1
|
Desilets A, Repetto M, Yang SR, Drilon A. Targeting ROS1 rearrangements in non-small cell lung cancer: Current insights and future directions. Cancer 2025; 131 Suppl 1:e35784. [PMID: 40171848 DOI: 10.1002/cncr.35784] [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: 10/23/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 04/04/2025]
Abstract
ROS1 rearrangements define a molecular subset of non-small cell lung cancer (NSCLC) by accounting for 1%-2% of cases. Targeted therapy with ROS1 tyrosine kinase inhibitors (TKIs) has significantly improved the outcomes for these patients. First-generation inhibitors, such as crizotinib and entrectinib, have demonstrated impressive efficacy, with objective response rates exceeding 60%-70%. However, the emergence of resistance mechanisms, including solvent-front mutations such as ROS1 G2032R, and limited blood-brain barrier penetration have limited the long-term efficacy of early-generation agents. Next-generation TKIs, including lorlatinib, taletrectinib, and repotrectinib, have been developed to overcome these challenges. These agents show enhanced central nervous system (CNS) penetration and activity against on-target ROS1 resistance mutations. Repotrectinib, a potent, CNS-penetrant ROS1 inhibitor, has demonstrated superior activity in both TKI-naive and -resistant tumors, including those harboring the G2032R mutation. Zidesamtinib, a highly selective next-generation ROS1 inhibitor, further addresses TRK-mediated off-target neurological toxicities seen with prior agents, and is poised to offer improved tolerability. Ongoing research is focused on optimizing sequencing strategies for ROS1 inhibitors and exploring combination approaches to prevent or overcome resistance. In addition, the development of novel diagnostic tools, including RNA-based next-generation sequencing, has enhanced the detection of functional ROS1 fusions by ensuring that patients with actionable mutations receive appropriate targeted therapies. These advances highlight the evolving landscape of treatment for ROS1-positive NSCLC, with the aim of maximizing long-term survival and quality of life.
Collapse
Affiliation(s)
- Antoine Desilets
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Matteo Repetto
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Soo-Ryum Yang
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alexander Drilon
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medicine and New York Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
2
|
Mc Leer A, Mondet J, Magnat N, Mersch M, Giovannini D, Emprou C, Toffart AC, Sturm N, Lantuéjoul S, Benito D. Rearranged During Transfection Rearrangement Detection by Fluorescence In Situ Hybridization Compared With Other Techniques in NSCLC. JTO Clin Res Rep 2024; 5:100714. [PMID: 39507413 PMCID: PMC11539407 DOI: 10.1016/j.jtocrr.2024.100714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/25/2024] [Accepted: 07/29/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction RET rearrangements occur in 1% to 2% NSCLCs. Since no clinically validated RET antibody is currently available, fluorescence in situ hybridization (FISH) is often used as a screening tool to identify patients likely to benefit from RET-targeted therapy. In this study, we performed a comprehensive review of publications in which RET-rearrangement testing was performed by FISH and compared the methods and results with our data. Methods The findings of an electronic search for publications using RET-FISH in lung cancer were compared with the results obtained at the Grenoble University Hospital where 784 EGFR -, KRAS -, ALK-, and ROS1-negative NSCLCs were tested by RET break-apart FISH and confirmed by RNA-sequencing (RNA-seq). Results Out of the 85 publications using RET-FISH analysis, 52 pertained to patients with lung cancer. The most often used positivity threshold was 15%. Six publications compared RET-FISH with at least one other molecular technique on at least eight samples, and the concordance was variable, from 5.9% to 66.7% for FISH-positive cases. Regarding our data, out of the 784 analyzed samples, 32 (4%) were positive by RET-FISH. The concordance between RET-FISH and RNA-seq in RET-FISH positive samples was 69%. Conclusions Overall, both existing literature and our data suggest that RET-FISH testing can be used for rapid screening of RET rearrangements in NSCLC. Nevertheless, using an orthogonal technique such as RNA-seq to confirm RET-FISH-positive cases is essential for ensuring that only patients likely to benefit from RET-target therapy receive the treatment.
Collapse
Affiliation(s)
- Anne Mc Leer
- Université Grenoble Alpes, Grenoble, France
- Service d’Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
- Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France
| | - Julie Mondet
- Université Grenoble Alpes, Grenoble, France
- Service d’Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
- Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France
| | - Nelly Magnat
- Service d’Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
| | - Mailys Mersch
- Université Grenoble Alpes, Grenoble, France
- Service d’Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
| | - Diane Giovannini
- Service d’Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
- TIMC-IMAG, Université Grenoble-Alpes, La Tronche, France
| | - Camille Emprou
- Service d’Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
- Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France
| | - Anne-Claire Toffart
- Université Grenoble Alpes, Grenoble, France
- Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France
- Clinique Hospitalo-Universitaire de Pneumologie Physiologie, Pôle Thorax et Vaisseaux, CHU Grenoble Alpes, Grenoble, France
| | - Nathalie Sturm
- Université Grenoble Alpes, Grenoble, France
- Service d’Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
- TIMC-IMAG, Université Grenoble-Alpes, La Tronche, France
| | - Sylvie Lantuéjoul
- Université Grenoble Alpes, Grenoble, France
- Cancer Research Center Lyon, Centre Léon Bérard, Lyon, France
| | - David Benito
- Medical Affairs - Oncology, Eli Lilly (Suisse) S.A., Dubai, United Arab Emirates
| |
Collapse
|
3
|
Mersiades AJ, Solomon BJ, Thomas DM, Lee CK, Cummins MM, Sebastian L, Ballinger ML, Collignon E, Turnbull OM, Yip S, Morton RL, Brown C, Wheeler PJ, Itchins M, Simes RJ, Pavlakis N. ASPiRATION: Australian observational cohort study of comprehensive genomic profiling in metastatic lung cancer tissue. Future Oncol 2024; 20:361-371. [PMID: 37767626 DOI: 10.2217/fon-2023-0366] [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] [Indexed: 09/29/2023] Open
Abstract
ASPiRATION is a national prospective observational cohort study assessing the feasibility, clinical and economic value of up-front tissue-based comprehensive genomic profiling (CGP) to identify actionable genomic alterations in participants with newly diagnosed metastatic non-squamous non-small-cell lung cancer in Australia. This study will enrol 1000 participants with tumor available for CGP and standard of care molecular testing (EGFR/ALK/ROS1). Participants with actionable variants may receive novel targeted treatments through ASPiRATION-specific substudies, other trials/programs. Clinical outcome data will be collected for a minimum of 2 years. Study outcomes are descriptive, including the ability of CGP to identify additional actionable variants, leading to personalized treatment recommendations, and will describe the feasibility, efficiency, cost and utility of implementation of CGP nationally.
Collapse
Affiliation(s)
- Antony J Mersiades
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
- Department of Medical Oncology, Northern Beaches Hospital, Frenchs Forest, NSW, 2086, Australia
| | - Benjamin J Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3001, Australia
| | - David M Thomas
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Randwick, NSW, 2031, Australia
| | - Chee K Lee
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
- Department of Medical Oncology, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Michelle M Cummins
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Lucille Sebastian
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Mandy L Ballinger
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Randwick, NSW, 2031, Australia
| | - Emily Collignon
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
| | - Olivia Mh Turnbull
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
| | - Sonia Yip
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Rachael L Morton
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Chris Brown
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Patrick J Wheeler
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Malinda Itchins
- Department of Medical Oncology, Royal North Shore Hospital, University of Sydney, St Leonards, NSW, 2065, Australia
| | - R John Simes
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, University of Sydney, St Leonards, NSW, 2065, Australia
| |
Collapse
|
4
|
de Oliveira Cavagna R, de Andrade ES, Tadin Reis M, de Paula FE, Noriz Berardinelli G, Bonatelli M, Ramos Teixeira G, Garbe Zaniolo B, Mourão Dias J, da Silva FAF, Baston Silva CE, Xavier Reis M, Lopes Maia E, de Alencar TS, Jacinto AA, da Nóbrega Oliveira REN, Molina-Vila MA, Ferro Leal L, Reis RM. Detection of NTRK fusions by RNA-based nCounter is a feasible diagnostic methodology in a real-world scenario for non-small cell lung cancer assessment. Sci Rep 2023; 13:21168. [PMID: 38036758 PMCID: PMC10689426 DOI: 10.1038/s41598-023-48613-4] [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: 09/03/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
NTRK1, 2, and 3 fusions are important therapeutic targets for NSCLC patients, but their prevalence in South American admixed populations needs to be better explored. NTRK fusion detection in small biopsies is a challenge, and distinct methodologies are used, such as RNA-based next-generation sequencing (NGS), immunohistochemistry, and RNA-based nCounter. This study aimed to evaluate the frequency and concordance of positive samples for NTRK fusions using a custom nCounter assay in a real-world scenario of a single institution in Brazil. Out of 147 NSCLC patients, 12 (8.2%) cases depicted pan-NTRK positivity by IHC. Due to the absence of biological material, RNA-based NGS and/or nCounter could be performed in six of the 12 IHC-positive cases (50%). We found one case exhibiting an NTRK1 fusion and another an NTRK3 gene fusion by both RNA-based NGS and nCounter techniques. Both NTRK fusions were detected in patients diagnosed with lung adenocarcinoma, with no history of tobacco consumption. Moreover, no concomitant EGFR, KRAS, and ALK gene alterations were detected in NTRK-positive patients. The concordance rate between IHC and RNA-based NGS was 33.4%, and between immunohistochemistry and nCounter was 40%. Our findings indicate that NTRK fusions in Brazilian NSCLC patients are relatively rare (1.3%), and RNA-based nCounter methodology is a suitable approach for NRTK fusion identification in small biopsies.
Collapse
Affiliation(s)
- Rodrigo de Oliveira Cavagna
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Antenor Duarte Villela, Barretos, São Paulo, 14784-400, Brazil
| | - Edilene Santos de Andrade
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Antenor Duarte Villela, Barretos, São Paulo, 14784-400, Brazil
- Molecular Diagnostic Laboratory, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | | | - Murilo Bonatelli
- Molecular Diagnostic Laboratory, Barretos Cancer Hospital, Barretos, Brazil
| | - Gustavo Ramos Teixeira
- Department of Pathology, Barretos Cancer Hospital, Barretos, Brazil
- Barretos School of Health Sciences Dr. Paulo Prata-FACISB, Barretos, Brazil
| | - Beatriz Garbe Zaniolo
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Antenor Duarte Villela, Barretos, São Paulo, 14784-400, Brazil
- Barretos School of Health Sciences Dr. Paulo Prata-FACISB, Barretos, Brazil
| | | | | | | | - Marina Xavier Reis
- Department of Medical Oncology, Barretos Cancer Hospital, Barretos, Brazil
| | - Erika Lopes Maia
- Department of Medical Oncology, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | | | - Miguel A Molina-Vila
- Laboratory of Oncology/Pangaea Oncology, Dexeus University Hospital, Barcelona, Spain
| | - Letícia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Antenor Duarte Villela, Barretos, São Paulo, 14784-400, Brazil
- Barretos School of Health Sciences Dr. Paulo Prata-FACISB, Barretos, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Antenor Duarte Villela, Barretos, São Paulo, 14784-400, Brazil.
- Molecular Diagnostic Laboratory, Barretos Cancer Hospital, Barretos, Brazil.
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
5
|
Mohebnasab M, Li P, Hong B, Dunlap J, Traer E, Fan G, Press RD, Moore SR, Xie W. Cytogenetically Cryptic Acute Promyelocytic Leukemia: A Diagnostic Challenge. Int J Mol Sci 2023; 24:13075. [PMID: 37685882 PMCID: PMC10488174 DOI: 10.3390/ijms241713075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 09/10/2023] Open
Abstract
Cytogenetically cryptic acute promyelocytic leukemia (APL) is rare, characterized by typical clinical and morphological features, but lacks t(15;17)(q24;q21)/PML::RARA translocation seen in conventional karyotyping or FISH. The prompt diagnosis and treatment of APL are critical due to life-threatening complications associated with this disease. However, cryptic APL cases remain a diagnostic challenge that could mislead the appropriate treatment. We describe four cryptic APL cases and review reported cases in the literature. Reverse transcriptase polymerase chain reaction (RT-PCR) is the most efficient diagnostic modality to detect these cases, and alternative methods are also discussed. This study highlights the importance of using parallel testing methods to diagnose cryptic APL cases accurately and effectively.
Collapse
Affiliation(s)
- Maedeh Mohebnasab
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR 97239, USA
- Division of Molecular Genomic Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Molecular and Medical Genetics and Knight Diagnostics Laboratory, Oregon Health and Science University, Portland, OR 97239, USA
| | - Peng Li
- Division of Hematopathology, Department of Pathology, University of Utah Health, Salt Lake City, UT 84112, USA
| | - Bo Hong
- Division of Hematopathology, Department of Pathology, University of Utah Health, Salt Lake City, UT 84112, USA
| | - Jennifer Dunlap
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Elie Traer
- Department of Molecular and Medical Genetics and Knight Diagnostics Laboratory, Oregon Health and Science University, Portland, OR 97239, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Guang Fan
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Richard D. Press
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Molecular and Medical Genetics and Knight Diagnostics Laboratory, Oregon Health and Science University, Portland, OR 97239, USA
| | - Stephen R. Moore
- Department of Molecular and Medical Genetics and Knight Diagnostics Laboratory, Oregon Health and Science University, Portland, OR 97239, USA
| | - Wei Xie
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
6
|
Salokas K, Dashi G, Varjosalo M. Decoding Oncofusions: Unveiling Mechanisms, Clinical Impact, and Prospects for Personalized Cancer Therapies. Cancers (Basel) 2023; 15:3678. [PMID: 37509339 PMCID: PMC10377698 DOI: 10.3390/cancers15143678] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer-associated gene fusions, also known as oncofusions, have emerged as influential drivers of oncogenesis across a diverse range of cancer types. These genetic events occur via chromosomal translocations, deletions, and inversions, leading to the fusion of previously separate genes. Due to the drastic nature of these mutations, they often result in profound alterations of cellular behavior. The identification of oncofusions has revolutionized cancer research, with advancements in sequencing technologies facilitating the discovery of novel fusion events at an accelerated pace. Oncofusions exert their effects through the manipulation of critical cellular signaling pathways that regulate processes such as proliferation, differentiation, and survival. Extensive investigations have been conducted to understand the roles of oncofusions in solid tumors, leukemias, and lymphomas. Large-scale initiatives, including the Cancer Genome Atlas, have played a pivotal role in unraveling the landscape of oncofusions by characterizing a vast number of cancer samples across different tumor types. While validating the functional relevance of oncofusions remains a challenge, even non-driver mutations can hold significance in cancer treatment. Oncofusions have demonstrated potential value in the context of immunotherapy through the production of neoantigens. Their clinical importance has been observed in both treatment and diagnostic settings, with specific fusion events serving as therapeutic targets or diagnostic markers. However, despite the progress made, there is still considerable untapped potential within the field of oncofusions. Further research and validation efforts are necessary to understand their effects on a functional basis and to exploit the new targeted treatment avenues offered by oncofusions. Through further functional and clinical studies, oncofusions will enable the advancement of precision medicine and the drive towards more effective and specific treatments for cancer patients.
Collapse
Affiliation(s)
- Kari Salokas
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00790 Helsinki, Finland
| | - Giovanna Dashi
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00790 Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00790 Helsinki, Finland
| |
Collapse
|
7
|
Damiola F, Alberti L, Mansuet-Lupo A, Damotte D, Hofman V, Tixier L, Penault-Llorca F, Rouquette I, Vignaud JM, Cazes A, Forest F, Begueret H, Gibault L, Badoual C, Cayre A, Taranchon-Clermont E, Duc A, Mc Leer A, Lantuejoul S. Usefulness of an RNA extraction-free test for the multiplexed detection of ALK, ROS1, and RET Gene Fusions in Real Life FFPE Specimens of Non-Small Cell Lung Cancers. Expert Rev Mol Diagn 2023; 23:1283-1291. [PMID: 37906110 DOI: 10.1080/14737159.2023.2277367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND ALK, ROS1 and RET rearrangements occur, respectively, in 5%, 2%, and 1% non-small cell lung cancers (NSCLC). ALK and ROS1 fusion proteins detection by immunohistochemistry (IHC) has been validated for rapid patient screening, but ROS1 fusions need to be confirmed by another technique and no RET IHC test is available for clinical use. RESEARCH DESIGN AND METHODS We report herein the usefulness of the HTG EdgeSeq Assay, an RNA extraction-free test combining a quantitative nuclease protection assay with NGS, for the detection of ALK, ROS1 and RET fusions from 'real-life' small NSCLC samples. A total of 203 FFPE samples were collected from 11 centers. They included 143 rearranged NSCLC (87 ALK, 39 ROS1, 17 RET) and 60 ALK-ROS1-RET negative controls. RESULTS The assay had a specificity of 98% and a sensitivity for ALK, ROS1 and RET fusions of 80%, 94% and 100% respectively. Among the 19 HTG-assay false negative samples, the preanalytical conditions were identified as the major factors impacting the assay efficiency. CONCLUSIONS Overall, the HTG EdgeSeq assay offers comparable sensitivities and specificity than other RNA sequencing techniques, with the advantage that it can be used on very small and old samples collected multicentrically.
Collapse
Affiliation(s)
- Francesca Damiola
- Department of Biopathology, CLCC UNICANCER Léon Bérard, Lyon, France
- Anatomopathology Research Platform and Team Genetics, Epigenetics and Biology of Sarcomas, INSERM 1052, CNRS 5286 of Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Laurent Alberti
- Department of Biopathology, CLCC UNICANCER Léon Bérard, Lyon, France
- Integrated analysis of the dynamics of cancer team, Cancer Research Center of Lyon (CRCL), UMR Inserm 1052, CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Audrey Mansuet-Lupo
- Department of Pathology, Groupe Hospitalier Universitaire Paris Centre, Cochin Hospital, APHP.centre, Université Paris Cité, Paris, France
| | - Diane Damotte
- Department of Pathology, Groupe Hospitalier Universitaire Paris Centre, Cochin Hospital, APHP.centre, Université Paris Cité, Paris, France
| | - Véronique Hofman
- Department of Biopathology, Nice University Hospital, Nice, France
| | - Lucie Tixier
- UF of Pathology, Centre Jean Perrin UNICANCER, INSERM UMR1240 IMoST, University Clermont Auvergne, Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- UF of Pathology, Centre Jean Perrin UNICANCER, INSERM UMR1240 IMoST, University Clermont Auvergne, Clermont-Ferrand, France
| | - Isabelle Rouquette
- Department of Biopathology, Toulouse University Hospital, Toulouse, France
| | | | - Aurélie Cazes
- Department of Biopathology, Bichat Hospital, Paris, France
| | - Fabien Forest
- Saint Etienne Department of Pathology University Hospital, Saint Etienne, France
| | - Hugues Begueret
- Department of Biopathology, Bordeaux University Hospital, Bordeaux, France
| | - Laure Gibault
- Service d'Anatomie et Cytologie Pathologiques, Hôpital Européen Georges Pompidou, APHP, Paris Cité University, Paris, France
| | - Cécile Badoual
- Service d'Anatomie et Cytologie Pathologiques, Hôpital Européen Georges Pompidou, APHP, Paris Cité University, Paris, France
| | - Anne Cayre
- UF of Pathology, Centre Jean Perrin UNICANCER, INSERM UMR1240 IMoST, University Clermont Auvergne, Clermont-Ferrand, France
| | | | - Adeline Duc
- Department of Biopathology, CLCC UNICANCER Léon Bérard, Lyon, France
- Anatomopathology Research Platform and Team Genetics, Epigenetics and Biology of Sarcomas, INSERM 1052, CNRS 5286 of Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Anne Mc Leer
- CHU Grenoble Alpes Pathology Department, Institute for Advanced Biosciences UGA/INSERM U1209/CNRS 5309, Grenoble Alpes University, Grenoble, France
- Université Grenoble Alpes, Grenoble, France
| | - Sylvie Lantuejoul
- Department of Biopathology, CLCC UNICANCER Léon Bérard, Lyon, France
- Anatomopathology Research Platform and Team Genetics, Epigenetics and Biology of Sarcomas, INSERM 1052, CNRS 5286 of Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
8
|
Chu YH. This is Your Thyroid on Drugs: Targetable Mutations and Fusions in Thyroid Carcinoma. Surg Pathol Clin 2023; 16:57-73. [PMID: 36739167 DOI: 10.1016/j.path.2022.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review aims to provide an overview of the molecular pathogenesis thyroid carcinomas, emphasizing genetic alterations that are therapeutically actionable. The main pathways in thyroid carcinogenesis are the MAPK and PI3K pathways. Point mutations and gene rearrangements affecting the pathway effectors and receptor tyrosine kinases are well-known drivers of thyroid cancer. Research over the past few decades has successfully introduced highly effective treatments for unresectable thyroid cancer, evolving from multi-kinase inhibitors to structurally selective agents, with constantly improving toxicity profiles and coverage of resistance mechanisms. The pros and cons of major laboratory techniques for therapeutic target identification are discussed.
Collapse
Affiliation(s)
- Ying-Hsia Chu
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan.
| |
Collapse
|
9
|
Chu YH, Barbee J, Yang SR, Chang JC, Liang P, Mullaney K, Chan R, Salazar P, Benayed R, Offin M, Drilon A, Ladanyi M, Nafa K, Arcila ME. Clinical Utility and Performance of an Ultrarapid Multiplex RNA-Based Assay for Detection of ALK, ROS1, RET, and NTRK1/2/3 Rearrangements and MET Exon 14 Skipping Alterations. J Mol Diagn 2022; 24:642-654. [PMID: 35430374 DOI: 10.1016/j.jmoldx.2022.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/14/2022] [Accepted: 03/03/2022] [Indexed: 11/19/2022] Open
Abstract
Several kinase fusions are established targetable drivers in lung cancers. However, rapid and comprehensive detection remains challenging because of diverse partner genes and breakpoints. We assess the clinical utility and performance of a rapid microfluidic multiplex real-time PCR-based assay for simultaneous query of fusions involving ALK, ROS1, RET, and NTRK1/2/3, as well as MET exon 14 skipping, using a 3-hour automated process. Dual analytic strategies were utilized: fusion-specific amplification and 3' to 5' expression imbalance. One-hundred and forty-three independent, formalin-fixed, paraffin-embedded tumor samples (112 surgical specimens, 31 cytologic cell blocks) were analyzed: 133 with known kinase gene alterations and 10 negative samples based on clinically validated next-generation sequencing. Testing was successful in 142 (99%) cases. The assay demonstrated a sensitivity of 97% (28/29), 100% (31/31), 92% (22/24), 81% (22/27), and 100% (20/20) for ALK, RET, ROS1, and NTRK1/2/3 rearrangements and MET exon 14 skipping alterations, respectively, with 100% specificity for all. Concordant results were achieved in specimens aged up to 5 years, with >10% tumor, and inputs of at least 9 mm2 (surgical specimens) and 9000 cells (cytologic cell blocks). The assay enables rapid screening for clinically actionable kinase alterations with quicker turnaround and lower tissue requirements compared with immunohistochemistry and molecular methods, while also circumventing the infrastructure dependencies associated with next-generation sequencing and fluorescence in situ hybridization.
Collapse
Affiliation(s)
- Ying-Hsia Chu
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jada Barbee
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Soo-Ryum Yang
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason C Chang
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Priscilla Liang
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kerry Mullaney
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Roger Chan
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paulo Salazar
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryma Benayed
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Offin
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander Drilon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Khedoudja Nafa
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria E Arcila
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
10
|
Haley L, Parimi V, Jiang L, Pallavajjala A, Hardy M, Yonescu R, Morsberger L, Stinnett V, Long P, Zou YS, Gocke CD. Diagnostic Utility of Gene Fusion Panel to Detect Gene Fusions in Fresh and Formalin-Fixed, Paraffin-Embedded Cancer Specimens. J Mol Diagn 2021; 23:1343-1358. [PMID: 34358677 DOI: 10.1016/j.jmoldx.2021.07.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/04/2021] [Accepted: 07/08/2021] [Indexed: 11/19/2022] Open
Abstract
Somatic gene fusions are common in leukemias/lymphomas and solid tumors. The detection of gene fusions is crucial for diagnosis. NanoString fusion technology is a multiplexed hybridization method that interrogates hundreds of gene fusions in a single reaction. This study's objective was to determine the performance characteristics and diagnostic utility of NanoString fusion assay in a clinical diagnostics laboratory. Validation using 100 positive specimens and 15 negative specimens by a combined reference standard of fluorescence in situ hybridization (FISH)/RT-PCR/next-generation sequencing (NGS) assays achieved 100% sensitivity in leukemias/lymphomas and 95.0% sensitivity and 100% specificity in solid tumors. Subsequently, 214 consecutive clinical cases, including 73 leukemia/lymphoma specimens and 141 formalin-fixed, paraffin-embedded solid tumor specimens, were analyzed by gene fusion panels across 638 unique gene fusion transcripts. A variety of comparator tests, including FISH panels, conventional karyotyping, a DNA-based targeted NGS assay, and custom RT-PCR testing, were performed in parallel. The gene fusion assay detected 31 gene fusions, including 16 in leukemia/lymphoma specimens and 15 in solid tumor specimens. The overall sensitivity, specificity, and accuracy of gene fusions detected by the gene fusion panel in all 329 specimens (validation and consecutive clinical specimens) tested in this study were 94.8%, 100%, and 97.9%, respectively, compared with FISH/RT-PCR/NGS assays. The gene fusion panel is a reliable approach that maximizes molecular detection of fusions among both fresh and formalin-fixed, paraffin-embedded cancer specimens.
Collapse
Affiliation(s)
- Lisa Haley
- Johns Hopkins Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vamsi Parimi
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Liqun Jiang
- Johns Hopkins Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aparna Pallavajjala
- Johns Hopkins Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Melanie Hardy
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Cytogenetics Laboratory, Johns Hopkins University Hospital, Baltimore, Maryland
| | - Raluca Yonescu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Cytogenetics Laboratory, Johns Hopkins University Hospital, Baltimore, Maryland
| | - Laura Morsberger
- Johns Hopkins Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Cytogenetics Laboratory, Johns Hopkins University Hospital, Baltimore, Maryland
| | - Victoria Stinnett
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Cytogenetics Laboratory, Johns Hopkins University Hospital, Baltimore, Maryland
| | - Patty Long
- Johns Hopkins Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Cytogenetics Laboratory, Johns Hopkins University Hospital, Baltimore, Maryland
| | - Ying S Zou
- Johns Hopkins Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Cytogenetics Laboratory, Johns Hopkins University Hospital, Baltimore, Maryland.
| | - Christopher D Gocke
- Johns Hopkins Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
11
|
Pisapia P, Pepe F, Sgariglia R, Nacchio M, Russo G, Gragnano G, Conticelli F, Salatiello M, De Luca C, Girolami I, Eccher A, Iaccarino A, Bellevicine C, Vigliar E, Malapelle U, Troncone G. Methods for actionable gene fusion detection in lung cancer: now and in the future. Pharmacogenomics 2021; 22:833-847. [PMID: 34525844 DOI: 10.2217/pgs-2021-0048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/26/2021] [Indexed: 12/28/2022] Open
Abstract
Although gene fusions occur rarely in non-small-cell lung cancer (NSCLC) patients, they represent a relevant target in treatment decision algorithms. To date, immunohistochemistry and fluorescence in situ hybridization are the two principal methods used in clinical trials. However, using these methods in routine clinical practice is often impractical and time consuming because they can only analyze single genes and the quantity of tissue material is often insufficient. Thus, novel technologies, able to test multiple genes in a single run with minimal sample input, are being under investigation. Here, we discuss the utility of next-generation sequencing and nCounter technologies in detecting simultaneous gene fusions in NSCLC patients.
Collapse
Affiliation(s)
- Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Roberta Sgariglia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Mariantonia Nacchio
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Gragnano
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Floriana Conticelli
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Maria Salatiello
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Caterina De Luca
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Ilaria Girolami
- Division of Pathology, Central Hospital Bolzano, Bolzano, Italy
| | - Albino Eccher
- Department of Pathology & Diagnostics, University & Hospital Trust of Verona, Verona, Italy
| | - Antonino Iaccarino
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Claudio Bellevicine
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Elena Vigliar
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
12
|
Garrido P, Hladun R, de Álava E, Álvarez R, Bautista F, López-Ríos F, Colomer R, Rojo F. Multidisciplinary consensus on optimising the detection of NTRK gene alterations in tumours. Clin Transl Oncol 2021; 23:1529-1541. [PMID: 33620682 PMCID: PMC8238709 DOI: 10.1007/s12094-021-02558-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/24/2021] [Indexed: 12/21/2022]
Abstract
The recent identification of rearrangements of neurotrophic tyrosine receptor kinase (NTRK) genes and the development of specific fusion protein inhibitors, such as larotrectinib and entrectinib, have revolutionised the diagnostic and clinical management of patients presenting with tumours with these alterations. Tumours that harbour NTRK fusions are found in both adults and children; and they are either rare tumours with common NTRK fusions that may be diagnostic, or more prevalent tumours with rare NTRK fusions. To assess currently available evidence on this matter, three key Spanish medical societies (the Spanish Society of Medical Oncology (SEOM), the Spanish Society of Pathological Anatomy (SEAP), and the Spanish Society of Paediatric Haematology and Oncology (SEHOP) have brought together a group of experts to develop a consensus document that includes guidelines on the diagnostic, clinical, and therapeutic aspects of NTRK-fusion tumours. This document also discusses the challenges related to the routine detection of these genetic alterations in a mostly public Health Care System.
Collapse
Affiliation(s)
- P. Garrido
- Sociedad Española de Oncología Médica (SEOM), Departamento de Oncología Médica, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, IRYCIS, CIBERONC, Madrid, Spain
| | - R. Hladun
- Sociedad Española de Hematología y Oncologías Pediátricas (SEHOP), Departamento de Oncología, Hematología y Trasplante de Progenitores Hematopoyéticos Pediátricos, Hospital Universitario Vall d’Hebron, Barcelona, Spain
| | - E. de Álava
- Sociedad Española de Anatomía Patológica (SEAP), Departamento de Citología e Histología Normal y Patológica, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), CSIC, Facultad de Medicina, Universidad de Sevilla, CIBERONC, Sevilla, Spain
| | - R. Álvarez
- Sociedad Española de Oncología Médica (SEOM), Departamento de Oncología Médica, Hospital Universitario Gregorio Marañón. Instituto Investigación Sanitaria Gregorio Marañon (IISGM), Madrid, Spain
| | - F. Bautista
- Sociedad Española de Hematología y Oncologías Pediátricas (SEHOP), Oncología Pediátrica, Departamento de Hematología y Trasplante de Células Madre Hematopoyéticas, Hospital Universitario Infantil Niño Jesús, Madrid, Spain
| | - F. López-Ríos
- Sociedad Española de Anatomía Patológica (SEAP), Departamento de Patología, Laboratorio de Dianas Terapéuticas, Hospital Universitario HM Sanchinarro, CIBERONC, Madrid, Spain
| | - R. Colomer
- Sociedad Española de Oncología Médica (SEOM), Departamento de Oncología Médica, Hospital Universitario La Princesa, Universidad Autónoma de Madrid, Cátedra UAM-Fundación Instituto Roche de Medicina Personalizada de Precisión, Madrid, Spain
| | - F. Rojo
- Sociedad Española de Anatomía Patológica (SEAP), Departamento de Patología, IIS-Fundación Universitaria Jiménez Díaz, CIBERONC, Madrid, Spain
| |
Collapse
|
13
|
Ji X, Liu Y, Mei F, Li X, Zhang M, Yao B, Wu R, You J, Pei F. SPP1 overexpression is associated with poor outcomes in ALK fusion lung cancer patients without receiving targeted therapy. Sci Rep 2021; 11:14031. [PMID: 34234236 PMCID: PMC8263595 DOI: 10.1038/s41598-021-93484-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/17/2021] [Indexed: 12/22/2022] Open
Abstract
The screening of non-small cell lung cancer (NSCLC) tumors for anaplastic lymphoma receptor tyrosine kinase (ALK) gene rearrangements is important because of the dramatically favorable therapy response to ALK inhibitor. However, the exact mechanism of poor survival in ALK fusion lung cancer patients without receiving targeted therapy is unclear. In this study, total of 521 tumor specimens from Chinese patients with lung cancer were screened for ALK fusion by immunohistochemistry (IHC) and confirmed by fluorescence in situ hybridization (FISH). As results, there were no cases of coexisting EGFR and ALK mutations identified. Fourteen cases (2.7%) harbored ALK fusion, including eight solid adenocarcinomas with signet ring cell features, four acinar adenocarcinomas with cribriform pattern containing mucin, one adenosquamous carcinoma and one micropapillary adenocarcinoma with mucin. Six (42.9%) of fourteen patients with ALK-positive lung cancer had stage IV disease, and five ALK-positive patients treated with platinum-based chemotherapy had poor outcome (all patients were dead and the mean survival time was 12 months), compared to 72 months for patients with ALK inhibitor therapy. Furthermore, Five ALK-positive cases were analyzed by whole exome sequencing (WES) and via direct transcript counting using a digital probe-base (NanoString) to explore the driver genes. Deregulation of PI3K/AKT signaling pathway in ALK-positive lung cancer was demonstrated by WES analysis, and significantly increased mRNA of ALK, ROS1, MET, SPP1 and PI3K signaling pathway was identified by NanoString assay. The concordance between NanoString, IHC and FISH methodologies for detecting ALK fusion was 100%. Significant overexpression of SPP1 protein in ALK-positive lung cancer was confirmed by IHC compared to paired adjacent normal tissues and ALK-negative cancers. Thus we concluded that SPP1 overexpression is associated with poor outcomes for patients with ALK fusion lung cancer without receiving targeted therapy and PI3K/AKT/SPP1 pathway may become the promising targets in patients with aggressive lung cancer.
Collapse
Affiliation(s)
- Xiaolin Ji
- Department of Pathology, Peking University Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Yan Liu
- Department of Pathology, Peking University Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Fang Mei
- Department of Pathology, Peking University Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Xinyang Li
- Department of Pathology, Peking University Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Mengxue Zhang
- Department of Pathology, Peking University Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Buwen Yao
- Department of Pathology, Peking University Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Rui Wu
- Department of Pathology, Peking University Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Jiangfeng You
- Department of Pathology, Peking University Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Fei Pei
- Department of Pathology, Peking University Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Haidian District, Beijing, 100191, People's Republic of China.
| |
Collapse
|
14
|
Uguen A. Each RET Break-Apart Fluorescence In Situ Hybridization Probe Requires Proper Interpretation Criteria. J Thorac Oncol 2021; 16:e55. [PMID: 34154792 DOI: 10.1016/j.jtho.2021.03.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Arnaud Uguen
- Lymphocytes B and Auto-Immunity, UMR 1227, CHRU Brest, INSERM, Brest University, Brest, France.
| |
Collapse
|
15
|
Zito Marino F, Alì G, Facchinetti F, Righi L, Fontanini G, Rossi G, Franco R. Fusion proteins in lung cancer: addressing diagnostic problems for deciding therapy. Expert Rev Anticancer Ther 2021; 21:887-900. [PMID: 33715580 DOI: 10.1080/14737140.2021.1903875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Gene fusions are frequent chromosomal aberrations in solid tumors. In Lung cancer (LC) several druggable-fusions involving tyrosine kinase receptor genes have been described, including ALK, ROS1, RET and NTRK. In non-small cell lung cancer, testing for targetable fusions has become a part of routine clinical practice, greatly impacting therapeutic choice for patients with these aberrations. Although substantial technologies for gene fusion detection have been implemented over time including; cytogenetic, Fluorescence in situ hybridization (FISH), Immunohistochemistry (IHC), Retro-transcription Real-Time PCR (RT-qPCR), to Next Generation Sequencing (NGS), nCounter system (Nanostring technology), several critical issues remain. To date, only the companion diagnostic tests FISH and IHC for ALK-rearrangements and NGS for ROS1-rearrangments were approved. Other fusion approved tests are currently unavailable.Areas covered: In this review, we explore current diagnostic problems of gene fusion detection relative to the technologies available, in order to clarify future standardization of analyses which determine therapeutic choices.Expert opinion: The establishment of a gold standard, an effective diagnostic algorithm, and a standardized interpretation for the analysis of each druggable-fusions in lung cancer is essential for adequate therapeutic management.
Collapse
Affiliation(s)
- Federica Zito Marino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Greta Alì
- Department of Surgical Pathology, Medical, Molecular, and Critical Area, University of Pisa, Pisa, Italy
| | - Francesco Facchinetti
- Université Paris-Saclay, Institut Gustave Roussy, INSERM, Biomarqueurs prédictifs et nouvelles stratégies thérapeutiques en oncologie, Villejuif, France.,Medical Oncology Unit, University Hospital of Parma, Italy
| | - Luisella Righi
- Department of Oncology, University of Turin, Pathology Division, San Luigi Hospital, University of Turin, Turin, Italy
| | - Gabriella Fontanini
- Department of Surgical Pathology, Medical, Molecular, and Critical Area, University of Pisa, Pisa, Italy
| | - Giulio Rossi
- Operative Unit of Pathologic Anatomy, Azienda Della Romagna, Teaching Hospital S. Maria Delle Croci, Ravenna, Italy
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| |
Collapse
|
16
|
Belli C, Penault-Llorca F, Ladanyi M, Normanno N, Scoazec JY, Lacroix L, Reis-Filho JS, Subbiah V, Gainor JF, Endris V, Repetto M, Drilon A, Scarpa A, André F, Douillard JY, Curigliano G. ESMO recommendations on the standard methods to detect RET fusions and mutations in daily practice and clinical research. Ann Oncol 2021; 32:337-350. [PMID: 33455880 DOI: 10.1016/j.annonc.2020.11.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 12/17/2022] Open
Abstract
Aberrant activation of RET is a critical driver of growth and proliferation in diverse solid tumours. Multikinase inhibitors (MKIs) showing anti-RET activities have been tested in RET-altered tumours with variable results. The low target specificity with consequent increase in side-effects and off-target toxicities resulting in dose reduction and drug discontinuation are some of the major issues with MKIs. To overcome these issues, new selective RET inhibitors such as pralsetinib (BLU-667) and selpercatinib (LOXO-292) have been developed in clinical trials, with selpercatinib recently approved by the Food and Drug Administration (FDA). The results of these trials showed marked and durable antitumour activity and manageable toxicity profiles in patients with RET-altered tumours. The European Society for Medical Oncology (ESMO) Translational Research and Precision Medicine Working Group (TR and PM WG) launched a collaborative project to review the available methods for the detection of RET gene alterations, their potential applications and strategies for the implementation of a rational approach for the detection of RET fusion genes and mutations in human malignancies. We present here recommendations for the routine clinical detection of targetable RET rearrangements and mutations.
Collapse
Affiliation(s)
- C Belli
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - F Penault-Llorca
- University Clermont Auvergne, INSERM U1240, Centre Jean Perrin, Department of BioPathology, Clermont-Ferrand, France
| | - M Ladanyi
- Department of Pathology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - N Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - J-Y Scoazec
- AMMICa, CNRS-UMS 3655 and INSERM-US23, Gustave Roussy, Villejuif, France; Department of Pathology and Translational Research, Gustave Roussy Cancer Centre, Villejuif, France
| | - L Lacroix
- Translational Research Laboratory and Biobank, Gustave Roussy, Villejuif, France; Inserm U981, Gustave Roussy, Villejuif, France; Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - J S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - V Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J F Gainor
- Massachusetts General Hospital, Boston, USA
| | - V Endris
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - M Repetto
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - A Drilon
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - A Scarpa
- ARC-Net Research Centre and Department of Diagnostics and Public Health - Section of Pathology, University of Verona, Verona, Italy
| | - F André
- Gustave Roussy Cancer Center, Villejuif, France
| | - J-Y Douillard
- Scientific and Medical Division, European Society for Medical Oncology, Lugano, Switzerland
| | - G Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
17
|
Novaes LAC, Sussuchi da Silva L, De Marchi P, Cavagna RDO, de Paula FE, Zanon MF, Evangelista AF, Albino da Silva EC, Duval da Silva V, Leal LF, Reis RM. Simultaneous analysis of ALK, RET, and ROS1 gene fusions by NanoString in Brazilian lung adenocarcinoma patients. Transl Lung Cancer Res 2021; 10:292-303. [PMID: 33569313 PMCID: PMC7867767 DOI: 10.21037/tlcr-20-740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Gene fusions have been successfully employed as therapeutic targets for lung adenocarcinoma. However, tissue availability for molecular testing of multiples alterations is frequently unfeasible. We aimed to detect the presence of ALK, RET, and ROS1 rearrangements by a RNA-based single assay in Brazilian lung adenocarcinomas and to associate with clinicopathological features and genetic ancestry. Methods From a FFPE series of 444 molecularly characterized lung adenocarcinomas, 253 EGFR/KRAS wild-type cases were eligible for gene rearrangement analysis. Following RNA isolation, ALK, RET, and ROS1 rearrangements were simultaneously analyzed employing the ElementsXT Custom panel (NanoString Technologies). Rearrangements were further associated with clinicopathological features and genetic ancestry of the patients. Results The NanoString platform was performed in subset of 142 cases. Gene fusion results were conclusive for 94.4% (n=134) cases (failure rate =5.6%). ALK rearrangements were observed in 21 out of 134 cases, and associated with younger, never smokers, metastatic disease, and metastases in the central nervous system. RET and ROS1 fusions were detected in two and one out of 134 cases, respectively. Genetic ancestry was not associated with gene fusions. Overall, considering all cases for which a molecular analysis was conclusive (EGFR/KRAS/ALK/RET/ROS1), ALK fusions frequency was observed in 6.5% (21/325), RET in 0.6% (2/325), and ROS1 in 0.3% (1/325). Conclusions This study successfully used a RNA-based single assay for the simultaneous analysis of ALK, RET, and ROS1 fusions employing routine biopsies from Brazilian patients lung adenocarcinoma allowing an extensive molecular testing for actionable rearrangements contributing to guide clinical strategies.
Collapse
Affiliation(s)
| | | | - Pedro De Marchi
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Department of Medical Oncology, Barretos Cancer Hospital, Barretos, Brazil.,Oncoclinicas Group, Rio de Janeiro, Brazil
| | - Rodrigo de Oliveira Cavagna
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Department of Molecular Diagnosis, Barretos Cancer Hospital, Barretos, Brazil
| | | | - Maicon Fernando Zanon
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Department of Molecular Diagnosis, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | - Vinícius Duval da Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Department of Pathology, Barretos Cancer Hospital, Barretos, Brazil.,Barretos School of Medicine Dr. Paulo Prata - FACISB, Barretos, Brazil
| | - Letícia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos School of Medicine Dr. Paulo Prata - FACISB, Barretos, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Department of Molecular Diagnosis, Barretos Cancer Hospital, Barretos, Brazil.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
18
|
D’Angelo A, Sobhani N, Chapman R, Bagby S, Bortoletti C, Traversini M, Ferrari K, Voltolini L, Darlow J, Roviello G. Focus on ROS1-Positive Non-Small Cell Lung Cancer (NSCLC): Crizotinib, Resistance Mechanisms and the Newer Generation of Targeted Therapies. Cancers (Basel) 2020; 12:3293. [PMID: 33172113 PMCID: PMC7694780 DOI: 10.3390/cancers12113293] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 12/18/2022] Open
Abstract
The treatment of patients affected by non-small cell lung cancer (NSCLC) has been revolutionised by the discovery of druggable mutations. ROS1 (c-ros oncogene) is one gene with druggable mutations in NSCLC. ROS1 is currently targeted by several specific tyrosine kinase inhibitors (TKIs), but only two of these, crizotinib and entrectinib, have received Food and Drug Administration (FDA) approval. Crizotinib is a low molecular weight, orally available TKI that inhibits ROS1, MET and ALK and is considered the gold standard first-line treatment with demonstrated significant activity for lung cancers harbouring ROS1 gene rearrangements. However, crizotinib resistance often occurs, making the treatment of ROS1-positive lung cancers more challenging. A great effort has been undertaken to identify a new generation or ROS1 inhibitors. In this review, we briefly introduce the biology and role of ROS1 in lung cancer and discuss the underlying acquired mechanisms of resistance to crizotinib and the promising new agents able to overcome resistance mechanisms and offer alternative efficient therapies.
Collapse
Affiliation(s)
- Alberto D’Angelo
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK; (S.B.); (J.D.)
| | - Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Robert Chapman
- University College London Hospitals NHS Foundation Trust, 235 Euston Rd, London NW1 2BU, UK;
| | - Stefan Bagby
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK; (S.B.); (J.D.)
| | - Carlotta Bortoletti
- Department of Dermatology, University of Padova, via Vincenzo Gallucci 4, 35121 Padova, Italy;
| | - Mirko Traversini
- Unità Operativa Anatomia Patologica, Ospedale Maggiore Carlo Alberto Pizzardi, AUSL Bologna, Largo Bartolo Nigrisoli 2, 40100 Bologna, Italy;
| | - Katia Ferrari
- Respiratory Medicine, Careggi University Hospital, 50139 Florence, Italy;
| | - Luca Voltolini
- Thoracic Surgery Unit, Careggi University Hospital, Largo Brambilla, 1, 50134 Florence, Italy;
| | - Jacob Darlow
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK; (S.B.); (J.D.)
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy;
| |
Collapse
|
19
|
Macerola E, Poma AM, Basolo F. NanoString in the screening of genetic abnormalities associated with thyroid cancer. Semin Cancer Biol 2020; 79:132-140. [PMID: 33091600 DOI: 10.1016/j.semcancer.2020.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 05/08/2020] [Accepted: 10/12/2020] [Indexed: 11/24/2022]
Abstract
In the setting of cancer pathology, molecular characterization of tumors providing diagnostic and predictive information is acquiring more and more relevance. Moreover, the advent of innovative technologies continuously improves the knowledge of the molecular landscape of tumors and strengthens the links between clinics, tumor pathology and molecular features. In the clinical management of patients with thyroid nodules and thyroid tumors, the aid of molecular testing is encouraged but still not strongly recommended by current guidelines. Also for this reason this field of study is attracting much interest. The nCounter system is a relatively new technology based on a direct hybridization of fluorescent probes to specific nucleic acid targets, followed by digital measurement of signals; the reaction is highly multiplexable and results are robust and reproducible. This review reports and discusses the available data related to the application of this specific technique to thyroid nodules and thyroid tumors samples. The available data indicate that nCounter system represents a solid approach for the research of relevant diagnostic and prognostic biomarkers in thyroid pathology.
Collapse
Affiliation(s)
- Elisabetta Macerola
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, via Savi, 10, 56126, Pisa, Italy.
| | - Anello Marcello Poma
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, via Savi, 10, 56126, Pisa, Italy.
| | - Fulvio Basolo
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, via Savi, 10, 56126, Pisa, Italy.
| |
Collapse
|
20
|
Comprehensive analysis of ALK, ROS1 and RET rearrangements in locally advanced rectal cancer. J Genet 2020. [DOI: 10.1007/s12041-020-01239-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
21
|
Bruno R, Fontanini G. Next Generation Sequencing for Gene Fusion Analysis in Lung Cancer: A Literature Review. Diagnostics (Basel) 2020; 10:E521. [PMID: 32726941 PMCID: PMC7460167 DOI: 10.3390/diagnostics10080521] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 02/07/2023] Open
Abstract
Gene fusions have a pivotal role in non-small cell lung cancer (NSCLC) precision medicine. Several techniques can be used, from fluorescence in situ hybridization and immunohistochemistry to next generation sequencing (NGS). Although several NGS panels are available, gene fusion testing presents more technical challenges than other variants. This is a PubMed-based narrative review aiming to summarize NGS approaches for gene fusion analysis and their performance on NSCLC clinical samples. The analysis can be performed at DNA or RNA levels, using different target enrichment (hybrid-capture or amplicon-based) and sequencing chemistries, with both custom and commercially available panels. DNA sequencing evaluates different alteration types simultaneously, but large introns and repetitive sequences can impact on the performance and it does not discriminate between expressed and unexpressed gene fusions. RNA-based targeted approach analyses and quantifies directly fusion transcripts and is more accurate than DNA panels on tumor tissue, but it can be limited by RNA quality and quantity. On liquid biopsy, satisfying data have been published on circulating tumor DNA hybrid-capture panels. There is not a perfect method for gene fusion analysis, but NGS approaches, though still needing a complete standardization and optimization, present several advantages for the clinical practice.
Collapse
Affiliation(s)
- Rossella Bruno
- Unit of Pathological Anatomy, University Hospital of Pisa, Via Roma 67, 56126 Pisa, Italy;
| | - Gabriella Fontanini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy
| |
Collapse
|
22
|
Guo Z, Niu X, Fu G, Yang B, Chen G, Sun S. SLC14A1 (UT-B) gene rearrangement in urothelial carcinoma of the bladder: a case report. Diagn Pathol 2020; 15:94. [PMID: 32703295 PMCID: PMC7376696 DOI: 10.1186/s13000-020-01009-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/14/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Bladder cancer (BC) is a common and deadly disease. Over the past decade, a number of genetic alterations have been reported in BC. Bladder urothelium expresses abundant urea transporter UT-B encoded by Slc14a1 gene at 18q12.3 locus, which plays an important role in preventing high concentrated urea-caused cell injury. Early genome-wide association studies (GWAS) showed that UT-B gene mutations are genetically linked to the urothelial bladder carcinoma (UBC). In this study, we examined whether Slc14a1 gene has been changed in UBC, which has never been reported. CASE PRESENTATION A 59-year-old male was admitted to a hospital with the complaint of gross hematuria for 6 days. Ultrasonography revealed a size of 2.8 × 1.7 cm mass lesion located on the rear wall and dome of the bladder. In cystoscopic examination, papillary tumoral lesions 3.0-cm in total diameter were seen on the left wall of the bladder and 2 cm to the left ureteric orifice. Transurethral resection of bladder tumor (TURBT) was performed. Histology showed high-grade non-muscle invasive UBC. Immunostaining was negative for Syn, CK7, CK20, Villin, and positive for HER2, BRCA1, GATA3. Using a fluorescence in situ hybridization (FISH), Slc14a1 gene rearrangement was identified by a pair of break-apart DNA probes. CONCLUSIONS We for the first time report a patient diagnosed with urothelial carcinoma accompanied with split Slc14a1 gene abnormality, a crucial gene in bladder.
Collapse
Affiliation(s)
- Zhongying Guo
- Department of Pathology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, 223300, China
| | - Xiaobing Niu
- Department of Urology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, 223300, China
| | - Guangbo Fu
- Department of Urology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, 223300, China
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Guangping Chen
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Su'an Sun
- Department of Pathology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, 223300, China.
| |
Collapse
|
23
|
López-Ríos F, Paz-Ares L, Sanz J, Isla D, Pijuan L, Felip E, Gómez-Román JJ, de Castro J, Conde E, Garrido P. [Updated guidelines for predictive biomarker testing in advanced non-small-cell lung cancer: A National Consensus of the Spanish Society of Pathology and the Spanish Society of Medical Oncology]. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2020; 53:167-181. [PMID: 32650968 DOI: 10.1016/j.patol.2019.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 11/25/2019] [Indexed: 01/15/2023]
Abstract
In 2011, the Spanish Society of Medical Oncology (SEOM) and the Spanish Society of Pathology (SEAP) initiated a joint project to establish guidelines for biomarker testing in patients with advanced non-small-cell lung cancer based on the information available at the time. As this field is constantly evolving, these guidelines were updated in 2012 and 2015 and now in 2019. Current evidence suggests it should be mandatory to test all patients with this kind of advanced lung cancer for EGFR and BRAF mutations, ALK and ROS1 rearrangements and PD-L1 expression. The growing need to study other emerging biomarkers has promoted the routine use of massive sequencing (next-generation sequencing, NGS). However, the coordination of every professional involved and the prioritisation of the most suitable tests and technologies for each case remain a challenge.
Collapse
Affiliation(s)
- Fernando López-Ríos
- Departamento de Patología-Laboratorio de Dianas Terapéuticas, Hospital Universitario HM Sanchinarro, CIBERONC, Madrid, España.
| | - Luis Paz-Ares
- Servicio de Oncología Médica, Hospital Universitario 12 de Octubre, Madrid, España
| | - Julián Sanz
- Departamento de Patología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, España
| | - Dolores Isla
- Servicio de Oncología Médica, Hospital Clínico Universitario Lozano Blesa, Zaragoza, España
| | - Lara Pijuan
- Departamento de Patología, Hospital del Mar, Barcelona, España
| | - Enriqueta Felip
- Departamento de Oncología Médica, Hospital Universitari Vall d'Hebron, Barcelona, España
| | - José Javier Gómez-Román
- Departamento de Patología, Hospital Universitario Marqués de Valdecilla, Universidad de Cantabria, IDIVAL, Santander, España
| | - Javier de Castro
- Departamento de Oncología Médica, Hospital Universitario La Paz, Madrid, España
| | - Esther Conde
- Departamento de Patología-Laboratorio de Dianas Terapéuticas, Hospital Universitario HM Sanchinarro, CIBERONC, Madrid, España
| | - Pilar Garrido
- Departamento de Oncología Médica, Hospital Universitario Ramón y Cajal, Universidad Alcalá, IRYCIS, CIBERONC, Madrid, España
| |
Collapse
|
24
|
Heyer EE, Blackburn J. Sequencing Strategies for Fusion Gene Detection. Bioessays 2020; 42:e2000016. [DOI: 10.1002/bies.202000016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/11/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Erin E. Heyer
- The Kinghorn Cancer CentreGarvan Institute of Medical Research 384 Victoria Street Darlinghurst NSW 2010 Australia
| | - James Blackburn
- The Kinghorn Cancer CentreGarvan Institute of Medical Research 384 Victoria Street Darlinghurst NSW 2010 Australia
- Faculty of Medicine, St. Vincent's Clinical SchoolUNSW, St Vincent's Hospital Victoria Street Darlinghurst NSW 2010 Australia
| |
Collapse
|
25
|
Kirchhoff T, Ferguson R. Germline Genetics in Immuno-oncology: From Genome-Wide to Targeted Biomarker Strategies. Methods Mol Biol 2020; 2055:93-117. [PMID: 31502148 DOI: 10.1007/978-1-4939-9773-2_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In immuno-oncology (IO), the baseline host factors attract significant clinical interest as promising predictive biomarker candidates primarily due to the feasibility of noninvasive testing and the personalized potential of IO outcome prediction catered to individual patients. Growing evidence from experimental or population-based studies suggests that the host genetic factors contribute to the immunological status of a patient as it plays out at the multiple rate-limiting steps of the cancer immunity cycle. Recent observations suggest that germline genetics may be associated with tumor microenvironment phenotypes, autoimmune toxicities, and/or efficacy of immunotherapy regimens and overall cancer survival. Despite these highly intriguing indications, the potential of germline genetic factors as personalized biomarkers of immune-checkpoint inhibition (ICI) remains vastly unexplored. In this chapter, we review the rationale for exploring the germline genetic factors as novel biomarkers predictive of IO outcomes, including ICI efficacy, toxicity, or survival, and discuss the approaches for the identification of such germline genetic surrogates. Specifically, we focus on strategies for mapping the germline genetic biomarkers of ICI using genome-wide scans (genome-wide association analyses, next-generation sequencing technologies), followed by targeted assays, to be applied in clinical use. As we discuss the limitations, we highlight a need for large collaborative consortia in these efforts and sketch possible avenues for incorporating germline genetic factors into emerging multifactorial approaches for more personalized prediction of ICI outcomes.
Collapse
Affiliation(s)
- Tomas Kirchhoff
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA.
| | - Robert Ferguson
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
26
|
Hosono Y, Masuishi T, Mitani S, Yamaguchi R, Kato S, Yoshino T, Ebi H. Evaluation of ALK Fusion Newly Identified in Colon Cancer by a Comprehensive Genomic Analysis. JCO Precis Oncol 2019; 3:1-5. [PMID: 35100727 DOI: 10.1200/po.19.00268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
| | | | | | - Rui Yamaguchi
- Aichi Cancer Center, Nagoya, Japan.,Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | - Hiromichi Ebi
- Aichi Cancer Center, Nagoya, Japan.,Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
27
|
Pagani F, Randon G, Guarini V, Raimondi A, Prisciandaro M, Lobefaro R, Di Bartolomeo M, Sozzi G, de Braud F, Gasparini P, Pietrantonio F. The Landscape of Actionable Gene Fusions in Colorectal Cancer. Int J Mol Sci 2019; 20:ijms20215319. [PMID: 31731495 PMCID: PMC6861915 DOI: 10.3390/ijms20215319] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023] Open
Abstract
The treatment scenario of metastatic colorectal cancer (mCRC) has been rapidly enriched with new chemotherapy combinations and biological agents that lead to a remarkable improvement in patients’ outcome. Kinase gene fusions account for less than 1% of mCRC overall but are enriched in patients with high microsatellite instability, RAS/BRAF wild-type colorectal cancer. mCRC patients harboring such alterations show a poor prognosis with standard treatments that could be reversed by adopting novel therapeutic strategies. Moving forward to a positive selection of mCRC patients suitable for targeted therapy in the era of personalized medicine, actionable gene fusions, although rare, represent a peculiar opportunity to disrupt a tumor alteration to achieve therapeutic goal. Here we summarize the current knowledge on potentially actionable gene fusions in colorectal cancer available from retrospective experiences and promising preliminary results of new basket trials.
Collapse
Affiliation(s)
- Filippo Pagani
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy; (F.P.); (G.R.); (V.G.); (A.R.); (M.P.); (R.L.); (M.D.B.); (F.d.B.)
| | - Giovanni Randon
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy; (F.P.); (G.R.); (V.G.); (A.R.); (M.P.); (R.L.); (M.D.B.); (F.d.B.)
| | - Vincenzo Guarini
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy; (F.P.); (G.R.); (V.G.); (A.R.); (M.P.); (R.L.); (M.D.B.); (F.d.B.)
| | - Alessandra Raimondi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy; (F.P.); (G.R.); (V.G.); (A.R.); (M.P.); (R.L.); (M.D.B.); (F.d.B.)
| | - Michele Prisciandaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy; (F.P.); (G.R.); (V.G.); (A.R.); (M.P.); (R.L.); (M.D.B.); (F.d.B.)
| | - Riccardo Lobefaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy; (F.P.); (G.R.); (V.G.); (A.R.); (M.P.); (R.L.); (M.D.B.); (F.d.B.)
| | - Maria Di Bartolomeo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy; (F.P.); (G.R.); (V.G.); (A.R.); (M.P.); (R.L.); (M.D.B.); (F.d.B.)
| | - Gabriella Sozzi
- Unit of Molecular Cytogenetics, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy; (G.S.); (P.G.)
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy; (F.P.); (G.R.); (V.G.); (A.R.); (M.P.); (R.L.); (M.D.B.); (F.d.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Patrizia Gasparini
- Unit of Molecular Cytogenetics, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy; (G.S.); (P.G.)
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy; (F.P.); (G.R.); (V.G.); (A.R.); (M.P.); (R.L.); (M.D.B.); (F.d.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- Correspondence:
| |
Collapse
|
28
|
Updated guidelines for predictive biomarker testing in advanced non-small-cell lung cancer: a National Consensus of the Spanish Society of Pathology and the Spanish Society of Medical Oncology. Clin Transl Oncol 2019; 22:989-1003. [PMID: 31598903 PMCID: PMC7260262 DOI: 10.1007/s12094-019-02218-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023]
Abstract
In 2011 the Spanish Society of Medical Oncology (SEOM) and the Spanish Society of Pathology (SEAP) started a joint project to establish guidelines on biomarker testing in patients with advanced non-small-cell lung cancer (NSCLC) based on current evidence. As this field is constantly evolving, these guidelines have been updated, previously in 2012 and 2015 and now in 2019. Current evidence suggests that the mandatory tests to conduct in all patients with advanced NSCLC are for EGFR and BRAF mutations, ALK and ROS1 rearrangements and PD-L1 expression. The growing need to study other emerging biomarkers has promoted the routine use of massive sequencing (next-generation sequencing, NGS). The coordination of every professional involved and the prioritisation of the most suitable tests and technologies for each case remains a challenge.
Collapse
|
29
|
Fluorescence in Situ Hybridization (FISH) for Detecting Anaplastic Lymphoma Kinase ( ALK) Rearrangement in Lung Cancer: Clinically Relevant Technical Aspects. Int J Mol Sci 2019; 20:ijms20163939. [PMID: 31412611 PMCID: PMC6720438 DOI: 10.3390/ijms20163939] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/08/2019] [Accepted: 08/11/2019] [Indexed: 02/06/2023] Open
Abstract
In 2011, the Vysis Break Apart ALK fluorescence in situ hybridization (FISH) assay was approved by the United States Food and Drug Administration as a companion diagnostic for detecting ALK rearrangement in lung cancer patients who may benefit from treatment of tyrosine kinase inhibitor therapy. This assay is the current “gold standard”. According to updated ALK testing guidelines from the College of American Pathologists, the International Association for the Study of Lung Cancer and the Association for Molecular Pathology published in 2018, ALK immunohistochemistry is formally an alternative to ALK FISH, and simultaneous detection of multiple hot spots, including, at least, ALK, ROS1, RET, MET, ERBB2, BRAF and KRAS genes is also recommended while performing next generation sequencing (NGS)-based testing. Therefore, ALK status in a specimen can be tested by different methods and platforms, even in the same institution or laboratory. In this review, we discuss several clinically relevant technical aspects of ALK FISH, including pros and cons of the unique two-step (50- to 100-cell) analysis approach employed in the Vysis Break Apart ALK FISH assay, including: the preset cutoff value of ≥15% for a positive result; technical aspects and biology of discordant results obtained by different methods; and incidental findings, such as ALK copy number gain or amplification and co-existent driver mutations. These issues have practical implications for ALK testing in the clinical laboratory following the updated guidelines.
Collapse
|
30
|
Heydt C, Ruesseler V, Pappesch R, Wagener S, Haak A, Siebolts U, Riedel R, Michels S, Wolf J, Schultheis AM, Rehker J, Buettner R, Merkelbach-Bruse S. Comparison of in Situ and Extraction-Based Methods for the Detection of ROS1 Rearrangements in Solid Tumors. J Mol Diagn 2019; 21:971-984. [PMID: 31382035 DOI: 10.1016/j.jmoldx.2019.06.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 05/07/2019] [Accepted: 06/12/2019] [Indexed: 11/19/2022] Open
Abstract
Clinical data confirmed that patients with ROS1 rearrangement are sensitive to specific inhibitors. Therefore, reliable detection of ROS1 rearrangements is essential. Several diagnostic techniques are currently available. However, previous studies were hampered by the low number of ROS1-positive samples. Thirty-five samples, including 32 ROS1 fluorescent in situ hybridization (FISH)-positive and three ROS1 FISH-negative samples were evaluated by ROS1 chromogenic in situ hybridization, ROS proto-oncogene 1, receptor tyrosine kinase (ROS1) immunohistochemistry (IHC), an Agilent SureSelectXT HS custom panel, the Archer FusionPlex Comprehensive Thyroid and Lung panel, and a custom NanoString fusion panel. Some samples were additionally analyzed with the Illumina TruSight Tumor 170 assay. Eleven samples were ROS1 FISH positive by a break-apart signal pattern. In all 11 samples, a ROS1 fusion was confirmed by at least one other method. The other 21 samples tested ROS1 FISH positive by an isolated 3' green signal pattern. Ten of 21 samples could be confirmed by at least two other methods. The other 11 samples tested negative by ROS1 IHC and at least one other method, indicating a false-positive ROS1 FISH result. Our study found that all ROS1 FISH-positive samples with isolated 3' green signals should be confirmed by another method. When sufficient material is available, extraction-based parallel sequencing approaches for the verification of these cases might be preferable.
Collapse
Affiliation(s)
- Carina Heydt
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Network Genomic Medicine, Cologne, Germany
| | - Vanessa Ruesseler
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Network Genomic Medicine, Cologne, Germany
| | - Roberto Pappesch
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Network Genomic Medicine, Cologne, Germany
| | - Svenja Wagener
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Network Genomic Medicine, Cologne, Germany
| | - Anja Haak
- Institute of Pathology, University Hospital Halle (Saale), Halle, Germany
| | - Udo Siebolts
- Institute of Pathology, University Hospital Halle (Saale), Halle, Germany
| | - Richard Riedel
- Network Genomic Medicine, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Köln-Bonn, University Hospital of Cologne, Cologne, Germany
| | - Sebastian Michels
- Network Genomic Medicine, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Köln-Bonn, University Hospital of Cologne, Cologne, Germany
| | - Juergen Wolf
- Network Genomic Medicine, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Köln-Bonn, University Hospital of Cologne, Cologne, Germany
| | - Anne M Schultheis
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Network Genomic Medicine, Cologne, Germany
| | - Jan Rehker
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Network Genomic Medicine, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Network Genomic Medicine, Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Network Genomic Medicine, Cologne, Germany.
| |
Collapse
|
31
|
Targeting ROS1 Rearrangements in Non-small Cell Lung Cancer: Crizotinib and Newer Generation Tyrosine Kinase Inhibitors. Drugs 2019; 79:1277-1286. [DOI: 10.1007/s40265-019-01164-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
32
|
Carapezza G, Cusi C, Rizzo E, Raddrizzani L, Di Bella S, Somaschini A, Leone A, Lupi R, Mutarelli M, Nigro V, di Bernardo D, Magni P, Isacchi A, Bosotti R. Comprehensive kinome NGS targeted expression profiling by KING-REX. BMC Genomics 2019; 20:307. [PMID: 31014245 PMCID: PMC6480677 DOI: 10.1186/s12864-019-5676-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 04/08/2019] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Protein kinases are enzymes controlling different cellular functions. Genetic alterations often result in kinase dysregulation, making kinases a very attractive class of druggable targets in several human diseases. Existing approved drugs still target a very limited portion of the human 'kinome', demanding a broader functional knowledge of individual and co-expressed kinase patterns in physiologic and pathologic settings. The development of novel rapid and cost-effective methods for kinome screening is therefore highly desirable, potentially leading to the identification of novel kinase drug targets. RESULTS In this work, we describe the development of KING-REX (KINase Gene RNA EXpression), a comprehensive kinome RNA targeted custom assay-based panel designed for Next Generation Sequencing analysis, coupled with a dedicated data analysis pipeline. We have conceived KING-REX for the gene expression analysis of 512 human kinases; for 319 kinases, paired assays and custom analysis pipeline features allow the evaluation of 3'- and 5'-end transcript imbalances as readout for the prediction of gene rearrangements. Validation tests on cell line models harboring known gene fusions demonstrated a comparable accuracy of KING-REX gene expression assessment as in whole transcriptome analyses, together with a robust detection of transcript portion imbalances in rearranged kinases, even in complex RNA mixtures or in degraded RNA. CONCLUSIONS These results support the use of KING-REX as a rapid and cost effective kinome investigation tool in the field of kinase target identification for applications in cancer biology and other human diseases.
Collapse
Affiliation(s)
| | - Carlo Cusi
- NMS Oncology, Nerviano Medical Sciences Srl, Nerviano, MI Italy
| | - Ettore Rizzo
- Department of Electrical, Computer, and Biomedical Engineering, University of Pavia, Pavia, Italy
- enGenome s.r.l., Pavia, Italy
| | | | | | | | - Antonella Leone
- NMS Oncology, Nerviano Medical Sciences Srl, Nerviano, MI Italy
| | - Rosita Lupi
- NMS Oncology, Nerviano Medical Sciences Srl, Nerviano, MI Italy
| | | | - Vincenzo Nigro
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, NA Italy
- Medical Genetics, Department of Biochemistry, Biophysics and General Pathology, University of Campania ‘Luigi Vanvitelli’, Naples, Italy
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, NA Italy
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
| | - Paolo Magni
- Department of Electrical, Computer, and Biomedical Engineering, University of Pavia, Pavia, Italy
| | | | - Roberta Bosotti
- NMS Oncology, Nerviano Medical Sciences Srl, Nerviano, MI Italy
| |
Collapse
|
33
|
Heyer EE, Deveson IW, Wooi D, Selinger CI, Lyons RJ, Hayes VM, O'Toole SA, Ballinger ML, Gill D, Thomas DM, Mercer TR, Blackburn J. Diagnosis of fusion genes using targeted RNA sequencing. Nat Commun 2019; 10:1388. [PMID: 30918253 PMCID: PMC6437215 DOI: 10.1038/s41467-019-09374-9] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 02/22/2019] [Indexed: 01/05/2023] Open
Abstract
Fusion genes are a major cause of cancer. Their rapid and accurate diagnosis can inform clinical action, but current molecular diagnostic assays are restricted in resolution and throughput. Here, we show that targeted RNA sequencing (RNAseq) can overcome these limitations. First, we establish that fusion gene detection with targeted RNAseq is both sensitive and quantitative by optimising laboratory and bioinformatic variables using spike-in standards and cell lines. Next, we analyse a clinical patient cohort and improve the overall fusion gene diagnostic rate from 63% with conventional approaches to 76% with targeted RNAseq while demonstrating high concordance for patient samples with previous diagnoses. Finally, we show that targeted RNAseq offers additional advantages by simultaneously measuring gene expression levels and profiling the immune-receptor repertoire. We anticipate that targeted RNAseq will improve clinical fusion gene detection, and its increasing use will provide a deeper understanding of fusion gene biology. Rapid and accurate detection of fusion genes is important in cancer diagnostics. Here, the authors demonstrate that targeted RNA sequencing provides fast, sensitive and quantitative gene fusion detection and overcomes the limitations of approaches currently in clinical use.
Collapse
Affiliation(s)
- Erin E Heyer
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, 2010, NSW, Australia
| | - Ira W Deveson
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, 2010, NSW, Australia.,St. Vincent's Clinical School, UNSW Australia, Sydney, 2031, NSW, Australia
| | - Danson Wooi
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, 2010, NSW, Australia.,St. Vincent's Clinical School, UNSW Australia, Sydney, 2031, NSW, Australia
| | - Christina I Selinger
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, 2050, NSW, Australia
| | - Ruth J Lyons
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, 2010, NSW, Australia
| | - Vanessa M Hayes
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, 2010, NSW, Australia.,St. Vincent's Clinical School, UNSW Australia, Sydney, 2031, NSW, Australia.,Faculty of Health Sciences, University of Limpopo, Turfloop Campus, Mankweng, 0727, South Africa.,School of Health Systems and Public Health, University of Pretoria, Pretoria, 0002, South Africa.,Central Clinical School, University of Sydney, Sydney, 2006, NSW, Australia
| | - Sandra A O'Toole
- St. Vincent's Clinical School, UNSW Australia, Sydney, 2031, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, 2050, NSW, Australia.,Central Clinical School, University of Sydney, Sydney, 2006, NSW, Australia.,The Kinghorn Cancer Centre and Cancer Division, Garvan Institute of Medical Research, Sydney, 2010, NSW, Australia.,Australian Clinical Labs, Sydney, 2010, NSW, Australia
| | - Mandy L Ballinger
- The Kinghorn Cancer Centre and Cancer Division, Garvan Institute of Medical Research, Sydney, 2010, NSW, Australia
| | - Devinder Gill
- Department of Haematology, Princess Alexandra Hospital, Brisbane, 4102, QLD, Australia
| | - David M Thomas
- The Kinghorn Cancer Centre and Cancer Division, Garvan Institute of Medical Research, Sydney, 2010, NSW, Australia
| | - Tim R Mercer
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, 2010, NSW, Australia. .,St. Vincent's Clinical School, UNSW Australia, Sydney, 2031, NSW, Australia. .,Altius Institute for Biomedical Sciences, Seattle, 98121, WA, USA.
| | - James Blackburn
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, 2010, NSW, Australia. .,St. Vincent's Clinical School, UNSW Australia, Sydney, 2031, NSW, Australia.
| |
Collapse
|
34
|
Blidner RA, Haynes BC, Hyter S, Schmitt S, Pessetto ZY, Godwin AK, Su D, Hurban P, van Kempen LC, Aguirre ML, Gokul S, Cardwell RD, Latham GJ. Design, Optimization, and Multisite Evaluation of a Targeted Next-Generation Sequencing Assay System for Chimeric RNAs from Gene Fusions and Exon-Skipping Events in Non-Small Cell Lung Cancer. J Mol Diagn 2019; 21:352-365. [PMID: 30529127 PMCID: PMC7057224 DOI: 10.1016/j.jmoldx.2018.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/11/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022] Open
Abstract
Lung cancer accounts for approximately 14% of all newly diagnosed cancers and is the leading cause of cancer-related deaths. Chimeric RNA resulting from gene fusions (RNA fusions) and other RNA splicing errors are driver events and clinically addressable targets for non-small cell lung cancer (NSCLC). The reliable assessment of these RNA markers by next-generation sequencing requires integrated reagents, protocols, and interpretive software that can harmonize procedures and ensure consistent results across laboratories. We describe the development and verification of a system for targeted RNA sequencing for the analysis of challenging, low-input solid tumor biopsies that includes reagents for nucleic acid quantification and library preparation, run controls, and companion bioinformatics software. Assay development reconciled sequence discrepancies in public databases, created predictive formalin-fixed, paraffin-embedded RNA qualification metrics, and eliminated read misidentification attributable to index hopping events on the next-generation sequencing flow cell. The optimized and standardized system was analytically verified internally and in a multiphase study conducted at five independent laboratories. The results show accurate, reproducible, and sensitive detection of RNA fusions, alternative splicing events, and other expression markers of NSCLC. This comprehensive approach, combining sample quantification, quality control, library preparation, and interpretive bioinformatics software, may accelerate the routine implementation of targeted RNA sequencing of formalin-fixed, paraffin-embedded samples relevant to NSCLC.
Collapse
Affiliation(s)
| | | | - Stephen Hyter
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Sarah Schmitt
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Ziyan Y Pessetto
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas; University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas
| | - Dan Su
- Q Squared Solutions Expression Analysis LLC, Morrisville, North Carolina
| | - Patrick Hurban
- Q Squared Solutions Expression Analysis LLC, Morrisville, North Carolina
| | - Léon C van Kempen
- The Molecular Pathology Centre, Jewish General Hospital, Montreal, Quebec, Canada
| | - Maria L Aguirre
- The Molecular Pathology Centre, Jewish General Hospital, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
35
|
Schröder J, Kumar A, Wong SQ. Overview of Fusion Detection Strategies Using Next-Generation Sequencing. Methods Mol Biol 2019; 1908:125-138. [PMID: 30649725 DOI: 10.1007/978-1-4939-9004-7_9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Structural gene fusion rearrangements leading to aberrant signaling are frequently detected in many cancer types. Gene fusions have significant prognostic and predictive value and are screened as part of molecular pathology testing for patient management. Many bioinformatic approaches have been developed to detect fusion mutations including whole-genome sequencing, targeted-based hybridization capture, and transcriptome-based approaches. Here we describe the most commonly used experimental methods to sequence and identify gene fusions using either DNA or RNA. We contrast experimental approaches both in the research and diagnostic setting and describe typical bioinformatic pipelines and software packages used to identify fusions.
Collapse
Affiliation(s)
- Jan Schröder
- Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | - Amit Kumar
- Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | - Stephen Q Wong
- Peter MacCallum Cancer Center, Melbourne, VIC, Australia.
| |
Collapse
|
36
|
Alì G, Bruno R, Fontanini G. In Reply. Arch Pathol Lab Med 2018; 142:1452. [PMID: 30500281 DOI: 10.5858/arpa.2018-0357-le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Rossella Bruno
- 2 Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Gabriella Fontanini
- 3 Program of Pleuropulmonary Pathology, Azienda Ospedaliero Universitaria Pisana, AOUP, Pisa, Italy.,2 Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| |
Collapse
|
37
|
Ginestet F, Lambros L, Le Flahec G, Marcorelles P, Uguen A. Evaluation of a Dual ALK/ROS1 Fluorescent In Situ Hybridization Test in Non–Small-cell Lung Cancer. Clin Lung Cancer 2018; 19:e647-e653. [DOI: 10.1016/j.cllc.2018.04.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/17/2018] [Accepted: 04/24/2018] [Indexed: 10/17/2022]
|
38
|
Davies KD, Le AT, Sheren J, Nijmeh H, Gowan K, Jones KL, Varella-Garcia M, Aisner DL, Doebele RC. Comparison of Molecular Testing Modalities for Detection of ROS1 Rearrangements in a Cohort of Positive Patient Samples. J Thorac Oncol 2018; 13:1474-1482. [PMID: 29935306 DOI: 10.1016/j.jtho.2018.05.041] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/25/2018] [Accepted: 05/31/2018] [Indexed: 12/16/2022]
Abstract
INTRODUCTION ROS1 gene fusions are a well-characterized class of oncogenic driver found in approximately 1% to 2% of NSCLC patients. ROS1-directed therapy in these patients is more efficacious and is associated with fewer side effects compared to chemotherapy and is thus now considered standard-of-care for patients with advanced disease. Consequently, accurate detection of ROS1 rearrangements/fusions in clinical tumor samples is vital. In this study, we compared the performance of three common molecular testing approaches on a cohort of ROS1 rearrangement/fusion-positive patient samples. METHODS Twenty-three ROS1 rearrangement/fusion-positive clinical samples were assessed by at least two of the following molecular testing methodologies: break-apart fluorescence in situ hybridization, DNA-based hybrid capture library preparation followed by next-generation sequencing (NGS), and RNA-based anchored multiplex polymerase chain reaction library preparation followed by NGS. RESULTS None of the testing methodologies demonstrated 100% sensitivity in detection of ROS1 rearrangements/fusions. Fluorescence in situ hybridization results were negative in 2 of 20 tested samples, the DNA-based NGS assay was negative in 4 of 18 tested samples, and the RNA-based NGS assay was negative in 3 of 19 tested samples. For all three testing approaches, we identified assay characteristics that likely contributed to false-negative results. Additionally, we report that genomic breakpoints are an unreliable predictor of breakpoints at the transcript level, likely due to alternative splicing. CONCLUSIONS ROS1 rearrangement/fusion detection in the clinical setting is complex and all methodologies have inherent limitations of which users must be aware to correctly interpret results.
Collapse
Affiliation(s)
- Kurtis D Davies
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Anh T Le
- Department of Medicine - Division of Medical Oncology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Jamie Sheren
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Hala Nijmeh
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Katherine Gowan
- Department of Pediatrics - Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Kenneth L Jones
- Department of Pediatrics - Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Marileila Varella-Garcia
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado; Department of Medicine - Division of Medical Oncology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Dara L Aisner
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Robert C Doebele
- Department of Medicine - Division of Medical Oncology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
39
|
Letovanec I, Finn S, Zygoura P, Smyth P, Soltermann A, Bubendorf L, Speel EJ, Marchetti A, Nonaka D, Monkhorst K, Hager H, Martorell M, Sejda A, Cheney R, Hernandez-Losa J, Verbeken E, Weder W, Savic S, Di Lorito A, Navarro A, Felip E, Warth A, Baas P, Meldgaard P, Blackhall F, Dingemans AM, Dienemann H, Dziadziuszko R, Vansteenkiste J, O'Brien C, Geiger T, Sherlock J, Schageman J, Dafni U, Kammler R, Kerr K, Thunnissen E, Stahel R, Peters S. Evaluation of NGS and RT-PCR Methods for ALK Rearrangement in European NSCLC Patients: Results from the European Thoracic Oncology Platform Lungscape Project. J Thorac Oncol 2018; 13:413-425. [PMID: 29191776 DOI: 10.1016/j.jtho.2017.11.117] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/14/2017] [Accepted: 11/19/2017] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The reported prevalence of ALK receptor tyrosine kinase gene (ALK) rearrangement in NSCLC ranges from 2% to 7%. The primary standard diagnostic method is fluorescence in situ hybridization (FISH). Recently, immunohistochemistry (IHC) has also proved to be a reproducible and sensitive technique. Reverse-transcriptase polymerase chain reaction (RT-PCR) has also been advocated, and most recently, the advent of targeted next-generation sequencing (NGS) for ALK and other fusions has become possible. This study compares anaplastic lymphoma kinase (ALK) evaluation with all four techniques in resected NSCLC from the large European Thoracic Oncology Platform Lungscape cohort. METHODS A total of 96 cases from the European Thoracic Oncology Platform Lungscape iBiobank, with any ALK immunoreactivity were examined by FISH, central RT-PCR, and NGS. An H-score higher than 120 defines IHC positivity. RNA was extracted from the same formalin-fixed, paraffin-embedded tissues. For RT-PCR, primers covered the most frequent ALK translocations. For NGS, the Oncomine Solid Tumour Fusion Transcript Kit (Thermo Fisher Scientific, Waltham, MA) was used. The concordance was assessed using the Cohen κ coefficient (two-sided α ≤ 5%). RESULTS NGS provided results for 77 of the 95 cases tested (81.1%), whereas RT-PCR provided results for 77 of 96 (80.2%). Concordance occurred in 55 cases of the 60 cases tested with all four methods (43 ALK negative and 12 ALK positive). Using ALK copositivity for IHC and FISH as the criterion standard, we derived a sensitivity for RT-PCR/NGS of 70.0%/85.0%, with a specificity of 87.1%/79.0%. When either RT-PCR or NGS was combined with IHC, the sensitivity remained the same, whereas the specificity increased to 88.7% and 83.9% respectively. CONCLUSION NGS evaluation with the Oncomine Solid Tumour Fusion transcript kit and RT-PCR proved to have high sensitivity and specificity, advocating their use in routine practice. For maximal sensitivity and specificity, ALK status should be assessed by using two techniques and a third one in discordant cases. We therefore propose a customizable testing algorithm. These findings significantly influence existing testing paradigms and have clear clinical and economic impact.
Collapse
Affiliation(s)
- Igor Letovanec
- Institute of Pathology, Centre Hospitalier Universitaire Vaudois CHUV, Lausanne, Switzerland.
| | - Stephen Finn
- Department of Histopathology, St James's Hospital and Trinity College, Dublin, Ireland
| | | | - Paul Smyth
- Department of Histopathology, St James's Hospital and Trinity College, Dublin, Ireland
| | - Alex Soltermann
- Institute of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Ernst-Jan Speel
- Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Antonio Marchetti
- Center of Predicitve Molecular Medicine, CeSI, University of Chieti-Pescara, Chieti, Italy
| | - Daisuke Nonaka
- Department of Histopathology, The Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Kim Monkhorst
- Division of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Henrik Hager
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Miguel Martorell
- Department of Pathology, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Aleksandra Sejda
- Department of Pathomorphology, Medical University of Gdansk, Gdansk, Poland
| | - Richard Cheney
- Department of Pathology, State University of New York at Buffalo, Buffalo, New York
| | | | - Eric Verbeken
- Department of Pathology, University Hospital KU Leuven, Leuven, Belgium
| | - Walter Weder
- Department of Thoracic Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Spasenija Savic
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Alessia Di Lorito
- Center of Predicitve Molecular Medicine, CeSI, University of Chieti-Pescara, Chieti, Italy
| | - Atilio Navarro
- Department of Pathology, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Enriqueta Felip
- Medical Oncology Department, Vall d'Hebrone University Hospital, Barcelona, Spain
| | - Arne Warth
- Department of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Paul Baas
- Department of Thoracic Oncology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Peter Meldgaard
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Fiona Blackhall
- Deparment of Medical Oncology, The Chrisite NHS Foundation Trust, Manchester, United Kingdom
| | - Anne-Marie Dingemans
- Department of Pulmonology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Hendrik Dienemann
- Department of Surgery, Thoraxklinik at Heidelberg University, Heidelberg, Germany
| | - Rafal Dziadziuszko
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Poland
| | - Johan Vansteenkiste
- Department of Respiratory Oncology, University Hospital KU Leuven, Leuven, Belgium
| | - Cathal O'Brien
- Department of Histopathology, St James's Hospital and Trinity College, Dublin, Ireland
| | - Thomas Geiger
- European Thoracic Oncology Platform, Bern, Switzerland
| | - Jon Sherlock
- Thermo Fisher Scientific, Paisley, United Kingdom
| | | | - Urania Dafni
- Frontier Science Foundation-Hellas & University of Athens, Athens, Greece
| | | | - Keith Kerr
- Department of Pathology, Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Rolf Stahel
- Clinic of Oncology, University Hospital Zürich, Zürich, Switzerland
| | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois CHUV, Lausanne, Switzerland
| |
Collapse
|
40
|
Alì G, Bruno R, Savino M, Giannini R, Pelliccioni S, Menghi M, Boldrini L, Proietti A, Chella A, Ribechini A, Fontanini G. Analysis of Fusion Genes by NanoString System: A Role in Lung Cytology? Arch Pathol Lab Med 2018; 142:480-489. [PMID: 29372843 DOI: 10.5858/arpa.2017-0135-ra] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - Patients with non-small cell lung cancer harboring ALK receptor tyrosine kinase ( ALK), ROS proto-oncogene 1 ( ROS1), and ret proto-oncogene ( RET) gene rearrangements can benefit from specific kinase inhibitors. Detection of fusion genes is critical for determining the best treatment. Assessing rearrangements in non-small cell lung cancer remains challenging, particularly for lung cytology. OBJECTIVE - To examine the possible application of the multiplex, transcript-based NanoString system (NanoString Technologies, Seattle, Washington) in the evaluation of fusion genes in lung adenocarcinoma samples. DATA SOURCES - This study is a narrative literature review. Studies about NanoString, gene fusions, and lung adenocarcinoma were collected from PubMed (National Center for Biotechnology Information, Bethesda, Maryland). We found 7 articles about the application of the NanoString system to detect fusion genes on formalin-fixed, paraffin-embedded tumor tissues and one article evaluating the adequacy of lung cytologic specimens for NanoString gene expression analysis. CONCLUSIONS - To maximize the yield of molecular tests on small lung biopsies, the NanoString nCounter system has been suggested to detect fusion genes. NanoString fusion gene assays have been successfully applied on formalin-fixed, paraffin-embedded tissues. Although there are only a few studies available, the application of NanoString assays may also be feasible in lung cytology. According to available data, the NanoString system could strengthen the routine molecular characterization of lung adenocarcinoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Gabriella Fontanini
- From the Unit of Pathological Anatomy (Drs Alì and Proietti and Ms Pelliccioni) and Pneumology (Dr Chella), the Endoscopic Section of Pneumology (Dr Ribechini), and the Program of Pleuropulmonary Pathology (Dr Fontanini), Azienda Ospedaliero Universitaria Pisana, Pisa, Italy; the Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy (Drs Bruno, Giannini, and Boldrini); and Diatech Pharmacogenetics srl, Jesi, Italy (Drs Savino and Menghi)
| |
Collapse
|
41
|
Troncone G. All-in-one: The dream and reality of molecular cytopathology testing on routine lung cancer smears. Cancer Cytopathol 2018; 126:155-157. [PMID: 29364571 DOI: 10.1002/cncy.21962] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/05/2017] [Accepted: 12/08/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
42
|
Wu HT, Li K, Wang G, Yang XX, Zhu A, Xu XP, Li M, Wu YS, Liu TC. Development of a high-throughput and sensitive assay of fusion genes in lung cancer by array-based MALDI-TOFMS. RSC Adv 2018; 8:27935-27945. [PMID: 35548167 PMCID: PMC9087866 DOI: 10.1039/c8ra05165h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 07/07/2018] [Indexed: 11/21/2022] Open
Abstract
ALK (anaplastic lymphoma kinase gene), ROS1 (ros proto-oncogene 1) and RET (ret proto-oncogene) fusions are oncogenic drivers in non-small cell lung cancer (NSCLC). Methods like fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC) are highly sensitive but subjectively analyzed, labor intensive, expensive and unsuitable for multiple fusion gene screening. This study aimed to establish a high-throughput, sensitive and cost-effective screening method (array-based matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, array-based MALDI-TOFMS) for ALK, ROS1 and RET fusion detection. This method was established with three fusion gene positive cell lines (H2228, ALK positive; HCC78, ROS1 positive; LC-2/AD, RET positive) and negative samples. Then, 34 clinical samples were selected and detected by Sanger sequencing, next generation sequencing (NGS) and array-based MALDI-TOFMS. The results were compared and analyzed and Sanger sequencing was considered the standard. 7 cases showed ALK fusions, 1 case showed ROS1 fusions, no case showed RET fusions and 4 cases were both ALK and ROS1 fusions. Results showed that array-based MALDI-TOFMS was 100% concordant with Sanger sequencing and NGS 82.3%. In this study, we reported the utility of array-based MALDI-TOFMS in the assessment of ALK, ROS1 and RET fusions in routine lung biopsies of FFPE and fresh tissue specimens. Besides, this method may also be applied to the diagnosis, monitoring and prognosis of illness. This study established a high-throughput, sensitive and cost-effective method to detect three lung fusion genes of 96 samples at one time.![]()
Collapse
Affiliation(s)
- Han-Tao Wu
- Institute of Antibody Engineering
- School of Laboratory Medicine and Biotechnology
- Southern Medical University
- Guangzhou 510515
- P. R. China
| | - Kun Li
- Institute of Antibody Engineering
- School of Laboratory Medicine and Biotechnology
- Southern Medical University
- Guangzhou 510515
- P. R. China
| | - Gang Wang
- Institute of Antibody Engineering
- School of Laboratory Medicine and Biotechnology
- Southern Medical University
- Guangzhou 510515
- P. R. China
| | - Xue-Xi Yang
- Institute of Antibody Engineering
- School of Laboratory Medicine and Biotechnology
- Southern Medical University
- Guangzhou 510515
- P. R. China
| | - Anna Zhu
- Guangzhou Darui Biotechnology Co. Ltd
- Guangzhou 510665
- China
| | - Xu-Ping Xu
- Guangzhou Darui Biotechnology Co. Ltd
- Guangzhou 510665
- China
| | - Ming Li
- Institute of Antibody Engineering
- School of Laboratory Medicine and Biotechnology
- Southern Medical University
- Guangzhou 510515
- P. R. China
| | - Ying-Song Wu
- Institute of Antibody Engineering
- School of Laboratory Medicine and Biotechnology
- Southern Medical University
- Guangzhou 510515
- P. R. China
| | - Tian-Cai Liu
- Institute of Antibody Engineering
- School of Laboratory Medicine and Biotechnology
- Southern Medical University
- Guangzhou 510515
- P. R. China
| |
Collapse
|
43
|
Vendrell JA, Taviaux S, Béganton B, Godreuil S, Audran P, Grand D, Clermont E, Serre I, Szablewski V, Coopman P, Mazières J, Costes V, Pujol JL, Brousset P, Rouquette I, Solassol J. Detection of known and novel ALK fusion transcripts in lung cancer patients using next-generation sequencing approaches. Sci Rep 2017; 7:12510. [PMID: 28970558 PMCID: PMC5624911 DOI: 10.1038/s41598-017-12679-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/04/2017] [Indexed: 12/25/2022] Open
Abstract
Rearrangements of the anaplastic lymphoma kinase (ALK) gene in non-small cell lung cancer (NSCLC) represent a novel molecular target in a small subset of tumors. Although ALK rearrangements are usually assessed by immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH), molecular approaches have recently emerged as relevant alternatives in routine laboratories. Here, we evaluated the use of two different amplicon-based next-generation sequencing (NGS) methods (AmpliSeq and Archer®FusionPlex®) to detect ALK rearrangements, and compared these with IHC and FISH. A total of 1128 NSCLC specimens were screened using conventional analyses, and a subset of 37 (15 ALK-positive, and 22 ALK-negative) samples were selected for NGS assays. Although AmpliSeq correctly detected 25/37 (67.6%) samples, 1/37 (2.7%) and 11/37 (29.7%) specimens were discordant and uncertain, respectively, requiring further validation. In contrast, Archer®FusionPlex® accurately classified all samples and allowed the correct identification of one rare DCTN1-ALK fusion, one novel CLIP1-ALK fusion, and one novel GCC2-ALK transcript. Of particular interest, two out of three patients harboring these singular rearrangements were treated with and sensitive to crizotinib. These data show that Archer®FusionPlex® may provide an effective and accurate alternative to FISH testing for the detection of known and novel ALK rearrangements in clinical diagnostic settings.
Collapse
Affiliation(s)
- Julie A Vendrell
- CHU Montpellier, Arnaud de Villeneuve Hospital, Department of Pathology, Montpellier, Université de Montpellier, Montpellier, France
| | - Sylvie Taviaux
- CHU Montpellier, Arnaud de Villeneuve Hospital, Department of Pathology, Montpellier, Université de Montpellier, Montpellier, France
| | - Benoît Béganton
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier (ICM), Montpellier, France
| | - Sylvain Godreuil
- CHU Montpellier, Arnaud de Villeneuve Hospital, Department of Bacteriology, Université de Montpellier, Montpellier, France
| | - Patricia Audran
- Institut du Cancer de Montpellier (ICM), Department of Biopathology, Montpellier, France
| | - David Grand
- Department of Pathology, Institut Universitaire du Cancer Toulouse Oncopole, CHU de Toulouse, Toulouse, France
| | - Estelle Clermont
- Department of Pathology, Institut Universitaire du Cancer Toulouse Oncopole, CHU de Toulouse, Toulouse, France
| | - Isabelle Serre
- CHU Montpellier, Arnaud de Villeneuve Hospital, Department of Pathology, Montpellier, Université de Montpellier, Montpellier, France
| | - Vanessa Szablewski
- CHU Montpellier, Arnaud de Villeneuve Hospital, Department of Pathology, Montpellier, Université de Montpellier, Montpellier, France
| | - Peter Coopman
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier (ICM), Montpellier, France
| | - Julien Mazières
- Thoracic Oncology Department, Larrey Hospital, University Hospital of Toulouse, Toulouse, France
| | - Valérie Costes
- CHU Montpellier, Arnaud de Villeneuve Hospital, Department of Pathology, Montpellier, Université de Montpellier, Montpellier, France
| | - Jean-Louis Pujol
- CHU Montpellier, Arnaud de Villeneuve Hospital, Department of Thoracic Oncology, Université de Montpellier, Montpellier, France
| | - Pierre Brousset
- Department of Pathology, Institut Universitaire du Cancer Toulouse Oncopole, CHU de Toulouse, Toulouse, France.,Laboratoire d'excellence Labex TOUCAN, Toulouse, France
| | - Isabelle Rouquette
- Department of Pathology, Institut Universitaire du Cancer Toulouse Oncopole, CHU de Toulouse, Toulouse, France
| | - Jérôme Solassol
- CHU Montpellier, Arnaud de Villeneuve Hospital, Department of Pathology, Montpellier, Université de Montpellier, Montpellier, France. .,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier (ICM), Montpellier, France.
| |
Collapse
|
44
|
ALK in Non-Small Cell Lung Cancer (NSCLC) Pathobiology, Epidemiology, Detection from Tumor Tissue and Algorithm Diagnosis in a Daily Practice. Cancers (Basel) 2017; 9:cancers9080107. [PMID: 28805682 PMCID: PMC5575610 DOI: 10.3390/cancers9080107] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/08/2017] [Accepted: 08/10/2017] [Indexed: 12/25/2022] Open
Abstract
Patients with advanced-stage non-small cell lung carcinoma (NSCLC) harboring an ALK rearrangement, detected from a tissue sample, can benefit from targeted ALK inhibitor treatment. Several increasingly effective ALK inhibitors are now available for treatment of patients. However, despite an initial favorable response to treatment, in most cases relapse or progression occurs due to resistance mechanisms mainly caused by mutations in the tyrosine kinase domain of ALK. The detection of an ALK rearrangement is pivotal and can be done using different methods, which have variable sensitivity and specificity depending, in particular, on the quality and quantity of the patient’s sample. This review will first highlight briefly some information regarding the pathobiology of an ALK rearrangement and the epidemiology of patients harboring this genomic alteration. The different methods used to detect an ALK rearrangement as well as their advantages and disadvantages will then be examined and algorithms proposed for detection in daily routine practice.
Collapse
|
45
|
Pisapia P, Lozano MD, Vigliar E, Bellevicine C, Pepe F, Malapelle U, Troncone G. ALK and ROS1 testing on lung cancer cytologic samples: Perspectives. Cancer Cytopathol 2017; 125:817-830. [DOI: 10.1002/cncy.21899] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/10/2017] [Accepted: 07/10/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Pasquale Pisapia
- Department of Public Health; University of Naples Federico II; Naples Italy
| | - Maria D. Lozano
- Department of Pathology; University Clinic of Navarra; Pamplona Spain
| | - Elena Vigliar
- Department of Public Health; University of Naples Federico II; Naples Italy
| | | | - Francesco Pepe
- Department of Public Health; University of Naples Federico II; Naples Italy
| | - Umberto Malapelle
- Department of Public Health; University of Naples Federico II; Naples Italy
| | - Giancarlo Troncone
- Department of Public Health; University of Naples Federico II; Naples Italy
| |
Collapse
|
46
|
Evangelista AF, Zanon MF, Carloni AC, de Paula FE, Morini MA, Ferreira-Neto M, Soares IC, Miziara JE, de Marchi P, Scapulatempo-Neto C, Reis RM. Detection of ALK fusion transcripts in FFPE lung cancer samples by NanoString technology. BMC Pulm Med 2017; 17:86. [PMID: 28549458 PMCID: PMC5446704 DOI: 10.1186/s12890-017-0428-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 05/19/2017] [Indexed: 11/29/2022] Open
Abstract
Background ALK-rearranged lung cancers exhibit specific pathologic and clinical features and are responsive to anti-ALK therapies. Therefore, the detection of ALK-rearrangement is fundamental for personalized lung cancer therapy. Recently, new molecular techniques, such as NanoString nCounter, have been developed to detect ALK fusions with more accuracy and sensitivity. Methods In the present study, we intended to validate a NanoString nCounter ALK-fusion panel in routine biopsies of FFPE lung cancer patients. A total of 43 samples were analyzed, 13 ALK-positive and 30 ALK-negative, as previously detected by FISH and/or immunohistochemistry. Results The NanoString panel detected the presence of the EML4-ALK, KIF5B-ALK and TFG-ALK fusion variants. We observed that all the 13 ALK-positive cases exhibited genetic aberrations by the NanoString methodology. Namely, six cases (46.15%) presented EML-ALK variant 1, two (15.38%) presented EML-ALK variant 2, two (15.38%) presented EML-ALK variant 3a, and three (23.07%) exhibited no variant but presented unbalanced expression between 5’/3’ exons, similar to other positive samples. Importantly, for all these analyses, the initial input of RNA was 100 ng, and some cases displayed poor RNA quality measurements. Conclusions In this study, we reported the great utility of NanoString technology in the assessment of ALK fusions in routine lung biopsies of FFPE specimens. Electronic supplementary material The online version of this article (doi:10.1186/s12890-017-0428-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adriane F Evangelista
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Maicon F Zanon
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Adriana Cruvinel Carloni
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Flávia E de Paula
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Mariana Andozia Morini
- Department of Pathology, Barretos Cancer Hospital, Rua Antenor Duarte Villela 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Maressa Ferreira-Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Iberê Cauduro Soares
- Department of Pathology, Barretos Cancer Hospital, Rua Antenor Duarte Villela 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Jose Elias Miziara
- Department of Thoracic Surgery, Barretos Cancer Hospital, Rua Antenor Duarte Villela 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Pedro de Marchi
- Department of Clinical Oncology, Barretos Cancer Hospital, Rua Antenor Duarte Villela 1331, Barretos, CEP 14784-400, Sao Paulo, Brazil
| | - Cristovam Scapulatempo-Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil.,Department of Pathology, Barretos Cancer Hospital, Rua Antenor Duarte Villela 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Rui M Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil. .,Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, 4710-057, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, 4710-057, Portugal.
| |
Collapse
|
47
|
Leong TL, Christie M, Kranz S, Pham K, Hsu A, Irving LB, Asselin-Labat ML, Steinfort DP. Evaluating the Genomic Yield of a Single Endobronchial Ultrasound-guided Transbronchial Needle Aspiration in Lung Cancer: Meeting the Challenge of Doing More With Less. Clin Lung Cancer 2017; 18:e467-e472. [PMID: 28576592 DOI: 10.1016/j.cllc.2017.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/27/2017] [Accepted: 05/02/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Minimally invasive techniques, including endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA), yield small specimens that are adequate for cytologic diagnosis of lung cancer, but also need to provide material for molecular analysis to guide treatment. The number of EBUS-TBNA passes needed for mutation testing remains unclear. We sought to assess the adequacy of a single pass for genomic profiling of actionable mutations. METHODS In a prospective observational study, paired samples from the same lesion were obtained from patients undergoing EBUS-TBNA for lung cancer diagnosis/staging. Following tumor cell confirmation by rapid on-site evaluation, a "reference" sample comprising ≥ 3 passes was obtained and formalin-fixed paraffin-embedded. A "study" sample comprising a single pass was taken and snap-frozen. The primary outcome was DNA yield and quality from a single pass. The secondary outcome was diagnostic accuracy of a single pass for detecting actionable mutations. RESULTS In 40 patients, single-pass specimens yielded a mean 3.98 μg of highly intact DNA, well above the minimum threshold for targeted sequencing, which was performed in adenocarcinoma cases (n = 24). In 23 cases, there was 100% agreement in mutation status between reference and study samples. In 1 case, the reference sample failed to generate a molecular diagnosis owing to insufficient tumor cells; however, the study specimen identified a KRAS mutation. Tumor cell percentage in mutation-positive specimens was 1% to 70%, suggesting that single-pass samples detect mutations even when tumor cell content is low. CONCLUSION Single EBUS-TBNA passes yield DNA of high quantity and quality with high accuracy for molecular profiling, irrespective of tumor cell content.
Collapse
Affiliation(s)
- Tracy L Leong
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, University of Melbourne, Parkville, Victoria, Australia.
| | - Michael Christie
- Department of Anatomical Pathology, Royal Melbourne Hospital, Parkville, Victoria, Australia; Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Sevastjan Kranz
- Department of Anatomical Pathology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Kym Pham
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Arthur Hsu
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Louis B Irving
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia; Department of Respiratory Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Marie-Liesse Asselin-Labat
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Daniel P Steinfort
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia; Department of Respiratory Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|