo Z, Guo C, Tang F, Xu X, Wang X, Wang M, Chen J, Abliz Z. In situ biomarker discovery and label-free molecular histopathological diagnosis of lung cancer by ambient mass spectrometry imaging. Sci Rep 2015; 5:14089. [PMID: 26404114 PMCID: PMC4585892 DOI: 10.1038/srep14089] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 08/17/2015] [Indexed: 12/16/2022] Open
Abstract
Sensitive and spatial exploration of the metabolism of tumors at the metabolome level is highly challenging. In this study, we developed an in situ metabolomics method based on ambient mass spectrometry imaging using air flow-assisted desorption electrospray ionization (AFADESI), which can spatially explore the alteration of global metabolites in tissues with high sensitivity. Using this method, we discovered potential histopathological diagnosis biomarkers (including lipids, amino acids, choline, peptides, and carnitine) from 52 postoperative lung cancer tissue samples and then subsequently used these biomarkers to generate images for rapid and label-free histopathological diagnosis. These biomarkers were validated with a sensitivity and a specificity of 93.5% and 100%, respectively. Moreover, a single imaging analysis of a cryosection that visualized all these biomarkers, taking tens of minutes, revealed the type and subtype of the cancer. This method could potentially be used as a molecular pathological tool for rapid clinical lung cancer diagnosis and immediate image-guided surgery.
Collapse
Affiliation(s)
- Tiegang Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Jiuming He
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Xinxin Mao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P. R. China
| | - Ying Bi
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing 100084, P. R. China
| | - Zhigang Luo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Chengan Guo
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing 100084, P. R. China
| | - Fei Tang
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing 100084, P. R. China
| | - Xin Xu
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Xiaohao Wang
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing 100084, P. R. China
| | - Mingrong Wang
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Jie Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P. R. China
| | - Zeper Abliz
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| |
Collapse
|
32
|
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
|
33
|
Roemen GM, zur Hausen A, Speel EJM. Adequate tissue for adequate diagnosis: what do we really need? Lung Cancer 2015. [DOI: 10.1183/2312508x.10010014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
34
|
Appenzeller S, Gilissen C, Rijntjes J, Tops BBJ, Kastner-van Raaij A, Hebeda KM, Nissen L, Dutilh BE, van Krieken JHJM, Groenen PJTA. Immunoglobulin rearrangement analysis from multiple lesions in the same patient using next-generation sequencing. Histopathology 2015; 67:843-58. [DOI: 10.1111/his.12714] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/14/2015] [Indexed: 01/16/2023]
Affiliation(s)
- Silke Appenzeller
- Center for Molecular and Biomolecular Informatics; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Nijmegen The Netherlands
- Core Unit Systems Medicine; University of Wuerzburg; Wuerzburg Germany
| | - Christian Gilissen
- Department of Genetics; Radboud University Medical Center; Nijmegen The Netherlands
| | - Jos Rijntjes
- Department of Pathology; Radboud University Medical Center; Nijmegen The Netherlands
| | - Bastiaan B J Tops
- Department of Pathology; Radboud University Medical Center; Nijmegen The Netherlands
| | | | - Konnie M Hebeda
- Department of Pathology; Radboud University Medical Center; Nijmegen The Netherlands
| | - Loes Nissen
- Department of Gastroenterology and Hepatology; Radboud University Medical Center; Nijmegen The Netherlands
| | - Bas E Dutilh
- Center for Molecular and Biomolecular Informatics; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Nijmegen The Netherlands
| | - J Han J M van Krieken
- Department of Pathology; Radboud University Medical Center; Nijmegen The Netherlands
| | | |
Collapse
|
35
|
The homogeneous mutation status of a 22 gene panel justifies the use of serial sections of colorectal cancer tissue for external quality assessment. Virchows Arch 2015; 467:273-8. [PMID: 26047774 PMCID: PMC4552773 DOI: 10.1007/s00428-015-1789-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 05/13/2015] [Indexed: 10/27/2022]
Abstract
Testing for treatment related biomarkers in clinical care, like Ras mutation status in colorectal cancer (CRC), has increased drastically over recent years. Reliable testing of these markers is pivotal for optimal treatment of patients. Participation in external quality assessment (EQA) programs is an important element in quality management and often obligatory to comply with regulations or for accreditation. Formalin-fixed paraffin-embedded (FFPE) clinical specimens would ideally form the basis for these assessments, as they represent the most common starting material for molecular testing. However, molecular heterogeneity of a lesion in a FFPE tissue block could potentially affect test results of participating laboratories, which might compromise reliability of the quality assessment results. To assess the actual impact of this potential problem, we determined the mutation status of 22 genes commonly mutated in colon cancer in four levels covering 360 μm of 30 FFPE tissue blocks, by Next Generation Sequencing. In each block, the genotype of these genes was identical at all four levels, with only little variation in mutation load. This result shows that the mutation status of the selected 22 genes in CRC specimens is homogeneous within a 360 μm segment of the tumor. These data justify the use of serial sections, within a defined segment of a CRC tissue block, for external quality assessment of mutation analysis.
Collapse
|
36
|
Chang KC, Marton MJ. Past, current and future approaches to querying MAPK pathway activation: status and clinical implications. Per Med 2014; 11:745-760. [PMID: 29764047 DOI: 10.2217/pme.14.52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
MAPK pathway activation related to cancer development has drawn a great deal of attention in the field of personalized medicine in recent years. Many different approaches and assays have been developed to query the activation of this pathway and to develop life-saving treatments. The goal of this review article is threefold. First, to provide a brief overview of the many mutation assays that have been used to detect MAPK pathway activation, and to compare pros and cons of these assay platforms. Second, to focus on one custom-designed multiplexing mutation assay that is currently used to support an ongoing clinical trial and to show the novel features of this assay and its relevance in addressing unmet clinical needs. Third, to provide future perspectives of these MAPK pathway gene mutation detection efforts and to suggest how what we have learned from past and current approaches should guide future developments.
Collapse
Affiliation(s)
- Ken Cn Chang
- Molecular Biomarkers & Diagnostics, Merck & Co, Inc., Rahway, NJ, USA
| | - Matthew J Marton
- Molecular Biomarkers & Diagnostics, Merck & Co, Inc., Rahway, NJ, USA
| |
Collapse
|
37
|
Sie D, Snijders PJF, Meijer GA, Doeleman MW, van Moorsel MIH, van Essen HF, Eijk PP, Grünberg K, van Grieken NCT, Thunnissen E, Verheul HM, Smit EF, Ylstra B, Heideman DAM. Performance of amplicon-based next generation DNA sequencing for diagnostic gene mutation profiling in oncopathology. Cell Oncol (Dordr) 2014; 37:353-61. [PMID: 25209392 DOI: 10.1007/s13402-014-0196-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2014] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Next generation DNA sequencing (NGS) holds promise for diagnostic applications, yet implementation in routine molecular pathology practice requires performance evaluation on DNA derived from routine formalin-fixed paraffin-embedded (FFPE) tissue specimens. The current study presents a comprehensive analysis of TruSeq Amplicon Cancer Panel-based NGS using a MiSeq Personal sequencer (TSACP-MiSeq-NGS) for somatic mutation profiling. METHODS TSACP-MiSeq-NGS (testing 212 hotspot mutation amplicons of 48 genes) and a data analysis pipeline were evaluated in a retrospective learning/test set approach (n = 58/n = 45 FFPE-tumor DNA samples) against 'gold standard' high-resolution-melting (HRM)-sequencing for the genes KRAS, EGFR, BRAF and PIK3CA. Next, the performance of the validated test algorithm was assessed in an independent, prospective cohort of FFPE-tumor DNA samples (n = 75). RESULTS In the learning set, a number of minimum parameter settings was defined to decide whether a FFPE-DNA sample is qualified for TSACP-MiSeq-NGS and for calling mutations. The resulting test algorithm revealed 82% (37/45) compliance to the quality criteria and 95% (35/37) concordant assay findings for KRAS, EGFR, BRAF and PIK3CA with HRM-sequencing (kappa = 0.92; 95% CI = 0.81-1.03) in the test set. Subsequent application of the validated test algorithm to the prospective cohort yielded a success rate of 84% (63/75), and a high concordance with HRM-sequencing (95% (60/63); kappa = 0.92; 95% CI = 0.84-1.01). TSACP-MiSeq-NGS detected 77 mutations in 29 additional genes. CONCLUSION TSACP-MiSeq-NGS is suitable for diagnostic gene mutation profiling in oncopathology.
Collapse
Affiliation(s)
- Daoud Sie
- Departments of Pathology, VU University Medical Center, PO Box 7057, 1007, MB, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cree IA, Deans Z, Ligtenberg MJL, Normanno N, Edsjö A, Rouleau E, Solé F, Thunnissen E, Timens W, Schuuring E, Dequeker E, Murray S, Dietel M, Groenen P, Van Krieken JH. Guidance for laboratories performing molecular pathology for cancer patients. J Clin Pathol 2014; 67:923-31. [PMID: 25012948 PMCID: PMC4215286 DOI: 10.1136/jclinpath-2014-202404] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Molecular testing is becoming an important part of the diagnosis of any patient with cancer. The challenge to laboratories is to meet this need, using reliable methods and processes to ensure that patients receive a timely and accurate report on which their treatment will be based. The aim of this paper is to provide minimum requirements for the management of molecular pathology laboratories. This general guidance should be augmented by the specific guidance available for different tumour types and tests. Preanalytical considerations are important, and careful consideration of the way in which specimens are obtained and reach the laboratory is necessary. Sample receipt and handling follow standard operating procedures, but some alterations may be necessary if molecular testing is to be performed, for instance to control tissue fixation. DNA and RNA extraction can be standardised and should be checked for quality and quantity of output on a regular basis. The choice of analytical method(s) depends on clinical requirements, desired turnaround time, and expertise available. Internal quality control, regular internal audit of the whole testing process, laboratory accreditation, and continual participation in external quality assessment schemes are prerequisites for delivery of a reliable service. A molecular pathology report should accurately convey the information the clinician needs to treat the patient with sufficient information to allow for correct interpretation of the result. Molecular pathology is developing rapidly, and further detailed evidence-based recommendations are required for many of the topics covered here.
Collapse
Affiliation(s)
- Ian A Cree
- Warwick Medical School, University Hospital Coventry and Warwickshire, Coventry, UK
- Institute of Ophthalmology, University College London, London, UK
| | - Zandra Deans
- UK NEQAS for Molecular Genetics, Department of Laboratory Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Marjolijn J L Ligtenberg
- Department of Pathology 824, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, INT-Fondazione Pascale, Naples, Italy
| | - Anders Edsjö
- Clinical Molecular Pathology Unit, Clinical Pathology and Genetics, Sahlgrenska University Hospital and Sahlgrenska Cancer Center, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Etienne Rouleau
- Service de Génétique, Unités de Génétique constitutionnelle et somatique, Paris, France
| | - Francesc Solé
- Institut de Recerca contra la Leucèmia Josep Carreras (IJC), Barcelona, Spain
| | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth Dequeker
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, KU Leuven—University of Leuven, Leuven, Belgium
| | | | - Manfred Dietel
- Institute of Pathology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Patricia Groenen
- Department of Pathology 824, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - J Han Van Krieken
- Department of Pathology 824, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
39
|
Dubbink HJ, Deans ZC, Tops BBJ, van Kemenade FJ, Koljenović S, van Krieken HJM, Blokx WAM, Dinjens WNM, Groenen PJTA. Next generation diagnostic molecular pathology: critical appraisal of quality assurance in Europe. Mol Oncol 2014; 8:830-9. [PMID: 24704265 DOI: 10.1016/j.molonc.2014.03.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/10/2014] [Indexed: 01/08/2023] Open
Abstract
Tumor evaluation in pathology is more and more based on a combination of traditional histopathology and molecular analysis. Due to the rapid development of new cancer treatments that specifically target aberrant proteins present in tumor cells, treatment decisions are increasingly based on the molecular features of the tumor. Not only the number of patients eligible for targeted precision medicine, but also the number of molecular targets per patient and tumor type is rising. Diagnostic molecular pathology, the discipline that determines the molecular aberrations present in tumors for diagnostic, prognostic or predictive purposes, is faced with true challenges. The laboratories have to meet the need of comprehensive molecular testing using only limited amount of tumor tissue, mostly fixed in formalin and embedded in paraffin (FFPE), in short turnaround time. Choices must be made for analytical methods that provide accurate, reliable and cost-effective results. Validation of the test procedures and results is essential. In addition, participation and good performance in internal (IQA) and external quality assurance (EQA) schemes is mandatory. In this review, we critically evaluate the validation procedure for comprehensive molecular tests as well as the organization of quality assurance and assessment of competence of diagnostic molecular pathology laboratories within Europe.
Collapse
Affiliation(s)
- Hendrikus J Dubbink
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Zandra C Deans
- UK NEQAS for Molecular Genetics, Department of Laboratory Medicine, The Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Bastiaan B J Tops
- Department of Pathology, Radboud University Medical Center, PO Box 9101, Nijmegen, The Netherlands
| | - Folkert J van Kemenade
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S Koljenović
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Han J M van Krieken
- Department of Pathology, Radboud University Medical Center, PO Box 9101, Nijmegen, The Netherlands
| | - Willeke A M Blokx
- Department of Pathology, Radboud University Medical Center, PO Box 9101, Nijmegen, The Netherlands
| | - Winand N M Dinjens
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Patricia J T A Groenen
- Department of Pathology, Radboud University Medical Center, PO Box 9101, Nijmegen, The Netherlands.
| |
Collapse
|