1
|
Fu Y, Liu Q, Wang X, Sun L, Han X, Meng X. Clinical difference on the variants and co-mutation in a Chinese cohort with ALK-positive advanced non-small cell lung cancer. Clin Transl Oncol 2024; 26:2513-2521. [PMID: 38637357 DOI: 10.1007/s12094-024-03481-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024]
Abstract
PURPOSE Despite the generally favourable prognoses observed in patients with ALK-positive non-small cell lung cancer (NSCLC), there remains significant variability in clinical outcomes. The objective of this study is to enhance patient stratification by examining both the specific sites of gene fusion and the presence of co-occurring mutations. METHODS We collected retrospective clinical and pathological data on ALK-positive patients with locally advanced or metastatic disease. ALK fusion variants and concomitant mutations were identified through next-generation sequencing technology. We then assessed treatment efficacy via tumor response and survival metrics. RESULTS This study included a total of 59 patients, with 49 harboring echinoderm microtubule-associated protein-like 4 (EML4)-ALK fusions and 10 presenting with rare fusions. The median follow-up period was 33 months. Clinical outcomes between non-EML4-ALK and EML4-ALK patients were comparable. Among the EML4-ALK cohort, patients with longer variants (v1, v2, v8) demonstrated superior progression-free survival (PFS) (median PFS: 34 months vs. 11 months; hazard ratio [HR]: 2.28; P = 0.05) compared to those with shorter variants (v3, v5). Furthermore, patients treated with second-generation ALK inhibitors (ALKi) displayed a progression-free survival advantage (median PFS: not reached [NR] vs. 9 months; HR: 5.37; P = 0.013). Baseline TP53 co-mutation were linked with a substantially shorter OS (median OS,37 months vs. NR; HR 2.74; P = 0.047). CONCLUSIONS In ALK+ NSCLC, longer EML4-ALK variants correlate with improved prognosis and enhanced response to second-generation ALKi, while TP53 co-mutations indicate a negative prognosis.
Collapse
Affiliation(s)
- Ying Fu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qing Liu
- Department of Oncology, Changqing District People's Hospital, Jinan, Shandong, China
| | - Xiaohan Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Liangchao Sun
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiao Han
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
2
|
Alidousty C, Becker A, Binot E, Hillmer AM, Merkelbach-Bruse S, Budde B, Bäßmann I, Rappl G, Wolf J, Eich ML, Noh KW, Buettner R, Schultheis AM. Frequency and functional characterization of fusion genes in squamous cell carcinoma of the lung. Gene 2024; 895:148018. [PMID: 37981082 DOI: 10.1016/j.gene.2023.148018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/23/2023] [Accepted: 11/17/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION In contrast to lung adenocarcinoma (LUAD), targetable genetic alterations are less frequently detected in squamous cell carcinoma of the lung (LUSC). Over the last years, gene fusions have become promising targets in many solid cancers. Here, we analysed a cohort of LUSC, identified recurrent fusion genes and functionally characterised these tumour genomes. METHODS A subset of 1608 squamous cell carcinomas of the lung was analysed by means of the FusionPlex® Lung Panel to identify potentially targetable gene fusions using targeted next-generation sequencing. Cases harbouring recurrent gene fusions were further analysed using FISH, Cytoscan HD arrays and cell culture experiments. RESULTS We found both, known and novel gene fusions in about 3 % of the cases. Known fusions occurring in lung cancer included ALK::EML4, EGFRvIII, EZR::ROS1 and FGFR3::TACC. We further identified recurrent gene fusions of currently unknown biological function, involving EGFR::VSTM2A and NSD3::FGFR1 and showed that the occurrence of the EGFR::VSTM2A fusion is accompanied by high-level amplification of EGFR. Our analyses further revealed that the genomes of these LUSC patients are chromosomally unstable, which leads us to believe that such non-actionable genomic rearrangements may be a result of "chromosomal chaos" most probably not representing exclusive cancer-driving genes in this cancer entity. CONCLUSIONS We emphasise that caution should be taken when novel fusions are found and that the appearance of new gene fusions should always be interpreted in the molecular context of the respective disease.
Collapse
Affiliation(s)
- Christina Alidousty
- University Hospital and Medical Faculty, University of Cologne, Institute of Pathology, Kerpener Straße 62, 50937 Cologne, Germany; Network Genomic Medicine, Cologne, Germany
| | - Arvid Becker
- University Hospital and Medical Faculty, University of Cologne, Institute of Pathology, Kerpener Straße 62, 50937 Cologne, Germany
| | - Elke Binot
- University Hospital and Medical Faculty, University of Cologne, Institute of Pathology, Kerpener Straße 62, 50937 Cologne, Germany
| | - Axel M Hillmer
- University Hospital and Medical Faculty, University of Cologne, Institute of Pathology, Kerpener Straße 62, 50937 Cologne, Germany; Network Genomic Medicine, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Robert Koch Strasse 21, 50931 Cologne, Germany
| | - Sabine Merkelbach-Bruse
- University Hospital and Medical Faculty, University of Cologne, Institute of Pathology, Kerpener Straße 62, 50937 Cologne, Germany; Network Genomic Medicine, Cologne, Germany
| | - Birgit Budde
- Cologne Center for Genomics, Medical Faculty of the University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Ingelore Bäßmann
- Cologne Center for Genomics, Medical Faculty of the University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Gunter Rappl
- Center for Molecular Medicine Cologne, University of Cologne, Robert Koch Strasse 21, 50931 Cologne, Germany
| | - Jürgen Wolf
- Lung Cancer Group Cologne, Department I for Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Marie-Lisa Eich
- University Hospital and Medical Faculty, University of Cologne, Institute of Pathology, Kerpener Straße 62, 50937 Cologne, Germany
| | - Ka-Won Noh
- University Hospital and Medical Faculty, University of Cologne, Institute of Pathology, Kerpener Straße 62, 50937 Cologne, Germany
| | - Reinhard Buettner
- University Hospital and Medical Faculty, University of Cologne, Institute of Pathology, Kerpener Straße 62, 50937 Cologne, Germany; Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I for Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Anne Maria Schultheis
- University Hospital and Medical Faculty, University of Cologne, Institute of Pathology, Kerpener Straße 62, 50937 Cologne, Germany.
| |
Collapse
|
3
|
Dagogo-Jack I, Kiedrowski LA, Lennerz JK. Molecular heterogeneity and co-altered genes in MET-amplified ALK-positive lung cancer: Implications for MET targeted therapy. Lung Cancer 2023; 186:107383. [PMID: 37813016 DOI: 10.1016/j.lungcan.2023.107383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/11/2023]
Abstract
OBJECTIVES MET amplification is a common mechanism of resistance to second- and third-generation anaplastic lymphoma kinase (ALK) inhibitors. In case series of MET-amplified ALK-rearranged (ALK + ) lung cancer, durability of responses to combinations targeting ALK and MET is variable, suggesting heterogeneity across tumors. However, little is known about the molecular composition of this subset of ALK-rearranged (ALK + ) NSCLC. MATERIALS AND METHODS We queried tissue and plasma databases to compile a group of > 50 specimens with ALK rearrangements and concurrent MET amplification. Fluorescence in-situ hybridization (FISH) and next-generation sequencing (NGS) were utilized to quantify the range of MET copies and describe the global molecular landscape of co-altered genes. RESULTS By FISH, high-level amplification (overall MET/centromere 7 probe ratio ≥ 5) was detected in 75 % of MET-amplified ALK + NSCLC tissue specimens. Intralesional heterogeneity of MET copies was observed, with high-level amplification identified even in cells from tumors with overall low-level MET amplification. Analysis of 48 MET-amplified ALK + NSCLC plasma specimens suggested that high-level amplification is rarely (17 %) detected in plasma. In both tissue and plasma, EML4-ALK variant 1 was the predominant variant (51 %) identified in MET-amplified specimens. ALK kinase domain mutations were only present in a minority of MET-amplified ALK + NSCLCs. MET-amplified ALK + NSCLC plasma specimens were enriched for TP53 mutations (81 % vs 45 %, p = 0.002), EGFR amplification (17 % vs 4 %, p < 0.001), and MYC amplification (21 % vs 3 %, p < 0.001) compared to ALK + NSCLC specimens lacking MET amplification. CONCLUSIONS MET-amplified, ALK + NSCLC often presents with high-level and heterogeneous amplification in tissue, seldom overlaps with ALK mutations, and frequently co-occurs with alterations associated with aggressive tumor biology.
Collapse
Affiliation(s)
- Ibiayi Dagogo-Jack
- Massachusetts General Hospital Cancer Center and Department of Medicine, Massachusetts General Hospital, USA; Harvard Medical School, Center for Integrated Diagnostics, Massachusetts General Hospital, USA.
| | - Lesli A Kiedrowski
- Department of Medical Affairs, Guardant Health, Inc, Center for Integrated Diagnostics, Massachusetts General Hospital, USA
| | - Jochen K Lennerz
- Harvard Medical School, Center for Integrated Diagnostics, Massachusetts General Hospital, USA; Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital, USA
| |
Collapse
|
4
|
Brik A, Wichert K, Weber DG, Szafranski K, Rozynek P, Meier S, Ko YD, Büttner R, Gerwert K, Behrens T, Brüning T, Johnen G. Assessment of MYC and TERT copy number variations in lung cancer using digital PCR. BMC Res Notes 2023; 16:279. [PMID: 37858127 PMCID: PMC10585721 DOI: 10.1186/s13104-023-06566-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
OBJECTIVE Lung cancer is the second most frequent cancer type and the most common cause of cancer-related deaths worldwide. Alteration of gene copy numbers are associated with lung cancer and the determination of copy number variations (CNV) is appropriate for the discrimination between tumor and non-tumor tissue in lung cancer. As telomerase reverse transcriptase (TERT) and v-myc avian myelocytomatosis viral oncogene homolog (MYC) play a role in lung cancer the aims of this study were the verification of our recent results analyzing MYC CNV in tumor and non-tumor tissue of lung cancer patients using an independent study group and the assessment of TERT CNV as an additional marker. RESULTS TERT and MYC status was analyzed using digital PCR (dPCR) in tumor and adjacent non-tumor tissue samples of 114 lung cancer patients. The difference between tumor and non-tumor samples were statistically significant (p < 0.0001) for TERT and MYC. Using a predefined specificity of 99% a sensitivity of 41% and 51% was observed for TERT and MYC, respectively. For the combination of TERT and MYC the overall sensitivity increased to 60% at 99% specificity. We demonstrated that a combination of markers increases the performance in comparison to individual markers. Additionally, the determination of CNV using dPCR might be an appropriate tool in precision medicine.
Collapse
Affiliation(s)
- Alexander Brik
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany.
| | - Katharina Wichert
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Daniel G Weber
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Katja Szafranski
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Peter Rozynek
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Swetlana Meier
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Yon-Dschun Ko
- Department of Internal Medicine, Johanniter-Kliniken Bonn GmbH, Bonn, Germany
| | - Reinhard Büttner
- Institute of Pathology, Medical Faculty and Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Klaus Gerwert
- Center for Protein Diagnostics (PRODI), Department of Biophysics, Ruhr University Bochum, Bochum, Germany
| | - Thomas Behrens
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Georg Johnen
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| |
Collapse
|
5
|
Jiang B, Hu L, Dong D, Guo Z, Wei W, Wang C, Shao W, Ma T, Chen Y, Li Q, Hu W. TP53 or CDKN2A/B covariation in ALK/RET/ROS1-rearranged NSCLC is associated with a high TMB, tumor immunosuppressive microenvironment and poor prognosis. J Cancer Res Clin Oncol 2023; 149:10041-10052. [PMID: 37261522 DOI: 10.1007/s00432-023-04924-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
INTRODUCTION ALK-rearranged lung adenocarcinomas with TP53 mutations have more unstable genomic features, poorer ALK-TKI efficacy and a worse prognosis than ALK-rearranged lung adenocarcinomas with wild-type TP53. Here, we examine the gene variations that co-occur with ALK/RET/ROS1 rearrangements in NSCLC and the corresponding tumor immune microenvironment, as well as their association with prognosis. METHODS A total of 155 patients with ALK/RET/ROS1 fusions were included retrospectively. Tumor genome mutation analysis was performed by next-generation sequencing. PD-L1 expression and tumor-infiltrating lymphocytes were assessed by multiplex immunohistochemistry. The correlations among gene covariation, the tumor immune microenvironment, and clinicopathological characteristics were analyzed. RESULTS Among the 155 patients, concomitant TP53 mutation appeared most frequently (31%), followed by CDKN2A/B copy number loss (15%). The ALK/RET/ROS1 fusion and TP53 or CDKN2A/B covariation group had more males and patients with stage IV disease (p < 0.001, p = 0.0066). Patients with TP53 or CDKN2A/B co-occurrence had higher tumor mutation burdens and more neoantigens (p < 0.001, p = 0.0032). PD-L1 expression was higher in the tumor areas of the TP53 or CDKN2A/B co-occurring group (p = 0.00038). However, the levels of CD8+, CD8+PD1-, and CD8+PD-L1- TILs were lower in the tumor areas of this group (p = 0.043, p = 0.029, p = 0.025). In the TCGA NSCLC cohorts, the top 2 mutated genes were CDKN2A/B (24%) and TP53 (16%). The TP53 or CDKN2A/B co-occurring group had higher tumor mutation burdens and shorter OS (p < 0.001, p < 0.001). CONCLUSIONS Patients with co-occurring TP53/CDKN2A/B variations and ALK/RET/ROS1 rearrangements are associated with high TMB, more neoantigens, an immunosuppressive microenvironment and a worse prognosis.
Collapse
Affiliation(s)
- Bin Jiang
- Department of Cardiothoracic Surgery, Guiqian International General Hospital, Guiyang, 550000, China
| | - Liwen Hu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Daling Dong
- Department of Cardiothoracic Surgery, Guiqian International General Hospital, Guiyang, 550000, China
| | - Zixin Guo
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Wei Wei
- Department of Cardiothoracic Surgery, Guiqian International General Hospital, Guiyang, 550000, China
| | - Chao Wang
- Department of Cardiothoracic Surgery, Guiqian International General Hospital, Guiyang, 550000, China
| | - Weikang Shao
- Genecast Biotechnology Co., Ltd., Wuxi, 214000, China
| | - Ting Ma
- Genecast Biotechnology Co., Ltd., Wuxi, 214000, China
| | - Yanhui Chen
- Genecast Biotechnology Co., Ltd., Wuxi, 214000, China
| | - Qingyun Li
- Genecast Biotechnology Co., Ltd., Wuxi, 214000, China
| | - Weidong Hu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, China.
| |
Collapse
|
6
|
Malapelle U, Tabbò F, Muscarella LA. Editorial: Concomitant pathogenic mutations in oncogene-driven subgroups: when next generation biology meets targeted therapy in NSCLC. Front Oncol 2023; 13:1239304. [PMID: 37427122 PMCID: PMC10325716 DOI: 10.3389/fonc.2023.1239304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/13/2023] [Indexed: 07/11/2023] Open
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Fabrizio Tabbò
- SC Oncologia ASLCN2 Alba e BRA, PO Michele e Pietro Ferrero, Verduno, Italy
| | - Lucia Anna Muscarella
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
7
|
Han Q, Zhang Z, He X, Chen M, Pang X, Chen C, Du T, Zhang H. Primary inflammatory myofibroblastic tumour of the liver: a clinicopathological and genetic study including a subset with ETV6::NTRK3 fusion. Histopathology 2023; 82:925-936. [PMID: 36748182 DOI: 10.1111/his.14881] [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: 10/25/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023]
Abstract
AIMS Inflammatory myofibroblastic tumour (IMT) is an intermediate neoplasm and rarely occurs in the liver. The aim of this study was to analyse the clinicopathological and genetic features of the largest primary hepatic IMT. METHODS AND RESULTS A total of 10 cases were identified (four males and six females aged 1-48 years, median = 35 years) from 2011 to 2021, which accounted for 2.5% of IMTs occurring in all organ systems. Histological findings revealed that myofibroblastic/fibroblastic cells with inflammatory infiltration and focal hypocellularity were observed in three children. Immunostaining showed ALK-diffuse cytoplasmic positive in six cases (six of 10; 60%) and pan-TRK nuclear positive in three cases (three of 10; 30%). Hypercellular pattern was detected in ALK-positive IMTs and obvious collagenous/myxoid matrix was observed in the pan-TRK-positive subgroup. ALK rearrangement was demonstrated in three of five interpretable ALK-positive IMTs by fluorescence in-situ hybridisation (FISH), and one case failed due to poor sample quality. Next-generation sequencing indicated an IMT with TFG::ALK and FCHSD2::ALK fusion and TP53 mutation. ETV6::NTRK3 fusion was confirmed by RT-PCR, but FISH-negative results were found in two of three cases with pan-TRK-positive IMTs. No genetic alteration was detected in one tumour. One patient died 1 year after biopsy, while nine patients survived without evidence of disease in the follow-up surveillance (17-119 months). CONCLUSIONS This article describes the first example of primary paediatric hepatic IMTs with ETV6::NTRK3 fusion. Besides the common ALK-positive subgroup, the proportion of NTRK3 fusion is high. Recognising the association between clinicopathological and molecular alterations is critical to accurate diagnosis of hepatic IMTs.
Collapse
Affiliation(s)
- Qianqian Han
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhang Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin He
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaojun Pang
- Department of Pathology, Mian yang Hospital of Traditional Chinese Medicine, Mian yang, Sichuan, China
| | - Chen Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianhai Du
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongying Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Guan M, Xu J, Shi Q. Molecular determinants of clinical outcomes for anaplastic lymphoma kinase-positive non-small cell lung cancer in Chinese patients: A retrospective study. Cancer Genet 2023; 270-271:32-38. [PMID: 36502690 DOI: 10.1016/j.cancergen.2022.11.005] [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: 02/19/2022] [Revised: 10/30/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022]
Abstract
Gene complexity affects the clinical outcomes of anaplastic lymphoma kinase (ALK)-rearranged non-small cell lung cancer (NSCLC). Here, we reviewed the medical records of patients with NSCLC between September 2015 and December 2020 in a single institution. We examined the clinical and genomic predictors of these outcomes using multivariate Cox proportional hazards analysis. Overall, 105 patients with ALK-rearranged NSCLC were included. Echinoderm microtubule-associated protein-like 4 (EML4) was the predominant fusion partner (96.2%). Five patients (4.8%) had non-EML4 fusion partners; three had novel partners. EML4::ALK variant 3 (36.5%) was predominant. One patient had the following three subtypes: E13::A20, E6ins33::A20, and E20::A20. Median progression-free survival (PFS), but not overall survival (OS), was significantly different between patients with variants 3 and 1. TP53 was the most common concomitant mutation (21.4%). The presence of TP53 mutations was associated with shorter PFS among patients who received ALK-TKI. Patients with concomitant oncogene mutations presented significantly shorter OS and PFS than those with only ALK rearrangement. In a multivariate Cox regression model, concomitant oncogene mutations and variant 3 carrier status were prognostic factors for PFS, whereas baseline brain metastasis was a prognostic factor for OS.
Collapse
Affiliation(s)
- Maojing Guan
- Department of Oncology, Anhui Chest Hospital, 397 Jixi Road, Hefei 230022, China; Anhui Medical University Clinical College of Chest, 397 Jixi Road, Hefei 230022, China
| | - Jianping Xu
- Department of Pathology, Anhui Chest Hospital, 397 Jixi Road, Hefei 230022, China; Anhui Medical University Clinical College of Chest, 397 Jixi Road, Hefei 230022, China
| | - Qingming Shi
- Department of Oncology, Anhui Chest Hospital, 397 Jixi Road, Hefei 230022, China; Anhui Medical University Clinical College of Chest, 397 Jixi Road, Hefei 230022, China.
| |
Collapse
|
9
|
Cao J, J Gu J, Liang Y, Wang B. Evaluate the Prognosis of MYC/TP53 Comutation in Chinese Patients with EGFR-Positive Advanced NSCLC Using Next-Generation Sequencing: A Retrospective Study. Technol Cancer Res Treat 2022; 21:15330338221138213. [PMID: 36524293 PMCID: PMC9761218 DOI: 10.1177/15330338221138213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Purpose: The purpose of this study was to investigate the effect of MYC and TP53 comutations on the clinical efficacy of EGFR tyrosine kinase inhibitors (TKIs) in Chinese patients with advanced EGFR-positive nonsmall-cell lung cancer (NSCLC). Patients and methods: Tissue samples and information from 65 patients with advanced NSCLC in Northern Jiangsu People's Hospital were collected and analyzed by next-generation sequencing (NGS). Progression-free survival (PFS) and total survival (OS) were the main endpoints, and the objective response rate (ORR) and disease control rate (DCR) were the secondary endpoints. Result: Among 65 patients, 17 had TP53 and MYC wild-type mutations (WT/WT), 36 had TP53 mutant and MYC wild-type mutations (TP53/WT), and 12 had coexisting MYC/TP53 mutations (MYC/TP53). When 12 patients with MYC/TP53 comutation were compared with the other two groups (TP53/WT, WT/WT), mPFS and mOS are significantly lower than those in the other two groups (mPFS: 4.1 months vs 6.0 months, 12.3 months, HR: 0.769, 95% CI: 4.592-7.608, P = .047. mOS: 14.6 months vs 24.1 months, 31.5 months, HR: 3.170, 95% CI: 18.786-31.214, P < .001), and the ORR, DCR of patients with MYC/TP53 comutation was lower than that of the other two groups (ORR, 25% vs 44.4%, 70.6%, P = .045. DCR, 58.3% vs 72.2%, 82.4%, P = .365). Conclusion: Patients with MYC/TP53 comutations with EGFR-positive advanced NSCLC are more likely to develop drug resistance after early treatment with EGFR-TKIs and have a worse clinical outcome.
Collapse
Affiliation(s)
- Jin Cao
- Medical College, Yangzhou
University, Yangzhou, Jiangsu, China
| | - Juan J Gu
- Medical College, Yangzhou
University, Yangzhou, Jiangsu, China,Institute of Oncology, Northern Jiangsu People's
Hospital, Yangzhou, Jiangsu, China,Department of Oncology, Northern Jiangsu People's
Hospital, Yangzhou, Jiangsu, China
| | - Yichen Liang
- Institute of Oncology, Northern Jiangsu People's
Hospital, Yangzhou, Jiangsu, China,Department of Oncology, Northern Jiangsu People's
Hospital, Yangzhou, Jiangsu, China
| | - Buhai Wang
- Medical College, Yangzhou
University, Yangzhou, Jiangsu, China,Institute of Oncology, Northern Jiangsu People's
Hospital, Yangzhou, Jiangsu, China,Department of Oncology, Northern Jiangsu People's
Hospital, Yangzhou, Jiangsu, China,Buhai Wang, MD, PhD, Medical College,
Yangzhou University, Yangzhou, Jiangsu, 225000, China.
Yichen Liang, MD, PhD, Institute of
Oncology, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225000, China.
| |
Collapse
|
10
|
Chen X, Liu T, Wu J, Zhu C, Guan G, Zou C, Guo Q, Ren X, Li C, Cheng P, Cheng W, Wu A. Molecular profiling identifies distinct subtypes across TP53 mutant tumors. JCI Insight 2022; 7:156485. [PMID: 36256461 PMCID: PMC9746906 DOI: 10.1172/jci.insight.156485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 10/14/2022] [Indexed: 01/12/2023] Open
Abstract
Tumor protein 53 mutation (TP53mut) is one of the most important driver events facilitating tumorigenesis, which could induce a series of chain reactions to promote tumor malignant transformation. However, the malignancy progression patterns under TP53 mutation remain less known. Clarifying the molecular landscapes of TP53mut tumors will help us understand the process of tumor development and aid precise treatment. Here, we distilled genetic and epigenetic features altered in TP53mut cancers for cluster-of-clusters analysis. Using integrated classification, we derived 5 different subtypes of TP53mut patients. These subtypes have distinct features in genomic alteration, clinical relevance, microenvironment dysregulation, and potential therapeutics. Among the 5 subtypes, COCA3 was identified as the subtype with worst prognosis, causing an immunosuppressive microenvironment and immunotherapeutic resistance. Further drug efficacy research highlighted olaparib as the most promising therapeutic agents for COCA3 tumors. Importantly, the therapeutic efficacy of olaparib in COCA3 and immunotherapy in non-COCA3 tumors was validated via in vivo experimentation. Our study explored the important molecular events and developed a subtype classification system with distinct targeted therapy strategies for different subtypes of TP53mut tumors. These multiomics classification systems provide a valuable resource that significantly expands the knowledge of TP53mut tumors and may eventually benefit in clinical practice.
Collapse
Affiliation(s)
- Xin Chen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tianqi Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jianqi Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chen Zhu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Gefei Guan
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Cunyi Zou
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qing Guo
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaolin Ren
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Neurosurgery, Shenyang Red Cross Hospital, Shenyang, Liaoning, China
| | - Chen Li
- Department of Orthodontics, Stomatological Hospital of China Medical University, Shenyang, Liaoning, China
| | - Peng Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wen Cheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Anhua Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
11
|
Gray ER, Mordaka JM, Christoforou ER, von Bargen K, Potts ND, Xyrafaki C, Silva AL, Stolarek-Januszkiewicz M, Anton K, Powalowska PK, Andreazza S, Tomassini A, Palmer RN, Cooke A, Osborne RJ, Balmforth BW. Ultra-sensitive molecular detection of gene fusions from RNA using ASPYRE. BMC Med Genomics 2022; 15:215. [PMID: 36224552 PMCID: PMC9555097 DOI: 10.1186/s12920-022-01363-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/23/2022] [Indexed: 12/04/2022] Open
Abstract
Background RNA is a critical analyte for unambiguous detection of actionable mutations used to guide treatment decisions in oncology. Currently available methods for gene fusion detection include molecular or antibody-based assays, which suffer from either being limited to single-gene targeting, lack of sensitivity, or long turnaround time. The sensitivity and predictive value of next generation sequencing DNA-based assays to detect fusions by sequencing intronic regions is variable, due to the extensive size of introns. The required depth of sequencing and input nucleic acid required can be prohibitive; in addition it is not certain that predicted gene fusions are actually expressed. Results Herein we describe a method based on pyrophosphorolysis to include detection of gene fusions from RNA, with identical assay steps and conditions to detect somatic mutations in DNA [1], permitting concurrent assessment of DNA and RNA in a single instrument run. Conclusion The limit of detection was under 6 molecules/ 6 µL target volume. The workflow and instrumentation required are akin to PCR assays, and the entire assay from extracted nucleic acid to sample analysis can be completed within a single day. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01363-0.
Collapse
Affiliation(s)
- Eleanor R Gray
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Justyna M Mordaka
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | | | - Kristine von Bargen
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Nicola D Potts
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Christina Xyrafaki
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Ana-Luisa Silva
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | | | - Katarzyna Anton
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Paulina K Powalowska
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Simonetta Andreazza
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Alessandro Tomassini
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Rebecca N Palmer
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Aishling Cooke
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Robert J Osborne
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Barnaby W Balmforth
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England.
| |
Collapse
|
12
|
Rosenquist R, Cuppen E, Buettner R, Caldas C, Dreau H, Elemento O, Frederix G, Grimmond S, Haferlach T, Jobanputra V, Meggendorfer M, Mullighan CG, Wordsworth S, Schuh A. Clinical utility of whole-genome sequencing in precision oncology. Semin Cancer Biol 2022; 84:32-39. [PMID: 34175442 DOI: 10.1016/j.semcancer.2021.06.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/02/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022]
Abstract
Precision diagnostics is one of the two pillars of precision medicine. Sequencing efforts in the past decade have firmly established cancer as a primarily genetically driven disease. This concept is supported by therapeutic successes aimed at particular pathways that are perturbed by specific driver mutations in protein-coding domains and reflected in three recent FDA tissue agnostic cancer drug approvals. In addition, there is increasing evidence from studies that interrogate the entire genome by whole-genome sequencing that acquired global and complex genomic aberrations including those in non-coding regions of the genome might also reflect clinical outcome. After addressing technical, logistical, financial and ethical challenges, national initiatives now aim to introduce clinical whole-genome sequencing into real-world diagnostics as a rational and potentially cost-effective tool for response prediction in cancer and to identify patients who would benefit most from 'expensive' targeted therapies and recruitment into clinical trials. However, so far, this has not been accompanied by a systematic and prospective evaluation of the clinical utility of whole-genome sequencing within clinical trials of uniformly treated patients of defined clinical outcome. This approach would also greatly facilitate novel predictive biomarker discovery and validation, ultimately reducing size and duration of clinical trials and cost of drug development. This manuscript is the third in a series of three to review and critically appraise the potential and challenges of clinical whole-genome sequencing in solid tumors and hematological malignancies.
Collapse
Affiliation(s)
- Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Genetics, Karolinska University Hospital, Solna, Sweden
| | - Edwin Cuppen
- Hartwig Medical Foundation, Amsterdam, The Netherlands; Center for Molecular Medicine and Oncode Institute, University Medical Center, Utrecht, The Netherlands
| | | | - Carlos Caldas
- Cancer Research UK Cambridge Institute and Department of Oncology, University of Cambridge, United Kingdom
| | - Helene Dreau
- NIHR Oxford Biomedical Research Centre and Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Olivier Elemento
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, United States; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, United States
| | - Geert Frederix
- Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht, The Netherlands
| | - Sean Grimmond
- Centre for Cancer Research, University of Melbourne, Melbourne, Australia
| | | | - Vaidehi Jobanputra
- New York Genome Center, 101 Avenue of the Americas, New York, NY 100132, United States; Columbia University Medical Center, 650 W 168th St, New York, NY 10032, United States
| | | | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, United States
| | - Sarah Wordsworth
- Nuffield Department of Population Health and Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Anna Schuh
- NIHR Oxford Biomedical Research Centre and Department of Oncology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
13
|
Alternative Treatment Options to ALK Inhibitor Monotherapy for EML4-ALK-Driven Lung Cancer. Cancers (Basel) 2022; 14:cancers14143452. [PMID: 35884511 PMCID: PMC9325236 DOI: 10.3390/cancers14143452] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 02/01/2023] Open
Abstract
EML4-ALK is an oncogenic fusion protein that accounts for approximately 5% of NSCLC cases. Targeted inhibitors of ALK are the standard of care treatment, often leading to a good initial response. Sadly, some patients do not respond well, and most will develop resistance over time, emphasizing the need for alternative treatments. This review discusses recent advances in our understanding of the mechanisms behind EML4-ALK-driven NSCLC progression and the opportunities they present for alternative treatment options to ALK inhibitor monotherapy. Targeting ALK-dependent signalling pathways can overcome resistance that has developed due to mutations in the ALK catalytic domain, as well as through activation of bypass mechanisms that utilise the same pathways. We also consider evidence for polytherapy approaches that combine targeted inhibition of these pathways with ALK inhibitors. Lastly, we review combination approaches that use targeted inhibitors of ALK together with chemotherapy, radiotherapy or immunotherapy. Throughout this article, we highlight the importance of alternative breakpoints in the EML4 gene that result in the generation of distinct EML4-ALK variants with different biological and pathological properties and consider monotherapy and polytherapy approaches that may be selective to particular variants.
Collapse
|
14
|
Ubiquitin-Specific Protease 6 n-Terminal-like Protein (USP6NL) and the Epidermal Growth Factor Receptor (EGFR) Signaling Axis Regulates Ubiquitin-Mediated DNA Repair and Temozolomide-Resistance in Glioblastoma. Biomedicines 2022; 10:biomedicines10071531. [PMID: 35884836 PMCID: PMC9312792 DOI: 10.3390/biomedicines10071531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/13/2022] [Accepted: 06/21/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant glioma, with a 30–60% epidermal growth factor receptor (EGFR) mutation. This mutation is associated with unrestricted cell growth and increases the possibility of cancer invasion. Patients with EGFR-mutated GBM often develop resistance to the available treatment modalities and higher recurrence rates. The drug resistance observed is associated with multiple genetic or epigenetic factors. The ubiquitin-specific protease 6 N-terminal-like protein (USP6NL) is a GTPase-activating protein that functions as a deubiquitinating enzyme and regulates endocytosis and signal transduction. It is highly expressed in many cancer types and may promote the growth and proliferation of cancer cells. We hypothesized that USP6NL affects GBM chemoresistance and tumorigenesis, and that its inhibition may be a novel therapeutic strategy for GBM treatment. The USP6NL level, together with EGFR expression in human GBM tissue samples and cell lines associated with therapy resistance, tumor growth, and cancer invasion, were investigated. Its pivotal roles and potential mechanism in modulating tumor growth, and the key mechanism associated with therapy resistance of GBM cells, were studied, both in vitro and in vivo. Herein, we found that deubiquitinase USP6NL and growth factor receptor EGFR were strongly associated with the oncogenicity and resistance of GBM, both in vitro and in vivo, toward temozolomide, as evidenced by enhanced migration, invasion, and acquisition of a highly invasive and drug-resistant phenotype by the GBM cells. Furthermore, abrogation of USP6NL reversed the properties of GBM cells and resensitized them toward temozolomide by enhancing autophagy and reducing the DNA damage repair response. Our results provide novel insights into the probable mechanism through which USP6NL/EGFR signaling might suppress the anticancer therapeutic response, induce cancer invasiveness, and facilitate reduced sensitivity to temozolomide treatment in GBM in an autolysosome-dependent manner. Therefore, controlling the USP6NL may offer an alternative, but efficient, therapeutic strategy for targeting and eradicating otherwise resistant and recurrent phenotypes of aggressive GBM cells.
Collapse
|
15
|
Kwon M, Ku BM, Olsen S, Park S, Lefterova M, Odegaard J, Jung HA, Sun JM, Lee SH, Ahn JS, Park K, Ahn MJ. Longitudinal monitoring by next-generation sequencing of plasma cell-free DNA in ALK rearranged NSCLC patients treated with ALK tyrosine kinase inhibitors. Cancer Med 2022; 11:2944-2956. [PMID: 35437925 PMCID: PMC9359877 DOI: 10.1002/cam4.4663] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 12/11/2022] Open
Abstract
Background Patients with ALK‐rearranged non‐small cell lung cancer (ALK+ NSCLC) inevitably acquire resistance to ALK inhibitors. Longitudinal monitoring of cell‐free plasma DNA (cfDNA) next‐generation sequencing (NGS) could predict the response and resistance to tyrosine kinase inhibitor (TKI) therapy in ALK+ NSCLC. Methods Patients with ALK+ NSCLC determined by standard tissue testing and planned to undergo TKI therapy were prospectively recruited. Plasma was collected at pretreatment, 2 months‐post therapy, and at progression for cfDNA‐NGS analysis, Guardant 360. Results Among 92 patients enrolled, circulating tumor DNA (ctDNA) was detected in 69 baseline samples (75%): 43 ALK fusions (62.3%) and two ALK mutations without fusion (2.8%). Two patients showed ALK‐resistance mutations after ceritinib; G1202R, and co‐occurring G1202R and T1151R. Eight patients developed ALK resistance mutations after crizotinib therapy; L1196M (n = 5), G1269A (n = 1), G1202R (n = 1), and co‐occurring F1174L, G1202R, and G1269A (n = 1). Absence of ctDNA at baseline was significantly associated with longer progression‐free survival (PFS; median 36.1 vs. 11.4 months, p = 0.0049) and overall survival (OS; not reached vs. 29.3 months, p = 0.0200). ctDNA clearance at 2 months (n = 29) was associated with significantly longer PFS (25.4 vs. 11.6 months, p = 0.0012) and OS (not reached vs. 26.1 months, p = 0.0307) than those without clearance (n = 22). Patients with co‐occurring TP53 alterations and ALK fusions at baseline (n = 16) showed significantly shorter PFS (7.28 vs. 13.0 months, p = 0.0307) than those without TP53 alterations (n = 25). Conclusions cfDNA‐NGS facilitates detection of ALK fusions and resistance mutations, assessment of prognosis, and monitoring dynamic changes of genomic alterations in ALK+ NSCLC treated with ALK‐TKI.
Collapse
Affiliation(s)
- Minsuk Kwon
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, South Korea
| | - Bo Mi Ku
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Steve Olsen
- Department of Medical and Clinical Affairs, Guardant Health AMEA, Singapore, Singapore
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Martina Lefterova
- Department of Medical and Clinical Affairs, Guardant Health AMEA, Singapore, Singapore
| | - Justin Odegaard
- Department of Medical and Clinical Affairs, Guardant Health AMEA, Singapore, Singapore
| | - Hyun-Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
16
|
Tu Y, Yao S, Chen Q, Li W, Song Y, Zhang P. 5-Hydroxytryptamine activates a 5-HT/c-Myc/SLC6A4 signaling loop in non–small cell lung cancer. Biochim Biophys Acta Gen Subj 2022; 1866:130093. [DOI: 10.1016/j.bbagen.2022.130093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 10/19/2022]
|
17
|
Meldrum K, Evans SJ, Vogel U, Tran L, Doak SH, Clift MJD. The influence of exposure approaches to in vitro lung epithelial barrier models to assess engineered nanomaterial hazard. Nanotoxicology 2022; 16:114-134. [PMID: 35343373 DOI: 10.1080/17435390.2022.2051627] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Exposure to engineered nanomaterials (ENM) poses a potential health risk to humans through long-term, repetitive low-dose exposures. Currently, this is not commonplace within in vitro lung cell cultures. Therefore, the purpose of this study was to consider the optimal exposure approach toward determining the stability, sensitivity and validity of using in vitro lung cell mono- and co-cultures to determine ENM hazard. A range of exposure scenarios were conducted with DQ12 (previously established as a positive particle control) (historic and re-activated), TiO2 (JRC NM-105) and BaSO4 (JRC NM-220) on both monocultures of A549 cells as well as co-cultures of A549 cells and differentiated THP-1 cells. Cell cultures were exposed to either a single, or a repeated exposure over 24, 48- or 72-hours at in vivo extrapolated concentrations of 0-5.2 µg/cm2, 0-6 µg/cm2 and 0-1µg/cm2. The focus of this study was the pro-inflammatory, cytotoxic and genotoxic response elicited by these ENMs. Exposure to DQ12 caused pro-inflammatory responses after 48 hours repeat exposures, as well as increases in micronucleus frequency. Neither TiO2 nor BaSO4 elicited a pro-inflammatory response at this time point. However, there was induction of IL-6 after 24 hours TiO2 exposure. In conclusion, it is important to consider the appropriateness of the positive control implemented, the cell culture model, the time of exposure as well as the type of exposure (bolus or fractionated) before establishing if an in vitro model is appropriate to determine the level of response to the specific ENM of interest.
Collapse
Affiliation(s)
- Kirsty Meldrum
- In Vitro Toxicology Group, Swansea University, Swansea, UK
| | | | - Ulla Vogel
- The National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Lang Tran
- Institute of Occupational Medicine (IOM), Edinburgh, UK
| | - Shareen H Doak
- In Vitro Toxicology Group, Swansea University, Swansea, UK
| | | |
Collapse
|
18
|
Janning M, Süptitz J, Albers-Leischner C, Delpy P, Tufman A, Velthaus-Rusik JL, Reck M, Jung A, Kauffmann-Guerrero D, Bonzheim I, Brändlein S, Hummel HD, Wiesweg M, Schildhaus HU, Stratmann JA, Sebastian M, Alt J, Buth J, Esposito I, Berger J, Tögel L, Saalfeld FC, Wermke M, Merkelbach-Bruse S, Hillmer AM, Klauschen F, Bokemeyer C, Buettner R, Wolf J, Loges S. Treatment outcome of atypical EGFR mutations in the German National Network Genomic Medicine Lung Cancer (nNGM). Ann Oncol 2022; 33:602-615. [PMID: 35263633 DOI: 10.1016/j.annonc.2022.02.225] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Atypical EGFR mutations occur in 10-30% of NSCLC patients with EGFR mutations and their sensitivity to classical EGFR-tyrosine kinase inhibitors (TKI) is highly heterogeneous. Patients harboring one group of uncommon, recurrent EGFR mutations (G719X, S768I, L861Q) respond to EGFR-TKI. Exon 20 insertions are mostly insensitive to EGFR-TKI but display sensitivity to exon 20 inhibitors. Clinical outcome data of patients with very rare point and compound mutations upon systemic treatments are still sparse to date. PATIENTS AND METHODS In this retrospective, multi-center study of the national Network Genomic Medicine (nNGM) in Germany, 856 NSCLC cases with atypical EGFR mutations including co-occuring mutations were reported from 12 centers. Clinical follow-up data after treatment with different EGFR-TKI, chemotherapy and immune checkpoint inhibitors were available from 260 patients. Response to treatment was analyzed in three major groups: (1) uncommon mutations (G719X, S7681, L861Q and combinations), (2) exon 20 insertions and (3) very rare EGFR mutations (very rare single point mutations, compound mutations, exon 18 deletions, exon 19 insertions). RESULTS Our study comprises the largest thus far reported real-world cohort of very rare EGFR single point and compound mutations treated with different systemic treatments. We validated higher efficacy of EGFR-TKI in comparison to chemotherapy in group 1 (uncommon), while most exon 20 insertions (group 2) were not EGFR-TKI responsive. In addition, we found TKI sensitivity of very rare point mutations (group 3) and of complex EGFR mutations containing exon 19 deletions or L858R mutations independent of the combination partner. Notably, treatment responses in group 3 (very rare) were highly heterogeneous. Co-occurring TP53 mutations exerted a non-significant trend for a detrimental effect on outcome in EGFR-TKI treated patients in groups 2 and 3 but not in group 1. CONCLUSIONS Based on our findings we propose a novel nNGM classification of uncommon EGFR mutations.
Collapse
Affiliation(s)
- M Janning
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. https://in.linkedin.com/linkedin.com/in/melanie-janning-a48a32153
| | - J Süptitz
- Department of Internal Medicine I, Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - C Albers-Leischner
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - P Delpy
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Federated Information Systems, German Cancer Research Center (DKFZ), Heidelberg, Germany; Complex Data Processing in Medical Informatics, University Medical Centre Mannheim, Mannheim, Germany
| | - A Tufman
- Department of Internal Medicine V, University of Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - J-L Velthaus-Rusik
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - M Reck
- LungenClinic Grosshansdorf, Thoracic Oncology, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany
| | - A Jung
- Pathology Institute, Ludwig Maximilians University of Munich, Munich, Germany; German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - D Kauffmann-Guerrero
- Department of Internal Medicine V, University of Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - I Bonzheim
- Institute of Pathology and Neuropathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - S Brändlein
- Institute of Pathology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - H-D Hummel
- Translational Oncology/Early Clinical Trial Unit (ECTU), Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| | - M Wiesweg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - H-U Schildhaus
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - J A Stratmann
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany
| | - M Sebastian
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany
| | - J Alt
- Department of Hematology, Medical Oncology & Pneumology, University Medical Center Mainz, Mainz, Germany
| | - J Buth
- Institute of Pathology, Heinrich Heine University and University Hospital of Duesseldorf, Duesseldorf, Germany
| | - I Esposito
- Institute of Pathology, Heinrich Heine University and University Hospital of Duesseldorf, Duesseldorf, Germany
| | - J Berger
- Charité Comprehensive Cancer Center, Berlin, Germany
| | - L Tögel
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - F C Saalfeld
- Clinic for Internal Medicine I, University Hospital, TU Dresden, Dresden, Germany
| | - M Wermke
- Clinic for Internal Medicine I, University Hospital, TU Dresden, Dresden, Germany
| | - S Merkelbach-Bruse
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - A M Hillmer
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - F Klauschen
- Pathology Institute, Ludwig Maximilians University of Munich, Munich, Germany; German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - R Buettner
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - J Wolf
- Department of Internal Medicine I, Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - S Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
19
|
Rivera-Concepcion J, Uprety D, Adjei AA. Challenges in the Use of Targeted Therapies in NSCLC. Cancer Res Treat 2022; 54:315-329. [PMID: 35209703 PMCID: PMC9016301 DOI: 10.4143/crt.2022.078] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 11/21/2022] Open
Abstract
Precision oncology has fundamentally changed how we diagnose and treat cancer. In recent years, there has been a significant change in the management of patients with oncogene-addicted advanced-stage non–small cell lung cancer (NSCLC). Increasing amounts of identifiable oncogene drivers have led to the development of molecularly targeted drugs. Undoubtedly, the future of thoracic oncology is shifting toward increased molecular testing and the use of targeted therapies. For the most part, these novel drugs have proven to be safe and effective. As with all great innovations, targeted therapies pose unique challenges. Drug toxicities, resistance, access, and costs are some of the expected obstacles that will need to be addressed. This review highlights some of the major challenges in the use of targeted therapies in NSCLC and provides guidance for the future strategies.
Collapse
|
20
|
Druzhinin VG, Baranova ED, Volobaev VP, Ivanov VI, Larionov AV, Minina VI, Smagulova F, Legoff L, Titov VA, Fucic A. The Length of Telomeres and the Baseline Level of Cytogenetic Damage in Leukocytes of Lung Cancer Patients. RUSS J GENET+ 2022. [DOI: 10.1134/s1022795422010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Noh KW, Buettner R, Klein S. Shifting Gears in Precision Oncology-Challenges and Opportunities of Integrative Data Analysis. Biomolecules 2021; 11:biom11091310. [PMID: 34572523 PMCID: PMC8465238 DOI: 10.3390/biom11091310] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
For decades, research relating to modification of host immunity towards antitumor response activation has been ongoing, with the breakthrough discovery of immune-checkpoint blockers. Several biomarkers with potential predictive value have been reported in recent studies for these novel therapies. However, with the plethora of therapeutic options existing for a given cancer entity, modern oncology is now being confronted with multifactorial interpretation to devise “the best therapy” for the individual patient. Into the bargain come the multiverse guidelines for established and emerging diagnostic biomarkers, as well as the complex interplay between cancer cells and tumor microenvironment, provoking immense challenges in the therapy decision-making process. Through this review, we present various molecular diagnostic modalities and techniques, such as genomics, immunohistochemistry and quantitative image analysis, which have the potential of becoming powerful tools in the development of an optimal treatment regime when analogized with patient characteristics. We will summarize the underlying complexities of these methods and shed light upon the necessary considerations and requirements for data integration. It is our hope to provide compelling evidence to emphasize on the need for inclusion of integrative data analysis in modern cancer therapy, and thereupon paving a path towards precision medicine and better patient outcomes.
Collapse
Affiliation(s)
- Ka-Won Noh
- Institute for Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (K.-W.N.); (R.B.)
| | - Reinhard Buettner
- Institute for Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (K.-W.N.); (R.B.)
| | - Sebastian Klein
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, 48149 Münster, Germany
- Correspondence: ; Tel.: +49-251-83-57670
| |
Collapse
|
22
|
Liu D, Xu X, Wen J, Zhang C, Fan M. Identification of a EML4-ALK exon 19 fusion variant in lung adenocarcinoma and alectinib resistance. Lung Cancer 2021; 160:32-35. [PMID: 34391065 DOI: 10.1016/j.lungcan.2021.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 01/13/2023]
Abstract
Alectinib, a highly selective inhibitor of anaplastic lymphoma kinase (ALK), has shown a high response rate and long progression-free survival in primary treatment of ALK-positive non-small-cell lung cancer (NSCLC). De novo resistance or refractory subtype is rare event. Herein, we identify the first case with serial next-generation sequencing (NGS) results that harboured a rare echinoderm microtubule associated protein like 4 gene (EML4) -ALK (breaking site at exon 19) fusion in a lung adenocarcinoma (LUAD) patient who acquired alectinib resistance rapidly (less than 3 months), followed by multi-drug resistance and short survival time.
Collapse
Affiliation(s)
- Di Liu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, People's Republic of China
| | - Xinyan Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, People's Republic of China
| | - Junmiao Wen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, People's Republic of China
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska, Lincoln, NE, USA.
| | - Min Fan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, People's Republic of China.
| |
Collapse
|
23
|
Yang L, Li N, Wang M, Zhang YH, Yan LD, Zhou W, Yu ZQ, Peng XC, Cai J. Tumorigenic effect of TERT and its potential therapeutic target in NSCLC (Review). Oncol Rep 2021; 46:182. [PMID: 34278503 DOI: 10.3892/or.2021.8133] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/25/2021] [Indexed: 12/24/2022] Open
Abstract
Non‑small cell lung cancer (NSCLC), which accounts for ~85% of all lung cancer cases, is commonly diagnosed at an advanced stage and has a high patient mortality rate. Despite the increasing availability of treatment strategies, the prognosis of patients with NSCLC remains poor, with a low 5‑year survival rate. This poor prognosis may be associated with the tumor heterogeneity of NSCLC, as well as its acquisition and intrinsic resistance to therapeutic drugs. It has been suggested that combination therapy with telomerase inhibition may be an effective strategy for the treatment of drug‑sensitive and drug‑resistant types of cancer. Telomerase is the key enzyme for cell survival, and ~90% of human cancers maintain telomeres by activating telomerase, which is driven by the upregulation of telomerase reverse transcriptase (TERT). Several mechanisms of telomerase reactivation have been described in a variety of cancer types, including TERT promoter mutation, epigenetic modifications via a TERT promoter, TERT amplification, and TERT rearrangement. The aim of the present study was to comprehensively review telomerase activity and its association with the clinical characteristics and prognosis of NSCLC, as well as analyze the potential mechanism via which TERT activates telomerase and determine its potential clinical application in NSCLC. More importantly, current treatment strategies targeting TERT in NSCLC have been summarized with the aim to promote discovery of novel strategies for the future treatment of NSCLC.
Collapse
Affiliation(s)
- Liu Yang
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Na Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Meng Wang
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Yan-Hua Zhang
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Lu-Da Yan
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Wen Zhou
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Zhi-Qiong Yu
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Jun Cai
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| |
Collapse
|
24
|
He F, Song K, Guan G, Huo J, Xin Y, Li T, Liu C, Zhu Q, Fan N, Guo Y, Wu L. The Phenomenon of Gene Rearrangement is Frequently Associated with TP53 Mutations and Poor Disease-Free Survival in Hepatocellular Carcinoma. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2021; 14:723-736. [PMID: 34188519 PMCID: PMC8233541 DOI: 10.2147/pgpm.s313848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/03/2021] [Indexed: 01/05/2023]
Abstract
Purpose Gene rearrangements (GRs) have been reported to be related to adverse prognosis in some tumours, but the relationship in hepatocellular carcinoma (HCC) remains less studied. The objective of our study was to explore the clinicopathological characteristics and prognosis of HCC patients (HCCs) with GRs (GR-HCCs). Patients and Methods This retrospective study included 297 HCCs who underwent hepatectomy and had their tumours sequenced by next-generation sequencing. Categorical variables between groups were compared by the chi-square test. The impact of variables on disease-free survival (DFS) and survival after relapse (SAR) was analysed by the Kaplan–Meier method and Cox regression. Results We observed four repetitive GR events in 297 HCCs: BRD9/TERT, ARID2/intergenic, CDKN2A/intergenic and OBSCN truncation. GR-HCCs frequently presented with low tumour differentiation, tumour necrosis, microvascular invasion, elevated AFP and gene mutations (TP53, NTRK3 and BRD9). The 1-, 2-, and 3-year cumulative DFS rates in GR-HCCs were 45.1%, 31.9%, 31.9%, respectively, which were significantly lower than those of GR-negative HCCs (NGR-HCCs) (72.5%, 57.9%, and 49.0%, respectively; P = 0.001). GR was identified as an independent risk factor for inferior DFS in HCCs (HR = 1.980, 95% CI = 1.246–3.147; P = 0.004). However, there was no significant difference in SAR between GR-HCCs and NGR-HCCs receiving targeted therapy or immunotherapy. Conclusion GR is frequently associated with TP53 mutations and significantly affects DFS following radical resection for HCC. We recommend that GR-HCCs should be closely followed up as a high-risk group for postoperative recurrence.
Collapse
Affiliation(s)
- Fu He
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China.,Department of Clinical Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Kangjian Song
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China.,Department of Clinical Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Ge Guan
- Organ Transplant Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Junyu Huo
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China.,Department of Clinical Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Yang Xin
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Tianxiang Li
- Organ Transplant Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Chao Liu
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Qingwei Zhu
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China.,Department of Clinical Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Ning Fan
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Yuan Guo
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Liqun Wu
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| |
Collapse
|
25
|
Sánchez-Herrero E, Serna-Blasco R, Ivanchuk V, García-Campelo R, Dómine Gómez M, Sánchez JM, Massutí B, Reguart N, Camps C, Sanz-Moreno S, Calabuig-Fariñas S, Jantus-Lewintre E, Arnal M, Fernández-Orth D, Calvo V, González-Rumayor V, Provencio M, Romero A. NGS-based liquid biopsy profiling identifies mechanisms of resistance to ALK inhibitors: a step toward personalized NSCLC treatment. Mol Oncol 2021; 15:2363-2376. [PMID: 34058070 PMCID: PMC8410554 DOI: 10.1002/1878-0261.13033] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/21/2021] [Accepted: 05/28/2021] [Indexed: 01/08/2023] Open
Abstract
Despite impressive and durable responses, nonsmall cell lung cancer (NSCLC) patients treated with anaplastic lymphoma kinase (ALK) inhibitors (ALK-Is) ultimately progress due to development of resistance. Here, we have evaluated the clinical utility of circulating tumor DNA (ctDNA) profiling by next-generation sequencing (NGS) upon disease progression. We collected 26 plasma and two cerebrospinal fluid samples from 24 advanced ALK-positive NSCLC patients at disease progression to an ALK-I. These samples were analyzed by NGS and digital PCR. A tool to retrieve variants at the ALK locus was developed (VALK tool). We identified at least one resistance mutation in the ALK locus in ten (38.5%) plasma samples; the G1269A and G1202R mutations were the most prevalent among patients progressing to first- and second-generation ALK-Is, respectively. Overall, 61 somatic mutations were detected in 14 genes: TP53, ALK, PIK3CA, SMAD4, MAP2K1 (MEK1), FGFR2, FGFR3, BRAF, EGFR, IDH2, MYC, MET, CCND3, and CCND1. Specifically, a deletion in exon 19 in EGFR, a non-V600 BRAF mutation (G466V), and the F129L mutation in MAP2K1 were identified in four patients who showed no objective survival benefit from ALK-Is. Potential ALK-I-resistance mutations were also found in PIK3CA and IDH2. Finally, a c-MYC gain, along with a loss of CCND1 and FGFR3, was detected in a patient progressing on a first-line treatment with crizotinib. We conclude that NGS analysis of liquid biopsies upon disease progression identified different putative ALK-I-resistance mutations in most cases and could be a valuable approach for therapy decision making.
Collapse
Affiliation(s)
- Estela Sánchez-Herrero
- Liquid Biopsy Laboratory, Biomedical Sciences Research Institute Puerta de Hierro-Majadahonda, Spain.,Atrys Health, Barcelona, Spain
| | - Roberto Serna-Blasco
- Liquid Biopsy Laboratory, Biomedical Sciences Research Institute Puerta de Hierro-Majadahonda, Spain
| | - Vadym Ivanchuk
- Liquid Biopsy Laboratory, Biomedical Sciences Research Institute Puerta de Hierro-Majadahonda, Spain
| | | | - Manuel Dómine Gómez
- Medical Oncology Department, Hospital Universitario Fundación Jiménez Díaz, Oncohealth Institute, Universidad Autónoma de Madrid, Spain
| | - José M Sánchez
- Medical Oncology Department, Hospital La Princesa, Madrid, Spain
| | - Bartomeu Massutí
- Medical Oncology Department, Hospital Universitario de Alicante, ISABIAL, Alicante, Spain
| | - Noemi Reguart
- Medical Oncology Department, Hospital Clinic of Barcelona, Spain
| | - Carlos Camps
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Spain.,CIBERONC, Valencia, Spain.,Department of Medical Oncology, Hospital General Universitario de Valencia, Spain.,Department of Medicine, Universitat de València, Spain
| | - Sandra Sanz-Moreno
- Liquid Biopsy Laboratory, Biomedical Sciences Research Institute Puerta de Hierro-Majadahonda, Spain
| | - Silvia Calabuig-Fariñas
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Spain.,CIBERONC, Valencia, Spain.,Department of Pathology, Universitat de València, Spain
| | - Eloísa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Spain.,CIBERONC, Valencia, Spain.,Department of Biotechnology, Universitat de València, Spain
| | - Magdalena Arnal
- MARGenomics, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | | | - Virginia Calvo
- Medical Oncology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Spain
| | | | - Mariano Provencio
- Liquid Biopsy Laboratory, Biomedical Sciences Research Institute Puerta de Hierro-Majadahonda, Spain.,Medical Oncology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Spain
| | - Atocha Romero
- Liquid Biopsy Laboratory, Biomedical Sciences Research Institute Puerta de Hierro-Majadahonda, Spain.,Medical Oncology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Spain
| |
Collapse
|
26
|
Gregorc V, Lazzari C, Mandalá M, Ippati S, Bulotta A, Cangi MG, Khater A, Viganò MG, Mirabile A, Pecciarini L, Ogliari FR, Arrigoni G, Grassini G, Veronesi G, Doglioni C. Intratumoral Cellular Heterogeneity: Implications for Drug Resistance in Patients with Non-Small Cell Lung Cancer. Cancers (Basel) 2021; 13:cancers13092023. [PMID: 33922215 PMCID: PMC8122731 DOI: 10.3390/cancers13092023] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The number of druggable tumor-specific molecular alterations in the treatment of non-small cell lung cancer (NSCLC) has grown significantly in the past decade. Emerging technologies such as liquid biopsy and single-cell methods allow for studying targetable drivers and develop personalized treatments. However, although new therapies confer prolonged disease control and high tumor response rates, most patients eventually progress on targeted treatments. Intratumoral heterogeneity is a frequent event in NSCLC, driving the tumor cells to develop adaptive or new resistance mechanisms within the drug environment. This review summarizes the current and upcoming research on the biological role of tumor heterogeneity, highlighting the link between early and acquired drug resistance and tumoral heterogeneity in targetable driver mutated NSCLC. Abstract Tailored therapies based on the identification of molecular targets currently represent a well-established therapeutic scenario in the treatment of non-small cell lung cancer (NSCLC) patients. However, while aiming to improve patients’ response to therapy, development of resistance is frequently observed in daily clinical practice. Intratumoral heterogeneity is a frequent event in NSCLC, responsible for several critical issues in patients’ diagnosis and treatment. Advances in single-cell sequencing technologies have allowed in-depth profiling of tumors and attributed intratumoral heterogeneity to genetic, epigenetic, and protein modification driven diversities within cancer cell populations. This review highlights current research on the biological role of tumor heterogeneity and its impact on the development of acquired resistance in NSCLC patients.
Collapse
Affiliation(s)
- Vanesa Gregorc
- Department of Oncology, IRCCS San Raffaele, 20132 Milan, Italy; (C.L.); (S.I.); (A.B.); (M.G.V.); (A.M.); (F.R.O.)
- Correspondence:
| | - Chiara Lazzari
- Department of Oncology, IRCCS San Raffaele, 20132 Milan, Italy; (C.L.); (S.I.); (A.B.); (M.G.V.); (A.M.); (F.R.O.)
| | - Mario Mandalá
- Division of Pathological Anatomy, Papa Giovanni XXIII Hospital, 24100 Bergamo, Italy;
- Unit of Medical Oncology, University of Perugia, 06123 Perugia, Italy
| | - Stefania Ippati
- Department of Oncology, IRCCS San Raffaele, 20132 Milan, Italy; (C.L.); (S.I.); (A.B.); (M.G.V.); (A.M.); (F.R.O.)
| | - Alessandra Bulotta
- Department of Oncology, IRCCS San Raffaele, 20132 Milan, Italy; (C.L.); (S.I.); (A.B.); (M.G.V.); (A.M.); (F.R.O.)
| | - Maria Giulia Cangi
- Pathology Unit, San Raffaele Scientific Institute, IRCCS, 20132 Milan, Italy; (M.G.C.); (L.P.); (G.A.); (G.G.); (C.D.)
| | - Abdelrahman Khater
- San Raffaele Hospital, IRCCS, University Vita Salute, 20132 Milan, Italy;
| | - Maria Grazia Viganò
- Department of Oncology, IRCCS San Raffaele, 20132 Milan, Italy; (C.L.); (S.I.); (A.B.); (M.G.V.); (A.M.); (F.R.O.)
| | - Aurora Mirabile
- Department of Oncology, IRCCS San Raffaele, 20132 Milan, Italy; (C.L.); (S.I.); (A.B.); (M.G.V.); (A.M.); (F.R.O.)
| | - Lorenza Pecciarini
- Pathology Unit, San Raffaele Scientific Institute, IRCCS, 20132 Milan, Italy; (M.G.C.); (L.P.); (G.A.); (G.G.); (C.D.)
| | - Francesca Rita Ogliari
- Department of Oncology, IRCCS San Raffaele, 20132 Milan, Italy; (C.L.); (S.I.); (A.B.); (M.G.V.); (A.M.); (F.R.O.)
| | - Gianluigi Arrigoni
- Pathology Unit, San Raffaele Scientific Institute, IRCCS, 20132 Milan, Italy; (M.G.C.); (L.P.); (G.A.); (G.G.); (C.D.)
| | - Greta Grassini
- Pathology Unit, San Raffaele Scientific Institute, IRCCS, 20132 Milan, Italy; (M.G.C.); (L.P.); (G.A.); (G.G.); (C.D.)
| | - Giulia Veronesi
- Division of Thoracic Surgery, San Raffaele Scientific Institute, IRCCS, 20132 Milan, Italy;
| | - Claudio Doglioni
- Pathology Unit, San Raffaele Scientific Institute, IRCCS, 20132 Milan, Italy; (M.G.C.); (L.P.); (G.A.); (G.G.); (C.D.)
| |
Collapse
|
27
|
Decoding the Evolutionary Response to Ensartinib in Patients With ALK-Positive NSCLC by Dynamic Circulating Tumor DNA Sequencing. J Thorac Oncol 2021; 16:827-839. [PMID: 33588113 DOI: 10.1016/j.jtho.2021.01.1615] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/10/2020] [Accepted: 01/09/2021] [Indexed: 02/05/2023]
Abstract
INTRODUCTION By implementing dynamic circulating tumor DNA (ctDNA) analysis, we explored the impact of TP53 mutations on tumor evolution and resistance mechanisms to ensartinib in patients with ALK-positive NSCLC. METHODS In a multicenter phase 2 trial, patients with ALK-positive NSCLC who progressed on crizotinib were treated with ensartinib. Blood samples for ctDNA analysis were collected at baseline, cycle 3 day 1, and progression disease (PD) and analyzed with a 212-gene panel. RESULTS A total of 440 samples were collected from 168 patients. Baseline TP53 mutations (20.2%) significantly correlated with inferior progression-free survival (4.2 mo versus 11.7 mo, p < 0.0001). Patients with TP53 mutations had higher mutation load than those without TP53 mutations at baseline (13.79 ± 3.72 versus 4.67 ± 0.39, p < 0.001). Although there was no significant difference in mutation load between these groups at cycle 3 day 1 (5.89 ± 2.25 versus 3.72 ± 0.62, p = 0.425), patients with mutated TP53 developed more mutations at PD (7.07 ± 1.25 versus 3.20 ± 0.33, p = 0.003). Frequency and abundance of secondary ALK mutations G1269A, G1202R, and E1210K increased markedly at PD than baseline. In patients without secondary ALK mutations, we identified ALK-independent resistance mechanisms including bypass signaling activation, downstream effector protein reactivation, epithelial-mesenchymal transformation, and epigenetic dysregulation. CONCLUSIONS Our study highlighted the advantage of ctDNA analysis for monitoring tumor evolution. TP53 mutations promoted genetic evolution and accelerated occurrence of resistance. We also unveiled ALK-dependent resistance mechanisms, mainly by G1269A, G1202R, and E1210K mutations, and ALK-independent resistance mechanisms to ensartinib.
Collapse
|
28
|
Redl E, Sheibani-Tezerji R, Cardona CDJ, Hamminger P, Timelthaler G, Hassler MR, Zrimšek M, Lagger S, Dillinger T, Hofbauer L, Draganić K, Tiefenbacher A, Kothmayer M, Dietz CH, Ramsahoye BH, Kenner L, Bock C, Seiser C, Ellmeier W, Schweikert G, Egger G. Requirement of DNMT1 to orchestrate epigenomic reprogramming for NPM-ALK-driven lymphomagenesis. Life Sci Alliance 2021; 4:e202000794. [PMID: 33310759 PMCID: PMC7768196 DOI: 10.26508/lsa.202000794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
Malignant transformation depends on genetic and epigenetic events that result in a burst of deregulated gene expression and chromatin changes. To dissect the sequence of events in this process, we used a T-cell-specific lymphoma model based on the human oncogenic nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) translocation. We find that transformation of T cells shifts thymic cell populations to an undifferentiated immunophenotype, which occurs only after a period of latency, accompanied by induction of the MYC-NOTCH1 axis and deregulation of key epigenetic enzymes. We discover aberrant DNA methylation patterns, overlapping with regulatory regions, plus a high degree of epigenetic heterogeneity between individual tumors. In addition, ALK-positive tumors show a loss of associated methylation patterns of neighboring CpG sites. Notably, deletion of the maintenance DNA methyltransferase DNMT1 completely abrogates lymphomagenesis in this model, despite oncogenic signaling through NPM-ALK, suggesting that faithful maintenance of tumor-specific methylation through DNMT1 is essential for sustained proliferation and tumorigenesis.
Collapse
Affiliation(s)
- Elisa Redl
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | | | | | - Patricia Hamminger
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerald Timelthaler
- Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Melanie Rosalia Hassler
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Maša Zrimšek
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Sabine Lagger
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Dillinger
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics (LBI AD), Vienna, Austria
| | - Lorena Hofbauer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Kristina Draganić
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Andreas Tiefenbacher
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics (LBI AD), Vienna, Austria
| | - Michael Kothmayer
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Charles H Dietz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bernard H Ramsahoye
- Centre for Genetic and Experimental Medicine, Institute of Genomic and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
- Christian Doppler Laboratory for Applied Metabolomics (CDL-AM), Medical University of Vienna, Vienna, Austria
- Center for Biomarker Research in Medicine (CBmed), CoreLab 2, Medical University of Vienna, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christian Seiser
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gabriele Schweikert
- Max Planck Institute for Intelligent Systems, Tübingen, Germany
- Division of Computational Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics (LBI AD), Vienna, Austria
| |
Collapse
|
29
|
Bu L, Hameed NUF, Luo C, Hong P, Zhou X, Wang S, Wu S. Germline ALK variations are associated with a poor prognosis in glioma and IDH-wildtype glioblastoma. J Neurooncol 2021; 152:27-36. [PMID: 33486679 DOI: 10.1007/s11060-020-03676-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/12/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Glioma is the most common primary brain tumor. Clear classification is crucial for accurate diagnosis and individualized treatment. Histopathological characteristics and genetic alterations have shown to be related to prognosis and treatment response. Germline variants are important components of genetic alterations. However, the distribution of germline variations in glioma patients and their association with survival remain unknown. METHODS We carried out whole-exome sequencing on 99 cases to explore germline variants in glioma. We also analyzed the association of germline variants with clinicopathological features and other prognostic indicators. RESULTS All the glioma cases harbored rare germline variants. Germline ALK variants (gALK-Mut) were identified in 12/99 (12.12%) patients. The gALK-Mut patients had significantly shorter overall survival than germline ALK wildtype (gALK-WT) patients in the all glioma group (99 cases) and the subset of patients with IDH-wildtype glioblastoma (IDH-WT-GBM, 39 cases) (P = 0.013 and 0.027, respectively). The gALK-Mut patients also had higher frequency of BIRC5, PIK3CA and RPN1 somatic mutations than the gALK-WT patients in IDH-WT-GBM. Other confounding factors appeared to contribute to patient survival. The subgroup of patients in IDH-WT-GBM with gALK-Mut/TP53-Mut had worse prognosis than the gALK-WT/TP53-Mut subgroup (P = 0.031); The gALK-Mut/TERT-WT and gALK-Mut/TERT-Mut subgroups both had a worse prognosis than the gALK-WT/TERT-Mut subgroup (P = 0.031 and 0.018, respectively). CONCLUSIONS Our study revealed ALK variation was an independent indicator of poor prognosis in glioma and IDH-WT-GBM. It could be a promising biomarker and tractable therapeutic target for this deadly disease.
Collapse
Affiliation(s)
- Linghao Bu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - N U Farrukh Hameed
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Chen Luo
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Pengjie Hong
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Xiaoyu Zhou
- GenomiCare Biotechnology (Shanghai) Co. Ltd.,, Shanghai, China
| | - Shengzhou Wang
- GenomiCare Biotechnology (Shanghai) Co. Ltd.,, Shanghai, China
| | - Shuai Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China. .,Neurosurgical Institute of Fudan University, Shanghai, China. .,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China. .,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
| |
Collapse
|
30
|
Alidousty C, Duerbaum N, Wagener-Ryczek S, Baar T, Martelotto LG, Heydt C, Siemanowski J, Holz B, Binot E, Fassunke J, Merkelbach-Bruse S, Wolf J, Kron A, Buettner R, Schultheis AM. Prevalence and potential biological role of TERT amplifications in ALK translocated adenocarcinoma of the lung. Histopathology 2020; 78:578-585. [PMID: 32946634 DOI: 10.1111/his.14256] [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: 06/10/2020] [Accepted: 09/11/2020] [Indexed: 12/20/2022]
Abstract
AIMS The advent of specific ALK-targeting drugs has radically changed the outcome of patients with ALK translocated non-small-cell lung cancer (NSCLC). However, emerging resistance to treatment with ALK inhibitors in these patients remains a major concern. In previous studies, we analysed two ALK+ patient cohorts (TP53 wild-type/TP53 mutated) in terms of copy number alterations. All patients belonging to the TP53 wild-type group had mainly genetically stable genomes, with one exception showing chromosomal instability and amplifications of several gene loci, including TERT. Here, we aimed to determine the prevalence of TERT amplifications in these ALK+ lung cancer patients by analysing an independent cohort of 109 ALK translocated cases. We further analysed the copy numbers of numerous cancer-relevant genes and other genetic aberrations. METHODS AND RESULTS The prevalence of TERT amplifications was determined by means of FISH analyses. Copy numbers of 87 cancer-relevant genes were determined by NanoString nCounter® technology, FoundationOne® and lung-specific NGS panels in some of these TERT-amplified samples, and clinical data on patients with TERT-amplified tumours were collected. Our data revealed that five (4.6%) of all 109 analysed ALK+ patients harboured amplification of TERT and that these patients had genetically unstable genomes. CONCLUSIONS Our preliminary study shows that ALK+ adenocarcinomas should be evaluated in the context of their genomic background in order to more clearly understand and predict patients' individual course of disease.
Collapse
Affiliation(s)
| | - Nicolai Duerbaum
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | - Till Baar
- Faculty of Medicine, Institute of Medical Statistics and Computational Biology, University of Cologne, Cologne, Germany
| | | | - Carina Heydt
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Janna Siemanowski
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Barbara Holz
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Elke Binot
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Jana Fassunke
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | - Jürgen Wolf
- Network Genomic Medicine, Cologne, Germany.,Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,Center for Integrated Oncology Koeln Bonn, Cologne, Germany
| | - Anna Kron
- Network Genomic Medicine, Cologne, Germany.,Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,Center for Integrated Oncology Koeln Bonn, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany.,Network Genomic Medicine, Cologne, Germany.,Center for Integrated Oncology Koeln Bonn, Cologne, Germany
| | - Anne M Schultheis
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
31
|
Nygård L, Ahlborn LB, Persson GF, Chandrananda D, Langer JW, Fischer BM, Langer SW, Gabrielaite M, Kjær A, Rosenfeld N, Mouliere F, Østrup O, Vogelius IR, Bentzen SM. Circulating cell free DNA during definitive chemo-radiotherapy in non-small cell lung cancer patients - initial observations. PLoS One 2020; 15:e0231884. [PMID: 32343749 PMCID: PMC7188247 DOI: 10.1371/journal.pone.0231884] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 04/02/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The overall aim was to investigate the change over time in circulating cell free DNA (cfDNA) in patients with locally advanced non-small cell lung cancer (NSCLC) undergoing concurrent chemo-radiotherapy. Furthermore, to assess the possibility of detecting circulating cell free tumor DNA (ctDNA) using shallow whole genome sequencing (sWGS) and size selection. METHODS Ten patients were included in a two-phase study. The first four patients had blood samples taken prior to a radiation therapy (RT) dose fraction and at 30 minutes, 1 hour and 2 hours after RT to estimate the short-term dynamics of cfDNA concentration after irradiation. The remaining six patients had one blood sample taken on six treatment days 30 minutes post treatment to measure cfDNA levels. Presence of ctDNA as indicated by chromosomal aberrations was investigated using sWGS. The sensitivity of this method was further enhanced using in silico size selection. RESULTS cfDNA concentration from baseline to 120 min after therapy was stable within 95% tolerance limits of +/- 2 ng/ml cfDNA. Changes in cfDNA were observed during therapy with an apparent qualitative difference between adenocarcinoma (average increase of 0.69 ng/ml) and squamous cell carcinoma (average increase of 4.0 ng/ml). Tumor shrinkage on daily cone beam computer tomography scans during radiotherapy did not correlate with changes in concentration of cfDNA. CONCLUSION Concentrations of cfDNA remain stable during the first 2 hours after an RT fraction. However, based on the sWGS profiles, ctDNA represented only a minor fraction of cfDNA in this group of patients. The detection sensitivity of genomic alterations in ctDNA strongly increases by applying size selection.
Collapse
Affiliation(s)
- Lotte Nygård
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lise B. Ahlborn
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Gitte F. Persson
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Dineika Chandrananda
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, England, United Kingdom
| | - Jonathan W. Langer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Barbara M. Fischer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
- PET Centre, School of Biomedical Engineering and Imaging Sciences, Kings College London, St Thomas' Hospital, London, England, United Kingdom
| | - Seppo W. Langer
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Miglė Gabrielaite
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, England, United Kingdom
| | - Florent Mouliere
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Olga Østrup
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ivan R. Vogelius
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Søren M. Bentzen
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, University of Maryland Greenebaum Comprehensive Cancer Center, and University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
32
|
Qin K, Hou H, Liang Y, Zhang X. Prognostic value of TP53 concurrent mutations for EGFR- TKIs and ALK-TKIs based targeted therapy in advanced non-small cell lung cancer: a meta-analysis. BMC Cancer 2020; 20:328. [PMID: 32299384 PMCID: PMC7164297 DOI: 10.1186/s12885-020-06805-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 03/30/2020] [Indexed: 12/26/2022] Open
Abstract
Background The prognostic significance of TP53 concurrent mutations in patients with epidermal growth factor receptor (EGFR)- or anaplastic lymphoma kinase (ALK)- mutated advanced non–small-cell lung cancer (NSCLC) who received EGFR-tyrosine kinase inhibitors (TKIs) or ALK-TKIs based targeted therapy remains controversial. Therefore, the present meta-analysis was performed to investigate the association between TP53 concurrent mutations and prognosis of patients with advanced NSCLC undergoing EGFR-TKIs or ALK-TKIs treatments. Methods Eligible studies were identified by searching the online databases PubMed, Embase, Medline, The Cochrane library and Web of Science. Hazard ratios (HRs) with 95% confidence intervals (CIs) were calculated to clarify the correlation between TP53 mutation status and prognosis of patients. This meta-analysis was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. Results In total, 15 studies with 1342 patients were included for final analysis. Overall, concurrent TP53 mutation was associated with unfavorable progression-free survival (PFS) (HR = 1.88, 95%CI: 1.59–2.23, p < 0.001, I2 = 0.0%, P = 0.792) and overall survival (OS) (HR = 1.92, 95%CI: 1.55–2.38, p < 0.001, I2 = 0.0%, P = 0.515). Subgroup analysis based on type of targeted therapy (EGFR-TKIs or ALK-TKIs, pathological type of cancer (adenocarcinoma only or all NSCLC subtypes) and line of treatment (first-line only or all lines) all showed that TP53 mutations was associated with shorter survivals of patients with EGFR-TKIs or ALK-TKIs treatments. Particularly, in patients with first-line EGFR-TKIs treatment, significantly poorer prognosis was observed in patients with TP53 concurrent mutations (pooled HR for PFS: 1.69, 95% CI 1.25–2.27, P < 0.001, I2 = 0.0%, P = 0.473; pooled HR for OS: 1.94, 95% CI 1.36–2.76, P < 0.001, I2 = 0.0%, P = 0.484). Begg’s funnel plots and Egger’s tests indicated no significant publication bias in this study. Conclusions This meta-analysis indicated that concurrent TP53 mutations was a negative prognostic factor and associated with poorer outcomes of patients with EGFR-TKIs or ALK-TKIs treatments in advanced NSCLC. In addition, our study provided evidence that TP53 mutations might be involved in primary resistance to EGFR-TKIs treatments in patients with sensitive EGFR mutations in advanced NSCLC.
Collapse
Affiliation(s)
- Kang Qin
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266005, Shandong Province, China
| | - Helei Hou
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266005, Shandong Province, China
| | - Yu Liang
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266005, Shandong Province, China
| | - Xiaochun Zhang
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266005, Shandong Province, China.
| |
Collapse
|
33
|
Wagner BJ, Buettner R. [Immunohistochemical and molecular pathological diagnosis of lung carcinoma]. DER PATHOLOGE 2020; 41:3-13. [PMID: 31932944 DOI: 10.1007/s00292-019-00732-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The therapy of lung cancer has revolutionarily changed within the last 15 years. The prognosis of patients has dramatically improved due to targeted therapies, for instance tyrosine kinase inhibitors (TKI). Current recruiting studies are testing new MET-, KRAS-, ROS1-, RET- and NTRK-inhibitors. The first clinical data are promising, emphasizing that it remains a future challenge for pathologists and oncologists to keep an eye on all facets of therapy options. Today, precise lung cancer classification via immunohistochemistry and molecular pathology is crucial for the therapy and prognosis of patients. Further, new biopsy technologies lead to very small tumor tissue samples and cytological samples of high diagnostic quality. Therefore, the complexity of diagnosis needs a strategic procedure to minimize loss of tissue material. This assay gives an overview of efficient and target-orientated diagnostic procedures in consideration of current clinical trials.
Collapse
Affiliation(s)
- Britta Janina Wagner
- Institut für Pathologie, Universitätsklinikum und Centrum für Integrierte Onkologie (CIO) Köln, Kerpener Straße 62, 50937, Köln, Deutschland.
| | - Reinhard Buettner
- Institut für Pathologie, Universitätsklinikum und Centrum für Integrierte Onkologie (CIO) Köln, Kerpener Straße 62, 50937, Köln, Deutschland
| |
Collapse
|
34
|
YAP1 mediates survival of ALK-rearranged lung cancer cells treated with alectinib via pro-apoptotic protein regulation. Nat Commun 2020; 11:74. [PMID: 31900393 PMCID: PMC6941996 DOI: 10.1038/s41467-019-13771-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Despite the promising clinical efficacy of the second-generation anaplastic lymphoma kinase (ALK) inhibitor alectinib in patients with ALK-rearranged lung cancer, some tumor cells survive and eventually relapse, which may be an obstacle to achieving a cure. Limited information is currently available on the mechanisms underlying the initial survival of tumor cells against alectinib. Using patient-derived cell line models, we herein demonstrate that cancer cells survive a treatment with alectinib by activating Yes-associated protein 1 (YAP1), which mediates the expression of the anti-apoptosis factors Mcl-1 and Bcl-xL, and combinatorial inhibition against both YAP1 and ALK provides a longer tumor remission in ALK-rearranged xenografts when compared with alectinib monotherapy. These results suggest that the inhibition of YAP1 is a candidate for combinatorial therapy with ALK inhibitors to achieve complete remission in patients with ALK-rearranged lung cancer.
Collapse
|
35
|
Wang T, Sun H, Bao Y, En R, Tian Y, Zhao W, Jia L. POM121 overexpression is related to a poor prognosis in colorectal cancer. Expert Rev Mol Diagn 2019; 20:345-353. [PMID: 31858845 DOI: 10.1080/14737159.2020.1707670] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: Nuclear pore membrane protein 121 (POM121) plays a crucial role in nucleocytoplasmic transport, but its significance in tumorigenesis and the progression of colorectal cancer (CRC) remains unknown. The aim of this study was to evaluate the relationship between POM121 and CRC.Methods: POM121 expression in colorectal tissues was analyzed at both the gene and protein levels. We investigated the connection between POM121 expression and clinicopathological features, as well as overall survival. A gene set enrichment analysis (GSEA) was performed, and a protein-protein interaction (PPI) network was constructed to determine the mechanism of POM121 in CRC.Results: CRC tissues displayed a striking increase in POM121 expression compared with colonitis and pericarcinomatous mucosa tissues (66.61% vs 24.36% vs 24.11%, respectively, p < 0.0167). POM121 overexpression was significantly associated with lymph node metastasis, distant metastasis, TNM stage, venous invasion, perineural invasion, preoperative CEA and CA19-9 levels, and Ki67 expression. CRC patients with high POM121 levels tended to have poor overall survival rates. POM121 may participate in the regulation of the cell cycle and DNA repair in CRC.Conclusions: Our results suggest that POM121 has the potential to serve as a novel prognostic biomarker in CRC patients.
Collapse
Affiliation(s)
- Tengqi Wang
- Cancer Center, Banyannur Hospital, Bayannur, China
| | - Haibin Sun
- Gastrointestinal Surgery, Banyannur Hospital, Bayannur, China
| | | | - Riletu En
- Gastrointestinal Surgery, Banyannur Hospital, Bayannur, China
| | - Yongjing Tian
- Gastrointestinal Surgery, Banyannur Hospital, Bayannur, China
| | - Wei Zhao
- Department of Pathology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Lizhou Jia
- Cancer Center, Banyannur Hospital, Bayannur, China
| |
Collapse
|
36
|
A Small Compound KJ-28d Enhances the Sensitivity of Non-Small Cell Lung Cancer to Radio- and Chemotherapy. Int J Mol Sci 2019; 20:ijms20236026. [PMID: 31795418 PMCID: PMC6928747 DOI: 10.3390/ijms20236026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/22/2019] [Accepted: 11/29/2019] [Indexed: 01/07/2023] Open
Abstract
We previously reported on a poly (ADP-ribose) polymerase (PARP) 1/2 inhibitor N-(3-(hydroxycarbamoyl)phenyl)carboxamide (designated KJ-28d), which increased the death of human ovarian cancer BRCA1-deficient SNU-251 cells. In the present study, we further investigated the antitumor activities of KJ-28d in BRCA-proficient non-small cell lung cancer (NSCLC) cells to expand the use of PARP inhibitors. KJ-28d significantly inhibited the growth of NSCLC cells in vitro and in vivo, and induced DNA damage and reactive oxygen species in A549 and H1299 cells. Combined treatment with KJ-28d and ionizing radiation led to increased DNA damage responses in A549 and H1299 cells compared to KJ-28d or ionizing radiation alone, resulting in apoptotic cell death. Moreover, the combination of KJ-28d plus a DNA-damaging therapeutic agent (carboplatin, cisplatin, paclitaxel, or doxorubicin) synergistically inhibited cell proliferation, compared to either drug alone. Taken together, the findings demonstrate the potential of KJ-28d as an effective anti-cancer therapeutic agent for BRCA-deficient and -proficient cancer cells. KJ-28d might have potential as an adjuvant when used in combination with radiotherapy or DNA-damaging agents, pending further investigations.
Collapse
|
37
|
Kron A, Alidousty C, Scheffler M, Merkelbach-Bruse S, Seidel D, Riedel R, Ihle MA, Michels S, Nogova L, Fassunke J, Heydt C, Kron F, Ueckeroth F, Serke M, Krüger S, Grohe C, Koschel D, Benedikter J, Kaminsky B, Schaaf B, Braess J, Sebastian M, Kambartel KO, Thomas R, Zander T, Schultheis AM, Büttner R, Wolf J. Impact of TP53 mutation status on systemic treatment outcome in ALK-rearranged non-small-cell lung cancer. Ann Oncol 2019; 29:2068-2075. [PMID: 30165392 PMCID: PMC6225899 DOI: 10.1093/annonc/mdy333] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background We analyzed whether co-occurring mutations influence the outcome of systemic therapy in ALK-rearranged non-small-cell lung cancer (NSCLC). Patients and methods ALK-rearranged stage IIIB/IV NSCLC patients were analyzed with next-generation sequencing and fluorescence in situ hybridization analyses on a centralized diagnostic platform. Median progression-free survival (PFS) and overall survival (OS) were determined in the total cohort and in treatment-related sub-cohorts. Cox regression analyses were carried out to exclude confounders. Results Among 216 patients with ALK-rearranged NSCLC, the frequency of pathogenic TP53 mutations was 23.8%, while other co-occurring mutations were rare events. In ALK/TP53 co-mutated patients, median PFS and OS were significantly lower compared with TP53 wildtype patients [PFS 3.9 months (95% CI: 2.4–5.6) versus 10.3 months (95% CI: 8.6–12.0), P < 0.001; OS 15.0 months (95% CI: 5.0–24.9) versus 50.0 months (95% CI: 22.9–77.1), P = 0.002]. This difference was confirmed in all treatment-related subgroups including chemotherapy only [PFS first-line chemotherapy 2.6 months (95% CI: 1.3–4.1) versus 6.2 months (95% CI: 1.8–10.5), P = 0.021; OS 2.0 months (95% CI: 0.0–4.6) versus 9.0 months (95% CI: 6.1–11.9), P = 0.035], crizotinib plus chemotherapy [PFS crizotinib 5.0 months (95% CI: 2.9–7.2) versus 14.0 months (95% CI: 8.0–20.1), P < 0.001; OS 17.0 months (95% CI: 6.7–27.3) versus not reached, P = 0.049] and crizotinib followed by next-generation ALK-inhibitor [PFS next-generation inhibitor 5.4 months (95% CI: 0.1–10.7) versus 9.9 months (95% CI: 6.4–13.5), P = 0.039; OS 7.0 months versus 50.0 months (95% CI: not reached), P = 0.001). Conclusions In ALK-rearranged NSCLC co-occurring TP53 mutations predict an unfavorable outcome of systemic therapy. Our observations encourage future research to understand the underlying molecular mechanisms and to improve treatment outcome of the ALK/TP53 co-mutated subgroup.
Collapse
Affiliation(s)
- A Kron
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - C Alidousty
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - M Scheffler
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - S Merkelbach-Bruse
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - D Seidel
- Center for Integrated Oncology Köln Bonn, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - R Riedel
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - M A Ihle
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - S Michels
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - L Nogova
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - J Fassunke
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - C Heydt
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - F Kron
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - F Ueckeroth
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - M Serke
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Lungenklinik Hemer des Deutschen Gemeinschafts-Diakonieverbandes GmbH, Hemer, Germany
| | - S Krüger
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Florence Nightingale Hospital, Düsseldorf, Germany
| | - C Grohe
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Evangelische Lungenklinik Berlin (Paul Gerhardt Diakonie), Berlin, Germany
| | - D Koschel
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Fachkrankenhaus Coswig, Coswig, Germany
| | - J Benedikter
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Klinikum Bogenhausen, Munich, Germany
| | - B Kaminsky
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Krankenhaus Bethanien, Solingen, Germany
| | - B Schaaf
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Center, Klinikum Dortmund GmbH, Dortmund, Germany
| | - J Braess
- Network Genomic Medicine, Cologne, Germany; Department of Oncology and Hematology, Krankenhaus Barmherzige Brueder, Regensburg, Germany
| | - M Sebastian
- Network Genomic Medicine, Cologne, Germany; Department of Oncology and Hematology, University Hospital Frankfurt (Johannes-Wolfgang Goethe Institute), Frankfurt am Main, Germany
| | - K-O Kambartel
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Bethanien Hospital Moers-Lungenzentrum, Moers, Germany
| | - R Thomas
- Network Genomic Medicine, Cologne, Germany; Cologne Center for Genomics, University Hospital of Cologne, Cologne, Germany
| | - T Zander
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - A M Schultheis
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - R Büttner
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - J Wolf
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany.
| |
Collapse
|
38
|
Heydt C, Becher AK, Wagener-Ryczek S, Ball M, Schultheis AM, Schallenberg S, Rüsseler V, Büttner R, Merkelbach-Bruse S. Comparison of in situ and extraction-based methods for the detection of MET amplifications in solid tumors. Comput Struct Biotechnol J 2019; 17:1339-1347. [PMID: 31762957 PMCID: PMC6861603 DOI: 10.1016/j.csbj.2019.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 01/22/2023] Open
Abstract
In EGFR-treatment naive NSCLC patients, high-level MET amplification is detected in approximately 2-3% and is considered as adverse prognostic factor. Currently, clinical trials with two different inhibitors, capmatinib and tepotinib, are under way both defining different inclusion criteria regarding MET amplification from proven amplification only to defining an exact MET copy number. Here, 45 patient samples, including 10 samples without MET amplification, 5 samples showing a low-level MET amplification, 10 samples with an intermediate-level MET amplification, 10 samples having a high-level MET amplification by a MET/CEN7 ratio ≥2.0 and 10 samples showing a high-level MET amplification with GCN ≥6, were evaluated by MET FISH, MET IHC, a ddPCR copy number assay, a NanoString nCounter copy number assay and an amplicon-based parallel sequencing. The MET IHC had the best concordance with MET FISH followed by the NanoString copy number assay, the ddPCR copy number assay and the custom amplicon-based parallel sequencing assays. The concordance was higher in the high-level amplified cohorts than in the low- and intermediate-level amplified cohorts. In summary, currently extraction-based methods cannot replace the MET FISH for the detection of low-level, intermediate-level and high-level MET amplifications, as the number of false negative results is very high. Only for the detection of high-level amplified samples with a gene copy number ≥6 extraction-based methods are a reliable alternative.
Collapse
Affiliation(s)
- Carina Heydt
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Ann-Kathrin Becher
- Institute of Pathology, University Hospital Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Svenja Wagener-Ryczek
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Markus Ball
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Anne M. Schultheis
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Simon Schallenberg
- Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Vanessa Rüsseler
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
39
|
Skoulidis F, Heymach JV. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat Rev Cancer 2019; 19:495-509. [PMID: 31406302 PMCID: PMC7043073 DOI: 10.1038/s41568-019-0179-8] [Citation(s) in RCA: 586] [Impact Index Per Article: 117.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2019] [Indexed: 02/07/2023]
Abstract
The impressive clinical activity of small-molecule receptor tyrosine kinase inhibitors for oncogene-addicted subgroups of non-small-cell lung cancer (for example, those driven by activating mutations in the gene encoding epidermal growth factor receptor (EGFR) or rearrangements in the genes encoding the receptor tyrosine kinases anaplastic lymphoma kinase (ALK), ROS proto-oncogene 1 (ROS1) and rearranged during transfection (RET)) has established an oncogene-centric molecular classification paradigm in this disease. However, recent studies have revealed considerable phenotypic diversity downstream of tumour-initiating oncogenes. Co-occurring genomic alterations, particularly in tumour suppressor genes such as TP53 and LKB1 (also known as STK11), have emerged as core determinants of the molecular and clinical heterogeneity of oncogene-driven lung cancer subgroups through their effects on both tumour cell-intrinsic and non-cell-autonomous cancer hallmarks. In this Review, we discuss the impact of co-mutations on the pathogenesis, biology, microenvironmental interactions and therapeutic vulnerabilities of non-small-cell lung cancer and assess the challenges and opportunities that co-mutations present for personalized anticancer therapy, as well as the expanding field of precision immunotherapy.
Collapse
Affiliation(s)
- Ferdinandos Skoulidis
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - John V Heymach
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
40
|
Büttner R, Wolf J, Kron A. [The national Network Genomic Medicine (nNGM) : Model for innovative diagnostics and therapy of lung cancer within a public healthcare system]. DER PATHOLOGE 2019; 40:276-280. [PMID: 31101971 DOI: 10.1007/s00292-019-0605-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Since 2012, the Network Genomic Medicine (NGM) has been providing a large number of lung cancer patients from referring partner sites with comprehensive molecular-pathological diagnostics on the single diagnostic platform at the University Hospital Cologne. In addition, the network headquarters in Cologne interprets the findings in close interdisciplinary coordination between pathologists and oncologists, provides information on innovative treatment options, and evaluates the personalized therapies using the central database. As part of one of its largest single grants in 2018, the German Cancer Aid (DKH) rolled out this interdisciplinary and intersectoral care model to all existing DKH-funded German comprehensive cancer centers at the time of the initial application. GOAL Presentation of the treatment reality within the national Network Genomic Medicine (nNGM) with its core elements and actors (network centers and intersectoral clinical partners sites). METHODS This article is based on our own experience in NGM and nNGM and includes a summary of the currently applicable guidelines for reimbursement and an overview of the treatment landscape in the field of molecular-pathological diagnostics in Germany. RESULTS The focus of nNGM is on the implementation of innovative molecular diagnostics and personalized therapy in broad clinical routine in Germany. This is enabled by developing molecular-pathological diagnostics within the network centers on an ongoing basic, interdisciplinary counseling of referring partner sites, offering innovative clinical trials, and performing central evaluation. In particular, a focus of nNGM is the development of regional networks to treat the affected lung cancer patients close to home at the partner sites whenever possible. DISCUSSION Interdisciplinary teams are essential for the success of a broad implementation of molecular-pathological diagnostics. nNGM addresses a severe deficit in German lung cancer care and in the future will be expanded to further network centers while meeting the defined quality criteria.
Collapse
Affiliation(s)
- R Büttner
- Institut für Pathologie, Centrum für Integrierte Onkologie Aachen Bonn Köln Düsseldorf, Universität zu Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
| | - J Wolf
- Klinik I für Innere Medizin, Centrum für Integrierte Onkologie Aachen Bonn Köln Düsseldorf, Universität zu Köln, Köln, Deutschland
| | - A Kron
- Klinik I für Innere Medizin, Centrum für Integrierte Onkologie Aachen Bonn Köln Düsseldorf, Universität zu Köln, Köln, Deutschland
| | | |
Collapse
|
41
|
Michels S, Massutí B, Schildhaus HU, Franklin J, Sebastian M, Felip E, Grohé C, Rodriguez-Abreu D, Abdulla DS, Bischoff H, Brandts C, Carcereny E, Corral J, Dingemans AMC, Pereira E, Fassunke J, Fischer RN, Gardizi M, Heukamp L, Insa A, Kron A, Menon R, Persigehl T, Reck M, Riedel R, Rothschild SI, Scheel AH, Scheffler M, Schmalz P, Smit EF, Limburg M, Provencio M, Karachaliou N, Merkelbach-Bruse S, Hellmich M, Nogova L, Büttner R, Rosell R, Wolf J. Safety and Efficacy of Crizotinib in Patients With Advanced or Metastatic ROS1-Rearranged Lung Cancer (EUCROSS): A European Phase II Clinical Trial. J Thorac Oncol 2019; 14:1266-1276. [DOI: 10.1016/j.jtho.2019.03.020] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/26/2019] [Accepted: 03/01/2019] [Indexed: 12/21/2022]
|
42
|
Next-generation Sequencing for ALK and ROS1 Rearrangement Detection in Patients With Non-small-cell Lung Cancer: Implications of FISH-positive Patterns. Clin Lung Cancer 2019; 20:e421-e429. [PMID: 30898567 DOI: 10.1016/j.cllc.2019.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/13/2019] [Accepted: 02/17/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Detection of ALK and ROS1 gene rearrangements in non-small-cell lung cancer is required for directing patient care. Although fluorescence in situ hybridization (FISH) and immunohistochemistry have been established as gold standard methods, next-generation sequencing (NGS) platforms are called to be at least equally successful. Comparison of these methods for translation into daily use is currently under investigation. PATIENTS AND METHODS Forty non-small-cell lung cancer paraffin-embedded samples with previous ALK (n = 33) and ROS1 (n = 7) FISH results were examined with the Oncomine Focus Assay and tested for ALK and ROS1 immunoreactivity. Clinical implications of concurrent molecular alterations and concordance between methods were evaluated. RESULTS NGS was successful in 32 (80%) cases: 25 ALK and 7 ROS1. Few concomitant alterations were detected: 1 ALK rearranged case had an ALK p.L1196M-resistant mutation, 4 had CDK4, MYC, and/or ALK amplifications, and 1 ROS1 rearranged case showed a FGFR4 amplification. Comparison between techniques revealed 5 (16%) discordant cases that had lower progression-free survival than concordant cases: 7.6 (95% confidence interval, 2.2-13) versus 19.4 (95% confidence interval, 10.1-28.6). Remarkably, 4 of these cases had isolated 3' signal FISH pattern (P = .026). CONCLUSION Our data support that the identification of 3' isolated signal FISH pattern in ALK and ROS1 cases might suggest a false-positive result. NGS seems a reliable technique to assess ALK and ROS1 rearrangements, offering the advantage over immunohistochemistry of detecting other molecular alterations with potential therapeutic implications.
Collapse
|
43
|
Alidousty C, Baar T, Heydt C, Wagener-Ryczek S, Kron A, Wolf J, Buettner R, Schultheis AM. Advance of theragnosis biomarkers in lung cancer: from clinical to molecular pathology and biology. J Thorac Dis 2019; 11:S3-S8. [PMID: 30775023 DOI: 10.21037/jtd.2018.12.03] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
One distinct molecular subtype of non-small cell lung cancer (NSCLC) is defined by rearrangement of the anaplastic lymphoma kinase (ALK). The increasing knowledge over the last years has enabled the continuous improvement of ALK inhibitors; however, resistance in these patients remains a major concern. In this review, we summarize recent findings in ALK+-adenocarcinoma of the lung, highlighting the role of TP53 mutations in this specific cancer type and suggest new diagnostic strategies for the future, in order to improve patient's outcome.
Collapse
Affiliation(s)
| | - Till Baar
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Carina Heydt
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | - Anna Kron
- Network Genomic Medicine, Cologne, Germany.,Lung Cancer Group Cologne, Department I for Internal Medicine, University Hospital of Cologne, Cologne, Germany.,Center for Integrated Oncology Cologne Bonn, Cologne, Germany
| | - Juergen Wolf
- Network Genomic Medicine, Cologne, Germany.,Lung Cancer Group Cologne, Department I for Internal Medicine, University Hospital of Cologne, Cologne, Germany.,Center for Integrated Oncology Cologne Bonn, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany.,Network Genomic Medicine, Cologne, Germany.,Lung Cancer Group Cologne, Department I for Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | | |
Collapse
|
44
|
TP53 mutations as potential prognostic markers for specific cancers: analysis of data from The Cancer Genome Atlas and the International Agency for Research on Cancer TP53 Database. J Cancer Res Clin Oncol 2018; 145:625-636. [PMID: 30542790 DOI: 10.1007/s00432-018-2817-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/05/2018] [Indexed: 10/27/2022]
Abstract
PURPOSE Mutations in the tumor suppressor gene TP53 are associated with a variety of cancers. Therefore, it is important to know the occurrence and prognostic effects of TP53 mutations in certain cancers. METHODS Over 29,000 cases from the April 2016 release of the International Agency for Research on Cancer (IARC) TP53 Database were analyzed to determine the distribution of germline and somatic mutations in the TP53 gene. Subsequently, 7,893 cancer cases were compiled in cBioPortal for Cancer Genomics from the 33 most recent The Cancer Genome Atlas (TCGA) studies to determine the prevalence of TP53 mutations in cancers and their effects on survival and disease-free survival times. RESULTS The data were analyzed, and it was found that the majority of TP53 mutations were missense and the major mutational hotspots were located at codons 248, 273, 175, and 245 in exons 4-8 for somatic mutations with the addition of codon 337 and other mutations in exons 9-10 for germline mutations. Out of 33 TGCA studies, the effects of TP53 mutations were statistically significant in nine cancers (lung adenocarcinoma, hepatocellular carcinoma, head and neck squamous cell carcinoma, acute myeloid leukemia, clear cell renal cell carcinoma (RCC), papillary RCC, chromophobe RCC, uterine endometrial carcinoma, and thymoma) for survival time and in five cancers (pancreatic adenocarcinoma, hepatocellular carcinoma, chromophobe RCC, acute myeloid leukemia, and thymoma) for disease-free survival time. It was also found that the most common p53 mutation in hepatocellular carcinomas (R249S) was a much better indicator for poor prognosis than TP53 mutations as a whole. In addition, in cases of ovarian serous cystadenocarcinoma, the co-occurrence of TP53 and BRCA mutations resulted in longer survival and disease-free survival times than the presence of neither TP53 nor BRCA mutations. CONCLUSION TP53 mutations are potential prognostic markers that can be used to further improve the accuracy of predicting survival and disease-free survival times of cancer patients.
Collapse
|
45
|
Christopoulos P, Kirchner M, Bozorgmehr F, Endris V, Elsayed M, Budczies J, Ristau J, Penzel R, Herth F, Heussel C, Eichhorn M, Muley T, Meister M, Fischer J, Rieken S, Lasitschka F, Bischoff H, Sotillo R, Schirmacher P, Thomas M, Stenzinger A. Identification of a highly lethal V3+TP53+subset in ALK+lung adenocarcinoma. Int J Cancer 2018; 144:190-199. [DOI: 10.1002/ijc.31893] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 12/24/2022]
Affiliation(s)
- P. Christopoulos
- Department of Thoracic Oncology; Thoraxklinik at Heidelberg University Hospital; Heidelberg Germany
- Translational Lung Research Center Heidelberg (TLRC-H); Heidelberg Germany, member of the German Center for Lung Research (DZL)
| | - M. Kirchner
- Institute of Pathology, Heidelberg University Hospital; Heidelberg Germany
| | - F. Bozorgmehr
- Department of Thoracic Oncology; Thoraxklinik at Heidelberg University Hospital; Heidelberg Germany
- Department of Radiation Oncology; Heidelberg University Hospital; Heidelberg Germany
| | - V. Endris
- Institute of Pathology, Heidelberg University Hospital; Heidelberg Germany
| | - M. Elsayed
- Department of Thoracic Oncology; Thoraxklinik at Heidelberg University Hospital; Heidelberg Germany
| | - J. Budczies
- Institute of Pathology, Heidelberg University Hospital; Heidelberg Germany
| | - J. Ristau
- Department of Radiation Oncology; Heidelberg University Hospital; Heidelberg Germany
| | - R. Penzel
- Institute of Pathology, Heidelberg University Hospital; Heidelberg Germany
| | - F.J. Herth
- Translational Lung Research Center Heidelberg (TLRC-H); Heidelberg Germany, member of the German Center for Lung Research (DZL)
- Department of Pneumology; Thoraxklinik at Heidelberg University Hospital; Heidelberg Germany
| | - C.P. Heussel
- Translational Lung Research Center Heidelberg (TLRC-H); Heidelberg Germany, member of the German Center for Lung Research (DZL)
- Department of Diagnostic and Interventional Radiology with Nuclear Medicine; Thoraxklinik at Heidelberg University Hospital; Heidelberg Germany
| | - M. Eichhorn
- Translational Lung Research Center Heidelberg (TLRC-H); Heidelberg Germany, member of the German Center for Lung Research (DZL)
- Department of Surgery; Thoraxklinik at Heidelberg University Hospital; Heidelberg Germany
| | - T. Muley
- Translational Lung Research Center Heidelberg (TLRC-H); Heidelberg Germany, member of the German Center for Lung Research (DZL)
- Translational Research Unit; Thoraxklinik at Heidelberg University Hospital; Heidelberg Germany
| | - M. Meister
- Translational Lung Research Center Heidelberg (TLRC-H); Heidelberg Germany, member of the German Center for Lung Research (DZL)
- Translational Research Unit; Thoraxklinik at Heidelberg University Hospital; Heidelberg Germany
| | - J.R. Fischer
- Department of Thoracic Oncology; Lungenklinik Löwenstein; Löwenstein Germany
| | - S. Rieken
- Department of Radiation Oncology; Heidelberg University Hospital; Heidelberg Germany
| | - F. Lasitschka
- Translational Lung Research Center Heidelberg (TLRC-H); Heidelberg Germany, member of the German Center for Lung Research (DZL)
- Institute of Pathology, Heidelberg University Hospital; Heidelberg Germany
| | - H. Bischoff
- Department of Thoracic Oncology; Thoraxklinik at Heidelberg University Hospital; Heidelberg Germany
| | - R. Sotillo
- Translational Lung Research Center Heidelberg (TLRC-H); Heidelberg Germany, member of the German Center for Lung Research (DZL)
- Division of Molecular Thoracic Oncology; German Cancer Research Center; Heidelberg Germany
| | - P. Schirmacher
- Institute of Pathology, Heidelberg University Hospital; Heidelberg Germany
| | - M. Thomas
- Department of Thoracic Oncology; Thoraxklinik at Heidelberg University Hospital; Heidelberg Germany
- Translational Lung Research Center Heidelberg (TLRC-H); Heidelberg Germany, member of the German Center for Lung Research (DZL)
| | - A. Stenzinger
- Translational Lung Research Center Heidelberg (TLRC-H); Heidelberg Germany, member of the German Center for Lung Research (DZL)
- Institute of Pathology, Heidelberg University Hospital; Heidelberg Germany
| |
Collapse
|