1
|
Murciano-Goroff YR, Uppal M, Chen M, Harada G, Schram AM. Basket Trials: Past, Present, and Future. ANNUAL REVIEW OF CANCER BIOLOGY 2024; 8:59-80. [PMID: 38938274 PMCID: PMC11210107 DOI: 10.1146/annurev-cancerbio-061421-012927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Large-scale tumor molecular profiling has revealed that diverse cancer histologies are driven by common pathways with unifying biomarkers that can be exploited therapeutically. Disease-agnostic basket trials have been increasingly utilized to test biomarker-driven therapies across cancer types. These trials have led to drug approvals and improved the lives of patients while simultaneously advancing our understanding of cancer biology. This review focuses on the practicalities of implementing basket trials, with an emphasis on molecularly targeted trials. We examine the biologic subtleties of genomic biomarker and patient selection, discuss previous successes in drug development facilitated by basket trials, describe certain novel targets and drugs, and emphasize practical considerations for participant recruitment and study design. This review also highlights strategies for aiding patient access to basket trials. As basket trials become more common, steps to ensure equitable implementation of these studies will be critical for molecularly targeted drug development.
Collapse
Affiliation(s)
| | - Manik Uppal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Monica Chen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Guilherme Harada
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alison M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
2
|
Chiarle R, Cheong TC, Jang A, Wang Q, Leonardi G, Ricciuti B, Alessi J, Federico AD, Awad M, Lehtinen M, Harris M. Mechanistic patterns and clinical implications of oncogenic tyrosine kinase fusions in human cancers. RESEARCH SQUARE 2024:rs.3.rs-3782958. [PMID: 38313284 PMCID: PMC10836111 DOI: 10.21203/rs.3.rs-3782958/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Tyrosine kinase (TK) fusions are frequently found in cancers, either as initiating events or as a mechanism of resistance to targeted therapy. Partner genes and exons in most TK fusions are typical and recurrent, but the underlying mechanisms and clinical implications of these patterns are poorly understood. Here, we investigated structures of > 8,000 kinase fusions and explore their generative mechanisms by applying newly developed experimental framework integrating high-throughput genome-wide gene fusion sequencing and clonal selection called Functionally Active Chromosomal Translocation Sequencing (FACTS). We discovered that typical oncogenic TK fusions recurrently seen in patients are selected from large pools of chromosomal rearrangements spontaneously occurring in cells based on two major determinants: active transcription of the fusion partner genes and protein stability. In contrast, atypical TK fusions that are rarely seen in patients showed reduced protein stability, decreased downstream oncogenic signaling, and were less responsive to inhibition. Consistently, patients with atypical TK fusions were associated with a reduced response to TKI therapies, as well as a shorter progression-free survival (PFS) and overall survival (OS) compared to patients with typical TK fusions. These findings highlight the principles of oncogenic TK fusion formation and their selection in cancers, with clinical implications for guiding targeted therapy.
Collapse
Affiliation(s)
| | | | - Ahram Jang
- Boston Children's Hospital and Harvard Medical School
| | - Qi Wang
- Boston Children's Hospital and Harvard Medical School
| | | | | | | | | | | | | | - Marian Harris
- Boston Children's Hospital and Harvard Medical School
| |
Collapse
|
3
|
Batra U, Nathany S, Sharma M, Jain P, Mehta A, Bansal A. A novel EML4-NTRK3 fusion in lung adenocarcinoma with dramatic response to entrectinib. J Cancer Res Ther 2023; 19:2045-2047. [PMID: 38376315 DOI: 10.4103/jcrt.jcrt_231_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 04/25/2022] [Indexed: 02/21/2024]
Abstract
ABSTRACT In-frame fusions in NTRK genes, with intact kinase domain, have been reported to occur at higher frequencies in rare tumors like infantile fibrosarcoma, congenital mesoblastic nephroma, and secretory carcinoma, whereas they occur at very low frequencies in common malignancies like NSCLC and colon cancers (0.1%-1%). Despite the rare occurrence, these alterations have gained importance owing to approval of drugs like entrectinib and larotrectinib targeting the kinase domain of the gene. More than 50 fusion partners have been described, and only in-frame fusions result in constitutive ligand-independent kinase activity leading to oncogenesis. The commonly reported NTRK fusions in the lung include SQSTM1-NTRK1, ETV6-NTRK3, and SQSTM1-NTRK3. Detection of these rests on the use of conventional modalities like Immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH); however, accurate characterization requires direct sequencing methods. We report an interesting case of an NTRK fusion-positive NSCLC, exhibiting good response to entrectinib.
Collapse
Affiliation(s)
- Ullas Batra
- Department of Medical Oncology, Section of Molecular Diagnostics, Pathology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Shrinidhi Nathany
- Department of Medical Oncology, Section of Molecular Diagnostics, Pathology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Mansi Sharma
- Department of Medical Oncology, Section of Molecular Diagnostics, Pathology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Parveen Jain
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Anurag Mehta
- Department of Medical Oncology, Section of Molecular Diagnostics, Pathology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Abhishek Bansal
- Department of Medical Oncology, Section of Molecular Diagnostics, Pathology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| |
Collapse
|
4
|
Dülger O, Öz B. Comparison of Different ROS1 Immunohistochemistry Clones and Consistency with Fluorescence In Situ Hybridization Results in Non-Small Cell Lung Carcinoma. Balkan Med J 2023; 40:344-350. [PMID: 37318131 PMCID: PMC10500138 DOI: 10.4274/balkanmedj.galenos.2023.2022-12-88] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 06/01/2023] [Indexed: 06/16/2023] Open
Abstract
Background The study of ROS1 rearrangement in non-small cell lung carcinoma (NSCLC) has gained importance as it enables personalized treatment of NSCLC with tyrosine kinase inhibitors. Therefore, it is important that the ROS1 assessment tests become more standardized. In this study, we compared the two immunohistochemistry (IHC) antibodies (D4D6 and SP384 clones) and consistency with the fluorescence in situ hybridization (FISH) results in NSCLC. Aims To investigate the effectiveness of the commonly used two IHC antibodies (SP384 and D4D6 clones) to detect ROS1 rearrangement in NSCLC. Study Design A retrospective cohort study. Methods The study included 103 samples diagnosed with NSCLC, confirmed using IHC and FISH ROS1 results (14 positives, four discordant, and 85 consecutive negatives), with sufficient tissue samples (≥ 50 tumor cells). All samples were initially tested with ROS1-IHC antibodies (D4D6 and SP384 clones); their ROS1 status was then analyzed using the FISH method. Finally, samples with discordant IHC and FISH results were confirmed using the reverse transcription polymerase chain reaction method. Results The sensitivity of SP384 and D4D6 clones of ROS1 antibody was 100% with a ≥ 1 + cut-off. When the ≥ 2 + cut-off was used, the sensitivity rate for the SP384 clone was 100%, whereas the sensitivity for the D4D6 clone was 42.86%. ROS1 FISH rearranged samples were positive for both clones, but SP384 had generally higher intensity than D4D6. The mean IHC score was + 2 for SP384 and + 1.17 for D4D6. SP384 mostly tended to have a higher IHC score intensity, which made the evaluation easier than D4D6. SP384 has a higher sensitivity than D4D6. However, false positives were found in both clones. There was no significant correlation between ROS1 FISH-positivity percentage with SP384 (p = 0.713, p = 0.108) and D4D6 (p = 0.26, p = -0.323) IHC staining intensity. The staining patterns of both clones were similar (homogeneity/heterogeneity). Conclusion Our findings show that the SP384 clone is more sensitive than D4D6. However, SP384 can also cause false positive results like D4D6. Knowing the variable diagnostic performance of different ROS1 antibodies before using them in clinical applications is necessary. IHC-positive results should be confirmed using FISH.
Collapse
Affiliation(s)
- Onur Dülger
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, İstanbul University, İstanbul, Turkey
- Institute of Graduate Studies in Health Sciences, İstanbul University, İstanbul, Turkey
| | - Büge Öz
- Department of Pathology, Cerrahpaşa Medical Faculty, İstanbul University-Cerrahpaşa, İstanbul, Turkey
| |
Collapse
|
5
|
Gilson P, Pouget C, Belmonte R, Fadil S, Demange J, Rouyer M, Lacour J, Betz M, Dardare J, Witz A, Merlin JL, Harlé A. Validation of the Idylla GeneFusion assay to detect fusions and MET exon-skipping in non-small cell lung cancers. Sci Rep 2023; 13:12909. [PMID: 37558711 PMCID: PMC10412571 DOI: 10.1038/s41598-023-39749-4] [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: 03/07/2023] [Accepted: 07/30/2023] [Indexed: 08/11/2023] Open
Abstract
Gene fusions and MET exon skipping drive oncogenesis in 8-9% and 3% of non-small cell lung cancers (NSCLC) respectively. Their detection are essential for the management of patients since they confer sensitivity to specific targeted therapies with significant clinical benefit over conventional chemotherapy. Immunohistochemistry (IHC) and fluorescent in situ hybridization (FISH) account for historical reference techniques however molecular-based technologies (RNA-based sequencing and RT-PCR) are emerging as alternative or complementary methods. Here, we evaluated the analytical performance of the fully-automated RT-PCR Idylla GeneFusion assay compared to reference methods using 35 fixed NSCLC samples. Idylla demonstrated overall agreement, sensitivity and specificity of 100% compared to RNASeq. Interestingly, it succeeded in retrieving 10 out of 11 samples with inconclusive results due to insufficient RNA quality for sequencing. Idylla showed an overall agreement, sensitivity and specificity of 90.32%, 91.67% and 89.47% compared to IHC/FISH respectively. Using commercial standards, the limit of detection of the Idylla system for the most frequent fusions and exon skipping ranges between 5 and 10 ng RNA input. These results support that the Idylla assay is a reliable and rapid option for the detection of these alterations, however a particular attention is needed for the interpretation of the expression imbalance.
Collapse
Affiliation(s)
- Pauline Gilson
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France.
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France.
| | - Celso Pouget
- Service d'Anatomocytopathologie, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Richard Belmonte
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Smahane Fadil
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Jessica Demange
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Marie Rouyer
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Julien Lacour
- Service d'Anatomocytopathologie, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Margaux Betz
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Julie Dardare
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Andréa Witz
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Jean-Louis Merlin
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Alexandre Harlé
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| |
Collapse
|
6
|
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
|
7
|
Baskaran AB, Buerki RA, Khan OH, Gondi V, Stupp R, Lukas RV, Villaflor VM. Building Team Medicine in the Management of CNS Metastases. J Clin Med 2023; 12:3901. [PMID: 37373596 DOI: 10.3390/jcm12123901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/30/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
CNS metastases are often terminal for cancer patients and occur at an approximately 10-fold higher rate than primary CNS tumors. The incidence of these tumors is approximately 70,000-400,000 cases annually in the US. Advances that have occurred over the past two decades have led to more personalized treatment approaches. Newer surgical and radiation techniques, as well as targeted and immune therapies, have enanled patient to live longer, thus increasing the risk for the development of CNS, brain, and leptomeningeal metastases (BM and LM). Patients who develop CNS metastases have often been heavily treated, and options for future treatment could best be addressed by multidisciplinary teams. Studies have indicated that patients with brain metastases have improved survival outcomes when cared for in high-volume academic institutions using multidisciplinary teams. This manuscript discusses a multidisciplinary approach for both parenchymal brain metastases as well as leptomeningeal metastases implemented in three academic institutions. Additionally, with the increasing development of healthcare systems, we discuss optimizing the management of CNS metastases across healthcare systems and integrating basic and translational science into our clinical care to further improve outcomes. This paper summarizes the existing therapeutic approaches to the treatment of BM and LM and discusses novel and emerging approaches to optimizing access to neuro-oncologic care while simultaneously integrating multidisciplinary teams in the care of patients with BM and LM.
Collapse
Affiliation(s)
- Archit B Baskaran
- Department of Neurology, The University of Chicago, Chicago, IL 60637, USA
| | - Robin A Buerki
- Health System Clinician of Neurology (Neuro-Oncology), Northwestern Medicine Regional Medical Group, Warrenville, IL 60555, USA
| | - Osaama H Khan
- Surgical Neuro-Oncology, Northwestern Medicine Central DuPage Hospital, Winfield, IL 60190, USA
| | - Vinai Gondi
- Department of Radiation Oncology, Nothwestern Medicine West Region, Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Warrenville, IL 60555, USA
| | - Roger Stupp
- Neuro-Oncology Division, Neurological Surgery, Medicine (Hematology and Oncology), Neurology, Department of Neurology, Lou & Jean Malnati Brain Tumor Institute Northwestern University, Chicago, IL 60611, USA
| | - Rimas V Lukas
- Neuro-Oncology Division, Department of Neurology, Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL 60611, USA
| | - Victoria M Villaflor
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
8
|
Englmeier F, Bleckmann A, Brückl W, Griesinger F, Fleitz A, Nagels K. Clinical benefit and cost-effectiveness analysis of liquid biopsy application in patients with advanced non-small cell lung cancer (NSCLC): a modelling approach. J Cancer Res Clin Oncol 2023; 149:1495-1511. [PMID: 35532791 PMCID: PMC10020305 DOI: 10.1007/s00432-022-04034-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/19/2022] [Indexed: 12/25/2022]
Abstract
PURPOSE Targeted therapies are effective therapeutic approaches in advanced stages of NSCLC and require precise molecular profiling to identify oncogenic drivers. Differential diagnosis on a molecular level contributes to clinical decision making. Liquid biopsy (LB) use has demonstrated its potential to serve as an alternative to tissue biopsy (TB) particularly in cases where tissue sampling is not feasible or insufficient. We aimed at evaluating the cost-effectiveness of ctDNA-based LB use (molecular multigene testing) according to German care guidelines for metastatic NSCLC. METHODS A Markov model was developed to compare the costs and clinical benefits associated with the use of LB as an add-on to TB according to the guidelines for NSCLC patients. Usual care TB served as comparator. A microsimulation model was used to simulate a cohort of non-squamous NSCLC patients stage IV. The parameters used for modelling were obtained from the literature and from the prospective German CRISP registry ("Clinical Research platform Into molecular testing, treatment, and outcome of non-Small cell lung carcinoma Patients"). For each pathway, average direct medical costs, and QALYs gained per patient were used for calculating incremental cost-effectiveness ratios (ICER). RESULTS The use of LB as an add-on was costlier (€144,981 vs. €144,587) but more effective measured in QALYs (1.20 vs. 1.19) for the care pathway of NSCLC patients (ICER €53,909/QALY). Cost-effectiveness was shown for EGFR-mutated patients (ICER €-13,247/QALY). CONCLUSION Including LB as an add-on into the care pathway of advanced NSCLC has positive clinical effects in terms of QALYs accompanied by a moderate cost-effectiveness.
Collapse
Affiliation(s)
- Fabienne Englmeier
- Chair of Healthcare Management and Health Services Research, University of Bayreuth, Parsifalstraße 25, 95445, Bayreuth, Germany
| | - Annalen Bleckmann
- Medical Clinic A, Haematology, Haemostaseology, Oncology and Pulmonology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
- Department of Haematology and Medical Oncology, University of Medicine Goettingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Wolfgang Brückl
- Department of Respiratory Medicine, Allergology and Sleep Medicine, Nuremberg Lung Cancer Center, General Hospital Nuremberg, Paracelsus Medical University, Prof.-Ernst-Nathan-Straße 1, 90419, Nuremberg, Germany
| | - Frank Griesinger
- Pius-Hospital Oldenburg, University Clinic Internal Medicine, Georgstraße 12, 26121, Oldenburg, Germany
| | - Annette Fleitz
- Clinical Epidemiology and Health Economics, iOMEDICO, Ellen-Gottlieb-Straße 19, 79108, Freiburg, Germany
| | - Klaus Nagels
- Chair of Healthcare Management and Health Services Research, University of Bayreuth, Parsifalstraße 25, 95445, Bayreuth, Germany.
| |
Collapse
|
9
|
Lai M, Li S, Li H, Hu Q, Li J, Zhou J, Ai R, Zhen J, Zhou Z, Wang L, Zhang Y, Hu W, Yuan L, Ma X, Zhang X, Song C, Li Z, Cai L. Lorlatinib for
ALK
‐fused, infant‐type hemispheric glioma with lung metastasis: a case report. Ann Clin Transl Neurol 2023; 10:836-841. [PMID: 37000961 DOI: 10.1002/acn3.51766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/24/2023] [Accepted: 03/14/2023] [Indexed: 04/03/2023] Open
Abstract
Infant-type hemispheric glioma, a new subtype of pediatric high-grade glioma, arises in the cerebral hemispheres. Despite better survival outcomes, the treatment of infant-type hemispheric glioma is still facing challenges. Here, we reported a case of QKI-ALK fusion, infant-type hemispheric glioma with lung metastasis who achieved a complete clinical response after lorlatinib treatment. This typical case demonstrated the importance of appropriate molecularly targeted treatments in ALK-fused tumors, and lorlatinib may serve as an effective complement to conventional chemotherapy and radiotherapy in primary glioma harboring ALK fusions and its metastasis.
Collapse
|
10
|
Krämer AS, Adeberg S, Kronsteiner D, König L, Schunn F, Bozorgmehr F, Christopoulos P, Eichkorn T, Schiele A, Hahnemann L, Rieken S, Debus J, Shafie RAE. Upfront and Repeated Stereotactic Radiosurgery in Patients With Brain Metastases From NSCLC. Clin Lung Cancer 2023; 24:269-277. [PMID: 36803615 DOI: 10.1016/j.cllc.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Approximately 40% of non-small-cell lung cancer (NSCLC) patients develop brain metastases (BM). Stereotactic radiosurgery (SRS) instead of whole-brain radiotherapy (WBRT) is increasingly administered as an upfront treatment to patients with a limited number of BM. We present outcomes and validation of prognostic scores for these patients treated with upfront SRS. METHODS We retrospectively analyzed 199 patients with a total of 268 SRS courses for 539 brain metastases. Median patient age was 63 years. For larger BM, dose reduction to 18 Gy or hypofractionated SRS in 6 fractions was applied. We analyzed the BMV-, the RPA-, the GPA- and the lung-mol GPA score. Cox proportional hazards models with univariate and multivariate analyses were fitted for overall survival (OS) and intracranial progression-free survival (icPFS). RESULTS Sixty-four patients died, 7 of them of neurological causes. Thirty eight patients (19,3%) required a salvage WBRT. Median OS was 38, 8 months (IQR: 6-NA). In univariate analysis as well as multivariate analysis, the Karnofsky performance scale index (KPI) ≥90% (P = 0, 012 and P = 0, 041) remained as independent prognostic factor for longer OS. All 4 prognostic scoring indices could be validated for OS assessment (BMV P = 0, 007; RPA P = 0, 026; GPA P = 0, 003; lung-mol GPA P = 0, 05). CONCLUSION In this large cohort of NSCLC patients with BM treated with upfront and repeated SRS, OS was markedly favourable, in comparison to literature. Upfront SRS is an effective treatment approach in those patients and can decidedly reduce the impact of BM on overall prognosis. Furthermore, the analysed scores are useful prognostic tools for OS prediction.
Collapse
Affiliation(s)
- Anna S Krämer
- Universitätsklinikum Heidelberg, Abteilung RadioOnkologie und Strahlentherapie, Heidelberg, Baden-Württemberg, Germany.
| | - Sebastian Adeberg
- Universitätsklinikum Heidelberg, Abteilung RadioOnkologie und Strahlentherapie, Heidelberg, Baden-Württemberg, Germany
| | - Dorothea Kronsteiner
- Institut für Medizinische Biometrie (IMB), Universitätsklinikum Heidelberg, Heidelberg, Baden-Württemberg, Germany
| | - Laila König
- Universitätsklinikum Heidelberg, Abteilung RadioOnkologie und Strahlentherapie, Heidelberg, Baden-Württemberg, Germany
| | - Fabian Schunn
- Universitätsklinikum Heidelberg, Abteilung RadioOnkologie und Strahlentherapie, Heidelberg, Baden-Württemberg, Germany
| | | | | | - Tanja Eichkorn
- Universitätsklinikum Heidelberg, Abteilung RadioOnkologie und Strahlentherapie, Heidelberg, Baden-Württemberg, Germany
| | - Annabella Schiele
- Universitätsklinikum Heidelberg, Abteilung RadioOnkologie und Strahlentherapie, Heidelberg, Baden-Württemberg, Germany
| | - Laura Hahnemann
- Universitätsklinikum Heidelberg, Abteilung RadioOnkologie und Strahlentherapie, Heidelberg, Baden-Württemberg, Germany
| | - Stefan Rieken
- Universitätsmedizin Göttingen, Klinik für Strahlentherapie und Radioonkologie, Göttingen, Lower Saxony, Germany
| | - Jürgen Debus
- Universitätsklinikum Heidelberg, Abteilung RadioOnkologie und Strahlentherapie, Heidelberg, Baden-Württemberg, Germany
| | - Rami A El Shafie
- Universitätsklinikum Heidelberg, Abteilung RadioOnkologie und Strahlentherapie, Heidelberg, Baden-Württemberg, Germany; Universitätsmedizin Göttingen, Klinik für Strahlentherapie und Radioonkologie, Göttingen, Lower Saxony, Germany
| |
Collapse
|
11
|
Shen Z, Qiu B, Li L, Yang B, Li G. Targeted therapy of RET fusion-positive non-small cell lung cancer. Front Oncol 2022; 12:1033484. [PMID: 36582799 PMCID: PMC9793070 DOI: 10.3389/fonc.2022.1033484] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022] Open
Abstract
Lung cancer has very high morbidity and mortality worldwide, and the prognosis is not optimistic. Previous treatments for non-small cell lung cancer (NSCLC) have limited efficacy, and targeted drugs for some gene mutations have been used in NSCLC with considerable efficacy. The RET proto-oncogene is located on the long arm of chromosome 10 with a length of 60,000 bp, and the expression of RET gene affects cell survival, proliferation, growth and differentiation. This review will describe the basic characteristics and common fusion methods of RET genes; analyze the advantages and disadvantages of different RET fusion detection methods; summarize and discuss the recent application of non-selective and selective RET fusion-positive inhibitors, such as Vandetanib, Selpercatinib, Pralsetinib and Alectinib; discuss the mechanism and coping strategies of resistance to RET fusion-positive inhibitors.
Collapse
Affiliation(s)
- Zixiong Shen
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Binxu Qiu
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Lin Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Bo Yang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China,*Correspondence: Bo Yang, ;; Guanghu Li,
| | - Guanghu Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China,*Correspondence: Bo Yang, ;; Guanghu Li,
| |
Collapse
|
12
|
Lim KHT, Kong HL, Chang KTE, Tan DSW, Tan IBH, Mohamad F, Soh SY, Pang BN, Soo RA, Choo SP, Hsieh W, Aung L. Recommended testing algorithms for NTRK gene fusions in pediatric and selected adult cancers: Consensus of a Singapore Task Force. Asia Pac J Clin Oncol 2022; 18:394-403. [PMID: 34806337 PMCID: PMC9541932 DOI: 10.1111/ajco.13727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/27/2021] [Indexed: 01/09/2023]
Abstract
The occurrence of neurotrophic tyrosine receptor kinase (NTRK) gene fusions in a wide range of tumor types presents an attractive opportunity for using a tropomyosin receptor kinase (TRK) inhibitor as cancer therapy. Recent clinical studies have demonstrated highly efficacious outcomes associated with the use of TRK inhibitors, such as larotrectinib and entrectinib in NTRK fusion-bearing cancers, in both adult and pediatric populations. While NTRK gene fusions are commonly found in some uncommon adult and pediatric malignancies, they are also found, albeit rarely, in a wide range of more common malignancies. The potential value of testing for NTRK gene fusions in practically all advanced malignancies is underpinned by the remarkable therapeutic outcomes that TRK inhibitors offer. This requirement presents practical and financial challenges in real-world oncological practice. Furthermore, different testing platforms exist to detect NTRK gene fusions, each with its advantages and disadvantages. It is, therefore, imperative to develop strategies for NTRK gene fusion testing in an attempt to optimize the use of limited tissue specimen and financial resources, and to minimize the turnaround time. A multidisciplinary task force of Singapore medical experts in both public and private sectors was convened in late 2020 to propose testing algorithms for adult colorectal tumors, sarcomas, non-small cell lung cancer, and pediatric cancers, with particular adaptation to the Singapore oncological practice. The recommendations presented here highlight the heterogeneity of NTRK-fusion positive cancers, and emphasize the need to customize the testing methods to each tumor type to optimize the workflow.
Collapse
Affiliation(s)
- Kiat Hon Tony Lim
- Division of PathologyDepartment of Anatomical PathologySingapore General HospitalSingapore
| | - Hwai Loong Kong
- ICON Cancer CentreMount Elizabeth Medical Centre OrchardSingapore
| | - Kenneth Tou En Chang
- Department of Pathology and Laboratory MedicineKK Women's and Children's HospitalSingapore
| | | | - Iain Bee Huat Tan
- Division of Medical OncologyNational Cancer Centre SingaporeSingapore
| | - Farid Mohamad
- Division of Medical OncologyNational Cancer Centre SingaporeSingapore
| | - Shui Yen Soh
- Haematology‐Oncology ServiceDepartment of Paediatrics SubspecialtiesKK Women's and Children's HospitalSingapore
| | | | - Ross Andrew Soo
- Department of Haematology‐OncologyNational University Cancer InstituteNational University Health SystemSingapore
| | | | | | - LeLe Aung
- Paediatric Haematology‐OncologyICON Cancer CentreSingapore
| |
Collapse
|
13
|
Depoilly T, Garinet S, van Kempen LC, Schuuring E, Clavé S, Bellosillo B, Ercolani C, Buglioni S, Siemanowski J, Merkelbach-Bruse S, Tischler V, Demes MC, Paridaens H, Sibille C, de Montpreville VT, Rouleau E, Bartczak A, Pasieka-Lis M, Wei Teo RY, Chuah KL, Barbosa M, Quintana C, Biscuola M, Delgado-Garcia M, Vacirca D, Rappa A, Cashmore M, Smith M, Jasionowicz P, Meeney A, Desmeules P, Terris B, Mansuet-Lupo A. Multicenter Evaluation of the Idylla GeneFusion in Non-Small-Cell Lung Cancer. J Mol Diagn 2022; 24:1021-1030. [PMID: 35718095 DOI: 10.1016/j.jmoldx.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/20/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022] Open
Abstract
Targeted therapy in lung cancer requires the assessment of multiple oncogenic driver alterations, including fusion genes. This retrospective study evaluated the Idylla GeneFusion prototype, an automated and ease-of-use (<2 minutes) test, with a short turnaround time (3 hours) to detect fusions involving ALK, ROS1, RET, and NTRK1/2/3 genes and MET exon 14 skipping. This multicenter study (18 centers) included 313 tissue samples from lung cancer patients with 97 ALK, 44 ROS1, 20 RET, and 5 NTRKs fusions, 32 MET exon 14 skipping, and 115 wild-type samples, previously identified with reference methods (RNA-based next generation sequencing/fluorescence in situ hybridization/quantitative PCR). Valid results were obtained for 306 cases (98%), overall concordance between Idylla and the reference methods was 89% (273/306); overall sensitivity and specificity were 85% (165/193) and 96% (108/113), respectively. Discordances were observed in 28 samples, where Idylla did not detect the alteration identified by the reference methods; and 5 samples where Idylla identified an alteration not detected by the reference methods. All of the ALK-, ROS1-, and RET-specific fusions and MET exon 14 skipping identified by Idylla GeneFusion were confirmed by reference method. To conclude, Idylla GeneFusion is a clinically valuable test that does not require a specific infrastructure, allowing a rapid result. The absence of alteration or the detection of expression imbalance only requires additional testing by orthogonal methods.
Collapse
Affiliation(s)
- Thomas Depoilly
- Department of Pathology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Simon Garinet
- Department of Biochemistry, Unit of Pharmacogenetics and Molecular Oncology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Léon C van Kempen
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ed Schuuring
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sergi Clavé
- Department of Pathology, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Beatriz Bellosillo
- Department of Pathology, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Cristiana Ercolani
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Simonetta Buglioni
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Janna Siemanowski
- Faculty of Medicine and University Hospital Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Faculty of Medicine and University Hospital Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Verena Tischler
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | | | | | | | | | - Etienne Rouleau
- Service de Génétique des Tumeurs, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Artur Bartczak
- Department of Pathomorphology, Public Specialist Hospital of Lung Diseases in Zakopane, Zakopane, Poland
| | - Monika Pasieka-Lis
- Department of Pathomorphology, Public Specialist Hospital of Lung Diseases in Zakopane, Zakopane, Poland
| | - Ryan Yee Wei Teo
- Department of Pathology, Tan Tock Seng Hospital, Novena, Republic of Singapore
| | - Khoon Leong Chuah
- Department of Pathology, Tan Tock Seng Hospital, Novena, Republic of Singapore
| | - Marta Barbosa
- Serviço de Anatomia Patológica, Hospital do Espírito Santo de Évora, Évora, Portugal
| | - Carlos Quintana
- Serviço de Anatomia Patológica, Hospital do Espírito Santo de Évora, Évora, Portugal
| | - Michele Biscuola
- Department of Pathology, Molecular Pathology Laboratory, Hospital Universitario Virgen del Rocío-IBIS, Seville, Spain
| | - Mercedes Delgado-Garcia
- Department of Pathology, Molecular Pathology Laboratory, Hospital Universitario Virgen del Rocío-IBIS, Seville, Spain
| | - Davide Vacirca
- Division of Pathology and Laboratory Medicine, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Alessandra Rappa
- Division of Pathology and Laboratory Medicine, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Matthew Cashmore
- Black Country Pathology Services, New Cross Hospital, The Royal Wolverhampton NHS Trust, Wolverhampton, United Kingdom
| | - Matthew Smith
- Pathology Department, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Piotr Jasionowicz
- Pathology Department, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Adam Meeney
- Sheffield Teaching Hospitals NHS Foundation Trust, Pathology Laboratory, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Patrice Desmeules
- Departement d'Anatomopathologie et Cytologie, Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Québec, Québec, Canada
| | - Benoit Terris
- Department of Pathology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Audrey Mansuet-Lupo
- Department of Pathology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France.
| |
Collapse
|
14
|
Luan Q, Becker JH, Macaraniag C, Massad MG, Zhou J, Shimamura T, Papautsky I. Non-small cell lung carcinoma spheroid models in agarose microwells for drug response studies. LAB ON A CHIP 2022; 22:2364-2375. [PMID: 35551303 DOI: 10.1039/d2lc00244b] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
There is a growing interest in developing personalized treatment strategies for each cancer patient, especially those with non-small cell lung carcinoma (NSCLC) which annually accounts for the majority of cancer related deaths in the US. Yet identifying the optimal NSCLC treatment strategy for each cancer patient is critical due to a multitude of mutations, some of which develop following initial therapy and can result in drug resistance. A key difficulty in developing personalized therapies in NSCLC is the lack of clinically relevant assay systems that are suitable to evaluate drug sensitivity using a minuscule amount of patient-derived material available following biopsies. Herein we leverage 3D printing to demonstrate a platform based on miniature microwells in agarose to culture cancer cell spheroids. The agarose wells were shaped by 3D printing molds with 1000 microwells with a U-shaped bottom. Three NSCLC cell lines (HCC4006, H1975 and A549) were used to demonstrate size uniformity, spheroid viability, biomarker expressions and drug response in 3D agarose microwells. Results show that our approach yielded spheroids of uniform size (coefficient of variation <22%) and high viability (>83% after 1 week-culture). Studies using epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKIs) drugs gefitinib and osimertinib showed clinically relevant responses. Based on the physical features, cell phenotypes, and responses to therapy of our spheroid models, we conclude that our platform is suitable for in vitro culture and drug evaluation, especially in cases when tumor sample is limited.
Collapse
Affiliation(s)
- Qiyue Luan
- Department of Biomedical Engineering, University of Illinois Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
| | - Jeffrey H Becker
- Department of Surgery, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Celine Macaraniag
- Department of Biomedical Engineering, University of Illinois Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
| | - Malek G Massad
- Department of Surgery, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Jian Zhou
- Department of Biomedical Engineering, University of Illinois Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Takeshi Shimamura
- Department of Surgery, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
15
|
Raman R, Villefranc JA, Ullmann TM, Thiesmeyer J, Anelli V, Yao J, Hurley JR, Pauli C, Bareja R, Wha Eng K, Dorsaint P, Wilkes DC, Beg S, Kudman S, Shaw R, Churchill M, Ahmed A, Keefer L, Misner I, Nichol D, Gumpeni N, Scognamiglio T, Rubin MA, Grandori C, Solomon JP, Song W, Mosquera JM, Dephoure N, Sboner A, Elemento O, Houvras Y. Inhibition of FGF receptor blocks adaptive resistance to RET inhibition in CCDC6-RET-rearranged thyroid cancer. J Exp Med 2022; 219:e20210390. [PMID: 35510953 PMCID: PMC9082625 DOI: 10.1084/jem.20210390] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 11/23/2021] [Accepted: 03/18/2022] [Indexed: 11/18/2022] Open
Abstract
Genetic alterations in RET lead to activation of ERK and AKT signaling and are associated with hereditary and sporadic thyroid cancer and lung cancer. Highly selective RET inhibitors have recently entered clinical use after demonstrating efficacy in treating patients with diverse tumor types harboring RET gene rearrangements or activating mutations. In order to understand resistance mechanisms arising after treatment with RET inhibitors, we performed a comprehensive molecular and genomic analysis of a patient with RET-rearranged thyroid cancer. Using a combination of drug screening and proteomic and biochemical profiling, we identified an adaptive resistance to RET inhibitors that reactivates ERK signaling within hours of drug exposure. We found that activation of FGFR signaling is a mechanism of adaptive resistance to RET inhibitors that activates ERK signaling. Combined inhibition of FGFR and RET prevented the development of adaptive resistance to RET inhibitors, reduced cell viability, and decreased tumor growth in cellular and animal models of CCDC6-RET-rearranged thyroid cancer.
Collapse
Affiliation(s)
- Renuka Raman
- Department of Surgery, Weill Cornell Medical College, New York, NY
| | | | | | | | - Viviana Anelli
- Department of Surgery, Weill Cornell Medical College, New York, NY
| | - Jun Yao
- Department of Surgery, Weill Cornell Medical College, New York, NY
| | - James R. Hurley
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Chantal Pauli
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Rohan Bareja
- The Caryl and Israel Englander Institute for Precision Medicine and the Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY
| | - Kenneth Wha Eng
- The Caryl and Israel Englander Institute for Precision Medicine and the Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY
| | - Princesca Dorsaint
- The Caryl and Israel Englander Institute for Precision Medicine and the Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY
| | - David C. Wilkes
- The Caryl and Israel Englander Institute for Precision Medicine and the Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY
| | - Shaham Beg
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Sarah Kudman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Reid Shaw
- SEngine Precision Medicine, Seattle, WA
| | | | - Adnan Ahmed
- Department of Biochemistry, Weill Cornell Medical College, New York, NY
| | | | - Ian Misner
- Personal Genome Diagnostics, Inc., Baltimore, MD
| | - Donna Nichol
- Personal Genome Diagnostics, Inc., Baltimore, MD
| | - Naveen Gumpeni
- Department of Radiology, Weill Cornell Medical College, New York, NY
| | - Theresa Scognamiglio
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Mark A. Rubin
- Bern Center for Precision Medicine, University of Bern, Bern, Switzerland
| | | | - James Patrick Solomon
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Wei Song
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Noah Dephoure
- Department of Biochemistry, Weill Cornell Medical College, New York, NY
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medical College, New York, NY
| | - Andrea Sboner
- The Caryl and Israel Englander Institute for Precision Medicine and the Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medical College, New York, NY
| | - Olivier Elemento
- The Caryl and Israel Englander Institute for Precision Medicine and the Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medical College, New York, NY
| | - Yariv Houvras
- Department of Surgery, Weill Cornell Medical College, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medical College, New York, NY
| |
Collapse
|
16
|
Randon G, Maddalena G, Germani MM, Pircher CC, Manca P, Bergamo F, Giordano M, Sposetti C, Montagna A, Vetere G, Zambelli L, Rasola C, Boccaccino A, Pagani F, Ambrosini M, Massafra M, Fontanini G, Milione M, Fassan M, Cremolini C, Lonardi S, Pietrantonio F. Negative Ultraselection of Patients With RAS/ BRAF Wild-Type, Microsatellite-Stable Metastatic Colorectal Cancer Receiving Anti-EGFR-Based Therapy. JCO Precis Oncol 2022; 6:e2200037. [PMID: 35544729 PMCID: PMC9200389 DOI: 10.1200/po.22.00037] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Several uncommon genomic alterations beyond RAS and BRAFV600E mutations drive primary resistance to anti–epidermal growth factor receptors (EGFRs) in metastatic colorectal cancer (mCRC). Our PRESSING panel (including PIK3CA exon 20/AKT1/PTEN mutations, ERBB2/MET amplifications, gene fusions, and microsatellite instability-high status) represented a paradigm of negative hyperselection with more precise tailoring of EGFR blockade. However, a modest proportion of hyperselected mCRC has intrinsic resistance potentially driven by even rarer genomic alterations. Beyond RAS/BRAF: rarer alterations drive negative ultraselection for anti-EGFR therapy in mCRC
Collapse
Affiliation(s)
- Giovanni Randon
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Giulia Maddalena
- Oncology Unit 1, Veneto Institute of Oncology-IRCCS, Padova, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padua, Padova, Italy
| | - Marco Maria Germani
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Chiara Carlotta Pircher
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Paolo Manca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | | | - Mirella Giordano
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Caterina Sposetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Aldo Montagna
- Oncology Unit 1, Veneto Institute of Oncology-IRCCS, Padova, Italy
| | - Guglielmo Vetere
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Luca Zambelli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Cosimo Rasola
- Oncology Unit 1, Veneto Institute of Oncology-IRCCS, Padova, Italy
| | - Alessandra Boccaccino
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Filippo Pagani
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Margherita Ambrosini
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Marco Massafra
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Gabriella Fontanini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Massimo Milione
- First Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padova, Italy.,Veneto Institute of Oncology-IRCCS, Padova, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Sara Lonardi
- Oncology Unit 3, Veneto Institute of Oncology-IRCCS, Padova, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| |
Collapse
|
17
|
Li W, Wan R, Guo L, Chang G, Jiang D, Meng L, Ying J. Reliability analysis of exonic-breakpoint fusions identified by DNA sequencing for predicting the efficacy of targeted therapy in non-small cell lung cancer. BMC Med 2022; 20:160. [PMID: 35534835 PMCID: PMC9087946 DOI: 10.1186/s12916-022-02362-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/04/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Diverse genomic breakpoints of fusions that localize to intronic, exonic, or intergenic regions have been identified by DNA next-generation sequencing (NGS), but the role of exonic breakpoints remains elusive. We investigated whether exonic-breakpoint fusions could predict matched targeted therapy efficacy in non-small cell lung cancer (NSCLC). METHODS NSCLC samples were analyzed by DNA NGS, RNA NGS, immunohistochemistry (IHC), and fluorescence in situ hybridization. RESULTS Using DNA NGS, kinase fusions were identified in 685 of 7148 (9.6%) NSCLCs, with 74 harboring exonic-breakpoint fusions, mostly anaplastic lymphoma kinase (ALK) fusions. RNA NGS and IHC revealed that 11 of 55 (20%) exonic-breakpoint fusions generated no aberrant transcript/protein, possibly due to open reading frame disruption or different gene transcriptional orientations. Four cases of genomic-positive but RNA/protein-negative fusions were treated with matched targeted therapy, but progressive disease developed within 2 months. Nevertheless, 44 of 55 (80%) exonic-breakpoint fusions produced chimeric transcripts/proteins, possibly owing to various alternative splicing patterns, including exon skipping, alternative splice site selection, and intron retention. Most of these genomic- and RNA/protein-positive fusion cases showed a clinical response to matched targeted therapy. Particularly, there were no differences in objective response rate (P = 0.714) or median progression-free survival (P = 0.500) between intronic-breakpoint (n = 56) and exonic-breakpoint ALK fusion subtypes (n = 11) among ALK RNA/protein-validated patients who received first-line crizotinib. CONCLUSIONS Exonic-breakpoint fusions may generate in-frame fusion transcripts/proteins or not, and thus are unreliable for predicting the efficacy of targeted therapy, which highlights the necessity of implementing RNA or protein assays for functional validation in exonic-breakpoint fusion cases.
Collapse
Affiliation(s)
- Weihua Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuan Nanli, Beijing, 100021, China.
| | - Rui Wan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuan Nanli, Beijing, 100021, China
| | - Geyun Chang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong Jiang
- Beijing Novogene Bioinformatics Technology Co., Ltd., Beijing, China
| | - Lin Meng
- Beijing Novogene Bioinformatics Technology Co., Ltd., Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuan Nanli, Beijing, 100021, China.
| |
Collapse
|
18
|
Xu X, Wang H, Yu Z, Chen X. A novel RET fusion in non-small cell lung cancer identified by next-generation sequencing: a case report. J Cancer Res Clin Oncol 2022; 148:1825-1827. [PMID: 35364706 DOI: 10.1007/s00432-022-03969-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/22/2022] [Indexed: 12/01/2022]
Abstract
REarranged during Transfection (RET) gene fusion is one of the common oncogenic variants in non-small cell lung cancers (NSCLCs). However, few RET fusion-positive cases have partner intergenic-breakpoint fusions, in which the partner breakpoint localizes to intergenic regions. Here, we report a 40-year-old Chinese female non-smoker diagnosed with minimally invasive lung adenocarcinomas (pT1bN0M0, stage IA). Targeted next-generation sequencing revealed a rare form of RET fusion in the cancerous tissue, in which an intergenic fragment upstream multiple inositol-polyphosphate phosphatase 1 gene was fused with the tyrosine kinase domain in RET. The result was validated by fluorescence in situ hybridization. To our knowledge, this novel form of RET fusion in NSCLC is reported for the first time, which expands the alteration spectrum and paves the way for the future development of specific targeted therapies.
Collapse
Affiliation(s)
- Xiaoyi Xu
- Department of Thoracic Surgery, Jinhua Hospital of Zhejiang University, Jinhua, 330700, China
| | - Haoyi Wang
- Hangzhou D.A. Medical Laboratory, Hangzhou, 310030, China
| | - Zhaonan Yu
- Hangzhou D.A. Medical Laboratory, Hangzhou, 310030, China.
| | - Xianguo Chen
- Department of Thoracic Surgery, Jinhua Hospital of Zhejiang University, Jinhua, 330700, China.
| |
Collapse
|
19
|
Dong OM, Poonnen PJ, Winski D, Reed SD, Vashistha V, Bates J, Kelley MJ, Voora D. Cost-Effectiveness of Tumor Genomic Profiling to Guide First-Line Targeted Therapy Selection in Patients With Metastatic Lung Adenocarcinoma. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2022; 25:582-594. [PMID: 35365302 PMCID: PMC8976872 DOI: 10.1016/j.jval.2021.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/18/2021] [Accepted: 09/30/2021] [Indexed: 06/04/2023]
Abstract
OBJECTIVES A cost-effectiveness analysis comparing comprehensive genomic profiling (CGP) of 10 oncogenes, targeted gene panel testing (TGPT) of 4 oncogenes, and no tumor profiling over the lifetime for patients with metastatic lung adenocarcinoma from the Centers for Medicare and Medicaid Services' perspective was conducted. METHODS A decision analytic model used 10 000 hypothetical Medicare beneficiaries with metastatic lung adenocarcinoma to simulate outcomes associated with CGP (ALK, BRAF, EGFR, ERBB2, MET, NTRK1, NTRK2, NTRK3, RET, ROS1), TGPT (ALK, BRAF, EGFR, ROS1), and no tumor profiling (no genes tested). First-line targeted cancer-directed therapies were assigned if actionable gene variants were detected; otherwise, nontargeted cancer-directed therapies were assigned. Model inputs were derived from randomized trials (progression-free survival, adverse events), the Veterans Health Administration and Medicare (drug costs), published studies (nondrug cancer-related management costs, health state utilities), and published databases (actionable variant prevalences). Costs (2019 US$) and quality-adjusted life-years (QALYs) were discounted at 3% per year. Probabilistic sensitivity analyses used 1000 Monte Carlo simulations. RESULTS No tumor profiling was the least costly/person ($122 613 vs $184 063 for TGPT and $188 425 for CGP) and yielded the least QALYs/person (0.53 vs 0.73 for TGPT and 0.74 for CGP). The costs per QALY gained and corresponding 95% confidence interval were $310 735 ($278 323-$347 952) for TGPT vs no tumor profiling and $445 545 ($322 297-$572 084) for CGP vs TGPT. All probabilistic sensitivity analysis simulations for both comparisons surpassed the willingness-to-pay threshold ($150 000 per QALY gained). CONCLUSION Compared with no tumor profiling in patients with metastatic lung adenocarcinoma, tumor profiling (TGPT, CGP) improves quality-adjusted survival but is not cost-effective.
Collapse
Affiliation(s)
- Olivia M Dong
- Duke Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Department of Veterans Affairs, Durham VA Medical Center, Durham, NC, USA
| | - Pradeep J Poonnen
- Department of Veterans Affairs, Durham VA Medical Center, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | - David Winski
- Department of Veterans Affairs, VA Boston Healthcare System, Boston, MA, USA
| | - Shelby D Reed
- Department of Veterans Affairs, Durham VA Medical Center, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA; Duke Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Vishal Vashistha
- Section of Hematology/Oncology, Raymond G. Murphy New Mexico Veterans Affairs Medical Center, Albuquerque, NM, USA
| | - Jill Bates
- Department of Veterans Affairs, Durham VA Medical Center, Durham, NC, USA; Division of Practice Advancement and Clinical Education, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Veterans Affairs, National Oncology Program, Durham, NC, USA
| | - Michael J Kelley
- Department of Veterans Affairs, Durham VA Medical Center, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA; Department of Veterans Affairs, National Oncology Program, Durham, NC, USA
| | - Deepak Voora
- Duke Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Department of Veterans Affairs, Durham VA Medical Center, Durham, NC, USA; Department of Veterans Affairs, National Oncology Program, Durham, NC, USA.
| |
Collapse
|
20
|
Rocco D, Della Gravara L, Maione P, Palazzolo G, Gridelli C. Identification of drug combinations for lung cancer patients whose tumors are unresponsive to targeted therapy: clinical bases and future directions. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2022. [DOI: 10.1080/23808993.2022.2050369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Danilo Rocco
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, Naples, Italy
| | - Luigi Della Gravara
- Department of Experimental Medicine, Università degli studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Paolo Maione
- Division of Medical Oncology, “S.g. Moscati” Hospital, Avellino, Italy
| | | | - Cesare Gridelli
- Division of Medical Oncology, “S.g. Moscati” Hospital, Avellino, Italy
| |
Collapse
|
21
|
Zhang C, Mei W, Zeng C. Oncogenic Neuregulin 1 gene (NRG1) fusions in cancer: A potential new therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2022; 1877:188707. [PMID: 35247506 DOI: 10.1016/j.bbcan.2022.188707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/27/2022] [Accepted: 02/27/2022] [Indexed: 10/19/2022]
Abstract
It is widely established that chromosomal rearrangements induce oncogenesis in solid tumors. However, discovering chromosomal rearrangements that are targetable and actionable remains a difficulty. Targeting gene fusion or chromosomal rearrangement seems to be a powerful strategy to address malignancies characterized by gene rearrangement. Oncogenic NRG1 fusions are relatively rare drivers that infrequently occur across most tumor types. NRG1 fusions exhibit unique biological properties and are difficult to identify owing to their large intronic regions. NRG1 fusions can be detected using a variety of techniques, including fluorescence in situ hybridization, immunohistochemistry, or next-generation sequencing (NGS), with NGS-based RNA sequencing being the most sensitive. Previous studies have shown that NRG1 fusion protein induces tumorigenesis, and numerous therapies targeting the ErbB signaling pathway, such as ErbB kinase inhibitors and monoclonal antibodies, have initially demonstrated encouraging anticancer efficacy in malignant tumors carrying NRG1 fusions. In this review, we present the characteristics and prevalence of NRG1 fusions in solid tumors. Additionally, we discuss the laboratory approaches for diagnosing NRG1 gene fusions. More importantly, we outline promising strategies for treating malignancies with NRG1 fusion.
Collapse
Affiliation(s)
- Congwang Zhang
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen 518110, China
| | - Wuxuan Mei
- Clinical Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen 518110, China.
| |
Collapse
|
22
|
Li W, Ye L, Huang Y, Zhou F, Wu C, Wu F, He Y, Li X, Wang H, Xiong A, Gao G, Wang L, Su C, Ren S, Chen X, Zhou C. Characteristics of Notch signaling pathway and its correlation with immune microenvironment in SCLC. Lung Cancer 2022; 167:25-33. [DOI: 10.1016/j.lungcan.2022.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/10/2022] [Accepted: 03/27/2022] [Indexed: 12/24/2022]
|
23
|
Copia Sperandio R, Luiza Teixeira Tostes F, Vidal Campregher P, Ribeiro Paes V, Moura F, Schvartsman G. EGFR-RAD51 fusion in lung adenocarcinoma with systemic and intracranial response to osimertinib: a case report and review of the literature. Lung Cancer 2022; 166:94-97. [DOI: 10.1016/j.lungcan.2022.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 10/19/2022]
|
24
|
Kazdal D, Hofman V, Christopoulos P, Ilié M, Stenzinger A, Hofman P. Fusion-positive non-small cell lung carcinoma: Biological principles, clinical practice, and diagnostic implications. Genes Chromosomes Cancer 2022; 61:244-260. [PMID: 34997651 DOI: 10.1002/gcc.23022] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
Based on superior efficacy and tolerability, targeted therapy is currently preferred over chemotherapy and/or immunotherapy for actionable gene fusions that occur in late-stage non-small cell lung carcinoma (NSCLC). Consequently, current clinical practice guidelines mandate testing for ALK, ROS1, NTRK, and RET gene fusions in all patients with newly diagnosed advanced non-squamous NSCLC (NS-NSCLC). Gene fusions can be detected using different approaches, but today RNA next-generation sequencing (NGS) or combined DNA/RNA NGS is the method of choice. The discovery of other gene fusions (involving, eg, NRG1, NUT, FGFR1, FGFR2, MET, BRAF, EGFR, SMARC fusions) and their partners has increased progressively in recent years, leading to the development of new and promising therapies and mandating the development and implementation of comprehensive detection methods. The purpose of this review is to focus on recent data concerning the main gene fusions identified in NSCLC, followed by the discussion of major challenges in this domain.
Collapse
Affiliation(s)
- Daniel Kazdal
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center (TLRC) Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Heidelberg, Germany
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, FHU OncoAge, Nice, France.,Centre Antoine Lacassagne Cancer Center, Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France.,Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Petros Christopoulos
- Translational Lung Research Center (TLRC) Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Heidelberg, Germany.,Thoraxklinik and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, FHU OncoAge, Nice, France.,Centre Antoine Lacassagne Cancer Center, Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France.,Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, FHU OncoAge, Nice, France.,Centre Antoine Lacassagne Cancer Center, Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France.,Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| |
Collapse
|
25
|
Deligiorgi MV, Trafalis DT. The Intriguing Thyroid Hormones-Lung Cancer Association as Exemplification of the Thyroid Hormones-Cancer Association: Three Decades of Evolving Research. Int J Mol Sci 2021; 23:436. [PMID: 35008863 PMCID: PMC8745569 DOI: 10.3390/ijms23010436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 12/21/2022] Open
Abstract
Exemplifying the long-pursued thyroid hormones (TH)-cancer association, the TH-lung cancer association is a compelling, yet elusive, issue. The present narrative review provides background knowledge on the molecular aspects of TH actions, with focus on the contribution of TH to hallmarks of cancer. Then, it provides a comprehensive overview of data pertinent to the TH-lung cancer association garnered over the last three decades and identifies obstacles that need to be overcome to enable harnessing this association in the clinical setting. TH contribute to all hallmarks of cancer through integration of diverse actions, currently classified according to molecular background. Despite the increasingly recognized implication of TH in lung cancer, three pending queries need to be resolved to empower a tailored approach: (1) How to stratify patients with TH-sensitive lung tumors? (2) How is determined whether TH promote or inhibit lung cancer progression? (3) How to mimic the antitumor and/or abrogate the tumor-promoting TH actions in lung cancer? To address these queries, research should prioritize the elucidation of the crosstalk between TH signaling and oncogenic signaling implicated in lung cancer initiation and progression, and the development of efficient, safe, and feasible strategies leveraging this crosstalk in therapeutics.
Collapse
Affiliation(s)
- Maria V. Deligiorgi
- Department of Pharmacology—Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens, Building 16, 1st Floor, 75 Mikras Asias Str, 11527 Athens, Greece;
| | | |
Collapse
|
26
|
Park HJ, Baek I, Cheang G, Solomon JP, Song W. Comparison of RNA-Based Next-Generation Sequencing Assays for the Detection of NTRK Gene Fusions. J Mol Diagn 2021; 23:1443-1451. [PMID: 34756276 DOI: 10.1016/j.jmoldx.2021.07.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/28/2021] [Accepted: 07/16/2021] [Indexed: 12/29/2022] Open
Abstract
Recently, the US Food and Drug Administration approved several targeted therapies directed against oncogenic fusions. One of the most effective such targeted therapies is Vitrakvi (larotrectinib), highly specific oral tropomyosin receptor kinase inhibitor indicated for the treatment of patients with any solid tumor harboring a fusion involving one of the neurotrophic receptor tyrosine kinase (NTRK) genes. Although several diagnostic approaches can be used to detect these NTRK fusions, RNA-based next-generation sequencing remains one of the most sensitive methods, as it can directly detect the transcribed end product of gene fusion at the mRNA level. In this study, performance characteristics of three RNA-based next-generation sequencing assays with distinct mechanisms and chemistries were investigated: anchored multiplex PCR, amplicon-based multiplex PCR, and hybrid capture-based enrichment method. Analytical sensitivity analysis shows that the amplicon-based multiplex PCR method has the lowest limit of detection. However, both hybrid-capture and anchored multiplex PCR methods can detect NTRK fusions with uncommon or novel fusion partners, which is challenging for the amplicon-based multiplex method. As for clinical sensitivity, all three methods were highly concordant in detecting NTRK fusions in patient samples. Additionally, they all presented equivalent high-level performance in specificity, suggesting that all three platforms can detect NTRK fusions in clinical samples with similar performance characteristics.
Collapse
Affiliation(s)
- Hyeon J Park
- Department of Laboratory Medicine and Pathology, Weill Cornell Medicine, New York, New York
| | - Inji Baek
- Department of Laboratory Medicine and Pathology, Weill Cornell Medicine, New York, New York
| | - Gloria Cheang
- Department of Laboratory Medicine and Pathology, Weill Cornell Medicine, New York, New York
| | - James P Solomon
- Department of Laboratory Medicine and Pathology, Weill Cornell Medicine, New York, New York
| | - Wei Song
- Department of Laboratory Medicine and Pathology, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
27
|
Drilon A, Duruisseaux M, Han JY, Ito M, Falcon C, Yang SR, Murciano-Goroff YR, Chen H, Okada M, Molina MA, Wislez M, Brun P, Dupont C, Branden E, Rossi G, Schrock A, Ali S, Gounant V, Magne F, Blum TG, Schram AM, Monnet I, Shih JY, Sabari J, Pérol M, Zhu VW, Nagasaka M, Doebele R, Camidge DR, Arcila M, Ou SHI, Moro-Sibilot D, Rosell R, Muscarella LA, Liu SV, Cadranel J. Clinicopathologic Features and Response to Therapy of NRG1 Fusion-Driven Lung Cancers: The eNRGy1 Global Multicenter Registry. J Clin Oncol 2021; 39:2791-2802. [PMID: 34077268 PMCID: PMC8407651 DOI: 10.1200/jco.20.03307] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/03/2021] [Accepted: 04/16/2021] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Although NRG1 fusions are oncogenic drivers across multiple tumor types including lung cancers, these are difficult to study because of their rarity. The global eNRGy1 registry was thus established to characterize NRG1 fusion-positive lung cancers in the largest and most diverse series to date. METHODS From June 2018 to February 2020, a consortium of 22 centers from nine countries in Europe, Asia, and the United States contributed data from patients with pathologically confirmed NRG1 fusion-positive lung cancers. Profiling included DNA-based and/or RNA-based next-generation sequencing and fluorescence in situ hybridization. Anonymized clinical, pathologic, molecular, and response (RECIST v1.1) data were centrally curated and analyzed. RESULTS Although the typified never smoking (57%), mucinous adenocarcinoma (57%), and nonmetastatic (71%) phenotype predominated in 110 patients with NRG1 fusion-positive lung cancer, further diversity, including in smoking history (43%) and histology (43% nonmucinous and 6% nonadenocarcinoma), was elucidated. RNA-based testing identified most fusions (74%). Molecularly, six (of 18) novel 5' partners, 20 unique epidermal growth factor domain-inclusive chimeric events, and heterogeneous 5'/3' breakpoints were found. Platinum-doublet and taxane-based (post-platinum-doublet) chemotherapy achieved low objective response rates (ORRs 13% and 14%, respectively) and modest progression-free survival medians (PFS 5.8 and 4.0 months, respectively). Consistent with a low programmed death ligand-1 expressing (28%) and low tumor mutational burden (median: 0.9 mutations/megabase) immunophenotype, the activity of chemoimmunotherapy and single-agent immunotherapy was poor (ORR 0%/PFS 3.3 months and ORR 20%/PFS 3.6 months, respectively). Afatinib achieved an ORR of 25%, not contingent on fusion type, and a 2.8-month median PFS. CONCLUSION NRG1 fusion-positive lung cancers were molecularly, pathologically, and clinically more heterogeneous than previously recognized. The activity of cytotoxic, immune, and targeted therapies was disappointing. Further research examining NRG1-rearranged tumor biology is needed to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Alexander Drilon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Michael Duruisseaux
- Respiratory Department, Louis Pradel Hospital, Hospices Civils de Lyon Cancer Institute, Lyon, France
- Anticancer Antibodies Laboratory, Cancer Research Center of Lyon, Lyon, France
- Université Claude Bernard Lyon UMR INSERM 1052 CNRS 5286, Université de Lyon, Lyon, France
| | - Ji-Youn Han
- National Cancer Center, Korea, Goyang-si, South Korea
| | - Masaoki Ito
- Pangaea Oncology, Quiron-Dexeus University Institute, Barcelona, Spain
- Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Christina Falcon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Soo-Ryum Yang
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | | | - Haiquan Chen
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | - Morihito Okada
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Miguel Angel Molina
- Pangaea Oncology, Laboratory of Molecular Biology, Quiron-Dexeus University Institute, Barcelona, Spain
| | - Marie Wislez
- Université de Paris, Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Paris, France
- Team Inflammation, Complement, and Cancer, and Oncology Thoracic Unit Pulmonology Department, AP-HP, Hôpital Cochin, Paris, France
| | - Philippe Brun
- Department of Pneumology, Lungenklinik Heckeshorn, Helios Klinikum Emil von Behring, Valence, France
| | - Clarisse Dupont
- Respiratory Department, Louis Pradel Hospital, Hospices Civils de Lyon Cancer Institute, Lyon, France
| | - Eva Branden
- Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden
- Centre for Research and Development, Uppsala University/Region Gävleborg, Gävle, Sweden
| | - Giulio Rossi
- Local Health Authority of Romagna, Infermi Hospital, Rimini, Italy
- Local Health Authority of Romagna, St Maria delle Croci Hospital, Ravenna, Italy
| | | | - Siraj Ali
- Foundation Medicine Inc, Cambridge, MA
| | - Valérie Gounant
- Department of Pulmonology, Hôpital Tenon, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Fanny Magne
- Hopital Nord Ouest Villefranche sur Saône, Gleizé, France
| | | | | | - Isabelle Monnet
- Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Jin-Yuan Shih
- National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Joshua Sabari
- New York University Langone Health Perlmutter Cancer Center, New York, NY
| | | | - Viola W. Zhu
- Chao Family Comprehensive Cancer Center, Department of Medicine, Division of Hematology/Oncology, University of California, Irvine School of Medicine, Orange, CA
| | - Misako Nagasaka
- Karmanos Cancer Institute, Wayne State University, Detroit, MI
- Division of Neurology, Department of Internal Medicine, St Marianna University, Kawasaki, Japan
| | - Robert Doebele
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, CO
| | - D. Ross Camidge
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, CO
| | - Maria Arcila
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, University of California Irvine Medical Center, Orange, CA
| | - Denis Moro-Sibilot
- Clinique de Pneumologie, Pôle Médecine Aiguë Communautaire, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Rafael Rosell
- Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Lucia Anna Muscarella
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Stephen V. Liu
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Jacques Cadranel
- Department of Pneumology and Thoracic Oncology, Assistance Publique-Hopitaux de Paris, Tenon Hospital and GRC Theranoscan Sorbonne Université, Paris, France
| |
Collapse
|
28
|
Sonkar A, Kumar P, Gautam A, Maity B, Saha S. New Scope of Targeted Therapies in Lung Carcinoma. Mini Rev Med Chem 2021; 22:629-639. [PMID: 34353252 DOI: 10.2174/1389557521666210805104714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/30/2020] [Accepted: 04/27/2021] [Indexed: 11/22/2022]
Abstract
Lung cancer (LC) is the leading cause of cancer deaths worldwide. Recent research has also shown LC as a genomic disease, causing somatic mutations in patients. Tests related to mutational analysis and genome profiles have lately expanded significantly in the genetics/genomics field of LC. This review summarizes the current knowledge about different signalling pathways of LC based on the clinical impact of molecular targets. It describes the main molecular pathways and changes involved in the development, progression, and cellular breakdown of LC and the molecular changes. This review focuses on approved and targeted experimental therapies such as immunotherapy and clinical trials that examine the different targeted approaches to treating LC. We aimto clarify the differences in the extent of various genetic mutations in several areas for LC patients. Targeted molecular therapies for LC can be continued with advanced racial differences in genetic changes, which have a significant impact on the choice of drug treatment and our understanding of the profile of drug susceptibility/resistance. The most relevant genes described in this review are EGFR, KRAS, MET, BRAF, PIK3CA, STK11, ERBB3, PTEN, and RB1. Combined research efforts in this field are required to understand the genetic difference in LC outcomes in the future.
Collapse
Affiliation(s)
- Archana Sonkar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| | - Pranesh Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| | - Anurag Gautam
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| | - Biswanath Maity
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh. India
| | - Sudipta Saha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| |
Collapse
|
29
|
Tang H, Pang P, Qin Z, Zhao Z, Wu Q, Song S, Li F. The CPNE Family and Their Role in Cancers. Front Genet 2021; 12:689097. [PMID: 34367247 PMCID: PMC8345009 DOI: 10.3389/fgene.2021.689097] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Despite significant advances in cancer research and treatment, the overall prognosis of lung cancer patients remains poor. Therefore, the identification for novel therapeutic targets is critical for the diagnosis and treatment of lung cancer. CPNEs (copines) are a family of membrane-bound proteins that are highly conserved, soluble, ubiquitous, calcium dependent in a variety of eukaryotes. Emerging evidences have also indicated CPNE family members are involved in cancer development and progression as well. However, the expression patterns and clinical roles in cancer have not yet been well understood. In this review, we summarize recent advances concerning CPNE family members and provide insights into new potential mechanism involved in cancer development.
Collapse
Affiliation(s)
- Haicheng Tang
- Department of Respiratory and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Pei Pang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhu Qin
- Department of Respiratory and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhangyan Zhao
- Department of Respiratory and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Qingguo Wu
- Department of Respiratory and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shu Song
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Feng Li
- Department of Respiratory and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Das D, Wang J, Hong J. Next-Generation Kinase Inhibitors Targeting Specific Biomarkers in Non-Small Cell Lung Cancer (NSCLC): A Recent Overview. ChemMedChem 2021; 16:2459-2479. [PMID: 33929777 DOI: 10.1002/cmdc.202100166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/27/2021] [Indexed: 12/25/2022]
Abstract
Lung cancer causes many deaths globally. Mutations in regulatory genes, irregularities in specific signal transduction events, or alterations of signalling pathways are observed in cases of non-small cell lung cancer (NSCLC). Over the past two decades, a few kinases have been identified, validated, and studied as biomarkers for NSCLC. Among them, EGFR, ALK, ROS1, MET, RET, NTRK, and BRAF are regarded as targetable biomarkers to cure and/or control the disease. In recent years, the US Food and Drug Administration (FDA) approved more than 15 kinase inhibitors targeting these NSCLC biomarkers. The kinase inhibitors significantly improved the progression-free survival (PFS) of NSCLC patients. Challenges still remain for metastatic diseases and advanced NSCLC cases. New discoveries of potent kinase inhibitors and rapid development of modern medical technologies will help to control NSCLC cases. This article provides an overview of the discoveries of various types of kinase inhibitors against NSCLC, along with medicinal chemistry aspects and related developments in next-generation kinase inhibitors that have been reported in recent years.
Collapse
Affiliation(s)
- Debasis Das
- Discovery Chemistry Research, Arromax Pharmatech Co., Ltd., Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou, 215123, China
| | - Jingbing Wang
- Discovery Chemistry Research, Arromax Pharmatech Co., Ltd., Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou, 215123, China
| | - Jian Hong
- Discovery Chemistry Research, Arromax Pharmatech Co., Ltd., Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou, 215123, China
| |
Collapse
|
31
|
Wang X, Huang L, Cai J, Liu A. A Novel KIF5B-EGFR Fusion Variant in Non-Small-Cell Lung Cancer and Response to Afatinib: A Case Report. Onco Targets Ther 2021; 14:3739-3744. [PMID: 34163176 PMCID: PMC8214534 DOI: 10.2147/ott.s313896] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/03/2021] [Indexed: 12/24/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) fusions are rare genomic events in non-small-cell lung cancer (NSCLC). With advances in detection technology, some uncommon genomic mutation events, including EGFR fusions, have been detected. There are no standard treatment options for NSCLC patients harboring EGFR fusion. Herein, we report a case of KIF5B-EGFR fusion in NSCLC responding to tyrosine kinase inhibitors (TKIs). A 50-year-old male underwent left upper lobectomy followed by adjuvant chemotherapy for pathological stage IA3 lung adenocarcinoma. The tumor tissue was subjected to next-generation sequencing (NGS) and showed a KIF5B-EGFR fusion. When cancer recurrence occurred thirteen months later, the patient received afatinib (40 mg qd) as second-line treatment, and a partial response was observed, which resulted in an 11-month progression-free survival (PFS). This case provides valuable information on the response to afatinib in an NSCLC patient with a novel KIF5B-EGFR fusion. The NGS assay provides a powerful tool for identifying rare or atypical EGFR gene mutations in patients with NSCLC, which should be encouraged in clinical practice.
Collapse
Affiliation(s)
- Xia Wang
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, People's Republic of China
| | - Long Huang
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, People's Republic of China
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, People's Republic of China
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, People's Republic of China.,Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, 330000, Jiangxi Province, People's Republic of China
| |
Collapse
|
32
|
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: 1.3] [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
|
33
|
Tan AC, Chan J, Khasraw M. The role of immunotherapy in fusion-driven lung cancer. Expert Rev Anticancer Ther 2021; 21:461-464. [PMID: 33682589 DOI: 10.1080/14737140.2021.1899816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Johan Chan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Mustafa Khasraw
- Duke Center for Cancer Immunotherapy, Duke University, Durham, NC, USA
| |
Collapse
|
34
|
Ramani NS, Patel KP, Routbort MJ, Alvarez H, Broaddus R, Chen H, Rashid A, Lazar A, San Lucas FA, Yao H, Manekia J, Dang H, Barkoh BA, Medeiros LJ, Luthra R, Roy-Chowdhuri S. Factors Impacting Clinically Relevant RNA Fusion Assays Using Next-Generation Sequencing. Arch Pathol Lab Med 2021; 145:1405-1412. [PMID: 33493304 DOI: 10.5858/arpa.2020-0415-oa] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— RNA-based next-generation sequencing (NGS) assays are being used with increasing frequency for comprehensive molecular profiling of solid tumors. OBJECTIVE.— To evaluate factors that might impact clinical assay performance. DESIGN.— A 4-month retrospective review of cases analyzed by a targeted RNA-based NGS assay to detect fusions was performed. RNA extraction was performed from formalin-fixed, paraffin-embedded tissue sections and/or cytology smears of 767 cases, including 493 in-house and 274 outside referral cases. The types of samples included 422 core needle biopsy specimens (55%), 268 resection specimens (35%), and 77 cytology samples (10%). RESULTS.— Successful NGS fusion testing was achieved in 697 specimens (90.9%) and correlated positively with RNA yield (P < .001) and negatively with specimen necrosis (P = .002), decalcification (P < .001), and paraffin block age of more than 2 years (P = .001). Of the 697 cases that were successfully sequenced, 50 (7.2%) had clinically relevant fusions. The testing success rates and fusion detection rates were similar between core needle biopsy and cytology samples. In contrast, RNA fusion testing was often less successful using resection specimens (P = .007). Testing success was independent of the tumor percentage in the specimen, given that at least 20% tumor cellularity was present. CONCLUSIONS.— The success of RNA-based NGS testing is multifactorial and is influenced by RNA quality and quantity. Identification of preanalytical factors affecting RNA quality and yield can improve NGS testing success rates.
Collapse
Affiliation(s)
- Nisha S Ramani
- From the Departments of Pathology (Ramani, Broaddus, Chen, Rashid, Lazar, Roy-Chowdhuri)
| | - Keyur P Patel
- Hematopathology (Patel, Routbort, Alvarez, San Lucas, Manekia, Dang, Barkoh, Medeiros, Luthra)
| | - Mark J Routbort
- Hematopathology (Patel, Routbort, Alvarez, San Lucas, Manekia, Dang, Barkoh, Medeiros, Luthra)
| | - Hector Alvarez
- Hematopathology (Patel, Routbort, Alvarez, San Lucas, Manekia, Dang, Barkoh, Medeiros, Luthra)
| | - Russell Broaddus
- From the Departments of Pathology (Ramani, Broaddus, Chen, Rashid, Lazar, Roy-Chowdhuri)
| | - Hui Chen
- From the Departments of Pathology (Ramani, Broaddus, Chen, Rashid, Lazar, Roy-Chowdhuri)
| | - Asif Rashid
- From the Departments of Pathology (Ramani, Broaddus, Chen, Rashid, Lazar, Roy-Chowdhuri)
| | - Alex Lazar
- From the Departments of Pathology (Ramani, Broaddus, Chen, Rashid, Lazar, Roy-Chowdhuri)
| | - Francis A San Lucas
- Hematopathology (Patel, Routbort, Alvarez, San Lucas, Manekia, Dang, Barkoh, Medeiros, Luthra)
| | - Hui Yao
- and Bioinformatics and Computational Biology (Yao), The University of Texas MD Anderson Cancer Center, Houston. Broaddus is currently with the Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill
| | - Jawad Manekia
- Hematopathology (Patel, Routbort, Alvarez, San Lucas, Manekia, Dang, Barkoh, Medeiros, Luthra)
| | - Hyvan Dang
- Hematopathology (Patel, Routbort, Alvarez, San Lucas, Manekia, Dang, Barkoh, Medeiros, Luthra)
| | - Bedia A Barkoh
- Hematopathology (Patel, Routbort, Alvarez, San Lucas, Manekia, Dang, Barkoh, Medeiros, Luthra)
| | - L Jeffrey Medeiros
- Hematopathology (Patel, Routbort, Alvarez, San Lucas, Manekia, Dang, Barkoh, Medeiros, Luthra)
| | - Rajyalakshmi Luthra
- Hematopathology (Patel, Routbort, Alvarez, San Lucas, Manekia, Dang, Barkoh, Medeiros, Luthra)
| | - Sinchita Roy-Chowdhuri
- From the Departments of Pathology (Ramani, Broaddus, Chen, Rashid, Lazar, Roy-Chowdhuri)
| |
Collapse
|
35
|
van de Ven M, Koffijberg H, Retèl V, Monkhorst K, Smit E, van Harten W, IJzerman M. Real-World Utilization of Biomarker Testing for Patients with Advanced Non-Small Cell Lung Cancer in a Tertiary Referral Center and Referring Hospitals. J Mol Diagn 2021; 23:484-494. [PMID: 33493663 DOI: 10.1016/j.jmoldx.2021.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 12/21/2020] [Accepted: 01/12/2021] [Indexed: 12/26/2022] Open
Abstract
The continued introduction of biomarkers and innovative testing methods makes already complex diagnosis in patients with stage IV non-small-cell lung cancer (NSCLC) even more complex. This study primarily analyzed variations in biomarker testing in clinical practice in patients referred to a comprehensive cancer center in the Netherlands. The secondary aim was to compare the cost of biomarker testing with the cost of whole-genome sequencing. The cohort included 102 stage IV NSCLC patients who received biomarker testing in 2017 or 2018 at the comprehensive cancer center. The complete biomarker testing history of the cohort was identified using linked data from the comprehensive cancer center and the nationwide network and registry of histopathology and cytopathology in the Netherlands. Unique biomarker-test combinations, costs, turnaround times, and test utilization were examined. The results indicate substantial variation in test utilization and sequences. The mean cost per patient of biomarker testing was 2259.92 ± 1217.10 USD, or 1881.23 ± 1013.15 EUR. Targeted gene panels were most frequently conducted, followed by IHC analysis for programmed cell death protein ligand 1. Typically, the most common biomarkers were assessed within the first tests, and emerging biomarkers were tested further down the test sequence. At the cost of current biomarker testing, replacing current testing with whole-genome sequencing would have led to cost-savings in only two patients (2%).
Collapse
Affiliation(s)
- Michiel van de Ven
- Health Technology and Services Research, TechMed Center, University of Twente, Enschede, the Netherlands
| | - Hendrik Koffijberg
- Health Technology and Services Research, TechMed Center, University of Twente, Enschede, the Netherlands
| | - Valesca Retèl
- Health Technology and Services Research, TechMed Center, University of Twente, Enschede, the Netherlands; Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Kim Monkhorst
- Department of Pathology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Egbert Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Wim van Harten
- Health Technology and Services Research, TechMed Center, University of Twente, Enschede, the Netherlands; Rijnstate General Hospital, Arnhem, the Netherlands
| | - Maarten IJzerman
- Health Technology and Services Research, TechMed Center, University of Twente, Enschede, the Netherlands; Centre for Cancer Research and Centre for Health Policy, University of Melbourne, Melbourne, Australia; Peter MacCallum Cancer Centre, Melbourne, Australia.
| |
Collapse
|
36
|
Yamamoto H, Nozaki Y, Sugii A, Taguchi K, Hongo T, Jiromaru R, Sato M, Nakano T, Hashimoto K, Fujiwara M, Oda Y. Pan-tropomyosin receptor kinase immunoreactivity, ETV6-NTRK3 fusion subtypes, and RET rearrangement in salivary secretory carcinoma. Hum Pathol 2020; 109:37-44. [PMID: 33301751 DOI: 10.1016/j.humpath.2020.11.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 10/22/2022]
Abstract
Salivary secretory carcinoma (SASC) is frequently associated with ETV6-neurotrophic tyrosine receptor kinase (NTRK) 3 fusion and more rarely with RET, MET, or ALK rearrangement. We aimed to elucidate the potential diagnostic utility of pan-tropomyosin receptor kinase (Trk) immunohistochemistry and its relationship with the fusion gene subtype in SASC. We examined 33 cases of SASC for immunoexpression of pan-Trk, ALK and ROS1, and gene rearrangement of the ETV6, NTRK3, and RET genes using fluorescence in situ hybridization (FISH) and reverse transcription-polymerase chain reaction (RT-PCR). Thirty (90.9%) of 33 SASCs harbored ETV6-NTRK3 fusion gene transcripts by RT-PCR and/or both ETV6 and NTRK3 gene rearrangements by FISH, and 3 cases (9.1%) had RET gene rearrangement. Most NTRK3-rearranged SASCs (27/33 cases; 81.8%) had conventional ETV6 exon 5-NTRK3 exon 15 fusion, whereas 2 cases (6.1%) had both the conventional fusion and a novel ETV6 exon 4-NTRK3 exon 15 fusion variant. In the remaining one case (3%), only FISH revealed both ETV6 and NTRK3 rearrangements, suggesting an ETV6-NTRK3 fusion with an as yet undetermined break point. All 30 SASCs with ETV6-NTRK3 fusion and/or NTRK3 rearrangement showed nuclear and cytoplasmic immunoreactivity for pan-Trk. In contrast, 3 SASCs with RET rearrangement showed negative or only weak cytoplasmic staining for pan-Trk. There was no case harboring ALK and ROS1 rearrangements. All 17 non-SASC tumors were negative for pan-Trk. The results suggest that nuclear and cytoplasmic immunoreactivity for pan-TRK may be helpful to identify ETV6-NTRK3-fused SASCs and to distinguish them from RET-rearranged SASCs and morphological mimics.
Collapse
Affiliation(s)
- Hidetaka Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan.
| | - Yui Nozaki
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| | - Azusa Sugii
- Departments of Pathology, National Kyushu Cancer Center, Fukuoka, 811-1395 Japan
| | - Kenichi Taguchi
- Departments of Pathology, National Kyushu Cancer Center, Fukuoka, 811-1395 Japan
| | - Takahiro Hongo
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan; Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| | - Rina Jiromaru
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan; Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| | - Masanobu Sato
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| | - Takafumi Nakano
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| | - Kazuki Hashimoto
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| | - Minako Fujiwara
- Departments of Pathology, National Kyushu Medical Center, Fukuoka, 810-8563 Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| |
Collapse
|
37
|
Lakkaniga NR, Gunaganti N, Zhang L, Belachew B, Frett B, Leung YK, Li HY. Pyrrolo[2,3-d]pyrimidine derivatives as inhibitors of RET: Design, synthesis and biological evaluation. Eur J Med Chem 2020; 206:112691. [PMID: 32823007 PMCID: PMC10536156 DOI: 10.1016/j.ejmech.2020.112691] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/17/2022]
Abstract
Gene fusions and point mutations of RET kinase are crucial for driving thoracic cancers, including thyroid cancer and non-small cell lung cancer. Various scaffolds based on different heterocycles have been synthesized and evaluated as RET inhibitors. In this work, we investigate pyrrolo[2,3-d]pyrimidine derivatives for inhibition of RET-wt, drug resistant mutant RET V804M and RET gene fusion driven cell lines. Several compounds were synthesized and the structure activity relationship was extensively studied to optimize the scaffold. Thieno[2,3-d]pyrimidine, a bioisostere of pyrrolo[2,3-d]pyrimidine, was also explored for the effect on RET inhibition. We identified a lead compound, 59, which shows low nanomolar potency against RET-wt and RET V804M. Further 59 shows growth inhibition of LC-2/ad cells which RET-CCDC6 driven. We also determined that 59 is a type 2 inhibitor of RET and demonstrated its ability to inhibit migration of tumor cells. Based on computational studies, we proposed a binding pose of 59 in RET pocket and have quantified the contributions of individual residues for its binding. Together, 59 is an important lead compound which needs further evaluation in biological studies.
Collapse
Affiliation(s)
- Naga Rajiv Lakkaniga
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Naresh Gunaganti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lingtian Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Binyam Belachew
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yuet-Kin Leung
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
38
|
Sreedurgalakshmi K, Srikar R, Rajkumari R. CRISPR-Cas deployment in non-small cell lung cancer for target screening, validations, and discoveries. Cancer Gene Ther 2020; 28:566-580. [PMID: 33191402 DOI: 10.1038/s41417-020-00256-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/14/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022]
Abstract
Continued advancements in CRISPR-Cas systems have accelerated genome research. Use of CRISPR-Cas in cancer research has been of great interest that is resulting in development of orthogonal methods for drug target validations and discovery of new therapeutic targets through genome-wide screens of cancer cells. CRISPR-based screens have also revealed several new cancer drivers through alterations in tumor suppressor genes (TSGs) and oncogenes inducing resistance to targeted therapies via activation of alternate signaling pathways. Given such dynamic status of cancer, we review the application of CRISPR-Cas in non-small cell lung cancer (NSCLC) for development of mutant models, drug screening, target validation, novel target discoveries, and other emerging potential applications. In addition, CRISPR-based approach for development of novel anticancer combination therapies is also discussed in this review.
Collapse
Affiliation(s)
- K Sreedurgalakshmi
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India.,Division of Biosimilars and Gene Therapy, R&D, Levim Biotech LLP, Chennai, Tamilnadu, India
| | - R Srikar
- Division of Biosimilars and Gene Therapy, R&D, Levim Biotech LLP, Chennai, Tamilnadu, India.
| | - Reena Rajkumari
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India.
| |
Collapse
|
39
|
Solomon BJ, Zhou CC, Drilon A, Park K, Wolf J, Elamin Y, Davis HM, Soldatenkova V, Sashegyi A, Lin AB, Lin BK, F Loong HH, Novello S, Arriola E, Pérol M, Goto K, Santini FC. Phase III study of selpercatinib versus chemotherapy ± pembrolizumab in untreated RET positive non-small-cell lung cancer. Future Oncol 2020; 17:763-773. [PMID: 33150799 DOI: 10.2217/fon-2020-0935] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Selpercatinib, a novel, highly selective and potent, inhibitor of RET, demonstrated clinically meaningful antitumor activity with manageable toxicity in heavily pretreated and treatment-naive RET fusion-positive non-small-cell lung cancer patients in a Phase I/II clinical trial. LIBRETTO-431 (NCT04194944) is a randomized, global, multicenter, open-label, Phase III trial, evaluating selpercatinib versus carboplatin or cisplatin and pemetrexed chemotherapy with or without pembrolizumab in treatment-naive patients with locally advanced/metastatic RET fusion-positive nonsquamous non-small-cell lung cancer. The primary end point is progression-free survival by independent review. Key secondary end points include overall survival, response rate, duration of response and progression-free survival. Clinical trial registration: NCT04194944 (ClinicalTrials.gov).
Collapse
Affiliation(s)
| | | | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, Manhattan, NY 10065, USA
| | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jürgen Wolf
- Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
| | - Yasir Elamin
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | - Boris K Lin
- Eli Lilly and Company, Indianapolis, IN 46225, USA
| | | | - Silvia Novello
- Department of Oncology, AOU San Luigi-Orbassano, University of Turin, Italy
| | | | | | - Koichi Goto
- National Cancer Center Hospital East, Chiba, Japan
| | | |
Collapse
|
40
|
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]
|
41
|
Upadhya A, Yadav KS, Misra A. Targeted drug therapy in non-small cell lung cancer: Clinical significance and possible solutions-Part I. Expert Opin Drug Deliv 2020; 18:73-102. [PMID: 32954834 DOI: 10.1080/17425247.2021.1825377] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) comprises of 84% of all lung cancer cases. The treatment options for NSCLC at advanced stages are chemotherapy and radiotherapy. Chemotherapy involves conventional nonspecific chemotherapeutics, and targeted-protein/receptor-specific small molecule inhibitors. Biologically targeted therapies such as an antibody-based immunotherapy have been approved in combination with conventional therapeutics. Approved targeted chemotherapy is directed against the kinase domains of mutated cellular receptors such as epidermal growth factor receptor (EGFR), anaplastic lymphoma kinases (ALK), neurotrophic receptor kinases (NTRK) and against downstream signaling molecules such as BRAF (v-raf murine sarcoma viral oncogene homolog B1). Approved biologically targeted therapy involves the use of anti-angiogenesis antibodies and antibodies against immune checkpoints. AREAS COVERED The rationale for the employment of targeted therapeutics and the resistance that may develop to therapy are discussed. Novel targeted therapeutics in clinical trials are also included. EXPERT OPINION Molecular and histological profiling of a given tumor specimen to determine the aberrant onco-driver is a must before deciding a targeted therapeutic regimen for the patient. Periodic monitoring of the patients response to a given therapeutic regimen is also mandatory so that any semblance of resistance to therapy can be deciphered and the regimen may be accordingly altered.
Collapse
Affiliation(s)
- Archana Upadhya
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS , Mumbai, Maharashtra, India
| | - Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS , Mumbai, Maharashtra, India
| | - Ambikanandan Misra
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS , Mumbai, Maharashtra, India
| |
Collapse
|
42
|
Nozaki Y, Yamamoto H, Iwasaki T, Sato M, Jiromaru R, Hongo T, Yasumatsu R, Oda Y. Clinicopathological features and immunohistochemical utility of NTRK-, ALK-, and ROS1-rearranged papillary thyroid carcinomas and anaplastic thyroid carcinomas. Hum Pathol 2020; 106:82-92. [PMID: 32980422 DOI: 10.1016/j.humpath.2020.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 02/08/2023]
Abstract
NTRK1/3, ALK, and ROS1 translocations have been reported in a minor subset of papillary thyroid carcinomas (PTCs). We aimed to elucidate the prevalence and clinicopathological characteristics of these gene rearrangements and the utility of immunohistochemistry (IHC) in PTC and anaplastic thyroid carcinoma (ATC). We screened nonradiation-exposed cases of 307 PTCs and 16 ATCs by IHC for pan-Trk, ALK, and ROS1, followed by fluorescence in situ hybridization (FISH). In the PTC group, IHC for pan-Trk, ALK, and ROS1 was positive in 18 cases (5.9%), 1 case (0.3%), and 12 cases (3.9%), respectively. Among the pan-Trk IHC-positive cases (n = 18), 2 cases (11.1%; 0.7% of all PTCs) had NTRK1 or NTRK3 gene rearrangement with conventional PTC histology. The ALK IHC-positive case (n = 1) was the follicular variant of PTC with consistent ALK gene rearrangement. ROS1 gene rearrangement was not detectable in the ROS1 IHC-positive PTCs (0/12) by FISH. Most (approximately 70%) of the pan-Trk or ROS1 IHC-positive/FISH-negative cases had BRAF gene mutation with conventional PTC morphology. In the ATC group, neither ALK nor ROS1 IHC was positive, whereas pan-Trk IHC was positive in 1 case (6.3%) in which NTRK1 gene rearrangement was confirmed by FISH. These results suggest that NTRK, ALK, and ROS1 rearrangements are rare molecular events in nonradiation-exposed Japanese patients with PTC and ATC. Although IHC is not an entirely specific surrogate for these abnormalities and does not serve as a stand-alone companion diagnosis, the combined use of IHC and molecular testing may be helpful for determining promising therapeutic strategies with tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Yui Nozaki
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Takeshi Iwasaki
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Masanobu Sato
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan; Department of Otorhinolaryngology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Rina Jiromaru
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan; Department of Otorhinolaryngology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Takahiro Hongo
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Ryuji Yasumatsu
- Department of Otorhinolaryngology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Otorhinolaryngology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan.
| |
Collapse
|
43
|
Shih JY, Inoue A, Cheng R, Varea R, Kim SW. Does Pemetrexed Work in Targetable, Nonsquamous Non-Small-Cell Lung Cancer? A Narrative Review. Cancers (Basel) 2020; 12:cancers12092658. [PMID: 32957736 PMCID: PMC7563381 DOI: 10.3390/cancers12092658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/18/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The chemotherapy agent pemetrexed is currently considered in combination with other therapies for the treatment of advanced nonsquamous non-small-cell lung cancer (NSCLC) in patients negative for gene mutations/rearrangements. The aim of this review was to highlight data from clinical studies with pemetrexed in patients with advanced nonsquamous NSCLC positive for gene mutations/rearrangements. The results of the review suggest that pemetrexed could be a treatment option in patients with advanced nonsquamous NSCLC positive for certain gene mutations/rearrangements. Abstract Pemetrexed is currently mainly considered for the treatment of advanced nonsquamous non-small-cell lung cancer (NSCLC) negative for gene mutations/rearrangements (wild-type disease (WTD)). This narrative review aimed to highlight the role of pemetrexed in the treatment of onco-driven nonsquamous advanced NSCLC by reviewing published clinical studies. For epidermal growth factor receptor (EGFR) mutations, patient survival following first-line pemetrexed–platinum was longer than for WTD. Later-line pemetrexed-based treatment after tyrosine kinase inhibitor (TKI) failure provided greater benefits than non-pemetrexed regimens. First- and later-line pemetrexed-based therapy also provided survival benefits in patients with anaplastic lymphoma kinase (ALK) or ROS proto-oncogene 1 (ROS1) rearrangements. In patients with rearranged during transfection (RET) proto-oncogene rearrangements, survival with pemetrexed was similar to that in ALK- and ROS1-positive patients and longer than that in patients with Kirsten rat sarcoma (KRAS) virus proto-oncogene mutations or WTD, although the available studies were limited. For Erb-b2 receptor tyrosine kinase 2 (ERRB2) mutations, first-line pemetrexed showed outcomes similar to those for EGFR and KRAS alterations. Data on pemetrexed in patients with KRAS mutations or MNNG HOS-transforming (MET) expression were limited. Pemetrexed could be an option for first- and second-line treatment for TKI failure in nonsquamous advanced NSCLC with select targetable driver mutations.
Collapse
Affiliation(s)
- Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital, No. 7 Zhongshan South Road, Zhongzheng District, Taipei City 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University, 7 Chung-Shan S. Rd., Taipei 100, Taiwan
- Correspondence: ; Tel.: +886-2-2312-3456 (ext. 62905)
| | - Akira Inoue
- Department of Palliative Medicine, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan;
| | - Rebecca Cheng
- Eli Lilly and Company, Songshan District, Fuxing North Road 365, Taipei 105, Taiwan; (R.C.); (R.V.)
| | - Rocio Varea
- Eli Lilly and Company, Songshan District, Fuxing North Road 365, Taipei 105, Taiwan; (R.C.); (R.V.)
| | - Sang-We Kim
- Department of Oncology, Asan Medical Center, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul 05505, Korea;
| |
Collapse
|
44
|
Li H, Yan S, Liu Y, Ma L, Liu X, Liu Y, Cheng Y. Analysis of NTRK mutation and clinicopathologic factors in lung cancer patients in northeast China. Int J Biol Markers 2020; 35:36-40. [PMID: 32921229 DOI: 10.1177/1724600820949883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE NTRK mutations and clinicopathological factors in patients with lung cancer in northeast China were analyzed by next-generation sequencing (NGS), and references were provided for patients with NTRK mutations undergoing targeted therapy in northeast China. METHODS A total of 224 specimens in 173 patients with lung cancer were collected. This included 51 patients with matched tissue and whole blood samples,133 tissue samples, 84 whole blood samples, and 7 pleural effusion samples. NGS (520 genes) was used to detected NTRK mutations and clinicopathologic factors. RESULTS NTRK mutation was detected in eight patients (8/173, 4.6%), including four NTRK missense mutations (4/173, 2.3%), two NTRK fusion gene mutations (2/173, 1.2%), and two NTRK copy number deletions (2/173, 1.2%). Among the eight patients with NTRK mutations, four were associated with lung cancer driver gene mutations (3/4 EGFR, 1/4ALK); NTRK in two patients was inconsistent in tissue and paired whole blood testing; NTRK missense mutation was detected in one patient, and NTRK copy number deletion was detected in the other; and NTRK wild type was detected in two patients. There was no correlation between NTRK mutation and clinicopathologic factors (including gender, age, pathological type, smoking status, metastasis site). CONCLUSION NTRK mutation was only 4.6%, effective fusion gene mutation was 1.2%, and common driver gene mutation in lung cancer was evident in 50% of patients. The results of NTRK were inconsistent with matched tissues and whole blood. Therefore, patients with NTRK mutation should use a variety of specimen types and large target area sequencing (panel) analysis method to provide individualized treatment.
Collapse
Affiliation(s)
- Hui Li
- Translational Medical Research Lab, Jilin Cancer Hospital, Changchun, China
| | - Shi Yan
- Translational Medical Research Lab, Jilin Cancer Hospital, Changchun, China
| | - Ying Liu
- Department of Medical Oncology, Jilin Cancer Hospital, Changchun, China
| | - Lixia Ma
- Department of Medical Oncology, Jilin Cancer Hospital, Changchun, China
| | - Xianhong Liu
- Department of Medical Oncology, Jilin Cancer Hospital, Changchun, China
| | - Yan Liu
- Translational Medical Research Lab, Jilin Cancer Hospital, Changchun, China
| | - Ying Cheng
- Translational Medical Research Lab, Jilin Cancer Hospital, Changchun, China.,Department of Medical Oncology, Jilin Cancer Hospital, Changchun, China
| |
Collapse
|
45
|
Matsukawa T, Aplan PD. Clinical and molecular consequences of fusion genes in myeloid malignancies. Stem Cells 2020; 38:1366-1374. [PMID: 32745287 DOI: 10.1002/stem.3263] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/12/2020] [Accepted: 05/17/2020] [Indexed: 11/07/2022]
Abstract
Leukemias are heterogeneous diseases characterized by aberrant hematopoietic stem and progenitor cells (HSPCs). Oncogenic fusion genes and proteins, produced via gross chromosomal rearrangements, such as chromosomal translocation, insertion, and inversion, play important roles in hematologic malignancies. These oncoproteins alter fundamental cellular properties, such as self-renewal, differentiation, and proliferation, and confer leukemogenic potential to HSPCs. In addition to providing fundamental insights into the process of leukemic transformation, these fusion genes provide targets for treatment and monitoring of myeloid leukemias. Furthermore, new technologies such as next-generation sequencing have allowed additional insights into the nature of leukemic fusion genes. In this review, we discuss the history, biologic effect, and clinical impact of fusion genes in the field of myeloid leukemias.
Collapse
Affiliation(s)
- Toshihiro Matsukawa
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter D Aplan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
46
|
Peters S, Reck M, Smit EF, Mok T, Hellmann MD. How to make the best use of immunotherapy as first-line treatment of advanced/metastatic non-small-cell lung cancer. Ann Oncol 2020; 30:884-896. [PMID: 30912805 DOI: 10.1093/annonc/mdz109] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Antibodies that target programmed death 1 (PD-1) or its ligand [programmed death ligand 1 (PD-L1)] have become a mainstay of first-line treatment of advanced/metastatic non-small-cell lung cancer (NSCLC) without targetable genetic alterations. In this review, we summarize results from recent clinical trials that have evaluated the anti-PD-1 antibodies pembrolizumab and nivolumab and the anti-PD-L1 antibodies atezolizumab and durvalumab as first-line treatment as monotherapy and in combination with chemotherapy, other immunotherapies, and antiangiogenesis agents. We discuss factors that may influence treatment selection, including patient baseline clinical and demographic characteristics, tumor histology, and biomarkers such as PD-L1 expression and tumor mutation burden. While immunotherapy has become a central component of first-line treatment of most patients with advanced NSCLC, important questions remain about how treatment should be managed for individual patients.
Collapse
Affiliation(s)
- S Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland.
| | - M Reck
- Lung Clinic Grosshansdorf, Airway Research Center North (ARCN), German Center of Lung Research (DZL), Grosshansdorf, Germany
| | - E F Smit
- Department of Pulmonary Diseases, VU University Medical Centre, Amsterdam; Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - T Mok
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Chinese University of Hong Kong, Hong Kong, China
| | - M D Hellmann
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York; Parker Institute for Cancer Immunotherapy, New York, USA
| |
Collapse
|
47
|
Burgess JT, Rose M, Boucher D, Plowman J, Molloy C, Fisher M, O'Leary C, Richard DJ, O'Byrne KJ, Bolderson E. The Therapeutic Potential of DNA Damage Repair Pathways and Genomic Stability in Lung Cancer. Front Oncol 2020; 10:1256. [PMID: 32850380 PMCID: PMC7399071 DOI: 10.3389/fonc.2020.01256] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
Despite advances in our understanding of the molecular biology of the disease and improved therapeutics, lung cancer remains the most common cause of cancer-related deaths worldwide. Therefore, an unmet need remains for improved treatments, especially in advanced stage disease. Genomic instability is a universal hallmark of all cancers. Many of the most commonly prescribed chemotherapeutics, including platinum-based compounds such as cisplatin, target the characteristic genomic instability of tumors by directly damaging the DNA. Chemotherapies are designed to selectively target rapidly dividing cells, where they cause critical DNA damage and subsequent cell death (1, 2). Despite the initial efficacy of these drugs, the development of chemotherapy resistant tumors remains the primary concern for treatment of all lung cancer patients. The correct functioning of the DNA damage repair machinery is essential to ensure the maintenance of normal cycling cells. Dysregulation of these pathways promotes the accumulation of mutations which increase the potential of malignancy. Following the development of the initial malignancy, the continued disruption of the DNA repair machinery may result in the further progression of metastatic disease. Lung cancer is recognized as one of the most genomically unstable cancers (3). In this review, we present an overview of the DNA damage repair pathways and their contributions to lung cancer disease occurrence and progression. We conclude with an overview of current targeted lung cancer treatments and their evolution toward combination therapies, including chemotherapy with immunotherapies and antibody-drug conjugates and the mechanisms by which they target DNA damage repair pathways.
Collapse
Affiliation(s)
- Joshua T Burgess
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Maddison Rose
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Didier Boucher
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Jennifer Plowman
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Christopher Molloy
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Mark Fisher
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Connor O'Leary
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Derek J Richard
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Kenneth J O'Byrne
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Emma Bolderson
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Princess Alexandra Hospital, Brisbane, QLD, Australia
| |
Collapse
|
48
|
Clarke M, Mackay A, Ismer B, Pickles JC, Tatevossian RG, Newman S, Bale TA, Stoler I, Izquierdo E, Temelso S, Carvalho DM, Molinari V, Burford A, Howell L, Virasami A, Fairchild AR, Avery A, Chalker J, Kristiansen M, Haupfear K, Dalton JD, Orisme W, Wen J, Hubank M, Kurian KM, Rowe C, Maybury M, Crosier S, Knipstein J, Schüller U, Kordes U, Kram DE, Snuderl M, Bridges L, Martin AJ, Doey LJ, Al-Sarraj S, Chandler C, Zebian B, Cairns C, Natrajan R, Boult JKR, Robinson SP, Sill M, Dunkel IJ, Gilheeney SW, Rosenblum MK, Hughes D, Proszek PZ, Macdonald TJ, Preusser M, Haberler C, Slavc I, Packer R, Ng HK, Caspi S, Popović M, Faganel Kotnik B, Wood MD, Baird L, Davare MA, Solomon DA, Olsen TK, Brandal P, Farrell M, Cryan JB, Capra M, Karremann M, Schittenhelm J, Schuhmann MU, Ebinger M, Dinjens WNM, Kerl K, Hettmer S, Pietsch T, Andreiuolo F, Driever PH, Korshunov A, Hiddingh L, Worst BC, Sturm D, Zuckermann M, Witt O, Bloom T, Mitchell C, Miele E, Colafati GS, Diomedi-Camassei F, Bailey S, Moore AS, Hassall TEG, Lowis SP, Tsoli M, Cowley MJ, Ziegler DS, Karajannis MA, Aquilina K, Hargrave DR, Carceller F, Marshall LV, von Deimling A, Kramm CM, Pfister SM, Sahm F, Baker SJ, Mastronuzzi A, Carai A, Vinci M, Capper D, Popov S, Ellison DW, Jacques TS, Jones DTW, Jones C. Infant High-Grade Gliomas Comprise Multiple Subgroups Characterized by Novel Targetable Gene Fusions and Favorable Outcomes. Cancer Discov 2020; 10:942-963. [PMID: 32238360 PMCID: PMC8313225 DOI: 10.1158/2159-8290.cd-19-1030] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 03/03/2020] [Accepted: 03/20/2020] [Indexed: 11/16/2022]
Abstract
Infant high-grade gliomas appear clinically distinct from their counterparts in older children, indicating that histopathologic grading may not accurately reflect the biology of these tumors. We have collected 241 cases under 4 years of age, and carried out histologic review, methylation profiling, and custom panel, genome, or exome sequencing. After excluding tumors representing other established entities or subgroups, we identified 130 cases to be part of an "intrinsic" spectrum of disease specific to the infant population. These included those with targetable MAPK alterations, and a large proportion of remaining cases harboring gene fusions targeting ALK (n = 31), NTRK1/2/3 (n = 21), ROS1 (n = 9), and MET (n = 4) as their driving alterations, with evidence of efficacy of targeted agents in the clinic. These data strongly support the concept that infant gliomas require a change in diagnostic practice and management. SIGNIFICANCE: Infant high-grade gliomas in the cerebral hemispheres comprise novel subgroups, with a prevalence of ALK, NTRK1/2/3, ROS1, or MET gene fusions. Kinase fusion-positive tumors have better outcome and respond to targeted therapy clinically. Other subgroups have poor outcome, with fusion-negative cases possibly representing an epigenetically driven pluripotent stem cell phenotype.See related commentary by Szulzewsky and Cimino, p. 904.This article is highlighted in the In This Issue feature, p. 890.
Collapse
Affiliation(s)
- Matthew Clarke
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Alan Mackay
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Britta Ismer
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Jessica C Pickles
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Ruth G Tatevossian
- Department of Neuropathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Scott Newman
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Tejus A Bale
- Department of Neuropathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Iris Stoler
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany
| | - Elisa Izquierdo
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Sara Temelso
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Diana M Carvalho
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Valeria Molinari
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Anna Burford
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Louise Howell
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Alex Virasami
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Amy R Fairchild
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Aimee Avery
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Jane Chalker
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Mark Kristiansen
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Kelly Haupfear
- Department of Neuropathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - James D Dalton
- Department of Neuropathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Wilda Orisme
- Department of Neuropathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ji Wen
- Department of Neuropathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Michael Hubank
- Molecular Diagnostics, Royal Marsden Hospital NHS Trust, Sutton, United Kingdom
| | - Kathreena M Kurian
- Brain Tumour Research Centre, University of Bristol, Bristol, United Kingdom
| | - Catherine Rowe
- Brain Tumour Research Centre, University of Bristol, Bristol, United Kingdom
| | - Mellissa Maybury
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia
- Oncology Service, Queensland Children's Hospital, Brisbane, Australia
- Child Health Research Centre, The University of Queensland, South Brisbane, Australia
| | - Stephen Crosier
- Newcastle Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Jeffrey Knipstein
- Division of Pediatric Hematology/Oncology/BMT, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ulrich Schüller
- Department of Neuropathology, University Hospital Hamburg-Eppendorf, and Research Institute Children's Cancer Center, Hamburg, Germany
- Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Uwe Kordes
- Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - David E Kram
- Section of Pediatric Hematology-Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Matija Snuderl
- Department of Neuropathology, NYU Langone Health, New York, New York
| | - Leslie Bridges
- Department of Neuropathology, St George's Hospital NHS Trust, London, United Kingdom
| | - Andrew J Martin
- Department of Neurosurgery, St George's Hospital NHS Trust, London, United Kingdom
| | - Lawrence J Doey
- Department of Clinical Neuropathology, Kings College Hospital NHS Trust, London, United Kingdom
| | - Safa Al-Sarraj
- Department of Clinical Neuropathology, Kings College Hospital NHS Trust, London, United Kingdom
| | - Christopher Chandler
- Department of Neurosurgery, Kings College Hospital NHS Trust, London, United Kingdom
| | - Bassel Zebian
- Department of Neurosurgery, Kings College Hospital NHS Trust, London, United Kingdom
| | - Claire Cairns
- Department of Neurosurgery, Kings College Hospital NHS Trust, London, United Kingdom
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Jessica K R Boult
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Simon P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Martin Sill
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Stephen W Gilheeney
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Marc K Rosenblum
- Department of Neuropathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Debbie Hughes
- Molecular Diagnostics, Royal Marsden Hospital NHS Trust, Sutton, United Kingdom
| | - Paula Z Proszek
- Molecular Diagnostics, Royal Marsden Hospital NHS Trust, Sutton, United Kingdom
| | - Tobey J Macdonald
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Matthias Preusser
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christine Haberler
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Irene Slavc
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Roger Packer
- Center for Neuroscience and Behavioural Medicine, Children's National Medical Center, Washington, DC
| | - Ho-Keung Ng
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, China
| | - Shani Caspi
- Cancer Research Center, Sheba Medical Center, Tel Aviv, Israel
| | - Mara Popović
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Barbara Faganel Kotnik
- Department of Hematology and Oncology, University Children's Hospital, Ljubljana, Slovenia
| | - Matthew D Wood
- Department of Pathology, Oregon Health & Science University, Portland, Oregon
| | - Lissa Baird
- Department of Neurosurgery, Oregon Health & Science University, Portland, Oregon
| | - Monika Ashok Davare
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon
| | - David A Solomon
- Department of Pathology, University of California, San Francisco, California
- Clinical Cancer Genomics Laboratory, University of California, San Francisco, California
| | - Thale Kristin Olsen
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Petter Brandal
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Michael Farrell
- Department of Histopathology, Beaumont Hospital, Dublin, Ireland
| | - Jane B Cryan
- Department of Histopathology, Beaumont Hospital, Dublin, Ireland
| | - Michael Capra
- Paediatric Oncology, Our Lady's Children's Hospital, Dublin, Ireland
| | - Michael Karremann
- Department of Pediatrics, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jens Schittenhelm
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Germany
| | | | - Martin Ebinger
- Department of Pediatric Hematology and Oncology, University Hospital Tübingen, Germany
| | - Winand N M Dinjens
- Department of Pathology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Hospital Muenster, Germany
| | - Simone Hettmer
- Department of Pediatric Hematology and Oncology, University Hospital Freiburg, Germany
| | - Torsten Pietsch
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Felipe Andreiuolo
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Pablo Hernáiz Driever
- Department of Paediatric Haematology/Oncology Charité Universitätsmedizin, Berlin, Germany
| | - Andrey Korshunov
- Department of Neuropathology, University Hospital Heidelberg, Germany
| | - Lotte Hiddingh
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara C Worst
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Dominik Sturm
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marc Zuckermann
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Olaf Witt
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Tabitha Bloom
- BRAIN UK, University of Southampton, Southampton, United Kingdom
| | - Clare Mitchell
- BRAIN UK, University of Southampton, Southampton, United Kingdom
| | - Evelina Miele
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Giovanna Stefania Colafati
- Oncological Neuroradiology Unit, Department of Diagnostic Imaging, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | | | - Simon Bailey
- Newcastle Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Andrew S Moore
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia
- Oncology Service, Queensland Children's Hospital, Brisbane, Australia
- Child Health Research Centre, The University of Queensland, South Brisbane, Australia
| | - Timothy E G Hassall
- Oncology Service, Queensland Children's Hospital, Brisbane, Australia
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Stephen P Lowis
- Brain Tumour Research Centre, University of Bristol, Bristol, United Kingdom
| | - Maria Tsoli
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
| | - Mark J Cowley
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
| | - David S Ziegler
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
| | - Matthias A Karajannis
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Kristian Aquilina
- Department of Neurosurgery, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Darren R Hargrave
- Department of Paediatric Oncology, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Fernando Carceller
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
- Children & Young People's Unit, Royal Marsden Hospital NHS Trust, Sutton, United Kingdom
| | - Lynley V Marshall
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
- Children & Young People's Unit, Royal Marsden Hospital NHS Trust, Sutton, United Kingdom
| | - Andreas von Deimling
- Department of Neuropathology, University Hospital Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christof M Kramm
- Division of Pediatric Hematology and Oncology, University Medical Centre Göttingen, Germany
| | - Stefan M Pfister
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Sahm
- Department of Paediatric Haematology/Oncology Charité Universitätsmedizin, Berlin, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Suzanne J Baker
- Department of Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Angela Mastronuzzi
- Neuro-oncology Unit, Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Andrea Carai
- Oncological Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Maria Vinci
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - David Capper
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sergey Popov
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
- Department of Pathology, University of Wales Hospital NHS Trust, Cardiff, United Kingdom
| | - David W Ellison
- Department of Neuropathology, St. Jude Children's Research Hospital, Memphis, Tennessee.
| | - Thomas S Jacques
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom.
| | - David T W Jones
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
49
|
Li J, Li X, Wang W, Shao Y, Zhang Y, Song Z. Gene Alterations in Paired Supernatants and Precipitates from Malignant Pleural Effusions of Non-Squamous Non-Small Cell Lung Cancer. Transl Oncol 2020; 13:100784. [PMID: 32428850 PMCID: PMC7236047 DOI: 10.1016/j.tranon.2020.100784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE: This study investigated the feasibility of using malignant pleural effusion (MPE) supernatant and paired cell blocks (precipitate) for gene profiling in patients with non-small cell lung cancer (NSCLC) using next-generation sequencing (NGS) technique. METHODS: Stage IV non-squamous NSCLC patients with MPE were eligible in this prospective study and recruited from Zhejiang Cancer Hospital between May 2014 and October 2015. MPE supernatant and paired precipitate sample gene alterations were determined with NGS containing 14 cancer-related genes. Progression free survival (PFS) was evaluated using Kaplan–Meier method and compared using log-rank test. RESULTS: A total of 102 patients were enrolled in the present study. All pleural effusions were confirmed as malignant with cytological smears. A total of 77 paired MPE supernatant and precipitate samples were acquired from the 102 patients. The results revealed that there were no statistically significant differences in the detection rate and maximum allelic fraction between supernatant and precipitate samples (P = 1.0 and P = .6). Collectively, 172 and 158 genomic alterations with 112 shared mutations were identified in supernatant and precipitate samples, respectively. Comparable PFS was found in EGFR mutation patients according to the supernatant and precipitate sample results (14.0 vs.13.9 months, P = .90). CONCLUSIONS: These results demonstrated that MPE supernatants were comparable to precipitate samples for detection of genetic alterations. However, gene mutation heterogeneity was found between these two media types.
Collapse
Affiliation(s)
- Jianqiang Li
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou 310022, China; Department of Thoracic Oncology Surgery, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China; Department of Thoracic Oncology Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Xingliang Li
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang 314000, China
| | - Wenxian Wang
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou 310022, China; Department of Thoracic Oncology Surgery, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China; Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yang Shao
- Translational Medicine Research Centre, Geneseeq Technology Inc., Toronto, Ontario, Canada
| | - Yiping Zhang
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou 310022, China; Department of Thoracic Oncology Surgery, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China; Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Zhengbo Song
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou 310022, China; Department of Thoracic Oncology Surgery, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China; Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China.
| |
Collapse
|
50
|
Li W, Liu Y, Li W, Chen L, Ying J. Intergenic Breakpoints Identified by DNA Sequencing Confound Targetable Kinase Fusion Detection in NSCLC. J Thorac Oncol 2020; 15:1223-1231. [PMID: 32151779 DOI: 10.1016/j.jtho.2020.02.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Next-generation sequencing (NGS) based on genomic DNA has been widely applied for gene rearrangement detection in patients with NSCLC. However, intergenic-breakpoint fusions, in which one or both genomic breakpoints localize to intergenic regions, confound kinase fusion detection. We evaluated the function of intergenic-breakpoint fusions with multiplex molecular testing approaches. METHODS NSCLCs with intergenic-breakpoint fusion identified by DNA-based NGS were analyzed by RNA-based NGS, immunohistochemistry (IHC), and fluorescence in situ hybridization. RESULTS A total of 26 cases with single intergenic-breakpoint fusion were identified from a large cohort of NSCLCs using DNA-based NGS. Of the 26 cases, RNA-based NGS detected expressed fusion transcripts in 11 cases, but the genomic breakpoint position did not logically predict breakpoint of the fusion transcript in these cases, possibly owing to complex rearrangements (n = 5), alternative splicing (n = 2), and reciprocal rearrangement (n = 4). Nonetheless, no expressed fusion transcript was detected in five cases. Moreover, positive anaplastic lymphoma receptor tyrosine (ALK) finding was observed in three of the remaining 10 cases with IHC but not with RNA-based NGS. Three patients with intergenic-breakpoint ALK fusion with or without RNA-based NGS or IHC confirmation who received crizotinib treatment were found to have partial responses. However, one patient with intergenic-breakpoint ROS1, given the positive fluorescence in situ hybridization result, received crizotinib but developed progressive disease within 1 month, possibly owing to no functional fusion transcript detected by RNA-based NGS. CONCLUSIONS Intergenic-breakpoint fusions detected by DNA sequencing confound kinase fusion detection in NSCLC, as functional fusion transcripts may be generated or not. Additional validation testing using RNA/protein assay should be performed in intergenic-breakpoint fusion cases to guide optimal treatment.
Collapse
Affiliation(s)
- Weihua Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Wenbin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Li Chen
- HeliTec Biotechnologies, Shenzhen, Guangdong Province, People's Republic of China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|